You are on page 1of 2486

RETINA

Editor-In-Chief
Stephen J. Ryan MD
President, Doheny Eye Institute, Los Angeles, CA, USA

Volume One
Part 1 Retinal Imaging and Diagnostics
Edited by
SriniVas R. Sadda MD

Part 2 Basic Science and Translation to Therapy


Edited by
David R. Hinton MD

Volume Two
Medical Retina
Edited by
Andrew P. Schachat MD and SriniVas R. Sadda MD

Volume Three
Part 1 Surgical Retina
Edited by
C. P. Wilkinson MD and Peter Wiedemann MD

Part 2 Tumors of the Retina, Choroid, and Vitreous


Edited by
Andrew P. Schachat MD
Copyright © 2013, 2006, 2001, 1994, 1989 by Saunders, an imprint of Elsevier Inc.
Chapter 15: “Function and Anatomy of the Mammalian Retina” by Ronald G. Gregg, Maureen A. McCall, Stephen C. Massey:
Stephen C. Massey retains copyright to his portion of the contribution and his original figures.
Chapter 25: “Inflammatory Response and Mediators in Retinal Injury”: Chapter is in the public domain.
Chapter 66: “Neovascular (Exudative or “Wet”) Age-Related Macular Degeneration”: Neil M. Bressler’s figures and tables
© Johns Hopkins University
Chapter 77: “Autoimmune Retinopathies”: Chapter is in the public domain.
Video: “Peripheral Retinal Detachment during Macular Hole Surgery”: Stanley Chang retains copyright to his original video.
Video: “Optimal Procedures For Retinal Detachment Repair [Cryo (vity air), Fluid air2, Scleral sutures]; © EyeMovies Ltd

First edition 1989


Second edition 1994
Third edition 2001
Fourth edition 2006
Fifth edition 2013

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including
photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher.
Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with
organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.
elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may
be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding,
changes in research methods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any
information, methods, compounds, or experiments described herein. In using such information or methods they should be
mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the most current information
provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to verify the recommended
dose or formula, the method and duration of administration, and contraindications. It is the responsibility of practitioners,
relying on their own experience and knowledge of their patients, to make diagnoses, to determine dosages and the best
treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any
injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or
operation of any methods, products, instructions, or ideas contained in the material herein.

British Library Cataloguing in Publication Data

Retina.
  1.  Retina–Diseases.  2.  Retina–Surgery.  3.  Retina–Physiology.
  I. Ryan, Stephen J., 1940-617.7′35-dc23
  ISBN-13: 9781455707379

Ebook ISBN: 9781455737802


Printed in China
Last digit is the print number: 9  8  7  6  5  4  3  2

The
Working together to grow publisher’s
policy is to use
libraries in developing countries paper manufactured
from sustainable forests
www.elsevier.com | www.bookaid.org | www.sabre.org

Senior Content Strategist: Russell Gabbedy


Content Development Specialist: Nani Clansey
Content Coordinator: Emma Cole/Trinity Hutton/Sam Crowe/Humayra Rahman
Project Manager: Caroline Jones
Design: Miles Hitchen
Illustration Manager: Jennifer Rose
Illustrator: Antbits
Marketing Manager: Helena Mutak
Cover image photographer: Tamera Schoenholz, CRA, OCT-C
Contributors
Michael Abràmoff MD, PhD Marcos Ávila MD Alan C. Bird MD
Associate Professor Professor Emeritus Professor
Retina Service Head of the Department of Ophthalmology Department of Ophthalmic Genetics
Department of Ophthalmology and Visual Federal University of Goias Institute of Ophthalmology
Sciences Goiânia, GO, Brazil University College London
Biomedical Engineering, and Electrical and Chapter 86 London, UK
Computer Engineering Chapter 64
University of Iowa G. William Aylward FRCS, FRCOphth, MD
Iowa City, IA, USA Consultant Vitreoretinal Surgeon Barbara A. Blodi MD
Chapter 6 Moorfields Eye Hospital Professor of Ophthalmology and Visual
London, UK Sciences
Gary W. Abrams MD Chapter 105 University of Wisconsin School of Medicine
The David Barsky MD Professor and Chair and Public Health
Department of Ophthalmology Matthew Bedell MD Madison, WI, USA
Wayne State University Clinical Research Fellow Chapter 48
Director, Kresge Eye Institute Department of Ophthalmology
Detroit, MI, USA Shiley Eye Center Mark S. Blumenkranz MD
Chapter 108 University of California, San Diego Professor and Chair
La Jolla, CA, USA Department of Ophthalmology
Anita Agarwal MD Chapter 46 Stanford University
Associate Professor of Ophthalmology Palo Alto, CA, USA
Retina, Vitreous and Uveitis Rubens Belfort Jr MD, PhD, MBA Chapter 39, Chapter 67
Vanderbilt Eye Institute Head Professor of Ophthalmology
Nashville, TN, USA Department of Ophthalmology H. Culver Boldt MD
Chapter 129 Federal University of São Paulo Professor of Ophthalmology
São Paulo, Brazil Department of Ophthalmology
Everett Ai MD Chapter 85 University of Iowa
Director, Ophthalmic Diagnostic Center Iowa City, IA, USA
California Pacific Medical Center Jean Bennett MD, PhD Chapter 145
San Francisco, CA, USA F.M. Kirby Professor of Ophthalmology
Chapter 1 Professor of Cell and Developmental Biology Norbert Bornfeld MD
Department of Ophthalmology Professor
Lloyd M. Aiello MD Scheie Eye Institute Center for Ophthalmology
Director Emeritus University of Pennsylvania Perelman School University Hospital of Essen
Beetham Eye Institute of Medicine Essen, Germany
Joslin Diabetes Center Research Scientist Chapter 148
Boston, MA, USA Center for Cellular and Molecular
Chapter 48 Therapeutics Ferdinando Bottoni MD, FEBO
The Children’s Hospital of Philadelphia Staff Member
Lloyd Paul Aiello MD, PhD Philadelphia, PA, USA Eye Clinic
Professor of Ophthalmology Chapter 34 Department of Clinical Science Luigi Sacco
Harvard Medical School Sacco Hospital, University of Milan
Vice Chair, Centers of Excellence Chris Bergstrom MD, OD Milan, Italy
Harvard Department of Ophthalmology Assistant Professor of Ophthalmology Chapter 2
Medical Director of Ophthalmology Emory University Eye Center
Brigham & Women’s Hospital Emory University School of Medicine Michael E. Boulton PhD
Section Head, Eye Research Atlanta, GA, USA Professor
Vice President of Ophthalmology Chapter 133 Department of Anatomy and Cell Biology
Director, Beetham Eye Institute University of Florida
Joslin Diabetes Center Cagri G. Besirli MD, PhD Gainesville, FL, USA
Boston, MA, USA Fellow Chapter 24
Chapter 48 Retina and Uveitis
Department of Ophthalmology and Visual Sara J. Bowne PhD
Daniel M. Albert MD, MS Sciences Faculty Associate, Human Genetics Center
RRF Emmett A. Humble Distinguished University of Michigan Division of Epidemiology
Director of the UW Eye Research Institute Ann Arbor, MI, USA Human Genetics and Environmental Sciences
F.A. Davis and Lorenz E. Zimmerman Chapter 73 School of Public Health
Professor The University of Texas Health Science Center
Department of Ophthalmology and Visual Pramod S. Bhende MS at Houston
Sciences Senior Consultant Houston, TX, USA
University of Wisconsin-Madison Department of Vitreoretinal Surgery Chapter 31
Madison, WI, USA Sankara Nethralaya
Chapter 138 Nungambakkam, Chennai, India Milam A. Brantley Jr MD, PhD
Chapter 109 Assistant Professor
Mathew W. Aschbrenner MD Ophthalmology and Visual Sciences
Instructor in Ophthalmology and Visual Susanne Binder MD Vanderbilt Eye Institute
Science Professor Vanderbilt University
Department of Ophthalmology and Visual Department of Ophthalmology Nashville, TN, USA
Sciences Rudolph Foundation Hospital Chapter 22
Washington University School of Medicine (Rudolfstiftung)
St Louis, MO, USA Vienna, Austria
Chapter 119 Chapter 111
Neil M. Bressler MD Antonio Capone Jr MD, PhD Jeannie Chen PhD
The James P. Gills Professor of Clinical Professor Professor
Ophthalmology Oakland University/William Beaumont Department of Cell and Neurobiology
xii Chief of the Retina Division Hospital School of Medicine Zilkha Neurogenetic Institute
Wilmer Eye Institute Auburn Hills, MI Keck School of Medicine
Johns Hopkins University School of Medicine Professor, European School for Advanced University of Southern California
Contributors

Baltimore, MD, USA Studies in Ophthalmology Los Angeles, CA, USA


Chapter 65, Chapter 66 Lugano, Switzerland Chapter 14
Partner, Associated Retinal Consultants
Susan B. Bressler MD Royal Oak, MI Youxin Chen MD
The Julia G. Levy, PhD Professor of Director. Vision Research Foundation Professor
Ophthalmology Novi, MI, USA Department of Ophthalmology
Wilmer Eye Institute Chapter 137 Peking Union Medical College Hospital
Johns Hopkins University School of Medicine Beijing, PR China
Baltimore, MD, USA David Carruthers MBBS, PhD, FRCP Chapter 91
Chapter 65, Chapter 66 Consultant Rheumatologist
Birmingham City Hospital Carol Yim Lui Cheung PhD
Andreas Bringmann PhD Birmingham, UK Research Scientist
Associate Professor Chapter 80 Singapore Eye Research Institute
Department of Ophthalmology and Eye Singapore
Hospital Jerry D. Cavallerano OD, PhD Adjunct Assistant Professor
University of Leipzig Optometrist Office of Clinical Sciences
Leipzig, Germany Beetham Eye Institute Duke-NUS Graduate Medical School
Chapter 17 Joslin Diabetes Center Durham, NC, USA
Associate Professor Chapter 49
Daniel A. Brinton MD Department of Ophthalmology
Assistant Clinical Professor Harvard Medical School Emily Y. Chew MD
Department of Ophthalmology Boston, MA, USA Chief of Clinical Trials
University of California, San Francisco Chapter 48 Deputy Director
San Francisco, CA, USA Division of Epidemiology and Clinical
Chapter 103 Usha Chakravarthy MD, PhD Applications
Professor of Ophthalmology and Vision National Eye Institute
Gary C. Brown MD, MBA Science National Institutes of Health
Chief and Attending Surgeon The Queen’s University Bethesda, MD, USA
Retina Service, Wills Eye Institute Consultant in Ophthalmology Chapter 52, Chapter 55, Chapter 130
Professor of Ophthalmology The Belfast Health and Social Care Trust
Jefferson Medical College Belfast, UK Allen Chiang MD
Philadelphia, PA Chapter 94 Fellow in Vitreoretinal Surgery
Chief Medical Officer Retina Service
Center for Value-Based Medicine Chi-Chao Chan MD Wills Eye Institute
Flourtown, PA, USA Chief, Immunopathology Section Philadelphia, PA, USA
Chapter 59, Chapter 152 Laboratory of Immunology Chapter 103
Chief, Histology Core
Justin C. Brown MD National Eye Institute Michael F. Chiang MD
Medical and Surgical Retinal Fellow National Institutes of Health Professor of Ophthalmology
The Wilmer Eye Institute Bethesda, MD, USA Professor of Medical Informatics and Clinical
Johns Hopkins Hospital Chapter 25 Epidemiology
Baltimore, MD, USA Oregon Health & Science University
Chapter 92 Waiman Chan MBBS(HK), MRCP(UK), Portland, OR, USA
FRCS(Edin), FRCP(Edin), FRCOphth(HK), Chapter 61, Chapter 62
Simon Brunner MD FRCOphth(UK), FHKAM (Ophthalmology)
Professor of Ophthalmology Consultant Ian J. Constable FRANZCO, FRCSE, DSc
Department of Ophthalmology Department of Ophthalmology (Hon)
Rudolph Foundation Hospital Hong Kong Sanatorium & Hospital Professor of Ophthalmology
(Rudolfstiftung) Hong Kong Lions Eye Institute
Vienna, Austria Chapter 72 University of Western Australia
Chapter 111 Perth, Australia
Steven Charles MD Chapter 107
Ronald A. Bush PhD Clinical Professor of Ophthalmology
Staff Scientist University of Tennessee Gabriel Coscas MD, FEBO
Section on Translational Research on Retinal College of Medicine Professor Emeritus of Ophthalmology
and Macular Degeneration Memphis, TN, USA Department of Ophthalmology
National Institute of Deafness and Other Chapter 101 University of Paris-East Créteil Val de Marne
Communication Disorders Créteil, Paris, France
National Institutes of Health David G. Charteris MD Chapter 30
Bethesda, MD, USA Consultant Vitreoretinal Surgeon
Chapter 37 Moorfields Eye Hospital Alan F. Cruess MD, FRCSC
London, UK Professor and Head
Dingcai Cao PhD Chapter 29 District Chief, Capital Health
Associate Professor Department of Ophthalmology and Visual
Department of Ophthalmology and Visual Dong Feng Chen MD, PhD Sciences
Sciences Associate Professor Dalhousie University
University of Illinois at Chicago Schepens Eye Research Institute Halifax, Nova Scotia, Canada
Chicago, IL, USA Department of Ophthalmology Chapter 131
Chapter 10 Harvard Medical School
Boston, MA, USA
Chapter 33
Emmett T. Cunningham Jr MD, PhD, MPH Alastair K. Denniston PhD, MRCP, Amani A. Fawzi MD
Director, The Uveitis Service FRCOphth Associate Professor of Ophthalmology
California Pacific Medical Center Clinical Lecturer Northwestern University, Feinberg School of
San Francisco, CA Academic Unit of Ophthalmology Medicine xiii
Adjunct Clinical Professor of Ophthalmology University of Birmingham Chicago, IL, USA
Stanford University School of Medicine Birmingham, UK Chapter 44

Contributors
Stanford, CA Chapter 80
West Coast Retina Medical Group Sharon Fekrat MD, FACS
San Francisco, CA, USA Ranjit S. Dhaliwal MD, FRCSC, FACS Associate Professor
Chapter 1 Ophthalmologist Vitreoretinal Surgery
Adjunct Faculty Department of Ophthalmology
Christine A. Curcio PhD Department of Ophthalmology Duke Eye Center
Professor of Ophthalmology Queen’s University School of Medicine Chief of Ophthalmology
University of Alabama School of Medicine Kingston, Ontario, Canada Durham Veterans Affairs Medical Center
Birmingham, AL, USA Consultant Retinal Specialist and Founder Durham, NC, USA
Chapter 20 Retina Consultants, PC (The Retina Eye Chapter 54
Center)
Stephen P. Daiger PhD Augusta, GA, USA Steven E. Feldon MD
TS Matney Professor of Environmental and Chapter 134, Chapter 155 Director, Flaum Eye Institute
Genetic Sciences Professor and Chair
Human Genetics Center Xiaoyan Ding MD, PhD Department of Ophthalmology
Division of Epidemiology Associate Professor University of Rochester
Human Genetics and Environmental Sciences Department of Retina Rochester, NY, USA
School of Public Health Zhongshan Ophthalmic Center Chapter 12
The University of Texas Health Science Center Sun Yat-sen University
at Houston Guangzhou, PR China Rodrigo A. Brant Fernandes MD
Houston, TX, USA Chapter 41 Research Associate
Chapter 31 Doheny Eye Institute
Diana V. Do MD Keck School of Medicine
Bertil E. Damato MD, PhD, FRCOphth Associate Professor of Ophthalmology University of Southern California
Honorary Professor The Wilmer Eye Institute Los Angeles, CA, USA
Ocular Oncology Service Johns Hopkins University School of Medicine Chapter 126
Royal Liverpool University Hospital Baltimore, MD, USA
Liverpool, UK Chapter 124, Chapter 155 Henry A. Ferreyra MD
Chapter 147 Assistant Clinical Professor
Guorui Dou MD, PhD Department of Ophthalmology
Janet L. Davis MD, MA Professor Shiley Eye Center
Professor of Ophthalmology Department of Ophthalmology University of California, San Diego
University of Miami Miller School of Xijing Hospital La Jolla, CA, USA
Medicine Fourth Military Medical University Chapter 46
Miami, FL, USA Xi’an, PR China
Chapter 87 Chapter 16 Deborah A. Ferrington PhD
Associate Professor
Matthew D. Davis MD William A. Dunn Jr PhD Department of Ophthalmology
Emeritus Professor of Ophthalmology and Professor and Interim Chairman University of Minnesota
Visual Sciences Department of Anatomy and Cell Biology Minneapolis, MN, USA
University of Wisconsin School of Medicine University of Florida College of Medicine Chapter 32
and Public Health Gainesville, FL, USA
Madison, WI, USA Chapter 24 Frederick L. Ferris III MD
Chapter 48 Director
Justis P. Ehlers MD Division of Epidemiology and Clinical
Shelley Day MD Staff Physician Applications
Clinical Assistant Professor of Ophthalmology Retina Service Clinical Director,
Stanford University School of Medicine Cole Eye Institute National Eye Institute
Palo Alto, CA, USA Cleveland Clinic Foundation National Institutes of Health
Chapter 153 Cleveland, OH, USA Bethesda, MD, USA
Chapter 70, Chapter 120 Chapter 47
Patrick De Potter MD, PhD
Professor of Ophthalmology Michael Engelbert MD, PhD Paul T. Finger MD
Chairman Attending Physician and Research Assistant Clinical Professor of Ophthalmology
Ophthalmology Department Professor New York University School of Medicine
Cliniques Universitaires St-Luc Department of Ophthalmology Director
Catholic University of Leuven New York University School of Medicine The New York Eye Cancer Center
Brussels, Belgium New York, NY, USA New York, NY, USA
Chapter 58 Chapter 55 Chapter 149

Marc D. de Smet MDCM, PhD, FRCSC, Lisa J. Faia MD Steven K. Fisher PhD
FRCOpth Vitreoretinal Surgery Fellow Research Professor
Professor of Ophthalmology Associated Retinal Consultants Neuroscience Research Institute
University of Amsterdam William Beaumont Hospital Professor Emeritus
Amsterdam, The Netherlands Royal Oak, MI, USA Molecular, Cellular and Developmental
Director of the Retina and Inflammation Unit Chapter 114 Biology
Center for Specialized Ophthalmology University of California, Santa Barbara
Clinique de Montchoisi Benedetto Falsini MD Santa Barbara, CA, USA
GSMN Medical Networks Associate Professor of Ophthalmology Chapter 29
Lausanne, Switzerland Department of Ophthalmology
Chapter 112 Università Cattolica del S. Cuore,
Rome, Italy
Chapter 37
Gerald A. Fishman MD Arthur D. Fu MD Dan S. Gombos MD
Director Clinical Professor of Ophthalmology Associate Professor
Pangere Center for Inherited Retinal Diseases California Pacific Medical Center Chief, Section of Ophthalmology
xiv The Chicago Lighthouse for People Who Are San Francisco, CA, USA Department of Head and Neck Surgery
Blind or Visually Impaired Chapter 1 The University of Texas
Professor Emeritus of Ophthalmology MD Anderson Cancer Center
Contributors

Department of Ophthalmology and Visual Carlos Alexandre de Amorim Garcia Filho Houston, TX, USA
Sciences MD Chapter 128
UIC Eye Center Post Doctoral Associate
Chicago, IL, USA Department Of Ophthalmology Lingam Gopal MS, FRCS
Chapter 43 Bascom Palmer Eye Institute Consultant
University of Miami Miller School of Vitreo-retinal Services
Monika Fleckenstein MD Medicine Sankara Nethralaya
Ophthalmology Specialist Miami, FL, USA Medical and Vision Research foundations
Department of Ophthalmology Chapter 3 Chennai, India
University of Bonn Chapter 109
Bonn, Germany Enrique Garcia-Valenzuela MD, PhD
Chapter 4 Clinical Assistant Professor Caroline Gordon MD, FRCP
Department of Ophthalmology Professor of Rheumatology
Harry W. Flynn Jr MD University of Illinois Eye and Ear Infirmary School of Immunity and Infection
Associate Professor of Ophthalmology Consulting Staff, Vitreo-Retinal Surgery College of Medical and Dental Sciences
The J. Donald M. Gass Distinguished Chair in Midwest Retina Consultants, SC The Medical School
Ophthalmology Parkside Center University of Birmingham
Bascom Palmer Eye Institute Park Ridge, IL, USA Birmingham, UK
Miami, FL, USA Chapter 108 Chapter 80
Chapter 87
Alain Gaudric MD Hiroshi Goto MD
Andrew C. Fok MBChB, FRCS Professor of Ophthalmology Professor and Chairman
Resident Specialist Hôpital Lariboisière Department of Ophthalmology
Department of Ophthalmology Paris Diderot University Tokyo Medical University
Hong Kong Eye Hospital Paris, France Tokyo, Japan.
Kowloon, Hong Kong Chapter 117 Chapter 75
Chapter 72
Mary Gayed MBChB MRCP(UK) Evangelos S. Gragoudas MD
Wallace S. Foulds CBE, MD, ChM, DSc, Academic Clinical Fellow Charles Edward Whitten Professor of
FRCS, FRCOphth Rheumatology Ophthalmology
Emeritus Professor of Ophthalmology Division of Immunity and Infection Harvard Medical School
University of Glasgow Birmingham Medical School Director of Retina Service
Glasgow, Scotland Birmingham, UK Massachusetts Eye & Ear Infirmary
Senior Consultant Chapter 80 Boston, MA, USA
Singapore Eye Research Institute Chapter 146
Singapore Mohamed A. Genead MD
Chapter 147 Ophthalmologist and Retinal Specialist Maria B. Grant MD
Pangere Center for Hereditary Retinal Professor
William R. Freeman MD Diseases Department of Pharmacology and
Professor of Ophthalmology The Chicago Lighthouse for People Who Are Therapeutics
Director of the Jacobs Retina Center Blind or Visually Impaired University of Florida, Gainesville
Department of Ophthalmology, Shiley Eye Chicago, IL, USA Director of Translational Research
Center Chapter 43 Department of Ophthalmology
University of California, San Diego Gainesville, FL, USA
La Jolla, CA, USA Heinrich Gerding MD Chapter 18
Chapter 81 Professor of Ophthalmology
Department of Ophthalmology W. Richard Green MD (posthumously)
Aurélien Freton MD Pallas Clinic Former Independent Order of Odd Fellows
Clinical Assistant Professor Olten, Switzerland Professor of Ophthalmology
Department of Ophthalmology Chapter 136 Professor of Pathology Eye
Saint Roch Hospital Pathology Laboratory
Nice, France Andrea Giani MD Johns Hopkins University School of Medicine
Chapter 149 Ophthalmologist, Vitreo-Retinal Service Baltimore, MD, USA
Eye Clinic Chapter 21
Martin Friedlander MD, PhD Department of Clinical Science Luigi Sacco
Professor Sacco Hospital Ronald G. Gregg PhD
Department of Cell Biology University of Milan Professor of Biochemistry and Molecular
The Scripps Research Institute Milan, Italy Biology
Division of Ophthalmology Chapter 2 Professor of Ophthalmology and Visual
Department of Surgery Sciences
Scripps Clinic Morton F. Goldberg MD Director, Center for Genetics and Molecular
La Jolla, CA, USA Joseph E. Green Professor of Ophthalmology Medicine
Chapter 35 Director Emeritus University of Louisville
Wilmer Ophthalmological Institute Louisville, KY, USA
Laura J. Frishman PhD Department of Ophthalmology Chapter 15
Moores Professor, Vision Sciences Johns Hopkins University School of Medicine
College of Optometry Baltimore, MD, USA Zdenek Gregor FRCS (Eng), FRCOphth
University of Houston Chapter 57 Consultant Vitreoretinal Surgeon
Houston, TX, USA Moorfields Eye Hospital
Chapter 7 London, UK
Chapter 116
Giovanni Gregori PhD Paul Hahn MD, PhD David R. Hinton MD
Assistant Research Professor Fellow Gavin S. Herbert Professor of Retinal
Department of Ophthalmology Vitreoretinal Surgery Research
University of Miami Department of Ophthalmology Professor of Pathology and Ophthalmology xv
Miller School of Medicine Duke Eye Center Keck School of Medicine
Miami, FL, USA Durham, NC, USA University of Southern California

Contributors
Chapter 3 Chapter 54 Los Angeles, CA, USA
Chapter 16, Chapter 35
Kevin Gregory-Evans MD, PhD, FRCS, Julia A. Haller MD
FRCOphth Ophthalmologist-in-Chief Frank G. Holz MD, FEBO
Julia Levy BC Leadership Chair in Macular Professor and Chair Professor and Chairman
Research Department of Ophthalmology Department of Ophthalmology
Professor in Ophthalmology Jefferson Medical College University of Bonn
Department of Ophthalmology and Visual William Tasman, MD Endowed Chair in Bonn, Germany
Sciences Ophthalmology Chapter 4
University of British Columbia Eye Care Wills Eye Institute
Centre Philadelphia, PA, USA Samuel K. Houston MD
Vancouver, BC, Canada Chapter 56 Resident Physician
Chapter 40 Department of Ophthalmology
J. William Harbour MD Bascom Palmer Eye Institute
Seanna Grob MD, MAS Director, Center for Ocular Oncology University of Miami Miller School of
Clinical Research Fellow Assistant Professor of Ophthalmology Medicine
Department of Ophthalmology Washington University School of Medicine Miami, FL, USA
Shiley Eye Center St Louis, MO, USA Chapter 145
University of California, San Diego Chapter 128
La Jolla, CA, USA Yan-Nian Hui MD
Chapter 46 Christos Haritoglou MD Professor
Professor Department of Ophthalmology
Carl Groenewald MD Department of Ophthalmology Xijing Hospital
Consultant Ophthalmologist Ludwig Maximilians University Fourth Military Medical University
St Paul’s Eye Unit Munich, Germany Xi’an, PR China
Royal Liverpool University Hospital Chapter 127 Chapter 97
Liverpool, UK
Chapter 147 Mary E. Hartnett MD, FACS Mark S. Humayun MD, PhD
Associate Professor of Ophthalmology Professor of Ophthalmology
Hans E. Grossniklaus MD, MBA Department of Ophthalmology Biomedical Engineering, Cell and
F. Phinizy Calhoun Jr Professor of University of North Carolina Neurobiology
Ophthalmology Chapel Hill, NC, USA Keck School of Medicine
Director, L.F. Montgomery Pathology Chapter 95 University of Southern California
Laboratory Associate Director of Research
Director, Ocular Oncology and Pathology Barbara S. Hawkins PhD Doheny Eye Institute
Service Professor Emeritus Los Angeles, CA, USA
Department of Ophthalmology Professor of Ophthalmology, School of Chapter 36, Chapter 38, Chapter 126
Emory University School of Medicine Medicine
Atlanta, GA, USA Professor of Epidemiology, Bloomberg School Yasushi Ikuno MD
Chapter 142 of Public Health Associate Professor
Johns Hopkins University Department of Ophthalmology
Sandeep Grover MD Baltimore, MD, USA Osaka University Graduate School of
Assistant Professor of Ophthalmology Chapter 70, Chapter 94, Chapter 150 Medicine
Director, Inherited Retinal Diseases & Osaka, Japan
Electrophysiology Shikun He MD Chapter 68, Chapter 113
University of Florida College of Medicine Associate Professor
Jacksonville, FL, USA Department of Ophthalmology David Isaac MD
Chapter 43 Doheny Eye Institute Assistant Professor
University of Southern California Department of Ophthalmology
Vamsi K. Gullapalli MD, PhD Los Angeles, CA, USA Federal University of Goias
Associate Chapter 33 Goiânia, GO, Brazil
Coastal Bend Retina Chapter 86
Corpus Christi, TX, USA Martina C. Herwig MD
Chapter 125 Professor, Department of Ophthalmology Tatsuro Ishibashi MD, PhD
Emory University School of Medicine Professor and Chairman
Aditi Gupta MS, DNB Atlanta, GA, USA Department of Ophthalmology
Clinical Fellow Professor, Department of Ophthalmology Graduate School of Medical Sciences
Department of Vitreoretinal Services University of Bonn Kyushu University
Sankara Nethralaya Bonn, Germany Fukuoka City, Japan
Chennai, India Chapter 142 Chapter 68
Chapter 50
Florian M.A. Heussen MD Douglas A. Jabs MD, MBA
Rudolf F. Guthoff MD Resident Physician Professor and Chair
Head Department of Ophthalmology Department of Ophthalmology
University Eye Department Charité – University Medicine Berlin Mount Sinai School of Medicine
University of Rostock Berlin, Germany New York, NY, USA
Rostock, Germany Chapter 60 Chapter 78
Chapter 9
Glenn J. Jaffe MD
Professor of Ophthalmology
Chief, Vitreoretinal Service
Director, Duke Reading Center
Durham, NC, USA
Chapter 79
Lee M. Jampol MD Christine N. Kay MD Lazaros Konstantinidis MD
Louis Feinberg Professor in Ophthalmology Assistant Professor Professor
Northwestern University Department of Ophthalmology Vitreoretinal Department
xvi Chicago, IL, USA University of Florida St Paul’s Eye Unit
Chapter 76 Gainesville, FL, USA Royal Liverpool University Hospital
Chapter 6 Liverpool, UK
Contributors

Leonard Joffe MD, FCS(SA), FRCSEd Chapter 118


Clinical Professor Pearse A. Keane MD, MRCOphth, MRCSI
Department of Ophthalmology Clinical Lecturer in Ophthalmic Translational Igor Kozak MD, PhD
University of Arizona Research Assistant Clinical Professor
Tucson, AZ, USA NIHR Biomedical Research Centre for Department of Ophthalmology
Chapter 135 Ophthalmology Shiley Eye Center
Moorfields Eye Hospital NHS Foundation University of California, San Diego
Mark Johnson PhD Trust La Jolla, CA, USA
Professor UCL Institute of Ophthalmology Chapter 81
Departments of Biomedical Engineering, London, UK
Mechanical Engineering, and Ophthalmology Chapter 5 Baruch D. Kuppermann MD
Northwestern University Professor of Ophthalmology and Biomedical
Evanston, IL, USA M. Cristina Kenney MD, PhD Engineering
Chapter 20 Professor University of California, Irvine
Director of Research The Gavin Herbert Eye Institute
Mark W. Johnson MD Gavin Herbert Eye Institute University of California, Irvine
Professor and Director of Retina Service Ophthalmology Research Laboratories Irvine, CA, USA
Department of Ophthalmology and Visual University of California, Irvine Chapter 38
Sciences Irvine, CA, USA
Kellogg Eye Center Chapter 32 Leanne T. Labriola DO
University of Michigan Clinical Assistant Professor of Ophthalmology
Ann Arbor. MI, USA Khizer R. Khaderi MD Medical Retina Service
Chapter 73 Neuro-Ophthalmology Fellow University of Pittsburgh School of Medicine
Doheny Eye Institute Pittsburgh, PA, USA
Robert N. Johnson MD Keck School of Medicine Chapter 9
Clinical Professor of Ophthalmology University of Southern California
California Pacific Medical Center Los Angeles, CA, USA Timothy Y. Lai MD, FRCS, FRCOphth
San Francisco, CA, USA Chapter 93 Honorary Clinical Associate Professor
Chapter 1 Department of Ophthalmology and Visual
Mohamad A. Khodair MD, PhD Sciences
Antonia M. Joussen MD, PhD, FEBO Lecturer The Chinese University of Hong Kong
Professor Robertwood Johnson Medical School Hong Kong
Department of Ophthalmology New Brunswick, NJ, USA Chapter 72
Charité, University Medicine Berlin Chapter 125
Berlin, Germany Dennis S. Lam MD, FRCOphth
Chapter 28 Ivana K. Kim MD S.H. Ho Professor and Chairman
Assistant Professor, Department of Department of Ophthalmology and Visual
Karina Julian MD Ophthalmology Sciences
Senior Instructor Harvard Medical School, Retina Service The Chinese University of Hong Kong
Ophthalmology Massachusetts Eye and Ear Infirmary Hong Kong
Austral University School of Medicine Boston, MA, USA Chapter 72
Buenos Aires, Argentina Chapter 146
Chapter 112 Linda A. Lam MD
Tae Wan Kim MD, PhD Assistant Professor of Ophthalmology
J. Michael Jumper MD Assistant Professor Doheny Eye Institute
Retina Service Chief Department of Ophthalmology Keck School of Medicine
California Pacific Medical Center Seoul National University University of Southern California
San Francisco, CA, USA Seoul, South Korea Los Angeles, CA, USA
Chapter 1 Chapter 123 Chapter 69

Peter K. Kaiser MD Bernd Kirchhof MD, PhD Maurice B. Landers III MD


Professor of Ophthalmology Professor and Chairman Professor of Ophthalmology
Cleveland Clinic Lerner College of Medicine Department of Ophthalmology, University of Department of Ophthalmology
Cleveland, OH, USA Cologne University of North Carolina
Chapter 53 Cologne, Germany Chapel Hill, NC, USA
Chapter 95, Chapter 121 Chapter 95
Anselm Kampik MD
Chairman Barbara E.K. Klein MD, MPH Anne Marie Lane MPH
Department of Ophthalmology Professor Instructor in Ophthalmology
Ludwig Maximilians University Department of Ophthalmology and Visual Harvard Medical School
Munich, Germany Sciences Research Associate
Chapter 127 University of Wisconsin, School of Medicine Retina Service
and Public Health Massachusetts Eye & Ear Infirmary
Robert Katamay MD Madison, WI, USA Boston, MA, USA
Research Fellow Chapter 45 Chapter 146
Laboratory of Immunology
National Eye Institute Ronald Klein MD MPH Erin B. Lavik ScD
National Institutes of Health Professor Elmer Lindseth Associate Professor of
Bethesda, MD, USA Department of Ophthalmology and Visual Biomedical Engineering
Chapter 27 Sciences Case Western Reserve University
University of Wisconsin School of Medicine Cleveland, OH, USA
and Public Health Chapter 38
Madison, WI, USA
Chapter 45
James F. Leary PhD Phoebe Lin MD, PhD Nancy C. Mansfield (posthumously)
SVM Professor of Nanomedicine Vitreoretinal Surgical Fellow Family Counselor and Patient Ombudsman
Professor of Basic Medical Sciences and Department of Ophthalmology The Retinoblastoma Centre
Biomedical Engineering Duke University Children’s Hospital of Los Angeles xvii
Department of Basic Medical Sciences Durham, NC, USA Los Angeles, CA, USA
Weldon School of Biomedical Engineering Chapter 79 Chapter 128

Contributors
Purdue University
West Lafayette, IN, USA David T. Liu FRCOphth, FRCS, MSc Arnold M. Markoe MD
Chapter 36 Associate Professor Professor and Chairman Emeritus
Department of Ophthalmology and Visual Department of Radiation Oncology
Sun Young Lee MD, PhD Sciences University of Miami, Miller School of
Clinical Instructor The Chinese University of Hong Kong Medicine
Doheny Eye Institute Hong Kong Miami, FL, USA
Keck School of Medicine Chapter 72 Chapter 145
University of Southern California
Los Angeles, CA, USA Nikolas J.S. London MD Michael F. Marmor MD
Chapter 98 Fellow in Vitreoretinal Surgery Professor of Ophthalmology
Wills Eye Institute Stanford University School of Medicine
Thomas C. Lee MD Philadelphia, PA, USA Byers Eye Institute at Stanford
Director, Retina Institute Chapter 56 Palo Alto, CA, USA
The Vision Center Chapter 19
Children’s Hospital Los Angeles Brandon J. Lujan MD
Associate Professor of Ophthalmology Associate Daniel F. Martin MD
Director, Surgical Retina Fellowship West Coast Retina Barbara & A. Malachi Mixon III Institute
Doheny Eye Institute San Francisco, CA, USA Chair of Ophthalmology
Keck School of Medicine Chapter 1 Chairman
University of Southern California Cole Eye Institute Cleveland Clinic
Los Angeles, CA, USA Yan Luo MD, PhD Cleveland, OH, USA
Chapter 61, Chapter 128 Professor Chapter 67
State Key Laboratory of Ophthalmology
Loh-Shan B. Leung MD Zhongshan Ophthalmic Center Stephen C. Massey PhD
Postdoctoral Fellow Sun Yat-sen University Elizabeth Morford Chair and Research
Department of Ophthalmology Guangzhou, PR China Director
Stanford University Chapter 41 Ophthalmology and Visual Sciences
Palo Alto, CA, USA The University of Texas Medical School at
Chapter 67 Gerard A. Lutty PhD Houston
G. Edward and G. Britton Durell Professor of Houston, TX, USA
David A. Lewis MD Ophthalmology Chapter 15
Ocular Pathology Fellow Wilmer Ophthalmological Institute
Department of Ophthalmology Johns Hopkins Hospital Maureen A. McCall PhD
University of Wisconsin Baltimore, MD, USA Professor
Madison, WI, USA Chapter 18, Chapter 57 Department of Ophthalmology and Visual
Chapter 138 Sciences
Robert MacLaren FRCOph, MD, PhD University of Louisville
Geoffrey P. Lewis PhD Nuffield Laboratory of Ophthalmology Louisville, KY, USA
Research Biologist University of Oxford Chapter 15
Neuroscience Research Institute John Radcliffe Hospital
University of California, Santa Barbara Oxford, UK Tara A. McCannel MD, PhD
Santa Barbara, CA, USA Chapter 121 Assistant Professor
Chapter 29 Director of Ophthalmic Oncology Center
Steven Madreperla MD Department of Ophthalmology
Anita Leys MD, PhD Associate The Jules Stein Eye Institute
Professor of Ophthalmology Retina Associates of New Jersey University of California, Los Angeles
Department of Medical Retina Teaneck, NJ, USA Los Angeles, CA, USA
Department of Ophthalmology Chapter 125 Chapter 139, Chapter 140
Catholic University of Leuven
Leuven, Belgium Albert M. Maguire MD J. Allen McCutchan MD, MSc
Chapter 58 Associate Professor of Ophthalmology Professor of Medicine
Department of Ophthalmology Department of Medicine
Xiaoxin Li MD Scheie Eye Institute Division of Infectious Diseases
Professor of Ophthalmology University of Pennsylvania Perelman School University of California, San Diego
People’s Eye Centre of People’s Hospital of Medicine La Jolla, CA, USA
Peking University Investigator, Center for Cellular and Chapter 81
Beijing, PR China Molecular Therapeutics
Chapter 71 Retina Specialist, Pediatric Ophthalmology H. Richard McDonald MD
The Children’s Hospital of Philadelphia Clinical Professor of Ophthalmology
Sandra Liakopoulos MD Philadelphia, PA, USA California Pacific Medical Center
Professor Chapter 34 Director, Vitreoretinal Fellowship Program
Department of Ophthalmology California Pacific Medical Center
University of Cologne Martin A. Mainster PhD, MD, FRCOphth Director, San Francisco Retina Foundation
Cologne, Germany Luther and Ardis Fry Professor Emeritus of San Francisco, CA, USA
Chapter 60 Ophthalmology Chapter 1
University of Kansas School of Medicine
Chang-Ping Lin MD, PhD Kansas City, KS, USA Milap P. Mehta MD, MS
Professor Chapter 90 Oculoplastics Fellow
Department of Ophthalmology Department of Ophthalmology
Changhua Christian Hospital Cole Eye Institute
Kaohsiung, Changhua, Taiwan Cleveland Clinic
Chapter 96 Cleveland, OH, USA
Chapter 144
Petra Meier MD Carlo Montemagno PhD Timothy G. Murray MD, MBA
Associate Professor Dean Professor
Department of Ophthalmology College of Engineering and Applied Science Department of Ophthalmology
xviii University of Leipzig Geier Professor of Engineering Education Bascom Palmer Eye Institute
Leipzig, Germany University of Cincinnati Miami, FL, USA
Chapter 115 College of Engineering Chapter 145
Contributors

Cincinnati, OH, USA


Shannath Merbs MD, PhD Chapter 36 Manish Nagpal, MS, DO, FRCS(UK)
Associate Professor of Ophthalmology and Vitreoretinal Consultant
Oncology Ala Moshiri MD, PhD Retina Foundation
Wilmer Eye Institute Retina Fellow Shahibag, Ahmedabad
Johns Hopkins University School of Medicine Department of Ophthalmology Gujarat, India
Baltimore, MD, USA Wilmer Eye Institute Chapter 107
Chapter 141 Johns Hopkins University School of Medicine
Baltimore, MD, USA Perumalsamy Namperumalsamy MS, FAMS
Travis A. Meredith MD Chapter 92 Professor of Ophthalmology
Sterling A. Barrett Distinguished Professor Retina-Vitreous Service
Chairman Prithvi Mruthyunjaya MD Aravind Eye Hospital and Postgraduate
Department of Ophthalmology Assistant Professor of Ophthalmology Institute of Ophthalmology
University of North Carolina Department of Vitreoretinal Surgery and Madurai, India
Chapel Hill, NC, USA Ocular Oncology Chapter 82, Chapter 83
Chapter 122 Duke University Eye Center
Durham, NC, USA Sumit K. Nanda MD
Carsten H. Meyer MD Chapter 54, Chapter 153 Clinical Associate Professor
Professor of Ophthalmology Department of Ophthalmology
Department of Ophthalmology Cristina Muccioli MD, PhD University of Oklahoma
Pallas Clinic Associate Professor of Ophthalmology Oklahoma City, OK, USA
Olten, Switzerland Department of Ophthalmology Chapter 108
Chapter 136 Federal University of São Paulo
São Paulo, Brazil Quan Dong Nguyen MD, MSc
William F. Mieler MD Chapter 85 Associate Professor of Ophthalmology
Professor and Vice-Chairman Wilmer Eye Institute
Director, Residency Training Robert F. Mullins PhD Johns Hopkins University School of Medicine
Department of Ophthalmology and Visual Associate Professor Baltimore, MD, USA
Sciences Department of Ophthalmology and Visual Chapter 78, Chapter 124
University of Illinois at Chicago Sciences
Chicago, IL, USA The University of Iowa Robert B. Nussenblatt MD, MPH
Chapter 89 Iowa City, IA, USA Chief
Chapter 42 Laboratory of Immunology
Joan W. Miller MD National Eye Institute
Chair & Henry Willard Williams Professor of Toshinori Murata MD, PhD Associate Director (Clinical Director)
Ophthalmology Professor and Chairman NIH Center for Human Immunology
Department of Ophthalmology Department of Ophthalmology Acting Scientific Director
Harvard Medical School School of Medicine National Center for Complementary and
Chief of Ophthalmology Shinshu University Alternative Medicine
Massachusetts Eye and Ear Infirmary Matsumoto Nagano, Japan National Institutes of Health
Massachusetts General Hospital Chapter 68 Bethesda, MD, USA
Boston, MA, USA Chapter 27, Chapter 77
Chapter 26 A. Linn Murphree MD
Professor of Ophthalmology and Pediatrics Kean T. Oh MD
Rukhsana G. Mirza MD Keck School of Medicine Assistant Professor of Ophthalmology
Assistant Professor in Ophthalmology University of Southern California Department of Ophthalmology
Northwestern University Los Angeles, CA, USA University of North Carolina
Chicago, IL USA Chapter 128 Chapel Hill, NC, USA
Chapter 76 Chapter 95
Robert P. Murphy MD
Sayak K. Mitter MTech Private Practitioner Masahito Ohji MD
Graduate Student Retina Group of Washington Professor and Chairman
Department of Anatomy and Cell Biology Fairfax, VA, USA Department of Ophthalmology
University of Florida Chapter 52 Shiga University of Medical Science
Gainesville, FL, USA Otsu, Japan
Chapter 24 Philip I. Murray PhD, FRCP, FRCS, Chapter 113
FRCOphth
Robert A. Mittra MD Professor of Ophthalmology Kyoko Ohno-Matsui MD
Vitreoretinal Surgeon Academic Unit of Ophthalmology Associate Professor
Vitreoretinal Surgery School of Immunity and Infection Department of Ophthalmology and Visual
Minneapolis, MN, USA College of Medical and Dental Sciences Sciences
Chapter 89 University of Birmingham Tokyo Medical and Dental University
Birmingham and Midland Eye Centre Tokyo, Japan
Yozo Miyake MD, PhD Sandwell and West Birmingham Hospitals Chapter 68
Chairman of the Board of Directors NHS Trust
Aichi Medical University City Hospital Daniel Palanker PhD
Aichi, Japan Birmingham, UK Associate Professor
Chapter 8 Chapter 80 Department of Ophthalmology and Hansen
Experimental Physics Laboratory
Stanford University
Stanford, CA, USA
Chapter 39
Purnima S. Patel MD Rajeev S. Ramchandran MD Robert Ritch MD
Assistant Professor of Ophthalmology Assistant Professor Shelley and Steven Einhorn Distinguished
Emory University School of Medicine Retinal Services Chair
Atlanta Veterans Affairs Medical Center Flaum Eye Institute Surgeon Director and Chief xix
Atlanta, GA, USA University of Rochester Medical Center Glaucoma Services
Chapter 51 Rochester, NY, USA The New York Eye and Ear Infirmary

Contributors
Chapter 12 New York, NY
Anna C. Pavlick MD Professor of Ophthalmology
Associate Professor Haripriya Vittal Rao PhD New York Medical College
New York University Comprehensive Cancer Chennai, India Valhalla, NY, USA
Center Chapter 24 Chapter 36
New York University School of Medicine
New York, NY, USA Narsing A. Rao MD, PhD Philip J. Rosenfeld MD, PhD
Chapter 149 Preceptor of Ophthalmology and Pathology Professor of Ophthalmology
Universidade Federal de Minas Gerais Bascom Palmer Eye Institute
David M. Peereboom MD Belo Horizonte, MG, Brazil Miami, FL, USA
Associate Professor of Medicine Chapter 74, Chapter 75, Chapter 84 Chapter 3, Chapter 67
Cleveland Clinic Lerner College of Medicine
The Rose Ella Burkhardt Brain Tumor and P. Kumar Rao MD Gary S. Rubin PhD
Neuro-Oncology Center Associate Professor of Ophthalmology and Helen Keller Professor of Ophthalmology
Solid Tumor Oncology Visual Sciences UCL Institute of Ophthalmology
Cleveland, OH, USA Department of Ophthalmology London, UK
Chapter 156 School of Medicine Chapter 11
Washington University in St. Louis
Mark E. Pennesi MD, PhD St. Louis, MO, USA Humberto Ruiz-Garcia MD
Assistant Professor of Ophthalmology Chapter 75 Retina Specialist
Casey Eye Institute Clinica Santa Lucia
Oregon Health and Science University Sivakumar R. Rathinam MNAMS, PhD Universidad de Guadalajara
Portland, OR, USA Professor of Ophthalmology Guadalajara, Mexico
Chapter 40 Head of Uveitis Service Chapter 5
Uveitis Service
Jay S. Pepose MD, PhD Aravind Eye Hospital & Postgraduate Stephen J. Ryan MD
Professor of Clinical Ophthalmology and Institute of Ophthalmology President, Doheny Eye Institute
Visual Sciences Madurai, India Professor, University of Southern California
Washington University School of Medicine Chapter 82 Los Angeles CA, USA
St Louis, MO, USA Chapter 98
Chapter 88 Franco M. Recchia MD
Associate Professor of Ophthalmology and SriniVas R. Sadda MD
Julian D. Perry MD Visual Sciences Associate Professor of Ophthalmology
Director Chief, Retina Division Doheny Eye Institute
Orbital and Oculoplastic Surgery Director, Fellowship in Vitreoretinal Diseases Associate Professor
Cole Eye Institute and Surgery Department of Ophthalmology
Cleveland Clinic Foundation, Vanderbilt Eye Institute Keck School of Medicine
Cleveland, OH, USA Vanderbilt University School of Medicine University of Southern California
Chapter 144 Nashville, TN, USA Los Angeles, CA, USA
Chapter 110 Chapter 5, Chapter 51, Chapter 60
Carmen A. Puliafito MD, MBA
Dean Kristin J. Redmond MD Alfredo A. Sadun MD, PhD
Keck School of Medicine Instructor Professor of Ophthalmology
May S and John Hooval Dean’s Chair in Department of Radiation Oncology and Doheny Eye Institute
Medicine Molecular Radiation Sciences Keck School of Medicine
Professor of Ophthalmology and Health Johns Hopkins University School of Medicine University of Southern California
Management Baltimore, MD, USA Los Angeles, CA, USA
Doheny Eye Institute Chapter 151 Chapter 93
University of Southern California
Los Angeles, CA Thomas A. Reh PhD Taiji Sakamoto MD, PhD
Voluntary Professor of Ophthalmology Professor Professor and Chair
Bascom Palmer Eye Institute Department of Biological Structure Department of Ophthalmology
University of Miami Institute for Stem Cells and Regenerative Kagoshima University Graduate School of
Miami, FL, USA Medicine Medical and Dental Sciences
Chapter 3 University of Washington Kagoshima, Japan
Seattle, WA, USA Chapter 68
Polly A. Quiram MD, PhD Chapter 13
Pediatric Retina Specialist Alapakkam P. Sampath PhD
Vitreoretinal Surgery Andreas Reichenbach MD Associate Professor
Minneapolis, MN, USA Full Professor Zilkha Neurogenetic Institute
Chapter 137 Department of Pathophysiology of Neuroglia Department of Physiology and Biophysics
Paul Flechsig Institute of Brain Research University of Southern California School of
Rajiv Raman MS, DNB University of Leipzig Medicine
Consultant Leipzig, Germany Los Angeles, CA, USA
Department of Vitreo-Retinal Services Chapter 17 Chapter 14
Sankara Nethralaya
Chennai, India Andrew P. Schachat MD
Chapter 50 Vice Chairman
Cole Eye Institute
Cleveland Clinic Foundation
Cleveland, OH, USA
Chapter 70, Chapter 130, Chapter 134, Chapter 150,
Chapter 151, Chapter 155, Chapter 156
Steffen Schmitz-Valckenberg MD, FEBO Shwu-Jiuan Sheu MD Sylvia B. Smith PhD, FARVO
Professor Chair Professor of Cellular Biology/Anatomy,
Department of Ophthalmology Department of Ophthalmology Ophthalmology and Graduate Studies
xx University of Bonn Kaohsiung Veterans General Hospital Basic Science Co-Director, Vision Discovery
Bonn, Germany Professor Institute
Chapter 4 National Yang-Ming University Medical College of Georgia
Contributors

Taipei, Taiwan Augusta, GA, USA


Stephen G. Schwartz MD, MBA Chapter 102 Chapter 23
Associate Professor of Clinical
Ophthalmology Carol L. Shields MD Wendy M. Smith MD
Department of Ophthalmology Professor of Ophthalmology Clinical Fellow
Bascom Palmer Eye Institute Thomas Jefferson University Hospital Uveitis and Ocular Immunology
University of Miami Miller School of Co-Director, Oncology Service National Eye Institute
Medicine Wills Eye Institute National Institutes of Health
Medical Director Philadelphia, PA, USA Bethesda, MD, USA
Bascom Palmer Eye Institute at Naples Chapter 56, Chapter 132, Chapter 135, Chapter 143, Chapter 25
Naples, FL, USA Chapter 152
Chapter 87 Lucia Sobrin MD, MPH
Jerry A. Shields MD Assistant Professor of Ophthalmology
Adrienne W. Scott MD Director Massachusetts Eye and Ear Infirmary
Assistant Professor of Ophthalmology Oncology Service Harvard Medical School
Wilmer Eye Institute Wills Eye Institute Boston, MA, USA
Baltimore, MD, USA Professor of Ophthalmology Chapter 63
Chapter 57 Thomas Jefferson University Hospital
Philadelphia, PA, USA Akrit Sodhi MD, PhD
Jerry Sebag MD, FACS, FRCOphth, FARVO Chapter 132, Chapter 135, Chapter 143, Chapter 152 Assistant Professor
Professor of Clinical Ophthalmology Retina Division
VMR Institute Kei Shinoda MD, PhD Wilmer Eye Institute
Huntington Beach, CA, USA Associate Professor Johns Hopkins University
Chapter 21 Department of Ophthalmology Baltimore, MD, USA
Teikyo University School of Medicine Chapter 141
Johanna M. Seddon MD, ScM Tokyo, Japan
Professor of Ophthalmology Chapter 8 Elliott H. Sohn MD
Tufts University School of Medicine Assistant Professor
Director of Ophthalmic Epidemiology & Dhananjay Shukla MS, MAMS Department of Ophthalmology
Genetics Services Professor of Ophthalmology University of Iowa Hospitals and Clinics
Turfs Medical Center, Consultant, Retina-Vitreous Service Iowa City, IA, USA
Boston, MA, USA Aravind Eye Hospital and Postgraduate Chapter 42
Chapter 63, Chapter 139, Chapter 140 Institute
Tamil Nadu, India Gisèle Soubrane MD, PhD, FEBO, FARVO
H. Nida Sen MD, MHSc Chapter 83 Consultant Professor
Director Department of Ophthalmology
Uveitis and Ocular Immunology Fellowship Paul A. Sieving MD, PhD University of Paris-East Créteil Val de Marne
Program Director Créteil, Paris, France
National Eye Institute National Eye Institute Chapter 30
National Institutes of Health National Institutes of Health
Bethesda, MD Bethesda, MD, USA Leigh Spielberg MD
Associate Clinical Professor Chapter 37 Ophthalmology Resident
Department of Ophthalmology Rotterdam Eye Hospital
The George Washington University Paolo A.S. Silva MD Rotterdam, The Netherlands
Washington DC, USA Instructor in Ophthalmology Chapter 58
Chapter 77 Harvard Medical School
Associate Surgeon Sunil K. Srivastava MD
Yasir Jamal Sepah MBBS Brigham and Women’s Hospital Staff Physician
Research Fellow Attending Staff Cole Eye Institute
Retinal Imaging Research and Reading Center Beth Israel Deaconess Medical Center Cleveland Clinic Foundation
Wilmer Eye Institute Staff Ophthalmologist Cleveland, OH, USA
Johns Hopkins University School of Medicine Assistant Chief of Telemedicine Chapter 133
Baltimore, MD, USA Beetham Eye Institute
Chapter 78 Joslin Diabetes Center Oliver Stachs PhD
Boston, MA, USA Physicist
Sanjay Sharma BSc, MD, FRCS, MSc (Epid), Chapter 48 Department of Ophthalmology
MBA Faculty of Medicine
Professor of Ophthalmology and Claudio Silveira MD, PhD University of Rostock
Epidemiology Professor of Ophthalmology Rostock, Germany
Director, Cost Effective Ocular Health Policy Clinica Silveira Chapter 9
Unit Erechim, RS, Brazil
Queen’s University Chapter 85 Giovanni Staurenghi MD
Kingston, Ontario, Canada Professor of Ophthalmology
Chapter 59, Chapter 131, Chapter 152 Arun D. Singh MD Chairman Eye Clinic
Professor of Ophthalmology Department of Clinical Science Luigi Sacco
Tarun Sharma MS, FRCS Ed, MBA Director, Department of Ophthalmic Sacco Hospital
Director Oncology University of Milan
Shri Bhagwan Mahavir Vitreoretinal Services Cole Eye Institute, Cleveland Clinic Milan, Italy
Sankara Nethralaya Foundation Chapter 2
Chennai, India Cleveland, OH, USA
Chapter 50, Chapter 109 Chapter 144, Chapter 156
Paul Sternberg Jr MD Hiroko Terasaki MD, PhD J. Niklas Ulrich MD
G.W. Hale Professor and Chair Professor and Chair Assistant Professor
Vanderbilt Eye Institute Department of Ophthalmology Department of Ophthalmology
Associate Dean for Clinical Affairs Nagoya University Graduate School of University of North Carolina at Chapel Hill xxi
Vanderbilt School of Medicine Medicine Chapel Hill, NC, USA
Assistant Vice Chancellor for Adult Health Nagoya, Japan Chapter 122

Contributors
Affairs Chapter 102
Vanderbilt Medical Center Russell N. Van Gelder MD, PhD
Nashville, TN, USA Matthew A. Thomas MD Boyd K. Bucey Memorial Chair
Chapter 22, Chapter 110, Chapter 129 Clinical Professor of Ophthalmology Professor and Chair
The Retina Institute Department of Ophthalmology
Edwin M. Stone MD, PhD Washington University Adjunct Professor
Professor St Louis, MO, USA Department of Biological Structure
Department of Ophthalmology Chapter 119 University of Washington
University of Iowa Seattle, WA, USA
Iowa City, IA, USA John T. Thompson MD Chapter 88
Chapter 42 Assistant Professor
The Wilmer Institute Jan C. van Meurs MD, PhD
Ilene K. Sugino MA Johns Hopkins University Professor
Principal Research Associate Baltimore, MD, USA The Rotterdam Eye Hospital and Erasmus
Institute of Ophthalmology and Visual Chapter 99 University
Sciences Rotterdam, The Netherlands
New Jersey Medical School Gabriele Thumann MD Chapter 121
University of Medicine and Dentistry Professor
Newark, NJ, USA Department of Ophthalmology Daniel Vítor Vasconcelos-Santos MD, PhD
Chapter 125 RWTH Aachen University Preceptor of Ophthalmology and Pathology
Aachen, Germany Universidade Federal de Minas Gerais
Lori S. Sullivan PhD Chapter 16 Belo Horizonte, MG, Brazil
Faculty Associate, Human Genetics Center Chapter 74
Division of Epidemiology Cynthia A. Toth MD
Human Genetics and Environmental Sciences Professor Demetrios G. Vavvas MD, PhD
School of Public Health Department of Ophthalmology and Chief Resident in Ophthalmology
The University of Texas Health Science Center Biomedical Engineering Massachusetts Eye and Ear Infirmary
Houston, TX, USA Duke Universtiy Massachusetts General Hospital
Chapter 31 Durham, NC, USA Boston, MA, USA
Chapter 120 Chapter 26
Paul Sullivan MBBS, MD, FRCOphth
Consultant Ophthalmic Surgeon Michael T. Trese MD G. Atma Vemulakonda MD
Director of Education Clinical Professor of Biomedical Sciences Assistant Professor
Moorfields Eye Hospital Eye Research Institute Vitreoretinal Surgery and Disease
London, UK Oakland University Department of Ophthalmology
Chapter 100 Rochester, MI University of Washington
Chief of Pediatric and Adult Vitreoretinal Seattle, WA, USA
Jennifer K. Sun MD, MPH Surgery Chapter 88
Assistant Professor of Ophthalmology William Beaumont Hospital
Harvard Medical School Royal Oak, MI, USA Hao Wang MD
Chief, Center for Clinical Eye Research and Chapter 114 Ophthalmologist and Vitreoretinal Specialist
Trials Capital Region Retina
Staff Ophthalmologist Julie H. Tsai MD Albany, NY, USA
Beetham Eye Institute Assistant Professor Chapter 125
Joslin Diabetes Center Department of Ophthalmology
Boston, MA, USA SUNY-Stony Brook Yusheng Wang MD, PhD
Chapter 48 Stony Brook, NY, USA Professor
Chapter 84 Department of Ophthalmology
Janet S. Sunness MD Xijing Hospital
Medical Director Mary E. Turell MD Fourth Military Medical University
Richard E. Hoover Low Vision Rehabilitation Ophthalmic Oncology Fellow Xi’an, PR China
Services Cole Eye Institute Chapter 16, Chapter 97
Greater Baltimore Medical Center Cleveland Clinic
Baltimore, MD, USA Cleveland, OH, USA James D. Weiland PhD
Chapter 92 Chapter 156 Associate Professor
Ophthalmology and Biomedical Engineering
Ramin Tadayoni MD, PhD Patricia L. Turner MD University of Southern California
Professor Clinical Associate Professor of Los Angeles, CA, USA
Department of Ophthalmology, Ophthalmology Chapter 126
Hôpital Lariboisière University of Kansas School of Medicine
Paris, France Kansas City, KS, USA Richard G. Weleber MD, FACMG
Chapter 117 Chapter 90 Professor of Ophthalmology and Molecular
and Medical Genetics
Shibo Tang MD, PhD Nitin Udar PhD Casey Eye Institute
Professor Project Scientist Department of Ophthalmology
Department of Retina Department of Ophthalmology Oregon Health and Science University
Zhongshan Ophthalmic Center University of California, Irvine Portland, OR, USA
Sun Yat-Sen University Irvine, CA, USA Chapter 40
Guangdong, PR China Chapter 32
Chapter 41
Moody D. Wharam Jr MD, FACR, FASTRO Tien Yin Wong MD, PhD Glenn Yiu MD, PhD
Professor of Radiation Oncology Director Clinical Fellow in Ophthalmology
Department of Radiation Oncology and Singapore Eye Research Institute Department of Ophthalmology
xxii Molecular Radiation Sciences Professor Massachusetts Eye and Ear Infirmary
The Sidney Kimmel Comprehensive Cancer Department of Ophthalmology Harvard Medical School
Center National University Health System Boston, MA, USA
Contributors

Johns Hopkins CRB II Kent Ridge, Singapore Chapter 33


Baltimore, MD, USA Chapter 49
Chapter 151 Young Hee Yoon MD, PhD
David M. Wu MD, PhD Professor and Chair
Louisa Wickham FRCOphth, MD, MSc Clinical Instructor Department of Ophthalmology
Consultant Vitreoretinal Surgeon Doheny Eye Institute Asan Medical Center
Moorfields Eye Hospital Keck School of Medicine University of Ulsan College of Medicine
London, UK University of Southern California Seoul, South Korea
Chapter 29, Chapter 116 Los Angeles, CA, USA Chapter 102
Chapter 44
Peter Wiedemann MD Hyeong Gon Yu MD, PhD
Professor of Ophthalmology Yanors Yandiev MD Professor
Department of Ophthalmology Professor Department of Ophthalmology
University of Leipzig, Faculty of Medicine Department of Ophthalmology and Eye Seoul National University
Leipzig, Germany Hospital Seoul, South Korea
Chapter 97, Chapter 115 University of Leipzig Chapter 123
Leipzig, Germany
Henry E. Wiley MD Chapter 97 Alex Yuan MD, PhD
Staff Clinician Ophthalmologist
Division of Epidemiology and Clinical Chang-Hao Yang MD, PhD, EMBA Cole Eye Institute
Applications Associate Professor of Ophthalmology Cleveland Clinic
National Eye Institute Department of Ophthalmology Cleveland, OH, USA
National Institutes of Health National Taiwan University Hospital Chapter 53
Bethesda, MD, USA Taipei, Taiwan
Chapter 47 Chapter 96 Marco A. Zarbin MD, PhD
Alfonse A. Cinotti, MD/Lions Eye Research
C. P. Wilkinson MD Chung-May Yang MD Professor
Chairman Professor Chair
Department of Ophthalmology Department of Ophthalmology Institute of Ophthalmology and Visual
Greater Baltimore Medical Center National Taiwan University Hospital Science
Professor, Department of Ophthalmology National Taiwan University College of New Jersey Medical School
Johns Hopkins University Medicine Newark, NJ, USA
Baltimore, MD, USA Taipei, Taiwan Chapter 36, Chapter 67, Chapter 125
Chapter 106 Chapter 96
Jun Jun Zhang MD
David J. Wilson MD Lawrence A. Yannuzzi MD Professor
Thiele-Petti Chair Professor of Clinical Ophthalmology Department of Ophthalmology
Department of Ophthalmology Columbia University Medical School West China Hospital, Sichuan University
Director, Casey Eye Institute Vice-Chairman, Director Chengdu, PR China
Oregon Health and Science University Retinal Research Center Chapter 46
Portland, OR, USA Manhattan Eye, Ear and Throat Hospital
Chapter 154 President of the Macula Foundation Inc. Kang Zhang MD, PhD
New York, NY, USA Professor of Ophthalmology and Human
Thomas J. Wolfensberger MD, PD, MER Chapter 55 Genetics
Associate Professor Director, Institute for Genomic Medicine
Vitreoretinal Department Miho Yasuda MD Department of Ophthalmology
Jules Gonin Eye Hospital Assistant Professor University of California, San Diego
University of Lausanne Department of Ophthalmology La Jolla, CA
Lausanne, Switzerland Graduate School of Medical Sciences Shiley Eye Center
Chapter 28, Chapter 118 Kyushu University UCSD Zhang Lab
Fukuoka, Japan La Jolla, CA, USA
David Wong MBChB (Liverpool), FRCS Chapter 68 Chapter 46
(Eng), FRCP (UK), FRCOphth
Chair Professor Po-Ting Yeh MD Mingwei Zhao MD
Eye Institute Lecturer Professor of Ophthalmology
The University of Hong Kong Department of Ophthalmology Department of Ophthalmology
Hong Kong National Taiwan University College of People’s Hospital
Chapter 104 Medicine Peking University
Taipei, Taiwan Beijing, PR China
Ian Y. Wong MBBS (HKU), MMed Chapter 96 Chapter 91
(Singapore), MRCS (Edinburgh),
FCOphthHK, FHKAM (Oph) Zohar Yehoshua MD, MHA Peng Zhou MD, PhD
Assistant Professor Assistant Research Professor Attending Ophthalmologist
Eye Institute Department of Ophthalmology Department of Ophthalmology
The University of Hong Kong University of Miami Eye and ENT Hospital
Hong Kong Miller School of Medicine Fudan University
Chapter 104 Miami, FL, USA Shanghai, PR China
Chapter 3 Chapter 33, Chapter 91
Dedication
The original and all subsequent editions of RETINA are dedi- academicians and clinicians around the world. He was the
cated to the clinicians and scientists who have contributed to Professors’ Professor.
the education in our field of medical students, residents, and
fellows, and especially to retina specialists and all ophthalmolo- For the Fourth Edition we added a special dedication to Arnall
gists who participate in continuing medical education. We rec- Patz (1920–2010), who was an editor of the original edition.
ognize that we are all students and committed to lifelong Arnall was a pioneer and leader in the establishment of the field
learning, especially in our field of retina. of medical retina. He founded the Retinal Vascular Center at the
Wilmer Institute and, subsequently, he became the Director of
The Second Edition included a special dedication to Ronald the Wilmer Institute. He trained many of today’s leaders in the
G. Michels (1942–1991), who was vitally involved in the plan- field and many contributing authors to RETINA. Arnall was an
ning of the original edition and in the recruitment of our initial inspiration for the multitude of retinal specialists around the
team of editors and authors. Ron was an enthusiastic and tal- world.
ented leader in vitreoretinal surgery. His teaching and innova-
tions had a major impact on the other editors of RETINA For this present edition we wish to stress the development of
specifically and on the entire field of ophthalmology generally. knowledge and the contribution of the international community
We are thankful for the privilege of having known and worked of retinal specialists. From the time of the First Edition in 1989
with Ron. – more than 20 years ago – we have benefited from the rapid
evolution of science – basic and clinical – in all fields related
For the Third Edition we offered an additional special dedica- to biology and medicine, and especially in relation to ophthal-
tion to A. Edward Maumenee (1913–1998), a true giant who mology and our chosen specialty of retina. The evolution of
influenced virtually every field and subspecialty in ophthalmol- knowledge and contributions of colleagues from around the
ogy. While most of his later contributions involved anterior world have shown that there are no borders; the free exchange
segment surgery, his original observations regarding macular of information directly benefits our patients in the prevention
degeneration provided a basis for subsequent clinical and of the most common forms of blindness caused by retinal dis-
research investigations in this area. As a gifted teacher, relent- eases. Thus, it is wholly appropriate that this Fifth Edition of
less investigator, and treasured mentor, Ed inspired the editors RETINA is dedicated to the international community of retinal
and many authors of this textbook, as well as a multitude of clinicians and educators.
Foreword
“In the fields of observation, chance favors the prepared mind.” Volume 3 Retinal surgery, complicated forms of retinal
Louis Pasteur detachment, vitreous surgery, and tumors of
retina, choroid, and vitreous.
In science and medicine, it is said that the doubling time for
information is two and a half years. By this measure, information Authors of the 156 chapters in the three volumes are inter-
regarding the retina and the intimately interactive brain has nationally recognized leaders of biomedical science and
increased more than four-fold since the most recent edition of medical–surgical ophthalmology. Each chapter is authoritatively
RETINA in 2006. presented with the central purpose of providing current,
Related to the retina and the visual system, the massive evidence-based and clinically relevant information. Authorship
increase in knowledge extends from the approximately 125 by outstanding clinician–scientists is extremely important,
million rod and cone photoreceptors in each eye, through the because the value of RETINA stems in large part from the
hierarchical circuitry of the inner retina to nearly one-third of scholarship, content selection, and clinical experience of the
the 100 trillion neurons in the human brain that participate in chapter authors.
vision. Also embracing the glial and vascular structures, retinal In this Internet era with widespread electronic communication
pigment epithelium and contiguous vitreous and choroid, new and data access, RETINA serves at least two purposes. First, it
retina-related information is derived from genetics and the presents an organized framework for the vast amount of retina-
Human Genome Project, basic sciences such as neurobiology related data and builds the “prepared mind” for clinical exami-
and immunology, applied disciplines including pharmacology nation, ancillary testing, diagnosis, and treatment of retinal
and bioengineering, as well as the emerging sciences of regen- abnormalities. Second, RETINA is a peer-created reference for
erative medicine and nanotechnology. specific information regarding retinal, choroidal, and vitreal
Assimilating this wealth of discovery, the Editor-in-Chief, diagnostics, abnormalities, and therapies. As a comprehensive
Stephen J. Ryan MD, and Editors of RETINA are to be com- framework of organized information and an authoritative
mended for maintaining the focus on information that is essen- resource, RETINA is requisite to sustained competence in retinal
tial to clinical medicine and retina specialty practice through: clinical research and retinal specialty practice.

Volume 1  Retinal imaging and diagnostics, basic science,


genetics, and translation to therapy; Bradley R. Straatsma MD, JD
Volume 2 Retinal degeneration and dystrophy, vascular Jules Stein Eye Institute
disease, and chorioretinal inflammatory disease; David Geffen School of Medicine at UCLA
and University of California, Los Angeles, CA, USA
Preface
Retinal specialists, both clinicians and scientists, share a fascina- treatments for certain consequences of age-related macular
tion with the retina, a unique tissue. The retina forms the ana- degeneration. Around the time of the publication of the First
tomic and physiologic basis for the gift of sight and accounts for Edition, the understanding of molecular mechanisms of angio-
over 35% of the neurons entering and exiting the human brain. genesis was in its infancy; this edition documents how intravit-
In the 21st century we are fortunate to be in the middle of the real pharmacotherapy has become standard care for the leading
most exciting scientific revolution in the history of man. We all blinding retinal diseases. These diseases and treatments are
anticipate that this revolution in biomedicine and our present stressed in Volume 2.
“century of biology” predict a bright future for our specialty of Volume 3 addresses both surgical diseases and the oncology
retina. section. Human retina is a direct outgrowth of the forebrain
It is helpful to recall that although there had long been a fas- and, therefore, is the most accessible part of the brain for neu-
cination with the eye and sight, and prior speculation regarding roscience studies. Many of us consider the retina to be aestheti-
the retina, it was not until the 19th century that modern ophthal- cally the most beautiful tissue in nature. We hope the reader
mology and the study of the retina truly began. Everyone in our will enjoy and appreciate the major revisions to the figures and
field points with pride to the revolution brought about by von illustrations throughout the text. The new edition includes 20
Helmholtz in 1850 with his introduction of his innovative oph- new chapters and 1304 new figures.
thalmoscope in Berlin. Great contributions in ophthalmology Thus, this Fifth Edition features a significant reorganization as
and descriptions of the retina followed, particularly in Europe. well as a tremendous consolidation – for example, one chapter
In the 20th century, ophthalmologists from the United States in this new edition has replaced what were previously five chap-
joined in these contributions. Further advances in this 21st ters in the Fourth Edition. At the same time, we have added
century, with important contributions from every continent – whole new chapters in every section of the book. The field of
Asia, Latin America, North America, Australia, Africa, and retina is so broad and the contribution of knowledge is so rapid
Europe – have continued at breakneck speed with the full expec- that we cannot be comprehensive, but we have attempted to
tation that the entire world will participate in this expansion of weight the content to match the needs of the wide array of our
our knowledge of the retina and retinal diseases. readers worldwide. The authors have updated their references,
As scientific knowledge explodes, it is often at the interface of as the reader will note; most chapters include references from
different disciplines and of science and clinical observation that 2011.
great contributions are made. Ophthalmology is fortunate to be We have many authors from around the world contributing
at the forefront of the interaction of engineering, biology, and to this Fifth Edition, who are recognized for their leadership in
medicine. From the time the laser was invented in 1960, the retina throughout the world.
specialty of ophthalmology was the earliest adapter, with clini- As a truly international multi-authored text, there are multiple
cal applications developed within 10 years. literary styles. The editors have worked to provide a level of
While we can see a logical progression over the five editions conformity and scientific balance to preserve accuracy and
of RETINA, if one steps back to the publication of the first edition knowledge as of 2012 without sacrificing the originality and
in 1989, the progress in science since that time has truly been style of the individual authors.
breathtaking. It is remarkable to recall the vigorous debate as to The editors gratefully acknowledge the support of many of the
the chances of success of sequencing the human genome, whereas faculty and staff of the Wilmer Eye Institute and the Doheny Eye
now with the advent of high throughput whole genomic and Institute and many eye institutions from around the world who
epigenomic analyses we have entered a new era in discovering have contributed immensely to the completion of this project.
the genetic basis of retinal disease. Timothy Hengst, whose artistic talent is displayed in the illustra-
In revising RETINA for the Fifth Edition, we now begin tions, contributed substantially to the quality of these volumes.
Volume 1 with imaging, where there is a veritable revolution We thank the office staffs of the individual authors and editors
underway. The human eye cannot resolve what advanced and, in particular, Joy Roque, who contributed tremendously to
imaging technologies including ocular coherence tomography this effort. We have enjoyed working with Elsevier and, specifi-
(OCT) can detect. This is followed by the basic sciences, which cally, Russell Gabbedy, Nani Clansey, and Caroline Jones and
have been updated to include not only the fundamentals of their staff, who share our commitment to the highest standards
retinal anatomy and physiology, but also recent advances in of quality for RETINA.
cell injury, genetics (both nuclear and mitochondrial) and epi-
genetics, and their translation to novel therapies that encompass S.J.R.
stem cells, neuroprotection, gene therapy, nanotechnology, and A.P.S.
drug delivery to the posterior segment. C.P.W.
One constant is that the second volume remains devoted to D.R.H.
medical retina. Tremendous progress has been made in retinal S.R.S.
vascular diseases such as diabetic retinopathy and with P.H.W.
Video Table of Contents
Section 1: Retinal Reattachment: General Surgical Principles Section 4: Retinal Vascular Diseases
and Techniques
20. Vitrectomy and Membrane Dissection of Diabetic
1. Techniques of Scleral Buckling Traction Retinal Detachment
Paul Sullivan - Chapter 100 Jose Garcia-Arumi
2. A case of Primary Vitrectomy for Treating 21. Vitrectomy for Diffuse Diabetic Macular Edema
Rhegmatogenous Retinal Detachment Zdenek J. Gregor, Lyndon da Cruz
Young Hee Yoon - Chapter 102
Section 5: Complications in Vitreoretinal Surgery
3. Primary Vitrectomy for Rhegmatogenous Retinal
Detachment 22. Complications in Vitreoretinal Surgery – Introduction
Hiroko Terasaki - Chapter 102 (Text only)
4. Vitrectomy Repair of Pseudophakic Rhegmatogenous Kourous Rezaei
Retinal Detachment 23. Always Measure Prior to Trochar Insertion
Bernd Kirchhof, Heinrich Heimann Stanislao Rizzo
5. Vitrectomy Repair of Rhegmatogenous Retinal 24. Suprachoroidal Infusion
Detachment without Postoperative Tamponade Yusuke Oshima
Vicente Martinez Castillo 25. Subretinal Insertion of Endo-illuminator
6. Optimal Procedures for Retinal Detachment Repair Kourous A. Rezaei
G. William Aylward - Chapter 105 26. Dislocated IOL and Capsular Tension Ring
Section 2: Complicated Forms of Retinal Detachment Andre Vieira Gomes
27. Iatrogenic Breaks during the Induction of Posterior
7. Proliferative Vitreoretinopathy Vitreous Detachment
Ian Constable - Chapter 107 Manish Nagpal
8. Proliferative Vitreoretinopathy 28. Iatrogenic Macular Hole during VMT Surgery
John T. Thompson Paul Sullivan
9. Relaxing Retinopathy: PVR / Retinal Detachment 29. Iatrogenic Breaks during the Delamination of Diabetic
Gary W. Abrams, Enrique Garcia-Valenzuela, Sumit K. Nanda Traction Retinal Detachment
10.  Posterior Segment Reconstruction of a Perforating Stratos Gotzaridis
Ocular Injury 30. Point Pressure Hemostasis during Diabetic Vitrectomy
Franco M. Recchia Ehab N. EL Rayes
Section 3: Vitreous Surgery for Macular Disorders 31. Iatrogenic Retinal Break during ERM Peeling
Paul Sullivan
11. Macular Hole
32. Subretinal Brilliant Blue
Alain Gaudric - Chapter 117
J. Fernando Arevalo, Abdulaziz Adel Rushood
a.  Induction of Posterior Vitreous Detachment. Weiss’ ring
and Posterior Hyaloid 33. Peripheral Retinal Detachment during Macular Hole
b. Ablation of an Epimacular Membrane around the Macular Surgery
Hole Stanley Chang
c. Inner Limiting Membrane removal 34. Subretinal Hemorrhage
12. Macular Hole Surgery Manish Nagpal
Raymond N. Sjaarda 35. Macular Fold
13. Membrane Peeling: Epiretinal Membrane Andre J. Witkin, Jason Hsu
Robert N. Johnson, H. Richard McDonald, Everett Ai, 36. Subretinal Perfluorocarbon
J. Michael Jumper, Arthur D. Fu Carl Claes
14. Surgical Management of Submacular Hemorrhage 37. Subretinal Perfluorocarbon Injection during En Bloc
Nancy M. Holekamp, Henry C. Lee Perfluorodissection
15. 360 Macular Translocation J. Fernando Arevalo
Cynthia A. Toth, Prithvi Mruthyunjaya, Marcin Stopa 38. Intraocular Foreign Body Dislodged on the Macula
16. Submacular Surgery of Peripapillary Choroidal Martin Charles
Neovascular Membrane 39. Management of Massive Suprachoroidal Hemorrhage
Lawrence P. Chong, Michael Samuel Jose Garcia Arumi
17. Vitrectomy and Membrane Peeling: Epiretinal Membrane 40. Hemorrhagic Choroidal Detachment after “One Stitch”
Secondary to Inflammation and Retinal Detachment Vitrectomy Surgery
Allen E. Kreiger, Christine R. Gonzales Susanne Binder
18. Submacular Surgery with Autologous Transplantation of 41. Dislocated Phakic IOL
RPE Virgilio Morales-Canton
Jan Van Meurs 42. Dislocation of the Tip of the Soft Tip Cannula
19. Vitrectomy for Myopic Foveoschisis Andre Vieira Gomes
Yasushi Ikuno 43. Iatrogenic Peripheral Retinal Breaks during IOFB
Extraction
Martin Charles
44. Peri-silicone Proliferation 50. Intraocular Retinal Prosthesis Implantation
Jose Garcia Arumi Mark Humayun
x 45. Miragel Buckle Removal 51. Perflouron Placement and Subretinal Injection
Stanislao Rizzo Procedure
46. Subconjunctival Silicone Oil Removal Albert M. Maguire, Jean Bennett - Chapter 34
Video Table
of Contents

Susanne Binder 52. Pars Plana Implantation of a Sustained Steroid Drug


Delivery System
Section 6: Ocular Tumors
Glen Jaffe
47. Exoresection of Choroidal Melanoma 53. Diffuse Unilateral Subacute Neuronetinitis
Bertil Damato - Chapter 147 Eduardo C. de Souza, Yoshitaka Nakashima
48. Iridocyclectomy for Ciliary Body Leiomyome 54. Subretinal Trypan Blue for Identifying Retinal Breaks
Jennifer I. Lim V. Sivagnanavel, T. Jackson, A. Kwan, G. William Aylward
Section 7: Miscellaneous 55. Vitrectomy for Trapped Pre-retinal Blood: “Shaken
Baby” Syndrome
49. Vitreous Surgery for 4A Retinopathy of Prematurity Peter Wiedemann, Petra Meier
Michael T. Trese
Supporting videos in section 5 available online at
www.expertconsult.com

Complications in Vitreoretinal Surgery


Kourous Rezaei MD

Good judgment is to make the best decision based on the known It is important to always measure the distance of the trochar
information in hand, and it is a product of experience. However, insertion from the limbus (range 3-4 mm posterior to the limbus
experience usually comes from the lessons learned from previ- depending on the lens status). One may use either a caliper or
ously made bad judgments. As surgeons we are trained to the other side of the trochar inserter (which also serves as a
predict and treat expected events during surgery. Unexpected caliper) for measuring. If the trochar is not inserted at a correct
events; however, are fact of life and generally the question is not distance to the limbus it should be removed and re-inserted
if they will happen but when they will happen. They are fre- correctly.
quently dangerous and can lead to undesirable outcomes.
The knowledge of how to predict, treat, and prevent unex-
SUPRACHOROIDAL INFUSION
pected events during surgery is extremely valuable and would
make vitreoretinal surgery safer, leading to improved visual Yusuke Oshima MD
outcomes in patients. The more one is acquainted with unex-
pected events, the less they are considered unexpected since one Suprachoroidal infusion is a serious intra-operative complica-
has already seen these events happen and knows how they were tion during vitrectomy surgery. This complication has become
handled and therefore the factor of surprise is eliminated. more prevalent with the use of trochar systems.
In this chapter, experienced surgeons from around the world At the beginning of the surgery, it is important to visualize
share with you their unexpected experiences during retinal the tip of the infusion cannula in the vitreous cavity prior to
surgery and show how they handle some of the most unusual opening the infusion line. A well-constructed scleral wound
surgical cases. Further, they share their surgical pearls on how may also prevent the dislodgement of the infusion cannula
to predict, prevent, and treat these unusual surgical cases. during surgery.
If the tip of the infusion cannula is dislodged into the
suprachoroidal space during the surgery one should imme-
ALWAYS MEASURE PRIOR TO
diately stop the flow the infusion into the eye. Generally, the
TROCHAR INSERTION infusion needs to be re-inserted into the eye through a dif-
Stanislao Rizzo MD ferent trochar. Pulling back the original infusion trochar slightly
outwards so that its tip is placed in the suprachoroidal space
Vitrectomy surgery with trochar systems reduces conjunctival may help the draining of the suprachoroidal fluid. Many times
trauma, scleral manipulation, iatrogenic peripheral breaks, post- the residual fluid in the suprachoroidal space can be left
operative inflammation, and corneal astigmatism. alone.
Precise measuring of the location of the trochar insertion is
crucial to assure a pars plana entrance into the eye, especially
when utilizing an oblique insertion technique. Further, it is
SUBRETINAL INSERTION OF
important that the insertion tunnel is parallel to the limbus so ENDO-ILLUMINATOR
that the distance between the entrance point into the eye and
Kourous A. Rezaei MD
limbus is the same as the scleral insertion point and limbus.
An insertion that is too anterior to the limbus (less than Patient underwent combined vitrectomy surgery and scleral
3  mm posterior to the limbus) leads to an inadvertent insertion buckling for a total retinal detachment with PVR. The inferotem-
into the cilliary body which is very vascular and may cause poral trochar was inserted and after the visualization of the tip
bleeding as shown in the video (case 1). An anterior insertion of the infusion cannula, the infusion line was opened. As the
may also lead to cataract formation or intraocular lens implant endo-illuminator was inserted into the eye through the trochar
dislocation. In these cases the cannula needs to be removed (tunneled incision), it was noted that it was placed under the
and re-inserted. retina. The endo-illuminator was removed and the cannula was
An insertion that is too posterior to the limbus, (more than examined and the tip of the cannula was under the pars plana
4 mm posterior to the limbus) may lead to a subretinal trochar (video#1).
insertion inducing retinal breaks and retinal detachment (case To resolve this situation one option would be to remove
2). In these patients the iatrogenic breaks need to be treated the trochar, suture the sclerotomy, and re-insert the trochar
similar to other peripheral breaks: thorough peripheral vitrec- through a new sclerotomy. The simpler option is shown in
tomy, endolaser, and tamponade. the video #2:
The trochar was removed and re-inserted through the same VMT is characterised by the presence of a partial vitreous
sclerotomy without tunneling the incision (inserted almost detachment with strong adhesions to the macula, especially the
e2 perpendicular to the sclera). At this point, the tip of the endo- fovea.
illuminator could be easily visualized. At the conclusion of This patient had a long history of reduced vision. While using
the surgery the sclerotomy was sutured. active aspiration over the disc to detach the hyaloid the surgeon
In the presence of pars plana detachment or hypotony the is focused on looking for signs of vitreous separation from the
tunneled trochars could be placed under the pars plana. In these disc rather than the traction vectors at the fovea as the fovea
cases one may avoid tunneling the incision and insert the trochar tissue is stretched before finally separating. In fact the central
perpendicular to the sclera. These sclerotomies would require lucent area was assumed to be a cyst as no hole was present
suturing at the conclusion of surgery. pre-operatively.
To summarize, observe the fovea closely while detaching the
DISLOCATED IOL AND CAPSULAR hyaloid in conditions with foveal thinning and strong vitreo-
Complications in Vitreoretinal Surgery

TENSION RING retinal adhesions such as vitreomacular traction syndrome. In


the presence of significant vitreo-macular adhesion and/or
Andre Gomes MD cystic macular changes one may trim the vitreous around the
The patient was presented for the removal of a dislocated intra- macular area rather than peeling it, leading to the un-roofing
ocular lens implant (IOL) after a difficult phaco-emulsification. of macula.
During the surgery, after the IOL removal it was noted that the
patient also had a capsular tension ring. Removal of the ring can IATROGENIC BREAKS DURING
be challenging due to their large size and their elasticity. They
THE DELAMINATION OF DIABETIC
may also have extensions that can induce traction to the periph-
eral vitreous. It is important to remove the ring gently to mini- TRACTION RETINAL DETACHMENT
mize traction and to remove it by rotating it (rather than pulling Stratos Gotzaridis MD
it) out of the eye. In patients who present with a dislocated intra-
ocular lens, after the removal of the lens it is important to inspect The newly designed 25G+ vitrectomy probes (Alcon Laborato-
the anterior pars plana very carefully for the capsular tension ries) have their opening closer to the tip when compared to 23
ring. It can be difficult to detect and (if missed) may dislocate or 20 gauge probes. This proximity to the surface of the retina
into the vitreous cavity over time. allows it to be used similar to scissors during the delamination
of diabetic fibrous tissue. However, small iatrogenic retinal
IATROGENIC BREAKS DURING breaks may be formed during the delamination. The utilization
THE INDUCTION OF POSTERIOR of a bimanual technique would minimize this complication. In
the bimanual technique the use of chandelier light would free
VITREOUS DETACHMENT
up the surgeon’s hand to hold the forceps instead of the endo-
Manish Nagpal MD illuminator. Keeping the level of the delaminating tissue stable
with the forceps would allow the probe to cut the focal adhesion
Formation of peripheral breaks during the induction of posterior points more accurately. If these adhesion points are broad and
vitreous detachment (PVD) in a patient with macular hole is difficult to be accessed with the vitreous cutter, then scissors
demonstrated. If these patients generally have a very adhesive should be used to make a dissection plane for subsequent tissue
vitreous, the induction of PVD may induce peripheral breaks. removal. The iatrogenic breaks need to be marked and treated
The use of wide angle viewing system would allow the monitor- with endolaser followed by a gas tamponade. It is important to
ing of peripheral retina during the induction of PVD. alleviate all the traction around the breaks to prevent retinal
At the beginning, the surgeon is paying attention to the optic re-detachment.
nerve and the grasping of the Weiss ring. Once the Weiss ring
is grasped and the PVD is in process of expanding, attention
must be given to the peripheral retina to identify the location of POINT PRESSURE HEMOSTASIS DURING
the anterior vitreous base. This is the location where the vitreous DIABETIC VITRECTOMY
can no longer be separated and breaks will form if suction/trac-
tion is applied. Ehab El Rayes MD
In these patients it is extremely important to screen the periph- Intra-operative bleeding is common during diabetic dissection.
ery at the conclusion of the surgery. In the presence of peripheral Generally one can apply increased intra-ocular pressure or endo-
breaks, a thorough peripheral vitrectomy should be performed diathermy/endolaser to achieve hemostasis. However, none of
around the breaks to relive any residual traction, followed by these techniques are preferable since the retina is generally
retinopexy, and possibly a tamponade agent. already ischemic with poor circulation. Lack of good hemostasis;
however, can lead to significant bleeding during surgery and the
IATROGENIC MACULAR HOLE DURING removal of the pre-retinal blood can become challenging due to
VMT SURGERY its “stickiness” to the surface of retina.
In this video an elegant technique is demonstrated in which
Paul Sullivan MD
gentle pressure is applied to the bleeding points with the tip of
This video illustrates the dangers of excessive traction on the the cutter while simultaneously the blood is removed with the
fovea while detaching the posterior hyaloid in a case of vitreo- suction (if necessary). The intra-ocular pressure is not increased
macular traction syndrome, or VMT. during this maneuver.
IATROGENIC RETINAL BREAK DURING prevent displacement of subretinal fluid into the macula. Under
air, the infusion line was moved to another opening and the
ERM PEELING
fundus was examined by indirect ophthalmoscopy. A retinal e3
Paul Sullivan MD break was found at the ora serrata adjacent to the site of the
cannula. It was treated with cryopexy, and an expanding con-
Retinal laceration is a serious and preventable complication of centration of gas was used. The patient was rotated into the
epiretinal membrane peeling. This video demonstrates a chorio- prone position after surgery. In reviewing the video of this
retinal laceration during ERM peeling. surgery it was noted that the angle of insertion of the trocar-
Having engaged the membrane it is important to elevate the cannula was aimed too posteriorly as it entered the eye.
membrane slightly while pulling. In the video, the membrane is
peeled tangentially as shown by the proximity of the forceps tip SUBRETINAL HEMORRHAGE
to the shadow. This may be due to the inappropriate extrapola-

Complications in Vitreoretinal Surgery


tion from capsulorhexis where it is important not to lift the Manish Nagpal MD
rhexis edge. Because of the concavity of the posterior pole the Subretinal hemorrhage may occur during vitrectomy surgery.
forceps move closer to the retina as the flap extends. The source of blood may be iatrogenic or from already existing
It is important to regularly re-grasp the membrane rather than breaks. When this occurs it is important to increase the intraocu-
peeling with a single grasp. Otherwise it is difficult to pay suf- lar pressure immediately to stop the bleeding (using the foot
ficient simultaneous attention to the forceps tips and the point pedal). If the blood threatens the macula, perfluorocarbon liquid
at which the membrane is delaminating. In fact the surgeon is may be injected to protect the macula and push the subretinal
probably more focused on the delamination point rather than hemorrhage away from the macula. In cases of large subretinal
the location of the tip of the forceps. hemorrhage, the subretinal blood may be removed with a soft
To summarize: while peeling epiretinal membranes, lift the tip cannula through the original break or through a drainage
membrane rather than pulling tangentially as you would in a retinotomy site.
capsulorhexis and keep sight of the tip of the forceps at all times.
It is important to remember that the posterior pole is concave MACULAR FOLD
(especially in high myopia) while peeling away from the center
of the macula. Always assure that the forceps remain within the Andre J. Witkin MD and Jason Hsu MD
surgical field during the peeling. A 57-year-old patient presented with a macula-on rhegmatog-
SUBRETINAL BRILLIANT BLUE enous retinal detachment (RRD), with a posterior linear tear at
1 o’clock. C3F8 gas (0.3cc) was injected into the vitreous with
J. Fernando Arevalo MD, FACS, the goal of performing a staged pneumatic retinopexy; however,
Abdulaziz Adel Rushood MD the next day the patient had a bullous macula-splitting RRD,
with the gas bubble having moved into the subretinal space.
Approximately 0.3 ml of brilliant blue dye was used to stain the A 23-gauge pars plana vitrectomy (PPV) surgery was performed
ILM during macular hole surgery. A significant resistance was that day, with creation of an anterior retinotomy to drain the
noted at the beginning of the dye injection. To overcome this subretinal gas and drainage of subretinal fluid through the
resistance additional pressure was applied. Sudden jet stream of original posterior retinal tear.
brilliant blue was formed and it was noted that the dye has Two weeks after PPV, visual acuity was 20/400 due to a hori-
gained access to the subretinal space through the macular hole. zontal retinal fold that had formed through the macula, and a
The dye was immediately removed from the vitreous cavity, and repeat 23-gauge PPV was performed that week. During surgery,
the ILM was peeled in a circular fashion in the usual manner. a 41-gauge needle was used to inject balanced salt solution into
The eye was filled with 16% perfluoropropane gas. Post- the subretinal space around the retinal fold. A complete air/fluid
operative fundus examination and OCT indicated a closed exchange was then performed, and the sub-retinal fluid slowly
macular hole with areas of choroidal hypereflectivity due to RPE coalesced in the posterior region of the retinal fold. After 5
and choriocapillaris atrophy. The brilliant blue dye was present minutes under air, the retina had completely unfolded. 20% SF6
in the subretinal space more than 3 months after surgery. was exchanged with air, and the patient was instructed to lie in
During the injection of any type of fluid, it is critical to inject the prone position for 5 days. Three weeks after surgery, the
slowly and away from the macula. If dye gains access to the retinal fold had disappeared, and visual acuity had improved to
subretinal space, remove it as much as possible and complete 20/200. Three months after surgery, best-corrected visual acuity
the surgery in the usual fashion. had improved to 20/30 and the retina remained flat. The follow-
ing pearls maybe helpful in treating macular folds:
PERIPHERAL RETINAL DETACHMENT
DURING MACULAR HOLE SURGERY 1. Inject balanced salt solution (BSS) via a translocation needle
(e.g., 41-gauge cannula), preferably in an area near the
Stanley Chang MD
macula but away from the fold.
This 70-year-old woman underwent macular hole surgery and 2. For improved control, consider using the viscous fluid
after separation of the posterior hyaloid, a peripheral retinal injection system when injecting BSS into the subretinal
detachment was noted inferotemporally in the area of the infu- space. Titrate the injection pressure to provide a steady drip
sion cannula. The management of the hole proceeded as usual of BSS when the foot pedal is fully engaged. This may be
with staining of brilliant blue to assist in ILM peeling. At the end titrated under the microscope prior to entering the eye.
of the procedure, a partial fluid-air exchange was made to A steady dripping of BSS rather than a stream is desired.
Depending on the vitrectomy machine the injection pressure To avoid this complication one may inject perfluorocarbon
may vary (For Constellation, (Alcon Laboratories) this very slowly without causing undue pressure. In the presence of
e4 pressure is around 12-16 mm Hg). severe traction detachment, additional instruments may be used
3. Inject slowly to avoid the formation of macular hole. i.e. picks, forceps, and scissors to relieve traction prior to per-
4. Expect that the BSS will not coalesce and unfurl the fold fluorocarbon injection.
until fluid-air exchange is completed. Consider waiting To drain the subretinal perfluorocarbon one may make a
several minutes under air and/or gently shaking the eye peripheral drainage retinotomy site. Then inject perfluorocarbon
to encourage unfurling of the fold. gently over the optic nerve to push the subretinal perfluorocar-
5. While in this case SF6 gas was used, it is very possible that bon peripherally and flatten the posterior pole while simultane-
air tamponade may have been sufficient as the subretinal ously aspirating the subretinal perfluorocarbon with a soft tip
BSS tends to be resorbed within a few days, depending on cannula through the peripheral retinotomy.
the volume.
Complications in Vitreoretinal Surgery

6. Post-operative prone positioning should theoretically lower INTRAOCULAR FOREIGN BODY


the chances of recurrent fold formation.
DISLODGED ON THE MACULA
SUBRETINAL PERFLUOROCARBON Martin Charles MD
Carl Claes MD The patient presented with traumatic cataract and intraocular
foreign body (IOFB). Cataract surgery was performed followed
A 35-year-old man was referred 1 week after vitrectomy surgery
by vitrectomy. During the vitrectomy the IOFB was dislodged
for macula off retinal detachment. A large subfoveal perfluoro-
on the macula. Perfluoron was injected and the intraocular
carbon (PFO) bubble was present with a poor visual acuity.
foreign body was grasped with a magnet and extracted with
Considering the toxicity of subretinal PFO, the decision was
forceps. The posterior hyaloid was detached and peripheral
made to remove the subretinal bubble. The surgical strategy
shaving of the vitreous base was performed.
consisted of inducing an iatrogenic posterior retinal detachment
48 hours postoperatively the OCT indicated a small scar
by injecting BSS into the subretinal space and then displacing
without affecting the center of the macula. 4 weeks later a
the subretinal PFO bubble peripherally by injecting PFO over the
macular pucker has developed. Vitrectomy was performed and
posterior pole. The peripherally displaced subretinal PFO bubble
the ILM was peeled using brilliant blue.
was then aspirated together with the subretinal BSS through a
40 gauge subretinal needle. This was followed by air/fluid
exchange. SUPRACHOROIDAL HEMORRHAGE
To prevent subretinal PFO in retinal detachment surgery, one Jose Garcia Arumi MD
should avoid allowing PFO level passing the most posterior
break. Also, avoid filling the eye with PFO up to the level of the Suprachoroidal hemorrhage is a severe and devastating com-
tears when working in the periphery. In case of posterior breaks, plication of vitreoretinal surgery. Trans-scleral drainage of
these should be completely covered by the PFO. It is important liquefied blood is performed while maintaining relatively
to shorten the duration of PFO presence in the eye and always high intra-ocular pressure with a 25G cannula inserted in the
monitor the meniscus of the PFO bubble with a wide angle anterior chamber. After partial drainage and better visualiza-
viewing system. One should also avoid the accumulation of free tion, trochars were inserted into the pars plana. Additional
moving small bubbles (fish eggs) of PFO around the meniscus suprachoroidal hemorrhage was drained through the tro-
of the main bubble. These small bubbles can gain access to the chars. Pars plana vitrectomy was performed. Intravitreal per-
subretinal space through open breaks in the retina. Further, large fluorocarbon injection pushed the choroidal hemorrhage
breaks and traction associated with breaks can lead to the sub- peripherally allowing further drainage through the anterior
retinal access of PFO. sclerotomies. Silicone oil tamponade was used at the conclu-
sion of the surgery.
SUBRETINAL PERFLUOROCARBON
INJECTION DURING EN BLOC HEMORRHAGIC CHOROIDAL
PERFLUORODISSECTION DETACHMENT AFTER “ONE STITCH”
VITRECTOMY SURGERY
J. Fernando Arevalo MD, FACS
Susanne Binder MD
This video demonstrates perfluorocarbon dissection in a patient
with diabetic traction retinal detachment. Core vitrectomy was A 35-year-old diabetic patient presented with a history of
performed and an opening was made in the mid peripheral uneventful “one stitch” vitrectomy surgery for macular trac-
hyaloid. Perfluorocarbon was injected very slowly between the tion associated with diabetic macular edema. 24 hours after
posterior hyaloid and the retina to generate space and dissect surgery patient has developed a massive exudative/hemor-
the fibrotic tissues (“En Bloc Perfluorodissection”). However, rhagic choroidal detachment with a retinal tear at the supe-
perfluorocarbon gained access to the subretinal space and an rotemporal sclerotomy site. Patient was treated with laser since
iatrogenic retinal detachment was formed. At this point more the break was supported by the choroidals buckling effect.
perfluorocarbon was injected in the posterior pole and the sub- Few days later the patient developed a retinal detachment
retinal perfluorocarbon was drained with a soft tip cannula with persistent large hemorrhagic choroidals nasally and
through a peripherally made drainage retinotomy site. temporally.
It was decided to proceed with a repeat surgery 7 days after breaks must be treated with retinopexy i.e. endolaser and
the original vitrectomy. Infusion cannula was inserted into the adequate tamponade.
anterior chamber to avoid supra-choroidal infusion. External e5
choroidal drainage was performed while using internal perfluo- PERI-SILICONE PROLIFERATION
ron tamponade. Peripheral retina was treated with endolaser
and the eye was filled with silicone oil. Silicone oil was removed Jose Garcia Arumi MD
3 month after the second surgery and the retina remained
Peri-silicone proliferation is a subtype of epiretinal prolifera-
attached.
tion that occurs in eyes filled with silicone oil. In the eye,
silicone oil floats over a meniscus of fluid rich in cytokines
DISLOCATED PHAKIC IOL that could stimulate a severe inferior proliferation. Peri-silicone
proliferation is observed in more than 50% of these patients,
Virgilio Morales MD

Complications in Vitreoretinal Surgery


and may lead to proliferative vitreoretinopathy (PVR) and
A 28-year-old patient presented with a history of previous recurrent inferior retinal detachment requiring repeat vitreous
implantation of a phakic refractive lens in the right eye.  Six surgery.
months after implantation, the implanted lens dislocated into the A case of peri-silicone proliferation associated with retinal
vitreous cavity. Pars plana vitrectomy was performed to extract detachment involving the posterior pole is presented. Three-port
the lens. This was particularly difficult due to the size of the lens 23-gauge pars plana vitrectomy with two aspirating syringes
and the phakic status of the patient. The usage of a chandelier was performed to remove the silicone oil. Trypan blue staining
light enabled bimanual grasping and cutting the lens in the vitre- was used to visualize the epiretinal proliferation, followed by
ous cavity. The smaller lens fragments were removed via the Perfluorocarbon injection to reattach and stabilize the detached
pars plana sclerotomies. retina. All the scar tissue was peeled from the retinal surface
with forceps.
DISLOCATION OF THE TIP OF THE SOFT Excellent visualization of the peri-silicone proliferation with
trypan blue staining and careful and meticulous peeling
TIP CANNULA ensures the complete removal of all the scar tissue while
Andre Gomes MD avoiding iatrogenic retinal breaks, reducing the risk of recur-
rent PVR.
Patient underwent pars plana vitrectomy for proliferative dia-
betic retinopathy. During the injection of triamcinolone ace-
tonide into the eye (to better visualize the vitreous), the tip
MIRAGEL BUCKLE REMOVAL
of the silicone tipped cannula separated from the cannula Stanislao Rizzo MD
and was dislodged into the vitreous cavity. The surgery was
continued and the dislocated tip was left alone. Towards the An 83-year-old patient presented with a superior orbital mass
end of the surgery; however, the silicone tip moved between and a history of retinal surgery 16 years ago. The proptosis had
the lens and pars plana. This was a challenging situation increased in the past 6 months causing diplopia. It was decided
since any manipulation could lead to either cataract formation to remove the expanding Miragel buckle.
or iatrogenic peripheral breaks. Using the vitreous cutter at During surgery, the Miragel implant was gel-like, brittle, and
the suction mode, the silicone tip was re-located into the swollen. It measured 14.5x12.4x7.0 mm about two to three times
posterior pole. At this point, it was decided to remove the the original width and thickness.
cannula tip with forceps; however, the flow of the fluid inside Surgical removal was complicated by a thick, fibrous capsule,
the eye guided the cannula tip into the trochar and outside which has fixated the swollen buckle element to the tenon’s
the eye. capsule, limiting the mobility of the globe.
It is important to remember that the soft tip cannula is The implant was removed in its entirety as described
designed for aspiration of fluid rather than injection of fluid below:
into the eye. Further, always examine the tip of the soft tip After opening the conjunctiva and isolating the Miragel
cannula when exiting the eye to assure that it is not dislodged implant, it was removed by pushing it out: exerting pressure to
inside the eye. it with a blunt instrument rather than grasping it. Using sharp
forceps as in the conventional silicone buckle removal would
make the removal of the Miragel implant very difficult since
IATROGENIC PERIPHERAL RETINAL forceps would bite through it rather than grasping it leading to
BREAKS DURING IOFB EXTRACTION its disintegration.
The Miragel implant was associated with a massive periocular
Martin Charles MD
fibrosis. During the dissection of this fibrotic tissue, great care
Peripheral retinal tears are not an unusual finding in patients was given to avoid any damage to the peri-ocular muscles (blunt
with intraocular foreign body (IOFB) injury. These patients are dissection). Following the removal of the Miragel implant, the
generally young and present with an attached hyaloid. During underlying sclera was examined carefully to assure its full integ-
the removal of the foreign body the vitreous manipulation and rity and that there were not any areas of perforation. The pres-
traction may lead to peripheral tears. sure induced by the expansion of the Miragel implant may lead
It is important to perform a thorough vitrectomy prior to to scleral thinning and sclera rupture during this type of surgery.
IOFB removal. At the conclusion of the surgery the peripheral It is important to have graft or synthetic tissue available in case
retina must be examined very carefully. Any possible retinal of scleral rupture.
SUBCONJUNCTIVAL SILICONE OIL subconjunctival silicone oil, reduced globe motility, and an infe-
rior retinal detachment with a break at 6 o’clock.
REMOVAL
e6 The video demonstrates the removal of subconjunctival
Susanne Binder MD pockets of silicone oil.
When silicone is used during vitrectomy surgery, suturing the
A 48-year-old myopic patient presented with chronic pain and sclerotomies would prevent the subsequent leakage of oil that
redness in the right eye. Patient had a history of 2 sutureless can occur immediately after surgery or over time. This is spe-
vitrectomies in the past (latter surgery with silicone oil). cially the case in patients with high myopia with a thin sclera.
On exam patient had an elevated intraocular pressure,
Complications in Vitreoretinal Surgery
List of Video Contributors
Gary W. Abrams MD Carl Claes MD Nancy M. Holekamp MD
The David Barsky, M.D. Professor and Chair Head of VR Surgery Associate Clinical Professor
Department of Ophthalmology ST Augustinus Department of Ophthalmology and Visual
Wayne State University Antwerp, Belgium Science
Director, Kresge Eye Institute Washington University School of Medicine
Detroit, MI, USA Ian J. Constable FRANZCO, FRCSE, Barnes Retina Institute
DSc (Hon) St. Louis, MO, USA
Everett Ai MD Professor of Ophthalmology
Director, Ophthalmic Diagnostic Center Lions Eye Institute Jason Hsu MD
California Pacific Medical Center University of Western Australia Clinical Instructor
San Francisco, CA, USA Perth, Australia Retina Service of Wills Eye Institute
Thomas Jefferson University
J. Fernando Arevalo MD FACS Lyndon da Cruz MBBS, MA, FRCOphth, Philadelphia, PA, USA
Chief of Vitreoretinal Division PhD, FRACO
Senior Academic Consultant Consultant Ophthalmic Surgeon Mark S. Humayun MD, PhD
The King Khaled Eye Specialist Hospital Moorfields Eye Hospital Professor of Ophthalmology
Riyadh, Kingdom of Saudi Arabia Vitreo-Retinal Service Biomedical Engineering, Cell and
Adjunct Professor of Ophthalmology Wilmer London, UK Neurobiology
Eye Institute Keck School of Medicine of the University of
The Johns Hopkins University Bertil E. Damato MD, PhD, FRCOphth Southern California
Baltimore, MD, USA Honorary Professor Associate Director of Research,
Ocular Oncology Service Doheny Eye Institute
Jose Garcia Arumi MD Royal Liverpool University Hospital Los Angeles, CA, USA
Professor Liverpool, UK
Instituto Microcirugia Ocular Yasushi Ikuno MD
Universidad Autonoma Arthur D. Fu MD Associate Professor
Barcelona, Spain Clinical Professor of Ophthalmology Department of Ophthalmology
California Pacific Medical Center Osaka University Graduate School of
G. William Aylward FRCS, FRCOphth, MD San Francisco, CA, USA Medicine
Consultant Vitreoretinal Surgeon Osaka, Japan
Moorfields Eye Hospital Enrique Garcia-Valenzuela MD, PhD
London, UK Vitreo-Retinal Surgery Timothy L Jackson MB. ChB, PhD,
Midwest Retina Consultants, S.C. FRCOphth
Jean Bennett MD, PhD Clinical Assistant Professor of Ophthalmology Honorary Consultant Ophthalmic and Retinal
F.M. Kirby Professor of Ophthalmology University of Illinois Eye & Ear Infirmary Surgeon
Professor of Cell and Developmental Biology Parkside Center Department of Ophthalmology
Department of Ophthalmology Park Ridge, IL, USA King’s College Hospital
Scheie Eye Institute London, UK
University of Pennsylvania Perelman School Alain Gaudric MD
of Medicine Professor of Ophthalmology Glenn J. Jaffe MD
Research Scientist Hôpital Lariboisière Professor of Ophthalmology
Center for Cellular and Molecular Université Paris-Diderot Chief, Vitreoretinal Service
Therapeutics Paris, France Director
The Children’s Hospital of Philadelphia Duke Reading Center
Philadelphia, PA, USA Andre Vieira Gomes MD, PhD Durham, NC, USA
Colaborator Professor
Susanne Binder MD Department of Ophthalmology Robert N. Johnson MD
Professor University of Sao Paulo Clinical Professor of Ophthalmology
Department of Ophthalmology Sao Paulo, Brazil California Pacific Medical Center
Rudolph Foundation Hospital San Francisco, CA, USA
Vienna, Austria Christine R Gonzales MD
Assistant Professor of Ophthalmology J. Michael Jumper MD
Vicente Martinez Castillo MD Jules Stein Eye Institute Retina Service Chief
Department of Ophthalmology University of California Los Angeles School of California Pacific Medical Center
Vall d’Hebron Hospital Medicine San Francisco, CA, USA
Barcelona, Spain Los Angeles, CA, USA
Bernd Kirchhof MD, PhD
Stanley Chang MD Zdenek Gregor FRCS (Eng) FRCOphth Professor and Chairman
KK Tse and Ku The Ying Professor Consultant Vitreoretinal Surgeon University Eye Department Cologne
Department of Ophthalmology Moorfields Eye Hospital Koln, Germany
Columbia University London, UK
New York, NY, USA Allan E Kreiger MD
Stratos Gotzaridis MD Professor, Department of Ophthalmology
Martin Charles MD Director of Retinal Services The Jules Stein Eye Institute
Professor of Medicine Athinaiki General Hospital Los Angeles, CA, USA
Centro Oftalmologico Dr Charles Athens, Greece
Buenos Aires, Argentina Anthony Kwan
Heinrich Heimann MD Queensland Eye Institute in the Brisbane
Lawrence P. Chong MD Consultant Ophthalmic Surgeon Region,
VMR Institute St Paul’s Eye Unit Queensland Ophthalmic Surgeons
Huntington Beach, CA, USA Royal Liverpool University Hospital Brisbane, QLD, Australia
Liverpool, UK
Henry C. Lee MD Yoshitaka Nakashima MD Eduardo C de Souza MD
Barnes Retina Institute Department of Ophthalmology Ophthalmologist
Washington University School of Medicine Hospital das Clinicas Federal University of Santa Catarina
xxiv St. Louis, MO, USA University of Sao Paulo Florianopolis, Santa Catarina, Brasil
Sao Paulo, Brazil
Jennifer I. Lim MD Marcin Stopa MD PhD
List of Video
Contributors

Professor of Ophthalmology and Visual Sumit K. Nanda MD Consultant, Department of Ophthalmology


Sciences Clinical Associate Professor Poznan University of Medical Sciences
Lions of Illinois Eye Research Institute Department of Ophthalmology Poznan, Poland
University of Illinois at Chicago University of Oklahoma
Chicago, IL, USA Oklahoma City, Oklahoma, USA Paul Sullivan MBBS, MD, FRCOphth
Consultant Opthalmic Surgeon
Albert M. Maguire MD Yusuke Oshima MD, PhD Director of Education
Associate Professor of Ophthalmology Associate Professor Moorfields Eye Hospital
Department of Ophthalmology Department of Ophthalmology London, UK
Scheie Eye Institute Osaka University Graduate School of
University of Pennsylvania Perelman School Medicine Hiroko Terasaki MD, PhD
of Medicine Suita, Osaka, Japan Professor and Chair
Investigator Department of Ophthalmology
Center for Cellular and Molecular Ehab N EL Rayes MD PhD Nagoya University Graduate School of
Therapeutics Professor Medicine
Retina Specialist Retina Department Nagoya, Japan
Pediatric Ophthalmology Institute of Ophthalmology
The Children’s Hospital of Philadelphia Cairo, Egypt John T. Thompson MD
Philadelphia, PA, USA Assistant Professor
Franco M. Recchia MD The Wilmer Institute
H. Richard McDonald MD Associate Professor of Ophthalmology and Johns Hopkins University
Clinical Professor of Ophthalmology Visual Sciences Baltimore, MD, USA
California Pacific Medical Center Chief, Retina Division
Director, Vitreoretinal Fellowship Program Director, Fellowship in Vitreoretinal Diseases Cynthia A. Toth MD
California Pacific Medical Center and Surgery Professor
Director, San Francisco Retina Foundation Vanderbilt Eye Institute Department of Ophthalmology and
San Francisco, CA, USA Vanderbilt University School of Medicine Biomedical Engineering
Nashville, TN, USA Duke Universtiy
Petra Meier MD Durham, NC, USA
Assistant Professor Kourous A. Rezaei MD
Klivik und Poliklinik fur Augen Associate Professor Michael T. Trese MD
Leipzig, Germany Illinois Retina Associates, S.C Clinical Professor of Biomedical Sciences
Rush University Medical Center Eye Research Institute
Jan C. van Meurs MD PhD Harvey, IL, USA Oakland University
Professor Rochester, MI
The Rotterdam Eye Hospital and Erasmus Stanislao Rizzo MD Chief Pediatrid and Adult Vitreoretinal
University Director U.O. Chirurgia Oftalmica Surgery
Rotterdam, the Netherlands Azienda Ospedaliero Universitaria Pisana William Beaumont Hospital
Pisa, Italy Royal Oak, MI, USA
Virgilio Morales-Canton MD
Chief of the Retina Department Abdulaziz Adel Rushood MD Peter Wiedemann MD
Asociacion para Evitar la Ceguera Vitreoretinal Fellow Professor of Ophthalmology
Hospital Dr. Luis Sanchez Bulnes Department of ophthalmology Department of Ophthalmology
Mexico City King Fahad Hospital of the University, University of Leipzig Faculty of Medicine
Professor Al-Khobar, Saudi Arabia Leipzig, Germany
Universidad Nacional Autonoma de Mexico
Mexico City, Mexico Michael Samuel MD Andre J. Witkin MD
Attending Surgeon, Retina Service Assistant Professor
Prithvi Mruthyunjaya MD Wills Eye Hospital Department of Ophthalmology
Assistant Professor of Ophthalmology Thomas Jefferson University Tufts University School of Medicine
Department of Vitreoretinal Surgery and Philadelphia, PA, USA Boston, MA, USA
Ocular Oncology
Duke University Eye Center V Sivagnanavel MD Young Hee Yoon MD, PhD
Durham, NC, USA Department of Ophthalmology Professor and Chair
Kings College Hospital Department of Ophthalmology
Manish Nagpal MS, DO, FRCS(UK) University of London, Asan Medical Center
Vitreo Retinal Consultant London, UK University of Ulsan College of Medicine
Retina Foundation Seoul, South Korea
Ahmedabad, Gujarat, India Raymond N Sjaarda MD
Retina Specialist
Baltimore, MD, USA
Section I Optical Imaging Technologies
For additional online content visit http://www.expertconsult.com

Chapter Fluorescein Angiography: Basic Principles


1  and Interpretation
Robert N. Johnson, Arthur D. Fu, H. Richard McDonald, J. Michael Jumper,
Everett Ai, Emmett T. Cunningham Jr, Brandon J. Lujan

For nearly 50 years, fundus photography and fluorescein angi- absorbed and changed, is blue; the resultant fluorescence, or
ography have been valuable in expanding our knowledge of the emitted wavelength, is green-yellow. If blue light between 465
anatomy, pathology, and pathophysiology of the retina and and 490 nm is directed to unbound sodium fluorescein, it emits
choroid.1 Initially, fluorescein angiography was used primarily a light that appears green-yellow (520–530 nm).
as a laboratory and clinical research tool; only later was it used This is a fundamental principle of fluorescein angiography.
for the diagnosis of fundus diseases.1–5 An understanding of In the procedure, the patient, whose eyes have been dilated,
fluorescein angiography and the ability to interpret fluorescein is seated behind the fundus camera, on which a blue filter
angiograms are essential to accurately evaluate, diagnose, and has been placed in front of the flash. Fluorescein is then injected
treat patients with retinal vascular and macular disease. intravenously. Eighty percent of the fluorescein becomes bound
This chapter discusses the basic principles of fluorescein angi- to protein and is not available for fluorescence, but 20% remains
ography and the equipment and techniques needed to produce free in the bloodstream and is available for fluorescence. The
a high-quality angiogram. Potential side-effects and complica- blue flash of the fundus camera excites the unbound fluorescein
tions of fluorescein injection are also discussed. Finally, interpre- within the blood vessels or the fluorescein that has leaked out
tation of fluorescein angiography, including fundus anatomy of the blood vessels. The blue filter shields out (reflects or
and histology, the normal fluorescein angiogram, and conditions absorbs) all other light and allows through only the blue exci-
responsible for abnormal fundus fluorescence are described. tation light. The blue light then changes those structures in
the eye containing fluorescein to green-yellow light at 520–
BASIC PRINCIPLES 530  nm. In addition, blue light is reflected off the fundus
structures that do not contain fluorescein. The blue reflected
To understand fluorescein angiography, a knowledge of fluo-
light and the green-yellow fluorescent light are directed back
rescence is essential. Likewise, to understand fluorescence, one
toward the film of the fundus camera. Just in front of the film
must know the principles of luminescence. Luminescence is
a filter is placed that allows the green-yellow fluorescent light
the emission of light from any source other than high tem-
through but keeps out the blue reflected light. Therefore the
perature. Luminescence occurs when energy in the form of
only light that penetrates the filter is true fluorescent light
electromagnetic radiation is absorbed and then re-emitted at
(Fig. 1.1).
another frequency. When light energy is absorbed into a lumi-
nescent material, free electrons are elevated into higher energy
states. This energy is then re-emitted by spontaneous decay of
the electrons into their lower energy states. When this decay
occurs in the visible spectrum, it is called luminescence. Lumi-
100 Absorption
nescence therefore always entails a shift from a shorter wave-
Emission
length to a longer wavelength. The shorter wavelengths
represent higher energy, and the longer wavelengths represent 80
Percent transmission

lower energy.
and absorption

60
Fluorescence
Fluorescence is luminescence that is maintained only by continu-
ous excitation. In other words, excitation at one wavelength 40
occurs and is emitted immediately through a longer wavelength.
Emission stops at once when the excitation stops. Fluorescence 20
thus does not have an afterglow. A typical example of fluores-
cence is television. In the television tube, the excitation radiation
0
is the electron beam from the cathode-ray tube. This beam excites
the phosphors of the screen, which re-emit the beam as a glow 400 500 600 700
that constitutes a television picture. Wavelength (nm)
Sodium fluorescein is a hydrocarbon that responds to light
Fig. 1.1 Absorption and emission curves of sodium fluorescein dye.
energy between 465 and 490 nm and fluoresces at a wavelength The peak absorption (excitation) is at 465–490 nm (blue light). The
of 520–530 nm. The excitation wavelength, the type that is peak emission occurs at 520–530 nm (yellow-green light).
100 Exciter filter
3
Barrier filter
80 Pseudofluorescence
Percent transmission

Chapter 1
and absorption

60

40

Fluorescein Angiography: Basic Principles and Interpretation


20

0
400 500 600 700
Wavelength (nm)

Fig. 1.2 Pseudofluorescence. The blue exciter filter overlaps into the
yellow-green zone, and the yellow-green barrier filter overlaps into the
blue zone. The combination results in pseudofluorescence.

Pseudofluorescence
Pseudofluorescence occurs when nonfluorescent light passes
through the entire filter system. If green-yellow light penetrates
the original blue filter, it will pass through the entire system. If
blue light reflected from nonfluorescent fundus structures pen-
etrates the green-yellow filter, pseudofluorescence occurs (Fig.
1.2). Pseudofluorescence (i.e., fake fluorescence) causes nonfluo-
rescent structures to appear fluorescent. It can confuse the physi-
cian interpreting the fluorescein angiogram and lead him or her
to think that certain fundus structures or materials are fluoresc- Fig. 1.3 Digital fundus camera for color fundus and fluorescein
ing when they are not. Pseudofluorescence also causes decreased angiography.
contrast, as well as decreased resolution. Because fluorescein
angiography uses black-and-white film, the nonfluorescent or
pseudofluorescent light appears as a background illumination.
The background illumination from pseudofluorescence is espe- Box 1.1 Equipment and materials needed for angiography
cially heightened if there are white areas of the fundus, such as Fundus camera and auxiliary equipment
highly reflective, hard exudates. Pseudofluorescence must be Matched fluorescein filters (barrier and exciter)
avoided. Therefore the excitation (blue) and barrier (green- Digital photoprocessing unit (computer-based) and software user
interface
yellow) filters should be carefully matched so that the overlap 23-gauge scalp vein needle
of light between them is minimal. 5 mL syringe
5 mL of l0% fluorescein solution
EQUIPMENT 20-gauge, 1 12 -inch needle to draw the dye
Armrest for fluorescein injection
Film-based versus digital fluorescein Tourniquet
Alcohol swabs
angiography – historical perspectives Bandage
Standard emergency equipment
Fluorescein angiography finds its origins in the late 1960s with
the publication of an original article describing its use as well as
subsequent atlases and textbooks for a medical retinal specialty
in its infancy.1,6 The landmark text Atlas of Macular Diseases by
Dr J. Donald Gass set a new standard for the use of stereoscopic images is also time-consuming compared with digital images
fluorescein angiography in fundus diagnosis.7 (Box 1.1).8
As digital photography has evolved with improved resolu-
tion, the convenience of digital-based fluorescein angiography Camera and auxiliary equipment
has gained wider acceptance. Though film-based images Cameras differ in the degree of fundus area included in the
offer the highest amounts of resolution and 35-mm negatives photographs. Fundus cameras may range from 35° to 200° wide-
are often easier to view for stereo, images are relatively dif- field camera systems such as Optos.9,10 In clinical retinal practice,
ficult to manipulate, and training and effort are required to cameras ranging from 35° to 50° are routinely used (Fig. 1.3).
process and duplicate film. Transmitting or sharing film-based Regardless of range, a camera with the ability to yield high
4
Section I

A B C
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.4 Optos wide-field images. (A) Nonperfusion detected in the left eye with wide-field fluorescein imaging. (Courtesy of Umar Mian, MD.
Image taken by Carolina Costa.) (B, C) Wide-field angiography of the right and left eyes of a patient with diabetic retinopathy. Note the multiple
areas of leakage corresponding to areas of retinal neovascularization associated with capillary nonperfusion. It is often difficult in certain
wide-field images to determine the presence of small neovascular complexes versus leakage from capillary nonperfusion, unless areas are
magnified further. (Courtesy of Szilárd Kiss, MD.)

resolutions of the posterior pole is essential for most macular


problems, especially when laser treatment is to be done, as with
background diabetic retinopathy, branch-vein occlusion, or cho-
roidal neovascularization.
Wide-angle angiography has the benefit of capturing a single
image of the retina in high resolution well beyond the equator.
The potential for clinical efficiency and sensitivity in detecting
neovascularization in the far periphery as well as acquiring an
excellent clinical picture of the degree of capillary retinal non-
perfusion is an exciting development in fluorescein angiography
(Fig. 1.4).
Stereophotography
A subtle joystick movement by a trained photographer from
right to left photographing from one side of the pupil to the
opposite side of the pupil produces a stereoscopic image when
combined through hand-held stereo viewers. It is important to
note that, in the process of acquiring images for stereophotogra- Fig. 1.5 Fundus film camera with side wall removed to view inside
phy, individual images may be blurred or may contain artifacts. parts. Yellow arrow, viewing bulb; black arrow, flash bulb; red arrow,
exciter filter wheel. The light from the flash goes through the system in
The photographer should not attempt to discard images until
this photograph from right to left. The light is reflected off a mirror and
viewed with the stereo viewers. travels upward to another mirror; it is then reflected to the left, into the
patient’s eye. From there it is reflected directly to the right of the
Matched fluorescein filters fluorescein camera back (white arrow).
Typically included in modern camera units, fluorescein angiog-
raphy uses both exciter and barrier filters. The exciter filter must
transmit blue light at 465–490 nm, the absorption peak of fluo-
rescein excitation. The barrier filter transmits light at 525–530 nm, Fluorescein solution
the fluorescent, or emitted, peak of fluorescein. The filters should
Sodium fluorescein, an orange-red crystalline hydrocarbon
allow maximal transmission of light in the proper spectral range
(C20H12O5Na), has a low molecular weight (376.27 Da) and
to achieve a good image without the use of an excessively pow-
readily diffuses through most of the body fluids and through
erful flash unit. Most new cameras come with filters. When
the choriocapillaris, but it does not diffuse through the retinal
choosing a camera, one should request the transmission curves
vascular endothelium or the pigment epithelium.
of the filter combination to be sure that no significant overlap
Solutions containing 500 mg fluorescein are available in vials
exists; pseudofluorescence results when there is overlap.
of 10 mL of 5% fluorescein or 5 mL of 10% fluorescein. Also
After several years, filters become thin, emitting more light
available are 3 mL of 25% fluorescein solution (750 mg). The
and increasing the incidence and degree of pseudofluorescence.
greater the volume, the longer the injection time will be; the
The clinician should always check the control photograph of
smaller the volume, the more likely a significant percentage of
each angiogram for excessive pseudofluorescence. Filters should
fluorescein will remain in the venous dead space between the
be replaced once pseudofluorescence reduces the quality of the
arm and the heart (see Injecting the fluorescein, below). For this
angiophotograph.
reason we prefer 5 mL of 10% solution (500 mg fluorescein).
Light sources (viewing bulb and flash strobe) Fluorescein is eliminated by the liver and kidneys within 24
In film-based cameras, bulbs burn out, but are easily replaced. hours, although traces may be found in the body for up to a week
A supply of each should always be kept on hand (Fig. 1.5). after injection. Retention may increase if renal function is
impaired. The skin has a yellowish tinge for a few hours after Nausea is the most frequent side-effect of fluorescein injection,
injection, and the urine has a characteristic yellow-orange color occurring in about 5% of patients. It is most likely to occur in
for most of the first day after injection. patients under 50 years of age or when fluorescein is injected 5
Various side-effects and complications can occur with rapidly. When nausea occurs, it usually begins approximately 30
fluorescein injection (Box 1.2).11–15 seconds after injection, lasts for 2–3 minutes, and disappears

Chapter 1
A serious complication of the injection is extravasation of the slowly.
fluorescein under the skin. This can be extremely painful and Vomiting occurs infrequently, affecting only 0.3–0.4% of
may result in a number of uncomfortable symptoms. Necrosis patients.11,13 When it does occur, it usually begins 40–50 seconds
and sloughing of the skin may occur, although this is extremely after injection. By this time most of the initial-transit photo-
rare. Superficial phlebitis also has been noted. A subcutaneous graphs of the angiogram will have been taken. A receptacle and
granuloma has occurred in a few patients after fluorescein tissues should be available in case vomiting does occur. When
extravasation. In each instance, however, the granuloma has patients experience nausea or vomiting, they must be reassured

Fluorescein Angiography: Basic Principles and Interpretation


been small, cosmetically invisible, and painless. Toxic neuritis that the unpleasant and uncomfortable feeling will subside
caused by infiltration of extravasated fluorescein along a nerve rapidly. Photographs can be taken after the vomiting episode
in the antecubital area can result in considerable pain for up to has passed. A slower, more gradual injection may help to prevent
a few hours. The application of an ice pack at the site of extrava- vomiting.
sation may help relieve pain. For extremely painful reactions an Patients who have previously experienced nausea or vomiting
injection of a local anesthetic at the site of extravasation is effec- from fluorescein injection may be given an oral dose of 25–50 mg
tive but rarely necessary. of promethazine hydrochloride (Phenergan) by mouth approxi-
If extravasation occurs, the physician must decide whether to mately 1 hour before injection. Promethazine has proved to be
continue angiography. Extravasation may occur immediately, helpful in preventing or lessening the severity of nausea or vom-
and thus the serum concentration of the fluorescein will be insuf- iting. We have recently found that we can also reduce the inci-
ficient for angiography. In this case it usually is best to place the dence of nausea by warming the vial of fluorescein to body
needle in another vein and reinject a full dose of fluorescein, temperature and drawing it into the syringe through a needle
starting the process again from the beginning. Occasionally, only with a Millipore filter. Restriction of food and water for 4 hours
a small amount of fluorescein is extravasated at the end of the before the fluorescein injection may reduce the incidence of vom-
injection. In this case photography can continue without stop- iting; an empty stomach may prevent vomiting but will not
ping or reinjecting. affect nausea. If the patient still has a tendency to vomit despite
A common cause of extravasation is the use of a large, long taking all these measures, a lesser amount of fluorescein can be
needle directly attached to a syringe. It is difficult to hold the given and injected more slowly if the photographic results will
syringe in the dark. For this and other reasons we have discussed not be compromised.
earlier, a scalp-vein needle attached to a syringe by a flexible Vasovagal attacks occur much less frequently during fluores-
tube is the best choice for this procedure. Also, the patient’s own cein angiography than does nausea and are probably caused
blood can be drawn back into the tubing of the scalp-vein needle, more by patient anxiety than by the actual injection of fluores-
with the blood going all the way up to but not into the syringe. cein. We have seen vasovagal attacks even when the patient sees
When it is time to inject, the person giving the injection can look the needle or immediately after the skin has been penetrated by
at the tip of the needle to ensure that extravasation has not the needle but before the injection has begun. Occasionally a
occurred. If it has, the patient’s own blood is extravasated, and vasovagal reaction causes a patient to faint, but consciousness is
little chance of complication exists if the injection is stopped at regained within a few minutes. If early symptoms of a vasovagal
this point so that no fluorescein is injected. episode are noted, smelling salts usually reverse the reaction.
It is always important to watch for extravasation at the begin- The photographer must be alert for signs of fainting because the
ning of the injection so that, should it occur, the process can be patient could be injured if he or she falls.
halted; thus only a minimal amount of fluorescein will have been Shock and syncope (more severe vasovagal reaction) consist
injected and extravasated. The amount of extravasated fluores- of bradycardia, hypotension, reduced cardiovascular perfusion,
cein can be minimized by slow injection and constant observa- sweating, and the sense of feeling cold. If the photographer and
tion of the needle with a hand-held light or if injection is done person injecting see that the patient is getting “shocky” or light-
before turning off the room lights. headed, the patient should be allowed to bend over or lie down
with the feet elevated. The patient’s blood pressure and pulse
should be carefully monitored. It is important to differentiate
Box 1.2 Side-effects and complications of fluorescein injection this from anaphylaxis, in which hypotension, tachycardia, bron-
chospasm, hives, and itching occur.
Extravasation and local-tissue necrosis13
Inadvertent arterial injection Hives and itching are the most frequent allergic reactions,
Nausea11–13 occurring 2–15 minutes after fluorescein injection. Although
Vomiting hives usually disappear within a few hours, an antihistamine,
Vasovagal reaction (circulatory shock, myocardial infarction)12
Allergic reaction, anaphylaxis (hives and itching, respiratory such as diphenhydramine hydrochloride (Benadryl), may be
problems, laryngeal edema, bronchospasm) administered intravenously for an immediate response. Bron-
Nerve palsy chospasm and even anaphylaxis are other reactions that have
Neurologic problems (tonic-clonic seizures) been reported, but these are extremely rare. Epinephrine, sys-
Thrombophlebitis
Pyrexia temic steroids, aminophylline, and pressor agents should be
Death available to treat bronchospasm or any other allergic or ana-
phylactic reactions. Other equipment that should be readily
available in the event of a severe vasovagal or anaphylactic Occasionally, a patient has a peculiar corneal reflex or central
reaction includes oxygen, a sphygmomanometer, a stethoscope, lens opacity, and it may be impossible to follow the usual pro-
6 and a device to provide an airway. The skilled photographer cedure of aligning the camera through the central axis of the eye.
observes each patient carefully and is alert to any scratching, Moving the camera slightly off axis may help improve focus and
wheezing, or difficulty in breathing that the patient may have resolution.
after injection. Any abnormalities, such as an unusual light reflex or a poorly
Section I

There are a few published and unpublished reports of death resolved image the photographer sees through the camera
following intravenous fluorescein injection. The mechanism may system, will appear on the photograph. If the ophthalmoscopic
be a severe allergic reaction or a hypotensive episode induced view seen through the camera is not optimal, the photograph
by a vasovagal reaction in a patient with pre-existing cardiac or will not be optimal (Fig. 1.8). If the view is optimal, well aligned,
cerebral vascular disease. The cause of death in each case may in focus, and without reflexes, the photograph can be optimal.
have been coincidental. Acute pulmonary edema following fluo- A helpful concept for the photographer is “what you see is what
Optical Imaging Technologies
Retinal Imaging and Diagnostics

rescein injection has also been reported. you get (or worse – never better).”
There are no known contraindications to fluorescein injections
in patients with a history of heart disease, cardiac arrhythmia, Focusing
or cardiac pacemakers. Although there have been no reports of Achieving perfect focus is a major factor in the photographic
fetal complications from fluorescein injection during pregnancy, process. Both the eyepiece crosshairs and the fundus details
it is current practice to avoid angiography in women who are must be in sharp focus to obtain a well-resolved photograph.
pregnant, especially those in the first trimester. The proper position of the eyepiece is determined by the refrac-
tive error of the photographer and the degree to which he or she
TECHNIQUE accommodates while focusing the camera.
The photographer first turns the eyepiece counterclockwise
Aligning camera and photographing (toward the plus, or hyperopic, range) to relax his or her own
To align the fundus camera properly, the photographer must accommodation; this causes the crosshairs to blur. The photog-
first assess the “field of the eye.” The camera is equipped with rapher then turns the eyepiece slowly clockwise to bring the
a joystick with which the photographer can adjust the camera crosshairs into sharp focus. The eyepiece is focused properly
laterally and for depth. The camera is also equipped with a knob when the crosshairs appear sharp and clear (Fig. 1.9, online).
for vertical adjustment. The photographer finds the red fundus They must remain perfectly clear while the photographer focuses
reflex, which is an even, round, sharply defined, pink or red light on the fundus with the camera’s focusing detail. With experi-
reflex. If the camera is too close to the eye, a bright, crescent- ence, the photographer becomes expert in adjusting the eyepiece
shaped light reflex appears at the edge of the viewing screen or and in keeping the crosshairs in focus throughout the entire
a bright spot appears at its center. If the camera is too far away, photographic sequence.
a hazy, poorly contrasted photograph results. The best position for the eyepiece is the point at which the
The photographer moves the camera from side to side to ascer- crosshairs are in focus while the photographer’s accommodation
tain the width of the pupil and the focusing peculiarities of the is relaxed. Photographers learn to relax accommodation by
particular cornea and lens. The photographer studies the eye keeping both eyes open. The photographer focuses the eyepiece
through the camera lens, moving the camera back and forth and with one eye and, with the other eye, keeps a distant object, such
up and down, looking for fundus details (e.g., retinal blood as the eye chart, in sharp focus. This skill may be difficult for
vessels). The photographer then determines the single best posi- technicians without ophthalmic training, but it is seldom impos-
tion from which to photograph (Figs 1.6 and 1.7). sible to learn.

Fig. 1.6 The patient’s arm rests on an adjustable armrest that is Fig. 1.7 The patient’s head is kept steady in the chinrest and headrest of
elevated so that the patient’s arm is at or above the level of her heart. the fundus camera. The photographer aligns the camera and focuses on
The armrest also facilitates easy placement of the intravenous needle the patient’s right fundus. Each is in a comfortable position, facilitating the
and injection of fluorescein. stability necessary to achieve a good fluorescein angiogram.
6.e1

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
Fig. 1.9, online The photographer focuses the eyepiece of the camera
by initially turning the eyepiece counterclockwise, then clockwise, and
stopping when it is in exact focus. The photographer must be sure that
the eyepiece crosshairs remain in perfect focus throughout the
photographic procedure.
7

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

C D

Fig. 1.8 Fundus photograph and reflexes. (A) Photograph of right fundus without reflexes. The camera was properly aligned and focused.
(B) Note the bright red, yellow and blue arc on the right side. The flash is reflecting off the iris. This can be remedied by repositioning the
camera slightly to the right or left. (C) In this case the camera was placed at the proper distance from the fundus but was placed too far to one
side (down and to the right), which allowed the bright white arc reflex to the lower right. (D) Note white reflex, especially above, in, and below the
papillomacular bundle. In this case the camera was in proper alignment but was placed too far away from the patient’s eye.

Keeping the crosshairs in sharp focus, the photographer then Film-based images contain 10 000 lines of resolution, in contrast
turns the focusing dial on the camera to focus the fundus detail. to digital imaging, which may have as little as 1000 lines of reso-
Some photographers focus the crosshairs just once at the begin- lution.8 However, some argue that, despite higher resolution in
ning of each day and control their accommodation throughout film, the greater ability to magnify digital images makes the
the day. This is not a good idea because the photographer’s disadvantage of digital photography less clinically relevant.8
accommodation may change during a photographic session; the Digital angiography offers advantages, including the instanta-
photographer should be aware of this possibility and regularly neous availability of the angiogram, and the avoidance of the
check and readjust the eyepiece for focus. With the camera prop- equipment and time necessary to develop film. With instanta-
erly aligned and focused, the photographer is ready to start the neous images, digital angiography facilitates education and dis-
preliminary photographs and angiograms. cussion concerning the patient’s condition and treatment options.
Also, digital angiography facilitates training of ophthalmic per-
Digital angiography sonnel. We have found that it is useful to stay in the room during
In theory, film-based photography has advantages over the initial frames of the angiogram to ensure that the desired
digital imaging: image resolution and stereoscopic viewing.8 pathology is photographed. Any changes can be promptly made,
and the photographer can also learn from this prompt feedback. Photographing the periphery
Digital angiography, however, necessitates an ongoing invest-
Photographing the peripheral retina with a standard 50° fundus
8 ment of money both in software updates and storage of digital
camera demands precision and skills acquired only after many
electronic files. Also, excessive image manipulation with image-
hours of practice. Problems with patient position and camera
editing software may result in artifacts. Specifically, some areas
alignment and focus are compounded by marginal corneal astig-
may appear overly hyperfluorescent due to limited dynamic
Section I

matism, unsteadiness of patient fixation, light reflexes, and


range in images and software manipulation. Care should be
awkward camera placement. All steps necessary for taking pos-
taken in avoiding misinterpretation of hyperfluorecence and
terior photographs, such as alignment and focusing, must be
hypofluorescence in digital images.
employed to achieve good peripheral fundus photography. The
Using stereophotography Zeiss camera comes with an astigmatic dial to help neutralize
Stereophotography separates, photographically, the tissues of the induced astigmatism. A tilt mechanism, now standard on
Optical Imaging Technologies
Retinal Imaging and Diagnostics

the eye for the observer. Stereo fluorescein angiography facili- most cameras, helps position the camera for extreme superior
tates interpretation by separating in depth the retinal and cho- and inferior peripheral photography (Fig. 1.11).
roidal circulation.16,17 Stereo angiography is considered absolutely During photography of the periphery, the patient tends to turn
essential in certain situations.18 The photographic protocol for or move his or her head. Unsatisfactory photographs caused by
the Macular Photocoagulation Study required stereo fluorescein the movement of the head away from the camera or to the side
angiography. Without well-resolved stereo images, interpreta- can be avoided if the photographer is alert to these possibilities
tion of angiograms with, for instance, choroidal neovasculariza- and takes the necessary steps to prevent them. On the whole,
tion associated with age-related macular degeneration, can be achieving good peripheral photographs depends on photo-
extremely difficult, if not impossible (Fig. 1.10, online). On the graphic skill, of course, but also on patience on the part of both
other hand, stereophotography, although extremely helpful in photographer and patient.
cases that are difficult to interpret, is not always absolutely nec-
essary because other fundus features and characteristics usually Informing the patient
indicate the level at which abnormal fluorescence is located. An important step toward a successful angiogram is to inform
Adequate stereophotographs can be achieved with a pupillary the patient about the procedure. An informed patient is gener-
dilation of 4 mm, although dilation of 6 mm or more is best. The ally less anxious and more cooperative than one who is unsure
first photograph of any stereo pair is taken with the camera of the situation. Some institutions routinely provide a consent
positioned as far to the photographer’s right (the patient’s left) form to be signed by patients who are to have angiograms.
of the pupil’s center as possible (of course, without inducing However, this practice cannot replace the duty of the physician
reflexes). The second photograph of the stereo pair is taken with to inform the patient about the procedure and its potential com-
the camera held as far to the photographer’s left (the patient’s plications and to answer all questions.
right) of the pupil’s center as possible. This order is extremely The patient should be told that the eyes will be dilated, sodium
important because the photographs are taken and positioned on fluorescein will be injected in a vein in the arm or back of the
the film so that the angiogram is read from right to left. Thus the hand, and photographs will be taken. The patient should be
first photograph in the stereo sequence appears on the right on assured that the flash is a harmless, bright light (not an X-ray)
the contact sheet to correspond with the interpreter’s right eye; and that fluorescein dye is safe. The patient should be told that
the second is printed on the left for his or her left eye. It follows, injection of the dye can cause complications but that such occur-
then, that the first view of a stereo pair should be taken from the rences are rare. If the patient requests further details about com-
photographer’s right, followed by a view from the left. plications, the physician is obligated to supply the information.

A B

Fig. 1.11 Photographing the periphery. (A) The tilt mechanism of the camera allows the back of the camera to be lifted up (tilted to aim
downward) for photography of the inferior periphery. The same tilt mechanism can be used to bring the camera far down (tilted to aim upward)
to take pictures of the superior periphery (B). In photographing the inferior periphery, the photographer must sometimes stand. This photograph
was a mock situation. In a real situation, the photographer or an assistant would have to lift the patient’s upper lid to view the inferior periphery
properly.
8.e1

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

Fig. 1.10, online Viewing stereo fundus photographs. (A) Negatives are placed on a viewing back-lit display. Two negative images are then
viewed with an adjustable stereo lens. Reading the contact print of the angiogram. The stereo viewer can be easily made up using a trial frame.
In viewing negative images, “hyperfluorescence” corresponds to dark objects while corresponding “hypofluorescent” objects are lighter. (B)
Reading a stereo pair of digital angiograms. The special viewer allows the observer to focus both images. The software displayed is Ophthalmic
Imaging Systems.
Positioning the patient In this way, the time from the beginning of injection is recorded
on each subsequent angiographic photograph. When the injec-
Before the patient is seated at the camera, the photographer
makes sure that the front lens is free of any dirt or dust. The lens
tion is finished, the photographer may take another picture, 9
which shows how long the injection took.
should always be covered by a lens cap when the camera is not
A rapid injection of 2 or 3 seconds delivers a high concentra-
in use. The front of the lens should be kept clean using chloro-

Chapter 1
tion of fluorescein to the bloodstream for a short time and prob-
form and a tightly rolled rod of lens tissue. To clean the lens,
ably yields somewhat better photographs than does a slower
begin at the center and rotate out to the periphery.
injection. However, the more rapid the injection, the greater the
The patient is positioned at the camera with the chin in the
incidence of nausea from a highly concentrated bolus of fluores-
chinrest and the forehead against the head bar. Because the most
cein. For this reason a slower injection (4–6 seconds) is prefera-
common cause of poor fluorescein photographs is involuntary
ble; the photographs will still be of good quality. Because some
movement of the patient’s head, the photographer should
fluorescein dye remains in the tubing, the scalp-vein needle

Fluorescein Angiography: Basic Principles and Interpretation


prepare and make adjustment for this before the fluorescein is
should have short, rather than long, tubing to ensure that more
injected. The photographer should aim and focus the camera on
of the dye is injected.
the specific area of primary interest, at the same time noting the
patient’s responses. If the photographer finds that the camera Developing a photographic plan
must continually be moved closer to the patient while aligning
To photograph and print the fluorescein angiogram, we suggest
it or taking preliminary photographs, or if reflexes suddenly
the following comprehensive plan, designed to yield maximal
appear in the view even though the camera is steady, then
angiographic information from each fundus and to facilitate a
the patient’s head has moved away from the chinrest. If so, the
thorough and complete interpretation (Fig. 1.14). In contrast to
photographer can make some adjustment before injecting the
film-based angiograms which required multiple duplicate
fluorescein dye. Sometimes having an assistant hold the patient’s
attempts to assure at least one image would be optimal, fewer
head in the chinrest is helpful (Fig. 1.12, online). The photogra-
photographs are necessary in digital angiography. Although
pher either may lower the entire camera and chinrest or raise the
most angiograms will be complete by following this procedure,
patient’s chair. This causes the patient to lean forward in the
there will be exceptions. This plan must be modified if abnor-
chinrest and against the forehead bar, making it more difficult
malities occur in areas other than the macula and disc.
for the patient to pull back.
The photographic strategy essentially begins when the clini-
Before photography begins, and between shots, the photogra-
cian has identified a condition or finding that requires angio-
pher may ask the patient to blink several times. This usually
graphic study. The pathology dictates whether the photographic
makes the patient more comfortable and also moistens the
approach should image a magnified highly detailed finding
cornea and keeps it clear. When the pictures are actually being
versus a wider field of view for a more diffuse disease. Narrower
shot, the patient should be instructed to blink as infrequently as
field limits with higher magnification yield optimal images for
possible.
focal pathology in conditions such as maculopathies, optic nerve
The photographer should talk to the patient frequently during
disorders, and small focal lesions. Wider field of view often
the procedure, informing the patient of the progress of the
sacrifices magnification, but is effective in documenting condi-
testing and assuring him or her that all is going well. Explana-
tions involving the periphery, such as diabetic retinopathy and
tion and reinforcement help produce better photographs.
vascular occlusive disease. Peripheral retinal scans for areas of
neovascularization, as well as consideration for images that can
Injecting the fluorescein be later montaged for wide-field reproductions, must also be
The color stereoscopic fundus photographs are taken first, before incorporated into certain photographic plans. Elevated lesions
the fluorescein is injected. For injection, we recommend a syringe such as tumors require great care in capturing high-quality
with a 23-gauge scalp-vein needle (Fig. 1.13, online). The scalp- stereo images.
vein needle has several advantages: it is small enough to enter It is both essential and extremely cost-effective for the physi-
most visible veins, and an intravenous opening is then available cian to indicate specifically what areas to photograph. The pho-
in the event of an emergency. Once in the vein, it requires no tographer should be directed as to where to start the angiogram
further attention, and although it can be taped in place, this and the issues important for each specific angiogram. It is most
usually is unnecessary. Whenever an antecubital vein is not efficient to use a photographic instruction slip that indicates the
visible or accessible, the vein in the back of the hand or radial specific number of color photographs to take of each area, where
(thumb) side of the wrist can usually be used for injection. Inject- to start the angiogram, what the diagnosis is, and any other
ing the fluorescein into a hand or wrist vein increases the circula- information about the patient or fundus that is pertinent to the
tion time by a few seconds, but this seldom makes any photographic process (Fig. 1.15, online). Although digital color
difference. and angiograms avoid the issue of wasted film and developing
Injection of the fluorescein is coordinated with the photo- costs, unnecessary computer storage of images and patient
graphic process and is done after the first photographs (color inconvenience can be avoided with good technique and a repeat-
fundus and control photographs; see next section) have been able, accurate algorithm for angiography.
taken. With the needle in place, angiography can begin. By a Historically, because the roll of 35-mm negative film used for
predetermined, preferably silent, signal (such as a nod of the fluorescein angiography has 36 frames, it was convenient to
head), the photographer indicates to the physician to begin think of the photograph session in terms of six rows of six frames
injecting fluorescein. The photographer starts the timer on the each. Thus frame 1 appears in the upper right-hand corner
camera simultaneously with the start of injection and takes one and frame 36 in the lower left-hand corner. The angiogram
photograph. This frame will show zero time on the photograph. developed from 36-mm negatives thus reads from right to left
9.e1

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
Fig. 1.13, online Ten percent fluorescein solution, 5 mL syringe, and
23-gauge scalp-vein needle.

Fig. 1.12, online An assistant holds the patient’s head as a reminder


to the patient to keep the chin in the chinrest and forehead against the
bar.
Fig. 1.15, online Photographic request form.
In the top left portion of the form, the
physician indicates the number of color
9.e2 photographs required for each area in
the fundus. The physician also indicates
the diagnosis because experienced
PATIENT STICKER HERE photographers will know which type of
Section I

photographs to take for each particular


diagnosis. The physician also indicates in
ZEISS CANON TOPCON ICG FA which location of the fundus the initial-transit
phase of the angiogram should take place or,
DX: START: in other words, where the photographer
RIGHT EYE: LEFT EYE: should start the angiogram. In the lower right
portion of the photographic request form, the
MAC DISC MAC DISC
physician can indicate to the photographer
Optical Imaging Technologies
Retinal Imaging and Diagnostics

PMB OTHER PMB OTHER specifics about the patient that will facilitate
COLOR SCAN FA SCAN COLOR SCAN FA SCAN the photographic process and save the
photographer time. The physician can
indicate whether an eye can fixate on a light,
whether the media are clear (so that if the
photographer cannot get a clear view, he or
she can immediately understand that it is
caused by a problem of the eye), and what
the patient’s refraction is so that the
photographer can know which special lenses
to use in the photographic process.
STUDY: VISIT: UTZ: OCT: OD
AFTER OS

Patient is staying for results

COPY OF COLOR/FA TO REFERRING MD

Notes:

DATE: FA REPORT:

Laser: R/L Type: Informed Pt:

Return in: Where: Appt. Date:

Return Ref MD: Ref Low Vision:

I understand I will be called with my test results. If I'm unavailable, my test results
will be released to:

Family Member/Spouse Answering Machine Other

Signature
Inject fluorescein, Pre-injection Pre-injection Left eye macula Right eye macula
10 when injection photograph with photograph with
ends, begin angio- fluorescein filters fluorescein filters
photography of secondary macula primary macula
primary macula
Section I

Youth: 10 sec
Elderly 12 sec

Stereo pair 1–2 sec later 1–2 sec later 1–2 sec later 1–2 sec later 1–2 sec later
Secondary macula primary macula primary macula primary macula primary macula primary macula
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Stereo pair Stereo pair Stereo pair Stereo pair Stereo pair Stereo pair
Secondary macula Secondary macula Primary disc Primary disc Primary macula Primary macula

Patient rest period Angiophotograph Post venous filling Post venous filling Stereo pair Stereo pair
unless peripheral other areas of Secondary macula Primary macula Secondary disc Secondary disc
scans indicated importance in and disc and disc
either eye
according to
fluorescein
angioscopy or
nature of case

Late Late secondary Late secondary Late primary disc Late primary
angiophotographs disc macula macula
of other areas of
importance

Fig. 1.14 Photographic plan for fluorescein angiography of macular disease. Film-based images printed upside down because the fundus camera
inverts the image of the fundus, and, to read the angiogram upright, the film is printed with the frame numbers upside down. In digital
photography, no inversion is required.

and from top to bottom. With the advent of digital imaging, photographer begins the initial “injection” image. When the
theoretically, an unlimited number of frames can be acquired. injecting clinician has completed infusion, he or she announces
However, to maximize efficiency of resources, digital storage of “injection complete” and the photographer takes the “end-of-
20 frames per digital proof sheet is typically more than adequate injection” image. Because it is important to observe the site of
for most clinical scenarios. the needle tip for extravasation of fluorescein, the lights are
The first frame of the angiographic series is the color turned off only at the end of the injection. An alternative method
photograph of each eye. Then, a preinjection “control” photo- is to turn the lights off after the needle has been inserted in the
graph checks the dual-filter system for autofluorescence and vein. The person injecting can hold a hand light to observe the
pseudofluorescence. fluorescein flow into the vein to be sure extravasation is not
At this point the fluorescein injection is begun. The needle is occurring. With the lights off, the photographer can become
inserted in a vein in the patient’s arm (Fig. 1.16, online). The dark-adapted, which allows him or her to be better able to see
photographer waits for confirmation of successful venous access the flow of fluorescein into the fundus as it occurs.
and awaits verbal confirmation that infusion is about to begin. So as not to miss the appearance of fluorescein as it enters the
Once the injecting clinician starts the infusion of fluorescein, the fundus, the photographer should begin taking the initial-transit
10.e1

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
Fig. 1.16, online After the needle is placed in the vein, the lights can
be turned off so that the photographer can become dark-adapted and
see fluorescein flow in the eye. With the use of a hand light, the
person injecting can carefully observe the injection site so as to be
sure extravasation is not occurring. In this way the fluorescein solution
can be injected while the room lights are out.
fluorescein photographs 8 seconds after the beginning of the Box 1.3 Checklist for fluorescein angiography
injection of the dye if the patient is young and 12 seconds after
injection for older patients. This is done so that these early pho- • Inform patient about fluorescein angiography. Obtain written 11
informed consent
tographs will not miss the appearance of fluorescein as it enters • Dilate patient’s pupil if necessary
the fundus. Then, at intervals of 1.5–2 seconds, approximately • Prepare fluorescein solution, scalp-vein needle, and syringe

Chapter 1
six photographs should be taken in succession. • Prepare fundus camera
If the photographer does not see fluorescein entering and • Clean front lens
• Focus eyepiece crosshairs
filling the retinal vessels while the six initial-transit photographs • Input patient identification and demographic data in computer
are taken, he or she must continue to photograph the fundus database
until filling takes place and also should check to see why no • Position patient for alignment, focus, and comfort
• Align and focus camera
fluorescein is present. • Complete color photography
After the first six initial-transit photographs and approxi-

Fluorescein Angiography: Basic Principles and Interpretation


• Take red-free photos if indicated
mately 20–30 seconds after injection, with sufficient fluorescein • Insert scalp-vein needle
concentration in the eye, the photographer should take a photo- • Simultaneously start timer from zero and inject fluorescein dye
• Take preinjection photograph: these serve as controls in
graph of the fellow eye, a stereoscopic pair of photographs of detecting auto- and pseudofluorescence
the primary area of interest, followed by a stereoscopic pair of • Shoot at exact start of injection as timer is turned on and shoot
other pertinent areas. For example, in the suggested photo- second shot at exact finish of injection (length of time of injection
is automatically recorded)
graphic plan, after stereophotographs of the right macula are
• Start fluorescein photograph 8 seconds after the start of injection
taken, stereophotographs of the right disc are taken. The pho- in young patients and 12 seconds in older patients
tographer should then photograph in stereo the macula and disc • Follow fluorescein angiography plan
of the fellow eye. • When photography is done, reassure patient that urine will be
discolored for a day or so
Late-stage angiographs, preferably in stereo, are taken of the • At the discretion of the physician, have patient wait for
pertinent areas of each eye. It is important to photograph both observation for possible reactions to fluorescein
discs and macula and any other areas of abnormal fluorescence
and to note any areas that could not be photographed. This
ensures that the interpreter will have adequate information for
a complete interpretation of the angiogram.
The entire photographic process lasts 5–10 minutes. Angio- superior, temporal, and inferior quadrants, respectively, of the
photographs taken more than 10 minutes after injection are primary eye are studied. For the purpose of orientation, inter-
usually not necessary. In some cases of central serous chorioreti- pretation, and uniformity, the nasal, superior, and inferior
nopathy, or other rare situations, angiophotographs taken longer photographs are taken with the edge of the disc at the edge of
than 10 minutes after injection are helpful. This photographic the photograph and the temporal photograph with the fovea
algorithm is modified for specific conditions. For instance, in an at the edge of the photograph. A similar sequence is then
angiogram of a diabetic patient, peripheral scans may be included performed on the secondary eye.
at the request of the clinician surveying for neovascularization.
In a patient with possible choroidal neovascularization due to INTERPRETATION
age-related macular degeneration, additional angiophotographs
of the suspicious lesion may be useful. Fundus anatomy and histology
At the end of the session the patient is asked regarding any Fluorescein angiography has greatly increased our knowledge
sensations related to an allergic reaction and reminded that the of retinal and choroidal circulatory physiology and fundus
urine will be discolored for about a day. pathology. This clinical and research tool facilitates the in vivo
In the event of a technical difficulty, such as camera break- study of histopathologic characteristics of fundus disease. Before
down, repeat fluorescein injection or photography can be carried the advent of fluorescein angiography, conditions such as
out with satisfactory results after a waiting period of 30–60 pigment epithelial detachment, cystoid retinal edema, and sub-
minutes. retinal neovascularization could be evaluated and understood
The plan we have suggested allows the fluorescein angiogram only histologically. Now they are widely appreciated and recog-
to yield all the information necessary to make a proper and nized clinically. Because fluorescein angiography graphically
thorough interpretation. demonstrates fundus pathophysiology, and because we rely on
Box 1.3 provides a checklist of important steps in the fluores- histologic points of reference to interpret a fluorescein angio-
cein angiography procedure. gram, a thorough knowledge of the anatomy of the fundus and
its microscopic layers is necessary to interpret fluorescein angio-
Diabetic retinopathy grams correctly. To interpret a fluorescein angiogram, it is essen-
Diabetic retinopathy presents a unique challenge for the photog- tial to understand the microscopic layers of the fundus (i.e., the
rapher as significant pathology may be located both within the histology).
macula and the periphery. A photographic plan must yield A logical place to begin this study is at the vitreous. In its
information regarding leakage contributing to diabetic macular normal state, and in a normal angiogram, the vitreous is clear
edema and nonfilling from capillary nonperfusion. At the same and nonfluorescent. However, when it contains opacities that
time, peripheral scans confirming the presence of neovascular- block the view of retinal and choroidal fluorescence, hypofluo-
ization in preproliferative and proliferative diabetic retinopathy rescence occurs. The vitreous is also an important point of refer-
must also be obtained. In this setting, rather than stereo pairs of ence when intraocular inflammation or retinal neovascularization
macula and disc of the fellow eye, photographs of the nasal, is present. In these cases fluorescein leaks into the vitreous,
causing fluffy fluorescence as fluorescein molecules disperse The choriocapillaris is made up of discrete units called lobules,
into fluid vitreous and vitreous gel. thought to be approximately one-fourth to one-half of a disc
12 For the purpose of fluorescein angiographic interpretation, it diameter in size. The center of each lobule is fed by a precapillary
is convenient to divide the sensory retina into two layers: the arteriole (terminal choroidal arteriole), which comes from a short
inner vascular half and the outer half, which is avascular. The posterior ciliary artery. Each lobule functions independently in
inner vascular half extends from the internal limiting membrane the normal state. It has been assumed that angiographic zones
Section I

to the inner nuclear layer. This portion of the retina contains the of delayed or patchy choroidal filling gradually fill in a trans-
retinal blood vessels, which are located in two separate planes: verse fashion, with one lobule spilling over into another. Careful
the larger retinal arteries and veins are located in the nerve fiber inspection, however, indicates that these filling defects generally
layer; the retinal capillaries are located in the inner nuclear layer. remain the same size, indicating a delayed filling from a pos­
In a well-focused stereoscopic fluorescein angiogram, these two terior origin (its own arteriolar feeder). In the abnormal state, as
vascular layers can be seen as distinct planes in the retina. An when a choroidal vascular occlusion occurs, there is a freely
Optical Imaging Technologies
Retinal Imaging and Diagnostics

extremely important fluorescein angiographic concept is that connecting “spilling over” of blood flow from well-perfused
normal retinal blood vessels are impermeable to fluorescein choroid to the occluded area.
leakage; that is, fluorescein flows through the normal retinal Around the margin of each lobule is a ring of postcapillary
vessels without leakage into the retina. venules that drain each lobule. These postcapillary venules drain
The outer avascular half of the sensory retina comprises the into the vortex veins, which drain the entire choroid. There are
outer plexiform layer, the outer nuclear layer, and the rods and usually four vortex veins, and each functions as a well-defined
cones. The outer plexiform layer is the primary interstitial space quadrantic segmental drainage system for the entire uvea. In the
in the retina. When the retina becomes edematous, it is in this case of a posterior ciliary artery obstruction, this occluded
layer that fluid accumulates, causing cystoid spaces. Deep retinal portion of the choroid can fill by a retrograde mechanism from
hemorrhages and exudates (lipid deposits) may also be depos- an adjacent posterior ciliary artery by way of the choroidal
ited in the outer plexiform layer. venous system. This mechanism may provide adequate nourish-
The rods and cones are very loosely attached to the pigment ment to prevent extensive ischemic changes until the occluded
epithelium, especially in the macular region, whereas the artery reopens.
pigment epithelium is very firmly attached to Bruch’s mem- Knowledge of each of these layers of the fundus is important
brane. In fluorescein angiographic interpretation the pigment in understanding fundus histopathology. The following six
epithelium is an extremely important tissue because it prevents areas, however, are more important than others in the
fluorescein leakage from the choroid and blocks, to a greater or interpretation of abnormal fundus fluorescence:
lesser extent, visualization of choroidal fluorescence.
Bruch’s membrane separates the pigment epithelium from the 1. preretinal area, where contraction from an epiretinal
choriocapillaris, which is permeable to fluorescein. Fluorescein membrane may influence the retinal circulation and where
passes freely from the choriocapillaris and diffuses through hemorrhage may be located
Bruch’s membrane up to, but not into, the pigment epithelium. 2. vascular layers of the sensory retina, both superficial and
Beneath the choriocapillaris are the larger choroidal vessels, deep
which are impermeable to fluorescein. Melanocytes are dis- 3. avascular portion of the sensory retina, particularly the outer
persed throughout the choroid but are most heavily concen- plexiform layer, the principal site of intraretinal edema and
trated in the lamina fusca, the thin layer between the choroid exudate
and sclera. The sclera lies beneath the choroidal vessels. 4. retinal pigment epithelium, which has the potential for many
The ophthalmic artery gives rise usually to two main posterior manifestations, including proliferation, depigmentation,
ciliary arteries: the lateral and medial. However, three posterior hyperpigmentation, and detachment
ciliary arteries may be present, in which case the medial artery 5. choroidal circulation, including the choriocapillaris and the
is the one usually duplicated less frequently. In rare instances large choroidal vessels
there may be a superior posterior ciliary artery. 6. sclera, which lies beneath the choroid.
The posterior ciliary arteries supply the lateral and medial
halves of the disc and choroid. During angiography a vertical Throughout this chapter a modified schematic drawing relates
zone of slightly delayed filling may be seen passing through various fluorescein angiographic abnormalities to fundus histo-
the papillomacular region, including the disc. Occasionally, pathologic changes (Fig. 1.17). The size and proportion of these
there is an oblique orientation to this supply or even a supero- various layers have been modified to include various pathologic
inferior distribution. This border between the main posterior manifestations and to illustrate the effects of these abnormalities
ciliary arteries has been termed the watershed zone, where on the angiogram. Because of its importance and various patho-
patchy choroidal filling often can be seen on fluorescein logic changes, the pigment epithelium is drawn to a larger scale
angiograms. in relation to other fundus structures. Only the inner portion of
Each main posterior ciliary artery divides into numerous short the sclera is represented because the outer portion of the sclera
arteries and one long artery. On the temporal side the short is usually of little importance to angiographic interpretation. The
posterior ciliary arteries supply small, variously sized, wedge- retinal and choroidal vessels are drawn larger and more numer-
shaped choroidal segments, whose apices are centered near the ous than they appear in a normal histopathologic section to
macula. The lateral long posterior ciliary artery passes obliquely emphasize the contribution of circulatory pathophysiologic
through the sclera. It supplies a wedge of choroid that begins interpretation.
temporal to the macular region and participates in the formation Two specialized areas of the fundus warrant more detailed
of the greater circle of the iris. discussion: the macula (Fig. 1.18) and the optic nerve head.
Vitreous
13

Internal limiting membrane

Chapter 1
Nerve fiber and vessel layer
Ganglion cell layer
Inner plexiform layer

Inner nuclear layer and capillaries

Outer plexiform layer

Fluorescein Angiography: Basic Principles and Interpretation


Outer nuclear layer

Photoreceptors
Retinal pigment epithelium
Bruch’s membrane
Choriocapillaris

Choroid

Sclera

Fig. 1.17 Modified schematic drawing of a microscopic section of retina, pigment epithelium, and choroid.

Vitreous

Internal limiting membrane


Nerve fiber and vessel layer
Ganglion cell layer

Inner nuclear layer and capillaries

Outer plexiform layer

Outer nuclear layer

Photoreceptors
Retinal pigment epithelium
Bruch’s membrane
Choriocapillaris

Choroid

Sclera

Fig. 1.18 Modified schematic drawing of a microscopic section of the macula.

The fovea is the center of the macula and contains only four edema in the macula as opposed to the honeycomb appear-
layers of the retina: (1) the internal limiting membrane; ance of cystoid edema outside the macula. Beyond the macular
(2) the outer plexiform layer; (3) the outer nuclear layer; and region the outer plexiform layer is perpendicular rather than
(4) the rods and cones. No intermediate layers exist between oblique.
the internal limiting membrane and the outer plexiform layer The pigment epithelial cells in the macula are more columnar
in the fovea, which in the macula is oblique. This is an impor- and have a greater concentration of melanin and lipofuscin
tant factor in understanding the stellate appearance of cystoid granules than in the remainder of the fundus.
Xanthophyll is present in the fovea, located probably in the from the retina. They are, most accurately, retinovenous to cilio­
outer plexiform layer. These differences in pigmentation are the venous collaterals.
14 chief factors responsible for producing the characteristic dark In summary, fluorescein angiography provides an in vivo
zone in the macular region on normal angiograms. The absence understanding of the histopathologic and pathophysiologic
of retinal vessels in the fovea (i.e., the perifoveal capillary-free changes of various fundus abnormalities. Therefore an anatomic
zone), in most cases approximately 400–500 mm in diameter in and, more specifically, a histologic understanding of important
Section I

the center of the fovea, is another cause of the dark appearance fundus landmarks is essential to fluorescein angiographic
of the macula. interpretation.
The optic nerve head, or disc, is the other highly specialized
tissue of the posterior pole. The disc is fed by two circulatory Normal fluorescein angiogram
systems: the retinal vascular system and the posterior ciliary The normal fluorescein angiogram is distinguished by certain
vascular system. Widespread anastomotic channels exist specific characteristics. Knowledge of these characteristics pro-
Optical Imaging Technologies
Retinal Imaging and Diagnostics

between the posterior ciliary vasculature and the optic nerve and vides an essential frame of reference for interpreting abnormal
retinal vasculature and become exaggerated in certain patho- fluorescein angiograms.
logic conditions. The disc is made up of many layers of nerve In the normal fluorescein angiogram (Fig. 1.19), the first true
fibers and glial supporting columns that contain the large retinal fluorescence begins to show in the choroid approximately 10–12
vessels. seconds after injection in young patients (e.g., adolescents) and
The central retinal artery arises from the ophthalmic artery in 12–15 seconds after injection in older patients.
close proximity to the main posterior ciliary arteries. In about Fluorescence can appear even earlier than 8 seconds in very
45% of the population, the central retinal artery and the medial young patients. The choroid occasionally begins to fluoresce 1
posterior ciliary artery arise from a common trunk. In 12% of or 2 seconds before the initial filling of the central retinal artery.
persons the central retinal artery originates from the ciliary Early choroidal fluorescence is faint, patchy, and irregularly
artery. Therefore it is impossible to have a choroidal infarction, scattered throughout the posterior fundus. It is interspersed with
anterior ischemic optic neuropathy, and a central retinal artery scattered islands of delayed fluorescein filling. This early phase
occlusion all due to a single site of obstruction. is referred to as the choroidal flush. When adjacent areas of
The central retinal artery provides a major source of blood choroidal filling and nonfilling are quite distinct, the pattern is
supply to the axial portion of the anterior orbital portion of designated as patchy choroidal filling.
the optic nerve. In the intraneural or axial course, short cen- Within the next l0 seconds (approximately 20–25 seconds after
trifugal branches arise but usually end a short distance behind injection), the angiogram becomes very bright for about 5
the lamina cribrosa. There are then no further branches from seconds because of the extreme choroidal fluorescence. Choroi-
the central retinal artery until it reaches the retina. If a cilio- dal fluorescence, however, is not visible in the macula because
retinal artery is present, it supplies the corresponding segment of the taller, more pigmented epithelium present in the fovea
of the disc. (retina). Therefore the macula remains dark throughout the
The peripapillary nerve fiber layer is supplied by small, recur- angiogram.
rent branches from the retinal arterioles at the peripapillary If present, a cilioretinal artery usually begins to fluoresce as
region. Emanating from these arterioles at the disc are the radial the choroid fluoresces, rather than as the retina fluoresces.
papillary capillaries. These capillaries are rather straight and Within 1–3 seconds after choroidal fluorescence is visible, or
long, have few anastomoses, and lie in the superficial portion of approximately 10–15 seconds after injection, the central retinal
the peripapillary nerve fiber layer. The capillaries to the disc are artery begins to fluoresce. The less dense the concentration of
continuous with these retinal peripapillary capillaries. pigment in the pigment epithelium, the greater the time between
The short posterior ciliary arteries, or the recurrent branches the visibility of the choroidal fluorescence and the filling of the
from the peripapillary choroid, supply the retrolaminar portion retinal vessels. The lighter pigment presents less interference to
of the optic nerve. The laminar cribrosa portion of the nerve is choroidal fluorescence, allowing it to be evident earlier in its
supplied by centripetal branches of the short posterior ciliary filling phase. With a more densely pigmented pigment epithe-
arteries. In this region a partial, or, rarely, a complete Zinn’s lium, the blockage barrier effect is greater. Therefore choroidal
vascular circle is occasionally found. The prelaminar portion is fluorescence appears somewhat later because a greater concen-
supplied by centripetal branches from the peripapillary choroid. tration of fluorescein is required to overcome the increased
Because most of the disc is fed by the ciliary system, fluores- density of the pigment epithelial barrier.
cein appears simultaneously at the optic nerve head and the Because no barrier exists in front of the retinal vessels, the
choroid and before it is apparent in the retinal arteries. patient’s pigmentation has no effect on the visibility of the
The main venous drainage of the disc is into the central retinal retinal vessels, although the degree of pigmentation does affect
vein. The prelaminar portion empties into both the central the contrast of the angiophotographs. The darker the pigment
retinal vein and the peripapillary choroid, thus providing poten- epithelium is, the less visible the choroidal fluorescence will
tial collateral drainage in the case of obstruction of the central be and the greater the contrast of the retinal vascular fluo-
retinal vein behind the lamina cribrosa. Such large dilated col- rescence (i.e., the better they stand out). The lighter the pigment
laterals are frequently seen following central retinal vein occlu- epithelium is, the more visible the choroidal fluorescence will
sion and are called retinociliary veins. Some mistakenly call be and the less the contrast of the fluorescence from the retinal
them opticociliary shunts, a misnomer because they are not true vessels.
shunts (defined as a congenital artery that empties into a vein After the central retinal artery begins to fill, the fluorescein
and that skips the capillary bed, sometimes part of the Wyburn– flows into the retinal arteries, then into the precapillary arteri-
Mason syndrome), and they are not optico because they emanate oles, the capillaries, the postcapillary venules, and finally the
retinal veins. Because the fluorescein from the venules enters the the junction of two veins, the inner lamina of each vein may
veins along their walls, the flow of fluorescein in the veins is merge. This creates three laminae: one in the center and one on
laminar. Because vascular flow is faster in the center of a lumen each side of the vein. As fluorescein filling increases in the veins, 15
(tube) than on the sides, the fluorescein seems to stick to the the laminae eventually enlarge and meet, resulting in complete
sides, creating the laminar pattern of retinal venous flow. The fluorescence of the retinal veins.

Chapter 1
dark (nonfluorescent) central lamina is nonfluorescent blood Fluorescence of the disc emanates from the posterior ciliary
that comes from the periphery, which takes longer to fluoresce vascular system, both from the edge of the disc and from the
because of its more distant location. tissue between the center and the circumference of the disc.
In the next 5–10 seconds, fluorescence of the two parallel Filling also comes from the capillaries of the central retinal artery
laminae along the walls of the retinal veins becomes thicker. At on the surface of the disc. Because healthy disc tissue contains

Fluorescein Angiography: Basic Principles and Interpretation


A B

C D

Fig. 1.19 Normal fundus photos and fluorescein angiogram of left disc and macula taken with a 50° camera. (A) Montage photograph of multiple
fields shows normal macula, fovea, and retinal vessels. (B) Early arterial phase of the fluorescein angiogram. Note the ground-glass fluorescence
of the choriocapillaris. There is very little fluorescence in the retinal veins; just the margins of the veins are fluorescent. This is the earliest
portion of the laminar filling phase of the vein. Note some hyperfluorescence of choriocapillaris. These dark patches of the choroid are areas that
have not fully filled, referred to as patchy choroidal filling. (C) The retinal arteries and capillaries have filled and the retinal veins have filled more
substantially. Note the laminar flow in the retinal veins; this is indicated by the white line of fluorescence along the walls of the retinal veins.
(D) Late venous phase. Laminar filling is no longer detectable and uniform filling is seen in both arterial and venous circulation.
Continued
16
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

E F

Fig. 1.19 Cont’d (E) Mid to later arteriovenous phase of fluorescein angiogram. Note that the ground-glass fluorescence of the choriocapillaris is
complete. The retinal arteries and veins are completely filled. (F) Arteriovenous phase of fluorescein angiogram showing the disc. Again, there is
diffuse fluorescence of the choriocapillaris. The arteries and veins have completely filled, and optic nerve fluorescence is normal.

many capillaries, the disc becomes fairly hyperfluorescent on the To summarize, the angiogram is initially dark; choroidal and
angiogram. retinal filling is seen 10–15 seconds after fluorescein injection.
The perifoveal capillary net cannot always be seen on the fluo- The retinal and choroidal vasculatures fill maximally about
rescein angiogram. It can be seen best in young patients with 20–30 seconds after injection. Late angiophotographs show fluo-
clear ocular media about 20–25 seconds after a rapid fluorescein rescence of the choroid and sclera (if the pigment epithelium is
injection. This is called the “peak” phase of the fluorescein angio- light) and fluorescence of the optic cup and the edge of the disc,
gram. The photographer should be aware of this phase and be but otherwise the fundus is dark (nonfluorescent in the late
sure not to miss it by shooting as rapidly as possible as the fluo- phase).
rescein concentration increases and by continuing to shoot
rapidly until the concentration of fluorescein begins to decrease.
Approximately 30 seconds after injection, the first high-
ABNORMAL FLUORESCEIN ANGIOGRAM
concentration flush of fluorescein begins to empty from the cho- The purpose of this section is to offer a schema by which the
roidal and retinal circulations. Recirculation phases follow, interpretation of the fluorescein angiogram follows a simple and
during which fluorescein in a lower concentration continues to logical progression. The first step is to recognize areas of abnor-
pass through the circulation of the fundus. mal fluorescence and determine if they are hypofluorescent or
Generally, 3–5 minutes after injection, the choroidal and retinal hyperfluorescent (Fig. 1.20).
vasculatures slowly begin to empty of fluorescein and become
gray. Vessels of most normal patients almost completely empty Hypofluorescence
of fluorescein in approximately 10 minutes. The large choroidal Hypofluorescence is a reduction or absence of normal fluores-
vessels and the retinal vessels do not leak fluorescein. However, cence, whereas hyperfluorescence is abnormally excessive fluo-
because of large gaps in its endothelium, the choriocapillaris rescence. A systematic series of decisions follows this initial
does leak fluorescein. The extravasated fluorescein diffuses differentiation to arrive at a proper diagnosis. These decisions
through the choroidal tissue, Bruch’s membrane, and sclera. relate to: (l) the anatomic location of various abnormalities;
Leakage of fluorescein with retention in tissues is designated as (2) the quality and quantity of the abnormal fluorescence; and
staining. In the later phase of the angiogram, staining of Bruch’s (3) other unique characteristics, as indicated in Fig. 1.20.
membrane, the choroid, and especially the sclera may be visible Hypofluorescence is any abnormally dark area on the positive
if the pigment epithelium is lightly pigmented. The disc and print of an angiogram. There are two possible causes of hypo-
adjacent visible sclera remain hyperfluorescent because of stain- fluorescence: blocked fluorescence or a vascular filling defect.
ing. When the retinal pigment epithelium is especially lightly Blocked fluorescence is sometimes referred to as masked,
pigmented, the large choroidal vessels can be seen in silhouette obscured, or negative fluorescence or transmission decrease.
against the fluorescent (fluorescein-stained) sclera. The lamina Each of these terms indicates a reduction or absence of normal
cribrosa within the disc also remains hyperfluorescent because retinal or choroidal fluorescence because of a tissue or fluid
of staining. This depends on the cup-to-disc ratio and the pres- barrier located anterior to the respective retinal or choroidal
ence of any visible sclera, such as occurs within a conus adjacent circulation. For example, blood in the vitreous or a layer of blood
to the disc. The edge of the disc stains from the adjacent chorio- in front of the retina obscures the view of the retinal and choroi-
capillaris, which normally leaks. dal circulations and therefore blocks fundus fluorescence from
Anterior segment
17
Retinal material Vitreous
Inner retinal
Blocked

Chapter 1
Deep retinal
Choroidal material
Subretinal (or sub-RPE)

Artery
Hypofluorescence Vein

Fluorescein Angiography: Basic Principles and Interpretation


Capillary bed
Retinal (cap. nonperfusion)
Combination

Vascular
filling defect Disc Capillary nonfilling

Physiologic
Posterior ciliary
Choroidal artery obstruction
Absence (congenital or
atrophic) of choroidal
vascular tissue

Optic nerve head drusen


Autofluorescence
Preinjection Astrocytic hamartoma
fluorescence
Pseudofluorescence Poorly matched filters

Tortuosity and dilation


Anastomosis
Retinal
(abnormal vessels) Neovascularization
Aneurysms
Telangiectasis
Tumor vessels
Hyperfluorescence Early (vascular)

PE window Atrophy
defect Congenital reduction

Choroidal Subretinal
neovascularization
Abnormal
vessels Inflammation
Tumor vessels

Neovascularization
Vitreous Inflammation
Tumors
Disc
Late (leak, Cystoid edema
extravascular) Retinal
Noncystoid edema

Pooling
Choroidal
Staining

Fig. 1.20 Flow sheet for abnormal fluorescein angiography.


these tissues. Hemorrhage that lies under the retina or retinal Any anterior-segment material, such as a corneal opacity,
pigment epithelium, but in front of the choroidal circulation, anterior-chamber haziness, or lens opacity, obscuring the view
18 does not obstruct visibility of the retinal circulation but does of the ocular fundus will result in an angiogram of reduced bril-
block the view of the choroidal circulation. Therefore the approx- liance, contrast, and resolution. This affects the quality of the
imate histologic location of blocking material can be determined angiogram and is, in a sense, a type of blocked fluorescence.
by the presence or absence of visibility of one or both fundus Many conditions of the vitreous produce a hazy medium that
Section I

circulations. prevents visualizing fundus detail. The most common vitreous


Fluorescein is present but cannot be seen in blocked fluores- opacity to cause blockage is hemorrhage. Whether diffusely dis-
cence. With vascular filling defects, however, fluorescein cannot persed in the vitreous gel or more densely accumulated, vitreous
be seen because it is not present. hemorrhage reduces or completely blocks fundus fluorescence.
The key to differentiating blocked fluorescence from a vascu- In addition to hemorrhage, media haze may be caused by a
lar filling defect is to correlate the hypofluorescence on the variety of opacifications, including asteroid hyalosis, vitreous
Optical Imaging Technologies
Retinal Imaging and Diagnostics

angiogram with the ophthalmoscopic view. If there is material condensation resulting from vitreous degenerative disease,
visible ophthalmoscopically that corresponds in size, shape, and inflammatory debris, vitreous membranes, or opacification sec-
location to the hypofluorescence on the angiogram, then blocked ondary to amyloidosis. When anterior-segment and vitreous
fluorescence is present. If there is no corresponding material on opacities are present, the angiogram may be of higher resolution
the color photograph, then it must be assumed that fluorescein and quality than the color photograph because the light scat-
has not perfused the vessels and that the hypofluorescence is tered from the nonfluorescing opacities is not transmitted
caused by a vascular filling defect. through the barrier filter and therefore has no effect on the angio-
Hypofluorescence resulting from a vascular filling defect graphic photograph.
occurs when either of the two fundus circulations is not perfus- Any translucent or opacified material in the retina or in the
ing normally. This is caused by an absence of the vascular tissue nerve fiber layer blocks fluorescence from both planes of retinal
or by a complete or partial obstruction of the particular vessels. vessels, as well as from the choroidal vessels. The large retinal
In these situations an absence or delay of fluorescence of the vessels and precapillary arterioles are located in the nerve fiber
involved vessels will occur. This type of hypofluorescence has a layer in the anterior plane of the retina. The capillaries and post-
pattern that follows the geographic distribution of the vessels capillary venules are located deeper in the retina, in the inner
involved. Although the ophthalmoscopic picture will demon- nuclear layer. If a blocking material lies in front of the nerve fiber
strate the material blocking fluorescence, it may show nothing if layer, it blocks both planes of retinal vessels (Fig. 1.21). However,
the hypofluorescence is the result of a vascular filling defect. if the material lies beneath the nerve fiber layer but within or in
To summarize, after an area of hypofluorescence is recog- front of the inner nuclear layer (where the smaller retinal vessels
nized, one must refer to the ophthalmoscopic photograph to are located), it blocks only the retinal capillaries (and choroidal
determine the cause. If material is visible ophthalmoscopically vessels), leaving the view of the large retinal vessels unob-
and corresponds to the area of hypofluorescence, this is blocked structed. If a blocking material lies deeper than the retinal vas-
fluorescence. If no corresponding blocking material exists, the cular structures, deep to the inner nuclear layer, it does not block
hypofluorescence is therefore a vascular filling defect. the vessels but will block the choroidal vascular fluorescence. In
other words, deep intraretinal blocking material, such as hemor-
Anatomic location of hypofluorescence rhage or exudate, does not obstruct retinal vascular fluorescence,
After determining the cause of the hypofluorescence, the next since the retinal vessels are located in the inner half of the retina
step is (1) to determine the anatomic location of the material that (Fig. 1.22).
is blocking fluorescence or (2) to determine which of the two Therefore one can determine the location of a retinal abnor-
fundus circulations is involved in the filling defect. Blocking mality, such as hemorrhage, by the vessels that are blocked by
material affects the retinal and choroidal circulations if it is it and by the fluorescence of the vessels that are not blocked.
located in front of the retina. The material blocks only the cho- The most common cause of blocked retinal vascular fluores-
roidal circulation if it is located beneath the retinal circulation cence is hemorrhage. Subinternal limiting membrane hemor-
and in front of the choroid. Similarly, vascular filling defects rhage blocks fluorescence of all underlying retinal vessels and
occur in either the retinal or the choroidal vasculature or in the choroidal vasculature. Nerve fiber layer hemorrhage, which
vessels of the optic nerve head. usually is flame-shaped, blocks the smaller retinal vessels lying
deeper in the retina but only partially blocks the larger retinal
Blocked retinal fluorescence vessels in the nerve fiber layer. Blockage from hemorrhage is
Blocked retinal vascular hypofluorescence is caused by anything usually complete, as opposed to the partial blockage caused by
that reduces media clarity. An opacification in front of the retinal the myelinated nerve fibers.
vessels involving the cornea, anterior chamber, iris, lens, vitre- Various retinal vascular (arteriolar) occlusive diseases may
ous, or the most anterior portion of the retina or disc produces cause white ischemic thickening (nerve fiber edema), which
hypofluorescence. results in some opacification of the retina and blockage of
The further the opacification is in front of the fundus, the the remaining retinal vascular and choroidal fluorescence.
less it will block fluorescence and the more it will affect the Conditions such as arterial occlusion in hypertension or
overall quality of the photographs. The closer the material is Purtscher’s retinopathy cause enough intracellular “cloudy”
to the fundus, the more it will block, causing hypofluorescent swelling and opacification to block fluorescence. It should be
images on the angiogram. Any material that blocks retinal vas- noted that, because there is occlusion in this type of hypo-
cular fluorescence will, of course, block choroidal fluorescence fluorescence, the hypofluorescence is caused partly by the
as well. vascular filling defect. However, the opacified ischemic retina
19

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A

B C

Fig. 1.21 Preretinal hemorrhage causing hypofluorescent blockage of all retinal and choroidal fluorescence. (A) Schematic drawing of subhyaloid
(right), subinternal limiting membrane (central), and nerve fiber layer (left) hemorrhages. Each hemorrhage lies in front of the retinal, and
therefore choroidal, vasculature, causing hypofluorescence-blocked fluorescence. (B) Color photograph of the right disc showing substantial
preretinal hemorrhage. (C) Fluorescein angiogram of the right disc showing hypofluorescence caused by blockage as a result of the preretinal
hemorrhage. Comment: All fluorescence of the fundus is blocked because the hemorrhage lies in front of the retinal vasculature.

effectively blocks fluorescence from underlying retinal and inflammatory material, or the like accumulates in front of the
choroidal vasculature. choroidal vasculature and deep to the retinal vasculature
In summary, the concept of blocked retinal vascular hypofluo- (Fig. 1.23).
rescence is fairly easy to understand and to identify on the Deep retinal material
angiogram. When the retinal vessels do not fluoresce, the oph- Materials deposited in the deep retina that cause blockage of
thalmoscopic view should be studied to determine if blocking choroidal fluorescence are fluid, hard exudate, hemorrhage, and
material is located in front of the retinal vessels. pigment.
If blocking material is present, the next step is to determine its Fluid that accumulates in the deep retina has a predilection
anatomic location. for the tissue of least resistance, the outer plexiform layer. Depo-
sition of edema fluid, originating from leaking retinal vessels or
Blocked choroidal fluorescence migrating from subretinal space into the retina, most frequently
Hypofluorescence caused by blocked choroidal vasculature occurs in the outer plexiform layer. After reaching a certain
occurs when fluid, exudate, hemorrhage, pigment, scar, volume, the fluid tends to form spaces, or pockets, between
20
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

B C

Fig. 1.22 Intraretinal hemorrhages causing hypofluorescent blockage. (A) Schematic of retina showing hemorrhages located in most of the layers
of the retina from the internal limiting membrane to the outer nuclear layer. (B) Color photograph of left macula shows dot-and-blot, as well as
flame-shaped hemorrhages just above the fovea. This is a case of branch-vein occlusion. (C) Fluorescein angiogram of left macula shows that
the hemorrhage causes irregular hypofluorescent blockage. The flame-shaped hemorrhage located in the nerve fiber layer blocks all the retinal
vasculature. The dot-and-blot hemorrhages do not block the large retinal vessels and therefore can be localized deeper in the retina. The
hemorrhages that do not block retinal capillary fluorescence can be located deeper to the capillary layer, which is in the inner nuclear layer.
Comment: Once hypofluorescent blockage is determined, an anatomic localization of the blocking material can be made by determining which
normally fluorescent structures can be seen and which are being blocked.

compressed nerve and Müller’s fibers, which are pushed aside When retinal vessels bleed, the blood can be deposited anywhere
in the process. This pattern of fluid accumulation in the outer in the retina. When located deep to the retinal vessels beneath
plexiform layer is called cystoid retinal edema. Noncystoid the inner nuclear layer, retinal vascular fluorescence is visible,
retinal edema occurs when the volume of extracellular fluid is whereas choroidal fluorescence is blocked.
insufficient to produce pockets, or spaces, in the outer plexiform
layer or other layers of the retina. A significant amount of retinal Subretinal material
edema, whether cystoid or noncystoid, especially if turbid or Any opaque or translucent substance located beneath the retina
containing lipid-laden macrophages, partially blocks choroidal but in front of the choroid blocks fluorescence of the choroidal
fluorescence in the early phase of the fluorescein angiogram. vasculature but does not block retinal vascular fluorescence
Later in the angiogram, retinal edema fluoresces. Intraretinal (Fig. 1.23). Blood located under the retina causes complete
hard exudates and lipid-laden macrophages, usually located in blockage of choroidal fluorescence, with the retinal fluorescence
the outer plexiform layer, partially block choroidal fluorescence. showing through normally. Subretinal hemorrhage appears red,
21

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A

B C

Fig. 1.23 Subretinal hemorrhage causing hypofluorescence, specifically, blockage of choroidal fluorescence. (A) Schematic of retina with
subretinal hemorrhage (blood located between photoreceptors and pigment epithelium). (B) Color photograph of right macula of an eye
with angioid streaks showing large scattered areas of subretinal hemorrhage. (C) Fluorescein angiogram of right macula shows marked
hypofluorescence caused by blocked choroidal fluorescence (the retinal vessels are visible) that is due to the subretinal hemorrhage. Comment:
The subretinal hemorrhage completely obscures fluorescence from the choroid. The retinal vessels are clearly seen overlying the subretinal
hemorrhage.

and subpigment epithelial hemorrhage is dark. Subretinal hem- block much of the choroidal fluorescence (Fig. 1.25) and espe-
orrhage is generally scalloped with somewhat irregular margins, cially blocks the later hyperfluorescent staining of the sclera. The
whereas subpigment epithelial hemorrhage is often quite round choriocapillaris may be seen normally over the nevus.
and well demarcated (Fig. 1.23). To summarize, various materials located in the deep retinal
Accumulated pigment (melanin and lipofuscin) from diseased layers, or beneath the retina, block choroidal fluorescence and
retinal pigment epithelium causes blocked choroidal fluores- are evident ophthalmoscopically. These materials result from a
cence (Fig. 1.24). Any hyperpigmentation of the pigment epithe- variety of disease processes.
lium causes blocked choroidal fluorescence. Xanthophyll, the
pigment present in the outer layers of the fovea, blocks choroidal Vascular filling defect
fluorescence by selectively absorbing the blue exciting light, The second cause of abnormal hypofluorescence is vascular
which results in less fluorescence. Finally, a choroidal nevus may filling defect. With blocked fluorescence, the fluorescein is
22
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

B C

Fig. 1.24 Hypertrophy of the retinal pigment epithelium. (A) Schematic showing hypertrophic pigment epithelial cells. (B) Color photograph of the
macula shows a well-demarcated hyperpigmented lesion. (C) Fluorescein angiogram of the same area shows marked hypofluorescence of the
choroid resulting from blocked fluorescence. Comment: This patient had marked hypertrophy of the retinal pigment epithelium, which allowed
normal retinal fluorescence; it completely blocked choroidal fluorescence.

present in the circulations of the fundus but is not visible because caused by a vascular filling defect. In some instances both forms
a tissue or fluid barrier conceals it. With vascular filling defect, of hypofluorescent mechanisms play a role simultaneously, as
fluorescein cannot be seen because it is not present. Since fluo- with retinal arteriolar occlusion, when the retina is not only not
rescein reaches the retina and choroid by way of vessels, lack of perfused (vascular filling defect) but is ischemic and therefore
the fluorescein dye in either vascular system indicates an obstruc- white and opaque, causing blocked fluorescence.
tive problem or a lack of vessels (i.e., a vascular filling defect). Vascular filling defects result from vascular obstruction,
As previously indicated, when a hypofluorescent area is seen atrophy, or absence (congenital or otherwise) of vessels. Any of
on an angiogram, the best way to differentiate blocked fluores- these conditions can be total or partial. When the obstruction is
cence from a vascular filling defect is to compare the angiogram complete (occlusion) or the vascular tissue is atrophied com-
with the ophthalmoscopic picture. When blood, pigment, or pletely, the hypofluorescence is complete and lasts throughout
exudate can be seen ophthalmoscopically corresponding to the the angiogram. When the obstruction is only partial or the vas-
area of hypofluorescence, the material is causing blocked fluo- cular tissue is not entirely atrophied, the vascular fluorescein
rescence. When no material is visible ophthalmoscopically (on filling is delayed or reduced relative to corresponding areas that
the color photograph), one must assume that fluorescein has not fill normally. Whatever the cause of a partial vascular filling
perfused the vessels and that the abnormal hypofluorescence is defect, hypofluorescence will be seen in the early phases of the
23

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A
B

C D

Fig. 1.25 Choroidal nevus hypofluorescent blockage. (A) Schematic drawing of retina showing choroidal nevus. Note that the choriocapillaris is
intact. (B) Color photograph of nevus. (C) Arteriovenous phase of fluorescein angiogram shows hypofluorescence corresponding to the area of
the nevus. (D) Later arteriovenous phase of fluorescein angiogram shows that the nevus is still hypofluorescent, although the choriocapillaris
ground-glass fluorescence can be seen surrounding.

angiogram but may not persist throughout the entire angiogram. Retinal vascular filling defect
Some vascular filling, although delayed or reduced, will eventu- If a retinal vascular filling defect is present, the clinician then
ally occur. considers whether the defect results from obstruction of a retinal
Once it is determined that a vascular filling defect is the cause artery or vein, capillary bed, or any combination of these. Dis-
of an area of hypofluorescence, the next step is to determine tinguishing the cause of the obstruction is not difficult because
which of the retinal, disc, or choroidal vessels are involved. A the fluorescein angiographic process is dynamic and timed.
vascular filling defect of the disc is easy to discern angiographi- When nonfilling of a specific retinal vessel occurs, it is easy to
cally. Determining whether a vascular filling defect is retinal or differentiate an arterial occlusion from a venous occlusion
choroidal can be more difficult. Since retinal vessels are normally because the retinal arteries fill first, then the retinal capillary bed,
present, however, the absence of retinal vessels is usually readily followed by the retinal veins. In addition, retinal vascular filling
apparent. If, on the other hand, a vascular filling defect is found defects can be localized by tracing the course of a particular
but the retinal vessels are full and visible, the hypofluorescence vessel; these defects correspond anatomically to the normal dis-
must be choroidal in origin. Stereoscopic angiophotographs tribution of the retinal vasculature (Figs 1.26 and 1.27). Thus
allow one to distinguish between the planes of the retina and retinal vascular filling defects result from a variety of disease
choroid and enable exact determination of the location of the processes, but most are commonly associated with atherosclero-
hypofluorescence. sis and diabetes.
24
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.26 Branch retinal artery occlusion. (A) Color photograph


showing areas of retinal whitening inferior to the macula. An intra-
arterial embolus is seen proximal to the whitened retina. (B) Earliest
arterial filling. An area of hypofluorescence is located inferior to the
macula. A small retinal artery feeding this area is occluded at the site
of the embolus previously identified on Figure 1.26A. (C) Midarterial
venous filling with a small area of intra-arterial hyperfluorescence distal
to the site of embolic obstruction. (D) Late arteriovenous phase of
fluorescein angiogram shows that the occluded artery is still mostly
hypofluorescent. (E) Portions of the area did fill with fluorescein due to
E retrograde filling from surrounding areas. There is some mild staining
of the occluded retinal artery.
25

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

Fig. 1.27 Retinal branch-vein occlusion. (A) Color photograph of the right macula and disc. There are areas of retinal hemorrhage, retinal
whitening, and cotton-wool spots. (B) The fluorescein angiogram of the right disc and macula shows normal fluorescence of the superior portion
of the macula. The inferior portion shows substantial hypofluorescence due to retinal capillary nonperfusion. The very bright hyperfluorescent
areas are due to neovascularization. Comment: This patient had a very ischemic inferotemporal branch retinal vein occlusion of the right eye.
This was a severe occlusion, as evidenced by closure of large areas of the capillary bed. The hypofluorescence was caused not only by vascular
filling defect but also by the nonperfused retina, which becomes partially opaque and caused hypofluorescence of the choroid. (In other words,
there was blockage of choroidal fluorescence by the opaque retina, which was caused by the retinal capillary nonperfusion.)

Vascular filling defects of the disc procedure, although leakage from surrounding areas of normal
Vascular filling defects of the disc occur because of the failure of choriocapillaris extends into the occluded area. When sufficient
the capillaries of the optic nerve head to fill. This failure can be leakage occurs, the sclera retains fluorescein (stains) late in
caused by: (1) congenital absence of disc tissue, as in an optic pit the angiogram. When the involved area is large and the leakage
or optic nerve head coloboma (Fig. 1.28); (2) atrophy of the disc is minimal, the hypofluorescence remains throughout the
tissue and its vasculature, as in optic atrophy; or (3) vascular later stages.
occlusion, as in an ischemic optic neuropathy.19,20 Each condition A normal physiologic condition exists in many patients in
is characterized by early hypofluorescence caused by nonfilling which the choroid fills in a patchy manner. Areas adjacent to the
and late hyperfluorescence resulting from staining of the foci that are filling show early hypofluorescence but eventually
involved tissue. fill normally, usually 2–5 seconds later. This has been termed
patchy choroidal filling, and it is the most common form of
Choroidal vascular filling defect choroidal vascular filling defect. This form of filling follows a
The normal choroidal vasculature is usually difficult to docu- pattern in which the short posterior ciliary arteries enter the eye
ment with fluorescein angiography because of the pigment epi- perpendicularly through the sclera. These vessels then feed the
thelial barrier. If chronic choroidal vascular filling defects exist, choriocapillaris lobules.
the pigment epithelium is often secondarily depigmented or The prechoriocapillaris arterioles and lobules are end, or ter-
atrophied. In these cases the hypofluorescence caused by a vas- minal, vessels demonstrating no anastomoses with adjacent cho-
cular filling abnormality of the choroid and choriocapillaris can riocapillaris arterioles or lobules. Each choriocapillaris lobule is
be documented angiographically. connected to adjacent lobules on the venous, or emptying, side
When choroidal vessels do not fill, dark patches of hypo- of the circulation. Fluorescence in each choriocapillaris segment
fluorescence beneath the retina appear early in the angiogram. or lobule is in the form of a round, irregular, or hexagonal patch.
The distribution and morphology of the hypofluorescence vary When some of the channels fill late, a heterogeneous filling
according to the disease process. Because the choroidal cir- pattern results. The choriocapillaris fills most areas, whereas
culation is completely separate from the retinal circulation, dark hypofluorescent patches are present in other areas. These
choroidal vascular filling defects do not correlate with the dark areas are lobules from separate end channels that are not
retinal vascular distribution. If the choriocapillaris is absent filled simultaneously with adjacent choriocapillaris lobules.
and the large choroidal vessels are still present, the choroidal They are filled in a delayed fashion by the single feeder choroidal
and retinal vessels fluoresce, but hypofluorescent gaps appear arteriole.
because of the loss of the diffuse “ground-glass” fluorescence In general, vascular filling defects of the choroid are caused
from the choriocapillaris (Fig. 1.29). When the choroidal vas- by obstructive disorders or absence of tissue with the following
culature does not fill, as in total occlusion or in atrophy, fluorescein angiographic characteristics: (1) normal retinal vas-
hypofluorescence occurs early in the angiogram. The hypo- cular flow; (2) depigmentation of the pigment epithelium; (3)
fluorescence remains throughout the late stages of the reduction of choroidal blood flow; and (4) hypofluorescence in
26
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.28 Optic pit and sensory macula detachment. (A) Color
photograph of left macula. Note the dark area of the optic pit (arrows).
Cystic edema is present in the macula secondary to the macular
schisis detachment from the optic pit. (B) Early arteriovenous phase
of fluorescein angiogram shows hypofluorescence of the disc in the
area of the pit due to absence of tissue and vessels. (C) In the
C late arteriovenous phase fluorescein angiogram, the hypofluorescent
area of the pit is evident.

the early phases of angiography caused by loss of the normal naturally fluoresce (autofluorescence) or by poorly matched
ground-glass choriocapillaris fluorescence. In some conditions filters (pseudofluorescence).
the large choroidal vessels are also absent, resulting in total early Transmitted fluorescence and abnormal vascular fluorescence
hypofluorescence in the affected area, with scleral staining only occur in the early, or vascular, stage of the angiogram, when
on the circumference of the lesion because of the adjacent patent fluorescein fills patent blood vessels. Transmitted fluorescence
choriocapillaris. Choroidal vascular defects result from a variety appears when fluorescein fills the normal choriocapillaris, but it
of disease processes (Figs 1.30 and 1.31). is more noticeable when there is reduced pigment in the pigment
epithelium or loss of retinal pigment epithelium. This is
Hyperfluorescence designated pigment epithelial window defect.
Hyperfluorescence is any abnormally light area on the positive When abnormal retinal, disc, or choroidal vessels are present
print of an angiogram, that is, an area showing fluorescence in and fill with fluorescein, hyperfluorescence occurs. This type of
excess of what would be expected on a normal angiogram. There hyperfluorescence, abnormal vascular fluorescence, is also seen
are four possible causes of abnormal hyperfluorescence: (1) pre- in the early, or vascular, phase of the angiography.
injection fluorescence; (2) transmitted fluorescence; (3) abnormal Hyperfluorescence caused by leakage is seen predominantly
vessels; and (4) leakage. The appearance of fluorescence depends in the later, or extravascular, phase of angiography. In this
in part on the relationship of its appearance to the timing of the phase, fluorescein has emptied from normal and abnormal
fluorescein injection. vessels. Any significant fluorescein that remains in the eye is
Preinjection fluorescence is hyperfluorescence that can be seen fluorescein that has escaped or leaked from vascular or tissue
before fluorescein is injected and is caused by structures that barriers and is thus extravascular.
27

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

Fig. 1.29 Choroideremia: total loss of retinal pigment epithelium (RPE)


and choriocapillaris with much of the large choroidal vasculature
remaining. (A) Color photograph of the left disc and macula. The large
choroidal vasculature can be seen as pale, irregular lines. Dark
patches of pigment are located in the macula and around the disc.
(B) Late venous phase of fluorescein angiogram of the left disc and
macula. The large choroidal vessels can be seen filling, as can the
retinal arteries. The choriocapillaris is not seen. (C) Recirculation
phase of fluorescein angiogram. The large choroidal vessels and
retinal vessels can be seen, but the choriocapillaris (usually seen
as ground-glass fluorescence) is not seen except in the far edges
of the view. Comment: This patient had a total loss of RPE and
choriocapillaris in most areas of the fundus. The ground-glass
choroidal fluorescence was absent from most areas. The large
choroidal vessels could be seen. The large choroidal vessels do not
leak fluorescein, and therefore the sclera did not stain in these areas.
The RPE and choriocapillaris were partially intact in a few areas.
C These can be seen at the far extremes, where there is some mild
ground-glass appearance.

Therefore, to ascertain the type of hyperfluorescence, one Autofluorescence


must determine the time at which the hyperfluorescence Autofluorescence is the emission of fluorescent light from ocular
appears in relation to when the fluorescein was injected. Once structures in the absence of sodium fluorescein. Conditions that
the hyperfluorescence is determined to be caused by preinjec- cause autofluorescence are optic nerve head drusen and astro-
tion fluorescence, transmitted fluorescence, the presence of cytic hamartoma (Fig. 1.32).
abnormal vessels, or by leakage, the next step is to determine Pseudofluorescence occurs when the blue exciter and green
the anatomic location of the hyperfluorescence. Abnormal barrier filters overlap. The blue filter overlaps into the green
blood vessels may come from the retina and disc or from range, allowing the passage of green light, or the green barrier
the choroid. Leakage can occur in the vitreous, disc, retina, filter overlaps into the blue range, allowing the passage of blue
or choroid. light (Fig. 1.2). The overlapping light passes through the system,
reflects off highly reflective surfaces (light-colored or white
Preinjection fluorescence structures), and stimulates the film. This reflected nonfluores-
Each angiographic study should include one photograph of cent light is called pseudofluorescence.
the fundus taken with the fluorescein filters in place and Conditions that tend to produce pseudofluorescence include
before fluorescein is injected. This exposure is called the pre- any light-colored or white (reflective) fundus change (e.g., sclera,
injection, or control, fluorescein photograph. In normal situ- exudate, scar tissue, myelinated nerve fibers, foreign body).
ations this photograph is totally dark; it is completely Currently, fluorescein angiographic filters are usually very
hypofluorescent. When the photograph is not dark, autofluo- well matched; overlap is minimal, so pseudofluorescence is
rescence or pseudofluorescence is present. The conditions that faint and rarely a major problem. However, filters do tend to
cause autofluorescence occur infrequently, and the filter prob- get thin with time. The frequent flashes of light from the
lems that produce pseudofluorescence have in recent years fundus camera wear them down, and most filter pairs eventu-
been minimized by the development of more precisely matched ally allow pseudofluorescence. Therefore, depending on fre-
filter systems. quency of use, fluorescein filters must be changed occasionally.
28
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

B C

Fig. 1.30 Choroidal atrophy, with some remaining islands of choriocapillaris, due to choroideremia. (A) Schematic of retina shows loss of pigment
epithelium and choriocapillaris and some of the outer retina (especially photoreceptors). (B) Color photograph of left superior retina showing
areas of severe atrophy and more intact areas of retinal pigment epithelium (RPE) peripherally. Arrows delineate margins between normal RPE
and RPE atrophy producing window defect. (C) The arteriovenous phase of fluorescein angiogram shows normal fluorescence of the retinal
arteries. The large choroidal vessels can be seen temporally on the right side of the photograph. The ground-glass fluorescence of the
choriocapillaris can be seen more peripherally on the left side of the angiogram, where the RPE and choriocapillaris are more intact. Comment:
This patient had severe atrophy of the RPE and choriocapillaris. Large choroidal vessels could be seen causing hypofluorescence in relationship
to absence of ground-glass choroidal fluorescence. Some areas of choriocapillaris remained and showed normal hyperfluorescence (perhaps
increased hyperfluorescence caused by loss of overlying RPE).

A B C

Fig. 1.31 Choroidal hypoperfusion caused by photodynamic therapy with verteporfin. (A) Left macula. Color photograph shows widespread retinal
pigment epithelial alterations and drusen secondary to an occult choroidal neovascular membrane secondary to age-related macular degeneration.
This treatment was considered standard therapy prior to the advent of intraocular antivascular endothelial growth factor medications. (B) The late
arteriovenous phase of the fluorescein angiogram of the left macula shows hypofluorescence of the macula and a large area temporally. Larger
choroidal vessels are perfused. The macular hypofluorescence corresponds to the laser treated area. The large area of hypofluorescence
temporally represents an area of choroidal nonperfusion caused by selective choriocapillaris occlusion from photodynamic therapy. (C) Later phase
of the fluorescein angiogram shows continued hypofluorescence of the area temporal to the macula despite relative restoration of perfusion to
choriocapillaris temporal to macula.
29

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

Fig. 1.32 Autofluorescence of optic nerve drusen. (A) Right disc and macula show a blurred disc margin with nonhyperemic vessels. Blurring of
the central optic nerve is consistent with disc edema noted on stereophotos. (B) Preinjection or “control” photos are performed with filters in
place, prior to any injection of fluorescein. This allowed for the identification of optic nerve drusen, which autofluoresce.

Our experience indicates that change is required approximately


every 5 years.

Transmitted fluorescence (pigment epithelial


window defect)
This fluorescence is an accentuation of the visibility of
normal choroidal fluorescence. Transmitted fluorescence
occurs when fluorescence from the choroidal vasculature
appears to be increased because of the absence of pigment
in the pigment epithelium, which normally forms a visual
barrier to choroidal fluorescence. The major cause of pigment
epithelial window defect is atrophy of the pigment epithe-
lium (Figs 1.33–1.36).
When the pigment epithelium is dense, choroidal fluores-
cence is not clearly visible because the pigment blocks the
view of the choroid and acts as a barrier to fluorescein. The
density of the pigment determines the degree to which trans-
mission of the normal choroidal fluorescence is blocked. The
visibility of choroidal fluorescence is inversely proportional Fig. 1.33 Pigment epithelial window defect. This schematic of the
to the concentration of pigment in the pigment epithelium. retina shows that the pigment epithelium in the center of the section is
less pigmented than the normal pigment epithelium. This allows the
If the pigment epithelium contains less than the normal amount normal choroidal and choriocapillaris fluorescence to show through;
of pigment or is defective, the choriocapillaris appears to that is, this pathologic condition would create a typical pigment
fluoresce more brightly. The presence of hyperfluorescence epithelial window defect.
caused by a defect in the pigment epithelium depends on
the state of both the pigment epithelium and the choriocapil-
laris. The choriocapillaris must be intact for a depigmented 2. It increases in intensity as dye concentration increases in the
area of the pigment epithelium to be apparent. If the cho- choroid.
riocapillaris does not fill, a depigmented area of the pigment 3. It does not increase in size or shape during the later phases
epithelium does not fluoresce. of angiography.
Transmitted fluorescence has the following four basic 4. It tends to fade and sometimes disappear as the choroid
characteristics: empties of dye at the end of angiography.

1. It appears early in angiography, coincidental with choroidal In short, transmitted fluorescence appears, peaks early, and
filling. fades late without changing size or shape, as would any normal
30
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B C

Fig. 1.34 An eye with drusen demonstrating pigment epithelial window defects. (A) Color photograph: right macula shows multiple drusen
temporally. (B) Late arteriovenous phase of fluorescein angiogram shows marked hyperfluorescence in the areas of the drusen. (C) Late
recirculation phase of fluorescein angiogram shows fading of fluorescence. Comment: Note the degree of fluorescence of the entire fundus
vasculature. This is typical of a pigment epithelial window defect, which is a type of vascular fluorescence. The drusen allow a better view to the
choriocapillaris because of the thinning of the pigment epithelium overlying them.

A B

Fig. 1.35 Pigment epithelial window defect: choroidal folds. (A) Montage color photograph of right disc and macula. Note the pale lines (choroidal
folds) scattered throughout the posterior pole. (B) Arteriovenous phase of fluorescein angiogram of the disc and macula. Hyperfluorescent lines
correspond to the folds, and adjacent hypofluorescent lines are present throughout the macula and surrounding the disc. Comment: This patient
had pigment epithelial folds caused by prolonged hypotony from a filtering bleb. The hyperfluorescent lines are thought to be the hills of the
folds, in the apices of which the pigment epithelium is thinned, allowing hyperfluorescence in the early phases of the fluorescein angiogram
(pigment epithelial window defect). The dark lines are thought to be the valleys of the folds, with an increase in pigmentation causing blockage
of choroidal fluorescence. The later phases of fluorescein angiograms often show fading of fluorescence. Choroidal folds represent a type of
pigment epithelial window defect with early vascular fluorescence and late fading of fluorescence.

vascular fluorescence. When pigment epithelial depigmentation disc, or at the level of the choroid. Normal and abnormal retinal
is extensive, late fluorescein staining of the choroid and sclera and disc vessels are clearly visible on the angiogram because no
may be visible, although it is less intense than the fluorescence barrier obscures them from view. Gross abnormalities of the
of the window defect. retinal and disc vasculature and subtle microvascular changes
that cannot be appreciated adequately by ophthalmoscopic
Abnormal retinal and disc vessels examination will be well defined and easily distinguished
Abnormal vascular fluorescence occurs when abnormal vessels by fluorescein angiography. These changes in the retinal
are present. Such pathologic vessels may be in the retina, on the vasculature can be classified into six morphologic categories:
31

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A
B

C D

Fig. 1.36 Pigment epithelial window defect: macular hole. (A) Schematic drawing of macula showing loss of entire central foveal tissue. (B) Color
photograph of the left macula. This patient has a macular hole. Note a corona of lighter detached, swollen retinal tissue surrounding the foveal
center where the hole is located. (C) Late phase of fluorescein angiogram shows hyperfluorescence within the macular hole. (D) Later phase of the
fluorescein angiogram shows some fading of the hyperfluorescence within the macular hole. Comment: The choriocapillaris was intact. Therefore
the angiogram showed normal fluorescence of the choriocapillaris (early hyperfluorescence within the center of the fovea) and fading in the late
phase of the angiogram.

(1) tortuosity and dilation (Figs 1.37 and 1.38); (2) telangiectasis degree of the distinct pathologic process, and understanding the
(Figs 1.39 and 1.40); (3) neovascularization (Fig. 1.41); (4) anas- pathophysiology of retinal vascular disease.
tomosis (Fig. 1.38); (5) aneurysms (Figs 1.38 and 1.39); and (6) Abnormal choroidal vessels
tumor vessels (Figs 1.42 and 1.43). Abnormal vessels that may be present under the retina and
These aforementioned changes can be viewed in the early originate from the choroid are subretinal neovascularization and
(vascular) phases of angiography. Later, as the vessels empty, vessels within a tumor. When subretinal neovascularization is
some of these vascular abnormalities leak fluorescein, whereas present, the early angiogram often shows a lacy, irregular, and
others do not. nodular hyperfluorescence (Figs 1.44 and 1.45). With a choroidal
Vascular abnormalities of the retina and disc are readily tumor, the abnormal hyperfluorescence is a similar, early
apparent on the fluorescein angiogram. The changes are charac- vascular-type fluorescence, although it may be coarser, as seen
terized by early vascular-appearing hyperfluorescence. Each of in choroidal hemangioma (Fig. 1.46) and malignant melanoma
the six morphologic types indicates specific disease processes (Fig. 1.47).
that aid the clinician in making a diagnosis, determining the Text continued on page p. 38
32
Section I

A B C
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.37 Abnormal retinal vessels, tortuosity, and dilation: internal limiting membrane contraction. (A) Color photograph of right macula shows a
pale membrane overlying the right macula, producing contraction of the retina and tortuosity of the retinal vessels. (B) Arteriovenous phase of
fluorescein angiogram shows marked irregularity and tortuosity of the retinal vessels in association with the preretinal membrane (macular
pucker). (C) Late phase of fluorescein angiogram shows a small amount of vascular leakage due to contraction of the membrane and pulling on
the retinal vessels. Comment: This is tortuosity and dilation, a type of abnormal retinal vascular fluorescence. It is caused by the mechanical
traction of an epiretinal membrane.

A B

Fig. 1.38 Abnormal retinal vascular fluorescence: retinal vascular microaneurysms, telangiectasis, and anastomoses. (A) Color photography of
right eye shows numerous telangiectatic retinal vessels due to a superotemporal branch-vein occlusion. (B) Arteriovenous-phase fluorescein
angiogram shows multiple areas of smaller and larger microaneurysms and telangiectasis. Several small venous–venous anastomoses can be
seen just temporal to the macula. The venous system of the occluded area has collateralized with patent vessels in uninvolved areas.
33

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

C D

Fig. 1.39 Retinal telangiectasis and microaneurysms secondary to diabetic retinopathy. (A) Color photography of right macula showing retinal
exudate, retinal striae, and irregularly dilated retinal vessels (telangiectasis). (B) Arteriovenous-phase fluorescein angiogram shows extensive
hyperfluorescence from the numerous microaneurysms, and telangiectatic retinal vessels. (C) Later arteriovenous-phase fluorescein angiogram
of right macula showing leakage from many of these vessels. (D) Late-phase fluorescein angiogram of right macula shows multiple circular areas
of hyperfluorescence due to accumulation of dye in extensive cystoid spaces. Comment: This patient had significant retinal microvascular
changes due to diabetic retinopathy.
34
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 1.40 Abnormal retinal vessels: telangiectasis. (A) Color montage photograph demonstrating severe areas of exudation, as well as dilated
and telangiectatic vessels. The retina is very edematous. (B) Arteriovenous phase of fluorescein angiogram shows marked irregularity of the
retinal vasculature. There are areas of capillary nonperfusion, telangiectasis, and tortuosity. Comment: This patient had Coats disease with
a markedly abnormal retinal capillary bed, including telangiectasis and dilated vessels.

A B

Fig. 1.41 Abnormal retinal vessels: retinal neovascularization due to proliferative diabetic retinopathy. (A) Montage color photograph of the
posterior pole of the right eye. Extensive irregular tortuous vessels extend from the optic nerve along the vascular arcades and nasally. These
vessels lie on the surface of the retina. (B) Later arteriovenous phase of fluorescein angiogram montage shows increasing hyperfluorescence of
the retinal neovascularization. Comment: This patient had severe proliferative diabetic retinopathy with extensive neovascularization of the right
disc. The vessels fluoresced early (vascular fluorescence) and leaked late. This is very typical of retinal or disc neovascularization.
35

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

C D

Fig. 1.42 Abnormal retinal vessels: tumor–retinal angioma as part of von Hippel’s disease. (A) Color photograph of right macula and disc shows
exudate temporal and inferior to the disc. A very vascular, slightly elevated mass was noted on the temporal border of the disc. Ophthalmoscopy
showed that it has a reddish appearance. A large full-thickness macular hole is also observed. (B) Early arterial phase of the fluorescein
angiogram shows marked fluorescence of the mass. (C) Midarteriovenous phase of the fluorescein angiogram shows an increased fluorescence
of the mass. (D) Late phase of the fluorescein angiogram shows leakage of fluorescein within the mass. Comment: This patient had a peripapillary
retinal angioma. It was very vascular and showed early fluorescence and extensive late leakage.

A B

Fig. 1.43 Arteriovenous malformation: Wyburn–Mason type. (A) Color montage photograph of right macula and temporal retina showing
enlarged, dilated retinal artery, with direct connection to an engorged draining vein. There is no intervening capillary bed. (B) Fluorescein
angiogram showing marked hyperfluorescence of the abnormal, dilated retinal artery and vein. Two smaller arteriovenous malformations appear
to be present, one just above the macula, and the other just below.
36
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A
B

C D

Fig. 1.44 Abnormal choroidal vessels: subretinal neovascularization. (A) Schematic view of the retina shows a small break in Bruch’s membrane,
with a fine proliferation of capillaries through the break dissecting under and lifting up the pigment epithelium. There is a shallow sensory retinal
detachment. (B) Color photograph of the left macula. There is a dirty-gray membrane involving the central macula. Note the small area of
subretinal hemorrhage. There is a shallow sensory retinal detachment. (C) The arteriovenous phase of fluorescein angiogram shows fine, lacy,
irregular hyperfluorescence corresponding to a small, fine patch of subretinal neovascularization. (D) Late phase of fluorescein angiogram shows
leakage of these vessels into the subpigment epithelial and subretinal spaces. Comment: This patient had a small patch of subretinal
neovascularization involving the central fovea. The angiogram shows typical, early vascular fluorescence (in a nodular, irregular, lacelike fashion)
and late hyperfluorescent leakage.
37

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A

C D

Fig. 1.45 Abnormal choroidal vessels: subretinal neovascularization. (A) Schematic drawing of retina shows vascular proliferation from the
choriocapillaris dissecting under the pigment epithelium, with associated fibrous tissue. The pigment epithelium has become thinned and the
sensory retina detached. The outer plexiform layer of the sensory retina shows cystic spaces. (B) Red-free photograph of left macula shows
some hemorrhage and exudate. On the color photograph and slit-lamp biomicroscopy, a dirty-gray membrane was noted in the inferotemporal
portion of the macula. This is seen as a slightly pale lesion in the inferotemporal macula. (C) Early arteriovenous phase of fluorescein angiogram
shows a lacy, irregular, nodular area of hyperfluorescence in the inferotemporal macula. This is a flat patch of vessels that has proliferated from
the choriocapillaris under the pigment epithelium. (D) Late phase of the fluorescein angiogram shows leakage from the patch of subretinal
neovascularization. Most of the fluorescence is pooling of fluorescein under the sensory retinal detachment, although there is some cystic
change in the fovea. Comment: This patient had a patch of subretinal neovascularization that was nearly 4 disc diameters in size. It fluoresced
early with the vascular phase of the angiogram (typical for subretinal neovascularization) and leaked late. Actually, “subretinal neovascularization”
is a misnomer because the new vessels are initially located in the subpigment epithelial space.
38
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.46 Abnormal choroidal vessels in a patient with choroidal


hemangioma. (A) Color photos of left macula and disc with elevated
choroidal hemangioma. (B) Arteriovenous phase of the fluorescein
angiogram shows prominent hyperfluorescence in this area
demonstrating the tumor vessels. (C) Late phase of the fluorescein
angiogram shows marked leakage in this area. Comment: This patient
had a choroidal hemangioma, which is a very vascularized choroidal
C mass. The vascularity in this mass causes the marked
hyperfluorescence and leakage.

A B C

Fig. 1.47 Abnormal choroidal vascular fluorescence due to malignant melanoma. (A) Color photograph of left eye. Note the darkly pigmented
mass nasal to the optic nerve. There is some orange lipofuscin pigment overlying the surface of this as well. (B) Arteriovenous phase of
fluorescein angiogram of the mass shows hyperfluorescence over the surface of the tumor. This patient also had some macular drusen, which
show some early hyperfluorescence in the macula. (C) Late phase of the fluorescein angiogram shows leakage from the mass. There are
multiple “hot spots” overlying the tumor. Comment: This patient had a choroidal malignant melanoma. This was a medium-sized tumor that
showed the typical early fluorescence that is seen in a medium-sized melanoma.

Leak Four types of late extravascular hyperfluorescent leakage


Fluorescence of the retinal and choroidal vessels begins to dimin- occur in the normal eye: (1) fluorescence of the disc margins from
ish about 40–60 seconds after injection. Fluorescein empties the surrounding choriocapillaris; (2) fluorescence of the lamina
almost completely from the retinal and choroidal vasculature cribrosa; (3) fluorescence of the sclera at the disc margin if the
about 10–15 minutes after injection. Any fluorescence that retinal pigment epithelium terminates away from the disc, as in
remains in the fundus after the retinal and choroidal vessels an optic crescent; and (4) fluorescence of the sclera when the
have emptied of fluorescein is extravascular fluorescence and pigment epithelium is lightly pigmented. These are the only
represents leakage. forms of late hyperfluorescence or leakage that can be
considered “normal.” Any other hyperfluorescence observed 15 cribrosa and the surrounding margins of the disc (from the nor-
minutes after the fluorescein injection represents extravascular mally leaking peripapillary choriocapillaries). The difference
fluorescein and is referred to as leakage. between normal and abnormal leakage at the disc may be subtle. 39
Either or both of the two vascular systems of the fundus Papilledema and optic disc edema
can produce abnormal late hyperfluorescence (leakage) if defects Papilledema is swelling of the optic nerve head as a result of

Chapter 1
are present in their respective barriers to fluorescein. The barrier increased intracranial pressure. Edema of the optic disc is defined
to fluorescein leakage from the retinal vessels is the retinal as swelling of the optic nerve head secondary to local or systemic
vascular endothelium. The barrier to leakage from the choroidal causes (Fig. 1.48). The angiogram is similar in each case, demon-
circulation is the pigment epithelium. An abnormality of the strating leakage associated with swelling of the optic nerve head.
retinal vascular endothelium can result in permeability to fluo- In the early phases of the angiogram, dilation of the capillaries
rescein and leakage of fluorescein into the retinal tissue. Simi- on the optic nerve head may be seen; in the late angiogram, the
larly, an abnormality of the pigment epithelium can result in

Fluorescein Angiography: Basic Principles and Interpretation


dilated vessels leak, resulting in a fuzzy fluorescence of the disc
permeability to fluorescein, and fluorescein will leak from the margin.
choroidal tissue through the pigment epithelium. Abnormal
Retinal leak
late hyperfluorescence of the choroid, however, can occur
In the late stages of the normal angiogram, the retinal vessels
without damage to the pigment epithelium, as in cellular infil-
have emptied of fluorescein and the retina is dark. Any late
trates of the choroid that occur in choroidal inflammation or
retinal hyperfluorescence is abnormal and indicates leakage of
tumor.
retinal vessels. When the leakage is severe, the extracellular fluid
There are two other types of late abnormal fluorescence: one
may flow into cystic pockets, and the angiogram shows fluores-
occurs when fluorescein enters the vitreous, and the other when
cence of the cystic spaces. Fluorescein flows out of the patent
fluorescein leaks into the optic nerve head.
retinal vessels to lie in pools in the cystoid spaces or stains the
Vitreous leak edematous (noncystic) retinal tissue. Cystoid retinal edema is
Leakage of fluorescein into the vitreous creates a diffuse, white apparent as the fluorescein pools in small loculated pockets. In
haze in the late phase of the fluorescein angiogram. In some the macula, cystoid edema takes on a stellate appearance (Fig.
instances the haze is generalized and evenly dispersed, and in 1.49); elsewhere in the retina, it has a honeycombed appearance
other cases the white haze is localized. (Fig. 1.50). Fluorescent staining of noncystoid edema is diffuse,
Leakage of fluorescein into the vitreous is due to three irregular, and not confined to well-demarcated spaces (Figs 1.51
major causes: (l) neovascularization growing from the retinal and 1.52).
vessels on to the surface of the retina or disc or into the The amount of fluorescein leakage depends on the dysfunction
vitreous cavity; (2) intraocular inflammation; and (3) intraocular of the retinal vascular endothelium (Fig. 1.52). When leakage is
tumors. not pronounced, the cystoid spaces fill slowly and become visible
Vitreous hyperfluorescence secondary to retinal neovascular- only late in angiography. When this occurs, the area of cystoid
ization is usually localized and appears as a cotton-ball type of retinal edema may be somewhat hypofluorescent early in the
fluorescence surrounding the neovascularization (Fig. 1.41B). angiogram because the fluid in these spaces acts as a barrier and
The vitreous fluorescence secondary to intraocular inflammation blocks the underlying choroidal fluorescence. When there is
is often generalized, giving a diffuse, white haze to the vitreous heavy fluorescein leakage, the cystoid spaces fill rapidly, in some
because of generalized leakage of fluorescein from the iris and cases within a minute after injection. The large confluent cysts
ciliary body. The vitreous fluorescence secondary to tumors is seen with severe cystoid macular edema may fill late in the angio-
most often localized over the tumor. gram. The large retinal vessels can also leak. This is called perivas-
Disc leak cular staining and is seen in three distinct situations: inflammation
The optic nerve head normally has some fluorescein leakage (indicating a perivasculitis), traction (severe pulling on a large
(late hyperfluorescence) as a result of staining of the lamina retinal vessel, Fig. 1.52), and occlusion. When a large retinal

A B C

Fig. 1.48 Disc leakage. (A) Color photograph of right optic nerve. Note the dilation of the disc capillaries. (B) The arteriovenous-phase angiogram
of the right disc and macula shows the hyperfluorescence due to these dilated disc capillaries. (C) The late phase of the angiogram shows
significant leakage from these dilated optic disc capillaries. Comment: This patient had a papillopathy related to diabetes. This produced
significant dilation of the disc capillaries. The leakage from this abnormal disc is quite obvious.
40
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

C D

Fig. 1.49 Retinal leak: cystoid macular edema. (A) Schematic drawing of the macula shows large cystic spaces in the outer plexiform layer.
There are some cystic spaces in the inner nuclear layer. (B) Color photograph of left macula. Careful inspection of the retina is often necessary
on biomicroscopy to detect intraretinal cystoid. (C) Arteriovenous phase of fluorescein angiogram shows some dilation of the fine capillary
network around the fovea. (D) Late phase of fluorescein angiogram shows hyperfluorescence from the accumulation of dye filling the cystic
spaces. Note the stellate appearance of the cystoid macular edema. Comment: This patient had late hyperfluorescence (i.e., leakage) into the
retina that was severe enough to create cystic spaces. This is a typical example of cystoid macular edema.
41

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B C

Fig. 1.50 Retinal leakage: cystoid retinal edema. (A) Color photograph of the right macula. Large central cystoid cavity is seen corresponding
to fovea. (B) Arteriovenous phase of fluorescein angiogram shows well-defined telangiectatic retinal vessels. (C) Late phase of fluorescein
angiogram shows leakage from these vessels. In the center of the macula, the leakage is in stellate cystic pockets, and just outside the macula,
temporally, the leakage has taken a honeycomb form. Comment: This patient had leakage of telangiectatic vessels into the retina, and the
leakage formed cystoid spaces. Cystoid edema in the center of the macula takes on a stellate form because of the oblique nature of the outer
plexiform layer. The cystic spaces take on a honeycomb form in nonmacular areas of the retina because of the perpendicular nature of the fibers
of the outer plexiform layer.

A B C

Fig. 1.51 Retinal leakage, severe noncystoid edema. Branch-vein occlusion. (A) Color photograph of right macula shows multiple retinal
hemorrhages inferotemporally due to a retinal branch-vein occlusion. (B) Arteriovenous phase of fluorescein angiogram shows the vascular
abnormalities associated with the branch-vein occlusion. Hypofluorescence corresponds to areas previously treated with grid pattern laser
photocoagulation. (C) Late phase of fluorescein angiogram shows diffuse leakage of the fluorescein dye. Comment: This patient had generalized
leakage of the retinal vascular bed in the distribution of the blocked branch vein. The leakage was not yet severe enough, however, to form
clearly defined cystic spaces. Late hyperfluorescence indicates leakage, and this fluorescence is located in the retina; thus this was retinal
edema.

A B C

Fig. 1.52 Late hyperfluorescence, retinal leakage: severe epiretinal membrane contraction. (A) Color photograph of right macula showing thick
epiretinal membrane overlying the macula and producing severe traction and contraction of the retina and vessels. (B) Arteriovenous phase of
fluorescein angiogram shows that the retinal vasculature is tortuous and irregular. (C) Late arteriovenous phase of fluorescein angiogram shows
leakage from the retinal vessels. Comment: The marked preretinal membrane caused sufficient traction on the retina, resulting in marked retinal
vascular leakage.
42
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 1.53 Retinal leakage: perivascular staining. (A) In this late arteriovenous-phase fluorescein angiogram, note the beading of the large retinal
veins. There is also associated leakage from these vessels. (B) Later phase of fluorescein angiogram shows perivascular staining (leakage) from
the large retinal vessels that are traversing large zones of capillary nonperfusion. Comment: Typically, when a large retinal vessel (artery or vein)
is perfused but traverses an area of capillary nonperfusion, ischemic retinal factors will act adversely on the endothelium of the large vessel and
cause it to leak. This is called perivascular staining. Perivascular staining also occurs with traction or inflammation.

vessel leak is partially occluded, or when it traverses an area of The differences in the adherence and the angle of detachment
occlusion (and capillary nonperfusion), it will leak (Fig. 1.53). between a sensory retinal detachment and a pigment epithelial
detachment result in specific differences in fluorescent pooling
Choroidal leak patterns. The hyperfluorescent pooling of a sensory retinal
Late hyperfluorescence under the retina can be classified as detachment tends to fade gradually toward the site where the
either pooling or staining (Fig. 1.54). Pooling is defined as sensory retina is attached. This makes fluorescein angiographic
leakage of fluorescein into a distinct anatomic space; staining is determination of the extent of a sensory retinal detachment dif-
leakage of fluorescein diffused into tissue. ficult. In contrast, the hyperfluorescent pooling under a pigment
Fluorescein pools in the spaces created by detachment of the epithelial detachment extends to the edges of the detachment,
sensory retina from the pigment epithelium or in the space making the entire detachment and its margins hyperfluorescent
created by detachment of the pigment epithelium from Bruch’s and clearly discernible.
membrane. The posterior layer of the sensory retina is made up Pooling of fluorescein under a sensory retinal detachment in
of rods and cones that are loosely attached to the pigment epi- central serous retinopathy takes place slowly, since the dye
thelium. When a sensory retinal detachment occurs, the detached passes through one or more points of leakage in the defective
segment separates with little force, forming a very gradual angle pigment epithelium (Fig. 1.54). When leakage comes from sub-
at the point of attachment to the pigment epithelium. Because of retinal neovascularization (Fig. 1.55) or a tumor (Fig. 1.47), it is
this narrow angle, the exact limits of a sensory retinal detach- more rapid and complete. When the pigment epithelium is
ment are difficult to locate ophthalmoscopically or by slit-lamp detached from Bruch’s membrane, fluorescein passes freely and
biomicroscopy. rapidly through Bruch’s membrane from the choriocapillaris
Depending on the specific disease, the late angiogram may or into the subpigment epithelial space (Fig. 1.56).
may not portray the full fluorescent filling of the subretinal fluid. In some cases of central serous chorioretinopathy, there is an
For example, in central serous chorioretinopathy the leakage is associated pigment epithelial detachment, and pooling under
gradual, and fluorescence of the subsensory retinal fluid will not each (sensory retinal detachment and the pigment epithelial
be complete. In other conditions, such as subretinal neovascu- detachment) is evident. Occasionally, the edge of a pigment
larization, fluorescein leakage is profuse, and the subsensory epithelial detachment may tear, or rip, and allow fluorescein dye
fluid often completely fluoresces (Fig. 1.55). to pass freely into the subretinal space (Fig. 1.57). Drusen may
In contrast to the attachment of the sensory retina, the base- also show late hyperfluorescence similar to that seen with a
ment membrane of the pigment epithelium adheres firmly to pigment epithelial detachment (Fig. 1.58). In some cases of
the collagenous fibers of Bruch’s membrane. The firm adhesion pigment epithelial detachment, especially in older patients, sub-
and wide angle of detachment make it easy to discern a retinal neovascularization is also present. This combination of
pigment epithelial detachment ophthalmoscopically. Occasion- subretinal neovascularization and pigment epithelial detach-
ally a light-orange ring appears around the periphery of a ment results in an interesting angiogram that can be challenging
pigment epithelial detachment, further facilitating identification to interpret (Fig. 1.59).
(Fig. 1.56). Text continued on page p. 49
43

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A B

C D

Fig. 1.54 Late hyperfluorescence, subretinal pooling: central serous chorioretinopathy. (A) Schematic drawing of retina shows a sensory retinal
detachment. There is a break in the pigment epithelium. Fluorescein flows from the choriocapillaris through Bruch’s membrane, through the
break in the pigment epithelium, and into the subretinal space, under the detached retina. (B) Color photograph of left macula shows a shallow
sensory detachment (arrows). Just superonasal to the fovea is a small white area with a gray center. The fluorescein angiogram will reveal that
this is the area of the leak. (C) Arteriovenous phase of fluorescein angiogram shows a hyperfluorescent spot that was seen on
stereoangiography to be leakage of fluorescein coming from the pigment epithelium. (D) Late phase of fluorescein angiogram shows that the
spot of pigment epithelial leakage has enlarged and become fuzzy. This is the release of fluorescein molecules into the fluid under the detached
sensory retina. Comment: This patient had central serous chorioretinopathy. There was a break in the pigment epithelium that allowed leakage of
fluorescein through it and into the subretinal space. Late hyperfluorescence means leakage, and in this case, there is pooling of fluorescein
under the detached retina.
44
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

C D

Fig. 1.55 Late hyperfluorescence, leakage, and pooling under the sensory retina caused by subretinal neovascularization, resulting in a sensory
retinal detachment. (A) Schematic of the retina showing that it has detached (photoreceptors are separated from pigment epithelium). Vessels
have proliferated from the choriocapillaris through Bruch’s membrane. There is a fibrovascular scar involving the pigment epithelium. The
sensory retina is detached. (B) Color photograph of left macula shows a pale gray lesion in the inferior portion of the macula with some
associated hemorrhage. (C) Arteriovenous phase of fluorescein angiogram shows a patch of subretinal neovascularization inferior to the fovea;
this is evidenced by the lacy, irregular hyperfluorescence in this area. (D) Late phase of fluorescein angiogram shows fuzzy fluorescence. There
is pooling of fluorescein under the detached retina and some staining of the fibrous tissue associated with the subretinal neovascularization.
Comment: This patient had a patch of subretinal neovascularization with a great deal of leakage, causing a sensory detachment. The
early-phase angiogram showed the vascular nature of the lesion, and the late-phase angiogram showed the leakage and pooling in
the subretinal space.
45

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A
B

C D

Fig. 1.56 Late hyperfluorescent pooling under the retinal pigment epithelium (pigment epithelial detachment). (A) Schematic diagram illustrating
detachment and elevation of the pigment epithelium; the pigment epithelium is separated from Bruch’s membrane. Because the attachment of
the pigment epithelium to Bruch’s membrane is quite firm, the angle of detachment is quite large. (B) Color photography of right macula shows a
round detachment of the pigment epithelium. (C) Early arteriovenous phase of fluorescein angiogram shows early fluorescence from the area of
detachment pigment epithelium. (D) Late-phase angiogram of right macula shows well-demarcated hyperfluorescent pooling of fluorescein under
the detached pigment epithelium. Comment: Fluorescein flows freely through Bruch’s membrane and stops at the pigment epithelium. When the
pigment epithelium is detached, the fluorescein flows right through Bruch’s membrane into the space made by the detached pigment epithelium.
Therefore a pigment epithelial detachment fluoresces evenly and slowly (like a light bulb on a rheostat) and shows intense hyperfluorescent
pooling that is well demarcated (indicating its well-defined angle of attachment) late in the angiogram.
46
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A
B

C D

Fig. 1.57 Late hyperfluorescence under the retina – leakage from the choroid due to a retinal pigment epithelial (RPE) rip. (A) Schematic of a
pigment epithelial detachment that has developed a tear along one edge. The barrier function of the pigment epithelium is lost and fluorescein
dye can diffuse easily and rapidly in the subretinal space. (B) Color photography of right macula showing a round dark area under the fovea,
and light (depigmented area) area extending temporally. In the inferior portion of the macula, some subretinal hemorrhage is seen. (C) Early
arteriovenous-phase fluorescein angiogram shows bright hyperfluorescence of the depigmented area temporally, and hypofluorescence under the
fovea as well as inferiorly in the area of the subretinal blood. (D) Late-phase fluorescein angiogram shows pooling of fluorescein under the retina
where the dye has been able to diffuse freely through Bruch’s membrane in the sensory retinal detachment. Comment: This patient developed a
tear of the pigment epithelial detachment. The dark area under the fovea is where the pigment epithelium has rolled up after tearing away from
the area temporally. The area temporal to the macula appears light due to absence of the RPE in this area. Since the RPE barrier is absent in
this area, the dye diffuses readily and rapidly into overlying sensory retinal detachment, producing late pooling of fluorescein.
47

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
A
B

C D

Fig. 1.58 Late hyperfluorescent pooling (or staining) of large drusen. (A) Schematic section of retina shows progressively larger detachments
of pigment epithelium. Drusen deposit between the pigment epithelium and Bruch’s membrane and lift the pigment epithelium up, forming
small or large pigment epithelial detachments, depending on the size of the drusen. (B) Color photograph of right macula shows multiple, pale,
round, and variably sized drusen. (C) Arteriovenous phase of fluorescein angiogram shows some early hyperfluorescence of the areas of
the drusen. (D) Late phase of fluorescein angiogram shows marked hyperfluorescence of the drusen. The larger drusen take longer for the
hyperfluorescence to develop. Comment: The larger the drusen, the more similar they are to pigment epithelial detachments, and therefore the
more likely it is that they will show pooling of fluorescein (or staining of the drusen material).
48
Section I
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 1.59 Pigment epithelial detachment with associated (suspicious)


subretinal neovascularization. (A) Color photograph of right macula.
Note the pigment epithelial detachment temporally and sensory retinal
detachment. (B) Arteriovenous-phase fluorescein angiogram shows
early hyperfluorescence of the superotemporal pigment epithelial
detachment. (C) Late-phase fluorescein angiogram of left macula
shows that the fluorescence of the pigment epithelial detachment
temporally has increased significantly. Comment: This patient had an
irregularly shaped pigment epithelial detachment, which is a sign of
C possible choroidal neovascularization. The irregular, fuzzy
hyperfluorescence is due to likely occult choroidal neovascularization.
In summary, late hyperfluorescence beneath the retina should likely they will retain fluorescein and staining will occur. When
first be distinguished as pooling of fluorescein into a space or as drusen are large and have smooth edges, the late staining on the
tissue staining with fluorescein. When pooling is present, one angiogram is similar in appearance to that of pooling of fluores- 49
must determine whether a sensory retinal or a pigment epithelial cein under a pigment epithelial detachment. In many cases it is
detachment is present. Similarly, if staining is present, one must difficult, if not impossible, to differentiate large drusen from

Chapter 1
find out whether the tissue involved is the retinal pigment epi- small pigment epithelial detachments: they have a similar oph-
thelium and Bruch’s membrane, choroid, or sclera. From this thalmoscopic, fluorescein angiographic, and even microscopic
anatomic determination a more specific diagnosis can be appearance.
determined. Scar
Staining Scar tissue retains fluorescein and usually demonstrates well-
Staining refers to leakage of fluorescein into tissue or material demarcated hyperfluorescence because little, if any, fluid sur-

Fluorescein Angiography: Basic Principles and Interpretation


and is contrasted with pooling of the fluorescein into an ana- rounds the scar. Later in the healing process, when only a few
tomic space. Many abnormal subretinal structures and materials vessels remain, the early angiogram is hypofluorescent because
can retain fluorescein and demonstrate later hyperfluorescent of the paucity of vessels and blockage by the scar tissue. The
staining. most commonly seen scar tissue is the disciform scar, which is
Drusen the endstage of subretinal neovascularization. Scarring is also
The most common form of staining occurs with drusen. Most seen following numerous other insults to the pigment epithe-
drusen hyperfluoresce early in the angiogram because choroidal lium and choroid, especially inflammation (Fig. 1.60).
fluorescence is transmitted through defects in the pigment epi- Sclera
thelium overlying the drusen (Fig. 1.34). Fluorescence from most In several situations the sclera is visible ophthalmoscopically
small drusen diminishes as the dye leaves the choroidal circula- and exhibits late hyperfluorescent staining on fluorescein angi-
tion. However, some larger drusen display later hyperfluores- ography. Scleral staining is best seen when the retinal pigment
cence or staining (Fig. 1.58). The larger the drusen, the more epithelium is very pale (as in a blonde patient) or when the

Fig. 1.60 Late hyperfluorescence and


leakage – staining in geographic helicoid
peripapillary choroidopathy (GHPC). (A)
Color montage of left disc and macula shows
large geographic areas of atrophy of pigment
epithelium and choriocapillaris. There is
some hyperplasia of the pigment epithelium
noted as hyperpigmentation (especially in the
macula and papillomacular bundle). Some
fibrous scar tissue is present. (B)
Arteriovenous-phase fluorescein angiogram
shows that the geographic lesions are mostly
hypofluorescent; they are caused by loss of
pigment epithelium and choriocapillaris. Note
that the large choroidal vessels can be seen
within these lesions, indicating that the
pigment epithelium and choriocapillaris are
both gone. There is some hyperfluorescence
along the edges of the geographic lesions.
The pigment epithelial hyperplasia causes
blocked fluorescence. (C) Late fluorescein
angiogram of left macula shows
hyperfluorescent staining along the edges of
the geographic lesion. Comment: This patient
A had GHPC; inflammation of choroid and
pigment epithelium resulted in a loss of the
pigment epithelium and choriocapillaris and
some of the choroid. The angiogram showed
that only large choroidal vessels remained
within these lesions. The choriocapillaris was
intact, however, in the normal tissue adjacent
to the geographic atrophic tissue. The normal
choriocapillaris leaked into the atrophic area
in a horizontal fashion, causing late
hyperfluorescence of areas of scar tissue
and some scleral staining.

B C
choriocapillaris is fully intact. When the choriocapillaris is not dye tends to diffuse toward the center of the lesion. The entire
intact, fluorescein staining of the sclera can occur from the edges lesion may not stain if the distance from the edge of the sclera
50 of the atrophic area where fluorescein leaks from the intact cho- is more than 1 mm. When the choriocapillaris is intact or the
riocapillaris inward toward the atrophy (Fig. 1.60). lesion is not expansive, the sclera will stain completely.
In conditions such as physiologically light-colored (blonde) In summary, late hyperfluorescence beneath the retina should
fundus or in myopia, the choriocapillaris is usually sufficient to first be distinguished as pooling of fluorescein into a space or as
Section I

stain the sclera completely. After the choroidal vessels have tissue stained with fluorescein. When pooling is present, it must
emptied of fluorescein in the later phases of angiography, the be determined whether a sensory retinal or a pigment epithe-
large hypofluorescent choroidal vessels appear as dark lines in lium detachment is present. Similarly, if staining is present, it
silhouette against the stained sclera. must be determined whether the tissue involved is the retinal
When a loss of choroid and choriocapillaris has occurred, there pigment epithelium and Bruch’s membrane, choroid, or sclera.
is a consequent diminution of fluorescein flow in the choroid. From this anatomic differentiation, a more specific diagnosis can
Optical Imaging Technologies
Retinal Imaging and Diagnostics

When this occurs, the sclera stains with fluorescein only from be determined.
adjacent normal patent choriocapillaris vasculature. These For online acknowledgments visit http://www.
vessels stain the sclera on the borders of the lesion because the expertconsult.com

Bonus images for this chapter can be found online at http://www.expertconsult.com


Fig. 1.9 The photographer focuses the eyepiece of the camera by physician also indicates the diagnosis because
initially turning the eyepiece counterclockwise, then experienced photographers will know which type of
clockwise, and stopping when it is in exact focus. The photographs to take for each particular diagnosis. The
photographer must be sure that the eyepiece crosshairs physician also indicates in which location of the fundus the
remain in perfect focus throughout the photographic initial-transit phase of the angiogram should take place or,
procedure. in other words, where the photographer should start the
Fig. 1.10 Viewing stereo fundus photographs. (A) Negatives are angiogram. In the lower right portion of the photographic
placed on a viewing back-lit display. Two negative images request form, the physician can indicate to the
are then viewed with an adjustable stereo lens. Reading photographer specifics about the patient that will facilitate
the contact print of the angiogram. The stereo viewer can the photographic process and save the photographer time.
be easily made up using a trial frame. In viewing negative The physician can indicate whether an eye can fixate on
images, “hyperfluorescence” corresponds to dark objects a light, whether the media are clear (so that if the
while corresponding “hypofluorescent” objects are lighter. photographer cannot get a clear view, he or she can
(B) Reading a stereo pair of digital angiograms. The immediately understand that it is caused by a problem of
special viewer allows the observer to focus both images. the eye), and what the patient’s refraction is so that the
The software displayed is Ophthalmic Imaging Systems. photographer can know which special lenses to use in the
Fig. 1.12 An assistant holds the patient’s head as a reminder to the photographic process.
patient to keep the chin in the chinrest and forehead Fig. 1.16 After the needle is placed in the vein, the lights can be
against the bar. turned off so that the photographer can become dark-
Fig. 1.13 Ten percent fluorescein solution, 5 mL syringe, and adapted and see fluorescein flow in the eye. With the use
23-gauge scalp-vein needle. of a hand light, the person injecting can carefully observe
Fig. 1.15 Photographic request form. In the top left portion of the the injection site so as to be sure extravasation is not
form, the physician indicates the number of color occurring. In this way the fluorescein solution can be
photographs required for each area in the fundus. The injected while the room lights are out.

11. Moosbrugger KA, Sheidow TG. Evaluation of the side-effects and image
REFERENCES quality during fluorescein angiography comparing 2 mL and 5 mL sodium
1. Novotny HR, Alvis DL. A method of photographing fluorescence in circulating fluorescein. Can J Ophthalmol 2008;43:571–5.
blood in the human retina. Circulation 1961;24:82–6. 12. Pacurariu RI. Low incidence of side-effects following intravenous fluorescein
2. Rabb MF, Burton TC, Schatz H, et al. Fluorescein angiography of the angiography. Ann Ophthalmol 1982;14:32–6.
fundus: a schematic approach to interpretation. Surv Ophthalmol 1978;22: 13. Lipson BK, Yannuzzi LA. Complications of intravenous fluorescein injections.
387–403. Int Ophthalmol Clin 1989;29:200–5.
3. Schatz H. Letter: flow sheet for the interpretation of the fluorescein angiograms. 14. Lu VH, Ho IV, Lee V, et al. Complications from fluorescein angiography:
Arch Ophthalmol 1976;94:687. a prospective study. Clin Exp Ophthalmol 2009;37:826–7.
4. Gass JD, Sever RJ, Sparks D, et al. A combined technique of fluorescein fun- 15. Zografos L. [International survey on the incidence of severe or fatal complica-
duscopy and angiography of the eye. Arch Ophthalmol 1967;78:455–61. tions which may occur during fluorescein angiography.] J Fr Ophtalmol
5. Haining WM, Lancaster RC. Advanced techniques for fluorescein angiogra- 1983;6:495–506.
phy. Arch Ophthalmol 1968;79:10–5. 16. Chen LJ, Yeh SI. Computer-assisted image processing for a simulated stereo
6. Novotny HR, Alvis D. A method of photographing fluorescence in circulating effect of ocular fundus and fluorescein angiography photographs. Ophthalmic
blood of the human eye. Tech Doc Rep SAMTDR USAF Sch Aerosp Med Surg Lasers Imaging 2010;41:293–300.
1960;60-82:1–4. 17. Watzke RC, Klein ML, Hiner CJ, et al. A comparison of stereoscopic fluorescein
7. Gass JD. Atlas of macular diseases: diagnosis and treatment. St Louis: Mosby; angiography with indocyanine green videoangiography in age-related macular
1970. degeneration. Ophthalmology 2000;107:1601–6.
8. Yannuzzi LA, Ober MD, Slakter JS, et al. Ophthalmic fundus imaging: today 18. Patz A, Finkelstein D, Fine SL, et al. The role of fluorescein angiography in
and beyond. Am J Ophthalmol 2004;137:511–24. national collaborative studies. Ophthalmology 1986;93:1466–70.
9. Friberg TR, Gupta A, Yu J, et al. Ultrawide angle fluorescein angiographic 19. Arnold AC, Hepler RS. Fluorescein angiography in acute nonarteritic anterior
imaging: a comparison to conventional digital acquisition systems. Ophthalmic ischemic optic neuropathy. Am J Ophthalmol 1994;117:222–30.
Surg Lasers Imaging 2008;39:304–11. 20. Segato T, Piermarocchi S, Midena E. The role of fluorescein angiography in the
10. Manivannan A, Plskova J, Farrow A, et al. Ultra-wide-field fluorescein angiog- interpretation of optic nerve head diseases. Metab Pediatr Syst Ophthalmol
raphy of the ocular fundus. Am J Ophthalmol 2005;140:525–7. 1990;13:111–4.
ACKNOWLEDGMENTS
This work was supported by the San Francisco Retina Research 50.e1
Fund. The authors would like to thank Sean Grout, head of
photography at West Coast Retina Medical Group, for his
assistance in acquiring many of the images used in this text.

Chapter 1
Fluorescein Angiography: Basic Principles and Interpretation
Optical Imaging Technologies Section 1

Clinical Applications of Diagnostic Indocyanine Chapter

Green Angiography
Giovanni Staurenghi, Ferdinando Bottoni, Andrea Giani 2 
INTRODUCTION film initially restricted its angiographic application. The resolu-
tion of ICGA was improved in the mid-1980s by Hayashi and de
Intravenous fluorescein angiography provides excellent spatial Laey, who developed improved filter combinations with suffi-
and temporal resolution of the retinal circulation, with a high cient sensitivity for near-infrared wavelengths.13 They were also
degree of fluorescence efficiency and minimal penetration of the instrumental in the transition from still-frame to dynamic
retinal pigment epithelium (RPE). Unfortunately, imaging of the imaging by introducing videoangiography.14,15
choroidal circulation is prevented by secondary poor transmis- Although the sensitivity of the initial video camera system was
sion of fluorescence through ocular media opacities, pathologic a vast improvement over previous techniques, its inability to
manifestations such as serosanguineous fluid or lipid exudation, study individual images and the potential light toxicity using a
and fundus pigmentation, including that from the RPE layer. 300-W halogen bulb restricted the duration and quality of the
In medicine, a core principle for diagnostic imaging is the technique. In 1989, Destro and Puliafito16 performed ICGA using
selection of a technique that best visualizes the disease undergo- a system very similar to that described by Hayashi. In the same
ing investigation. Indocyanine green (ICG) has several advan- year, the use of the SLO for ICG videoangiography was intro-
tages over sodium fluorescein in imaging the choroidal duced by Scheider and Schroedel.17 In 1992, Guyer et al. intro-
vasculature. Its physical characteristics allow for visualization of duced the use of a 1024 × 1024-line digital imaging system to
the dye through overlying melanin, xanthophyll pigment, sero- produce high-resolution ICGA.18 However, this system lacked
sanguineous fluid, or lipid exudates. The use of high-resolution flash synchronization with the video camera.
infrared digital fundus cameras and confocal scanning laser oph- Finally, Yannuzzi and coworkers described a 1024-line resolu-
thalmoscopes (SLO), specifically designed for ICG angiography tion system, which was synthesized with the appropriate flash
(ICGA), has reflected a growing awareness and interest in cho- synchronization and image storage capability, permitting high-
roidal vascular lesions, and has facilitated the rapid diffusion of resolution, long-duration ICGA.19
ICGA in the ophthalmic community.
Even in the era of anti-vascular endothelial growth factor CHEMICAL AND PHARMACOKINETICS
(VEGF) intravitreal injections, a therapy for which accurate
localization of the choroidal neovascular membrane is not as ICG is a tricarbocyanine, anionic dye. Its structural formula is
critical, ICGA, among other imaging techniques, is still extremely 2,2’-indo-6,7,6’,7’-dibenzocarbocyanine sodium salt20 with a
useful in clinical practice.1 molecular weight of 774.96 Da.2 ICG is soluble in highly dis-
tilled water,21 even though in protein-free buffer it is difficult
to obtain stable and simple ICG solutions, because of the for-
HISTORY mation of reversible dimers/polymers.2 Binding to albumin or
ICGA was developed by Kodak Research Laboratories,2 on plasma proteins improves the stability of ICG solutions.2 ICG
request by cardiologists to be used as an indicator of cardiac is supplied with a solvent consisting of sterile water at pH
output which was not influenced by variations in blood oxygen 5.5–6.5. The final product contains 5–9.5% sodium iodine,22 to
saturation.3,4 Hepatologists subsequently began to use ICGA to prevent recrystallization.23
evaluate hepatic blood flow5 and hepatocellular function.6 ICG absorbs light in the near-infrared region of the spectrum.
In 1969, Kogure and Choromokos first used ICGA for studying The maximum absorption is at 790 nm,23 while the maximum
the cerebral circulation in a dog.7 The following year, Kogure emission occurs at approximately 835 nm.24 These optical prop-
et al. reported on intra-arterial ICG absorption of the choroid in erties allow penetration through macular pigment, melanin,25
monkeys.8 The first human ICG angiogram was of the carotid blood, and pigment.
artery.9 In 1971, Hochheimer modified the system for ICGA by About 98% of ICG is bound to plasma protein, in particular to
changing the color film which had previously been used to globulins, such as A1-lipoproteins.22 In pig plasma, lipoprotein
black-and-white infrared film.10 HDL3 is the major binding protein.2 ICG is excreted by the
In 1972, Flower and Hochheimer performed the first intrave- liver,5,26 with negligible extrahepatic removal.5,27 The presumed
nous ICGA to image the human choroid.11 In the following years, mechanism is active27 and depends on both liver blood flow and
Flower and coworkers evaluated the potential utility of ICGA in hepatocellular function.28 ICG is excreted into the bile without
the investigation of the normal and pathologic eye.11,12 The rela- metabolic process or enters the enteropathic circulation,27
tively poor fluorescence efficiency of the ICG molecule and its through three steps: (1) uptake over the hepatotocyte sinusoidal
limited ability to produce high-resolution images on infrared (basolateral) membrane (Na+-mediated); (2) passage through the
cell, with some role of the microfilaments and vesicular trans- ICG was extensively used as a chromodiagnostic agent in the
port; and (3) excretion over the canalicular (apical) membrane.2 evaluation of hemodynamic changes during pregnancy.35 Never­
52 Rate of ICG disappearance from vascular compartment is 18– theless there are concerns among ophthalmologists, since the
24% per minute, and after 20 minutes no more than 4% remains Food and Drug Administration has classified ICG as a preg-
in the plasma.29 Plasma decay curve is initially exponential, then nancy category C drug, meaning that adequate studies of safety
decelerates.26 No peripheral uptake has been described, in have not been conducted.30
Section 1

kidney,26 lungs, or placenta.30


ICG’s high molecular weight, in combination with the high Instrument comparison
percentage of dye bound to plasma proteins, reduces the amount There are several instruments at present that can be used to
of dye that exits from fenestrations in choroid vessels. This perform ICGA. All of them can be divided into two main cate­
feature and its optical properties make ICG suitable for choroi- gories: digital flash fundus cameras and SLOs.
dal vascular network visualization. Although it has been reported At the time of writing, the flash camera group includes the
Optical Imaging Technologies
Retinal Imaging and Diagnostics

that ICG can diffuse through the choroid and can accumulate in TRC-50DX ICG (Topcon, Tokyo, Japan), the FF 450plus (Carl
RPE cells, no dye should remain in the late phases (30–40 Zeiss Meditec, Dublin, CA, USA), and the VX-10i (Kowa, Tokyo,
minutes) of the angiogram.29 Japan). ICG-capable SLO systems include the Spectralis HRA
(Heidelberg Engineering, Heidelberg, Germany) and the F10
Toxicity (Nidek, Gamagori, Japan).
ICG is considered a safe and well-tolerated dye. Its LD50 is The differences between these instruments are largely related
60  mg/kg in mice.20 Constant infusions over a 3-hour period to the acquisition modality (Fig. 2.1). The light source for
with dosages as high as 50  mg/kg of body weight were well a digital camera is a white light with an excitation filter
tolerated.27 Subcutaneous extravasation also does not produce (640–780  nm) and a barrier filter (820–900  nm). In an SLO a
significant local effects.26,31 Overall, the side-effect rate is low: laser monochromatic light is used to excite (785–790  nm) with
0.15% with mild events (nausea, vomit, sneezing, pruritus), a barrier filter at 805  nm. The laser light for an SLO system is
0.2% with moderate events (urticarial, syncope, pyrexia, nerve moved on the fundus by two rotating mirrors and the image
palsy), 0.05% with severe events (bronchospasm, laryngospasm, is acquired point by point. For a typical 30° image, this takes
anaphylaxis).31 approximately 60–200  ms. The presence of a confocal aperture
The mechanism of these various adverse side-effects is in SLO systems allows selective acquisition of light from a par-
uncertain. For some, a dose-dependent pseudoallergic mecha- ticular tissue layer (from the focal plane), and blocks the light
nism has been proposed,32 though there does not appear to that is coming from the surrounding tissue.36,37 Flash systems,
be correlation with iodide or shellfish intolerance, suggesting in contrast, do not use a confocal aperture, and thus the fluo-
that the sodium iodide component is of little significance.22,33 rescent light returning to the camera will emanate from multiple
Nevertheless patients with a history of definite iodine allergy layers. However, even for clinical confocal SLO ICG systems,
should not be given the dye, because of concerns for possible the  confocal aperture  is much larger than one would choose for
anaphylaxis.31 Caution should also be observed in patients optimum z-resolution  imaging. The reason for this is that fluo-
affected by liver diseases29 and kidney diseases, since a 9.3% rescence light from different depth planes (i.e., from the choroid
incidence of adverse reactions has been reported in dialysis and from the retinal vessel) needs to be imaged simultaneously
patients.22,34 for many clinical applications.

Fig. 2.1 Schematic representation of


the differences between digital flash
fundus cameras (A), scanning laser
ophthalmoscopes (SLO) (B), and confocal
SLO instruments (C). In digital flash fundus
cameras, white light (with or without
excitation/barrier filters) is used. In SLO
systems the light source is a monochromatic
laser. In SLO confocal systems a pinhole
aperture blocks the reflected or fluorescent
light from areas outside the focal plane.

Digital camera Scanning laser Confocal scanning laser


ophthalmoscope ophthalmoscope
These characteristics are important to recognize the different
appearance of ICGA images obtained by different instruments Injection technique
(Fig. 2.2). Another difference is the number of images acquired The concentration and preparation for intravenous injection of 53
per second. With SLO systems the frame rate may reach 12 ICG vary with the instrument used. For fundus cameras the
images per second, thus permitting dynamic ICGA (Fig. 2.2). standard concentration is 25 mg of ICG dissolved in 5 mL

Chapter 2
With digital fundus cameras the maximum rate is one frame solvent.38 The dosage may be increased to 50 mg in patients with
per second. poorly dilated pupils and heavy pigmentation.39 For SLOs the

Clinical Applications of Diagnostic Indocyanine Green Angiography


A B

C D

Fig. 2.2 Comparison of dynamic (six frames per second) and conventional indocyanine green angiography of a stage II retinal angiomatous
proliferation lesion. A feeding retinal arteriole (A, arrow), filling of the vascular lesion, and a draining retinal vein are all characteristic features
visible in the dynamic sequences (A–C).
Continued
quite difficult to visualize, since the resolution of the cameras is
insufficient to resolve the size of its small lobular morphology.
54 Therefore the choriocapillaris is visualized as a diffuse indistinct
haze, more evident in the posterior pole and less evident in the
peripheral retina (Fig. 2.3).
Choroidal vessels are usually first observed emanating from
Section 1

the posterior ciliary arteries. A well-defined watershed zone is


present between the medial and lateral posterior ciliary artery
(Fig. 2.4).40 Compared to fluorescein, the watershed zone is more
difficult to visualize, since there is less contrast between per-
fused and nonperfused choroid.
Choroidal vortex veins are visible in the late phase of ICGA
Optical Imaging Technologies
Retinal Imaging and Diagnostics

and are usually four in number (Fig. 2.5). They drain the
corresponding segment of the iris, ciliary body, and choroid.
Sometimes, especially in myopic eyes, a vein may be seen
passing from the choroid through the sclera closely adjacent
to the optic nerve head and draining into the venous plexus
of the pial sheath of the optic nerve (choriovaginal vein)
(Fig. 2.5).40
In case of a thin sclera, such as in the setting of a choroidal
staphyloma, extrabulbar vessels may be visualized. These can be
E
distinguished from normal choroidal vessels because they
pulsate in accordance with the heartbeat. Moreover, they change
Fig. 2.2 Cont’d The filling sequence is missed during the first two shape and position with eye movements.41,42
images captured with a conventional flash fundus camera system (D, E).

Exudative age-related macular degeneration


Exudative age-related macular degeneration (AMD) is generally
standard dosage is 25 mg of ICG dissolved in 3 mL, and 1 mL classified based upon the axial location of the choroidal neovas-
of the solution is injected. The solvent may be either saline alone cularization (CNV). A type 1 CNV is a neovascular membrane
or fluorescein sodium solution at 10–20–25% concentrations, for that is located under the RPE, whereas a type 2 CNV has passed
combined fluorescein and ICGA. Intravenous ICG injection through the RPE and lies under the neurosensory retina.43
should be rapid and immediately followed by a 5-mL saline According to the Macular Photocoagulation Study, type 1 CNV
flush. is generally considered to correspond to “occult” CNV on fluo-
In patients with iodine allergy, infracyanine green is available: rescein angiography (as defined by the Macular Photocoagula-
this is the iodine-free formula of ICG. The technique of injection tion Study), and type 2 CNV generally corresponds to “classic”
is equivalent, but a glucosate solvent should be used for prepara- CNV.44,45 More recently, type 3 CNV has been defined to be CNV
tion. As a result, combined fluorescein angiography and infra- with a definite intraretinal component.46
cyanine green angiography is not possible. The fact that occult CNV accounts for the vast majority of the
exudative complications in AMD47 explains in part why ICGA,
INDOCYANINE GREEN
with its ability to delineate occult CNV, has become part of
ANGIOGRAPHY INTERPRETATION standard care in exudative AMD for many clinicians.19,48,49 None-
Normal eye theless, in peer-reviewed journals, the number of articles pub-
lished whose title included the terms “indocyanine green
To understand the manifestations of disease on ICGA, a recogni-
angiography (or videoangiography)” decreased between 1995
tion of the normal appearance of ICGA in normal subjects is
and 2010.1 One likely explanation might be the advent of anti-
essential.
VEGF therapy which inaugurated a new era in the management
Since ICGA is a dynamic examination, the characteristic find-
of exudative AMD: it was the first therapy to improve the mean
ings may vary depending on the time after dye injection or the
visual acuity of eyes treated with monthly injections of ranibi-
phase of the angiogram. It is well known that fluorescein angi-
zumab, regardless of whether the CNV lesion was predomi-
ography has an arterial retinal phase, an arteriovenous, and a
nantly classic50 or occult.51 Thus, the management of CNV has
venous phase. Similarly for ICGA, one can recognize an early
changed from the use of therapy for which accurate localization
phase when the retinal artery is not yet filled, a midphase when
of the membrane was crucial (i.e., laser photocoagulation, pho-
both arteries and veins are filled, and a late or recirculation phase
todynamic therapy) to the nonspecific intravitreal delivery of
more than 10 minutes after injection.
active and highly effective biologic drugs. Nonetheless, we
In ICGA, the early filling phase may best be correlated with
would still argue that the gold-standard diagnostic procedure
the filling of different layers of the choroid. The first choroidal
should be the one which best visualizes the disease (CNV) under
vessels to be filled are the ones of the deeper Haller’s layer, fol-
investigation.
lowed by the intermediate Sattler’s layer (Fig. 2.3). The chorio-
capillaris is the last layer to be filled (therefore the sequence Type 1 choroidal neovascularization
progresses from the biggest and outermost to the smallest and This type of CNV is by definition under the RPE, and corre-
innermost vessels). However, the choriocapillaris is typically sponds to the occult neovascular network on fluorescein
angiography. The Macular Photocoagulation Study recognized dynamic ICGA may differentiate an occult form of CNV from
two forms of occult CNV: (1) a fibrovascular pigment epithelial retinal angiomatous proliferation (RAP)52 (Fig. 2.8). Considering
detachment (PED); and (2) a late-phase leakage of an undeter- that one-fourth of patients with an LLUS do have a RAP,52 and 55
mined source (LLUS).44 As for the fibrovascular PED, its preva- that an early diagnosis of these lesions is crucial for the func-
lence may vary from 22% to 50%47–49,52 of occult CNV lesions; tional prognosis,57 the importance of an ICGA evaluation for

Chapter 2
dynamic ICGA may delineate the presence of a neovascular these cases becomes readily apparent.
network usually located along the edges of the PED49,53,54 In conclusion, ICGA faciliates a better and more complete
(Fig. 2.6). Moreover, dynamic ICGA may reveal a feeder vessel classification of occult CNV subtypes, compared to fluorescein
that can be successfully treated with laser photocoagulation, angiography. Of note, Yannuzzi et al.19 found that 39% of lesions
when it is located outside the foveal region55,56 (Fig. 2.7). In case classified as poorly demarcated occult lesions by fluorescein
of LLUS, which may represent 36–78% of occult CNV,47,48,52 angiography were well defined by ICGA.

Clinical Applications of Diagnostic Indocyanine Green Angiography


Fig. 2.3 The filling of vessels during an
indocyanine green angiogram follows a
A precise sequence. The first layer to be filled
is Haller’s layer (A, B), followed by Sattler’s
layer (C, D).
Continued

B
Choriocapillaris
Sattler’s layer

Haller’s layer

D
Choriocapillaris
Sattler’s layer

Haller’s layer
Fig. 2.3 Cont’d After Haller’s layer and
Sattler’s layer, the choriocapillaris is filled
E next (E, F). Simultaneous dynamic
56 fluorescein and indocyanine green
angiography (A, C, E) permits the different
phases to be resolved. The choriocapillaris
may be better appreciated when there
Section 1

are adjacent areas of focal loss, as in the


example of geographic atrophy shown in
panels (G) and (H).
Optical Imaging Technologies
Retinal Imaging and Diagnostics

F
Choriocapillaris
Sattler’s layer

Haller’s layer

H
Choriocapillaris
Sattler’s layer

Haller’s layer
57

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

Medial PCA Long PCA

Fig. 2.4 A case of occlusion of the medial long posterior ciliary artery (PCA). (A) Indocyanine green angiography (ICGA) allows one to appreciate
the different territories supplied by this artery and the lateral long PCA. (B) Venous phase of ICGA shows the filling of the lateral vortex veins.
In the upper left area, the vortex vein is partially filled due to drainage from the iris (B, arrow). (C) Schematic representation of the different
vascular territories of the two arteries. (Panel (C) modified with permission from Hayreh SS. Physiological anatomy of the choroidal vascular bed.
Int Ophthalmol 1983;6:85–93.)

inhibitor along with a platelet-derived growth factor (PDGF)


Type 2 choroidal neovascularization
inhibitor appeared to show a synergistic effect for controlling the
In classic CNV, ICGA improves visualization of the fine struc-
growth of mature vessels.61 This is likely because pericyte recruit-
ture of the neovascular network1 (Fig. 2.9), allowing the choroi-
ment is part of the maturation process in blood vessel develop-
dal and retinal circulation to be distinguished. This high spatial
ment. Once the pericyte cell population is well established, the
and temporal resolution permits identification of choroidal
effectiveness of anti-VEGF agents is greatly reduced. PDGF-B is
vessels that feed into the CNV.58 In early phases, ICGA shows a
a key requirement for the recruitment of pericytes to the newly
dark rim which corresponds to a whitish ring on infrared
formed vessels. Mature, larger choroidal vessels may be readily
imaging,59 and a discrete neovascular network surrounded by a
differentiated from immature choroidal capillaries on ICGA
hypocyanescent margin which is more visible after 15 minutes.60
(Fig. 2.10). Thus, in patients with chronic AMD or those who did
Watzke et al.54 showed that 87% of eyes with classic choroidal
not benefit from previous treatments with anti-VEGF, ICGA
neovascular membranes were hypercyanescent with distinct
might better delineate a more mature stage of CNV. This has
edges.
potential implications for therapeutic decision-making.
This ability to provide a clear delineation of the neovascular
network may confer an important advantage in the era of anti- Type 3 choroidal neovascularization
VEGF therapy. It has been reported that VEGF inhibitors were During the past decade, RAP has been labeled with a number of
more effective in controlling immature vessels, whereas a VEGF different terms, including “retinal vascular anomalous complex,”
58
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 2.5 Indocyanine green angiography allows visualization of the four vortex veins (A), and the choriovaginal vessels (B, arrow).

A B

Fig. 2.6 A pigment epithelium detachment (PED) (A, fluorescein angiography) with a well-delineated neovascular network located along the edges
of the PED (B, indocyanine green angiography).
59

Chapter 2
*

Clinical Applications of Diagnostic Indocyanine Green Angiography


A B

Fig. 2.7 Fibrovascular pigment epithelium detachment (PED). Fluorescein angiography demonstrates occult choroidal neovascularization with
PED (A). In the early phases of indocyanine green angiography (ICGA) a feeder vessel originating in the juxtapapillary area is clearly delineated
(B, asterisk). Feeder vessel and draining vein are indistinguishable in the late phases of indocyanine green angiography (C).
60
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

C D

Fig. 2.8 Late leakage of undetermined source (A, C). Indocyanine green angiography may clearly differentiate a subtype of occult choroidal
neovascularization (B) from retinal angiomatous proliferation (D).
61

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

Fig. 2.9 A case of type 2 choroidal neovascularization. In fluorescein angiography images (A), the leakage of the dye from the lesion is evident,
and obscures the boundaries of the neovascular network. In indocyanine green angiography (ICGA) (B), the limits of the neovascularization are
much more visible. Moreover, ICGA allows the visualization of a central feeder vessel, with a surrounding net of smaller neovessels.

“retinal choroidal anastomosis,” “retinal anastomosis to the diagnosis of RAP is based upon the temporal evidence of “dye
lesion,” and “chorioretinal anastomosis.” In a comprehensive filling of at least one retinal arteriole descending into the deep
article on this entity, Yannuzzi et al.62 provided evidence to retinal space to a vascular communication and at least one drain-
support the original concept of capillaries arising within the ing retinal vein.”64 In conventional angiography, images are
inner half of the retina, or “retinal angiomatous proliferation,” usually captured at 1 frame per second. This makes it virtually
thus suggesting the acronym of RAP as the appropriate descrip- impossible to visualize the progression of the dye through the
tor for the disease. Subsequently, Gass et al.63 suggested a pos- vascular complex, even though images are taken at very early
sible choroidal origin for these vessels, emanating from occult phases. By contrast, d-ICGA takes up to 12 frames per second
CNV and developing into an occult chorioretinal anastomosis. and captures the progressive filling of the lesion, allowing detec-
The new category, type 3 neovascularization, has been recently tion of very small and recent-onset cases of RAP (Fig. 2.2). The
proposed46 to harmonize these conflicting theories. Type 3 possibility of repeated viewing of the dynamic sequence of pro-
lesions would encompass the following disease manifestations: gression on ICGA may further increase our chances of an early
(1) focal neovascular proliferation arising from the deep retinal diagnosis of RAP (Fig. 2.13). In a recent series of RAP diagnosed
capillary plexus (the original RAP concept); (2) intraretinal neo- using d-ICGA,52 the incidence of stage 1 RAP (64.9%) and the
vascular extension from an underlying occult/type I CNV; and mean distance of the lesions from the fovea (682 ± 304 µm) were
(3) de novo breaks in Bruch’s membrane with neovascular infil- both consistent with an early-stage disease process, supporting
tration into the retina. the utility of this imaging procedure.
Whatever the origin or initial location might be, our under- Early and accurate diagnosis of RAP is important for at least
standing of the importance of RAP as a component of neovas- two reasons. First, RAP lesions are thought to be more aggres-
cular AMD has been enhanced by ICGA. The fluorescein sive,65 with a treatment response that is likely to diminish with
angiographic study generally shows manifestations of occult more advanced disease stages.57 Second, recent data from the
CNV, either fibrovascular PED (Fig. 2.11) or LLUS (Fig. 2.12) literature suggest that successful anatomic and functional results
without a characteristic feature to identify and delineate the with RAP may be achieved more consistently with combined
angiomatous process in the retina (indistinct zone of staining treatments (i.e., intravitreal injection of steroid or anti-VEGF +
within and beyond the retina). By contrast, ICGA may clearly photodynamic therapy) rather than with intravitreal injection of
delineate the vascular structure of the lesion. When associated anti-VEGF therapy alone.66,67 Dynamic ICGA is also extremely
with PED, the RAP is usually well within the area of detachment useful for monitoring the therapeutic effect: a complete remodel-
and not at the edges, as is typically the case with CNV which ing of the vascular structure may be achieved after successful
vascularizes a serous PED (so-called notched PED configuration closure and it is clearly highlighted in d-ICGA (Fig. 2.14).68
on fluorescein angiography). As previously reported, RAP may
be present in up to one-fourth of eyes thought to have occult Polypoidal choroidal vasculopathy
CNV with LLUS.52 Dynamic ICGA (d-ICGA) has further Polypoidal choroidal vasculopathy (PCV) is a primary abnor-
expanded our capability for an early diagnosis. By definition, a mality of the choroidal circulation characterized by an inner
62
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

C D

Fig. 2.10 Red-free image showing intraretinal blood within a central


atrophic area. A small subretinal hemorrhage is also present inferiorly
(A). Corresponding fluorescein angiogram demonstrating late leakage
of undetermined source along the inferior edge of the atrophic area
(B). Simultaneous early-phase indocyanine green angiography (C)
reveals mature, large choroidal vessel (asterisk) feeding the large net
of neovascularization along the inferior edge of the central atrophy. A
real chorioretinal anastomosis (arrow) is also present. Four minutes
after injection (D), the draining choroidal veins are well visualized, as
is the neovascular network inferiorly (D, open circles). Red-free image
after a 3-month loading phase with monthly ranibizumab (E) shows an
E increase in size of the central retinal hemorrhage despite anti-vascular
endothelial growth factor therapy.
63

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

Fig. 2.11 Late-phase fluorescein angiography (A) shows a pigment epithelium detachment (PED) with a “hot spot.” Indocyanine green
angiography (B) reveals the presence of retinal angiomatous proliferation overlying the PED. Feeding retinal arteriole (arrow) and draining retinal
venule (arrowhead) are clearly visualized.

*
V

A B

Fig. 2.12 Indocyanine green angiography (A) and fluorescein angiography (B) demonstrating an extrafoveal stage II retinal angiomatous
proliferation (RAP). One feeding first-order macular arteriole (arrow) shunts blood flow from the vascular arcade (*) to the RAP and to one
draining retinal vein (V). Cystoid macular edema is evident in late fluorescein phases.
64
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

V
A

A B

Fig. 2.13 Midphase fluorescein angiography (A) showing a small area of progressive extrafoveal staining at 6 o’clock. Dynamic indocyanine
green angiography (B) reveals a well-delineated stage I retinal angiomatous proliferation with a feeding retinal arteriole (A) and a draining retinal
vein (V).

choroidal vascular network of vessels ending in an aneurysmal as well as selective laser photocoagulation have been shown to
bulge or outward projection, visible clinically as a reddish- be effective treatments.78
orange, spheroid, polyp-like structure.69 It was first described in Given the facts that ICGA is the most sensitive and specific
the peripapillary area69,70 (Fig. 2.15) but it may affect the macula71 tool for PCV identification and the treatment for PCV may differ
(Fig. 2.16), and also extramacular areas (Fig. 2.17). from other diseases for which it is frequently confused, it would
The disorder is associated with multiple, recurrent, serosan- seem apparent that ICGA is an important tool in the evaluation
guineous detachments of the RPE and neurosensory retina sec- of all cases of exudative lesions suspected of harboring PCV.
ondary to leakage and bleeding from the peculiar choroidal
vascular abnormality. It has been reported that 85% of patients Central serous chorioretinopathy
with serosanguineous detachments of the RPE have evidence of CSC is characterized by multifocal areas of choroidal hyperper-
PCV.72 ICGA has been used to detect and characterize the PCV meability on ICGA,79–81 visible in the mid and late phases of the
abnormality with enhanced sensitivity and specificity.1,71 The angiogram82 (Fig. 2.18). These areas surround the active RPE
early phase of the ICG angiogram shows a distinct network of leaks but can also be found in areas apparently unaffected by
vessels within the choroid (Fig. 2.16B). In patients with juxtapap- leakage or abnormal fluorescence on fluorescence angiography,
illary lesions, the vascular channels may follow a radial, arching even in the fellow eyes.79 Zones of choroidal hyperpermeability
pattern. In PCV limited to the macula, a vascular network often tend to persist in cases of severe and chronic CSC83 (Fig. 2.19),
arises in the macula and follows an oval distribution pattern.73 and are of value for distinguishing CSC from AMD in older
Larger choroidal vessels of the PCV network begin to fill before patients with suspected occult neovascularization.80,84
retinal vessels, and PCV network fills also at a slower rate than Moreover, ICG assessment of the location of these areas of
retinal vessels. Shortly after the network can be identified by the hyperpermeability may be useful when considering treatment
ICG angiogram, small hyperfluorescent “polyps” become visible with verteporfin photodynamic therapy, using normal80 or half-
(Fig. 2.16C). These polypoidal structures correspond to the fluence laser energy.85 The treatment focused on these areas
reddish, orange choroidal excrescence seen clinically. They showed rapid reduction of fluid and improvement in visual
appear to leak slowly as the surrounding hypofluorescent area acuity,80 possibly by leading to hypoperfusion of choriocapillaris
becomes increasingly hyperfluorescent. In the later phase of the and vascular remodeling.86 In these studies, verteporfin photo-
angiogram there is uniform disappearance of dye (“washout”) dynamic therapy success rate seemed to be dependent on the
from the bulging polypoidal lesions. degree of hyperpermeability, as the treatment was less effective
PCV is often misdiagnosed or confused with chronic central or had more frequent recurrence of CSC in eyes without intense
serous chorioretinopathy (CSC)74,75 and with exudative age- hyperfluorescence.87
related maculopathy71,76 and may represent a transitional condi- Other findings in CSC using ICGA include multiple “occult”
tion between the two pathologies.1 Moreover, the treatment serous PED,79 punctate hyperfluorescent spots,88 delays in arte-
strategies for PCV differ from exudative AMD. The use of anti- rial filling of the choroidal arteries and choriocapillaris,89,90 and
VEGF agents is controversial in PCV,1 while verteporfin photo- venous congestion.90 ICGA is also useful in differentiating CSC
dynamic therapy, alone or in combination with bevacizumab,77 from PCV.75
65

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

C D

Fig. 2.14 Baseline fluorescein angiography (A) and dynamic indocyanine green angiography (ICGA) (B) shows a stage II extrafoveal retinal
angiomatous proliferation (RAP) located inferotemporal to the fovea. One feeding first-order macular arteriole (arrowhead) shunts blood flow from
the vascular arcade to the RAP and to the draining retinal vein (arrow). Two months after one combined treatment (intravitreal triamcinolone
acetonide + photodynamic therapy), the RAP is no longer detectable by either fluorescein angiography (C) or ICGA (D). There is an evident
reduction in the size of both the first-order macular arteriole and the draining retinal vein (barely visible) (C, D).

V
66
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 2.15 Late-phase fluorescein angiography (A) shows a neurosensory retinal detachment with multiple juxtapapillary “hot spots.” Early (B) and
late (C) indocyanine green angiography reveals the presence of juxtapapillary polypoidal choroidal vasculopathy.

fluid leading to exudative retinal detachment with macular


Choroidal tumors involvement is common in symptomatic cases. Retinal hard exu-
Choroidal hemangioma dates are minimal or absent.
Unlike diffuse choroidal hemangiomas, which are usually Angiographic studies such as fluorescein and ICG can be
evident at birth and typically occur as part of neuro- helpful in establishing the diagnosis and differentiating these
oculocutaneous hemangiomatosis or Sturge–Weber syndrome, benign lesions from other tumors, namely amelanotic malignant
circumscribed choroidal hemangioma may be more difficult to melanoma and choroidal metastases. Fluorescein angiography
diagnose. demonstrates a hyperfluorescent mass with a fine lacy vascular
Circumscribed choroidal hemangiomas are benign hamarto- network of intrinsic vessels in the early choroidal filling phase.
mas that typically present from the second to fourth decade of The hyperfluorescence increases throughout the angiogram, and
life.91 They usually occur sporadically in the absence of systemic there is variable leakage in late views94 (Fig. 2.20A, B). ICGA is
disease. Histopathology reveals that the tumor is composed by the most useful study for demonstrating the intrinsic vascular
vascular channels lined with endothelium. It involves the full pattern of circumscribed choroidal hemangioma.95 The advan-
thickness of the choroid with secondary changes of the overlying tage of ICG dye over sodium fluorescein dye is that it diffuses
RPE and the retina.92 Although commonly asymptomatic, cho- much more slowly out of fenestrated small choroidal vessels
roidal hemangiomas can be associated with exudative retinal than does sodium fluorescein. Within 30 seconds of injection of
detachment resulting in reduced visual function, metamorphop- the ICG dye, the tumor’s intrinsic vascular pattern becomes
sia, and photopsia. apparent. By 1 minute, choroidal hemangiomas completely fill
On ophthalmoscopic examination, a circumscribed choroidal with the dye, showing brilliant hyperfluorescence. This 1-minute
hemangioma appears as an orange choroidal mass with indis- stage of intense hyperfluorescence seen with choroidal heman-
tinct margins that blend with the surrounding choroid. They are giomas is brighter than any other tumor, and it is very sugges-
frequently located in the macular region of the posterior pole, tive of the diagnosis. In the following phases (6–10 minutes), the
and are not usually thicker than 6 mm.93 Surrounding subretinal hyperfluorescence can persist or begin to wane (Fig. 2.20C-E). In
67

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

Fig. 2.16 Late-phase fluorescein angiographic image revealing type 1 occult choroidal neovascularization with a subfoveal pigment epithelium
detachment (PED) (A). Early (B) and late (C) indocyanine green angiographic frames demonstrate a distinct network of vessels within the
macular choroid ending with two hyperfluorescent “polyps.” One of the two is located within the PED.
68
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 2.17 Early (A) and late (B) indocyanine green angiography showing extramacular polypoidal choroidal vasculopathy with a pigment
epithelium detachment.

the late phases of the ICG angiogram (30 minutes), a washout simulate a vitreous hemorrhage with suspicion of underlying
effect with reduction of the initial hyperfluorescence is observed retinal detachment or break, intraocular inflammatory process,
secondary to the outflow of dye from the hemangioma (Fig. retinal artery macroaneurysm, or choroidal melanoma.105 In fact,
2.20F).95 The low-resistance, high-flow properties of the tumor PEHCR often appears as a visible intraocular elevated mass with
allow rapid flow of the dye into and out of the tumor. The result- a mean basal dimension of 10 mm and a mean thickness of
ing final effect is that the tumor empties sooner than the normal 3 mm, consistent with the size of a small to medium-sized mela-
surrounding choroid and thus appears hypofluorescent in com- noma.106 The lesion is most often located temporally (77%)
parison. This washout sign is very helpful in differentiating cho- between the equator and the ora serrata (89%) and involves one
roidal hemangiomas from amelanotic malignant melanoma and (46%) or two (46%) quadrants.106 In comparison, eyes with uveal
choroidal metastases. melanoma show tumor location in the macula (5%), between the
macula and the equator (78%), and between the equator and the
Choroidal melanoma ora serrata (17%).106
ICGA findings in uveal melanoma are variable.96 No study Many eyes with PEHCR have features of macular or extra-
revealed any pathognomonic sign with ICGA to identify choroi- macular (peripheral) degeneration such as drusen, RPE altera-
dal melanoma.23,97 tions, or CNV.106 The majority of PEHCR lesions spontaneously
Nevertheless ICGA was found to be capable of identifying resolve, leaving RPE atrophy, hyperplasia, and fibrosis. These
tumor vessels (Fig. 2.21)98,99 which are usually irregularly tortu- features imply a bilateral generalized aging process within the
ous, with anarchic branching,97 dilated with a parallel course,98 eye and are consistent with the degenerative nature of the
and characterized by vasculogenic mimicry patterns.100 ICGA disease. Although almost half of patients may be asymptomatic,
was demonstrated to be superior to fluorescein angiography in a decrease in visual acuity related to PEHCR may occur in up to
detecting both tumor borders and vasculature.97,101 20% of cases.106 Both for this reason and for the fact that the acute
Mueller et al. found that different patterns of the microcircula- hemorrhagic form is typically mistaken for melanoma, PEHCR
tion within the tumor may be useful in the prognosis of the deserves an early and proper clinical diagnosis. Fluorescein
disease.101 The evidence of microcirculation patterns character- angiography is of little help because choroidal neovascular
ized by networks and a parallel course with cross-linking may lesions are visible in only 3% of cases.106 This is due to the block-
be associated with a higher risk of metastatic disease.101 Other age of choroidal fluorescence related to subretinal hemorrhage,
studies reported the possible role of ICGA in evaluating the sub-RPE hemorrhage, or RPE hyperplasia. Diffuse peripheral
outcome of brachytherapy,102 proton beam irradiation,103 and changes consistent with variable degrees of RPE hyperplasia or
transpupillary thermotherapy.104 atrophy are also common fluorescein angiographic features.
By contrast, ICGA may clearly delineate the choroidal neo-
Peripheral exudative hemorrhagic chorioretinopathy vascular process, which is often located at the edges of the
Peripheral exudative hemorrhagic chorioretinopathy (PEHCR) blood pool, even in the far periphery. Photodynamic therapy
is a bilateral peripheral exudative hemorrhagic retinal degenera- or laser photocoagulation applied to the choroidal new vessels
tive process of the eye.105,106 The condition is characterized by may further accelerate the reabsorption of subretinal blood
blood in the subretinal or subretinal pigment epithelial space. with decreased risk of subsequent visual acuity loss (Figs 2.22
PEHCR is most often found in older Caucasian patients and may and 2.23).
69

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

Fig. 2.18 A case of central serous chorioretinopathy. In the fluorescein


angiographic image (A) it is possible to visualize three distinct serous
detachments of the pigment epithelium, and one point of leakage.
Another hyperfluorescent area is visible (arrow), but it is not clear if it
corresponds to another point of leakage or represents an additional
detachment. Indocyanine green angiographic images (B, C) allow one
to distinguish clearly points of leakage as hyperfluorescent areas, with
C marked leakage in the late phases (C, arrow). The uncertain area
noted on fluorescein angiography is another leaky point.
70
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 2.19 A case of central serous chorioretinopathy. Fluorescein angiographic image (A) shows only an area of pigment epithelium disturbance.
Indocyanine green angiography (B) reveals a more extensive alteration of the choriocapillaris, with multiple areas of hyperfluorescence.

Varix of the vortex vein ampulla hypofluorescent areas at the posterior pole and peripheral
Varix of the vortex vein ampulla is a rare, benign, asymptomatic retina. These spots become visible in the mid to late phases,
condition, which may be confused with a choroidal nevus or range in size between 50 and 1000  µm,112 and are more appar-
melanoma.107 The choroidal veins drain into an average of four ent in ICGA images than by fundus examination and fluorescein
vortex veins, which exit the globe through scleral canals.108 angiography1,112 (Fig. 2.25). In addition, ICGA may show hypo-
About half of the vortex veins show dilatations of varying sizes fluorescence surrounding the disc area.111 The hypofluorescent
and shapes, and are referred as vortex vein ampullae. The varix spots disappear at the recovery stage of the disease, and some-
of the vortex vein ampulla is an unusually large dilatation of the times are more persistent with ICGA.113
vortex vein. The cause remains unclear. The gaze-dependent Multifocal choroiditis
dynamic nature of the lesion suggested gaze-evoked kinking of In multifocal choroiditis the white lesions are visualized as hypo-
the extrascleral vortex vein or narrowing of the scleral canal to fluorescent spots in ICGA images. These lesions may be followed
be considered as the possible cause.109 The varix may also be up with ICGA both in the natural course of the pathology and
enlarged by factors that increase ocular venous pressure, like in the response to treatment with oral prednisone.114 A reduction
Valsalva maneuver, head-down positioning, and jugular vein in size and number of hypofluorescent spots is observed after
compression.109 Biomicroscopically, the lesion appears as a successful treatment. Other findings visible on ICGA are hyper-
smooth red-brown elevation in the equatorial region, usually in fluorescent spots, which usually do not correspond with the
the nasal quadrants.107 It is usually a single lesion but may be hyperfluorescent foci seen on fluorescein angiography, and a
bilateral.109 A proper diagnosis may be achieved by pressure on large hypofluorescent area surrounding the optic nerve.
the globe that readily collapses the varix (Fig. 2.24).109 ICGA96,107,110
is particularly useful because it demonstrates the relationship of Birdshot chorioretinopathy
the varix to the choroidal vasculature and also allows visualiza- This disease is characterized by deep cream-colored dots scat-
tion of the pressure and gaze-dependent changes. Relatively tered diffusely throughout both fundi. The lesions appear as
early maximum fluorescence and a homogeneous filling pattern round-oval, hypofluorescent, symmetric dots on ICGA.115 These
may further help differentiate the varix from other choroidal lesions are typically not seen on fluorescein angiography, and
masses.107 therefore ICGA may detect birdshot lesions more rapidly.116
Other findings on ICGA include diffuse ICG hyperfluorescence
predominantly found in the posterior pole in the late phase of
Choroidal inflammation and
angiography, and an alteration of the vascular pattern of the
white-dot syndrome choroid with choroidal vessels appearing fuzzy and indistinct
Multiple evanescent white-dot syndrome in the intermediate phase of angiography.115 In the chronic phase
Multiple evanescent white-dot syndrome is a unilateral acute of the disease the hypofluorescent dots persist in the late phases
disease that affects young women, presenting with a transient, of the angiogram and correspond to RPE atrophy or choroidal
self-limiting visual loss. The disease involves the choroid granulomas.1,117
and the outer retina.111,112 ICGA shows a pattern of multiple Text continued on page 77
71

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
A B

C D

Fig. 2.20 Fluorescein angiography demonstrating a hyperfluorescent mass along the inferior vascular arcade (A). The hyperfluorescence
increases progressively throughout the angiogram with variable leakage in late views (B). Early-phase indocyanine green angiography (ICGA)
(49 seconds) revealing a fine lacy vascular network of intrinsic vessels (C). Increasing hyperfluorescence is detected after injection:
(D) at 2 minutes.
Continued
72
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

E F

Fig. 2.20 Cont’d Increasing hyperfluorescence after injection: (E) at 5 minutes. Of note, the margins of the tumor appear scalloped. (F) Late-phase
ICGA study demonstrating hypofluorescence within the tumor (washout effect). A halo of minimal hyperfluorescence surrounds the tumor. This may
result from staining of the retinal pigment epithelium or leakage of indocyanine green into the subneurosensory retinal space.

A B

Fig. 2.21 A case of choroidal melanoma. Indocyanine green angiography (A) allows visualization of the tumor’s intrinsic vasculature with irregular
tortuosity and anarchic branching. Note the strong fluorescence within the large choroidal vessels around the lesion, a possible sign of increased
flow due to the presence of the tumor. Melanoma-intrinsic vessels are leaky by fluorescein angiography (B), and therefore they cannot be
identified with this diagnostic tool. In fluorescein angiographic images (B), in the inferior quadrant it is possible to visualize damage to retinal
vessels with associated exudative detachment.
73

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
*

A B

C D

Fig. 2.22 A case of a midperipheral inferotemporal subretinal hemorrhage. Midphase fluorescein angiography showing a hyperfluorescent leaking
spot within the hemorrhage (A). Note the diffuse peripheral changes consistent with variable degrees of retinal pigment epithelial hyperplasia
or atrophy. Indocyanine green (ICG) angiography 31 seconds after injection (B): the choroidal neovascularization (CNV) is clearly outlined
(asterisk). Six months after laser photocoagulation of the CNV: fluorescein (C) and ICG (D) angiographies reveal fibrosis of the CNV with late
staining and no leakage.
74
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

C D

Fig. 2.23 Infrared image of a peripheral temporal subretinal hemorrhage (A). Midphase fluorescein angiography showing diffuse peripheral
changes consistent with variable degrees of retinal pigment epithelium hyperplasia or atrophy. Typical features of choroidal neovascularization
(CNV) are absent (B). Corresponding indocyanine green (ICG) angiography 2 minutes after injection (C): CNV is clearly visible (asterisk) while a
second choroidal new vessel is suspected (circle). Six minutes after injection, ICG angiography shows progressive leakage from both areas (D).
75

Chapter 2
Clinical Applications of Diagnostic Indocyanine Green Angiography
E F

Fig. 2.23 Cont’d Infrared image 3 months after photodynamic therapy applied to the two leaking spots: the reabsorption of the subretinal
hemorrhage is almost complete (E). Corresponding fluorescein (F) and ICG (G) angiograms reveal persistent obliteration of CNV with no late
leakage.
76
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 2.24 Indocyanine green angiography in a case of varix of the vortex vein in the nasal equatorial region (A). Application of sufficient pressure
on the globe readily collapses the varix (B).

A B

Fig. 2.25 A case of multiple evanescent white-dot syndrome. Late phases of the fluorescein angiogram reveal only mild alterations at the level of
the outer retina and retinal pigment epithelium (A). Early phases of the indocyanine green angiogram (B) begin to reveal areas of
hypofluorescence.
and healed, inactive lesions in AMPPE can both be imaged and
differentiated using ICGA.120
Serpiginous choroidopathy 77
ICG allows better staging and identification of active lesions in
serpiginous chorioretinopathy.121 The active phase of the patho­

Chapter 2
logy is characterized by hypofluorescent areas with poorly
defined margins (Fig. 2.26). These findings can predict the active
lesions observed by fluorescein angiography. The presence of
late hyperfluorescence in ICG images represents a sign of cho-
roidal hyperpermeability and may be associated with a more
aggressive evolution of the disease. The healed lesions appear

Clinical Applications of Diagnostic Indocyanine Green Angiography


hypofluorescent with well-defined margins. The atrophy of the
RPE and choriocapillaris allows better identification of large and
medium-sized choroidal vessels.
Punctate inner chorioretinopathy
The subretinal lesions observed in punctate inner chorioreti-
nopathy are visualized by ICG as hypofluorescent areas through-
out all the phases of the angiogram.122 These areas may
correspond to localized choroidal hypoperfusion,123 and are
greater in number compared to fluorescein angiography.124
C
Another finding in ICG images is the presence of hyperfluores-
cent points situated close to the vessel wall, representing a pos-
Fig. 2.25 Cont’d Areas of hypofluorescence become much more
evident in the mid to late phases of the angiogram (C).
sible sign of vasculitis.123

Acute zonal occult outer retinopathy


Acute multifocal placoid pigment epitheliopathy In acute zonal occult outer retinopathy, ICGA shows a variety
ICG of acute posterior multifocal placoid pigment epitheliopa- of patterns of presentations. Spaide reported that the peripapil-
thy (AMPPE) shows areas of hypofluorescence in both early and lary drusenoid material blocks the choroidal fluorescence in ICG
late phases that correlate with the placoid lesions. These lesions and therefore the involved areas appear hypofluorescent.125 The
may be caused by choroidal hypoperfusion, secondary to occlu- secondary atrophy of the choriocapillaris produces hypofluores-
sive vasculitis,118 and ICGA often shows partial or complete reso- cence as well, which does not affect the fluorescence from the
lution throughout the timecourse of the disease.119 New, active underlying larger choroidal vessels.125 In some cases, though,

A B

Fig. 2.26 A case of serpiginous chorioretinopathy. The lesion observed by indocyanine green angiography (A) occupies a greater extent than is
evident on fluorescein angiography (B). This may indicate a progression of the pathology that can be anticipated or predicted using indocyanine
green angiography.
78
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

B C

Fig. 2.27 A case of acute zonal occult outer retinopathy. Autofluorescence shows the typical features of this disorder, with hyperautofluorescent
areas around the optic disc (A). Simultaneous spectral-domain optical coherence tomography suggests that these areas (black arrows)
correspond to zones with loss of the outer segments of the photoreceptors (white arrows), thus resulting in less blockage of the autofluorescence
originating from the retinal pigment epithelium. Fluorescein angiography shows increased fluorescence corresponding to these areas (B). Early
phase of indocyanine green angiography (C) does not reveal alterations.
15. Hasegawa Y, Hayashi K, Tokoro T, et al. [Clinical use of indocyanine green
angiography in the diagnosis of choroidal neovascular diseases.] Fortschr
Ophthalmol 1988;85:410–2.
16. Destro M, Puliafito CA. Indocyanine green videoangiography of choroidal 79
neovascularization. Ophthalmology 1989;96:846–53.
17. Scheider A, Schroedel C. High resolution indocyanine green angiography
with a scanning laser ophthalmoscope. Am J Ophthalmol 1989;108:458–9.
18. Guyer DR, Puliafito CA, Mones JM, et al. Digital indocyanine-green angiog-

Chapter 2
raphy in chorioretinal disorders. Ophthalmology 1992;99:287–91.
19. Yannuzzi LA, Slakter JS, Sorenson JA, et al. Digital indocyanine green video-
angiography and choroidal neovascularization. Retina 1992;12:191–223.
20. Lutty GA. The acute intravenous toxicity of biological stains, dyes, and other
fluorescent substances. Toxicol Appl Pharmacol 1978;44:225–49.
21. Tripp MR, Cohen GM, Gerasch DA, et al. Effect of protein and electrolyte
on the spectral stabilization of concentrated solutions of indocyanine green.
Proc Soc Exp Biol Med 1973;143:879–83.
22. Benya R, Quintana J, Brundage B. Adverse reactions to indocyanine green: a

Clinical Applications of Diagnostic Indocyanine Green Angiography


case report and a review of the literature. Cathet Cardiovasc Diagn
1989;17:231–3.
23. Bischoff PM, Flower RW. Ten years experience with choroidal angiography
using indocyanine green dye: a new routine examination or an epilogue?
Doc Ophthalmol 1985;60:235–91.
24. Flower RW, Hochheimer BF. Indocyanine green dye fluorescence and infrared
absorption choroidal angiography performed simultaneously with fluores-
cein angiography. Johns Hopkins Med J 1976;138:33–42.
25. Geeraets WJ, Berry ER. Ocular spectral characteristics as related to hazards
from lasers and other light sources. Am J Ophthalmol 1968;66:15–20.
26. Cherrick GR, Stein SW, Leevy CM, et al. Indocyanine green: observations on
its physical properties, plasma decay, and hepatic extraction. J Clin Invest
1960;39:592–600.
D 27. Leevy CM, Bender J. Physiology of dye extraction by the liver: comparative
studies of sulfobromophtthalein and indocyanine green. Ann N Y Acad Sci
1963;111:161–76.
Fig. 2.27 Cont’d The late phase of the angiogram (D) allows the 28. Stehr A, Ploner F, Traeger K, et al. Plasma disappearance of indocyanine green:
affected areas to be distinguished by increased fluorescence. a marker for excretory liver function? Intens Care Med 2005;31:1719–22.
29. Costa DL, Huang SJ, Orlock DA, et al. Retinal-choroidal indocyanine green
dye clearance and liver dysfunction. Retina 2003;23:557–61.
30. Fineman MS, Maguire JI, Fineman SW, et al. Safety of indocyanine green
ICG may show an increase in fluorescence from the affected angiography during pregnancy: a survey of the retina, macula, and vitreous
societies. Arch Ophthalmol 2001;119:353–5.
areas, due to the lack of photoreceptor outer segments and the 31. Hope-Ross M, Yannuzzi LA, Gragoudas ES, et al. Adverse reactions due to
minor blocking effect from this layer (Fig. 2.27). indocyanine green. Ophthalmology 1994;101:529–33.
32. Speich R, Saesseli B, Hoffmann U, et al. Anaphylactoid reactions after
CONCLUSIONS indocyanine-green administration. Ann Intern Med 1988;109:345–6.
33. Michie DD, Wombolt DG, Carretta RF, et al. Adverse reactions associated
with the administration of a tricarbocyanine dye (Cardio-Green) to uremic
In summary, despite the changing landscape of retinal disease patients. J Allergy Clin Immunol 1971;48:235–9.
therapeutics with an increased emphasis on pharmacotherapies, 34. Iseki K, Onoyama K, Fujimi S, et al. Shock caused by indocyanine green dye
in chronic hemodialysis patients. Clin Nephrol 1980;14:210.
and the rise in prominence of noninvasive diagnostic technolo- 35. Robson SC, Mutch E, Boys RJ, et al. Apparent liver blood flow during preg-
gies such as optical coherence tomography, ICGA remains an nancy: a serial study using indocyanine green clearance. Br J Obstet Gynaecol
important tool in the diagnosis and management of a variety of 1990;97:720–4.
36. Flower RW, Csaky KG, Murphy RP. Disparity between fundus camera and
retinal disorders. scanning laser ophthalmoscope indocyanine green imaging of retinal pigment
epithelium detachments. Retina 1998;18:260–8.
REFERENCES 37. Wolf S, Wald KJ, Elsner AE, et al. Indocyanine green choroidal videoangio­
1. Cohen SY, Dubois L, Quentel G, et al. Is indocyanine green angiography still graphy: a comparison of imaging analysis with the scanning laser ophthal-
relevant? Retina 2011;31:209–21. moscope and the fundus camera. Retina 1993;13:266–9.
2. Ott P. Hepatic elimination of indocyanine green with special reference to 38. Stanga PE, Lim JI, Hamilton P. Indocyanine green angiography in chorioreti-
distribution kinetics and the influence of plasma protein binding. Pharmacol nal diseases: indications and interpretation: an evidence-based update.
Toxicol 1998;83(Suppl 2):1–48. Ophthalmology 2003;110:15–21; quiz 22-3.
3. Fox IJ, Brooker LG, Heseltine DW, et al. A tricarbocyanine dye for continuous 39. Yannuzzi LA, Flower RW, Slakter JS. Indocyanine green angiography.
recording of dilution curves in whole blood independent of variations in St Louis: Mosby Year Book; 1997. p. 46.
blood oxygen saturation. Proc Staff Meet Mayo Clin 1957;32:478–84. 40. Hayreh SS. Physiological anatomy of the choroidal vascular bed. Int
4. Fox IJ, Wood EH. Applications of dilution curves recorded from the right side Ophthalmol 1983;6:85–93.
of the heart or venous circulation with the aid of a new indicator dye. Proc 41. Mutoh T, Sakurai M, Tamai M. Indocyanine green fundus angiography of
Staff Meet Mayo Clin 1957;32:541–50. retrobulbar vasculature. Arch Ophthalmol 1995;113:631–3.
5. Caesar J, Shaldon S, Chiandussi L, et al. The use of indocyanine green in the 42. Ohno-Matsui K, Morishima N, Ito M, et al. Indocyanine green angiography
measurement of hepatic blood flow and as a test of hepatic function. Clin Sci of retrobulbar vascular structures in severe myopia. Am J Ophthalmol
1961;21:43–57. 1997;123:494–505.
6. Hunton DB, Bollman JL, Hoffman HN. Studies of hepatic function with indo- 43. Gass JD. Biomicroscopic and histopathologic considerations regarding the
cyanine green. Gastroenterology 1960;39:713–24. feasibility of surgical excision of subfoveal neovascular membranes. Am J
7. Kogure K, Choromokos E. Infrared absorption angiography. J Appl Physiol Ophthalmol 1994;118:285–98.
1969;26:154–7. 44. Macular Photocoagulation Study Group. Subfoveal neovascular lesions in
8. Kogure K, David NJ, Yamanouchi U, et al. Infrared absorption angiography age-related macular degeneration. Guidelines for evaluation and treatment in
of the fundus circulation. Arch Ophthalmol 1970;83:209–14. the macular photocoagulation study. Arch Ophthalmol 1991;109:1242–57.
9. Choromokos E, Kogure K, David NJ. Infrared absorption angiography. J Biol 45. Sadda SR, Liakopoulos S, Keane PA, et al. Relationship between angiographic
Photogr Assoc 1969;37:100–4. and optical coherence tomographic (OCT) parameters for quantifying choroi-
10. Hochheimer BF. Angiography of the retina with indocyanine green. dal neovascular lesions. Graefes Arch Clin Exp Ophthalmol 2010;248:175–84.
Arch Ophthalmol 1971;86:564–5. 46. Freund KB, Ho IV, Barbazetto IA, et al. Type 3 neovascularization: the
11. Flower RW, Hochheimer BF. Clinical infrared absorption angiography of the expanded spectrum of retinal angiomatous proliferation. Retina 2008;28:
choroid. Am J Ophthalmol. 1972;73:458–9. 201–11.
12. Hyvarinen L, Flower RW. Indocyanine green fluorescence angiography. 47. Olsen TW, Feng X, Kasper TJ, et al. Fluorescein angiographic lesion type
Acta Ophthalmol (Copenh) 1980;58:528–38. frequency in neovascular age-related macular degeneration. Ophthalmology
13. Hayashi K, de Laey JJ. Indocyanine green angiography of submacular choroi- 2004;111:250–5.
dal vessels in the human eye. Ophthalmologica 1985;190:20–9. 48. Guyer DR, Yannuzzi LA, Slakter JS, et al. Classification of choroidal neovas-
14. Hayashi K, Hasegawa Y, Tokoro T. Indocyanine green angiography of central cularization by digital indocyanine green videoangiography. Ophthalmology
serous chorioretinopathy. Int Ophthalmol 1986;9:37–41. 1996;103:2054–60.
49. Yannuzzi LA, Hope-Ross M, Slakter JS, et al. Analysis of vascularized pigment 79. Guyer DR, Yannuzzi LA, Slakter JS, et al. Digital indocyanine green videoan-
epithelial detachments using indocyanine green videoangiography. Retina giography of central serous chorioretinopathy. Arch Ophthalmol 1994;112:
1994;14:99–113. 1057–62.
80 50. Brown DM, Kaiser PK, Michels M, et al. Ranibizumab versus verteporfin for 80. Yannuzzi LA, Slakter JS, Gross NE, et al. Indocyanine green angiography-
neovascular age-related macular degeneration. N Engl J Med 2006;355: guided photodynamic therapy for treatment of chronic central serous
1432–44. chorioretinopathy: a pilot study. Retina 2003;23:288–98.
51. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizumab for neovascular age- 81. Piccolino FC, Borgia L. Central serous chorioretinopathy and indocyanine
related macular degeneration. N Engl J Med 2006;355:1419–31. green angiography. Retina 1994;14:231–42.
Section 1

52. Massacesi AL, Sacchi L, Bergamini F, et al. The prevalence of retinal angio- 82. Spaide RF, Hall L, Haas A, et al. Indocyanine green videoangiography of older
matous proliferation in age-related macular degeneration with occult choroi- patients with central serous chorioretinopathy. Retina 1996;16:203–13.
dal neovascularization. Graefes Arch Clin Exp Ophthalmol 2008;246:89–92. 83. Shiraki K, Moriwaki M, Matsumoto M, et al. Long-term follow-up of severe
53. Lim JI, Aaberg TM, Capone AJ, et al. Indocyanine green angiography-guided central serous chorioretinopathy using indocyanine green angiography.
photocoagulation of choroidal neovascularization associated with retinal Int Ophthalmol 1997;21:245–53.
pigment epithelial detachment. Am J Ophthalmol 1997;123:524–32. 84. Lafaut BA, Salati C, Priem H, et al. Indocyanine green angiography is of value
54. Watzke RC, Klein ML, Hiner CJ, et al. A comparison of stereoscopic fluores- for the diagnosis of chronic central serous chorioretinopathy in elderly
cein angiography with indocyanine green videoangiography in age-related patients. Graefes Arch Clin Exp Ophthalmol 1998;236:513–21.
Optical Imaging Technologies

macular degeneration. Ophthalmology 2000;107:1601–6. 85. Reibaldi M, Cardascia N, Longo A, et al. Standard-fluence versus low-fluence
Retinal Imaging and Diagnostics

55. Shiraga F, Ojima Y, Matsuo T, et al. Feeder vessel photocoagulation of subfo- photodynamic therapy in chronic central serous chorioretinopathy: a nonran-
veal choroidal neovascularization secondary to age-related macular degenera- domized clinical trial. Am J Ophthalmol 2010;149:307–15.e2.
tion. Ophthalmology 1998;105:662–9. 86. Chan WM, Lam DS, Lai TY, et al. Choroidal vascular remodelling in central
56. Staurenghi G, Orzalesi N, La Capria A, et al. Laser treatment of feeder vessels serous chorioretinopathy after indocyanine green guided photodynamic
in subfoveal choroidal neovascular membranes: a revisitation using dynamic therapy with verteporfin: a novel treatment at the primary disease level.
indocyanine green angiography. Ophthalmology 1998;105:2297–305. Br J Ophthalmol 2003;87:1453–8.
57. Bottoni F, Massacesi A, Cigada M, et al. Treatment of retinal angiomatous 87. Inoue R, Sawa M, Tsujikawa M, et al. Association between the efficacy of
proliferation in age-related macular degeneration: a series of 104 cases of photodynamic therapy and indocyanine green angiography findings for
retinal angiomatous proliferation. Arch Ophthalmol 2005;123:1644–50. central serous chorioretinopathy. Am J Ophthalmol 2010;149:441–6.e1-2.
58. Flower RW. Optimizing treatment of choroidal neovascularization feeder 88. Tsujikawa A, Ojima Y, Yamashiro K, et al. Punctate hyperfluorescent spots
vessels associated with age-related macular degeneration. Am J Ophthalmol associated with central serous chorioretinopathy as seen on indocyanine
2002;134:228–39. green angiography. Retina 2010;30:801–9.
59. Semoun O, Guigui B, Tick S, et al. Infrared features of classic choroidal 89. Kitaya N, Nagaoka T, Hikichi T, et al. Features of abnormal choroidal circula-
neovascularisation in exudative age-related macular degeneration. Br J tion in central serous chorioretinopathy. Br J Ophthalmol 2003;87:709–12.
Ophthalmol 2009;93:182–5. 90. Prunte C, Flammer J. Choroidal capillary and venous congestion in central
60. Schmidt-Erfurth U, Kriechbaum K, Oldag A. Three-dimensional angiography serous chorioretinopathy. Am J Ophthalmol 1996;121:26–34.
of classic and occult lesion types in choroidal neovascularization. Invest 91. Anand R, Augsburger JJ, Shields JA. Circumscribed choroidal hemangiomas.
Ophthalmol Vis Sci 2007;48:1751–60. Arch Ophthalmol 1989;107:1338–42.
61. Hlushchuk R, Baum O, Gruber G, et al. The synergistic action of a VEGF- 92. Shields CL, Honavar SG, Shields JA, et al. Circumscribed choroidal heman-
receptor tyrosine-kinase inhibitor and a sensitizing PDGF-receptor blocker gioma: clinical manifestations and factors predictive of visual outcome in 200
depends upon the stage of vascular maturation. Microcirculation 2007;14: consecutive cases. Ophthalmology 2001;108:2237–48.
813–25. 93. Witschel H, Font RL. Hemangioma of the choroid. A clinicopathologic study
62. Yannuzzi LA, Negrao S, Iida T, et al. Retinal angiomatous proliferation in of 71 cases and a review of the literature. Surv Ophthalmol 1976;20:
age-related macular degeneration. Retina 2001;21:416–34. 415–31.
63. Gass JD, Agarwal A, Lavina AM, et al. Focal inner retinal hemorrhages in 94. Singh AD, Kaiser PK, Sears JE. Choroidal hemangioma. Ophthalmol Clin
patients with drusen: an early sign of occult choroidal neovascularization and North Am 2005;18:151–61. ix.
chorioretinal anastomosis. Retina 2003;23:741–51. 95. Arevalo JF, Shields CL, Shields JA, et al. Circumscribed choroidal hemangi-
64. Hartnett ME, Weiter JJ, Staurenghi G, et al. Deep retinal vascular anomalous oma: characteristic features with indocyanine green videoangiography.
complexes in advanced age-related macular degeneration. Ophthalmology Ophthalmology 2000;107:344–50.
1996;103:2042–53. 96. Shields CL, Shields JA, De Potter P. Patterns of indocyanine green video­
65. Viola F, Massacesi A, Orzalesi N, et al. Retinal angiomatous proliferation: angiography of choroidal tumours. Br J Ophthalmol 1995;79:237–45.
natural history and progression of visual loss. Retina 2009;29:732–9. 97. Sallet G, Amoaku WM, Lafaut BA, et al. Indocyanine green angiography of
66. Rouvas AA, Papakostas TD, Vavvas D, et al. Intravitreal ranibizumab, intra- choroidal tumors. Graefes Arch Clin Exp Ophthalmol 1995;233:677–89.
vitreal ranibizumab with PDT, and intravitreal triamcinolone with PDT for 98. Andersen MV, Scherfig E, Prause JU. Differential diagnosis of choroidal mela-
the treatment of retinal angiomatous proliferation: a prospective study. Retina nomas and nevi using scanning laser ophthalmoscopical indocyanine green
2009;29:536–44. angiography. Acta Ophthalmol Scand 1995;73:453–6.
67. Saito M, Shiragami C, Shiraga F, et al. Comparison of intravitreal triamcino- 99. Mueller AJ, Bartsch DU, Folberg R, et al. Imaging the microvasculature of
lone acetonide with photodynamic therapy and intravitreal bevacizumab choroidal melanomas with confocal indocyanine green scanning laser
with photodynamic therapy for retinal angiomatous proliferation. Am J ophthalmoscopy. Arch Ophthalmol 1998;116:31–9.
Ophthalmol 2010;149:472–81.e1. 100. Frenkel S, Barzel I, Levy J, et al. Demonstrating circulation in vasculogenic
68. Bottoni F, Romano M, Massacesi A, et al. Remodeling of the vascular channels mimicry patterns of uveal melanoma by confocal indocyanine green angio­
in retinal angiomatous proliferations treated with intravitreal triamcinolone graphy. Eye (Lond) 2008;22:948–52.
acetonide and photodynamic therapy. Graefes Arch Clin Exp Ophthalmol 101. Mueller AJ, Freeman WR, Schaller UC, et al. Complex microcirculation
2006;244:1528–33. patterns detected by confocal indocyanine green angiography predict
69. Yannuzzi LA, Sorenson J, Spaide RF, et al. Idiopathic polypoidal choroidal time to growth of small choroidal melanocytic tumors: MuSIC Report II.
vasculopathy (IPCV). Retina 1990;10:1–8. Ophthalmology 2002;109:2207–14.
70. Spaide RF, Yannuzzi LA, Slakter JS, et al. Indocyanine green videoangio­ 102. Schaller UC, Mueller AJ, Bartsch DU, et al. Choroidal melanoma microcircula-
graphy of idiopathic polypoidal choroidal vasculopathy. Retina 1995;15: tion with confocal indocyanine green angiography before and 1 year after
100–10. radiation brachytherapy. Retina 2000;20:627–32.
71. Yannuzzi LA, Wong DW, Sforzolini BS, et al. Polypoidal choroidal vasculopa- 103. Krause L, Bechrakis NE, Heinrich S, et al. Indocyanine green angiography and
thy and neovascularized age-related macular degeneration. Arch Ophthalmol fluorescein angiography of malignant choroidal melanomas following proton
1999;117:1503–10. beam irradiation. Graefes Arch Clin Exp Ophthalmol 2005;243:545–50.
72. Ahuja RM, Stanga PE, Vingerling JR, et al. Polypoidal choroidal vasculopathy 104. Midena E, Pilotto E, de Belvis V et al. Choroidal vascular changes after trans-
in exudative and haemorrhagic pigment epithelial detachments. Br J Ophthal- pupillary thermotherapy for choroidal melanoma. Ophthalmology 2003;110:
mol 2000;84:479–84. 2216–22.
73. Ciardella AP, Donsoff IM, Huang SJ, et al. Polypoidal choroidal vasculopathy. 105. Annesley WHJ. Peripheral exudative hemorrhagic chorioretinopathy. Trans
Surv Ophthalmol 2004;49:25–37. Am Ophthalmol Soc 1980;78:321–64.
74. Ahuja RM, Downes SM, Stanga PE, et al. Polypoidal choroidal vasculopathy 106. Shields CL, Salazar PF, Mashayekhi A, et al. Peripheral exudative hemor-
and central serous chorioretinopathy. Ophthalmology 2001;108:1009–10. rhagic chorioretinopathy simulating choroidal melanoma in 173 eyes.
75. Yannuzzi LA, Freund KB, Goldbaum M, et al. Polypoidal choroidal vascu- Ophthalmology 2009;116:529–35.
lopathy masquerading as central serous chorioretinopathy. Ophthalmology 107. Gunduz K, Shields CL, Shields JA. Varix of the vortex vein ampulla simulat-
2000;107:767–77. ing choroidal melanoma: report of four cases. Retina 1998;18:343–7.
76. Maruko I, Iida T, Saito M, et al. Combined cases of polypoidal choroidal 108. Rutnin U. Fundus appearance in normal eyes. I. The choroid. Am J Ophthal-
vasculopathy and typical age-related macular degeneration. Graefes Arch mol 1967;64:821–39.
Clin Exp Ophthalmol 2010;248:361–8. 109. Osher RH, Abrams GW, Yarian D, et al. Varix of the vortex ampulla. Am J
77. Lai TY, Chan WM, Liu DT, et al. Intravitreal bevacizumab (Avastin) with or Ophthalmol 1981;92:653–60.
without photodynamic therapy for the treatment of polypoidal choroidal 110. Singh AD, De Potter P, Shields CL, et al. Indocyanine green angiography and
vasculopathy. Br J Ophthalmol 2008;92:661–6. ultrasonography of a varix of vortex vein. Arch Ophthalmol 1993;111:
78. Eandi CM, Ober MD, Freund KB, et al. Selective photodynamic therapy for 1283–4.
neovascular age-related macular degeneration with polypoidal choroidal 111. Gross NE, Yannuzzi LA, Freund KB, et al. Multiple evanescent white dot
neovascularization. Retina 2007;27:825–31. syndrome. Arch Ophthalmol 2006;124:493–500.
112. Dell’omo R, Wong R, Marino M, et al. Relationship between different fluores- 119. Howe LJ, Woon H, Graham EM, et al. Choroidal hypoperfusion in acute
cein and indocyanine green angiography features in multiple evanescent posterior multifocal placoid pigment epitheliopathy. An indocyanine green
white dot syndrome. Br J Ophthalmol 2010;94:59–63. angiography study. Ophthalmology 1995;102:790–8.
113. Tsukamoto E, Yamada T, Kadoi C, et al. Hypofluorescent spots on indocya- 120. Schneider U, Inhoffen W, Gelisken F. Indocyanine green angiography in a 81
nine green angiography at the recovery stage in multiple evanescent white case of unilateral recurrent posterior acute multifocal placoid pigment
dot syndrome. Ophthalmologica 1999;213:336–8. epitheliopathy. Acta Ophthalmol Scand 2003;81:72–5.
114. Slakter JS, Giovannini A, Yannuzzi LA et al. Indocyanine green angiography 121. Giovannini A, Mariotti C, Ripa E, et al. Indocyanine green angiographic find-
of multifocal choroiditis. Ophthalmology 1997;104:1813–9. ings in serpiginous choroidopathy. Br J Ophthalmol 1996;80:536–40.

Chapter 2
115. Fardeau C, Herbort CP, Kullmann N, et al. Indocyanine green angiography 122. Amer R, Lois N. Punctate inner choroidopathy. Surv Ophthalmol 2011;56:
in birdshot chorioretinopathy. Ophthalmology 1999;106:1928–34. 36–53.
116. Howe LJ, Stanford MR, Graham EM, et al. Choroidal abnormalities in birdshot 123. Tiffin PA, Maini R, Roxburgh ST, et al. Indocyanine green angiography in a
chorioretinopathy: an indocyanine green angiography study. Eye (Lond) case of punctate inner choroidopathy. Br J Ophthalmol 1996;80:90–1.
1997;11:554–9. 124. Levy J, Shneck M, Klemperer I, et al. Punctate inner choroidopathy: resolution
117. Trinh L, Bodaghi B, Fardeau C et al. Clinical features, treatment methods, after oral steroid treatment and review of the literature. Can J Ophthalmol
and evolution of birdshot chorioretinopathy in 5 different families. Am J 2005;40:605–8.
Ophthalmol 2009;147:1042–7. 1047.e1. 125. Spaide RF. Collateral damage in acute zonal occult outer retinopathy. Am J
118. Park D, Schatz H, McDonald HR, et al. Indocyanine green angiography of Ophthalmol 2004;138:887–9.

Clinical Applications of Diagnostic Indocyanine Green Angiography


acute multifocal posterior placoid pigment epitheliopathy. Ophthalmology
1995;102:1877–83.
Section 1 Optical Imaging Technologies

Chapter Optical Coherence Tomography


3  Carlos Alexandre de Amorim Garcia Filho, Zohar Yehoshua,
Giovanni Gregori, Carmen A. Puliafito, Philip J. Rosenfeld

PHYSICAL PRINCIPLES OF OPTICAL The scanning pattern with the commercial TD-OCT instru-
ment (Stratus OCT, Carl Zeiss Meditec, Dublin, CA) incorpo-
COHERENCE TOMOGRAPHY
rated six radial, concentric, 6-mm-long B-scans centered on the
During the past two decades, optical coherence tomography fovea. With the recent development of high-speed SD-OCT
(OCT) has become an essential tool in ophthalmology. Its ability systems, several novel and important imaging strategies have
to image detailed ocular structures noninvasively in vivo with been introduced based on acquiring three-dimensional datasets
high resolution has revolutionized patient care.1,2 OCT technol- and B-scan averaging (Fig. 3.1).
ogy is based on the principle of low-coherence interferometry, Three-dimensional datasets are obtained using a dense two-
where a low-coherence (high-bandwidth) light beam is directed dimensional raster array over a relatively large retinal region.
on to the target tissue and the scattered back-reflected light is The resulting datasets can be rendered as a volume image in
combined with a second beam (reference beam), which was split three dimensions and can be analyzed by showing two-
off from the original light beam. The resulting interference pat- dimensional slices (i.e., sequences of parallel B-scans). Three-
terns are used to reconstruct an axial A-scan, which represents dimensional datasets give detailed information about the retinal
the scattering properties of the tissue along the beam path. structure over large areas. In addition, it is possible to generate
Moving the beam of light along the tissue in a line results in a en face fundus-like images directly from the OCT datasets. These
compilation of A-scans with each A-scan having a different inci- OCT fundus images (OFIs) provide an accurate spatial colocal-
dence point. From all these A-scans, a two-dimensional cross- ization of retinal features observed on the en face and cross-
sectional image of the target tissue can be reconstructed and this sectional images. Therefore, exact correlations can be achieved
is known as a B-scan. between the retinal cross-sectional geometry seen on the OCT
Typically OCT instruments use an infrared light source cen- B-scans and the retinal landmarks seen on en face images,
tered at a wavelength of about 840  nm. For a given wavelength, known as the OFI. The potential exists for registration between
the axial resolution is dictated by the bandwidth of the light several SD-OCT datasets of the same eye and images obtained
source. The latest commercial instruments typically have an using other imaging modalities, such as color fundus photo­
axial resolution of approximately 5  µm, while research instru- graphy, fluorescein angiography, and fundus autofluorescence
ments have been built with a resolution as high as approxi- imaging. This holds the promise for an unprecedented ability
mately 2  µm.1 The lateral resolution is limited by the diffraction
caused by the pupil and it is normally about 20  µm. For clini-
cal purposes, the image acquisition time is limited by the
patient’s ability to avoid eye movements, i.e., less than 2 seconds
in the typical patient. The instrument’s scanning speed (number
of A-scans acquired per second) is then the crucial parameter
determining the amount of data available for a single OCT
dataset.
The early OCT instruments, known as time domain OCT (TD-
OCT), used a single photo detector, and an A-scan was created
by moving a mirror to change the optical path of the reference
beam in order to match different axial depths in the target tissue.
This setup limited the scanning speed to a few thousand A-scans
per second. A newer technique, known as spectral domain OCT
(SD-OCT), Fourier domain OCT (FD-OCT), or high-definition
OCT (HD-OCT), is able to acquire an entire A-scan by using an
array of detectors instead of using multiple reference beams
from a moving mirror. Scanning speeds with SD-OCT instru-
ments can exceed 100 000 A-scans per second, about 200 times
faster than TD-OCT. Currently available SD-OCT commercial
systems operate at a scanning rate of approximately 27 000 Fig. 3.1 Three-dimensional dataset. (Courtesy of Cirrus HD-OCT, Carl
A-scans per second.1 Zeiss Meditec, Dublin, CA.)
to describe and monitor changes in the local geometry of the improvement of SD-OCT. It is not an objective of this section to
retina.3 In addition to the OFI generated by a full OCT dataset, discuss the differences between each of the currently available
partial OFIs (or slabs) can be generated to produce en face ren- instruments since these instruments are continuously evolving. 83
derings that correspond to particular retinal layers or features.4 Table 3.1 lists currently available instruments.
These slabs can be very useful to visualize and quantify specific

Chapter 3
pathologies (Fig. 3.2). QUANTITATIVE ANALYSIS OF
The scanning speed of SD-OCT can also be used to produce
very high-quality individual B-scan images through a combina-
OCT DATASETS
tion of high sampling density and image averaging. One of the A crucial step towards a clinically useful, quantitative under-
main factors affecting the perceived quality of OCT images is standing of the retinal anatomy is the development of accurate,
noise, in particular the speckle noise which is responsible for the robust, reproducible segmentation algorithms that can automati-
characteristic “granular” appearance of OCT. Noise can be cally identify the boundaries between specific retinal layers

Optical Coherence Tomography


reduced through the acquisition, registration, and averaging of and/or other retinal features. The currently available SD-OCT
a number of B-scans at approximately the same retinal position instruments have several advantages over the previous genera-
(Fig. 3.3). tion of TD-OCT instruments. SD-OCT instruments generally
Although en face registration and B-scan averaging strategies have a higher axial resolution and can produce B-scans with
can be implemented in many ways, a particularly powerful and better image quality by increasing the A-scan density and
flexible solution is the use of a built-in laser eye-tracking system. through averaging techniques. Much more importantly, the
The main limitation of this approach is that the necessary acqui- higher scanning speed made possible by the SD-OCT technology
sition times can become very long and sometimes unmanage- reduces the effect of artifacts associated with eye motion
able, particularly for large raster scans and for subjects with poor and produces images that provide a true picture of the retinal
fixation. geometry. The large, dense raster scans make it possible to
Recently, different companies have invested in research in the obtain detailed surfaces of individual retina layers over large
field of retinal imaging, especially in the development and areas, resulting in segmentation maps. These maps allow for an

A B C

Fig. 3.2 (A) Color fundus image of a patient with geographic atrophy secondary to age-related macular degeneration. (B) Optical coherence
tomography (OCT) fundus image, which is the en face image from the reflected light from each A-scan, of the same patient obtained with a
scan pattern of 200 × 200 A-scans in the Cirrus high-definition OCT instrument. (C) Registration of the color fundus image with the OCT fundus
image. Since the area of the OCT fundus image is known to be 6 × 6 mm, it is possible to quantify lesion area and calibrate the fundus camera
to use this technique.

A B

Fig. 3.3 Averaging process. (A) Multiple B-scans acquired through the foveal center of a normal patient. (B) The registration and averaging of
these B-scans can reduce the speckled noise and improve the image quality. In these examples the image was averaged 20 times using the
Cirrus high-definition optical coherence tomograph.
Table 3.1 Commercially available spectral domain optical unprecedented visualization and quantitative evaluation of the
coherence tomography (OCT) instruments corresponding retinal structures.
84 Several commercially available SD-OCT instruments offer
Device Axial resolution; Special
some level of quantitative analysis using different, proprietary
(manufacturer) scanning rate characteristics
segmentation algorithms. The various segmentation algorithms
make different design choices and have been shown to have
Section 1

3D-OCT 2000 5 µm; 27 kHz Fundus camera


(Topcon, Tokyo, very different performance profiles in terms of accuracy,
Japan) reproducibility, and robustness.5–8 Care should be exercised
Bioptigen SD-OCT 4 µm; 20 kHz Designed for
when comparing measurements obtained from different OCT
(Bioptigen, Research research instruments.
Triangle Park, NC) applications The most commonly used quantitative parameter derived
from OCT datasets is retinal thickness, obtained by segmenting
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Cirrus HD-OCT 5 µm; 27 kHz


(Carl Zeiss Meditec,
the internal limiting membrane (ILM) and a boundary represent-
Dublin, CA) ing the retinal pigment epithelium (RPE). This information can
be used to generate surface maps of the ILM and the RPE as well
RTVue-100 5 µm; 26 kHz as two-dimensional and three-dimensional retinal thickness
(Optovue,
Fremont, CA)
maps. These maps can be very useful in identifying and describ-
ing deviations from the normal anatomy and changes over time.
SOCT Copernicus 6 µm; 27 kHz Registering OCT datasets acquired over time can give very
(Optopol, Zawiercie, precise information about the dynamics of disease progression
Poland)
and response to treatment based on changes in retinal anatomy
Spectral OCT SLO 6 µm; 27 kHz Microperimetry (Fig. 3.4).
(Opko, Miami, FL) It is important to keep in mind that there is some confu-
sion in the definition of the outer retinal boundary. In a
Spectralis OCT 8 µm; 40 kHz Eye-tracking, normal eye, the bright reflective band at the external aspect
(Heidelberg fluorescein
Engineering, angiography, ICG of the retina, often referred to as the RPE complex, can be
Heidelberg, angiography, resolved in ultrahigh-resolution images, and occasionally in
Germany) autofluorescence images acquired with a commercially available SD-OCT instru-
3D, three-dimensional; SD, spectral domain; HD, high-definition; ment, consisting of three individual layers.9 Different segmen-
SOCT, spectral optical coherence tomography; SLO, scanning laser tation algorithms from different instruments tend to follow
ophthalmoscope; ICG, indocyanine green.

B C D

Fig. 3.4 Segmentation process. (A) B-scan through the foveal center of a normal patient with a yellow line identifying the internal limiting
membrane (ILM) and a red line corresponding to the retinal pigment epithelium (RPE). Three-dimensional map of the ILM (B), RPE (C), and
the retinal thickness map (D) acquired with a 200 × 200 scan pattern with the Cirrus high-definition optical coherence tomography instrument.
85

A B C

Chapter 3
Optical Coherence Tomography
D E F

Fig. 3.5 Differences in segmentation and retinal thickness maps between instruments. The B-scan, the B-scan identifying the internal limiting
membrane (yellow line), and the retinal pigment epithelium (red line), and the retinal thickness map acquired with the Cirrus high-definition
optical coherence tomograph (A–C) and the Spectralis (D–F). Note that, using the Cirrus instrument, the segmentation algorithm identifies the
actual retinal pigment epithelium (B) and using the Spectralis the segmentation algorithm identifies Bruch’s membrane. This subtle difference in
the segmentation algorithm between each instrument can be responsible for different retinal thickness measurements.

different edges and therefore result in different measurements.


For example, the Spectralis SD-OCT instrument typically
NORMAL MACULAR ANATOMY
follows the posterior edge of the RPE complex, the Stratus The OCT image closely approximates the histological appear-
TD-OCT instrument typically follows the inner segment–outer ance of the macula and, for this reason, it has been referred to
segment (IS/OS) junction, which is anterior to the RPE as an in vivo optical biopsy. With the increase in the axial
complex, and the Cirrus SD-OCT instrument typically follows resolution of the new SD-OCT instruments (5–8  µm) and the
the anterior edge of the RPE layer (Fig. 3.5). This situation ultrahigh-resolution OCT (2  µm), it has become possible to cor-
becomes even more complicated and sometimes inconsistent relate OCT images accurately with histological features of the
when the normal retinal structure is deformed by the pres- retina.20 However, care must be taken when making assump-
ence of pathology.10 tions about these correlations because histological sections
In addition to total retinal thickness, a number of other require fixation and exogenous staining to produce contrast
quantitative parameters have been proposed. For example, it within tissue, and this can introduce artifacts, while OCT relies
is possible to obtain measurements of particular retinal layers, on intrinsic differences in tissue optical properties to produce
such as the thickness of the ganglion cell layer or the thick- image contrast.21 When light travels through the retinal tissue
ness of the photoreceptors’ outer segments, as well as mea- it can be reflected, scattered, or absorbed, and this creates the
surements of retinal lesions, like the area of geographic multilayered pattern of the retina. The angle of incidence of
atrophy (GA).9,11,12 the light, motion artifacts, speckled noise, and image contrast
An area of particular promise is the measurement of RPE can affect the axial resolution of the retinal imaging. Therefore,
deformations associated with drusen.13,14 These measurements one-to-one correspondence of histology with OCT images
are obtained by comparing the actual RPE geometry with the cannot be expected.2,21
geometry of a virtual RPE free of deformations. Parameters Although the interpretation of features of the inner retina,
like drusen area and volume can be generated in a fully auto- which can be defined for our purpose to span from the ILM to
mated manner and have been shown to be quite robust and the junction of the inner and outer segments of the photorecep-
reproducible (Fig. 3.6). tors, appears to correlate well with histology, the OCT features
The amount of information provided by each dataset, together of the outer retina are less well understood and remain a topic
with the possibility for image registration and longitudinal of discussion (Fig. 3.7).9,22,23
studies, makes SD-OCT a very promising new tool for the quan- The first detected layer in most OCT scans is the ILM that
titative study of retinal pathologies. These capabilities solve the appears as a hyperreflective layer at the vitreoretinal interface.
biggest problems associated with the Stratus TD-OCT retinal In some patients, the posterior hyaloid can be seen above the
thickness maps: the lack of precise correspondence between the ILM as a hyperreflective layer. Within the retina, the retinal
B-scans and the retinal topography, the difficulties associated nerve fiber layer and the plexiform layers (both inner and outer)
with eye movements, and the need for significant interpolation are seen as hyperreflective while the ganglion cell layer and the
of the data due to undersampling of the retina. Despite the nuclear layers (both inner and outer) are hyporeflective. A recent
advantages of SD-OCT, segmentation algorithms can produce study demonstrated that the incidence of the light beam could
artifacts, particularly in the presence of macular disorders with affect the appearance of Henle’s fiber layer by OCT, resulting in
complex morphology like neovascular age-related macular a thin hyperreflective layer corresponding to the photoreceptor
degeneration (AMD).15–19 Therefore, it is important to be vigilant synapses or a thicker hyperreflective layer corresponding to
and monitor the quality of the segmentation in order to eliminate photoreceptor axonal extensions enveloped by the outer cyto-
artifacts arising from flawed segmentation and associated plasm of Müller cells (Fig. 3.8).24 The retinal vessels may some-
measurements. times be seen on OCT images as circular hyperreflective
86
Section 1

B
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A C
S

D E F

Fig. 3.6 Retinal pigment epithelium (RPE) deformation algorithm. (A) Color fundus image of a patient with drusen. A 6 × 6 mm white box
was superimposed on the image to represent the scan area. (B) B-scan from the spectral domain optical coherence tomography dataset that
corresponds to the central line on the color fundus image. (C) B-scan with a yellow line representing the RPE segmentation and a red line
showing the RPE floor (virtual map of the RPE free of deformations). (D) En face image of the 6 × 6 mm scan pattern (optical coherence
tomography fundus image). (E) Three-dimensional RPE map delineating the drusen conformation. (F) RPE elevation map with drusen area
(1.41 mm2) and volume (0.08 mm3).

NFL
GCL
IPL

OPL
ONL
ELM
IS/OS
RPE
Choroid
A B

Fig. 3.7 Spectral domain optical coherence tomography (Spectralis, Heidelberg) image of a normal individual. The multilayered retinal
architecture can be observed and each retinal layer can be identified. NFL, nerve fiber layer; GCL, ganglion cell layer; IPL, inner plexiform
layer; OPL, outer plexiform layer; ONL, outer nuclear layer; ELM, external limiting membrane; IS/OS, junction of the inner and outer segments
of the photoreceptors; RPE, retinal pigment epithelium.
87

Chapter 3
*

Optical Coherence Tomography


Fig. 3.8 Spectral domain optical coherence tomography of the
same patient using a different light incidence. This results in a thin Fig. 3.9 Enhanced-depth spectral domain optical coherence
hyperreflective layer that corresponds to the photoreceptor synapses tomography image (Spectralis, Heidelberg) of a normal subject
(white arrow) or a thicker hyperreflective layer corresponding to showing the boundaries of the choroid (arrowheads).
photoreceptor axonal extensions enveloped by the outer cytoplasm
of Müller cells (white asterisk).

allows a better visualization of the posterior choroid and sclera


than currently available SD-OCT instruments.30,33,34
structures located in the inner retina, with a vertical shadow or The high axial resolution and the different scan patterns
reduced reflectivity extending into deeper layers. offered by SD-OCT provide comprehensive structural infor-
Outside the central fovea, commercially available SD-OCT mation that can be used to map retinal layer thicknesses and
instruments typically resolve four bands in the outer retina. perform volumetric analyses. Using different SD-OCT instru-
There is discordance between different authors regarding which ments, several authors have reported an approximate central
anatomical structure correlates with each band.20,21,25 The inner- retinal thickness of 265  µm in normal subjects.35 However,
most band has been attributed to the external limiting membrane caution is required, as errors in automated measurements
(ELM). This band is typically thinner and fainter than the others. may occur and are more often found in macular disorders
The nomenclature for the middle two bands has much less sup- with complex morphology like neovascular AMD, which
portive evidence. The second of the four bands has been com- alters the ability of segmentation algorithms to detect normal
monly ascribed to the boundary between the IS/OS of the boundaries.15–19 Therefore, care must be taken that high-
photoreceptors and the third band is referred to as either the OS quality and artifact-free scans are obtained before running
tips or as Verhoeff’s membrane.9,26 A recent study suggested that the retinal thickness algorithm.
the second band was the ellipsoid section of the photoreceptors It is essential to consider the following points when analyzing
(inner segment) instead of the IS/OS junction and that the third OCT images: location, shape, and reflectivity of the structure,
band appears to correspond to the contact cylinder between the along with its histological correlation. It is also important to
RPE apical process and the external portion of the cone outer remember that the alignment of the instrument with the pupil
segment. This band typically merges with the fourth band in the can generate signals that may lead to a misinterpretation of
central fovea and this is explained by a greater height of the exam.
the contact cylinder of the cones and RPE outside the fovea.25 Characteristic OCT findings in several common retinal disor-
The fourth hyperreflective outer retinal band is attributed ders which are frequently studied using OCT are discussed
to the RPE, with potential contribution from Bruch’s membrane below. For OCT findings, in other disorders, such as retinal
and choriocapillaris, with abundant experimental and clinical degenerations, the reader is directed to the specific chapters
evidence supporting this designation.9,22,27 describing these diseases.
Although the current SD-OCT uses a short wavelength of
approximately 840 nm, which results in light scattering at the
SD-OCT IN RETINAL DISORDERS
level of the RPE and a lower signal from the deep choroidal Vitreoretinal interface disorders
tissue, it is also possible to image the choroid and extract quan-
Abnormalities of the vitreoretinal interface are involved in the
titative information (Fig. 3.9).28–31 Choroidal thickness may be
pathogenesis of several macular conditions. In idiopathic epireti-
influenced by age, axial length, and perhaps refractive abnor-
nal membranes (ERMs), a layer of fibrotic tissue develops on the
malities. It also varies in different retinal regions within the same
surface of the retina, usually after a posterior vitreous detach-
normal subject, being thickest beneath the fovea,31 or in the supe-
ment. Contraction of this membrane can result in retinal dis­
rior outer macula (Early Treatment Diabetic Retinopathy Study
tortion, leading to vision loss. In other conditions, such as
(ETDRS) subfield), with the thinnest choroid being located in the
vitreomacular traction (VMT) syndrome or idiopathic macular
nasal outer ETDRS subfield.32 When centering the optic nerve
hole, there are abnormal attachments between the vitreous and
head as a reference point, the choroid appears thin in the peri-
the retina. The resulting traction exerted on the retina causes
papillary region and increases in thickness with eccentricity in
anatomical alteration and subsequent visual loss.
all directions, up to a certain point, except inferiorly.32 This is the
embryonic location of the optic fissure closure and thus may be Vitreomacular traction
responsible for the localized thinning.32,33 High-penetration OCT VMT syndrome results from persistent vitreoretinal adhesions
uses a light source with a wavelength around 1050 nm that in the setting of a partial posterior vitreous detachment.36 In
normal eyes, as the vitreous liquefies due to age, it detaches Epiretinal membrane
from the macula. This natural progression has been demon- ERM occurs in approximately 6% of patients over the age of 60,
88 strated using OCT.37 In some people, an unusually strong with incidence increasing with age.49,50 ERMs can be classified as
adhesion is present between the vitreous and macula, and as idiopathic or secondary to an initiating event. Most idiopathic
the vitreous detaches peripherally, it continues to pull on ERMs are thought to result from fibroglial proliferation on the
areas of the macula. The vitreoretinal adhesions transmit trac- inner surface of the retina secondary to a break in ILM occurring
Section 1

tional forces to the retina from the vitreous body, having the during posterior vitreous detachment.51,52 Secondary ERMs
potential to cause tensile deformation, foveal cavitations, cystoid result from an already-existing ocular pathology such as central
macular edema (CME), limited macular detachment, or a or branch retinal vein occlusion, diabetic retinopathy, uveitis,
macular hole.38,39 Patients can present with visual loss and and retinal breaks with or without detachment.53 Glial cells, RPE
metamorphopsia. cells, and myofibroblasts are shown to be mostly involved in
Diagnosis of VMT by biomicroscopy may be challenging, par- ERM formation.51,52 ERM may lead to loss of normal retinal
Optical Imaging Technologies
Retinal Imaging and Diagnostics

ticularly when the area of vitreoretinal attachment is broad. OCT anatomy, with the patient experiencing metamorphopsia,
better defines the vitreoretinal relationships in eyes with VMT micropsia, monocular diplopia, and decreased visual acuity.
and also documents concomitant ERM and macular edema.40–44 These symptoms vary in severity depending on the location,
With OCT imaging, the abnormal VMT bands from the promi- density, and contraction of the membrane.
nent posterior hyaloid are well delineated as reflective lines from On slit-lamp biomicroscopy, a mild ERM appears as a glisten-
the perifoveal area into the vitreous cavity, distorting the macular ing layer on the retinal surface. Denser membranes may be seen
contour with or without accumulation of intraretinal or subreti- as a gray sheet overlying the retina and causing distortion in the
nal fluid (Fig. 3.10). macular vascular architecture. Occasionally, ERMs can evolve
In recent years OCT has been most beneficial in diagnosing into macular pseudoholes and ERMs are often seen in conjunc-
VMT and subsequently directing treatment of this condition. tion with idiopathic full-thickness macular holes.38 Fluorescein
In some cases, spontaneous resolution can occur with separa- angiography may demonstrate macular leakage, which can be
tion of the vitreous from the macula, leading to subsequent variable from case to case.
resolution of the intraretinal and subretinal fluid and restora- OCT provides qualitative and quantitative information about
tion of normal vision.45,46 However, in most eyes, VMT persists the retinal anatomy, which can identify factors contributing to
and vitrectomy may be an effective treatment option for vision loss in patients with ERM. On OCT, ERMs are seen as a
patients with symptomatic VMT.40,47,48 Consequently, OCT is highly reflective layer on the inner retinal surface (Fig. 3.11). In
useful in monitoring subtle changes in vitreoretinal adhesions most eyes, the membrane is globally adherent to the retina but,
and retinal architecture and assisting the treatment decision- in some cases, it can be separated from the inner aspect of the
making process. retina, which enhances its visibility by OCT. In this situation, it

A B C

D E F

Fig. 3.10 Vitreomacular traction syndrome: color fundus image of the left eye of a 71-year-old woman superimposed with the retinal thickness
map (A) showing an increase in the retinal thickness (red areas). The B-scan of the macular region shows an increase in the retinal thickness
and the presence of subretinal fluid and intraretinal cysts due to vitreomacular traction and an epiretinal membrane (B). A three-dimensional
spectral domain optical coherence tomography (courtesy of Cirrus, Carl Zeiss Meditec) is presented (C). The patient underwent surgery and,
2 months after pars plana vitrectomy, the retinal thickness decreased, with resolution of the intraretinal cysts (D–F).
89

Chapter 3
A B C

Fig. 3.11 Epiretinal membrane – color fundus image of the left eye of a 65-year-old man with grayish tissue over the retina (A). Cross-sectional

Optical Coherence Tomography


optical coherence tomography image showing a hyperreflective tissue overlying the retina, resulting in increased retinal thickness and cysts in
the retina (B and C).

is usually distinguishable from a detached posterior hyaloid.


Secondary effects of the membrane include loss of the normal
foveal contour, increased retinal thickness, and the presence of
cystoid changes, and these features may be observed in more
advanced membranes. OCT is useful for monitoring changes in
cases that are being observed and for documenting the response
to treatment in patients undergoing pars plana vitrectomy with A
membrane peeling.
Macular hole
Idiopathic macular holes typically occur in the sixth to seventh
decade of life with a 2 : 1 female preponderance. Symptoms
include decreased visual acuity, metamorphopsia, and central
scotoma. Bilateral involvement occurs in 15–20% of patients.21
A full-thickness defect in the neural retina as seen with OCT
B
can differentiate a true macular hole from a pseudohole seen
clinically. Pseudoholes are seen in the presence of a dense sheet
of ERM with a central defect that overlies the foveal center,
giving the ophthalmoscopic appearance of a true macular
hole.21,54
Gass described the stages of macular hole formation based on
biomicroscopic findings.55 A stage 1 impending hole is character-
ized by a foveal detachment seen as a yellow spot (1A) or ring
(1B) in the fovea (Fig. 3.12A). Spontaneous resolution will occur
in approximately 50% of these cases. In stages 2–4, there is a full- C
thickness retinal defect, with a complete absence of neural retinal
tissue overlying the foveal center. What differentiates these
Fig. 3.12 Macular hole. (A) Stage 1 macular hole in a 63-year-old
stages is the size of the retinal defect (<400 µm in stage 2 and woman with a 3-month history of decreased visual acuity (20/60).
>400 µm in stage 3) or the presence of a complete posterior vitre- An outer retinal defect can be observed in the B-scan (arrow). (B) A
ous detachment regardless of the hole size (stage 4) (Fig. 3.12B). full-thickness retinal defect developed after 2 months of follow-up with
worsening in the visual acuity (20/80). The posterior vitreous remains
OCT has enhanced our understanding of the pathogenesis of
adhered to the edge of the macular hole. (C) One month after surgery,
macular holes, the healing process after surgical repair, and the macular hole was closed and the visual acuity improved to 20/50,
helped in identifying pre- and postoperative features that are but a persistent foveal outer defect could be observed (arrowhead).
related to visual outcome. The anatomic changes identified on
OCT have been correlated with the various stages of macular
hole. In stage 1A, patients usually present with a localized foveo-
lar detachment, which can resolve spontaneously after the pos- OCT can demonstrate the complete hyaloid separation and occa-
terior vitreous detachment with resolution of the yellow foveal sionally a retinal operculum can be seen floating above the
spot, or it can progress to stage 1B with a development of a foveal center.
pseudocyst with loss of the outer retinal layers, and later develop Vitrectomy has become the standard treatment for macular
into a full-thickness macular hole.56,57 Generally, the retinal hole with anatomical success rates of 85–100%.58,59 OCT can be
defect is accompanied by a variable amount of intraretinal fluid used to confirm complete macular hole closure and restoration
appearing as cysts and a variable amount of subretinal fluid at of the normal foveal contour.60–63 In cases with suboptimal post-
the edge of the hole. The edge of the hole can appear elevated, operative visual outcomes, OCT can visualize persistent retinal
as a result of the significant intraretinal fluid accumulation or abnormalities despite anatomically successful macular hole
due to persistent vitreofoveal traction. In a stage 4 macular hole, surgery (Fig. 3.12C). Restoration of the ELM and the so-called
junction of the inner and outer segment of photoreceptors may vision, and are considered to be the precursors of GA and
reflect the morphologic and functional recovery of the photore- choroidal neovascularization (CNV).74,75
90 ceptors in surgically closed macular holes.62–65 A residual small
Early non-neovascular AMD: drusen and
defect in the ELM is often still evident in closed holes, particu-
pigmentary changes
larly in those that are spontaneously healed. The ability to
Drusen appear clinically as focal white-yellow excrescences
perform OCT imaging in eyes filled with gas or silicone oil
Section 1

deep to the retina. They vary in number, size, shape, and distri-
has also been useful as an adjunct to determine the extension of
bution. Several grading strategies have been developed to image
the face-down position in patients following vitrectomy for
drusen using color fundus imaging.76,77 Although color fundus
macular hole.66–68
imaging is useful for assessing the appearance of drusen, these
Age-related macular degeneration images only provide two-dimensional area information on the
geometry of the drusen and cannot be used to measure quantita-
AMD is a common cause of irreversible vision loss among the
Optical Imaging Technologies
Retinal Imaging and Diagnostics

tive properties such as drusen volume. Until the advent of high-


elderly worldwide. It is estimated that approximately 30% of
speed spectral domain technology, evaluation of drusen with
adults older than 75 years have some sign of AMD and that
OCT was often difficult as motion artifacts commonly resulted
approximately 10% of these patients have advanced stages of the
in apparent undulation of the RPE, mimicking the appearance
disease.69–72 AMD can be classified in two forms: non-neovascular
of drusen.78,79 SD-OCT can provide a three-dimensional, geomet-
(dry) and neovascular (wet or exudative). The non-neovascular
ric assessment of drusen.
form accounts for 80–90% of cases while the neovascular form
The high-definition B-scans obtained with SD-OCT are useful
accounts for 10–20% of cases, but was responsible for the major-
to assess the ultrastructure of drusen and to evaluate for evi-
ity of severe vision loss (80–90%) prior to the widespread use of
dence of disruption of adjacent retinal layers. Drusen are seen
vascular endothelial growth factor (VEGF) inhibitors.71,73
as discrete areas of RPE elevation with variable reflectivity,
Non-neovascular AMD (see Chapter 65, Dry AMD which is consistent with the variable composition of the underly-
– diagnosis and treatment) ing material (Fig. 3.13).80,81 In larger drusen or drusenoid retinal
Non-neovascular (dry) AMD is characterized by abnormalities of pigment epithelial detachments (PEDs), the RPE has a greater
the RPE, Bruch’s membrane, and choriocapillaris. These abnor- elevation with a dome-shaped configuration.82 Larger drusen
malities may be asymptomatic or accompanied by compromised may often become confluent and can sometimes be accompanied

S
3
T N
A B
I

C D E

Fig. 3.13 Early non-neovascular age-related macular degeneration. (A) Color fundus image of the right eye of a 61-year-old man with drusen and
pigmentary changes in the macula. (B) Foveal B-scan showing the drusen as elevations of the retinal pigment epithelium (RPE). The inner and outer
segment junction of the photoreceptors adjacent to the drusen appears disrupted (arrow). (C) Fundus autofluorescence illustrating that drusen cannot
be reliably identified by this imaging modality. (D) RPE segmentation map showing drusen in a unique three-dimensional perspective. (E) RPE
elevation map providing the drusen area (1.37 mm2) and volume (0.063 mm3).
91

Chapter 3
A B

Optical Coherence Tomography


C D E

Fig. 3.14 Drusenoid retinal pigment epithelium detachment (DPED). (A) Color fundus image of the right eye of a 66-year-old man with a DPED
and pigmentary changes in the macula. (B) Foveal B-scan showing the confluent drusenoid material as a large elevation of the retinal pigment
epithelium (RPE). Intraretinal pigment migration can be observed (arrow). (C) Fundus autofluorescence image (Heidelberg retina angiograph,
Heidelberg). (D) RPE segmentation map showing the DPED in a three-dimensional perspective. (E) RPE elevation map providing the DPED
area (3.87 mm2) and volume (0.508 mm3).

by fluid accumulation under the retina in the absence of CNV increase in volume and area. Drusen could also remain stable or
(Fig. 3.14).80 Recognition of this feature may avoid unnecessary they could dramatically decrease over time. When these drusen
treatment with anti-VEGF drugs. SD-OCT imaging has the reso- decreased, they could evolve into GA or neovascular AMD, or
lution to evaluate the retinal layers overlying drusen. A thinning they could decrease, resulting in no apparent residual anatomic
in the photoreceptor layer can be observed in up to 97% of cases, defect in the macula.
with average photoreceptor layer thickness reduced by 27% The RPE cells are capable of hypertrophy and proliferation in
compared to age-matched control eyes. The inner retinal layers response to different stimuli and in many cases an intraretinal
usually remain unchanged. These findings demonstrate a degen- pigment migration may occur (Fig. 3.14B). The Age-Related Eye
erative process with photoreceptor loss leading to visual Disease Study research group reported a severity scale defining
impairment.83 large drusen (≥125 µm) and pigment abnormality in the macula
The acquisition of dense raster scans comprised of a large as being a risk factor for disease progression in patients with
number of lower-density B-scans combined with the use of seg- intermediate AMD.84,85 This pigmentary abnormality can be
mentation algorithms results in the ability to generate maps of observed on OCT imaging as small discrete hyperreflective
the RPE, which provides information on RPE geometry and lesions within the neurosensory retina, usually within the outer
therefore a unique perspective of drusen. A novel algorithm nuclear layer.86
developed to identify RPE deformations such as drusen has been Typical drusen in AMD are seen as deposits between the RPE
shown to be highly reproducible in the measurement of drusen and the inner collagenous layer of Bruch’s membrane. OCT
area and volume.13 The algorithm creates a drusen map from a imaging is also useful for the assessment of a variety of condi-
scan pattern of 40 000 uniformly spaced A-scans organized as tions characterized by variant forms of drusen. These deposits
200 A-scans in each B-scan and 200 horizontal B-scans, covering can also be seen on top of the RPE and are known as “subretinal
an area of 6 × 6 mm centered in the fovea. The algorithm uses drusenoid deposits.”87,88 They appear on OCT imaging as granu-
the actual RPE geometry and compares this RPE map to a virtual lar hyperreflective material between the RPE and the IS/OS
map of the RPE free of any deformations (RPE floor). The algo- junction and are also well visualized on blue-light reflectance
rithm creates a difference map from these two maps, which imaging and autofluorescence imaging (Fig. 3.15).
permits reproducible measurements of drusen area and volume Another form of drusen, known as “cuticular drusen,” appears
(Fig. 3.6). This algorithm was used to study the natural history as numerous, uniform, round, yellow-white punctuate accumu-
of drusen in AMD.14 Drusen were shown to undergo three dif- lations under the RPE. Cuticular drusen are usually seen on OCT
ferent growth patterns. In most eyes, drusen were found to imaging as elevations of the RPE with occasional disruption of
Fig. 3.15 Subretinal drusenoid deposits. (A)
Color fundus image of a 76-year-old woman
shows multiple yellowish, small, and round
92 lesions. (B) Fundus autofluorescence clearly
shows these deposits as small and multiple
hyperautofluorescent spots. (C) On optical
coherence tomography, these lesions appear
Section 1

as multiple areas of granular hyperreflectivity


between the retinal pigment epithelium and
the inner segment–outer segment junction
(arrows).
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

the overlying IS/OS junction and ELM.81 Although cuticular and choriocapillaris loss in the initiation and propagation of this
drusen, subretinal drusenoid deposits, and soft drusen are com- condition.95 SD-OCT has been shown to be useful in detecting
posed of common components, they are distinguishable by mul- some of these morphologic alterations (Fig. 3.16D).
timodal imaging because of differences in location, morphology, With the use of SD-OCT enhanced depth imaging (EDI) pro-
and the optical properties of the drusenoid material and tocols, it is now possible to visualize the structure of the choroid
the RPE. in greater detail.29 EDI demonstrated that subfoveal choroidal
thickness decreases with age and axial length.31 In a subset of
Late non-neovascular AMD: geographic atrophy elderly patients complaining of unexplained vision loss, abnor-
The natural history of GA has been described as a progressive mal choroidal thinning was identified, and this condition was
condition that evolves through stages with loss of vision occur- named “age-related choroidal atrophy.”96 Future studies are nec-
ring over many years.89–91 Multiple imaging modalities have essary to confirm if this represents a new clinical entity or a
been used to document and quantify the area of GA. Until subtype of AMD. In contrast, the choroidal thickness appears to
recently, color fundus photography was used as the standard be unaffected in early non-neovascular AMD patients.97
method to image GA; however, the use of color photos can be SDOCT can also be used to quantify the areas of GA and
challenging due to the reported difficulty in detecting and accu- monitor the progression of the disease. GA is currently imaged
rately delineating GA.91,92 Other imaging modalities such as fluo- with SDOCT by using the OFI, which represents a virtual fundus
rescein angiography, fundus autofluorescence, and SD-OCT image resulting from the en face summation of the reflected light
imaging are now used to evaluate and quantify GA (Fig. 3.16). from each A-scan. This en face OCT fundus image identifies GA
Although these imaging modalities provide different informa- as a bright area due to the increased penetration of light into the
tion, none has been shown to be superior to the other. choroid where atrophy has occurred in the macula. The absence
GA is seen clinically as one or more well-demarcated areas of of the RPE and choriocapillaris is responsible for this increased
hypopigmentation or depigmentation due to the absence or penetration of light associated with GA. The OFI was shown to
severe attenuation of the underlying RPE. The larger, deeper cho- correlate well with the GA seen on clinical examination, color
roidal vessels are more readily visualized through the atrophic fundus imaging, and autofluorescence imaging (Fig. 3.16E).12,98,99
areas, and are accompanied by varying degrees of photoreceptor More recently, a newer algorithm provides an enhanced (partial)
and choriocapillaris loss. Associated retinal atrophy is seen as OFI, which is the summation of the reflected light from beneath
thinning or loss of the outer nuclear layer and the absence of ELM the RPE (Fig. 3.17). In addition, this new algorithm is able to
and IS/OS junctions.93,94 The loss of photoreceptors often extends quantify the area of GA automatically. The enhanced OFI has
beyond the margins of GA, with the ELM and IS/OS junctions advantages over the conventional OFI because the area of GA
disappearing while bridging across the GA margin.95 Evaluation appears brighter than in the conventional OFI due to a better
of these junctional zones may provide information about the contrast at the boundaries of the lesions and there is less interfer-
pathogenesis of GA, and the role of RPE, photoreceptor, ence from other macular pathologies such as ERMs.
93

Chapter 3
A B C

Optical Coherence Tomography


D E

Fig. 3.16 Geographic atrophy (GA). (A) Color fundus image of the left eye of a 74-year-old male with central GA secondary to age-related
macular degeneration. (B) Fundus autofluorescence (Spectralis, Heidelberg) showing a central area of hypoautofluorescence corresponding
to the GA seen on the color image. (C) Late-phase fluorescein angiography showing a central window defect corresponding to the GA.
(D) Horizontal B-scan through the foveal center demonstrating retinal thinning, loss of the retinal pigment epithelium (RPE), and photoreceptors.
The loss of photoreceptors (yellow arrows) often extends beyond the margins of the RPE loss (white arrows). Observe the increased light
penetration in the areas where the RPE is absent (bracket) and thin choroid (arrowhead). (E) Optical coherence tomography fundus image
(courtesy of Cirrus, Carl Zeiss Meditec), showing the GA as a bright area.

Fig. 3.17 Geographic atrophy (GA).


(A) Horizontal B-scan through the foveal
center of a 73-year-old man with GA showing
increased light penetration in the areas
where the retinal pigment epithelium (RPE) is
absent. The white arrow shows the junction
where the RPE is present and absent.
(B) Optical coherence tomography (OCT)
fundus image (OFI) represents a virtual
fundus image resulting from the en face
summation of the reflected light from each
A-scan. GA lesions are identified as a bright
area due to the increased penetration of light
into the choroid where atrophy has occurred.
A (C) Enhanced OCT fundus image (courtesy
of Cirrus, Carl Zeiss Meditec), which
represents the summation of the reflected
light from beneath the RPE (red lines and
arrows on A).

B C
Neovascular AMD (see Chapter 66, Wet AMD – diagnosis the subretinal space are associated with a larger volume of sub-
and treatment) retinal fluid compared with sub-RPE lesions.103
94 The neovascular (wet) form of AMD is characterized by the
Retinal pigment epithelium detachment
overproduction of VEGF and the growth of abnormal vessels in
In wet AMD, a retinal PED is formed by the separation of the
the macular region. These vessels may arise from the choroidal
RPE from Bruch’s membrane due to the presence of sub-RPE
circulation and penetrate Bruch’s membrane to form a fibrovas-
Section 1

fluid, blood, or fibrovascular tissue. A serous PED is defined as


cular tissue beneath or above the RPE, or these vessels may arise
an area of smooth, sharply demarcated dome-shaped elevation
primarily from the retinal circulation. In either case, the presence
of the RPE, often yellow-orange in color with a reddish halo of
of VEGF and abnormal vessels leads to structural changes in the
subretinal fluid. On fluorescein angiography, serous PEDs are
retina and choroid with the accumulation of fluid within the
associated with early hyperfluorescence with a well-defined
retina, in the subretinal space, or under the RPE. Furthermore,
border, which increases gradually throughout the study and
this neovascular invasion may lead to significant disorganiza-
Optical Imaging Technologies
Retinal Imaging and Diagnostics

classically demonstrates a pooling of dye rather than leakage.104,105


tion and remodeling of the retina, resulting in the loss of the RPE
Serous PEDs can be categorized as vascular or avascular.106 On
and photoreceptors with the formation of a disciform scar.100,101
OCT imaging, serous PEDs appear as a dome-shaped elevation
Intraretinal and subretinal fluid of the RPE typically seen overlying a homogeneously hypo­
In cases suspicious for exudative changes, OCT imaging can be reflective space, bound inferiorly by a visible Bruch’s membrane,
extremely useful in detecting intraretinal, subretinal, or sub-RPE which is seen as a thin hyperreflective line at the outer aspect of
fluid. In cases with active neovascular AMD, OCT imaging can the PED (Fig. 3.19).107,108 The appearance of vascularized serous
be used to establish baseline retinal thickness and volume, and PEDs is similar. However, in some cases, the apparent fibrovas-
determine the extent of neovascularization, fluid involvement, cular proliferation can be seen adjacent to the PED and even
and other lesion components (blood, fluid, pigment, and adherent to the outer surface of the RPE.
fibrosis). The fibrovascular PED usually produces an irregularly ele-
The growth of neovascularization is often accompanied by vated lesion visible on clinical examination and can be associated
VEGF-dependent leakage from both the mature vessels and the with RPE hyperpigmentation, subretinal hemorrhage, subretinal
growing immature vessels. Intraretinal edema can range from lipid exudation, and intra- or subretinal fluid collection.109 The
mild retinal thickening of the outer nuclear layer to large and elevation is often low and the borders are ill defined. The detailed
diffuse cystoid edema, seen as round or oval hyporeflective structural characteristics and precise mechanism of PED forma-
areas (Fig. 3.18).21 Lipid exudation can also be present in patients tion have not been completely resolved. Recent studies using
with profuse intraretinal edema and appear as small hyperre- SD-OCT imaging revealed that many of the fibrovascular PEDs
flective dots in the outer retina. The fluid may also accumulate appear to be filled with solid layers of material of medium reflec-
in the space between the RPE and the neurosensory retina. The tivity, separated by hyporeflective clefts (Fig. 3.20).110
subretinal fluid appears on OCT imaging as homogeneous hypo- Hemorrhagic PEDs occur when a CNV membrane bleeds into
reflective spaces when the fluid exudation is serous, or may be the sub-RPE space or as a result of an RPE tear. The hemorrhage
separated by fibrinous membranes when profuse proteinaceous can invade the subretinal space, with the sub-RPE blood having
exudation is present.102 Usually neovascular lesions growing in a typically darker appearance than subretinal blood. OCT

A B C

D E F

Fig. 3.18 Neovascular age-related macular degeneration. (A) Color fundus image of the right eye of an 81-year-old man with a 1-month history
of vision loss. Visual acuity was 20/100. (B) Horizontal B-scan through the foveal center showing retinal thickening and the presence of
intraretinal fluid with large cysts. (C) Retinal thickness map (courtesy of Cirrus, Carl Zeiss Meditec) showing the increase in retinal thickness
(red areas). After three intravitreal injections of antivascular endothelial growth factor, the intraretinal fluid was reabsorbed (D). This is better
observed in the B-scan and retinal thickness map (E, F).
Fig. 3.19 Vascularized serous retinal
pigmented epithelium detachment (PED).
(A) Color fundus image of the left eye of a
77-year-old woman with pigmentary changes 95
in the macula associated with an elevation of
the macula. (B) Horizontal B-scan through
the fovea showing a dome-shaped retinal

Chapter 3
pigment epithelium (RPE) elevation overlying
a homogeneous hyporeflective space.
Observe the presence of subretinal fluid
above the PED. (C) RPE segmentation map
A B showing a three-dimensional perspective of
the PED. (D) RPE elevation map showing
the area (5.84 mm2) and volume (0.83 mm3)
measurements of the PED.

Optical Coherence Tomography


C D

Fig. 3.20 Fibrovascular retinal pigmented


epithelium detachment (PED). Cross-
sectional B-scan of the right eye of an
87-year-old woman with a fibrovascular
PED. The space below the retinal pigment
epithelium is filled with solid layers of
medium reflectivity separated by
hyporeflective clefts. A small amount of
subretinal fluid can be identified over the
PED (arrow).

demonstrates a dome-shaped lesion, similar to serous PEDs, RPE tears may also be associated with central serous chorio-
although the slope of the elevation is more acute and the blood retinopathy (CSC), trauma, as well as other causes of CNV.115,116
under the RPE appears hyperreflective, attenuating the signal Although RPE tears can occur spontaneously in AMD patients,
from deeper structures, with the loss of choroidal detail (Fig. they have also been related temporally to various treatments
3.21).107,109,111 for AMD, such as verteporfin photodynamic therapy and intra-
In addition, the same algorithm used to measure drusen vitreal injection of anti-VEGF agents.117–121 Hemodynamic factors
can be used to measure PEDs, since both involve the deforma- play a role in the pathogenesis of the tear. The RPE layer is
tion of the RPE. This algorithm is able to measure both the put on stretch as a result of accumulating sub-RPE fluid and
area and volume of PEDs (Fig. 3.19D). In addition, algorithms this stress leads to a tear in the RPE. A sheet of RPE cells
may be developed to characterize the internal architecture of then contracts and scrolls up upon itself in a radial fashion,
the PEDs automatically.112 The qualitative appearance of the leaving an area of retina without underlying RPE.114,122 Sub-
B-scans and the qualitative and quantitative changes in the retinal and sub-RPE hemorrhages frequently accompany an
retinal thickness maps and RPE elevation map can be used RPE tear, which appears ophthalmoscopically as an area of
to appreciate better the natural history of the disease and to well-demarcated hyperpigmentation immediately adjacent to
monitor the effect of anti-VEGF therapy in patients with PEDs an area of relative hypopigmentation.
associated with wet AMD. On OCT imaging, an area of discontinuity in a large PED is
often seen, with the free edge of the RPE often curled under the
Tear of the retinal pigment epithelium PED. Adjacent to the tear, there is increased reflectivity from the
RPE tears are most commonly seen in association with CNV choroid vessels, due to the absence of the RPE. The overlying
secondary to AMD, especially when a PED is present.113,114 retina is typically intact, but may be separated from the area of
atrophy by subretinal fluid.114 The tear tends to occur at the base lesion becomes less active, and the fibrous components typically
of the PED, near or at the intersection of attached and detached increase, resulting in disciform scar formation. Clinically the scar
96 retina (Fig. 3.22).113 During anti-VEGF therapy, the height of the appears as smooth, elevated white or gray tissue in the subreti-
PED and the irregular surface contour may help in predicting nal space and on OCT imaging the scar corresponds to a highly
the risk for RPE tear, which may also occur without treatment reflective outer retinal or subretinal lesion (Fig. 3.23).21 Scar for-
as part of normal disease progression.123,124 The visual outcome mation may be associated with loss of the overlying photorecep-
Section 1

in patients with RPE tears is generally poor when the fovea is tor layer and irreversible reduction in visual acuity. This may be
involved. observed on OCT imaging as a disruption of the IS/OS junction
and ELM.125,126 In this stage of the disease, the OCT is very helpful
Disciform scarring in identifying the presence of subretinal fluid or intraretinal
Disciform scarring and subretinal fibrosis mark the endstage of cysts that are associated with the neovascular activity of the
CNV. The vascular components of CNV typically regress as the lesion, and may help in making the retreatment decision.
Optical Imaging Technologies
Retinal Imaging and Diagnostics

S
1
T N
A B
I
Fig. 3.21 Hemorrhagic retinal pigmented epithelium detachment (PED). (A) Color fundus image of the right eye of a 65-year-old woman with
a large subretinal pigment epithelium (RPE) hemorrhage secondary to neovascular age-related macular degeneration. (B) Optical coherence
tomography demonstrates a dome-shaped lesion, similar to serous PEDs. The blood under the RPE appears hyperreflective, attenuating the
signal from deeper structures. Subretinal fluid can be observed as hyporeflective spaces above the RPE (arrows).

Fig. 3.22 Retinal pigment epithelium (RPE)


tear. (A) Color image of the right eye of an
81-year-old man with an area of relative
hypopigmentation that corresponds
to the RPE tear. (B) Heidelberg
fundus autofluorescence showing
hypoautofluorescence in the area where the
RPE is absent. (C) On the B-scan there is
an area of discontinuity of the RPE near the
base of the pigmented epithelium detachment
with the free edge of the RPE curled under
the pigmented epithelium detachment
(arrow).

A B

C
Retinal angiomatous proliferation studies with SD-OCT imaging concluded that the initial neovas-
The term “retinal angiomatous proliferation” was introduced by cular process could originate from either the retinal or choroidal
Yannuzzi and coworkers to describe a form of neovascularization circulation; however, histopathological studies suggest that all 97
in AMD patients, which arises from within the retina with pos- the neovascularization is within the retina.129,130 On OCT imaging,
sible formation of a retinochoroidal anastomosis as the disease the most common feature is the presence of a serous PED with

Chapter 3
progresses.127 Whether the development of the retinochoroidal CME overlying the PED (Fig. 3.24).129,131,132 An intraretinal hyper-
anastomosis is a result of a primary intraretinal neovasculariza- reflective angiomatous complex consistent with the intraretinal
tion or a sub-RPE lesion remains controversial.127,128 Recently, neovascularization and subretinal fluid may also be seen.127

Optical Coherence Tomography


A B

Fig. 3.23 Disciform scar. (A) Color fundus image of the left eye of an 80-year-old woman with a white-grayish tissue involving the macula.
(B) Horizontal B-scan with a large hyperreflective lesion under the retina (arrow).

A B C

D E

Fig. 3.24 Retinal angiomatous proliferation. (A) Color fundus image of the right eye of a 90-year-old woman with a history of blurred vision and
metamorphopsia for 2 weeks. Visual acuity was 20/40. Fundus examination revealed multiple drusen, pigmentary changes, and hemorrhage
inferior to the fovea with a subtle elevation of the retina. (B) Fundus autofluorescence demonstrates hypoautofluorescence in the area
corresponding to the hemorrhage. (C) Fluorescein angiography demonstrates a focal area of leakage inferior to the fovea. (D) Late-phase
indocyanine green angiography reveals a hot spot. (E) B-scan through the lesion reveals a retinal pigment epithelium detachment (arrow) with
cystoid macular edema overlying the pigmented epithelium detachment.
Polypoidal choroidal vasculopathy of “outer retinal tubulation” since the latter represents a rear-
Polypoidal choroidal vasculopathy is considered a variant form rangement of photoreceptors in response to injury and RPE loss
98 of CNV characterized by the presence of multiple vascular and is usually present in patients with chronic and advanced
sacular dilations (polyps) in the choroidal circulation that mani- neovascular AMD (Fig. 3.26). Importantly, this tubulation does
fests clinically with variably sized serous and serosanguineous not respond to anti-VEGF therapy.145 In patients with PEDs, the
area and volume of the lesion can be assessed and used to
Section 1

detachments of the neurosensory retina and RPE, usually around


the optic nerve or in the central macula.133 Indocyanine green monitor the effect of anti-VEGF therapy in patients with wet
(ICG) angiography is particularly useful in imaging the polyp- AMD associated with PEDs, and an increase in the area and
oidal abnormalities seen in polypoidal choroidal vasculopathy, volume of PEDs could be used to indicate when retreatment is
with a branching vascular network of vessels ending in polyp- necessary.
like structures.134 SD-OCT images can demonstrate the polypoi-
Optical Imaging Technologies

dal structure beneath the RPE, which remains adherent to the


Retinal Imaging and Diagnostics

Central serous chorioretinopathy


RPE, even with increased exudation. It is especially useful to
CSC is an idiopathic syndrome that typically affects young to
detect the abnormalities surrounding the polypoidal lesions
middle-aged males and is characterized by serous detachment
such as intraretinal, subretinal, and sub-RPE fluid.135,136
of the neurosensory retina. Focal and multifocal areas of leakage
Choroidal neovascularization: response to treatment secondary to increased permeability of the choroidal vessels and
The combination of clinical examination, fluorescein angiogra- a barrier defect at the level of the RPE have been described in
phy, OCT images, and, less frequently, ICG angiography is the pathogenesis of this disorder.146–148
usually required to diagnose neovascular AMD and exclude Presenting symptoms include central vision loss, a decrease in
other macular conditions that can mimic the features of neo- vision that can be corrected with an increased hyperopic correc-
vascular AMD.137 With the use of anti-VEGF drugs the ideal tion, metamorphopsia, central scotoma, and decreased color
strategy for following eyes with wet AMD has evolved from saturation. The symptoms are usually self-limited but can recur
monthly injections to OCT imaging to determine whether the in the same or the opposite eye. In most cases, CSC resolves
treatment is effective in resolving the macular fluid.138,139 Many spontaneously within 6 months, with a good visual prognosis.
alternative treatment regimens have used OCT-guided strate- However, prolonged and recurrent macular detachment in some
gies, with good visual and anatomical results with fewer intra- cases may cause degenerative changes in the subfoveal RPE and
vitreal injections compared with monthly dosing.140–144 The neurosensory retina with poor visual outcome.149,150
macular fluid can be identified by examining the B-scans and The primary pathology of acute CSC is thought to begin
reviewing the retinal thickness maps, which calculate the retinal with disruption of the choroidal circulation. The RPE then
thickness between the ILM and the RPE segmentation maps. decompensates and exudation from the choroidal vasculature
The effect of anti-VEGF therapy can then be assessed based on passes into the subretinal space. These hypotheses were based
the qualitative appearance of the B-scans and the qualitative, on fluorescein angiography and ICG angiography find-
as well as quantitative, changes in the retinal thickness maps ings.147,151–154 The development of OCT imaging has provided a
(Figs 3.18 and 3.25). The presence or recurrence of intraretinal better understanding of CSC, especially the abnormalities in the
or subretinal fluid has to be differentiated from the appearance RPE layer.155–159

A B C

D E F

Fig. 3.25 Neovascular age-related macular degeneration (response to treatment). Color fundus image, horizontal B-scan, and retinal thickness
map of the right eye of a 65-year-old man with wet age-related macular degeneration before (A–C) and after (D–F) a single treatment with
intravitreal anti-vascular endothelial growth factor. Observe the improvement of the intraretinal fluid and cysts in the B-scans (B, E) and the
decrease in retinal thickness (C, F).
Fig. 3.26 Outer retinal tubulation: a 67-year-
old woman with wet age-related macular
degeneration who has received 13 intravitreal
injections over the last 2 years. The foveal 99
horizontal B-scans before (A) and after the
last (B) treatment are presented. The larger
intraretinal cyst present in the image before

Chapter 3
treatment (arrowhead) disappeared after
treatment. The small cyst (arrow) showed
no response to the intravitreal injection
of anti-vascular endothelial growth factor.
This smaller cyst corresponds to an outer
retinal tubulation which is frequently present
in patients with chronic and advanced
neovascular age-related macular

Optical Coherence Tomography


A degeneration.

B
S

There are two forms of the disease, acute and chronic. Acute changes.156,157,162–166 OCT can also visualize the subretinal yellow
CSC (Fig. 3.27) is classically unilateral and characterized by one deposits as highly reflective material. Precipitates are not only
or more focal leaks at the level of the RPE on fluorescein angi- on the posterior surface of the detached retina but also in the
ography. The chronic form (Fig. 3.28) is believed to be due to detached neurosensory retina. Photoreceptor segment morpho-
diffuse RPE disease and is usually bilateral. It presents with logic changes along the detached retina show elongation of the
diffuse RPE atrophic changes, varying degree of subretinal fluid, photoreceptor outer segments and decreased thickness of the
RPE alterations, and RPE tracks. It is characterized by diffuse outer nuclear layer.167 Accumulation of abnormal outer segments
RPE leakage on fluorescein angiography. in the neurosensory retina is related to clinical manifestation on
OCT imaging is helpful in diagnosing and managing patients OCT as a granulated shaggy profile of the outer surface of the
with CSC. OCT imaging can noninvasively identify the pres- detached retina.168
ence and extent of subretinal fluid and PEDs. OCT imaging En face OCT imaging has been found to detect alterations
is also useful for assessing the resolution of subretinal fluid of the RPE in the form of a PED or a small defect in the RPE.
and the morphological retinal changes during normal disease Most alterations of RPE are associated with choroidal abnor-
progression. OCT is more sensitive than clinical exam and malities.159 OCT imaging has been found to detect morphologic
fluorescein angiography in identifying small amounts of sub- changes at the point of dye leakage in eyes with CSC. Trans-
retinal fluid.160 OCT is useful in predicting the recovery of verse images (C-scans) have shown serous retinal detachments
visual acuity and explaining poor visual outcomes even after and irregular lesions of the RPE. These findings, along with
the resolution of the fluid. With SD-OCT imaging, topographic other findings on B-scans and segmentation maps, are consistent
changes in CSC can be visualized with two- and three- with location of lesions in areas of fluorescein angiographic
dimensional reconstructions. SD-OCT also offers the ability of leakage.169,170
exact localization of the pathology and accurate volumetric Visual prognosis in patients with CSC can be linked to retinal
measurements.161 morphological changes by OCT.171,172 Mastsumoto et al. corre-
OCT features of acute CSC include thickening of the neuro- lated the visual outcome with the preservation of outer nuclear
sensory retina within the area of retinal detachment, PED, the layer thickness and continuity of photoreceptor IS/OS in
presence of fibrinous exudates in the subretinal space, and the resolved CSC. The outer nuclear layer thickness was positively
shaggy outer segments of the neurosensory retina above correlated with visual acuity. Discontinuity of the IS/OS line
the leakage site. OCT features of the chronic form include was prevalent in eyes with thinner outer nuclear layer and lower
foveal atrophy, retinal thinning, and cystoid degenerative visual acuity.172 Ojima et al.171 reported that microstructural
100
Section 1

A B C D
Optical Imaging Technologies
Retinal Imaging and Diagnostics

E F G H

Fig. 3.27 Acute central serous chorioretinopathy. (A) Color photo shows a well-defined, circular area of retinal elevation. (B) Fluorescein
angiography shows an area of hyperfluorescence with “smokestack” leakage. (C) Retinal thickness map shows elevation of the retina.
(D) Spectral domain optical coherence tomography (OCT), horizontal, acquired through the fovea, shows serous detachment of the neurosensory
retina above an optically clear, fluid-filled cavity, associated with a pigment epithelial detachment. The retinal pigment epithelium detachment
corresponds to the area of hyperfluorescence seen on the angiogram. (E–H) Follow-up visit 1 month later. (E) Color photo shows resolution
of the retinal elevation in the area of the fovea but illustrates a well-defined, circular area of retinal elevation inferior to the fovea (small arrow).
(F) Fundus autofluorescence shows a well-defined, circular area of retinal elevation inferior to the fovea involving the inferior arcade (small
arrow). (G) Retinal thickness map shows decrease in the thickness of the retina in the fovea. (H) Spectral domain OCT, horizontal B-scan
acquired through the fovea, shows decrease in the amount of subretinal fluid.

A B C D

Fig. 3.28 Central serous chorioretinopathy. (A) Color photo shows a well-defined, circular area of retinal elevation: white line represents the
location of the B-scan. (B) Fundus autofluorescence shows an area of hyperfluorescence. (C, D) Fluorescein angiography shows an inkblot
appearance that leaks later. (E) Spectral domain optical coherence tomography shows serous detachment of the neurosensory retina associated
with an irregular, granulated retinal pigment epithelial layer and sagging/dipping of the posterior layer of the neurosensory retina (asterisk).

changes occur in the photoreceptor layer of the detached retina


and the visualization of the ELM and the photoreceptor layer Enhanced depth imaging OCT IN CSC
correlates with visual function. Foveal thickness can be a predic- Conventional SD-OCT has a limited ability to image the choroid
tor of visual outcome in patients with CSC.163 Both foveal thick- because of scattering by the pigment granules within the RPE
ness and visual acuity have been observed to be proportional to and by the pigment and blood within the choroid, and because
the duration of symptoms, Foveal attenuation, and atrophy, of a depth-dependent roll-off in sensitivity of SD-OCT instru-
which may be a consequence of prolonged absence of contact ments in general.29 A method to improve imaging of the
between photoreceptor and RPE cells.160 choroid, known as EDI OCT, showed that eyes with CSC
101

Chapter 3
A B D E

Optical Coherence Tomography


C F

Fig. 3.29 Central serous chorioretinopathy. (A) Color photo of the right eye shows area of retinal elevation with pigmentary changes; white line
represents the position of B-scan. (B) Fundus autofluorescence shows an area of hyperfluorescence and hypofluorescence. (C) Spectral domain
optical coherence tomography (OCT), enhanced depth imaging, shows serous detachment of the neurosensory retina along with pigmented
epithelium detachment, retinal pigment epithelial alterations, granulated posterior detached retina, and thick choroid (arrowheads represent the
outer boundary of the choroid). (D) Color photo of the left eye shows pigmentary changes without retinal elevation; white line represents location
of B-scan. (B) Fundus autofluorescence shows an area of hyperfluorescence and hypofluorescence. (C) Spectral domain OCT, enhanced depth
imaging, demonstrates a thick choroid (arrowheads represent the boundary of the choroid) without serous detachment of the neurosensory retina.

had a much thicker choroid compared with normal eyes (Fig. Diabetic retinopathy
3.29).173 Fellow eyes of patients with CSC were also found
Diabetic retinopathy is the leading cause of blindness in indi-
to have thicker choroids compared with age-matched normal
viduals under 65 years of age in the USA, with diabetic macular
eyes.174 Maruko et  al. reported a thickened choroid in CSC
edema (DME) being the principal cause of vision loss in these
and the association with choroidal vascular hyperpermeability
patients.180,181 Diabetic retinopathy can be classified into nonpro-
on ICG angiography.175
liferative diabetic retinopathy (NPDR) and proliferative diabetic
Verteporfin photodynamic therapy is one of the therapies
retinopathy (PDR).
used to treat leakage and subretinal fluid in eyes with CSC.
Maruko et al.175 reported that eyes treated with focal laser Nonproliferative diabetic retinopathy and diabetic
showed no alteration in choroidal thickness even though there macular edema
was fluid reabsorption, but eyes treated with verteporfin photo- The important role of OCT in DME management involves the
dynamic therapy showed a decrease in choroidal thickness by evaluation of retinal pathology, including retinal thickness,
SD-OCT imaging and a decrease in choroidal hyperpermeability CME, intraretinal exudates, vitreomacular interface abnormali-
seen during ICG angiography. The changes occurring in ties, subretinal fluid, and photoreceptor IS/OS junction abnor-
the choroid after photodynamic therapy may reflect a more malities. OCT is also important in monitoring the response to
normalized choroidal permeability. treatment of DME by laser, intravitreal pharmacotherapies, and
vitreoretinal surgery.
Cystoid macular edema Determination of macular edema can be difficult with biomi-
CME is an important cause of reduced visual acuity in a wide croscopy or color fundus imaging, especially when the edema is
variety of retinal diseases such as diabetic retinopathy, retinal mild.182–184 It has been suggested that OCT measurements may
vein occlusion, CNV, retinal dystrophies, uveitis, and following be a more sensitive and reproducible indicator of true change in
intraocular surgery. Regardless of the underlying etiology, CME retinal thickness than color fundus imaging, supporting the use
appears as retinal thickening with intraretinal cavities of reduced of OCT as the principal method for documenting retinal thick-
reflectivity on OCT (Fig. 3.30). ness. However, OCT is less suitable than fundus imaging for
Clinically significant pseudophakic CME is estimated to occur documenting the location and severity of other morphologic
in 1–2% of patients undergoing cataract extraction.176,177 Inflam- features of diabetic retinopathy, such as hard exudates, retinal
matory components induced by surgery along with mechanical hemorrhages, microaneurysms, and vascular abnormalities. Fur-
forces induced by a modified vitreous are responsible for the thermore, OCT cannot provide information on overall retinopa-
macular changes in these patients.178,179 The diagnosis based only thy severity, for which color photographs remain the gold
on fundus examination can be challenging and usually fluores- standard.185–188
cein angiographic imaging, which shows a classic petaloid OCT can be used to distinguish patients with normal retinal
pattern of leakage, or OCT imaging is needed for confirmation. contour and thickness despite extensive angiopathy from those
OCT has the advantage of being a faster and noninvasive with early retinal edema. In general, the DME can be classified
imaging technique which can also provide quantitative assess- into several categories: diffuse retinal thickening, CME, serous
ment of the macular thickness that can be used to monitor the retinal detachment or subretinal fluid, and vitreomacular inter-
clinical course and to make therapeutic decisions. face abnormality.189–191 Diffuse retinal thickening is usually
102
Section 1

A B C
Optical Imaging Technologies
Retinal Imaging and Diagnostics

D E
ILM - RPE

F G
ILM - RPE

Fig. 3.30 Cystoid macular edema. Left eye of a 64-year-old man 30 days after phacoemulsification. The visual acuity was 20/50. (A) Color
fundus image with some cystic changes: white line represents where the B-scan was acquired. (B and C) Fluorescein angiography showing the
classic petaloid leakage pattern. (D) B-scan showing the intraretinal cysts as hyporeflective spaces within the retina. (E) Retinal thickness map
showing increased retinal thickness due to the presence of cysts. (F and G) Same patient after 45 days of treatment with topical nonsteroid
anti-inflammatory medication. The retinal thickness decreased and the intraretinal cysts disappeared. ILM, internal limiting membrane;
RPE, retinal pigment epithelium.

defined as a sponge-like swelling of the retina with a general- fluid. Vitreomacular interface abnormalities include the pres-
ized, heterogeneous, mild hyporeflectivity compared with ence of ERMs, VMT, or both. Intraretinal focal hyperreflections
normal retina. CME is characterized by the presence of intrareti- that correspond clinically to retinal exudates are a frequent
nal cystoid areas of low reflectivity, which are typically sepa- finding in all the patterns described above.
rated by highly reflective septa (Fig. 3.31). Serous retinal OCT has become widely accepted in monitoring progression
detachment is defined on OCT as a focal elevation of neurosen- and treatment response in patients with DME. Prior to OCT
sory retina overlying a hyporeflective, dome-shaped space. The imaging, precision in central retinal thickness monitoring was
posterior border of the detached retina is usually highly reflec- not possible. The ETDRS provided guidelines for laser manage-
tive, which helps to differentiate subretinal from intraretinal ment of patients with DME.192–194 Although OCT was not
103

Chapter 3
A B

Optical Coherence Tomography


C D

Fig. 3.31 Diabetic macular edema. Right eye of a 43-year-old woman with type 2 diabetes and moderate nonproliferative diabetic retinopathy.
(A) B-scan and (B) retinal thickness map showing diffuse macular edema and the presence of intraretinal cysts with an increased retinal
thickness. (C) B-scan and (D) retinal thickness map of the same patient after 3 months of intensive blood sugar control and focal laser therapy.
The intraretinal cysts disappeared and the retinal thickness map shows an important decrease in the retinal thickness.

available for use in this study, quantitative retinal thickness Proliferative diabetic retinopathy
maps can be used to direct laser therapy and may be better than PDR can be visualized with OCT imaging as highly reflective
using biomicroscopy alone. In the era of pharmacotherapy, preretinal bands anterior to the retinal surface consistent with
many agents like triamcinolone and anti-VEGF agents (ranibi- preretinal fibrovascular or fibroglial proliferation. Diffuse retinal
zumab and bevacizumab) have been studied to treat DME. In thickening, distortion, and irregularity of the retinal contour can
these studies, OCT played an important role in determining the also occur as a result of the contraction of these preretinal mem-
retinal thickness and the treatment response.195,196 The treatment branes. An associated traction retinal detachment may be
response of each OCT pattern of DME has been shown to be observed as well. OCT imaging is valuable in determining the
different.197 Patients with diffuse retinal thickening may achieve extent of the tractional component as well as the presence of
a greater reduction in retinal thickness and a greater improve- foveal involvement, assisting in the decision to intervene surgi-
ment in visual acuity compared with patients exhibiting CME, cally (Fig. 3.32).21 The decision for surgery typically hinges on
subretinal fluid, or vitreomacular interface abnormality.197,198 the progressive nature of the traction and the degree to which
Macular traction has become increasingly recognized in the macula is affected by the traction.
patients with DME, especially in eyes with persistent edema
after focal laser or pharmacological treatment. These patients Retinal vein occlusion
often show the clinical appearance of a thick posterior hyaloid Retinal vein occlusions have been defined as retinal vascular
with diffuse fluorescein leakage. Recognition of this condition disorders characterized by engorgement and dilatation of the
can be difficult using the clinical exam alone. This is readily retinal veins with secondary, mostly intraretinal, hemorrhages
recognized on OCT imaging as diffuse cystoid retinal thicken- and mostly intraretinal (and partially subretinal) fluid, retinal
ing, a flat-appearing foveal contour, and a thickened hyperre- ischemia, including cotton-wool spots, and retinal exudates.205
flective linear vitreoretinal interface. Focal vitreoretinal adhesions Retinal vein occlusions are commonly divided into central retinal
that cannot be identified on clinical exam are also often evident vein occlusion and branch retinal vein occlusion, and as soon as
on OCT.199,200 These findings can direct the decision as to whether the foveal region is involved with macular edema, central visual
to proceed with pars plana vitrectomy and membrane peeling.201 acuity may be affected.
Furthermore, the improvement in axial resolution with In retinal vein occlusions, OCT can display intraretinal cysts
SD-OCT has enhanced the ability to evaluate foveal microstruc- responsible for the increase in retinal thickness often associated
tural abnormalities, including the photoreceptor IS/OS junction, with serous detachment of the neurosensory retina. Retinal cysts
which may reveal damage to macular photoreceptors. Several can be numerous and confluent, forming large central cystoid
studies have demonstrated that an intact IS/OS junction is pre- spaces. Associated findings can be observed, such as vitreous
dictive of a better visual acuity in patients after treatment macular adherence, ERM, and hyperreflectivity of the posterior
for DME.202–204 layer corresponding to atrophy or fibrosis of the RPE, subretinal
104
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 3.32 Diabetic retinal tractional detachment. (A) Color fundus image of the right eye of a 72-year-old woman with proliferative diabetic
retinopathy. (B) Foveal B-scan of the same patient showing a thick posterior hyaloid distorting the retinal architecture with traction and
accumulation of fluid under the retina.

A B

C D

Fig. 3.33 Central retinal vein occlusion. (A) Color photo of the right eye shows optic nerve head edema, dilated tortuous retinal veins, scattered
intraretinal hemorrhages in all quadrants, and macular edema: white line represents the location of the B-scan. (B) Spectral domain optical
coherence tomography (OCT) obtained through the fovea illustrates loss of normal foveal contour and marked and diffuse retinal thickening.
Large areas of low intraretinal reflectivity consistent with cystic fluid accumulation and edema were seen. A detachment of the neurosensory
retina with subretinal fluid was observed below the fovea. (C) Color photo of the right eye 1 month after bevacizumab injection shows dilated
tortuous retinal veins and scattered intraretinal hemorrhages in all quadrants: white line represents area of B-scan. (D) Spectral domain OCT,
1 month after bevacizumab injection obtained through the fovea, shows that macular edema almost completely disappeared with a small amount
of residual subretinal fluid. Improvement in the normal foveal contour and decrease in the retinal thickening and edema. Areas of high intraretinal
reflectivity consistent with the hemorrhages.

accumulation of material, subretinal fibrosis, lamellar macular proportion of eyes retain poor visual acuity despite treatment.
hole formation, intraretinal lipid exudates, and intraretinal hem- Several studies have shown that low visual acuity has been
orrhage (Fig. 3.33). associated with a poor functional outcome after treatment or
Ota et al. reported that, in branch retinal vein occlusion, visual during the natural course (Fig. 3.34). SD-OCT can help predict
function and recovery of vision are correlated with thickness of visual acuity based on the integrity of the neurosensory retina.
the central macula, and that is correlated with the integrity of
the inner and outer segments of the photoreceptors in the Central retinal artery occlusion
fovea.206 SD-OCT imaging helps to quantify the amount of CME. Central retinal artery occlusion shows a distinct pattern on
The accumulation of fluid can be located mostly within the OCT images. In the acute phase, OCT images demonstrate the
retinal layers or additionally in the subretinal space.207 Anti- increased reflectivity and thickness of the inner retina and a
VEGF therapy is increasingly used to treat macular edema in corresponding decrease of reflectivity in the outer layer of the
patients with retinal vein occlusions. Nevertheless, a significant retina and RPE/choriocapillaris layer. Follow-up OCT images
105
B C D

Chapter 3
A E F G

Optical Coherence Tomography


Fig. 3.34 Branch retinal vein occlusion. (A) Color photo of the right eye shows dilated tortuous retinal veins, flame-shaped hemorrhages in an
arcuate configuration in the distribution of inferotemporal branch retinal vein occlusion, and macular edema. (B) Retinal thickness map showing
increase in retinal thickness. (C, D) Spectral domain optical coherence tomography (OCT) horizontal and vertical scan respectively obtained
through the fovea revealed that marked retinal thickening, areas of low intraretinal reflectivity consistent with cystic fluid accumulation, and
edema were identified, especially in the outer plexiform layer. High reflectivity is noted in the inner layers from intraretinal hemorrhage.
(E) Retinal thickness map 1 month after bevacizumab injection, showing decrease in retinal thickness. (F, G) Spectral domain OCT horizontal
and vertical scan respectively, 1 month after bevacizumab injection, obtained through the fovea showed complete resolution of macular edema,
improvement in foveal contour, and decrease in retinal thickening.

A B

Fig. 3.35 Central retinal artery occlusion. (A) Color photo of the left eye shows cherry-red spot appearance, retinal opacity of posterior fundus,
most marked in the parafoveal region, and a small area of normal retina temporal to the optic disc corresponding to the patent cilioretinal retinal
artery. (B) Spectral domain optical coherence tomography horizontal scan through the fovea illustrates increased thickness and hyperreflectivity
of the inner retinal layers, denoting the presence of intracellular edema, with decreased reflectivity of photoreceptor and retinal pigment epithelial
layers because of the shadowing effect.

demonstrate a decrease in the reflectivity and thickness of the beneath the fovea shows a relative increase in reflectivity com-
inner retinal layers and a corresponding increase of reflectivity pared with the other regions of the RPE/choriocapillaris. An
in the outer retina and RPE/choriocapillaris layer compared additional finding on OCT imaging is the thinning and atrophy
with the baseline OCT image, suggesting a generalized atrophy in the affected area of the retina, which occurs after a period
of the neurosensory retina as a late finding. Therefore, the use of time (Fig. 3.35).208
of OCT may help facilitate prompt recognition of acute and
chronic central retinal artery occlusion. In patients with central Branch retinal artery occlusion
retinal artery occlusion, OCT images closely correspond with Branch retinal artery occlusions are usually embolic in nature.
known histopathologic changes. Histology following acute The embolic source is either a carotid artery atheroma or myo-
central retinal artery occlusion shows retinal changes limited to cardial thrombus. The embolus usually lodges at the bifurcation
the nerve fiber and ganglion cell layers. There are profound of the central retinal artery into the branch retinal artery. Histo-
losses of ganglion cells and diffuse edema of the inner retinal pathologically, acute branch retinal artery occlusions reveal isch-
layers with little change seen in the deeper retinal layers sup- emia in the corresponding retinal quadrant marked by inner
plied by choroidal vessels. OCT images provide an in vivo view retinal edema at the initial stage followed by atrophy in long-
of the retinal structure following central retinal artery occlusion. standing cases. SD-OCT imaging shows the edematous inner
Increased reflectivity of the inner retina, presumably because retina, comprising the inner nuclear layer, inner plexiform layer,
of opacification of the ganglion cell and nerve fiber layers, cor- and ganglion cell layer, as a hyperintense band with increased
responds to previously described histologic findings of “cloudy thickness, which is contrasted by the normal reflectivity and
swelling” of these layers. Attenuation of reflectivity in the outer thickness of the corresponding layers of the unaffected macular
layer of the retina and the RPE/choriocapillaris layer is due to regions. Prolonged ischemia results in consecutive atrophy of
the ganglion cell and nerve fiber changes allowing less light these layers with each layer exhibiting differential sensitivity to
reflected back from the outer portions of the retina. Further the underlying hypoxia. Animal experiments have revealed
evidence of this phenomenon is at the foveal depression where retinal ganglion cells to be relatively resistant to the ischemia
the ganglion cell layer is absent. As more light is allowed compared to the other retinal neurons.209 Similar findings in vivo
through the fovea, the RPE/choriocapillaris layer directly using SD-OCT imaging revealed the relative preservation of
106
Section 1

A B C D

Fig. 3.36 Branch retinal artery occlusion. (A) Color photo of the right eye shows area of whitening in the distribution of an inferotemporal
retinal arteriole: white vertical line represents location of B-scan; square dotted line represents area of embolus in arteriole which is magnified.
(B) Embolus was appreciated in the inferior retinal arteriole next to the optic nerve. (C) Spectral domain optical coherence tomography (OCT)
vertical scan through the fovea illustrates increased thickness and hyperreflectivity of the inner retinal layers in the inferior perifoveolar area,
denoting the presence of intracellular edema, with decreased reflectivity of photoreceptor and retinal pigment epithelial layers. The asymmetry
Optical Imaging Technologies
Retinal Imaging and Diagnostics

of optical reflectivity in perifoveal region is an important finding; OCT findings in the superior perifoveolar area are normal. (D) Retinal thickness
map shows increased thickness in the inferior perifoveal area.

ganglion cell layer as opposed to the thinning of the inner plexi- Disclosures
form and nuclear layers (Fig. 3.36).210 Drs Garcia Filho, Rosenfeld, and Yehoshua received research
support from Carl Zeiss Meditec. Dr Gregori and the University
FUTURE DIRECTIONS of Miami co-own a patent that is licensed to Carl Zeiss Meditec.
The recent advances in OCT technology have clearly revolution- Dr Rosenfeld has received honoraria for lectures from Carl Zeiss
ized the assessment of patients with retinal disorders. Although Meditec.
SD-OCT has changed the way we image macular diseases, the
future of OCT holds even more promise with the use of longer- REFERENCES
1. Gabriele ML, Wollstein G, Ishikawa H, et al. Optical coherence tomography:
wavelength light sources, faster scan times, functional assess- history, current status, and laboratory work. Invest Ophthalmol Vis Sci
ments, and higher image resolution. 2011;52:2425–36.
2. Drexler W, Fujimoto JG. State-of-the-art retinal optical coherence tomography.
Current commercial available SD-OCT instruments allow Prog Retin Eye Res 2008;27:45–88.
dense scanning of the macula with high axial resolution (approx- 3. Li Y, Gregori G, Knighton RW, et al. Registration of OCT fundus images with
imately 5–8 µm). Ultrahigh-resolution OCT may achieve axial color fundus photographs based on blood vessel ridges. Opt Express
2011;19:7–16.
image resolution of 2–3 µm enabling better visualization of 4. Jiao S, Knighton R, Huang X, et al. Simultaneous acquisition of sectional and
retinal structures. However, the price-versus-performance trade­ fundus ophthalmic images with spectral-domain optical coherence tomo­
graphy. Opt Express 2005;13:444–52.
off remains, limiting the use of this technology to research 5. Han IC, Jaffe GJ. Evaluation of artifacts associated with macular spectral-
applications.2 The use of adaptive optics to correct the ocular domain optical coherence tomography. Ophthalmology 2010;117:1177–89
e4.
aberrations may increase not only the axial resolution, but also 6. Ho J, Sull AC, Vuong LN, et al. Assessment of artifacts and reproducibility
the transverse resolution of OCT systems and provide cellular across spectral- and time-domain optical coherence tomography devices.
Ophthalmology 2009;116:1960–70.
level detail.211 7. Mylonas G, Ahlers C, Malamos P, et al. Comparison of retinal thickness
Swept-source OCT systems allow significant increases in measurements and segmentation performance of four different spectral and
imaging sensitivity and speed (>300 000 A-scans per second), time domain OCT devices in neovascular age-related macular degeneration.
Br J Ophthalmol 2009;93:1453–60.
with decreased motion artifacts, through the use of a tunable 8. Wolf-Schnurrbusch UE, Ceklic L, Brinkmann CK, et al. Macular thickness
laser and a photodetector. While swept-source OCT can achieve measurements in healthy eyes using six different optical coherence tomo­
graphy instruments. Invest Ophthalmol Vis Sci 2009;50:3432–7.
extremely high imaging speeds, the axial image resolution is less 9. Srinivasan VJ, Monson BK, Wojtkowski M, et al. Characterization of outer
than that achieved using SD-OCT.2,212 retinal morphology with high-speed, ultrahigh-resolution optical coherence
tomography. Invest Ophthalmol Vis Sci 2008;49:1571–9.
Clinically available SD-OCT instruments operate with a light 10. Krebs I, Smretschnig E, Moussa S, et al. Quality and reproducibility of retinal
source of approximately 840 nm. This wavelength is highly scat- thickness measurements in two spectral-domain optical coherence tomo­
tered and absorbed by the melanin in the RPE and choroid, graphy machines. Invest Ophthalmol Vis Sci 2011;52:6925–33.
11. Mwanza JC, Oakley JD, Budenz DL, et al. Macular ganglion cell-inner plexi-
reducing the light penetration into deeper tissues. Imaging the form layer: automated detection and thickness reproducibility with spectral
retina with a wavelength of 1050 nm enables greater light pen- domain-optical coherence tomography in glaucoma. Invest Ophthalmol Vis
Sci 2011;52:8323–9.
etration and thus a better visualization of choroidal struc- 12. Yehoshua Z, Rosenfeld PJ, Gregori G, et al. Progression of geographic atrophy
tures.213,214 The use of this wavelength also has the advantage of in age-related macular degeneration imaged with spectral domain optical
coherence tomography. Ophthalmology 2011;118:679–86.
less interference by media opacities such as cataract.215 13. Gregori G, Wang F, Rosenfeld PJ, et al. Spectral domain optical coherence
In the field of functional OCT, Doppler OCT systems are able tomography imaging of drusen in nonexudative age-related macular degen-
eration. Ophthalmology 2011;118:1373–9.
to detect the retinal blood flow by the assessment of light reflec- 14. Yehoshua Z, Wang F, Rosenfeld PJ, et al. Natural history of drusen morphol-
tivity in multiple successive datasets over short time periods.216 ogy in age-related macular degeneration using spectral domain optical
Polarization-sensitive OCT uses tissue birefringence properties coherence tomography. Ophthalmology 2011;118:2434–41.
15. Malamos P, Ahlers C, Mylonas G, et al. Evaluation of segmentation proce-
to detect the health of different retinal layers. The combination dures using spectral domain optical coherence tomography in exudative
of birefringence and thickness measurements may provide a age-related macular degeneration. Retina 2011;31:453–63.
16. Matt G, Sacu S, Buehl W, et al. Comparison of retinal thickness values and
more sensitive diagnostic tool than either alone.217,218 segmentation performance of different OCT devices in acute branch retinal
Much has been learned since the development of the first OCT vein occlusion. Eye (Lond) 2011;25:511–8.
17. Krebs I, Haas P, Zeiler F, et al. Optical coherence tomography: limits of
instrument, and OCT holds the promise for continuing advances the retinal-mapping program in age-related macular degeneration. Br J
in fundamental research and improvements in clinical care. Ophthalmol 2008;92:933–5.
18. Krebs I, Falkner-Radler C, Hagen S, et al. Quality of the threshold algorithm 50. McCarty DJ, Mukesh BN, Chikani V, et al. Prevalence and associations of
in age-related macular degeneration: Stratus versus Cirrus OCT. Invest epiretinal membranes in the visual impairment project. Am J Ophthalmol
Ophthalmol Vis Sci 2009;50:995–1000. 2005;140:288–94.
19. Patel PJ, Chen FK, da Cruz L, et al. Segmentation error in Stratus optical 51. Smiddy WE, Maguire AM, Green WR, et al. Idiopathic epiretinal membranes. 107
coherence tomography for neovascular age-related macular degeneration. Ultrastructural characteristics and clinicopathologic correlation. Ophthalmol-
Invest Ophthalmol Vis Sci 2009;50:399–404. ogy 1989;96:811–20; discussion 821.
20. Drexler W, Sattmann H, Hermann B, et al. Enhanced visualization of macular 52. Vinores SA, Campochiaro PA, Conway BP. Ultrastructural and electron-
pathology with the use of ultrahigh-resolution optical coherence tomography. immunocytochemical characterization of cells in epiretinal membranes.

Chapter 3
Arch Ophthalmol 2003;121:695–706. Invest Ophthalmol Vis Sci 1990;31:14–28.
21. Schuman JS, Pulliafito CA. Optical coherence tomography of ocular diseases. 53. Appiah AP, Hirose T. Secondary causes of premacular fibrosis. Ophthalmol-
Thorofare, NJ: Slack; 2004. ogy 1989;96:389–92.
22. Gloesmann M, Hermann B, Schubert C, et al. Histologic correlation of pig 54. Wilkins JR, Puliafito CA, Hee MR, et al. Characterization of epiretinal mem-
retina radial stratification with ultrahigh-resolution optical coherence tomo­ branes using optical coherence tomography. Ophthalmology 1996;103:
graphy. Invest Ophthalmol Vis Sci 2003;44:1696–703. 2142–51.
23. Anger EM, Unterhuber A, Hermann B, et al. Ultrahigh resolution optical 55. Gass JD. Reappraisal of biomicroscopic classification of stages of development
coherence tomography of the monkey fovea. Identification of retinal sublayers of a macular hole. Am J Ophthalmol 1995;119:752–9.
by correlation with semithin histology sections. Exp Eye Res 2004;78: 56. Takahashi A, Nagaoka T, Yoshida A. Stage 1-A macular hole: a prospective

Optical Coherence Tomography


1117–25. spectral-domain optical coherence tomography study. Retina 2011;31:127–47.
24. Lujan BJ, Roorda A, Knighton RW, et al. Revealing Henle’s fiber layer using 57. Takahashi A, Yoshida A, Nagaoka T, et al. Macular hole formation in fellow
spectral domain optical coherence tomography. Invest Ophthalmol Vis Sci eyes with a perifoveal posterior vitreous detachment of patients with a
2010;52:1486–92. unilateral macular hole. Am J Ophthalmol 2011;151:981–9 e4.
25. Spaide RF, Curcio CA. Anatomical correlates to the bands seen in the outer 58. Kelly NE, Wendel RT. Vitreous surgery for idiopathic macular holes. Results
retina by optical coherence tomography: literature review and model. Retina of a pilot study. Arch Ophthalmol 1991;109:654–9.
2011;31:1609–19. 59. Brooks HL, Jr. Macular hole surgery with and without internal limiting
26. Srinivasan VJ, Ko TH, Wojtkowski M, et al. Noninvasive volumetric imaging membrane peeling. Ophthalmology 2000;107:1939–48; discussion 1948–9.
and morphometry of the rodent retina with high-speed, ultrahigh-resolution 60. Bottoni F, De Angelis S, Luccarelli S, et al. The dynamic healing process
optical coherence tomography. Invest Ophthalmol Vis Sci 2006;47:5522–8. of idiopathic macular holes after surgical repair: a spectral-domain optical
27. Toth CA, Narayan DG, Boppart SA, et al. A comparison of retinal morphology coherence tomography study. Invest Ophthalmol Vis Sci 2011;52:4439–46.
viewed by optical coherence tomography and by light microscopy. 61. Hee MR, Puliafito CA, Wong C, et al. Optical coherence tomography of
Arch Ophthalmol 1997;115:1425–8. macular holes. Ophthalmology 1995;102:748–56.
28. Manjunath V, Taha M, Fujimoto JG, et al. Choroidal thickness in normal eyes 62. Ko TH, Witkin AJ, Fujimoto JG, et al. Ultrahigh-resolution optical coherence
measured using Cirrus HD optical coherence tomography. Am J Ophthalmol tomography of surgically closed macular holes. Arch Ophthalmol 2006;124:
2010;150:325–9 e1. 827–36.
29. Spaide RF, Koizumi H, Pozzoni MC. Enhanced depth imaging spectral- 63. Sano M, Shimoda Y, Hashimoto H, et al. Restored photoreceptor outer
domain optical coherence tomography. Am J Ophthalmol 2008;146:496–500. segment and visual recovery after macular hole closure. Am J Ophthalmol
30. Chen Y, Burnes DL, de Bruin M, et al. Three-dimensional pointwise compari- 2009;147:313–8 e1.
son of human retinal optical property at 845 and 1060 nm using optical 64 Oh J, Smiddy WE, Flynn HW, Jr, et al. Photoreceptor inner/outer segment
frequency domain imaging. J Biomed Opt 2009;14:024016. defect imaging by spectral domain OCT and visual prognosis after macular
31. Margolis R, Spaide RF. A pilot study of enhanced depth imaging optical hole surgery. Invest Ophthalmol Vis Sci 2009;51:1651–8.
coherence tomography of the choroid in normal eyes. Am J Ophthalmol 65. Ooka E, Mitamura Y, Baba T, et al. Foveal microstructure on spectral-domain
2009;147:811–5. optical coherence tomographic images and visual function after macular hole
32. Ouyang Y, Heussen FM, Mokwa N, et al. Spatial distribution of posterior surgery. Am J Ophthalmol 2011;152:283–90 e1.
pole choroidal thickness by spectral domain optical coherence tomography. 66. Muqit MM, Akram I, Turner GS, et al. Fourier-domain optical coherence
Invest Ophthalmol Vis Sci 2011;52:7019–26. tomography imaging of gas tamponade following macular hole surgery.
33. Ikuno Y, Kawaguchi K, Nouchi T, et al. Choroidal thickness in healthy Ophthalmic Surg Lasers Imaging 2011;41 Online: e1–6.
Japanese subjects. Invest Ophthalmol Vis Sci 2009;51:2173–6. 67. Sano M, Inoue M, Taniuchi S, et al. Ability to determine postoperative status
34. Ikuno Y, Maruko I, Yasuno Y, et al. Reproducibility of retinal and choroidal of macular hole in gas-filled eyes by spectral domain-optical coherence
thickness measurements in enhanced depth imaging and high-penetration tomography. Clin Exp Ophthalmol 2011;39:885–892.
optical coherence tomography. Invest Ophthalmol Vis Sci 2011;52:5536–40. 68. Eckardt C, Eckert T, Eckardt U, et al. Macular hole surgery with air
35. Giammaria D, Ioni A, Bartoli B, et al. Comparison of macular thickness mea- tamponade and optical coherence tomography-based duration of face-down
surements between time-domain and spectral-domain optical coherence positioning. Retina 2008;28:1087–96.
tomographies in eyes with and without macular abnormalities. Retina 69. Bressler NM, Bressler SB, Congdon NG, et al. Potential public health impact
2011;31:707–16. of Age-Related Eye Disease Study results: AREDS report no. 11. Arch
36. Gass J. Stereoscopic atlas of macular diseases: diagnosis and treatment. Ophthalmol 2003;121:1621–4.
St Louis: Mosby-Year Book; 1997. p. 903–14. 70. Congdon N, O’Colmain B, Klaver CC, et al. Causes and prevalence of visual
37. Uchino E, Uemura A, Ohba N. Initial stages of posterior vitreous detachment impairment among adults in the United States. Arch Ophthalmol 2004;122:
in healthy eyes of older persons evaluated by optical coherence tomography. 477–85.
Arch Ophthalmol 2001;119:1475–9. 71. Friedman DS, O’Colmain BJ, Munoz B, et al. Prevalence of age-related macular
38. Smiddy WE, Michels RG, Green WR. Morphology, pathology, and surgery of degeneration in the United States. Arch Ophthalmol 2004;122:564–72.
idiopathic vitreoretinal macular disorders. A review. Retina 1990;10:288–96. 72. Klein R, Peto T, Bird A, et al. The epidemiology of age-related macular
39. Johnson MW. Perifoveal vitreous detachment and its macular complications. degeneration. Am J Ophthalmol 2004;137:486–95.
Trans Am Ophthalmol Soc 2005;103:537–67. 73. Bird AC, Bressler NM, Bressler SB, et al. An international classification and
40. Witkin AJ, Patron ME, Castro LC, et al. Anatomic and visual outcomes of grading system for age-related maculopathy and age-related macular degen-
vitrectomy for vitreomacular traction syndrome. Ophthalm Surg Lasers eration. The International ARM Epidemiological Study Group. Surv Ophthal-
Imaging 2010;41:425–31. mol 1995;39:367–74.
41. Gallemore RP, Jumper JM, McCuen BW, 2nd, et al. Diagnosis of vitreoretinal 74. Hirvela H, Luukinen H, Laara E, et al. Risk factors of age-related maculopathy
adhesions in macular disease with optical coherence tomography. Retina in a population 70 years of age or older. Ophthalmology 1996;103:871–7.
2000;20:115–20. 75. Vingerling JR, Hofman A, Grobbee DE, et al. Age-related macular degenera-
42. Johnson MW. Tractional cystoid macular edema: a subtle variant of the tion and smoking. The Rotterdam Study. Arch Ophthalmol 1996;114:1193–6.
vitreomacular traction syndrome. Am J Ophthalmol 2005;140:184–92. 76. Seddon JM, Sharma S, Adelman RA. Evaluation of the clinical age-related
43. Do DV, Cho M, et al. Impact of optical coherence tomography on surgical maculopathy staging system. Ophthalmology 2006;113:260–6.
decision making for epiretinal membranes and vitreomacular traction. Retina 77. Bartlett H, Eperjesi F. Use of fundus imaging in quantification of age-related
2007;27:552–6. macular change. Surv Ophthalmol 2007;52:655–71.
44. Chang LK, Fine HF, Spaide RF, et al. Ultrastructural correlation of spectral- 78. Seddon JM, Sharma S, Adelman RA, et al. Optical coherence tomography
domain optical coherence tomographic findings in vitreomacular traction of age-related macular degeneration and choroidal neovascularization.
syndrome. Am J Ophthalmol 2008;146:121–7. Ophthalmology 1996;103:1260–70.
45. Hikichi T, Yoshida A, Trempe CL. Course of vitreomacular traction 79. Pieroni CG, Witkin AJ, Ko TH, et al. Ultrahigh resolution optical coherence
syndrome. Am J Ophthalmol 1995;119:55–61. tomography in non-exudative age related macular degeneration. Br J
46. Sulkes DJ, Ip MS, Baumal CR, et al. Spontaneous resolution of vitreomacular Ophthalmol 2006;90:191–7.
traction documented by optical coherence tomography. Arch Ophthalmol 80. Sikorski BL, Bukowska D, Kaluzny JJ, et al. Drusen with accompanying fluid
2000;118:286–7. underneath the sensory retina. Ophthalmology 2010;118:82–92.
47. Yamada N, Kishi S. Tomographic features and surgical outcomes of 81. Spaide RF, Curcio CA. Drusen characterization with multimodal imaging.
vitreomacular traction syndrome. Am J Ophthalmol 2005;139:112–7. Retina 2010;30:1441–54.
48. Sonmez K, Capone A, Jr, Trese MT, et al. Vitreomacular traction syndrome: 82. Roquet W, Roudot-Thoraval F, Coscas G, et al. Clinical features of drusenoid
impact of anatomical configuration on anatomical and visual outcomes. pigment epithelial detachment in age related macular degeneration. Br J Oph-
Retina 2008;28:1207–14. thalmol 2004;88:638–42.
49. Pearlstone AD. The incidence of idiopathic preretinal macular gliosis. 83. Schuman SG, Koreishi AF, Farsiu S, et al. Photoreceptor layer thinning over
Ann Ophthalmol 1985;17:378–80. drusen in eyes with age-related macular degeneration imaged in vivo with
spectral-domain optical coherence tomography. Ophthalmology 2009;116: 116. Ishida Y, Kato T, Minamoto A, et al. Retinal pigment epithelial tear in a
488–96 e2. patient with central serous chorioretinopathy treated with corticosteroids.
84. Davis MD, Gangnon RE, Lee LY, et al. The Age-Related Eye Disease Study Retina 2004;24:633–6.
108 severity scale for age-related macular degeneration: AREDS report no. 17. 117. Gelisken F, Inhoffen W, Partsch M, et al. Retinal pigment epithelial tear after
Arch Ophthalmol 2005;123:1484–98. photodynamic therapy for choroidal neovascularization. Am J Ophthalmol
85. Ferris FL, Davis MD, Clemons TE, et al. A simplified severity scale for age- 2001;131:518–20.
related macular degeneration: AREDS report no. 18. Arch Ophthalmol 118. Pece A, Introini U, Bottoni F, et al. Acute retinal pigment epithelial tear after
2005;123:1570–4. photodynamic therapy. Retina 2001;21:661–5.
Section 1

86. Ho J, Witkin AJ, Liu J, et al. Documentation of intraretinal retinal pigment 119. Meyer CH, Mennel S, Schmidt JC, et al. Acute retinal pigment epithelial
epithelium migration via high-speed ultrahigh-resolution optical coherence tear following intravitreal bevacizumab (Avastin) injection for occult
tomography. Ophthalmology 2010;118:687–93. choroidal neovascularisation secondary to age related macular degeneration.
87. Zweifel SA, Imamura Y, Spaide TC, et al. Prevalence and significance of Br J Ophthalmol 2006;90:1207–8.
subretinal drusenoid deposits (reticular pseudodrusen) in age-related macular 120. Carvounis PE, Kopel AC, Benz MS. Retinal pigment epithelium tears
degeneration. Ophthalmology 2010;117:1775–81. following ranibizumab for exudative age-related macular degeneration.
88. Zweifel SA, Spaide RF, Curcio CA, et al. Reticular pseudodrusen are Am J Ophthalmol 2007;143:504–5.
subretinal drusenoid deposits. Ophthalmology 2009;117:303–12 e1. 121. Gelisken F, Ziemssen F, Voelker M, et al. Retinal pigment epithelial tears after
Optical Imaging Technologies

89. Sarks JP, Sarks SH, Killingsworth MC. Evolution of geographic atrophy of the single administration of intravitreal bevacizumab for neovascular age-related
Retinal Imaging and Diagnostics

retinal pigment epithelium. Eye (Lond) 1988;2:552–77. macular degeneration. Eye (Lond) 2009;23:694–702.
90. Sunness JS. The natural history of geographic atrophy, the advanced atrophic 122. Gass JD. Pathogenesis of tears of the retinal pigment epithelium. Br J
form of age-related macular degeneration. Mol Vis 1999;5:25. Ophthalmol 1984;68:513–9.
91. Sunness JS, Bressler NM, Tian Y, et al. Measuring geographic atrophy in 123. Chan CK, Abraham P, Meyer CH, et al. Optical coherence tomography-
advanced age-related macular degeneration. Invest Ophthalmol Vis Sci measured pigment epithelial detachment height as a predictor for retinal
1999;40:1761–9. pigment epithelial tears associated with intravitreal bevacizumab injections.
92. Sunness JS, Margalit E, Srikumaran D, et al. The long-term natural history of Retina 2009;30:203–11.
geographic atrophy from age-related macular degeneration: enlargement of 124. Chiang A, Chang LK, Yu F, et al. Predictors of anti-VEGF-associated retinal
atrophy and implications for interventional clinical trials. Ophthalmology pigment epithelial tear using FA and OCT analysis. Retina 2008;28:1265–9.
2007;114:271–7. 125 Landa G, Su E, Garcia PM, et al. Inner segment–outer segment junctional layer
93. Schmitz-Valckenberg S, Fleckenstein M, Gobel AP, et al. Optical coherence integrity and corresponding retinal sensitivity in dry and wet forms of
tomography and autofluorescence findings in areas with geographic atrophy age-related macular degeneration. Retina 2011;31:364–70.
due to age-related macular degeneration. Invest Ophthalmol Vis Sci 126. Oishi A, Hata M, Shimozono M, et al. The significance of external limiting
2010;52:1–6. membrane status for visual acuity in age-related macular degeneration.
94. Fleckenstein M, Charbel Issa P, Helb HM, et al. High-resolution spectral Am J Ophthalmol 2010;150:27–32 e1.
domain-OCT imaging in geographic atrophy associated with age-related 127. Yannuzzi LA, Negrao S, Iida T, et al. Retinal angiomatous proliferation in
macular degeneration. Invest Ophthalmol Vis Sci 2008;49:4137–44. age-related macular degeneration. Retina 2001;21:416–34.
95. Bearelly S, Chau FY, Koreishi A, et al. Spectral domain optical coherence 128. Gass JD, Agarwal A, Lavina AM, et al. Focal inner retinal hemorrhages in
tomography imaging of geographic atrophy margins. Ophthalmology patients with drusen: an early sign of occult choroidal neovascularization and
2009;116:1762–9. chorioretinal anastomosis. Retina 2003;23:741–51.
96. Spaide RF. Age-related choroidal atrophy. Am J Ophthalmol 2009;147: 129. Truong SN, Alam S, Zawadzki RJ, et al. High resolution Fourier-domain
801–10. optical coherence tomography of retinal angiomatous proliferation. Retina
97. Wood A, Binns A, Margrain T, et al. Retinal and choroidal thickness in early 2007;27:915–25.
age-related macular degeneration. Am J Ophthalmol 2011;152:1030–8. 130. Freund KB, Ho IV, Barbazetto IA, et al. Type 3 neovascularization: the
98. Lujan BJ, Wang F, Gregori G, et al. Calibration of fundus images using spectral expanded spectrum of retinal angiomatous proliferation. Retina 2008;28:
domain optical coherence tomography. Ophthalmic Surg Lasers Imaging 201–11.
2008;39(Suppl):S15–20. 131. Querques G, Atmani K, Berboucha E, et al. Angiographic analysis of retinal-
99. Lujan BJ, Rosenfeld PJ, Gregori G, et al. Spectral domain optical coherence choroidal anastomosis by confocal scanning laser ophthalmoscopy technol-
tomographic imaging of geographic atrophy. Ophthalmic Surg Lasers ogy and corresponding (eye-tracked) spectral-domain optical coherence
Imaging 2009;40:96–101. tomography. Retina 2009;30:222–34.
100. Green WR. Clinicopathologic studies of treated choroidal neovascular 132. Krebs I, Glittenberg C, Hagen S, et al. Retinal angiomatous proliferation:
membranes. A review and report of two cases. Retina 1991;11:328–56. morphological changes assessed by Stratus and Cirrus OCT. Ophthalmic Surg
101. Green WR. Histopathology of age-related macular degeneration. Mol Vis Lasers Imaging 2009;40:285–9.
1999;5:27. 133. Yannuzzi LA, Sorenson J, Spaide RF, et al. Idiopathic polypoidal choroidal
102. Keane PA, Aghaian E, Ouyang Y, et al. Acute severe visual decrease after vasculopathy (IPCV). Retina 1990;10:1–8.
photodynamic therapy with verteporfin: spectral-domain OCT features. 134. Costa RA, Navajas EV, Farah ME, et al. Polypoidal choroidal vasculopathy:
Ophthalmic Surg Lasers Imaging 2010;41(Suppl):S85–8. angiographic characterization of the network vascular elements and a new
103. Keane PA, Liakopoulos S, Chang KT, et al. Relationship between optical treatment paradigm. Prog Retin Eye Res 2005;24:560–86.
coherence tomography retinal parameters and visual acuity in neovascular 135. Saito M, Iida T, Nagayama D. Cross-sectional and en face optical coherence
age-related macular degeneration. Ophthalmology 2008;115:2206–14. tomographic features of polypoidal choroidal vasculopathy. Retina
104. Yannuzzi LA, Hope-Ross M, Slakter JS, et al. Analysis of vascularized 2008;28:459–64.
pigment epithelial detachments using indocyanine green videoangiography. 136. Ojima Y, Hangai M, Sakamoto A, et al. Improved visualization of polypoidal
Retina 1994;14:99–113. choroidal vasculopathy lesions using spectral-domain optical coherence
105. Jager RD, Mieler WF, Miller JW. Age-related macular degeneration. N Engl J tomography. Retina 2009;29:52–9.
Med 2008;358:2606–17. 137. Harding SP. Neovascular age-related macular degeneration: decision making
106. Zayit-Soudry S, Moroz I, Loewenstein A. Retinal pigment epithelial detach- and optimal management. Eye (Lond) 2010;24:497–505.
ment. Surv Ophthalmol 2007;52:227–43. 138. Brown DM, Kaiser PK, Michels M, et al. Ranibizumab versus verteporfin for
107. Bloom SM, Singal IP. The outer Bruch membrane layer: a previously unde- neovascular age-related macular degeneration. N Engl J Med 2006;355:
scribed spectral-domain optical coherence tomography finding. Retina 2010; 1432–44.
31:316–23. 139. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizumab for neovascular age-
108. Coscas G, Coscas F, Zourdani A, et al. [Optical coherence tomography and related macular degeneration. N Engl J Med 2006;355:1419–31.
ARMD.] J Fr Ophtalmol 2004;27:3S7–30. 140. Engelbert M, Zweifel SA, Freund KB. Long-term follow-up for type 1 (sub-
109. Pepple K, Mruthyunjaya P. Retinal pigment epithelial detachments in retinal pigment epithelium) neovascularization using a modified “treat and
age-related macular degeneration: classification and therapeutic options. extend” dosing regimen of intravitreal antivascular endothelial growth factor
Semin Ophthalmol 2011;26:198–208. therapy. Retina 2010;30:1368–75.
110. Spaide RF. Enhanced depth imaging optical coherence tomography of 141. Martin DF, Maguire MG, Ying GS, et al. Ranibizumab and bevacizumab for
retinal pigment epithelial detachment in age-related macular degeneration. neovascular age-related macular degeneration. N Engl J Med 2011;364:
Am J Ophthalmol 2009;147:644–52. 1897–908.
111. Joeres S, Tsong JW, Updike PG, et al. Reproducibility of quantitative optical 142. Fung AE, Lalwani GA, Rosenfeld PJ, et al. An optical coherence tomography-
coherence tomography subanalysis in neovascular age-related macular guided, variable dosing regimen with intravitreal ranibizumab (Lucentis) for
degeneration. Invest Ophthalmol Vis Sci 2007;48:4300–7. neovascular age-related macular degeneration. Am J Ophthalmol 2007;143:
112. Lee SY, Stetson PF, Ruiz-Garcia H, et al. Automated characterization of 566–83.
pigment epithelial detachment using optical coherence tomography. 143. Engelbert M, Zweifel SA, Freund KB. “Treat and extend” dosing of intravitreal
Invest Ophthalmol Vis Sci 2012;53:164–70. antivascular endothelial growth factor therapy for type 3 neovascularization/
113. Hoskin A, Bird AC, Sehmi K. Tears of detached retinal pigment epithelium. retinal angiomatous proliferation. Retina 2009;29:1424–31.
Br J Ophthalmol 1981;65:417–22. 144. Lalwani GA, Rosenfeld PJ, Fung AE, et al. A variable-dosing regimen with
114. Chang LK, Sarraf D. Tears of the retinal pigment epithelium: an old problem intravitreal ranibizumab for neovascular age-related macular degeneration:
in a new era. Retina 2007;27:523–34. year 2 of the PrONTO Study. Am J Ophthalmol 2009;148:43–58 e1.
115. Levin LA, Seddon JM, Topping T. Retinal pigment epithelial tears associated 145. Zweifel SA, Engelbert M, Laud K, et al. Outer retinal tubulation: a novel
with trauma. Am J Ophthalmol 1991;112:396–400. optical coherence tomography finding. Arch Ophthalmol 2009;127:1596–602.
146. Gass JD. Pathogenesis of disciform detachment of the neuroepithelium. Am J 178. Irvine SR. A newly defined vitreous syndrome following cataract surgery.
Ophthalmol 1967;63:Suppl:1–139. Am J Ophthalmol 1953;36:499–619.
147. Spaide RF, Campeas L, Haas A, et al. Central serous chorioretinopathy in 179. Schepens CL, Avila MP, Jalkh AE, et al. Role of the vitreous in cystoid macular
younger and older adults. Ophthalmology 1996;103:2070–9; discussion edema. Surv Ophthalmol 1984;28(Suppl):499–504. 109
2079–80. 180. Moss SE, Klein R, Klein BE. Ten-year incidence of visual loss in a diabetic
148. Kitaya N, Nagaoka T, Hikichi T, et al. Features of abnormal choroidal circula- population. Ophthalmology 1994;101:1061–70.
tion in central serous chorioretinopathy. Br J Ophthalmol 2003;87:709–12. 181. Moss SE, Klein R, Klein BE. The 14-year incidence of visual loss in a diabetic
149. Jalkh AE, Jabbour N, Avila MP, et al. Retinal pigment epithelium decompen- population. Ophthalmology 1998;105:998–1003.

Chapter 3
sation. I. Clinical features and natural course. Ophthalmology 1984;91: 182. Strom C, Sander B, Larsen N, et al. Diabetic macular edema assessed
1544–8. with optical coherence tomography and stereo fundus photography.
150. Yannuzzi LA, Shakin JL, Fisher YL, et al. Peripheral retinal detachments and Invest Ophthalmol Vis Sci 2002;43:241–5.
retinal pigment epithelial atrophic tracts secondary to central serous pigment 183. Brown JC, Solomon SD, Bressler SB, et al. Detection of diabetic foveal edema:
epitheliopathy. Ophthalmology 1984;91:1554–72. contact lens biomicroscopy compared with optical coherence tomography.
151. Scheider A, Nasemann JE, Lund OE. Fluorescein and indocyanine green angi- Arch Ophthalmol 2004;122:330–5.
ographies of central serous choroidopathy by scanning laser ophthalmoscopy. 184. Browning DJ, Apte RS, Bressler SB, et al. Association of the extent of diabetic
Am J Ophthalmol 1993;115:50–6. macular edema as assessed by optical coherence tomography with visual
152. Guyer DR, Yannuzzi LA, Slakter JS, et al. Digital indocyanine green video­ acuity and retinal outcome variables. Retina 2009;29:300–5.

Optical Coherence Tomography


angiography of central serous chorioretinopathy. Arch Ophthalmol 1994;112: 185. Al-latayfeh MM, Sun JK, Aiello LP. Ocular coherence tomography and
1057–62. diabetic eye disease. Semin Ophthalmol 2010;25:192–7.
153. Piccolino FC, Borgia L. Central serous chorioretinopathy and indocyanine 186. Browning DJ, McOwen MD, Bowen RM, Jr, et al. Comparison of the clinical
green angiography. Retina 1994;14:231–42. diagnosis of diabetic macular edema with diagnosis by optical coherence
154. Prunte C, Flammer J. Choroidal capillary and venous congestion in central tomography. Ophthalmology 2004;111:712–5.
serous chorioretinopathy. Am J Ophthalmol 1996;121:26–34. 187. Virgili G, Menchini F, Dimastrogiovanni AF, et al. Optical coherence tomog-
155. Kamppeter B, Jonas JB. Central serous chorioretinopathy imaged by optical raphy versus stereoscopic fundus photography or biomicroscopy for diagnos-
coherence tomography. Arch Ophthalmol 2003;121:742–3. ing diabetic macular edema: a systematic review. Invest Ophthalmol Vis Sci
156. Montero JA, Ruiz-Moreno JM. Optical coherence tomography characterisation 2007;48:4963–73.
of idiopathic central serous chorioretinopathy. Br J Ophthalmol 2005;89: 188. Davis MD, Bressler SB, Aiello LP, et al. Comparison of time-domain OCT and
562–4. fundus photographic assessments of retinal thickening in eyes with diabetic
157. van Velthoven ME, Verbraak FD, Garcia PM, et al. Evaluation of macular edema. Invest Ophthalmol Vis Sci 2008;49:1745–52.
central serous retinopathy with en face optical coherence tomography. 189. Otani T, Kishi S, Maruyama Y. Patterns of diabetic macular edema with
Br J Ophthalmol 2005;89:1483–8. optical coherence tomography. Am J Ophthalmol 1999;127:688–93.
158. Hussain N, Baskar A, Ram LM, et al. Optical coherence tomographic pattern 190. Kim BY, Smith SD, Kaiser PK. Optical coherence tomographic patterns of
of fluorescein angiographic leakage site in acute central serous chorioreti- diabetic macular edema. Am J Ophthalmol 2006;142:405–12.
nopathy. Clin Experiment Ophthalmol 2006;34:137–40. 191. Soliman W, Sander B, Hasler PW, et al. Correlation between intraretinal
159. Hirami Y, Tsujikawa A, Sasahara M, et al. Alterations of retinal pigment changes in diabetic macular oedema seen in fluorescein angiography and
epithelium in central serous chorioretinopathy. Clin Experiment Ophthalmol optical coherence tomography. Acta Ophthalmol 2008;86:34–9.
2007;35:225–30. 192. Early Treatment Diabetic Retinopathy Study research group. Photocoagula-
160. Wang MS, Sander B, Larsen M. Retinal atrophy in idiopathic central serous tion for diabetic macular edema. Early Treatment Diabetic Retinopathy Study
chorioretinopathy. Am J Ophthalmol 2002;133:787–93. report number 1. Arch Ophthalmol 1985;103:1796–806.
161. Stock G, Ahlers C, Sayegh R, et al. [Three-dimensional imaging in central 193. Early Treatment Diabetic Retinopathy Study Research Group. Photocoagula-
serous chorioretinopathy]. Ophthalmologe 2008;105:1127–34. tion for diabetic macular edema: Early Treatment Diabetic Retinopathy Study
162. Eandi CM, Chung JE, Cardillo-Piccolino F, et al. Optical coherence tomo­ report no. 4. Int Ophthalmol Clin 1987;27:265–72.
graphy in unilateral resolved central serous chorioretinopathy. Retina 194. Early Treatment Diabetic Retinopathy Study Research Group. Treatment tech-
2005;25:417–21. niques and clinical guidelines for photocoagulation of diabetic macular edema.
163. Furuta M, Iida T, Kishi S. Foveal thickness can predict visual outcome in Early Treatment Diabetic Retinopathy Study report no. 2. Ophthalmology
patients with persistent central serous chorioretinopathy. Ophthalmologica 1987;94:761–74.
2009;223:28–31. 195. Elman MJ, Bressler NM, Qin H, et al. Expanded 2-year follow-up of ranibi-
164. Maruko I, Iida T, Sekiryu T, et al. Morphologic changes in the outer layer of zumab plus prompt or deferred laser or triamcinolone plus prompt laser for
the detached retina in rhegmatogenous retinal detachment and central serous diabetic macular edema. Ophthalmology 2011;118:609–14.
chorioretinopathy. Am J Ophthalmol 2009;147:489–94 e1. 196. Michaelides M, Kaines A, Hamilton RD, et al. A prospective randomized trial
165. Fujimoto H, Gomi F, Wakabayashi T, et al. Morphologic changes in acute of intravitreal bevacizumab or laser therapy in the management of diabetic
central serous chorioretinopathy evaluated by Fourier-domain optical coher- macular edema (BOLT study) 12-month data: report 2. Ophthalmology
ence tomography. Ophthalmology 2008;115:1494–500, 1500 e1–2. 2010;117:1078–86 e2.
166. Matsumoto H, Kishi S, Otani T, et al. Elongation of photoreceptor outer 197. Kim NR, Kim YJ, Chin HS, et al. Optical coherence tomographic patterns in
segment in central serous chorioretinopathy. Am J Ophthalmol 2008;145: diabetic macular oedema: prediction of visual outcome after focal laser pho-
162–168. tocoagulation. Br J Ophthalmol 2009;93:901–5.
167. Hee MR, Puliafito CA, Wong C, et al. Optical coherence tomography of central 198. Soliman W, Sander B, Soliman KA, et al. The predictive value of optical coher-
serous chorioretinopathy. Am J Ophthalmol 1995;120:65–74. ence tomography after grid laser photocoagulation for diffuse diabetic
168. Kon Y, Iida T, Maruko I, et al. The optical coherence tomography- macular oedema. Acta Ophthalmol 2008;86:284–91.
ophthalmoscope for examination of central serous chorioretinopathy with 199. Kaiser PK, Riemann CD, Sears JE, et al. Macular traction detachment
precipitates. Retina 2008;28:864–9. and diabetic macular edema associated with posterior hyaloidal traction.
169. Mitarai K, Gomi F, Tano Y. Three-dimensional optical coherence tomographic Am J Ophthalmol 2001;131:44–9.
findings in central serous chorioretinopathy. Graefes Arch Clin Exp 200. Ghazi NG, Ciralsky JB, Shah SM, et al. Optical coherence tomography findings
Ophthalmol 2006;244:1415–20. in persistent diabetic macular edema: the vitreomacular interface. Am J
170. Gupta P, Gupta V, Dogra MR, et al. Morphological changes in the retinal Ophthalmol 2007;144:747–754.
pigment epithelium on spectral-domain OCT in the unaffected eyes with 201. Haller JA, Qin H, Apte RS, et al. Vitrectomy outcomes in eyes with diabetic
idiopathic central serous chorioretinopathy. Int Ophthalmol 2009;30:175–81. macular edema and vitreomacular traction. Ophthalmology 2010;117:1087–93
171. Ojima Y, Hangai M, Sasahara M, et al. Three-dimensional imaging of the e3.
foveal photoreceptor layer in central serous chorioretinopathy using high- 202. Maheshwary AS, Oster SF, Yuson RM, et al. The association between percent
speed optical coherence tomography. Ophthalmology 2007;114:2197–207. disruption of the photoreceptor inner segment–outer segment junction and
172. Matsumoto H, Sato T, Kishi S. Outer nuclear layer thickness at the fovea visual acuity in diabetic macular edema. Am J Ophthalmol 2010;150:63–7
determines visual outcomes in resolved central serous chorioretinopathy. e1.
Am J Ophthalmol 2009;148:105–10 e1. 203. Otani T, Yamaguchi Y, Kishi S. Correlation between visual acuity and
173. Imamura Y, Fujiwara T, Margolis R, et al. Enhanced depth imaging optical foveal microstructural changes in diabetic macular edema. Retina 2010;30:
coherence tomography of the choroid in central serous chorioretinopathy. 774–80.
Retina 2009;29:1469–73. 204. Sakamoto A, Nishijima K, Kita M, et al. Association between foveal pho-
174. Maruko I, Iida T, Sugano Y, et al. Subfoveal choroidal thickness in fellow eyes toreceptor status and visual acuity after resolution of diabetic macular edema
of patients with central serous chorioretinopathy. Retina 2011;31:1603–8. by pars plana vitrectomy. Graefes Arch Clin Exp Ophthalmol 2009;247:
175. Maruko I, Iida T, Sugano Y, et al. One-year choroidal thickness results after 1325–30.
photodynamic therapy for central serous chorioretinopathy. Retina 2011; 205. Hayreh SS. Classification of central retinal vein occlusion. Ophthalmology
31:1921–7. 1983;90:458–74.
176. Henderson BA, Kim JY, Ament CS, et al. Clinical pseudophakic cystoid 206. Ota M, Tsujikawa A, Murakami T, et al. Foveal photoreceptor layer in eyes
macular edema. Risk factors for development and duration after treatment. with persistent cystoid macular edema associated with branch retinal vein
J Cataract Refract Surg 2007;33:1550–8. occlusion. Am J Ophthalmol 2008;145:273–280.
177. Wolf EJ, Braunstein A, Shih C, et al. Incidence of visually significant pseudo- 207. Shroff D, Mehta DK, Arora R, et al. Natural history of macular status in recent-
phakic macular edema after uneventful phacoemulsification in patients onset branch retinal vein occlusion: an optical coherence tomography study.
treated with nepafenac. J Cataract Refract Surg 2007;33:1546–9. Int Ophthalmol 2008;28:261–8.
208. Falkenberry SM, Ip MS, Blodi BA, et al. Optical coherence tomography find- 214. Povazay B, Hermann B, Hofer B, et al. Wide-field optical coherence tomo­
ings in central retinal artery occlusion. Ophthalmic Surg Lasers Imaging graphy of the choroid in vivo. Invest Ophthalmol Vis Sci 2009;50:
2006;37:502–5. 1856–63.
110 209. Goldenberg-Cohen N, Dadon S, Avraham BC, et al. Molecular and histologi- 215. Povazay B, Hermann B, Unterhuber A, et al. Three-dimensional optical coher-
cal changes following central retinal artery occlusion in a mouse model. ence tomography at 1050 nm versus 800 nm in retinal pathologies: enhanced
Exp Eye Res 2008;87:327–33. performance and choroidal penetration in cataract patients. J Biomed Opt
210. Murthy RK, Grover S, Chalam KV. Sequential spectral domain OCT 2007;12:041211.
documentation of retinal changes after branch retinal artery occlusion. 216. Wang Y, Lu A, Gil-Flamer J, et al. Measurement of total blood flow in the
Section 1

Clin Ophthalmol 2010;4:327–9. normal human retina using Doppler Fourier-domain optical coherence
211. Fernandez EJ, Hermann B, Povazay B, et al. Ultrahigh resolution optical tomography. Br J Ophthalmol 2009;93:634–7.
coherence tomography and pancorrection for cellular imaging of the living 217. Ahlers C, Gotzinger E, Pircher M, et al. Imaging of the retinal pigment
human retina. Opt Express 2008;16:11083–94. epithelium in age-related macular degeneration using polarization-sensitive
212. Huber R, Adler DC, Srinivasan VJ, et al. Fourier domain mode locking at optical coherence tomography. Invest Ophthalmol Vis Sci 2010;51:
1050 nm for ultra-high-speed optical coherence tomography of the human 2149–57.
retina at 236 000 axial scans per second. Opt Lett 2007;32:2049–51. 218. Cense B, Chen TC, Park BH, et al. Thickness and birefringence of healthy
213. Unterhuber A, Povazay B, Hermann B, et al. In vivo retinal optical coherence retinal nerve fiber layer tissue measured with polarization-sensitive optical
Optical Imaging Technologies

tomography at 1040 nm – enhanced penetration into the choroid. Opt Express coherence tomography. Invest Ophthalmol Vis Sci 2004;45:2606–12.
Retinal Imaging and Diagnostics

2005;13:3252–8.
Optical Imaging Technologies Section 1

Autofluorescence Imaging Chapter

Monika Fleckenstein, Steffen Schmitz-Valckenberg, Frank G. Holz


4 
BASIC PRINCIPLES suggested that products of the photo-oxidation of RPE LF com-
ponents could serve as a trigger for the complement system
Fundus autofluorescence which could predispose the macular area to a chronic, low-grade
Fundus autofluorescence (FAF) imaging is a noninvasive inflammatory process over time.
imaging method for in vivo mapping of naturally or pathologi- Detection of LF and its constituents is facilitated by its auto-
cally occurring fluorophores of the ocular fundus. The dominant fluorescent properties. When stimulated with light in the blue
sources are fluorophores accumulating in lipofuscin (LF) gran- range, LF granules typically emit a green-yellow fluorescence.16,17
ules in the retinal pigment epithelium (RPE).1 In the absence of The distribution of LF in postmitotic human RPE cells and its
RPE cells, minor fluorophores including collagen and elastin, accumulation with age have been extensively studied in vitro,
e.g., in choroidal blood vessel walls, may also become visible. applying fluorescence microscopic techniques.5,6,8
Bleaching phenomena and loss of photopigment may result in
increased FAF by reduced absorbance anterior to the RPE level. Near-infrared autofluorescence
Near-infrared autofluorescence (NIA) images can also be
Retinal pigment epithelium and lipofuscin obtained in vivo, most commonly and easily by using the indo-
The RPE constitutes a polygonal monolayer between the neuro- cyanine green angiography mode of the scanning laser ophthal-
sensory retina and the choroid and is essential for vision. Given moscope, i.e., without dye injection.18,19 Due to the excitation and
multiple essential physiological functions of the RPE, it is not emission in the red end of the spectrum, the topographic distri-
surprising that RPE dysfunction has been implicated in a variety bution of fluorophores other than LF may be studied by this
of retinal diseases (reviewed by Schmitz-Valckenberg et al.2). technique. It has been suggested that the NIA signal is largely
A hallmark of aging is the gradual accumulation of LF gran- melanin-derived.18–20 As such, Keilhauer and Delori18 further
ules in the cytoplasm of RPE cells. It is thought that progressive speculated that, to varying degrees, choroidal sources contrib-
LF accumulation is mainly a byproduct of the constant phago- uted to this signal. Gibbs et al.21 investigated NIA in humans and
cytosis of shed photoreceptor outer-segment discs.3–5 Several mice and suggested that melanosomes in the RPE and choroid
lines of evidence indicate that adverse effects of excessive LF were likely the dominant origin of the signal. Except for meas­
accumulation represent a common downstream pathogenetic urements in cell cultures at low magnification, their analyses
mechanism in various monogenic macular and retinal dystro- were limited to excitation at 633 nm, in contrast to in vivo NIA,
phies as well as in multifactorial complex retinal disease entities, which is generated at 795 nm. Using a customized magnification
including age-related macular degeneration (AMD).3,4,6–8 lens attached to the front of the confocal scanning laser ophthal-
Apparently, once formed, the RPE cell has no means of either moscope (cSLO), Schmitz-Valckenberg and coworkers studied
degrading or transporting LF material and granules into the the distribution of the NIA signal in retinal cross-sections of a
extracellular space via exocytosis. Subsequently, these granules human donor eye and correlated ex vivo autofluorescence meas­
are trapped in the cytoplasmic space of the postmitotic RPE urements to in vivo findings in a rat animal model.22 They
cells. Previous studies have shown that various LF components observed that the NIA signal was spatially confined to the RPE
such as A2-E (N-retinylidene-N-retinylethanol-amine), a domi- monolayer and melanin in the choroid.
nant fluorophore, possess toxic properties which may interfere
with normal cell function via various molecular mechanisms, Macular pigment imaging
including impairment of lysosomal degradation due to inhibi- Macular pigment, consisting of lutein and zeaxanthin, exten-
tion of the lysosomal adenosine triphosphate-dependent proton sively accumulates along the axons of the cone photoreceptors
pump.9–12 Other components of LF include precursors of A2-E, in the central retina.23–25 As has been reported, a number of func-
molecules formed by the mixture of oxygen-containing moieties tions have been proposed for macular pigment,24,25 including
within photo-oxidized A2-E, reactions between retinoids and filtration of blue light which may reduce photo damage and
other constituents other than ethanolamine, and peroxidation glare, minimization of the effects of chromatic aberration on
products of proteins and lipids.13,14 The molecular composition visual acuity, improvement in fine-detail discrimination, and
of LF may possibly be dependent on specific underlying molecu- enhancement of contrast sensitivity. Neutralization of reactive
lar mechanisms. Zhou and associates demonstrated with an in oxygen species by macular pigment may have a protective effect
vitro assay a link between inflammation, activation of the com- on the neurosensory retina. Although there may be a large varia-
plement system, oxidative damage, drusen, and RPE LF.15 They tion with regard to the concentration of macular pigment, the
pattern of distribution is relatively uniform in the normal popu- Along with autofluorescence recordings in patients with
lation. It generally shows a peak concentration at the foveal several pathological conditions, the initial work by Delori et al.1
112 center and rapidly decreases with eccentricity, with very little demonstrated that LF is the dominant source of intrinsic fluores-
present at about 8° of eccentricity. cence of the ocular fundus. However, the small area sampled by
Peak absorption of luteal pigment is at 460 nm. These absorp- the fundus spectrometer as well as the customized relatively
tion properties can be readily recorded in vivo by blue-light complex instrumentation and techniques were not practical for
Section 1

autofluorescence imaging.26 Therefore, blue FAF imaging can recording fundus autofluorescence from patients in a clinical
also be used to determine the topographic distribution of macular setting.
pigment. Compared to other methods, including heterochro-
matic flicker photometry, the advantage of FAF imaging is its Scanning laser ophthalmoscopy
objective acquisition technique which is not dependent on psy- Confocal scanning laser ophthalmoscopy (cSLO) optimally
chophysical cooperation by the examined individual. addresses the limitations of the low intensity of the autofluores-
Optical Imaging Technologies
Retinal Imaging and Diagnostics

cence signal and the interference of the crystalline lens. It was


TECHNIQUES OF FUNDUS used initially by von Rückmann and coworkers in a clinical
imaging system.27 The confocal scanning laser ophthalmoscope
AUTOFLUORESCENCE IMAGING projects a low-power laser beam on the retina which is swept
Recording of autofluorescence images is noninvasive and across the fundus in a raster pattern.28 The intensity of the
requires relatively little time. reflected light at each point, after passing through the confocal
The intensity of naturally occurring fluorescence of the ocular pinhole, is registered by means of a detector, and a two-
fundus is about 2 orders of magnitude lower than the back- dimensional image is subsequently generated. Confocal optics
ground of a fluorescein angiogram at the most intense part of insure that out-of-focus light (i.e., light originating outside the
the dye transit.1 Absorption of light with reduction of the fluo- adjusted focal plane, but within the light beam) is suppressed
rescence signal, or excitation and emission of light with an and, thus, the image contrast is enhanced. This suppression
increase in the fluorescence signal by anatomical structures ante- increases with the distance from the focal plane and signals from
rior to the retina, may further complicate or interfere with the sources anterior to the retina, i.e., the lens or the cornea, are
detection of the FAF signal. In the eye, the principal barrier is effectively reduced.
the crystalline lens which has highly fluorescent properties in In contrast to the 2° retinal field of the fundus spectrophotom-
the short-wavelength range (excitation between 400 and 600 nm eter, the cSLO allows imaging over larger retinal areas. To reduce
results in peak emission at c. 520 nm). With increasing age and background noise and to enhance image contrast, a series of
particularly the development of nuclear lens opacities, the fluo- several single images is usually recorded (reviewed by Schmitz-
rescence of the lens becomes even more prominent. Valckenberg et al.2). For the final fundus autofluorescence image,
Pioneering work on the spectral analysis of the origin of the a number of these frames (usually out of 4–32) are averaged and
autofluorescence signal was performed by Delori and cowork- pixel values are normalized. Given the high sensitivity of the
ers1 using a fundus spectrometer. In parallel, von Rückmann cSLO and the high frame rate of up to 16 frames per second, FAF
et al., in their landmark paper, described the use of cSLO for FAF imaging can be performed within seconds and at low excitation
imaging.27 energies which are well below the maximum retinal irradiance
limits of lasers established by the American National Standards
Fundus spectrophotometer Institute and other international standards.29
The fundus spectrophotometer by Delori and coworkers1 was With the cSLO, excitation is usually induced in the blue range
designed to analyse systematically the excitation and emission (λ = 488 nm), and an emission filter between 500 and 700 nm is
spectra of the autofluorescence signals originating from small used to detect emission of the autofluorescence signal. The most
retinal areas (2° diameter) of the fundus. By incorporating an widely used cSLO system for FAF imaging is the Heidelberg
image intensifier diode array as a detector, a beam separation in retina angiograph/Heidelberg Spectralis. One key advantage of
the pupil, and confocal detection to minimize contribution of the Spectralis system is the simultaneous acquisition of optical
autofluorescence from the crystalline lens, this device allowed coherence tomography (OCT) recordings that allows for both
the absolute measurements of autofluorescence. These authors averaging of several OCT B-scans in order to enhance the signal-
showed that fundus fluorescence is emitted across a broad band to-noise ratio and the synchronous topographic alignment of
from 500 to 800 nm. Both at the center of the fovea and at 7° FAF intensities with OCT findings.30 Other previous systems,
temporally, optimal excitation occurred at 510 nm with peak such as the Rodenstock cSLO and the Zeiss prototype SM 30 4024
emission at approximately 630 nm, indicating the predominance for FAF imaging, are no longer commercially available. Nidek
of a fluorophore at these excitation and emission spectra. There has recently introduced the F-10 cSLO platform that also allows
was a significant increase with age and the recording along a for FAF imaging (Fig. 4.1).
horizontal line through the fovea showed a minimum fluores-
cence at the fovea, a maximum intensity at 7–15° from the fovea, Fundus camera
and a decrease toward the periphery, most likely reflecting the The relatively weak fundus autofluorescence signal, absorption
concomitant distribution of macular pigment and melanin inter- effects of the crystalline lens, nonconfocality, and light-scattering
fering with the emission of the dominant fluorophore. The optic effects are important limitations of fundus camera-based systems
disc was characterized by a less intense signal. The relationship for FAF recordings. Delori and coworkers described a modified
with age and the topographic distribution of the dominant fundus camera for FAF imaging.31 Their design included the
fundus fluorophore were consistent with those of RPE LF as insertion of an aperture in the illumination optics of the camera
measured in the RPE of human donor eyes.3,5 in order to minimize the loss of contrast caused by light
scattering and fluorescence from the crystalline lens. However, the FAF detection. The visualization of subtle FAF alterations
the modification also resulted in the restriction of the field of is challenging with the modified fundus camera, as shown in
view to a 13° diameter circle; this, together with the complex one study of patients with geographic atrophy (GA) secondary 113
design, is the likely reason why this configuration has not been to AMD.34
further pursued. In 2003, Spaide32 reported the modification of
Wide-field imaging

Chapter 4
a commercially available fundus camera system by shifting the
excitation and emission wavelengths for fundus autofluores- The standard image field of the typical cSLO encompasses a
cence imaging towards the red end of the spectrum in order to retinal field of 30° × 30°. Additional lenses allow for imaging
suppress the fluorescence originating from the lens (Fig. 4.2). of a 55° field or, using the composite mode, imaging over
The relatively inexpensive purchase of an additional filter set, even larger retinal areas. Using the fundus camera, so-called
together with the broad availability of the flash fundus camera, montage images can be manually generated using image
may make thus an attractive alternative. These operate with analysis software on the basis of a seven-field panorama

Autofluorescence Imaging
excitation in the green spectrum and emission is recorded in the survey.
yellow-orange spectrum.33 Peripheral FAF images can also be recorded with a recently
In addition to the different excitation light (green versus introduced wide-field scanning laser ophthalmoscope (P200Tx,
blue) for FAF recording, other major technical differences Optos). This system allows for FAF acquisition in less than 2
between fundus camera systems and the cSLO setup must be seconds by using green light excitation (532  nm). FAF record-
considered (Table 4.1). In particular, the absence of confocal ings beyond the vascular arcades may be particularly helpful
optics makes the fundus camera prone to light scattering and for assessment of the peripheral extension of retinal diseases
generation of secondary reflectance light that interferes with (Fig. 4.3).

Table 4.1 Summary of technical differences between the confocal


scanning laser ophthalmoscope (cSLO) and the modified fundus
camera for fundus autofluorescence imaging

cSLO Modified fundus camera

One excitation wavelength (laser Bandwidth filters for


source) excitation and emission
Large emission spectrum (cutoff
filter)

Continuous scanning at low light One single flash at


intensities in a raster pattern maximum intensities

Confocal system Entire cone of light

Laser power fixed by Flash light intensity,


manufacturer, detector sensitivity gain and gamma of
adjustable detector adjustable

Imaging processing with averaging Manual contrast and


Fig. 4.1 Normal fundus autofluorescence image obtained with a Nidek of single frames and pixel brightness
F-10 scanning laser ophthalmoscope. normalization

cSLO (Heidelberg Engineering) modified FC (Topcon)

400 500 600 700 800 400 500 600 700 800
Wavelength (nm) Wavelength (nm)

SLO (Optos) modified FC (ZeissMediTec)

400 500 600 700 800 400 500 600 700 800
Wavelength (nm) Wavelength (nm)

Fig. 4.2 Range of excitation and emission for different camera systems. cSLO, confocal scanning laser ophthalmoscopy; FC, fundus camera.
INTERPRETATION OF FUNDUS Using pixel gray values, typical ratios between the intensity of
the fovea and perifoveal macula have been established in normal
AUTOFLUORESCENCE IMAGES
114 subjects (reviewed by Schmitz-Valckenberg et al.2). Based on
The FAF image shows the spatial distribution of the intensity these findings, qualitative descriptions of localized FAF changes
of the FAF signal for each pixel in gray values (arbitrary values are widely used. Usually, the FAF signal over a certain retinal
from 0 to 255). Per definition, low pixel values (dark) illustrate location is categorized in decreased, normal, or increased inten-
Section 1

low intensities and high pixel values (bright) high intensities, sities in comparison to the background signal of the same image.
respectively. The topographical distribution of FAF in normal In contrast, the quantification of absolute intensities and their
eyes demonstrates a consistent pattern, as illustrated in Fig. comparison between subjects or within longitudinal observation
4.4.27 A diffuse FAF signal over the posterior pole can be seen, in the same subject are more complicated and remain a challenge
while retinal vessels (due to an absorption phenomenon by in FAF imaging. Of note, as the pixel histogram in the usual
blood contents, i.e., hemoglobin) and the optic nerve head available cSLO images is normalized in order to visualize better
Optical Imaging Technologies
Retinal Imaging and Diagnostics

(absence of autofluorescent material) are characterized by a very the topographic distribution of the FAF intensity (see above), the
low signal and appear dark. Showing a high degree of inter­ pixel values are not absolute and these images must not be used
individual variability, decreased FAF intensities at the macular for absolute intensity analyses from the outset. Furthermore,
area with a minimum in the fovea are observed; these are caused when interpreting FAF images, one should take into account that
by absorption of short-wavelength light due to luteal pigment the digital resolution of the detector in current imaging devices
(lutein and zeaxanthin). exceeds the maximum spatial resolution of ocular media and the
optics of the system, mainly due to high-order aberrations.
Therefore, single pixel values of a standard FAF image do not
reflect the actual anatomical resolution of the image and should
not be used to compare intensities between different locations.
This also explains why increasing the digital resolution of the
detector does not improve the resolution of the actual image, but
rather results in an artificially high-resolution, posterized image.
When analyzing absolute intensities on averaged but non-
normalized FAF images (after ensuring that the normalization
of the pixel histogram is turned off), a great variability of the
mean gray value for a certain retinal location is usually noted
when FAF images are subsequently acquired from the same
subject directly one after the other using the same imaging
device. A systematic analysis by Lois and coworkers35 reported
good intraobserver and moderate interobserver reproducibility
when comparing the absolute mean pixel value of a 16 × 16
pixel square on the retina. In this report, the image resolution
is not provided. When assuming an image resolution of 256 ×
256 pixels and a 40° × 30° field (as these settings were published
in previous studies using the same cSLO by the same group),
Fig. 4.3 Patient with geographic atrophy due to age-related macular the 16 × 16 pixel box would encompass a retinal area of c. 2°
degeneration. The image was recorded by a wide-field scanning laser × 1.9°. Hence, moderate interobserver reproducibility would
ophthalmoscope (P200Tx, Optos). This system allows for fundus
just have been achieved over a rather large retinal area, but
autofluorescence acquisition in less than 2 seconds by using green
light excitation (532 nm). Note the peripheral extension of abnormal was not shown for the anatomical resolution of the imaging
fundus autofluorescence signal nasal to the optic disc. system.

Fig. 4.4 Color fundus photograph (A) and


fundus autofluorescence image (B) of the
right eye of a normal subject imaged with the
confocal scanning laser ophthalmoscope
(Heidelberg retina angiograph, HRA 2,
Heidelberg Engineering, Heidelberg,
Germany). Topographical distribution of
fundus autofluorescence intensity shows
typical background signal with a dark optic
disc (absence of autofluorescent material)
and retinal vessels (absorption). Further,
intensity is markedly decreased over the
fovea due to the absorption of the blue light
by yellow macular pigment. (Reproduced with
permission from Schmitz-Valckenberg S,
Fleckenstein M, Scholl HP, et al. Fundus
autofluorescence and progression of
A B age-related macular degeneration. Surv
Ophthalmol 2009;54:96–117.)
Several confounding factors have to be taken into account Multiple foci and/or irregular areas of FAF are observed when
when comparing absolute FAF intensities between different several small hard or soft drusen coalesce. Focal areas of
examinations and different individuals. This not only includes increased FAF are typically found in the vicinity of drusen 115
standardization of settings (laser power, detector sensitivity, cor- with overlying areas of pigment-clumping or adjacent to long-
rection of refractive errors, and image-processing steps, includ- standing and crystalline drusen.

Chapter 4
ing the number of averaged images), but also eye movements, Increased FAF signal adjacent to drusen fundoscopically cor-
position of the patient in the chin rest, orientation of the camera, responding to focal hyperpigmentation and pigment figures
distance between the camera and the cornea, fluctuations of laser has been attributed to the presence of melanolipofuscin or
power, and short-term dynamic changes in FAF intensities changes in the metabolic activity of the RPE. Areas of hypopig-
caused by prolonged exposure to the excitation light or previous mentation on fundus photographs tend to be associated with a
dark adaption (reviewed by Schmitz-Valckenberg et al.2). corresponding decreased FAF signal, suggesting the absence of
Recently, Delori and coworkers introduced a method for RPE cells or degenerating RPE cells with reduced content of LF

Autofluorescence Imaging
quantitative autofluorescence measurements by insertion of an granules.
internal FAF reference to account for variable laser power and So-called reticular pseudodrusen have been identified as a
detector sensitivity.36 Quantified autofluorescence is calculated risk factor for the development of late-stage AMD. In patients
accounting for the calibrated reference, the zero gray level, and with GA this specific phenotypic pattern, which is best rec-
the magnification (refractive error). For retinal degenerations ognizable by infrared reflectance and FAF imaging, can be
and related diseases, this approach may enhance the under- detected in over 60% of eyes with GA.41 The precise morpho-
standing of disease processes, and may serve as a diagnostic aid, logical correlate of this distinct pattern is controversial. Specu-
as a more sensitive marker of natural disease progression, and lations range from abnormalities in the inner choroid42 to
as a tool to monitor the effects of therapeutic interventions tar- subretinal deposits43; the latter speculation is based on the
geting LF accumulations. spectral domain (SD)-OCT changes recorded in the presence
of reticular pseudodrusen.30,43,44
CLINICAL APPLICATIONS The variability of the FAF phenotye in AMD contrasts with
young patients with monogenic disorders in whom the drusen
Age-related macular degeneration typically autofluoresce brightly, presumably reflecting a compo-
Early AMD sition of the accumulating material distinctly different from
Early manifestation of AMD include focal hypo- and hyperpig- age-related drusen.
mentation at the level of the RPE as well as drusen with extracel- The spectrum of FAF findings in patients with early AMD was
lular material accumulating in the inner aspects of Bruch’s classified by an international expert group.40 Pooling data from
membrane.37 Drusen may be distinguished based on size (small several retinal centers, a system with eight different FAF pat-
versus large) and morphology (hard versus soft). Postmortem terns was developed, including normal, minimal change, focal
analyses demonstrated that some molecular species in drusen increased, patchy, linear, lace-like, reticular, and speckled
material possess autofluorescent properties. pattern (Fig. 4.5). This classification demonstrates the relatively
In vivo FAF changes in early AMD have been described by poor correlation between visible alterations on fundus photog-
several authors using the cSLO and the fundus camera, respec- raphy and notable FAF changes. Based on these results, it was
tively (reviewed by Schmitz-Valckenberg et al.2). Interestingly, speculated that FAF findings in early AMD may indicate more
drusen visible on fundus photography are not necessarily cor- widespread abnormalities and a greater extent of disease than is
related with notable FAF changes and areas of increased FAF ophthalmoscopically visible. The changes seen in FAF imaging
may or may not correspond with areas of hyperpigmentation or at the RPE cell level may precede the occurrence of visible lesions
soft or hard drusen (Fig. 4.5). Overall, larger drusen are more as the disease progresses. This classification system may help to
frequently associated with notable FAF abnormalities than identify specific high-risk characteristics for disease progression
smaller ones, with the exception of basal laminar drusen. Crys- and may be of value in future interventional trials. Furthermore,
talline drusen typically demonstrate a corresponding decreased it may be of use in molecular genetic analysis to identify one or
FAF signal. several genes conferring risk for the development of certain
Delori and coworkers described a pattern of FAF distribu- AMD manifestations.
tion associated with drusen which consists of decreased FAF Recent approaches to investigate FAF findings in AMD
in the center of the druse with a surrounding annulus of patients have included the use of image analysis software to
increased FAF.31 It has been speculated that this appearance compare pixel values and topographically map and register
is caused by attenuated RPE at the center and tangential ori- alterations visible on FAF images with fundus photographs or
entation of RPE cells at the edges of the druse. A reduced reflectance images.32,38 Differences in the percentage of areas
turnover and a net increase in the amount of LF of the RPE with focally increased FAF intensity between eyes with various
cells at the edges would lead to the increased signal. Interest- AMD manifestations have been reported. One study reported
ingly, this ring-like appearance of drusen with FAF imaging that the fellow eyes of patients with unilateral exudative AMD
is much more pronounced when imaged with a flash fundus in the other eye tended not to exhibit FAF abnormalities. Another
camera. Several authors have consistently reported that conflu- analysis showed that patients with exudative AMD in one eye
ent drusen and large foveal soft drusen (drusenoid RPE detach- had larger amounts of areas with abnormal autofluorescence in
ments) topographically correspond well with mildly increased the fellow eye than did eyes of patients with early disease and
FAF using cSLO.38–40 With a fundus camera-based system, large without a history of exudative AMD. Unfortunately, because of
soft drusen have a slightly decreased FAF signal at their centers differences in imaging devices and the use of different image
and are surrounded by a faint ring of increased signal. analysis protocols, comparisons between these studies are
116
Section 1

A B I J
Optical Imaging Technologies
Retinal Imaging and Diagnostics

C D K L

E F M N

G H O P

Fig. 4.5 Classification of abnormal autofluorescence patterns in early age-related macular disease. Corresponding color fundus photographs and
fundus autofluorescence (FAF) images are shown. Eight phenotypic patterns are differentiated: (1) Normal (A, B): homogeneous-background
FAF and a gradual decrease in the inner macula toward the fovea due to the masking effect of macular pigment. Only small hard drusen are
visible in the corresponding fundus photograph. (2) Minimal change (C, D): only minimal variations from normal background FAF. There is limited
irregular increase or decrease in FAF intensity due to multiple small hard drusen. (3) Focal (E, F): several well-defined spots with markedly
increased FAF. Fundus photograph of the same eye with multiple hard and soft drusen. (4) Patchy (G, H): multiple large areas (over 200 µm
in diameter) of increased FAF corresponding to large, soft drusen and/or hyperpigmentation on the fundus photograph. (5) Linear (I, J):
characterized by the presence of at least one linear area of markedly increased FAF. A corresponding hyperpigmented line is visible on the
fundus photograph. (6) Lace-like (K, L): multiple branching linear structures of increased FAF. This pattern may correspond to hyperpigmentation
on the fundus photograph or to no visible abnormalities. (7) Reticular (M, N): multiple, specific small areas of decreased FAF with brighter lines
in between. The reticular pattern not only occurs in the macular area but is found more typically in a superotemporal location. There may be
visible reticular drusen in the corresponding fundus photograph. (8) Speckled (O, P): a variety of FAF abnormalities are noted to occupy a larger
area of the FAF image. There seem to be fewer pathologic areas in the corresponding fundus. (Reproduced with permission from Bindewald A,
Bird AC, Dandekar SS, et al. Classification of fundus autofluorescence patterns in early age-related macular disease. Invest Ophthalmol Vis Sci
2005;46:3309–14.)
difficult and further investigation is required (reviewed by Recently, a classification system of FAF patterns in the junc-
Schmitz-Valckenberg et al.2). tional zone of atrophy in GA patients has been proposed (Fig.
4.9).52 Studies of retinal sensitivity have underscored the impor- 117
Geographic atrophy tance of increased FAF surrounding areas of GA and, thus, the
Areas of GA are associated with RPE cell death as well as with pathophysiological role of increased RPE LF accumulation in

Chapter 4
loss or attenuation of adjacent layers, in particular the outer such patients. Scholl and coworkers have demonstrated that rod
neurosensory retina and the choriocapillaris. 45 With disappear- photoreceptor function is more severely affected than cone func-
ance of the RPE, LF is also lost, resulting in a corresponding tion over areas with increased FAF using fine matrix mapping.53
marked decrease in FAF intensity (Figs 4.6 and 4.7).27 Compared Combining SLO microperimetry and FAF imaging in another
to drusen which may also exhibit a decreased FAF signal, atro- study, impaired photopic sensitivity has been observed in areas
phic areas typically show an even more profound reduction of of abnormal FAF in the junctional zone54.
FAF. 31 The high-contrast difference between atrophic and non- Outer retinal atrophy in the context of AMD is a dynamic

Autofluorescence Imaging
atrophic regions of retina allows more easy and reliable delinea- process with gradual enlargement of atrophic areas over time.
tion of the area of atrophy than from conventional fundus Initial natural history studies on atrophy progression in GA
photographs. 46 These advantages of documenting and studying patients using FAF imaging demonstrated the occurrence of new
GA by FAF imaging have been used in many natural history atrophic patches and the spread of pre-existing atrophy in areas
studies47,48 (Figs 4.6 and 4.7). with abnormally high levels of FAF at baseline.49 Looking at
An even more striking finding of FAF imaging in GA patients larger patient groups with longer review periods, the signifi-
is the frequent presence of areas of hyperautoflourescence in the cance of increased junctional FAF for foreshadowing atrophy
junctional zone surrounding the patch of atrophy.49 Distinct pat- enlargement has been highlighted.47,48 In accordance with other
terns of abnormal FAF in the junctional zone of atrophy and a natural history studies, the FAM (Fundus Autofluorescence
high degree of intraindividual symmetry between fellow eyes Imaging in Age-related Macular Degeneration) study identified
have been described (Fig. 4.8).50,51 a large variability in the rate of atrophy enlargement between

Fig. 4.6 In atrophic age-related macular


degeneration, geographic atrophy appears
as a sharply demarcated area with
depigmentation and enhanced visualization
of deep choroidal vessels on the color fundus
photograph (A). On the corresponding fundus
autofluorescence (FAF) image (B), atrophic
patches are clearly delineated by decreased
intensity and high-contrast to adjacent
nonatrophic retina. Surrounding the atrophy,
in the junctional zone, foci and areas of
increased FAF intensity are observed which
are invisible on fundus photography. These
abnormalities tend to precede atrophy over
time and may serve as disease markers.
(Reproduced with permission from Holz FG,
Spaide RF. Essentials In ophthalmology:
A B Medical retina. Berlin: Springer; 2007,
Fig. 5.3.)

Fig. 4.7 Monitoring of atrophic progression over time with fundus autofluorescence imaging, showing the natural course of the disease over 5
years. (Reproduced with permission from Holz FG, Spaide RF. Essentials In ophthalmology: Medical retina. Berlin: Springer; 2007, Fig. 5.4.)
118
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Fig. 4.8 Fundus autofluorescence images of patients with bilateral geographic atrophy (images of the right and left eye are taken at the same
time point). There is a high degree of symmetry with respect of the configuration of atrophy while there is a high degree of interindividual
variability. (Reproduced with permission from Fleckenstein M, Adrion C, Schmitz-Valckenberg S, et al. Concordance of disease progression in
bilateral geographic atrophy due to AMD. Invest Ophthalmol Vis Sci 2010;51:637–42. Copyright ARVO.org.)

patients, which was neither explained by the extent of baseline which are not always detectable using conventional imaging
atrophy nor by any other comorbid factor such as smoking, lens techniques such as fundus photography, fluorescein, or indo-
status, or family history. Interestingly, the initial studies using cyanine green angiography (Fig. 4.10). The majority of PEDs
FAF imaging on patients with GA have already reported various show a corresponding marked, evenly distributed increase of
patterns of changes in FAF in the junctional zone of GA (reviewed the FAF signal over the lesion surrounded by a well-defined,
by Schmitz-Valckenberg et al.2). These investigators speculated less autofluorescent halo delineating the entire border of the
that their observations might reflect heterogeneity of the under- lesion. There are also PEDs with an intermediate or a decreased
lying disease process. In addition, Bellmann and coworkers FAF signal over the lesion which may or may not correspond
reported a high degree of symmetry of abnormal FAF patterns to areas of RPE atrophy or fibrovascular scarring. Additional
in patients with bilateral GA in the presence of a high degree of investigation, however, is necessary to categorize fully the
interindividual variability, suggesting that genetic determinants FAF features of PEDs and to correlate these findings with
rather than nonspecific aging changes may be involved. 50 other imaging studies. Rarely, a PED shows a cartwheel pattern
A more recent analysis of the FAM study of 195 eyes of 129 of increased autofluorescence corresponding with hyper­
patients shows that variable rates of progression of GA are pigmented radial lines. The hyperpigmented lines correlate
dependent on the specific phenotype of abnormal FAF pattern with subretinal hyperreflective structures, as demonstrated
at baseline.48 Atrophy enlargement was the slowest in eyes with by OCT.
no abnormal FAF pattern (median 0.38 mm2/year), followed by However, a systematic analysis of these FAF alterations with
eyes with the focal FAF pattern (median 0.81 mm2/year), then correlation to the underlying causes of PED such as choroidal
by eyes with the diffuse FAF pattern (median 1.77 mm2/year), neovascularization (CNV), retinal angiomatous proliferation,
and finally, by eyes with the banded FAF pattern (1.81 mm2/ polypoidal vasculopathy, or serous, nonexudative PED is lacking
year). The difference in atrophy progression between the groups to date. These changes are probably not only caused by increased
of no abnormal and focal FAF patterns and the groups of the or decreased amounts of LF, but may derive from other
diffuse and banded FAF patterns was statistically significant dominant fluorophores with similar excitation and emission
(P < 0.0001). These results have subsequently been confirmed in spectra, such as extracellular fluid or degraded photoreceptors
another large-scale study (the Natural History of Geographic (Fig. 4.10).
Atrophy Progression (GAP) study).55,56 These findings under-
Choroidal neovascularization
score the importance of abnormal FAF intensities around atrophy
Theoretical considerations would suggest that FAF imaging may
and the pathophysiological role of increased RPE LF accumula-
provide important clues to our understanding of CNV second-
tion in patients with GA due to AMD.
ary to AMD. For example, it may be helpful to assess the integ-
Pigment epithelium detachment rity of the RPE which may influence the development and
FAF imaging in eyes with pigment epithelium detachment behavior of new vascular complexes as well as photoreceptor
(PED) secondary to AMD show variable FAF phenotypes viability and potential therapeutic success.
Is there any increased
FAF in the junctional YES
zone of atrophy? 119

Chapter 4
NO

Only increased FAF adjacent FAF at the margin


directly to margin of GA and elsewhere (= diffuse)

Autofluorescence Imaging
Single or (Almost)
continuous Laminar, DIFFUSE
individual homogeneous
small spots ring-shaped
around GA

NONE FOCAL BANDED PATCHY

Fig. 4.9 Classification of fundus autofluorescence (FAF) patterns in the junctional zone in patients with geographic atrophy (GA) due to age-
related macular degeneration. Eyes with no apparent increased FAF intensity are graded as ‘‘none’’ (slow progressor). The eyes with increased
FAF are divided into two groups depending on the configuration of increased FAF surrounding atrophy. Eyes showing areas with increased FAF
directly adjacent to the margin of the atrophic patch(es) and elsewhere are called “diffuse” (rapid progressors) and are subdivided into five
groups. From left to right: (top row) fine granular, branching, (bottom row) trickling, reticular, and fine granular with punctuated spots. Eyes with
increased FAF only at the margin of GA are divided into three subtypes (focal (slow progressor), banded (rapid progressor), and patchy (no data,
occurs rarely)) according to their typical FAF pattern around atrophy. (Reproduced with permission from Schmitz-Valckenberg S, Fleckenstein M,
Scholl HP, et al. Fundus autofluorescence and progression of age-related macular degeneration. Surv Ophthalmol 2009;54:96–117.)

Patients with early CNV secondary to AMD tend to have most likely representing gravitational effects of fluid tracking. In
patches of “continuous” or “normal” autofluorescence corre- contrast to fluid, hemorrhages and intraretinal exudates typi-
sponding with areas of hyperfluorescence on the corresponding cally show a decreased FAF signal because of light absorption
fluorescein angiograms, implying that RPE viability is preserved obscuring the underlying retinal details. When retinal hemor-
at least initially in CNV development (Fig. 4.11).57 By contrast, rhages undergo organization and evolve into an ocher color on
eyes with long-standing CNV typically exhibit more areas of fundoscopy, they may become intensely autofluorescent. Later,
decreased FAF signal, which could be explained by photorecep- with disappearance of the yellowish material seen on biomicros-
tor loss and scar formation with increased melanin deposition copy, a large RPE scar and atrophy with decreased autofluores-
(Fig. 4.11). cence may be visible.59 Other imaging studies, such as fundus
One other important finding in eyes with CNV is that abnor- photography, are often required to differentiate between hemor-
mal FAF intensities typically extend beyond the edge of the rhages, exudates, and RPE atrophy.
angiographically defined lesion, indicating a more widespread Comparing FAF findings with the classification of occult and
involvement than is apparent from conventional imaging classic CNV based on fluorescein angiography, Spital et al.59a
studies. Increased FAF signal has also been described around the reported that focal areas of decreased FAF are more prevalent
edge of lesions. It has been speculated that this observation may in classic CNV in comparison to larger occult CNVs. McBain and
reflect the proliferation of RPE cells around the CNV.58 As in associates confirmed this finding and speculated that typical
other exudative retinal disease, such as central serous chorioreti- decreased FAF signals at the site of the CNV are related to
nopathy, areas with increased FAF adjacent to CNV are com- absorption phenomena caused by the CNV growing in the sub-
monly found inferior to the leakage on fluorescein angiography, retinal space, rather than being related to severe damage to the
120
Section 1

A
FAF FL-A ICG-A
Optical Imaging Technologies
Retinal Imaging and Diagnostics

B
FAF FL-A ICG-A

C
FAF FL-A ICG-A

D
FAF FL-A ICG-A

Fig. 4.10 Pigment epithelium detachment classification based on fundus autofluorescence (FAF) characteristics. (A) Increased FAF;
(B) decreased FAF; (C) FAF; and (D) cartwheel FAF. FL-A, fluorescein angiography; ICG-A, indocyanine green angiography. (Reproduced with
permission from Roth F, et al. Fundus autofluorescence imaging of pigment epithelial detachments. ARVO Meet Abstracts 2004;45:2962.)

RPE. 58 These observations would be in accordance with histo- These observations would suggest that the new vessel complex,
pathological studies which have shown that classic CNV mem- regardless of whether classic or occult, was either external
branes are composed of predominantly subretinal fibrovascular (sclerad) to the RPE or had little impact on the attenuation of the
changes as opposed to most occult lesions, which remain exter- FAF signal.
nal to the RPE. However, a more recent study could not demon- Recently, Heimes and coworkers analyzed the prognostic
strate any significant difference in FAF alterations between value of RPE autofluorescence with respect to the therapeutic
occult and classic CNVs secondary to AMD.57 A continuous outcome of anti-vascular endothelial growth factor therapy in
pattern of preserved autofluorescence in the central macula was exudative AMD.60 The analysis of 95 eyes showed a significant
observed in most patients and this was correlated with better difference in visual acuity outcomes in eyes with changes in FAF
visual acuity, shorter symptom length, and smaller lesion size. within the central 500 and 1000 µm.
Fig. 4.11 (A) The right central macula of this
73-year-old woman with a history of blurred
vision for 2 weeks (central visual acuity
20/30) showed fresh hemorrhages, subretinal 121
fluid, and a pigment epithelium detachment.
(C, D) Fluorescein angiography reveals an
active choroidal neovascular membrane with

Chapter 4
leakage in the inferior part of the lesion.
(B) On the autofluorescence image, the
borders (arrows) of the subretinal fluid can
be seen. Of note, the autofluorescence signal
appears to be normal at the site of the active
neovascularization, suggesting that the retinal
pigment epithelium is still viable.
A B (Reproduced with permission from Schmitz-

Autofluorescence Imaging
Valckenberg S, Holz FG, Bird AC, et al.
Fundus autofluorescence imaging: review
and perspectives. Retina 2008;28:385–409.)

C D

In patients with advanced atrophic AMD, the role of sur- The fundoscopically visible pale/yellowish lesions at the level
rounding areas with increased FAF signal with regard to the of RPE/Bruch’s membrane in Best macular dystrophy (Fig. 4.12),
development and progression of exudative AMD remains adult vitelliform macular dystrophy (Fig. 4.13), and other pattern
unclear, and large patient cohorts with longitudinal follow-up dystrophies, as well as Stargardt macular dystrophy/fundus
are still lacking. In a secondary analysis of one study of four flavimaculatus (Fig. 4.14), are associated with an intense focally
eyes,58 no abnormal FAF changes were observed prior to devel- increased FAF signal.
opment of CNV at the site or in the vicinity where the membrane The flecks with increased FAF signal in Stargardt macular
later developed. Looking at 125 eyes with soft drusen and no dystrophy and fundus flavimaculatus may fade over time, with
history of laser treatment, a longitudinal analysis (mean subsequent atrophy development. This finding is in accordance
follow-up 18 months) within the FAM study disclosed nine eyes with histopathological data which have shown that these flecks
which developed advanced exudative AMD during the follow-up represent aggregates of enlarged RPE cells engorged to 10 times
period.61 Six of these nine eyes exhibited the so-called patchy their normal size with LF.
FAF pattern at baseline, characterized by a homogeneous broad As in all forms of macular dystrophies examined systemati-
zone of increased FAF intensity. This finding, if confirmed in cally to date, background autofluorescence in Stargardt macular
other studies, would suggest that the “patchy” FAF pattern in dystrophy appears to be elevated, implying a generalized abnor-
early AMD may represent a high-risk marker for progression to mality of the RPE. This observation confirms the impression
advanced AMD. derived from histological studies that inherited macular dystro-
phies affect the entire RPE.
Macular and diffuse retinal dystrophies Lorenz and coworkers63 described absent or minimal FAF
In macular and diffuse retinal dystrophies, various associated intensities in patients with early-onset severe retinal dystrophy
abnormalities in FAF have been described (reviewed by von associated with mutations on both alleles of RPE65. The lack
Rückmann et al.62). In areas of atrophy, the FAF signal is typi- or severe decrease of FAF signal would be consistent with
cally markedly decreased due to loss of the RPE and conse- the biochemical defect and could be used as a clinical marker
quently a lack of autofluorescent LF. It is well established that of this genotype. Another study demonstrated that patients
autofluorescent material excessively accumulates in the RPE in with Leber congenital amaurosis having vision reduced to
association with various genetically determined retinal diseases. light perception and undetectable electroretinograms (ERGs)
Increased FAF due to excessive LF accumulation in RPE cells may still exhibit normal or minimally decreased FAF intensi-
may result from abnormally high turnover of photoreceptor ties.64 This suggests that the RPE–photoreceptor complex is,
outer segments or impaired RPE lysosomal degradation of at least in part, functionally and anatomically intact. This
normal or altered phagocytosed molecular substrates. finding would have implications for future treatment, sug-
The extent and pattern of increased FAF may show characteris- gesting that photoreceptor function may still be rescuable in
tic abnormal distributions in retinal dystrophy disease entities. such patients.
indicate that these lines in heterogeneous diseases share a
common underlying pathophysiological mechanism.67
122 In RP it could be demonstrated that patients with a larger-
diameter FAF ring also had larger preserved central visual
fields.65 Furthermore, Robson et al. firstly showed a high correla-
tion between the pattern ERG P50 amplitude – a valuable indica-
Section 1

tor of macula function – and the size of the abnormal ring. Later,
they could demonstrate by fine matrix mapping that photopic
sensitivity was preserved over the central macular areas, but
there was a gradient of sensitivity loss over high-density seg-
ments of the ring and severe threshold elevation outside the arc
of the ring. Scotopic sensitivity losses were more severe, and
Optical Imaging Technologies
Retinal Imaging and Diagnostics

they encroached on areas within the ring.65


Popovic et al.68 confirmed by fundus perimetry, among other
methods, that retinal sensitivity was preserved within the ring
and was lost outside the ring, regardless of whether the FAF
pattern was normal or whether atrophic lesions of the RPE were
already present or not. Figure 4.16A shows fundus perimetry in
a patient with autosomal dominant RP.
It has been concluded that the ring of increased FAF in RP
demarcates areas of preserved central photopic function and that
constriction of the ring may mirror progressive visual field loss
by advancing dysfunction that encroaches over areas of central
macular.65
Functional assessment in CRD, CD (cone dystrophy), and
macular dystrophies exhibiting a parafoveal ring of increased
FAF revealed inverted results compared to RP: Robson et al.
could demonstrate that the pattern ERG P50 amplitude was
inversely related to the size of the FAF ring; by fine matrix
mapping they revealed a gradient of sensitivity loss across the
arc of increased FAF.65
By fundus perimetry, in patients with macular dystrophy (Fig.
4.16), it could be demonstrated, that within the ring – indepen-
dent of a normal or abnormal FAF signal – there was severe
retinal dysfunction.67 Outside the ring, the FAF signal and retinal
sensitivity were almost normal.
Despite a different orientation of the line in PPCRA, fundus
perimetry revealed that photoreceptor dysfunction exceeded the
area of RPE cell loss and was precisely delineated (Fig. 4.17). In
the central retina and in the periphery that was not circum-
scribed by the FAF signal line, the FAF signal was normal and
photoreceptor sensitivity was preserved. Within the area out-
lined by increased FAF, independently of whether a normal or
abnormal FAF signal was present, there was severe photorecep-
tor impairment. Despite the variable orientation and morphol-
Fig. 4.12 Best macular dystrophy. Vitelliruptive stage. There is ogy of the line, these observations reflect the findings in RP and
prominent increased fundus autofluorescence (FAF) in the lower part
CD whereby the ring or line of increased FAF represented the
of the original vitelliform lesion that is still demarcated by a faint ring of
increased FAF. (Reproduced with permission from Ho AC, Brown GC, border between functional and dysfunctional retinal sensitivity.
McNamara JA, et al. Color atlas and synopsis of clinical Fleckenstein and coworkers69 first described the SD-OCT cor-
ophthalmology: Retina. New York: McGraw Hill; 2003.) relate of these lines of increased FAF. Specifically, these corre-
sponded with a discrete junctional zone between an area with
Discrete, well-defined lines of increased FAF may occur in preserved OCT layers and an area where the outer aspects of the
various forms of retinal dystrophies.65–67 These lines have no retina are lost and the external limiting membrane band appeared
prominent correlate on fundus biomicroscopy, although there is to rest directly on the RPE (Figs 4.18 and 4.19).
evidence that these lines precisely reflect the border of the More recently, the same SD-OCT correlate has been demon-
regions of retinal dysfunction.65,67,68 Despite the variable orienta- strated in patients with RP.70 This junction might be character-
tion of this line in different entities, e.g., orientation along the ized by progressively altered photoreceptor outer and inner
retinal veins in pigmented paravenous chorioretinal atrophy segments. While the pathophysiological mechanism is unknown,
(PPCRA) or as a ring-like structure in retinitis pigmentosa (RP) it may be hypothesized that the increased FAF signal observed
or macular dystrophies (Fig. 4.15), the similar appearance in various retinal dystrophies might result from an increased
on FAF images and the concordance of functional findings metabolic burden of corresponding RPE cells and subsequent
Fig. 4.13 Adult vitelliform macular dystrophy.
A sharply demarcated area of increased
fundus autofluorescence (FAF) is visible in
the macular area. In the center of the lesion 123
there is reduced FAF. (Reproduced with
permission from Ho AC, Brown GC,
McNamara JA, et al. Color atlas and

Chapter 4
synopsis of clinical ophthalmology: Retina.
New York: McGraw Hill; 2003.)

Autofluorescence Imaging
Fig. 4.14 Stargardt macular dystrophy/fundus
flavimaculatus. Fundoscopically visible focal
flecks show a bright, increased fundus
autofluorescence (FAF) signal. Focal areas
of decreased FAF seem to correspond with
retinal pigment epithelial atrophy.
(Reproduced with permission from Ho AC,
Brown GC, McNamara JA, et al. Color atlas
and synopsis of clinical ophthalmology:
Retina. New York: McGraw Hill; 2003.)

excessive accumulation of fluorophores in the lysosomal com- capillary network and progressive retinal cell death (see
partment due to phagocytosis of components of severely altered Chapter 55, MacTel/parafoveal telangiectasia).71–73 The disease
photoreceptors in such junctional zones. Changes in absorbtion typically manifests temporal to the fovea, and may later
of the FAF signal due to loss of photoreceptor outer segments encompass an oval-shaped area centered on the foveola.
may also contribute to this phenomenon. In the zone with a As outlined above, normal eyes show masking of the foveal
normal FAF signal but impaired retinal sensitivity, the structure 488-nm blue-light FAF due to the accumulation of luteal pigment.
of the photoreceptors seems to be severely distorted. A normal Reduced macular pigment density in MacTel type 2 affects this
FAF signal, therefore, does not necessarily reflect an intact masking. Eyes with MacTel type 2 show an abnormally increased
photoreceptor–RPE complex, but may rather correspond to a signal in the macular area to a variable degree with blue-light
structurally intact-appearing RPE cell monolayer with or without FAF imaging (Figs 4.20 and 4.21). 74 A loss of luteal pigment may
the presence of intact photoreceptors. initially occur in the area temporal to the foveal center (Fig.
4.22).74,75 Quantitative analysis confirmed that the loss of luteal
Macular telangiectasia pigment was more pronounced in the temporal compared with
Macular telangiectasia (MacTel) type 2 is a bilateral disease of the nasal parafoveolar area and suggested that zeaxanthin would
unknown cause with characteristic alterations of the macular be more reduced than lutein.75
124
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

C D

Fig. 4.15 Arcs and rings of increased fundus autofluorescence (FAF) in patients with various forms of retinal dystrophies. Patients 1–3 (A–C),
diagnosed with pigmented paravenous chorioretinal atrophy, demonstrates an arc of increased FAF orienting along the retinal veins. There is a
normal FAF signal between this arc and the atrophic areas (i.e., decreased FAF) and in the area that is not circumscribed by the line. In the left
eye of patient 1 (A) and the right eye of patient 3 (C), the arc of increased FAF almost merges to a parafoveal ring with a temporal opening. In
patient 4 (D) with sector retinitis pigmentosa, there is an arc with a semicircular configuration in the parafoveal region.
125

Chapter 4
Autofluorescence Imaging
E F

Fig. 4.15 Cont’d In typical retinitis pigmentosa (patient 5, E), there is a ring of increased FAF. Within and outside the ring, there is a normal
FAF signal. In patient 6 (F), who is diagnosed with macular dystrophy, there is a parafoveal ring of increased FAF. Centrally, there is reduced
FAF corresponding to the fundoscopically visible lesion. On both sides of the ring, there is a normal FAF signal. In patient 7 (G) with another
bull’s-eye macular dystrophy, a ring of increased FAF directly borders the central lesion. In the very center, there is a spot of preserved FAF.
Note that there is no significant correlate of the arc of increased FAF in the conventional color fundus photograph. (Reproduced with permission
from Fleckenstein M, Charbel Issa P, Fuchs HA, et al. Discrete arcs of increased fundus autofluorescence in retinal dystrophies and functional
correlate on microperimetry. Eye (Lond) 2009;23:567–75.)
126

A B C
Section 1

Attenuation scale (dB)


0 2 4 6 8 10 12 14 16 18 20

Fig. 4.16 Fundus-controlled microperimetric assessment of rings of increased fundus autofluorescence (FAF). The sensitivity map is
superimposed on the FAF image. Light increment sensitivity (LIS) varies from 0 to 20 dB (attenuation scale). Hollow red squares indicate testing
Optical Imaging Technologies

points where the brightest stimulus was not seen. (A) In the patient with autosomal dominant retinitis pigmentosa, LIS is preserved within the
Retinal Imaging and Diagnostics

ring; outside, there is severely impaired LIS despite a normal FAF signal. In patients with macular dystrophy exhibiting a ring of increased FAF
(B, C), LIS is significantly impaired within the ring independently of a normal or decreased FAF signal; outside, LIS is preserved. These findings
are the inverse of the findings in patients diagnosed with retinitis pigmentosa (A). (Reproduced with permission from Fleckenstein M, Charbel
Issa P, Fuchs HA, et al. Discrete arcs of increased fundus autofluorescence in retinal dystrophies and functional correlate on microperimetry.
Eye (Lond) 2009;23:567–75.)

A B

Attenuation scale (dB)


0 2 4 6 8 10 12 14 16 18 20

Fig. 4.17 Fundus-controlled microperimetric assessment in pigmented paravenous chorioretinal atrophy. The sensitivity map is superimposed on
the fundus autofluorescence (FAF) image. Light increment sensitivity (LIS) varies from 0 to 20 dB (attenuation scale). Hollow red squares
indicate testing points where the brightest stimulus was not seen. In the area that is framed by the arc of increased FAF, LIS is severely
impaired independently of a normal or abnormal FAF signal. The arc of increased FAF demarcates the area of preserved LIS. (Reproduced with
permission from Fleckenstein M, Charbel Issa P, Fuchs HA, et al. Discrete arcs of increased fundus autofluorescence in retinal dystrophies and
functional correlate on microperimetry. Eye (Lond) 2009;23:567–75.)

RNFL
GCL
IPL
INL
OPL
ONL
ELM*
IPRL*
RPE*

Fig. 4.18 Simultaneous fundus autofluorescence (FAF) and spectral domain-optical coherence tomography imaging. The line of increased FAF
corresponds to a junctional zone (within black lines) between involved and preserved retina. RNFL, retinal nerve fiber layer; GCL, ganglion cell
layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; ELM*, presumed correspondence
of the external limiting membrane; IPRL*, presumed correspondence of the interface of the inner/outer segments of photoreceptors; RPE*,
presumed correspondence of the retinal pigment epithelium. (Reproduced with permission from Fleckenstein M, Charbel Issa P, Helb HM, et al.
Correlation of lines of increased autofluorescence in macular dystrophy and pigmented paravenous retinochoroidal atrophy by optical coherence
tomography. Arch Ophthalmol 2008;126:1461–3.)
127

Chapter 4
RNFL
GCL
IPL
INL
OPL
ONL
ELM*

Autofluorescence Imaging
IPRL*
RPE*

Fig. 4.19 Simultaneous fundus autofluorescence (FAF) and spectral domain optical coherence tomography imaging. The line of increased FAF
corresponds to a junctional zone (within black lines) between preserved and involved retina. RNFL, retinal nerve fiber layer; GCL, ganglion cell
layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; ELM*, presumed correspondence
of the external limiting membrane; IPRL*, presumed correspondence of the interface of the inner/outer segments of photoreceptors; RPE*,
presumed correspondence of the retinal pigment epithelium. (Reproduced with permission from Fleckenstein M, Charbel Issa P, Helb HM, et al.
Correlation of lines of increased autofluorescence in macular dystrophy and pigmented paravenous retinochoroidal atrophy by optical coherence
tomography. Arch Ophthalmol 2008;126:1461–3.)

Fig. 4.20 Fundus autofluorescence image


obtained at 488 nm (excitation) of a normal
eye (left) and of a 59-year-old woman with
type 2 idiopathic macular telangiectasis
(right). (Reproduced with permission from
Helb HM, Charbel Issa P, van der Veen RLP,
et al. Macular pigment density and
distribution in patients with type II macular
telangiectasia. Retina 2008;28:808–16.)

Fig. 4.21 Fluorescein angiogram frame (left)


and fundus autofluorescence (FAF) image
obtained at 488 nm (right) of a patient with
type 2 idiopathic macular telangiectasia
showing an abnormal FAF distribution in the
macular area due to depletion of luteal
pigment. (Reproduced with permission from
Helb HM, Charbel Issa P, van der Veen RLP,
et al. Macular pigment density and
distribution in patients with type II macular
telangiectasia. Retina 2008;28:808–16.)
Fig. 4.22 Macular pigment optical density
(MPOD) maps obtained by digital subtraction
of log fundus reflectance maps (left column)
128 compared with corresponding fundus
autofluorescence maps (right column).
The independent measurement techniques
demonstrated the same rings of MPOD
Section 1

at 5–7° eccentricity. (Reproduced with


permission from Helb HM, Charbel Issa P,
van der Veen RLP, et al. Macular pigment
density and distribution in patients with type
II macular telangiectasia. Retina
2008;28:808–16.)
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Further proof for the depletion in macular pigment derived classes of macular pigment loss with the consecutive disease
from a postmortem analysis of an eye with MacTel type 2. Mac- stages of MacTel type 2 described by Gass and Blodi.71 Correla-
roscopic examination disclosed the absence of the central yel- tion studies with microperimetric data revealed a trend towards
lowish spot.76 A yellow ring of residual macular pigment was worse retinal function with increasing class of macular pigment
present eccentrically in accordance with the in vivo imaging changes.
observations. The loss of macular pigment was subsequently
divided into three classes based on a cross-sectional analysis of Pseudoxanthoma elasticum
two-wavelength (blue-light and green-light) FAF images77: Class Pseudoxanthoma elasticum (PXE) is caused by a mutation in the
1 shows a wedge-shaped loss of macular pigment restricted to ABCC6 gene. More than 300 distinct loss-of-function mutations
an area temporal to the foveal center. In class 2, the area is larger representative of over 1000 mutant alleles in ABCC6 have been
and also involves the foveal center. Class 3 is characterized by found. Many of the missense mutations occur at locations in the
loss of luteal pigment within an oval-shaped area centered on protein involving domain–domain interactions in the ABCC6
the foveola. There was a significant association of these three transporter. Even heterozygotes can show manifestations of
disease. FAF abnormalities are common in eyes affected by PXE. more chronic leaks can have decreased autofluorescence sur-
Typical phenotypic alterations, including angioid streak, and rounding the known leaks. This area of hypoautofluorescence
drusen of the optic nerve, have autofluorescence correlates. Peau appears to expand in size with increasing chronicity of the leak. 129
d’orange is hardly detectable on FAF, whereas comet-tail lesions Patients who were known to have a history of chronic central
are typically apparent. RPE atrophy can be widespread and serous chorioretinopathy that had been inactive for several years

Chapter 4
heterogeneous, located mostly adjacent to angioid streaks are left with hypoautofluorescent areas and no hyperautofluo-
or CNV.78,79 rescent regions.
Furthermore, irregular patterns of increased FAF at the pos­
terior pole with an appearance similar to that of pattern dystro- CHLOROQUINE AND
phies can be found in eyes with PXE. In these eyes, areas of HYDROXYCHLOROQUINE RETINOPATHY
yellowish deposits and hyperpigmentation on color photogra-
FAF imaging may show distinct alterations due to toxic retinal
phy corresponded to areas of increased FAF (Fig. 4.23). Agarwal

Autofluorescence Imaging
effects of long-term chloroquine and hydroxychloroquine
et al.80 suggested the following classification: a fundus appear-
therapy.84 Various methods have been proposed to detect early
ance similar to a pattern dystrophy of the fundus flavimaculatus,
stages of chloroquine retinopathy. Early on, a pericentral ring of
the reticular, the vitelliform and the fundus pulverulentus types,
increased FAF intensity may occur associated with pericentral
respectively. The pattern dystrophy-like changes in PXE may
reduction in multifocal ERG amplitudes and pericentral inter-
occur unilaterally or bilaterally.
ruption of the photoreceptor inner-segment–outer-segment
Abnormalities of the RPE–photoreceptor complex detected by
junction on SD-OCT imaging.85 More advanced stages are associ-
FAF imaging are more diverse and widespread than expected
ated with a more mottled appearance with increased and
from conventional fundus imaging. Such extensive alteration of
decreased FAF intensity in the pericentral macula (Fig. 4.25).
the RPE suggests an important role of pathological RPE changes
While electrophysiological examination has been thought to
in the evolution of visual loss in PXE.
represent an adequate tool to diagnose early chloroquine macu-
Recently, the abnormalities detected by multimodal imaging
lopathy, FAF imaging can be used as a highly sensitive tool.
suggest a centrifugal spread of the retinal pathologic features of
the Bruch’s membrane–RPE complex in PXE. These results FUNCTIONAL CORRELATES OF
suggest that three different areas centered on the posterior pole
can be defined in the fundus of patients with PXE. These areas
FAF ABNORMALITIES
are separated by two consecutive transition zones. The central The relevance of alterations in FAF images can further be
two areas may indicate different stages of pathologic fundus addressed by assessing corresponding retinal sensitivity. Severe
alterations.81 damage to the RPE such as atrophy, melanin pigment migra-
tion, or fibrosis leading to compromised photoreceptor function
Central serous chorioretinopathy as confirmed by microperimetry to correspond topographically
Central serous chorioretinopathy is a condition characterized by to areas of decreased autofluorescence.53,54 In patients with
idiopathic leaks at the level of the RPE leading to serous pigment GA secondary to AMD, it has been shown that, in addition
epithelial and neurosensory retinal detachments. In the early to absence of retinal sensitivity over atrophic areas, retinal
phases of the disease, the visual acuity may be good despite the function is relatively and significantly reduced over areas with
presence of the macular detachment, and after resolution the increased FAF intensities compared to areas with normal
acuity often shows improvement. More chronic forms of central background signal.54 Localized functional impairment over
serous chorioretinopathy are associated with atrophic and areas with increased FAF has also been recently confirmed
degenerative changes of the retina and RPE and consequently in patients with early AMD. Using fine matrix mapping, it
with visual acuity decline.82 has been demonstrated that, interestingly, rod function is more
Accordingly, FAF findings in central serous chorioretinopathy severely affected than cone function over areas with increased
are dependent on the extent of involvement of the RPE and the FAF in patients with AMD.53 These studies are in accordance
stage of the disease.83 Patients with acute leaks imaged within with the observation of increased accumulation of autofluo-
the first month have minimal abnormalities other than a slight rescent material at the level of the RPE prior to the occurrence
increase in autofluorescence in the area of the serous retinal of cell death. As normal photoreceptor function is dependent
detachment. Over time, the area of the detachment increasingly on normal RPE function, in particular with regard to the
exhibits more irregular increased autofluorescence. In some constant phagocytosis of shed distal outer-segment stacks for
patients, discrete granules with increased intensity within the photoreceptor cell renewal, a negative-feedback mechanism
detachment are observed which correspond with the pinpoint has been proposed, whereby cells with LF-loaded secondary
subretinal precipitates seen on fundoscopy. It has been sug- lysosomes would less efficiently phagocytose newly shed pho-
gested that these dots may represent macrophages engorged toreceptor outer segments, subsequently leading to impaired
with phagocytosed outer segments. Patients with chronic disease retinal sensitivity. This would also be in line with experimental
have irregular levels of autofluorescence with markedly data showing that compounds of LF such as A2-E possess
decreased intensity over areas of atrophy. A typical finding also toxic properties and may interfere with normal RPE cell
includes the visualization of fluid tracks in the inferior retina function.10
(Fig. 4.24). The area of the leak also undergoes change in auto- In patients with different retinal dystrophies, lines and rings
fluorescence over time. Soon after the development of central of increased FAF have been noted (see above).65 Interestingly,
serous chorioretinopathy, little or no change in the autofluores- functional testing by microperimetry and electrophysiology
cence pattern in the area around the leak is seen, although the indicates that these rings circumscribe areas of preserved pho-
leak site may be somewhat hypoautofluorescent. Patients with toreceptor function (Fig. 4.16A).65,67,68
130
Section 1

A B C
Optical Imaging Technologies
Retinal Imaging and Diagnostics

D E F

G H I

J K L

Fig. 4.23 Various types of retinal pigment epithelium degenerative patterns of the macula observed in pseudoxanthoma elasticum (first column:
fundus autofluorescence; second column: fluorescein angiography; third column: color fundus photograph). (A–C) Fundus changes resembling
reticular dystrophy; (D–F) fundus changes resembling fundus flavimaculatus; (G–I) fundus changes resembling vitelliform dystrophy; (J–L) fundus
changes resembling fundus pulverulentus. (Reproduced with permission from Finger RP, Charbel Issa P, Ladewig M, et al. Fundus
autofluorescence in pseudoxanthoma elasticum. Retina 2009;29:1496–505.)
REFERENCES
1. Delori FC, Dorey CK, Staurenghi G, et al. In vivo fluorescence of the
ocular fundus exhibits retinal pigment epithelium lipofuscin characteristics.
Invest Ophthalmol Vis Sci 1995;36:718–29. 131
2. Schmitz-Valckenberg S, Fleckenstein M, Scholl HP, et al. Fundus autofluores-
cence and progression of age-related macular degeneration. Surv Ophthalmol
2009;54:96–117.

Chapter 4
3. Weiter JJ, Delori FC, Wing GL, et al. Retinal pigment epithelial lipofuscin and
melanin and choroidal melanin in human eyes. Invest Ophthalmol Vis Sci
1986;27:145–52.
4. Feeney-Burns L, Berman ER, Rothmann H. Lipofuscin of human retinal
pigment epithelium. Am J Ophthalmol 1980;90:783–91.
5. Wing GL, Blanchard GC, Weiter JJ. The topography and age relationship of
lipofuscin concentration in the retinal pigment epithelium. Invest Ophthalmol
Vis Sci 1978;17:601–7.
6. Sparrow JR, Boulton M. RPE lipofuscin and its role in retinal pathobiology.

Autofluorescence Imaging
Exp Eye Res 2005;80:595–606.
7. Eldred GE, Lasky MR. Retinal age-pigments generated by self-assembling
lysosomotrophic detergents. Nature 1993;361:724–6.
8. Dorey CK, Wu G, Ebenstein D, et al. Cell loss in the aging retina. Relationship
to lipofuscin accumulation and macular degeneration. Invest Ophthalmol Vis
Sci 1989;30:1691–9.
9. Schütt F, Davies S, Kopitz J, et al. Photodamage to human RPE cells by A2-E,
a retinoid component of lipofuscin. Invest Ophthalmol Vis Sci 2000;41:
2303–8.
10. Bergmann M, Schutt F, Holz FG, et al. Inhibition of the ATP-driven proton
pump in RPE lysosomes by the major lipofuscin fluorophore A2-E may con-
tribute to the pathogenesis of age-related macular degeneration. FASEB J
2004;18:562–4.
11. Brunk UT, Wihlmark U, Wrigstad A, et al. Accumulation of lipofuscin
within retinal pigment epithelial cells results in enhanced sensitivity to
photo-oxidation. Gerontology 1995;41(Suppl 2):201–12.
12. Hammer M, Richter S, Guehrs KH, et al. Retinal pigment epithelium cell
damage by A2-E and its photo-derivatives. Mol Vis 2006;12:1348–54.
13. Liu J, Itagaki Y, Ben-Shabat S, et al. The biosynthesis of A2E, a fluorophore of
aging retina, involves the formation of the precursor, A2-PE, in the photorecep-
tor outer segment membrane. J Biol Chem 2000;275:29354–60.
14. Eldred GE, Katz ML. Fluorophores of the human retinal pigment epithelium:
separation and spectral characterization. Exp Eye Res 1988;47:71–86.
15. Zhou J, Jang YP, Kim SR, et al. Complement activation by photooxidation
Fig. 4.24 Chronic central serous retinopathy as imaged by confocal products of A2E, a lipofuscin constituent of the retinal pigment epithelium.
scanning laser ophthalmoscopy shows decreased fundus Proc Natl Acad Sci U S A 2006;103:16182–7.
autofluorescence (FAF) in the macula due to atrophy with surrounding 16. Lamb LE, Simon JD. A2E: a component of ocular lipofuscin. Photochem
Photobiol 2004;79:127–36.
increased FAF. Additional FAF abnormalities outside the central 17. Marmorstein AD, Marmorstein LY, Sakaguchi H, et al. Spectral profiling of
macula, including prominent descending tracts or gutters, are also autofluorescence associated with lipofuscin, Bruch’s membrane, and sub-RPE
observed. deposits in normal and AMD eyes. Invest Ophthalmol Vis Sci 2002;43:
2435–41.
18. Keilhauer CN, Delori FC. Near-infrared autofluorescence imaging of the
fundus: visualization of ocular melanin. Invest Ophthalmol Vis Sci 2006;
47:3556–64.
19. Weinberger AW, Lappas A, Kirschkamp T, et al. Fundus near infrared fluores-
cence correlates with fundus near infrared reflectance. Invest Ophthalmol Vis
Sci 2006;47:3098–108.
20. Kellner U, Kellner S, Weinitz S. Fundus autofluorescence (488 nm) and near-
infrared autofluorescence (787 nm) visualize different retinal pigment epithe-
lium alterations in patients with age-related macular degeneration. Retina
2010;30:6–15.
21. Gibbs D, Cideciyan AV, Jacobson SG, et al. Retinal pigment epithelium defects
in humans and mice with mutations in MYO7A: imaging melanosome-specific
autofluorescence. Invest Ophthalmol Vis Sci 2009;50:4386–93.
22. Schmitz-Valckenberg S, Lara D, Nizari S, et al. Localisation and significance of
in vivo near-infrared autofluorescent signal in retinal imaging. Br J Ophthalmol
2011;95:1134–9.
23. Snodderly DM, Brown PK, Delori FC, et al. The macular pigment. I. Absor-
bance spectra, localization, and discrimination from other yellow pigments in
primate retinas. Invest Ophthalmol Vis Sci 1984;25:660–73.
24. Whitehead AJ, Mares JA, Danis RP. Macular pigment: a review of current
knowledge. Arch Ophthalmol 2006;124:1038–45.
25. Davies NP, Morland AB. Macular pigments: their characteristics and putative
role. Prog Retin Eye Res 2004;23:533–59.
26. Wolf S, Wolf-Schnurrbusch U. Macular pigment measurement – theoretical
background. In: Holz FG, Schmitz-Valckenberg S, Spaide RF, et al. editors.
Atlas of autofluorescence imaging. Berlin: Springer; 2007.
27. von Rückmann A, Fitzke FW, Bird AC. Distribution of fundus autofluorescence
with a scanning laser ophthalmoscope. Br J Ophthalmol 1995;79:407–12.
28. Webb RH, Hughes GW, Delori FC. Confocal scanning laser ophthalmoscope.
Appl Optics 1987;26:1492–9.
29. American National Standard for the Safe Use of Lasers Z136.1; 1993.
30. Fleckenstein M, Charbel Issa P, Helb HM, et al. High-resolution spectral
domain-OCT imaging in geographic atrophy associated with age-related
macular degeneration. Invest Ophthalmol Vis Sci 2008;49:4137–44.
31. Delori FC, Fleckner MR, Goger DG, et al. Autofluorescence distribution associ-
ated with drusen in age-related macular degeneration. Invest Ophthalmol Vis
Fig. 4.25 Advanced stage of chloroquine retinopathy. There is Sci 2000;41:496–504.
a mottled appearance with increased and decreased fundus 32. Spaide RF. Fundus autofluorescence and age-related macular degeneration.
autofluorescence intensity in the pericentral macula. Ophthalmology 2003;110:392–9.
33. Spaide RF. Autofluorescence Imaging with the fundus camera. In: Holz FG, 58. McBain VA, Townend J, Lois N. Fundus autofluorescence in exudative
Schmitz-Valckenberg S, Spaide RF, et al, editors. Atlas of autofluorescence age-related macular degeneration. Br J Ophthalmol 2007;91:491–6.
imaging. Berlin: Springer; 2007. p. 49–53. 59. Sawa M, Ober MD, Spaide RF. Autofluorescence and retinal pigment epithelial
132 34. Schmitz-Valckenberg S, Fleckenstein M, Gobel AP, et al. Evaluation of atrophy after subretinal hemorrhage. Retina 2006;26:119–20.
autofluorescence imaging with the scanning laser ophthalmoscope and the 59a.  Spital G, Redermacher M, Müller C, et al. [Autofluorescence characteristics of
fundus camera in age-related geographic atrophy. Am J Ophthalmol 2008;146: lipofuscin components in different forms of late senile macular degeneration.]
183–92. Klin Monabl Augenheilkd 1998;213:23–31.
35. Lois N, Halfyard AS, Bunce C, et al. Reproducibility of fundus autofluorescence 60. Heimes B, Lommatzsch A, Zeimer M, et al. Foveal RPE autofluorescence as a
Section 1

measurements obtained using a confocal scanning laser ophthalmoscope. prognostic factor for anti-VEGF therapy in exudative AMD. Graefes Arch Clin
Br J Ophthalmol 1999;83:276–9. Exp Ophthalmol 2008;246:1229–34.
36. Delori F, Greenberg JP, Woods RL, et al. Quantitative measurements of auto- 61. Einbock W, Moessner A, Schnurrbusch UE, et al. Changes in fundus autofluo-
fluorescence with the scanning laser ophthalmoscope. Invest Ophthalmol Vis rescence in patients with age-related maculopathy. Correlation to visual func-
Sci 2011;52:9379–90. tion: a prospective study. Graefes Arch Clin Exp Ophthalmol 2005;243:300–5.
37. Bird A. Age-related macular disease. Br J Ophthalmol 1996;80:2–3. 62. von Rückmann A, Fitzke F, Schmitz-Valckenberg S, et al. Macular and retinal
38. Smith RT, Chan JK, Busuoic M, et al. Autofluorescence characteristics of dystrophies. In: Holz FG, Schmitz-Valckenberg S, Spaide R, et al, editors. Atlas
early, atrophic, and high-risk fellow eyes in age-related macular degeneration. of fundus and fluorescence imaging. Berlin: Springer; 2007.
Optical Imaging Technologies

Invest Ophthalmol Vis Sci 2006;47:5495–504. 63. Lorenz B, Wabbels B, Wegscheider E, et al. Lack of fundus autofluorescence to
Retinal Imaging and Diagnostics

39. Lois N, Owens SL, Coco R, et al. Fundus autofluorescence in patients 488 nanometers from childhood on in patients with early-onset severe retinal
with age-related macular degeneration and high risk of visual loss. Am J dystrophy associated with mutations in RPE65. Ophthalmology 2004;111:
Ophthalmol 2002;133:341–9. 1585–94.
40. Bindewald A, Bird AC, Dandekar SS, et al. Classification of fundus autofluo- 64. Scholl HP, Chong NH, Robson AG, et al. Fundus autofluorescence in patients
rescence patterns in early age-related macular disease. Invest Ophthalmol Vis with leber congenital amaurosis. Invest Ophthalmol Vis Sci 2004;45:2747–52.
Sci 2005;46:3309–14. 65. Robson AG, Michaelides M, Saihan Z, et al. Functional characteristics of
41. Schmitz-Valckenberg S, Alten F, Steinberg JS, et al. Reticular drusen patients with retinal dystrophy that manifest abnormal parafoveal annuli of
associated with geographic atrophy in age-related macular degeneration. high density fundus autofluorescence; a review and update. Doc Ophthalmol
Invest Ophthalmol Vis Sci 2011;52:5009–15. 2008;116:79–89.
42. Arnold JJ, Sarks SH, Killingsworth MC, et al. Reticular pseudodrusen. A risk 66. von Ruckmann A, Fitzke FW, Bird AC. In vivo fundus autofluorescence in
factor in age-related maculopathy. Retina 1995;15:183–91. macular dystrophies. Arch Ophthalmol 1997;115:609–15.
43. Zweifel SA, Spaide RF, Curcio CA, et al. Reticular pseudodrusen are subretinal 67. Fleckenstein M, Charbel Issa P, Fuchs HA, et al. Discrete arcs of increased
drusenoid deposits. Ophthalmology 2010;117:303–12 e1. fundus autofluorescence in retinal dystrophies and functional correlate on
44. Schmitz-Valckenberg S, Steinberg JS, Fleckenstein M, et al. Combined confocal microperimetry. Eye (Lond) 2009;23:567–75.
scanning laser ophthalmoscopy and spectral-domain optical coherence 68. Popovic P, Jarc-Vidmar M, Hawlina M. Abnormal fundus autofluorescence in
tomography imaging of reticular drusen associated with age-related macular relation to retinal function in patients with retinitis pigmentosa. Graefes Arch
degeneration. Ophthalmology 2010;117:1169–76. Clin Exp Ophthalmol 2005;243:1018–27.
45. Sarks SH. Ageing and degeneration in the macular region: a clinico-pathological 69. Fleckenstein M, Charbel Issa P, Helb HM, et al. Correlation of lines of increased
study. Br J Ophthalmol 1976;60:324–41. autofluorescence in macular dystrophy and pigmented paravenous retinocho-
46. Schmitz-Valckenberg S, Jorzik J, Unnebrink K, et al. Analysis of digital scan- roidal atrophy by optical coherence tomography. Arch Ophthalmol 2008;
ning laser ophthalmoscopy fundus autofluorescence images of geographic 126:1461–3.
atrophy in advanced age-related macular degeneration. Graefes Arch Clin Exp 70. Lima LH, Cella W, Greenstein VC, et al. Structural assessment of hyperauto-
Ophthalmol 2002;240:73–8. fluorescent ring in patients with retinitis pigmentosa. Retina 2009;29:1025–31.
47. Schmitz-Valckenberg S, Bindewald-Wittich A, Dolar-Szczasny J, et al. Correla- 71. Gass JD, Blodi BA. Idiopathic juxtafoveolar retinal telangiectasis. Update of
tion between the area of increased autofluorescence surrounding geographic classification and follow-up study. Ophthalmology 1993;100:1536–46.
atrophy and disease progression in patients with AMD. Invest Ophthalmol Vis 72. Hutton WL, Snyder WB, Fuller D, et al. Focal parafoveal retinal telangiectasis.
Sci 2006;47:2648–54. Arch Ophthalmol 1978;96:1362–7.
48. Holz FG, Bindewald-Wittich A, Fleckenstein M, et al. Progression of geo- 73. Gass JDM. Stereoscopic atlas of macular diseases: diagnosis and treatment.
graphic atrophy and impact of fundus autofluorescence patterns in age-related 2nd ed. St Louis: Mosby; 1977.
macular degeneration. Am J Ophthalmol 2007;143:463–72. 74. Helb HM, Charbel Issa P, van der Veen RL, et al. Abnormal macular pigment
49. Holz FG, Bellman C, Staudt S, et al. Fundus autofluorescence and development distribution in type 2 idiopathic macular telangiectasia. Retina 2008;28:808–16.
of geographic atrophy in age-related macular degeneration. Invest Ophthalmol 75. Charbel Issa P, van der Veen RL, Stijfs A, et al. Quantification of reduced
Vis Sci 2001;42:1051–6. macular pigment optical density in the central retina in macular telangiectasia
50. Bellmann C, Jorzik J, Spital G, et al. Symmetry of bilateral lesions in geographic type 2. Exp Eye Res 2009;89:25–31.
atrophy in patients with age-related macular degeneration. Arch Ophthalmol 76. Powner MB, Gillies MC, Tretiach M, et al. Perifoveal muller cell depletion in a
2002;120:579–84. case of macular telangiectasia type 2. Ophthalmology 2010;117:2407–16.
51. Holz FG, Bellmann C, Margaritidis M, et al. Patterns of increased in vivo 77. Zeimer MB, Padge B, Heimes B, et al. Idiopathic macular telangiectasia type 2:
fundus autofluorescence in the junctional zone of geographic atrophy of the distribution of macular pigment and functional investigations. Retina
retinal pigment epithelium associated with age-related macular degeneration. 2010;30:586–95.
Graefes Arch Clin Exp Ophthalmol 1999;237:145–52. 78. Charbel Issa P, Finger RP, Holz FG, et al. Multimodal imaging including spec-
52. Bindewald A, Schmitz-Valckenberg S, Jorzik JJ, et al. Classification of abnormal tral domain OCT and confocal near infrared reflectance for characterisation of
fundus autofluorescence patterns in the junctional zone of geographic atrophy outer retinal pathology in pseudoxanthoma elasticum. Invest Ophthalmol Vis
in patients with age related macular degeneration. Br J Ophthalmol 2005;89: Sci 2009;50:5913–8.
874–8. 79. Finger RP, Charbel Issa P, Ladewig M, et al. Fundus autofluorescence in
53. Scholl HP, Bellmann C, Dandekar SS, et al. Photopic and scotopic fine matrix pseudoxanthoma elasticum. Retina 2009;29:1496–505.
mapping of retinal areas of increased fundus autofluorescence in patients with 80. Agarwal A, Patel P, Adkins T, et al. Spectrum of pattern dystrophy in
age-related maculopathy. Invest Ophthalmol Vis Sci 2004;45:574–83. pseudoxanthoma elasticum. Arch Ophthalmol 2005;123:923–8.
54. Schmitz-Valckenberg S, Bultmann S, Dreyhaupt J, et al. Fundus autofluores- 81. Charbel Issa P, Finger RP, Gotting C, et al. Centrifugal fundus abnormalities
cence and fundus perimetry in the junctional zone of geographic atrophy in in pseudoxanthoma elasticum. Ophthalmology 2010;117:1406–14.
patients with age-related macular degeneration. Invest Ophthalmol Vis Sci 82. Spaide RF, Campeas L, Haas A, et al. Central serous chorioretinopathy in
2004;45:4470–6. younger and older adults. Ophthalmology 1996;103:2070–9; discussion 9-80.
55. Schmitz-Valckenberg S, Jaffe GJ, Fleckenstein M, et al. and GAP Study Group. 83. Spaide RF, Klancnik JM, Jr. Fundus autofluorescence and central serous
Lesion characteristics and progression in the Natural History of Geographic chorioretinopathy. Ophthalmology 2005;112:825–33.
Atrophy (GAP) Study. ARVO Meeting Abstracts 2009;50:3914. 84. Kellner U, Renner AB, Tillack H. Fundus autofluorescence and mfERG for
56. Holz FG, Schmitz-Valckenberg S, Fleckenstein M, et al. Lesion characteristics early detection of retinal alterations in patients using chloroquine/
and progression in the Natural History of Geographic Atrophy (GAP) Study. hydroxychloroquine. Invest Ophthalmol Vis Sci 2006;47:3531–8.
ARVO Meeting Abstracts 2010;51:94. 85. Kellner S, Weinitz S, Kellner U. Spectral domain optical coherence tomography
57. Vaclavik V, Vujosevic S, Dandekar SS, et al. Autofluorescence imaging in age- detects early stages of chloroquine retinopathy similar to multifocal electroreti-
related macular degeneration complicated by choroidal neovascularization: a nography, fundus autofluorescence and near-infrared autofluorescence. Br J
prospective study. Ophthalmology 2008;115:342–6. Ophthalmol 2009;93:1444–7.
Optical Imaging Technologies Section 1

Advanced Imaging Technologies Chapter

Pearse A. Keane, Humberto Ruiz-Garcia, SriniVas R. Sadda


5 
INTRODUCTION – RETINAL IMAGING development, fundus photography has incorporated angio-
graphic methodologies for the visualization of blood vessels.1,4
TO DATE
In the 1960s, the assessment of retinal disease was revolutionized
Assessment of chorioretinal disease is dependent on the ability by the introduction of fluorescein angiography (Fig. 5.2A). In the
to visualize pathologic changes occurring in the posterior 1970s, evaluation of the choroidal circulation was also improved
segment of the eye using optical instruments, termed ophthal- through the use of indocyanine green dye. The development of
moscopy (Fig. 5.1A).1 Ophthalmoscopy, in turn, has been greatly these angiographic techniques has also led, at least in part, to the
enhanced by the development of techniques that allow record- discovery that certain fundal structures fluoresce in the absence
ing of these changes.2 By the end of the 19th century the first of contrast. The subsequent evolution of fundus autofluores-
images of the living human retina had been obtained (Fig. 5.1B) cence imaging has greatly extended our ability to evaluate retinal
and, by the 1950s, with the advent of electronic flashes and degenerative and other disorders.2
35-mm cameras, the field of modern retinal/fundal imaging Since the 1990s, fundus photographic systems have also made
had been born.3 the transition from “analogue” image capture using film to
As well as documenting pathology, fundal imaging facilitates digital image capture using charge-coupled devices (CCDs),
the identification of morphologic features not visible to the clini- allowing for improved image processing and analysis.2 In paral-
cian on biomicroscopy. For example, from an early stage in its lel with this, new image capture technologies have emerged such

Fig. 5.1 Early attempts at retinal visualization and image capture.


(A) Early model of the Helmholtz ophthalmoscope, 1851. (Alcon
Laboratories Museum of Ophthalmology, The Sherman Collection,
Fort Worth, TX, USA. Historical image: Helmholtz ophthalmoscope,
1851. Ophthalmology 2002;109:729. Reproduced with permission.)
(B) The first published human fundus photograph involved this
apparatus, an albocarbon burner, and a 2 12 -minute exposure. Blood
vessels cannot be defined, but the optic disc can be seen as a white
area superiorly, while inferiorly, the larger light area is an artifact.
(Reproduced with permission from Same PJ. Landmarks in the
historical development of fluorescein angiography. J Ophthalm
Photography 1993;15:18.)

B
134
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

BV
Retina Vitreous

BV

SRF RPE

Sclera

300 µm

C
Log Reflection
Fig. 5.2 Fluorescein angiography, scanning laser ophthalmoscopy, and optical coherence tomography (OCT). (A) Early attempt at fundus
fluorescein angiography in a normal subject. (Reproduced with permission from Novotny HR, Alvis DL. A method of photographing fluorescence
in circulating blood in the human retina. Circulation 1961;24:82–6.) (B) A single frame from a fluorescein angiogram taken with a confocal
scanning laser ophthalmoscope (cSLO) showing a choroidal neovascular membrane. (Reproduced with permission from Bennett PJ, Barry CJ.
Ophthalmic imaging today: an ophthalmic photographer’s viewpoint – a review. Clin Exp Ophthalmol 2009;37:2–13.) (C) Early attempt at OCT
imaging of the human retina and optic nerve, obtained in vitro. SRF, subretinal fluid; RPE, retinal pigment epithelium; BV, blood vessel.
(Reproduced with permission from Huang D, Swanson EA, Lin CP, et al. Optical coherence tomography. Science 1991;254:1178–81.)
as scanning laser ophthalmoscopy (SLO) and optical coherence
tomography (OCT) (Fig. 5.2B, C). In particular, the widespread
adoption of OCT in recent years has greatly extended our knowl- 135
edge of chorioretinal disease pathophysiology, and proven
important for the provision of new pharmacotherapeutics.1
Light from retina

Chapter 5
Despite the unprecedented advances of recent years, con-
siderable deficiencies exist in our retinal imaging capability.
While angiographic techniques provide exquisite detail of vas-
cular structures, our ability to quantify chorioretinal blood flow
Aberrated wavefront
and subsequent oxygen saturations – in a noninvasive manner
– remains inadequate. While OCT provides cross-sectional
images of the neurosensory retina (and more recently the

Advanced Imaging Technologies


choroid) with high axial resolution, its transverse resolution
is limited, and our ability to assess many retinal cell types
Beamsplitter
remains poor (e.g., Müller cells, microglia, astrocytes, and indi-
vidual neuronal elements). Furthermore, many advances in Corrected
microscopic techniques that permit “molecular” imaging in Adaptive wavefront
basic science research have yet to make the transition to human Control mirror
clinical studies. system
In this chapter, we discuss the attempts that are underway to
address the shortcomings of current retinal imaging technolo-
gies. We begin by describing the use of adaptive optics to provide
cellular-level image resolution, before moving on to describe
Doppler and spectral imaging techniques for the assessment of High-resolution
retinal blood flow and oxygenation. We then describe a number Wavefront retinal image
sensor
of technologies with the potential for assessment of novel func-
tional parameters, such as photoacoustic and magnetic reso-
nance imaging (MRI). We conclude by introducing the emerging
field of nanotechnology and describing how the use of targeted Fig. 5.3 Schematic representation of a typical adaptive optics system.
(Courtesy of Joseph Carroll, Williams Laboratory, University of
nanoparticles may ultimately allow molecular-level imaging in Rochester.)
clinical practice.

ADAPTIVE OPTICS – IMAGING OF SINGLE


CELLS IN THE RETINA Technology
The adaptive optics devices first developed for ophthalmic use
Basic principles were conventional fundus cameras modified to incorporate
Our ability to obtain high-resolution images of the human retina wavefront sensing and correction.7,8 These systems typically
is limited by the presence of defects, or aberrations, in the optical focus a laser beam on the retina at a location of interest. The light
system of the eye (i.e., the cornea and lens).5 In addition to exhib- reflected from the retina is then distorted by the optical system
iting lower-order monochromatic aberrations such as defocus of the eye before returning to the wavefront sensor. Information
and astigmatism, normal eyes also exhibit higher-order mono- obtained from the wavefront sensor is then used to alter the
chromatic aberrations such as coma, trefoil, and spherical aber- shape of the deformable mirrors and compensate for the aberra-
ration. The combined effects of these aberrations limit the quality tion. This process continues in a closed loop until the ocular
of images obtainable for the diagnosis and management of aberrations have been reduced to near diffraction-limited levels;
retinal disease. Until the early 1990s, there existed no effective at this point, a flash from a separate incoherent light source is
way to obtain rapid, precise measurements of the human eye’s triggered by the wavefront sensor, illuminating the retina as an
optical aberrations. However, wavefront sensors based on the image is captured by the digital fundus camera.
Hartmann–Shack principle, originally developed for astronomi- Adaptive optics components have also been incorporated into
cal purposes, have now been adapted for use in the human eye. confocal SLO systems, offering the advantage of increased con-
These sensors consist of an array of lenses, each having the same trast, cross-sectional imaging, and the measurement of dynamic
focal length, which can be used to approximate the whole wave- changes such as blood flow.5,9 In an adaptive optics SLO, light
length of incident light. Once measured, the detected aberrations returning from the retina is split into a light detection path and
can then be corrected (i.e., rendered flat) using one or more a wavefront-sensing path. Again, deformable mirrors are used
deformable mirrors (mirrors have large numbers of small elec- to provide closed-loop dynamic compensation for ocular aber-
tronically controlled actuators on their rear surface that can push rations. In contrast to flood-illuminated adaptive optics devices,
and pull the mirror within a range of ±2 µm, allowing it to adopt the adaptive optics SLO uses the same source of light for the
any desired configuration) (Fig. 5.3). By incorporating wavefront wavefront sensor as for the retinal illumination; as a result, the
sensing and correction into existing optical imaging platforms – wavefront sensor is able to measure aberrations from the entire
“adaptive optics” – it is now possible to acquire images of the scanned area of the retina. When imaging the retina, SLO devices
retina with cellular-level resolution, and in a noninvasive provide a transverse resolution of approximately 15 µm but are
fashion.5,6 limited to an axial resolution of approximately 300 µm. With the
addition of adaptive optics correction to an SLO, the transverse on the retina – a factor limited by ocular aberrations. A suc-
resolution may be increased to less than 3 µm while the axial cessful combination of OCT and adaptive optics could poten-
136 resolution may be improved to 40 µm. tially demonstrate, therefore, the narrowest point-spread
Adaptive optics devices have also been incorporated into function of all in vivo retinal-imaging techniques (Fig. 5.4).
prototype OCT systems.10,11 The axial resolution achievable However, a number of technical challenges remain, in particular,
using OCT is many orders of magnitude greater than that the reductions in image quality that occur as a result of chro-
Section 1

achievable using an SLO device, with commercially available matic aberrations when large-bandwidth light sources are
devices commonly achieving axial resolutions of approximately used.12,13
5  µm. However, the transverse resolution of OCT systems is Finally, adaptive optics may be useful for the performance
determined by the size of the laser spot that can be focused of fundus perimetry (“microperimetry”) in the study of macular
Optical Imaging Technologies
Retinal Imaging and Diagnostics

AO-OCT Volume Image of Retina

NFL

GCL

0.25
mm

OPL

0.4
mm OS

0.3 mm

Fig. 5.4 Adaptive optics optical coherence tomography (AO-OCT) volume acquired over a 1° retinal region located temporal of the fovea, as
illustrated by the rectangle in the fundus photograph. The images on the right are en face views of particular retinal layers extracted from the
AO-OCT volume. Retinal layers from top to bottom are: nerve fiber layer (NFL), ganglion cell layer (GCL), outer plexiform layer (OPL), and
outer-segment (OS) layer of photoreceptors. (Reproduced with permission from Miller DT, Kocaoglu OP, Wang Q, et al. Adaptive optics and the
eye (super resolution OCT). Eye 2011;25:321–30.)
disease.1 In current microperimetry systems, macular light segment and is reflected by the retinal pigment epithelium
sensitivity is measured using a stimulus size that covers an (RPE), the most highly reflective surface in the retina. As light is
area containing in excess of 150 cones at the foveal center. reflected out of the eye, it is guided toward a small area of the 137
As with the imaging modalities described above, further reduc- pupil by the cone inner segments (in this respect cones act as
tions in stimulus size are hindered by aberrations of the optical “optical fibers,” rejecting stray light in the retina and maximiz-

Chapter 5
systems in the human eye, a shortcoming that may be over- ing efficiency in light collection). The human eye has three
come through the incorporation of adaptive optics.14 In addi- classes of cone photoreceptors: blue (S), green (M), and red (L).
tion, by analyzing the warping that occurs both within Adaptive optics has supplied the first data regarding the propor-
individual SLO frames and between frames, adaptive optics tion, and arrangement, of each cone class in the human eye.18 By
can be used to resolve the effects of eye movements at a fine quantifying cone receptor density, it has also been demonstrated
spatial scale. As a consequence, in adaptive optics micrope- that the density of cone packing appears to be lower in highly
rimetry, real-time stabilization of the retinal image is possible, myopic eyes versus emmetropic eyes – a finding consistent with

Advanced Imaging Technologies


allowed for targeted delivery of the small visual stimulus to the reduced visual acuity and contrast sensitivity observed in
the retina.15,16 the eyes of axial myopes.19
Rod photoreceptors are abundant in the peripheral retina but
Visualization of retinal structures are also seen in the macular region of human eyes. Rods are not
Cone photoreceptors are the dominant feature seen with both as easily visualized in the normal retina, even in areas of the
flood-illuminated and SLO devices (Fig. 5.5A).6,17 Light projected retina where they outnumber cones approximately 10-fold.6 The
into the eye from these devices passes through the cone outer difficulty in imaging rods is consistent with their smaller size

A B

C D

Fig. 5.5 Visualization of photoreceptors, retinal pigment epithelium (RPE), and retinal vasculature using adaptive optics. (A) The complete foveal
cone mosaic and (B) the complete peripheral photoreceptor mosaic showing both rods and cones, imaged at 10° temporal and 1° inferior. Scale
bars = 20 µm. (Reproduced with permission from Rossi EA, Chung M, Dubra A, et al. Imaging retinal mosaics in the living eye. Eye 2011;25:
301–8.) (C) Adaptive optics scanning laser ophthalmoscopy image revealing a patchy foveal cone mosaic with increased cone spacing, and
resulting visualization of the RPE cells (three RPE cells in the bottom left corner have been outlined, and the preferred retinal locus is also
indicated by the dashed circle in the image). (Reproduced with permission from Roorda A, Zhang Y, Duncan JL. High-resolution in vivo imaging
of the RPE mosaic in eyes with retinal disease. Invest Ophthalmol Vis Sci 2007;48:2297–303.) (D) Three successive frames demonstrate a
single leukocyte’s (1) change in position from left to right in each frame. A second leukocyte (2) has just come into view in the last frame. Scale
bar = 100 µm. (Reproduced with permission from Martin JA, Roorda A. Direct and noninvasive assessment of parafoveal capillary leukocyte
velocity. Ophthalmology 2005;112:2219–24.)
(approximately 2 µm in diameter), and their broad angular Although cone photoreceptors offer high contrast and can
tuning, which reflects less light back through the pupil where it now routinely be imaged using adaptive optics, other cell types
138 can be collected for imaging. However, recent advances, includ- in the retina are not as easily visualized. Unlike cones, other
ing the use of smaller confocal pinholes and improvements in neurons are not highly reflective of light and do not exhibit high
registration algorithms, have now allowed clear images of contrast (the reflected signal from ganglion cells is 60 times
single rods to be obtained in the living human eye (as well as lower than that from cones). These difficulties are especially dif-
Section 1

aiding visualization of the smallest cones at the foveal center) ficult to surmount given the safe limits for laser usage in humans,
(Fig. 5.5A).20 and the effects of involuntary eye movements. Despite these
Rod and cone photoreceptors rest on a monolayer of RPE cells. obstacles, recent animal studies have made progress toward in
Each RPE cell in the monolayer has an approximate diameter of vivo visualization of retinal ganglion cells using fluorescent
10 µm, dimensions well within the resolution limits of adaptive markers, and the development of similar capabilities in humans
optics systems; however, direct in vivo visualization of single may lead to improved understanding of optic neuropathies such
Optical Imaging Technologies
Retinal Imaging and Diagnostics

RPE cells remains challenging. Unlike cone photoreceptors, the as glaucoma.26


intrinsic contrast of RPE cells is poor. In addition, much of the
light scattered by the RPE is masked by the overlying photore- Early clinical applications
ceptors, which are also highly scattering. To date, direct imaging The cellular-level resolution provided by adaptive optics retinal
of the RPE has only been possible in human subjects with retinal imaging allows direct measurement of photoreceptor density
disease where absence of cone photoreceptors has allowed visu- and diameter and is, therefore, an ideal tool for the examination
alization of the underlying RPE (Fig. 5.5B).21 However, by the of patients with inherited retinal degenerations (Fig. 5.6). On
incorporation of adaptive optics with fundus autofluorescence, examination with adaptive optics, diseased cones do not show
images of the RPE cell mosaic have been achieved in primate the same reflectance pattern as healthy photoreceptors, resulting
eyes.22 in dark gaps or areas of “drop-out” within the cone photorecep-
The high scanning speed of SLO systems allows retinal images tor mosaic.27 Significant correlations have been found between
to be captured at video rates, thus allowing measurement of parafoveal cone density and a number of functional parameters,
dynamic retinal changes, such as blood flow. The use of an adap- including best-corrected visual acuity, contrast sensitivity, and
tive optics SLO thus allows direct visualization of white blood detection sensitivities for visual stimuli recorded using multifo-
cells as they transit through retinal capillaries (the smaller size cal electroretinography.28–30 In a recent phase II clinical trial
and different absorption characteristic of red blood cells make where patients with inherited retinal degenerations were treated
them more difficult to visualize) (Fig. 5.5C).23,24 As a consequence, with sustained-release ciliary neurotrophic factor, no changes in
direct noninvasive measurement of the velocity of retinal blood visual acuity, visual field, or electroretinography responses were
flow becomes possible. Retinal imaging with adaptive optics observed. However, using an adaptive optics SLO, significant
may also be useful for detection of structural changes in the differences in both cone density and spacing were detected
retinal vasculature not visible with other imaging modalities, between treatment and control groups.31
such as microaneurysms and hard exudates in diabetic High-resolution adaptive optics imaging has also been used
retinopathy.25 to aid the examination of patients with “macular microholes,”32

250 µm

A B C

80 µm 40 µm

Fig. 5.6 Adaptive optics scanning laser ophthalmoscope (AOSLO) images at different magnifications with corresponding features on the fundus
photograph (A). (B) Six-degree montage from 3° adaptive optics images of the central macula of the cone–rod dystrophy patient’s right eye. In
addition to the features detected in the fundus photograph, detailed structures in the granular pattern of the retinal pigment epithelium in the
atrophic bull’s-eye lesion are observed. Within the central relatively spared region (box), photoreceptors are seen as gray dots. (C) Three-degree
montage from 1.5° images. The photoreceptors are visible in the central relatively spared area of the retina. (Reproduced with permission from
Wolfing JI, Chung M, Carroll J, et al. High-resolution retinal imaging of cone–rod dystrophy. Ophthalmology 2006;113:1019.)
color blindness,33 foveal hypoplasia,34 and otherwise unex- Blood flow (Q) is the volume of blood passing through a vessel
plained visual disturbances.35 In patients with glaucoma and in a given time, and is determined by the velocity of the blood
other optic neuropathies, adaptive optics has also revealed (V) multiplied by the cross-sectional area of the blood vessel 139
evidence of structural changes in cone photoreceptors when through which it passes (πr2).38 Consequently, if blood flow
there is permanent damage to the overlying inner retinal velocity can be measured using the Doppler effect, and the diam-

Chapter 5
layers.36 eter of the blood vessel can also be measured, then absolute
values for blood flow may be determined. Measurements of
Conclusions retinal vascular diameter can be acquired from fundus images
It is clear that the addition of adaptive optics to existing retinal obtained with standard optical imaging techniques (e.g., fundus
imaging platforms is an important step for clinical ophthalmol- photography).39,40 However, in order to measure retinal vascular
ogy, particularly when such devices can be made inexpensive, diameters in real units of length, the magnification of the image
user-friendly, and reliable. In fact, the first commercially avail- induced by the eye, as well as the magnification of the camera,

Advanced Imaging Technologies


able adaptive optics system, for ophthalmic research purposes must be known (failure to account for such magnification will
only, has recently been launched (the rtx1 Adaptive Optics result in significant errors).
Retinal Camera, Imagine Eyes, France). Rapid progress is also
being made on the use of adaptive optics with other imaging Non-Doppler assessment of retinal
systems, and, although significant technical challenges must still blood flow
be overcome, the inclusion of adaptive optics in OCT systems Measurement of retinal blood flow is possible using quantitative
seems particularly promising. angiography, based on use of dye dilution techniques.37,39,40 In
this method, the concentration of fluorescent dye within the
DOPPLER IMAGING – ASSESSMENT OF blood at a specific observation point is graphed over time, pro-
BLOOD FLOW ducing a dye dilution curve. The enhanced contrast provided by
SLO systems is particularly suited to this approach, and it can
Basic principles be used to evaluate vascular parameters such as retinal arterio-
The Doppler effect, first described in the 19th century by the venous passage time and mean dye velocity. The increased con-
Austrian physicist Christian Doppler, is the change in frequency trast provided by SLO images also allows measurement of blood
of a wave as it is reflected off a moving object: if the reflecting velocities in the small vessels surrounding the fovea and the
object is moving away from the observer/transducer, the fre- optic nerve head (in the future, this technique is likely to be
quency of the reflected waves is lower than that of the waves enhanced through the incorporation of adaptive optics).40 Indo-
emitted, and vice versa. As the frequency shift is dependent on cyanine green angiography may also be used to evaluate the
the velocity of the moving object, this effect can be used to choroidal circulatory flow, as the infrared light sources employed
measure the velocity of blood flowing in the eye.37 Importantly, penetrate the RPE more efficiently than the shorter wavelengths
the Doppler effect is also dependent on the angle between the used in fluorescein angiography. In one analysis method, the
axis of the wave and the axis of movement of the object. There- choroid is divided into six regions, and dye dilution curves are
fore, if the observer/transducer is not parallel to the axis of the created for each region (Fig. 5.7A). Parameters such as 10%
moving object calculations must be performed using a Doppler filling time, and maximum brightness, can then be calculated.40
angle correction formula: SLO-based fluorescein and indocyanine green angiography
provide significant information regarding retinal and choroidal
V = ∆FC / 2F0 cos α blood velocities. However, without measurement of correspond-
ing vascular diameters, it is not possible to measure blood flow.
where V is the velocity of the moving object, ΔF is the Doppler In addition, the invasive nature of the dye injections required for
(frequency) shift, C is the velocity of the wave in the medium, these approaches precludes their routine use in many clinical
F0 is the frequency of the wave source, and cosα is the Doppler scenarios.
angle. Of note, accurate measurement of the Doppler angle is a
significant obstacle to the noninvasive assessment of ocular Doppler ultrasound
blood flow. Furthermore, as the Doppler angle increases between Color Doppler imaging (CDI) is an ultrasound technique that
60° and 90°, velocities calculations are subject to significant combines B-scan gray-scale imaging of tissue structure, color
errors (cosine 90° is equal to zero). representation of blood flow based on Doppler shifted frequen-
Utilization of the Doppler effect in ocular imaging systems cies, and pulsed-Doppler measurement of blood flow velocities
allows calculation of ocular blood flow velocities.37 Reductions (Fig. 5.7B).37,40 As in other Doppler-based methods, blood flow
in blood flow velocity may occur as a result of vascular degen- velocity is determined by the shift in the frequency of sound
erative changes in diseases such as diabetic retinopathy, or as a waves reflected from the moving blood column. Color is then
result of vascular occlusion in diseases such as central retinal added to the B-scan gray-scale image of the eye to represent the
vein occlusion (CRVO). However, changes in retinal blood flow motion of blood through the vessels. The color varies in propor-
velocity may also occur as a result of either constriction or dilata- tion to the flow velocity and is typically coded red-white for
tion of vessels during normal physiological autoregulation motion toward the probe, and blue-white for motion away from
(according to Bernoulli’s principle, constriction of a blood vessel the probe. Using the color Doppler image, the operator can then
causes a conversion of pressure into kinetic energy, thereby identify the vessel of interest and place the sampling window
increasing the velocity of the blood, but decreasing its pressure). for pulsed Doppler measurements (this window is generally
Therefore, measurements of the absolute quantities of “blood situated in the center of the vessel). At this point, the general
flow” may represent a more clinically relevant parameter. flow axis of the blood flow is detected simply by observation to
140
Section 1

A B C

D E F
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 5.7 Evaluation of chorioretinal blood flow. (A) Quantification of choroidal blood flow using indocyanine green angiography. Six locations,
each a 6° square, are identified on the image for dye dilution analysis. (B) Color Doppler image (CDI) of the central retinal artery and vein. The
Doppler shifted spectrum (time–velocity curve) is displayed at the bottom of the image. Red and blue pixels represent blood movement toward
and away from the transducer, respectively. (C) Confocal scanning laser Doppler flowmetry (Heidelberg retinal flowmeter) of optic nerve head
and peripapillary retina. The left arrow indicates a 1 × 1-pixel measurement window, which collects flow values from the entire retina except for
large vessels, for new pixel-by-pixel analysis. The right arrow indicates a 10 × 10-pixel measurement window used for conventional analysis.
(Reproduced with permission from Harris A, Chung HS, Ciulla TA, et al. Progress in measurement of ocular blood flow and relevance to our
understanding of glaucoma and age-related macular degeneration. Prog Retin Eye Res 1999;18:673–7.)

determine the Doppler flow angle for the appropriate calculation


of velocity. Flow–velocity data are then plotted against time, and Laser Doppler velocimetry
the computer identifies the peak and trough of the waves. Bidirectional laser Doppler velocimetry (LDV) is a technique
Current CDI analysis focuses primarily on the arteries located used to quantify maximum blood velocity in large retinal
behind the globe: ophthalmic artery, central retinal artery, and vessels.37,43 Instruments based on this principle typically consist
posterior ciliary arteries. CDI can be used to describe blood flow of modified fundus camera where the body of the camera has
to the eye in terms of a set of well-defined parameters including: been replaced by a fiberoptic unit. A low-powered laser light
(1) peak systolic velocity (PSV); (2) end-diastolic velocity; and (3) source casts a beam on to the ocular fundus which is positioned
resistance index.37,40 It does not provide absolute measurements by the operator on the retinal vessel of interest. The Doppler
of blood flow (no quantitative information on vessel diameter is shifted frequency spectra of the returning light can then be mea-
obtained). With a 7.5-MHz probe, CDI is able to resolve struc- sured. These spectra exhibit large fluctuations up to a clearly
tures 0.2 mm (200 µm) or larger, but can also be used to measure measurable maximum shift – this maximum shift arises from
Doppler shifts in smaller vessels such as the posterior ciliary scattered light from red blood cells flowing at the maximum
arteries (diameter of approximately 40 µm). CDI may be of par- speed at the center of the vessel. If the Doppler angles are then
ticular use for the primary evaluation and follow-up of orbital known, the maximum frequency shift can be used to calculate
vascular lesions, such as varices, arteriovenous malformations, the maximum vessel velocity; however, these angles are difficult
and carotid-cavernous sinus fistulas. It has also been used for to measure reproducibly. Therefore, a bidirectional technique
the semiquantitative assessment of perfusion in retinal and cho- was developed to expand the capability of LDV to provide abso-
roidal vascular disease. Specifically, in patients with CRVO, the lute measurements of maximum velocity.43 In the bidirectional
central retinal artery PSV and end-diastolic velocity have been technique, one incident beam is used to illuminate a sight along
found to be much lower than in unaffected fellow eyes and in a retinal vessel. The light scattered by the blood cells at the illu-
healthy control subjects.41 Patients with ocular ischemic syn- minated site is then detected simultaneously in two distinct
dromes also present reduced PSVs in the central retinal and directions separated by a known, fixed angle. Two Doppler
posterior ciliary arteries, as well as increased resistance.42 shifted frequency spectra are measured and the difference
between their maximum shifts is then used to calculate the number of Doppler OCT prototypes utilize the phase change
maximum velocity in units of speed. between sequential A-scans in order to generate information
By combining an eye-tracking system with a retinal laser regarding the Doppler shift.10 Although OCT can generate 141
instrument based on bidirectional LDV, Canon has developed a cross-sectional images of retinal blood flow, accurate quanti-
commercially available instrument (Canon Laser Blood Flow­ fication of this flow remains challenging: to achieve this it is

Chapter 5
meter (CLBF)-100, Tokyo, Japan).44 This device allows concomi- necessary to measure the geometry of the vessel and, in par-
tant measurement of blood vessel diameter; as a result, total ticular, the Doppler angle. Furthermore, current Doppler OCT
retinal blood flow in a single vessel can be calculated in absolute systems are limited in terms of the maximal velocities measur-
units. Thus far, the smallest vessels in which the blood flow rate able, and in terms of the smallest vessel diameters measurable
has been measured were approximately 40 µm in diameter.44 (capillary flow involves single erythrocyte movement rather
than continuous fluid flow).
Laser Doppler flowmetry

Advanced Imaging Technologies


Laser Doppler flowmetry (LDF) is a technique where laser light Conclusions
is not directed at a retinal vessel, but on an area of vascularized A number of commercial devices, allowing assessment of retinal
retinal tissue with no large vessels visible.37,39 LDF instruments blood flow, have been introduced. However, these devices have
are based on the theories of Bonner and Nossal, which describe significant issues which have prevented their widespread adop-
the characteristics of laser light injected at one point into capil- tion as of yet. The prospects for Doppler OCT appear good, yet
lary tissue and collected at an adjacent point.43 This theory relates it is still at an early stage in its development pathway. Further-
the Doppler shift of the light to the total number of blood cells more, due to the complexity of retinal hemodynamics, extensive
moving in the illuminated tissue volume, and to the flow rate of validation and reproducibility studies are required before accep-
the moving cells. By using this theory, relative measurements of tance of any new measurement approach.
the mean velocity of the red blood cells, and the blood volume,
can be obtained. Relative values of blood flow can then be cal- SPECTRAL IMAGING – ASSESSMENT OF
culated as the product of velocity and volume. However, such RETINAL OXYGENATION
measurements are not absolute, and variations in vascular
density and vessel orientation, even within relatively small Basic principles
volumes of tissue, can lead to considerable differences in the Spectroscopy is the study of the interaction between any form
scattering properties between subjects. of matter and radiated energy (e.g., visible light). By measuring
Scanning LDF combines the principles of scanning laser radiation intensity as a function of wavelength, and through the
tomography and LDF, with the commercially available Heidel- identification of characteristic signatures, it is possible to deter-
berg retina flowmeter (HRF) providing two-dimensional flow mine the constituents of a material. For example, in astronomy,
maps of the retina and optic nerve head (Fig. 5.7C).45 In this spectral analysis can be used to derive many properties of
system, the HRF images a 2880 × 720  µm area of the retina or distant stars and galaxies. As well as its uses in remote sensing,
optic nerve, at a resolution of approximately 10  µm/pixel. After spectroscopy has also been adapted for a number of laboratory-
the scan is performed, the HRF computer performs a fast Fourier based applications. In biological systems, the combination of
transform to extract the Doppler shift spectrum from each mea- spectroscopy with conventional imaging techniques – “spectral
sured pixel of reflected light. The HRF device is simple to use imaging” – allows determination of the spatial distribution of
and sensitive to small changes over time in the same eye. spectroscopic data.47 In clinical settings, the application of spec-
However, the flow measurements are displayed in arbitrary troscopic principles has been of particular use for oximetry – the
units and there is a lack of accuracy when dealing with very measurement of oxygen saturation in a patient’s blood. From
high and very low blood speeds. Furthermore, contributions to as early as 1935, transillumination of the ear has been used as
blood flow measurements from the underlying choriocapillaris a method of continuously measuring oxygen saturation in
cannot be excluded and previous experimental tests of the human blood. The subsequent incorporation of plethysmogra-
instrumentation have demonstrated that large-flow readings can phy (the analysis of pulsatile components of the arterial cycle)
be obtained from sample volumes even when they contain no has facilitated the development of pulse oximeters capable of
moving cells.43 measuring arterial oxygen saturation in isolation.
The use of spectral imaging to perform blood oximetry is
Doppler optical coherence tomography dependent on assumptions about the relationship between light
OCT, first described by Huang et al. in 1991, uses interferom- transmittance through the blood and its oxygen saturation.47
etry to generate high-resolution cross-sectional images of the According to the Lambert–Beer law, for any given wavelength
neurosensory retina.46 Since the mid-1990s, Doppler measure- of light, its transmittance through blood is dependent on the
ments have been incorporated into prototype OCT imaging extinction coefficient of the blood (ε), the concentration of blood
systems (Fig. 5.8).10 In the original time-domain OCT systems, (c), and the path length (d) through which the light travels:
the blood velocity can be determined by measurement of the
Doppler shift of the interference fringe frequency after Fourier IT = IO 10 − εcd ,
transformation of the data, whereas, in newer spectral domain
OCT systems, direct recording of the interference fringe where IT is the intensity of the light transmitted through the
frequency is possible. However, in this approach, blood veloc- blood, and Io is the intensity of incident light. By rearranging
ity sensitivity and image spatial resolution are coupled and the equation, the extinction coefficient of blood (ε) can be seen
inversely related (increasing the velocity sensitivity decreases to be proportional to the logarithm of (Io/IT), i.e., the optical
the spatial resolution). In order to overcome this limitation, a density (OD):
142
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

1.0

0.0

C
–1.0

Fig. 5.8 Doppler optical coherence tomography (OCT). (A) Fundus photograph showing the double circular pattern of the OCT beam scanning
retinal blood vessels emerging from the optic disc. (B) The relative position of a blood vessel in the two OCT cross-sections is used to calculate
the Doppler angle θ between the beam and the blood vessel. (C) Color Doppler OCT image showing the unfolded cross-section from a circular
scan. Arteries and veins can be distinguished by the direction of flow as determined by the signs (blue or red) of the Doppler shift and the
angle θ. (Reproduced with permission from Wang Y, Fawzi AA, Varma R, et al. Pilot study of optical coherence tomography measurement of
retinal blood flow in retinal and optic nerve diseases. Invest Ophthalmol Vis Sci 2011;52:841.)

εcd = log(IO / IT ) transmittance. Instead, measurements of light reflected from the


εcd = OD retina are typically used:

The extinction coefficient of blood (ε) is dependent on its main ODvessel = log(IV / IR )
absorbing component, hemoglobin, which, in turn, is dependent
on its oxygen concentration. As the extinction coefficient of where IV and IR are the intensity of light reflected from the
blood varies with wavelength, analyzing the optical density of retinal vessel and adjacent retina respectively (retinal oximetry
blood at multiple wavelengths compensates for variables such assumes that a large fraction of incident light is reflected back
as the concentration and path length and ultimately allows esti- from the tissues surrounding the vessels). Although the prin-
mation of oxygen levels in the blood (the exact details of these ciples described above require a number of assumptions that
calculations are beyond the scope of this chapter, but have been may not hold true in the real world, measurement of light
reviewed in detail by Harris et al.).48 However, in retinal oxim- reflected from the retina at multiple wavelengths, in this
etry, it is impractical to determine optical densities (and thus manner, forms the basis for a number of noninvasive retinal
oxygen concentrations) through the measurement of light oximetry technologies.
band-pass filter only allows light of specific wavelengths to
Technology pass). This simultaneously yields four fundus images, each with
Spectral imaging devices typically employ one of three different a specific wavelength of light. Specialized software automati- 143
approaches: (1) multispectral imaging; (2) hyperspectral imaging; cally selects measurement points on these images and calculates
and (3) imaging spectroscopy.47,48 Initial efforts by Hickam the optical density of retinal vessels at two wavelengths, 605 nm
et al. in the 1950s49 employed film-based fundus cameras; more

Chapter 5
and 586 nm (optical density is sensitive to oxygen saturation at
recently, digital fundus cameras and confocal scanning laser 605 nm but not at 586 nm). The ratio of these optical densities is
ophthalmoscopes have also been employed. Early attempts have approximately linearly related to the hemoglobin oxygen con-
also been made at the utilization of OCT in association with centration. The oximeter is then calibrated to yield relative
spectral analysis techniques. oxygen saturation values. This approach has been shown to be
Multispectral imaging involves the measurement of reflected sensitive to changes in oxygen concentration and to yield repro-
light from images obtained at discrete and somewhat narrow ducible results. Other commercially available multispectral

Advanced Imaging Technologies


spectral bands. Adopting a similar approach to that first devices have also recently been described (Imedos UG, Jena,
described by Beach et al.,50 Hardarson et al. have developed Germany).
Oxymap, a commercially available device approved for investi- Hyperspectral imaging involves the measurement of light
gational usage.51 This device consists of a fundus camera, which from images obtained at narrow spectral bands over a contigu-
is coupled with a beam splitter and a digital camera. The beam ous spectral range. Mordant et al. have recently described the
splitter separates the original image into four optical channels, validation of a hyperspectral fundus camera for noninvasive
each of which contains a different narrow band-pass filter (each retinal oximetry (Fig. 5.9).52 In their system, a commercial fundus

Arterioles Venules

OS
100%

90%

80%

70%

60%

50%

40%

30%

20%

10% B
0%

Fig. 5.9 Spectral imaging of the retina. (A) Color fundus photograph demonstrating a left inferotemporal branch retinal arteriole occlusion.
(B) Pseudocolor oxim map showing abnormally low oxygen saturation within the affected inferotemporal retinal arteriole in contrast to the normal
oxygen saturation levels in the unaffected superotemporal arteriole. The corresponding inferotemporal retinal venule has a normal level of
oxygen saturation. (Reproduced with permission from Mordant DJ, Al-Abboud I, Muyo G, et al. Spectral imaging of the retina. Eye 2011;25:317.)
camera is linked to a liquid crystal tunable filter in the optical absolute, measurements of oxygen saturation. Furthermore, all
path of the camera’s light source; this enable the electronic selec- devices rely, to a certain extent, on biophotonic assumptions that
144 tion of a combination of desired wavelengths between 400 and may not hold true for in vivo imaging. Therefore, it is vital that
700 nm. A CCD camera is then used to record a sequence of detailed validation and reproducibility assessments be per-
spectral images between 500 and 650 nm in 2-nm increments formed on all new retinal oximeters before such devices can be
(this process takes approximately 10–15 minutes in healthy vol- widely adopted for clinical or research purposes.
Section 1

unteers). Further image processing and analysis then take place:


the images are registered, the retinal vessel profiles extracted, PHOTOACOUSTIC IMAGING –
and their optical densities estimated. Oxygen saturations can ASSESSMENT OF RETINAL ABSORPTION
then be calculated. Use of hyperspectral imaging in this manner
may provide more accurate measurements of oxygen saturation Basic principles
than two-wavelength multispectral approaches (such approaches In large part, contemporary retinal imaging modalities are based
Optical Imaging Technologies
Retinal Imaging and Diagnostics

have been reported to overestimate oxygen saturation). However, on the measurement of light reflected from the retina, e.g.,
such an approach requires the use of sensitive detectors and fundus photography, SLO, OCT systems.2 In contrast, no oph-
powerful computers to enable fast and accurate processing of thalmic imaging modality exists that can directly measure the
images. absorption of light by retinal tissues – information of potentially
A third method utilized for the noninvasive measurement of great significance. Assessment of optical absorption profiles at
retinal oxygen saturation involves imaging spectroscopy. Sch- multiple wavelengths may improve the accuracy of retinal vas-
weitzer et al. have described an imaging ophthalmospectrome- cular oxygen saturation measurements (current “spectral
ter, which consists of a modified fundus camera and an attached imaging” determines optical density ratios indirectly, through
spectrograph.53 The instrument illuminates the retina with a the measurement of reflected light). Retinal absorption charac-
small slit of light and then simultaneous measurements are teristics may also provide contrast for generation of enhanced
made at 76 different wavelengths in this discrete area using a retinal angiographic maps. Assessment of optical absorption
spectrometer. This approach may be the most accurate; however could also be useful for providing contrast when imaging the
a major limitation is that measurements are made at one single highly pigmented RPE cell mosaic. Fortunately, through recent
cross-section of one or two retinal vessels. By contrast, a com- advances in microscopy, utilizing the photoacoustic effect, it has
plete two-dimensional mapping of oxygen saturation in the become possible to acquire optical absorption profiles in the
retinal vascular tree is needed for clinical diagnostics. context of noninvasive ophthalmic imaging – photoacoustic
ophthalmoscopy (PAOM).59,60
Clinical applications The photoacoustic effect was first recognized by Alexander
Using the Oxymap system, Hardarson and Stéfansson have Graham Bell in the 1880s when he discovered that thin discs of
evaluated retinal vasculature oxygen saturations in patients fol- selenium emit sound when exposed to a rapidly interrupted
lowing CRVO and branch retinal vein occlusion (BRVO).54,55 In beam of sunlight. This phenomenon occurs as energy absorbed
patients with CRVO, they demonstrated that retinal venular from the incident light is converted into kinetic energy, which
oxygen saturation is lower than in fellow eyes. However, they results in local heating and, thus, generation of a pressure wave
also demonstrated considerable variability within and between or sound. In the recently described photoacoustic microscopy
CRVO eyes. Similarly, in patients with BRVO, they found con- (PAM),61 a laser is used to irradiate a target tissue and thus
siderable variability in oxygen saturation between patients. They induce ultrasonic pressure waves as a result of specific optical
found evidence of hypoxia in some patients but not others and absorption. These pressure waves can then be recorded using a
speculated that this reflected variable disease severity in terms high-resolution ultrasonic transducer, and images generated.
of degree of occlusion, recanalization, collateral circulation, and Through the integration of OCT technology with a laser-
coexistent tissue atrophy. Hardarson et al. have also used this scanning, optical-resolution PAM, Jiao et al. have recently
system to examine the effects of glaucoma filtration surgery, and reported the use of PAOM in small animals.59,60
topical antiglaucoma medications, on retinal vascular oxygen
concentrations.56 Technology
Using the Imedos system, Hammer et al. found that, in patients In the system developed by Jiao et al., the illumination source is
with diabetes, increasing severity of retinopathy was associated a frequency-doubled, Q-switched, Nd:YAG laser, combined
with increased retinal venous oxygen saturations (from 63 ± 5% with the output laser beam of a fiber-based spectral domain OCT
for mild nonproliferative retinopathy to 75 ± 8% for proliferative system, and then scanned across the retina using a galvanom­
retinopathy). They suggest that these changes may occur second- eter.59 The photoacoustic waves induced from the retina are then
ary to the degeneration of capillary vascular beds with formation detected by an ultrasonic transducer placed in contact with the
of arteriovenous shunt vessels.57 Conversely, earlier work by eyelid (coupled by ultrasound gel). The resulting photoacoustic
Tiedeman et al. demonstrated evidence of increased oxygen con- images can then be registered with the images generated from
sumption (i.e., decreased retinal venous oxygen saturation) in the integrated OCT system (Fig. 5.10). As with conventional OCT
diabetic patients with acute hyperglycemia.58 image sets, maximum-amplitude “projection” images can be
generated from photoacoustic datasets allowing two-dimensional
Conclusions visualization of the retinal vasculature. Volumetric images can
Although major progress has been made in recent years, there also be generated following automated segmentation of the RPE
is currently no consensus on the optimal method for measure- and retinal vessels. More recently, the same group has tested the
ment of oxygen saturation in the retinal vasculature. A number acquisition of photoacoustic images in association with fundus
of spectral imaging devices provide relative, rather than autofluorescence signals (apart from generating heat, the
Fig. 5.10 Comparison of optical coherence
tomography (OCT) and photoacoustic
ophthalmoscopy (PAOM) images acquired
Blood vessel simultaneously in vivo. (A) PAOM B-scan 145
image in pseudocolor; (B) OCT B-scan
image; (C) projection image of the PAOM
data set. Scale: 100 µm. HA, hyaloid artery;

Chapter 5
RPE, retinal pigment epithelium. (Reproduced
with permission from Jiao S, Jiang M, Hu J,
et al. Photoacustic ophthalmoscopy for in
HA vivo retinal imaging. Optics Express
Blood vessel 2010;18:3971.)
A

Advanced Imaging Technologies


C

RPE RPE boundary


B

absorbed photons may undergo other physical processes, such causing the protons to spin and producing a rotating magnetic
as stimulating autofluorescence when fluorophores are present). field detectable by the scanner. Detectors in the MRI system then
Multimodal imaging in this manner may thus provide spatial evaluate a number of parameters (e.g., spin density, spin-lattice
information on the distribution of both melanin and lipofuscin relaxation time (T1), spin-spin relaxation times (T2)), which vary
via photoacoustic and autofluorescent signals respectively.62 depending on the local tissue environment. As a result, soft-
tissue images can be generated. Image contrast may be further
Conclusions enhanced through the use of exogenous paramagnetic contrast
The development of photoacoustic imaging techniques may agents such as gadolinium. In this manner, clinical MRI scanners
greatly extend the scope of future retinal imaging; however, at can produce high-resolution images of the entire body, both
present, the technology remains at an early stage in the develop- noninvasively and in a single setting.63
ment process. Much of the work to date, on photoacoustic As well as its anatomical imaging capability, MRI scanning
imaging in the eye, has been performed in tissue samples or in can be used to measure blood flow.63 MRI-derived quantification
animals. A number of technical hurdles remain before images of blood flow may be performed by the use of exogenous intra-
will be obtained from living human subjects, or commercial, venous contrast agents. However, it may also be performed non-
clinical devices introduced. invasively by magnetically labeling blood as a means of
providing endogenous contrast. These techniques have been
widely used to quantify blood flow to the brain and have been
MAGNETIC RESONANCE IMAGING
cross-validated using positron emission tomography. Relative
As previously described, the acquisition of retinal images is blood oxygen saturations can also be measured using the blood
largely dependent on optical techniques such as fundus photog- oxygenation level-dependent (BOLD) technique. This technique
raphy. However, many such techniques are constrained by a detects differences in magnetic resonance signal intensity that
relatively small field of view, and are often limited when there arise from changes in the oxygen saturation of hemoglobin
is disease-induced opacification of the ocular media, such as lens during brain activation – a local decrease in concentration of
opacity or vitreous hemorrhage. In the clinical setting, these deoxygenated hemoglobin will increase the BOLD signal, while
limitations are addressed, at least in part, by acoustic imaging an increase will decrease the BOLD signal. In the brain, when a
techniques such as ultrasonography. More recently however, specific region is activated in response to stimulation, local blood
advances in MRI offer the prospect of retinal application in flow increases in response to the increased metabolic demand;
humans.63,64 In addition to a wide field of view, and the ability such increases in blood flow will provide a boost in oxygen
to acquire images despite media opacification, retinal MRI may delivery and thus decrease the concentration of deoxygenated
also enable evaluation of novel functional parameters. hemoglobin. Techniques that measure blood flow and oxygen-
ation can thus be used to image brain function noninvasively –
Basic principles functional MRI (fMRI).63
In MRI systems, a powerful magnetic field is applied to the body
leading to alignment of the magnetization of its hydrogen nuclei Retinal imaging
or protons (the human body is largely made up of water mole- To date, most work on retinal imaging using magnetic resonance
cules which contain two hydrogen atoms). Radiofrequency has been reported in animal studies (Fig. 5.11).63 As the spatial
fields are then used to alter this alignment systematically, resolution of MRI is limited compared to that of OCT and other
large, nonhuman primates (baboons) using a standard clinical
vitreous scanner.66 Studies of this nature allow optimization of MRI scan-
146 T2-W ning parameters and represent a first step towards magnetic
resonance-derived retinal imaging in humans. Zhang et al. have
sclera
recently demonstrated, for the first time, the use of BOLD fMRI
to examine the changes associated with oxygen and carbogen
Section 1

challenges in the unanesthetized human retina.67 Although


DWIz much work has still to be done, such imaging may prove a useful
adjunct to more established optical imaging methods, particu-
larly when assessing the differential regulation of the retinal and
choroidal circulations.
Optical Imaging Technologies
Retinal Imaging and Diagnostics

DWIy NANOTECHNOLOGY
Basic principles
Nanotechnology involves the creation and use of materials and
devices on a nanometer scale, i.e., at the size scale of intracellular
DWIx structures and molecules.68 To comprehend the implications of
nanotechnology adequately, it is necessary to appreciate the rel-
Fig. 5.11 Magnetic resonance imaging (MRI) of the retina in vivo. ative sizes of important biological structures. For example: mac-
Higher-resolution T2-weighted (TE = 40 ms) and diffusion-weighted rophages are approximately 21 000 nm in diameter; red blood
(b = 504 s/mm2) images at 50 × 100 µm resolution. Diffusion-
sensitizing gradients were placed along the x, y, or z axis separately. cells are approximately 7000 nm in diameter; cone photorecep-
The small and large white arrows indicate the “inner” and “outer” tors measure between 500 and 4000 nm in diameter; the smallest
strips, respectively. (Reproduced with permission from Shen Q, Cheng single cellular organisms (bacteria of the genus Mycoplasma) are
H, Pardue MT, et al. Magnetic resonance imaging of tissue and
vascular layers in the cat retina. J Magn Reson Imaging 2006;23:470.)
200 nm in diameter; a strand of DNA is approximately 2 nm in
diameter; and the smallest molecule, H2, is less than 1/10 of 1 nm
in size. According to the National Nanotechnology Initiative,
nanoparticles are those with a diameter ranging from 1 to 100 nm
optical imaging modalities, current magnetic resonance-derived (within the biomedical community, slightly larger particles are
retinal imaging only allows delineation of three to four distinct often defined as nanoparticles owing to a similarity in size to
retinal layers. Using gadolinium for contrast provides increased important naturally occurring agents, such as viruses).69 At these
signal from the retinal and choroidal vasculature and thus aids dimensions, nanoparticles show unique properties that seem
in correlation of MRI retinal scans with histological sections (the surprising but are, in fact, attributable to the principles of
avascular photoreceptor layers do not show any enhancements quantum mechanics. As a result of these effects, slight deviations
using this method). Manganese has also been used as a contrast in the size, shape, and organization of nanoparticles can have
agent to improve the anatomic contrast between layers; using profound effects on their properties.
this approach, it has been possible to reveal seven distinct retinal The engineering of nanomaterials and/or devices, and their
bands of alternating hypo- and hyperintensity.63 application in “nanomedicine,” are likely to alter profoundly our
In 2008, the first report of retinal blood flow assessment in rats, approach to disease, with significant advances in biopharmaceu-
using MRI, was published.65 In this study, arterial spin labeling ticals (e.g., drug design and delivery), implantable materials and
was used to quantify basal blood flow levels and their responses devices (e.g., tissue regeneration scaffolds), and diagnostic tools
to physiological stimulation. With the improvements in spatial (e.g., genetic testing and imaging).68,69 In ophthalmology, the use
resolution afforded by new MRI devices, visualization of both of nanoparticles shows particular promise for use as contrast
retinal and choroidal blood flow has become possible. In animals, agents in retinal imaging. Nanoparticles can be “functionalized”
using BOLD fMRI techniques, differential responses of the (i.e., conjugated with targeting ligands) to facilitate their precise
retinal and choroidal circulations to physiological stimuli (e.g., and specific targeting. During their synthesis, the properties of
hyperoxia versus hypercapnia) have been demonstrated.63 In nanoparticles can also be finely tuned for use with multiple
animal studies, BOLD fMRI responses have also been assessed imaging modalities. Finally, the biocompatibility of many
in response to visual stimuli. The results of these studies suggest nanoparticles has been established and, thus, the potential exists
evidence that retinal vessels were very responsive to visual for their translation to clinical settings. In this section, we provide
stimulation but choroidal vessels only showed small percentage an overview of the properties and translation imaging potential
changes. for a number of nanoparticle groups.

Conclusions Iron oxide nanoparticles


Translation of retinal MRI from animal studies to human research Magnetic nanoparticles are particularly attractive for magnetic
and clinical practice faces a number of obstacles. The MRI scan- resonance-derived imaging.68,69 MRI is based on the behavior,
ners available in clinical environments currently have limited alignment, and interaction of protons in the presence of a mag-
spatial resolutions and low signal-to-noise ratios. In addition, netic field (see above). Within a strong magnetic field, protons
the issue of eye movements in awake humans is a major limiting are perturbed; magnetic nanoparticles can then be used to alter
factor. As a first step in addressing these issues, the feasibility their longitudinal (T1) or transverse (T2) relaxation times and,
of multimodal MRI has recently been tested on anesthetized thus, image contrast generated. Superparamagnetic iron oxide
(SPIO) nanoparticles have large magnetic moments and are well gold shell. Unlike colloidal gold, the optical resonance of these
suited as T2 contrast agents in this context. A significant benefit nanoshells can be precisely and systematically varied over a
associated with SPIO nanoparticles is their biocompatibility and broad range, extending from the near-ultraviolet to the mid- 147
ready detection at moderate concentrations; as a result, SPIO infrared. Thus, gold nanoshells can be designed and synthesized
nanoparticles have been approved by the Food and Drug to demonstrate light-scattering peaks in the near-infrared region

Chapter 5
Administration for use in clinical practice and are now commer- commonly utilized by ophthalmic imaging systems such as
cially available (e.g., ferucarbotran: Resovist, Bayer, Germany). OCT. Conversely, gold nanoshells with large absorption cross-
sections have been used for photothermal destruction of cancer
Gold nanoparticles cells, and may be useful in the future for photoacoustic ophthal-
Due to the phenomenon of localized surface plasmon reso- moscopy.74 In addition to their tunable spectral characteristics,
nance – the collective oscillation of their conduction electrons and their putative biocompatibility, the same conjugation proto-
in the presence of an incident light – gold nanoparticles can cols used to functionalize colloidal gold can also be used for gold

Advanced Imaging Technologies


show strong extinction peaks in the visible and near-infrared nanoshells. In addition to spherical gold nanoparticles and gold
regions of the electromagnetic spectrum (in simple terms, they nanoshells, gold nanorods and gold nanocages have also been
are capable of scattering or absorbing large amounts of light investigated as contrast agents for optical imaging in preclinical
when illuminated). As a result, gold nanoparticles are par- settings.75
ticularly well suited for use as contrast agents in optical
imaging.70 Quantum dots
Colloidal gold (suspensions of gold nanoparticles in fluid) has Quantum dots are fluorescent nanocrystals, 1–100 nm in size,
been used for hundreds of years to impart vibrant colors to the with unique optical and electrical properties, and with a syn-
stained-glass windows of Gothic churches, and for many decades thetic history of approximately 20 years.68,69 Quantum dots com-
in the treatment of patients with rheumatoid arthritis.68 Since monly consist of a metalloid crystalline core (e.g., cadmium
1971, when Faulk and Taylor71 invented the immunogold staining selenide) surrounded by a shell (e.g., zinc sulfide). The fluores-
procedure, colloidal gold has also been widely used in laboratory cent properties of quantum dots offer a number of advantages
settings, with the optical (and electron beam) contrast qualities for their use in optical imaging. The brightness of quantum dots
of gold providing excellent detection qualities for techniques is 10–100 times greater than most organic dyes or proteins.
such as immunoblotting, flow cytometry, and hybridization Quantum dots also show broad absorption characteristics with
assays.72 More recently, the creation of a new form of nanoparticle narrow-emission spectra that are continuous and tunable due to
– gold nanoshells – has reignited interest in gold nanoparticles as quantum size effects. Quantum dots also possess a long fluores-
contrast agents for clinical imaging (Fig. 5.12).73 Gold nanoshells cence lifetime and undergo negligible photobleaching. Most
consist of a dielectric core (typically silica), surrounded by a thin importantly perhaps, quantum dots can be labeled to allow

20nm 10nm BV Spot Magn


Extinction (Arb. units)

7nm Acc. Det WD 200 nm


5nm 30.0 kV 3.0 120000x SE 11.4 Hivac

Fig. 5.12 Gold nanoparticles, such as nanoshells, may be of use as


contrast agents for clinical imaging. (A) Visual demonstration of the
tunability of metal nanoshells. (Reproduced with permission from Loo
C, Lin A, Hirsch L, et al. Nanoshell-enabled photonics-based imaging
and therapy of cancer. Technol Cancer Res Treat 2004;3:34.)
500 600 700 800 900 1000 1100 1200 (B) Scanning electron microscopy image of nanoshell with 291-nm
Wavelength, nm core diameter and 15-nm shell thickness. (Reproduced with permission
from Agrawal A, Huang S, Wei Haw Lin A, et al. Quantitative
evaluation of optical coherence tomography signal enhancement with
gold nanoshells. J Biomed Opt 2006;11:041121–2.) (C) Optical
resonances of gold shell-silica core nanoshells as a function of their
core/shell ratio. Respective spectra correspond to the nanoparticles
60nm core radius 60nm core radius depicted beneath. (Reproduced with permission from Loo C, Lin A,
C Hirsch L, et al. Nanoshell-enabled photonics-based imaging and
20nm shell 5nm shell
therapy of cancer. Technol Cancer Res Treat 2004;3:34.)
Qdot 800
Qdot 565 Qdot 605
148 Qdot 705 Qdot 655
Section 1

Neck LNs

Neck LNs

Lat thoracic LN
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Lat. Thoracic
LNs Axillary LN
Axillary
LNs
Schema
White light imaging White + 5 colors

Qdot 565 (blue) Qdot 605 (yellow)

Qdot 655 (green) Qdot 705 (magenta)

In vivo imaging Qdot 800 (red)

Fig. 5.13 In vivo and intrasurgical spectral fluorescence imaging of a mouse injected with five-carboxyl quantum dots (565, blue; 605, yellow;
655, green; 705, magenta; 800, red), intracutaneously into the middle digits of the bilateral upper extremities, the bilateral ears, and at the
median chin, as shown in the schema. Five primary draining lymph nodes were simultaneously visualized with different colors through the skin
in the in vivo image and are more clearly seen in the image taken at the surgery. (Reproduced with permission from Kobayashi H, Hama Y,
Koyama Y, et al. Simultaneous multicolor imaging of five different lymphatic basins using quantum dots. Nano Lett 2007;7:1714.)

precise targeting of cellular structures. As a result of these fea- clinical trials of gold nanoparticle therapy in humans provide
tures, quantum dots are increasingly being used for in vivo grounds for optimism in this regard.
imaging in animal studies (Fig. 5.13), although concerns about
cytotoxicity must be addressed before they will be suitable for
CONCLUSIONS AND FUTURE DIRECTIONS
use in humans.
In the past 25 years, advances in retinal imaging have revo-
Conclusions lutionized the diagnosis and management of retinal disease.
A large number of other nanoparticle groups are currently being As recently as 1990, the conventional wisdom held that axial
investigated for their biomedical potential, with examples image resolution was fundamentally constrained by geometric
including carbon nanotubes, dendrimers, perfluorocarbons, and optics and the depth of focus.6 However, with the advent of
lipid-based nanoparticles.69 The unique and tunable optical OCT, axial resolution has now been improved 1000-fold over
properties of many nanoparticles, along with their small size and that previously thought possible. In the short to medium term,
capacity for cellular targeting, make them strong candidates for continued advances in OCT will be coupled with advances
use as contrast agents in retinal imaging. Using these agents in in adaptive optics technology to provide unprecedented non-
combination with techniques such as OCT may ultimately allow invasive cellular imaging. In parallel with this, functional
visualization of many retinal structures (e.g., Müller cells) and extensions of these and other imaging modalities will provide
cellular processes (e.g., apoptosis) in clinical practice. While such greatly enhanced information regarding parameters such as
usage has yet to be demonstrated in humans, the previous com- retinal blood flow and oxygenation. Increasing use of nano-
mercialization of magnetic nanoparticles for MRI and the early technology may provide “molecular” imaging capabilities, and
allow evaluation of biochemical processes such as apoptosis. 21. Roorda A, Zhang Y, Duncan JL. High-resolution in vivo imaging of the RPE
mosaic in eyes with retinal disease. Invest Ophthalmol Vis Sci 2007;48:
In the longer term, a number of fundamental limits will need 2297–303.
to be overcome, including: (1) constraints imposed by maximum 22. Gray DC, Merigan W, Wolfing JI, et al. In vivo fluorescence imaging of primate 149
retinal ganglion cells and retinal pigment epithelial cells. Optics Express
light exposure that can be delivered safely to the eye; (2) 2006;14:7144–58.
windows of spectral transmittance imposed by the cornea and 23. Martin J, Roorda A. Direct and noninvasive assessment of parafoveal capillary
leukocyte velocity. Ophthalmology 2005;112:2219–24.

Chapter 5
lens; and (3) diffraction limits imposed by the wave nature
24. Zhong Z, Petrig BL, Qi X, et al. In vivo measurement of erythrocyte velocity
of light.6 While many of these barriers seem impenetrable, and retinal blood flow using adaptive optics scanning laser ophthalmoscopy.
early breakthroughs have already taken place in each area. In Optics Express 2008;16:12746–56.
25. Doble N. High-resolution, in vivo retinal imaging using adaptive optics
particular, the diffraction limit has already been surpassed in and its future role in ophthalmology. Expert Rev Med Devices 2005;2:
the field of microscopy,75 and the use of such techniques may 205–16.
26. Gray DC, Wolfe R, Gee BP, et al. In vivo imaging of the fine structure of
allow a leap forward to much smaller spatial scales in future rhodamine-labeled macaque retinal ganglion cells. Invest Ophthalmol Vis Sci
retinal imaging. 2008;49:467–73.

Advanced Imaging Technologies


27. Wolfing JI, Chung M, Carroll J, et al. High-resolution retinal imaging of cone–
rod dystrophy. Ophthalmology 2006;113:1019 e1011.
Disclosure 28. Duncan JL, Talcott KE, Ratnam K, et al. Cone structure in retinal degeneration
Dr Keane has received a proportion of his funding from the UK associated with mutations in the peripherin/RDS gene. Invest Ophthalmol Vis
Sci 2011;52:1557–66.
Department of Health’s National Institute for Health Research 29. Duncan JL, Zhang Y, Gandhi J, et al. High-resolution imaging with adaptive
Biomedical Research Centre for Ophthalmology at Moorfields optics in patients with inherited retinal degeneration. Invest Ophthalmol Vis
Sci 2007;48:3283–91.
Eye Hospital and University College London Institute of Oph- 30. Carroll J. Adaptive optics retinal imaging: applications for studying retinal
thalmology. The views expressed in the publication are those of degeneration. Arch Ophthalmol 2008;126:857–8.
31. Talcott KE, Ratnam K, Sundquist SM, et al. Longitudinal study of cone photo-
the authors and not necessarily those of the Department of receptors during retinal degeneration and in response to ciliary neurotrophic
Health. factor treatment. Invest Ophthalmol Vis Sci 2011;52:2219–26.
Dr Sadda is a co-inventor of Doheny intellectual property 32. Kitaguchi Y, Bessho K, Yamaguchi T, et al. In vivo measurements of cone
photoreceptor spacing in myopic eyes from images obtained by an adaptive
related to optical coherence tomography that has been licensed optics fundus camera. Jpn J Ophthalmol 2007;51:456–61.
by Topcon Medical Systems, and is a member of the scientific 33. Carroll J, Neitz M, Hofer H, et al. Functional photoreceptor loss revealed
with adaptive optics: an alternate cause of color blindness. Proc Natl Acad Sci
advisory board for Heidelberg Engineering. Dr Sadda also U S A 2004;101:8461–6.
receives research support from Carl Zeiss Meditec, Optos, and 34. Marmor MF, Choi SS, Zawadzki RJ, et al. Visual insignificance of the foveal
pit: reassessment of foveal hypoplasia as fovea plana. Arch Ophthalmol
Optovue, Inc. 2008;126:907–13.
35. Joeres S, Jones SM, Chen DC, et al. Retinal imaging with adaptive optics
scanning laser ophthalmoscopy in unexplained central ring scotoma. Arch
REFERENCES Ophthalmol 2008;126:543–7.
1. Keane PA, Sadda SR. Imaging chorioretinal vascular disease. Eye (Lond) 36. Choi SS, Zawadzki RJ, Keltner JL, et al. Changes in cellular structures
2010;24:422–7. revealed by ultra-high resolution retinal imaging in optic neuropathies.
2. Yannuzzi LA, Ober MD, Slakter JS, et al. Ophthalmic fundus imaging: today Invest Ophthalmol Vis Sci 2008;49:2103–19.
and beyond. Am J Ophthalmol 2004;137:511–24. 37. Ciulla TA, Regillo CD, Harris A. Retina and optic nerve imaging. Philadelphia:
3. Bennett TJ, Barry C. Ophthalmic imaging today: an ophthalmic photographer’s Lippincott Williams & Wilkins; 2003.
viewpoint – a review. Clin Exp Ophthalmol 2009;37:2–13. 38. Williamson TH, Harris A. Ocular blood flow measurement. Br J Ophthalmol
4. Yannuzzi LA. Indocyanine green angiography: a perspective on use in the 1994;78:939–45.
clinical setting. Am J Ophthalmol 2011;151:745–51. 39. Schmetterer L, Garhofer G. How can blood flow be measured? Surv
5. Roorda A. Adaptive optics ophthalmoscopy. J Refract Surg 2000;16:S602–607. Ophthalmol 2007;52(Suppl 2):S134–138.
6. Williams DR. Imaging single cells in the living retina. Vision Res 2011;51: 40. Harris A, Chung H, Ciulla T, et al. Progress in measurement of ocular blood
1371–96. flow and relevance to our understanding of glaucoma and age-related macular
7. Liang J, Williams DR, Miller DT. Supernormal vision and high-resolution degeneration. Prog Retin Eye Res 1999;18:669–87.
retinal imaging through adaptive optics. J Opt Soc Am A 1997;14: 41. Keyser BJ, Flaharty PM, Sergott RC, et al. Color Doppler imaging of arterial
2884–92. blood flow in central retinal vein occlusion. Ophthalmology 1994;101:
8. Kitaguchi Y, Fujikado T, Bessho K, et al. Adaptive optics fundus camera 1357–61.
to examine localized changes in the photoreceptor layer of the fovea. 42. Ho AC, Lieb WE, Flaharty PM, et al. Color Doppler imaging of the ocular
Ophthalmology 2008;115:1771–7. ischemic syndrome. Ophthalmology 1992;99:1453–62.
9. Roorda A, Romero-Borja F, Donnelly W III. Adaptive optics scanning laser 43. Feke G, Yoshida A. Laser based instruments for ocular blood flow assessment.
ophthalmoscopy. Optics Express 2002;10:405–12. J Biomed Optics 1998;3:415–22.
10. Drexler W, Fujimoto JG. State-of-the-art retinal optical coherence tomography. 44. Yoshida A, Feke GT, Mori F, et al. Reproducibility and clinical application
Progr Retinal eye res 2008;27:45–88. of a newly developed stabilized retinal laser Doppler instrument. Am J
11. Miller DT, Kocaoglu OP, Wang Q, et al. Adaptive optics and the eye (super Ophthalmol 2003;135:356–61.
resolution OCT). Eye (Lond) 2011;25:321–30. 45. Sehi M. Basic technique and anatomically imposed limitations of confocal scan-
12. Fernández EJ, Hermann B, Povazay B, et al. Ultrahigh resolution optical coher- ning laser Doppler flowmetry at the optic nerve head level. Acta Ophthalmol
ence tomography and pancorrection for cellular imaging of the living human 2011;89:e1–11.
retina. Optics Express 2008;16:11083–94. 46. Huang D, Swanson E, Lin C, et al. Optical coherence tomography. Science
13. Zawadzki RJ, Cense B, Zhang Y, et al. Ultrahigh-resolution optical coherence 1991;254:1178–81.
tomography with monochromatic and chromatic aberration correction. 47. Mordant DJ, Al-Abboud I, Muyo G, et al. Spectral imaging of the retina.
Optics Express 2008;16:8126–43. Eye (Lond) 2011;25:309–20.
14. Poonja S, Patel S, Henry L, et al. Dynamic visual stimulus presentation in 48. Harris A, Dinn RB, Kagemann L, et al. A review of methods for human retinal
an adaptive optics scanning laser ophthalmoscope. J Refract Surg 2005;21: oximetry. Ophthalm Surg Lasers Imaging 2003;34:152–64.
S575–580. 49. Hickam JB, Sieker HO, Frayser R. . Studies of retinal circulation and A-V
15. Vogel CR, Arathorn DW, Roorda A, et al. Retinal motion estimation in oxygen difference in man. Trans Am Clin Climatol Assoc 1959;71:34–44.
adaptive optics scanning laser ophthalmoscopy. Optics Express 2006;14: 50. Beach JM, Schwenzer KJ, Srinivas S, et al. Oximetry of retinal vessels by dual-
487–97. wavelength imaging: calibration and influence of pigmentation. J Appl Physiol
16. Arathorn DW, Yang Q, Vogel CR, et al. Retinally stabilized cone-targeted 1999;86:748–58.
stimulus delivery. Optics Express 2007;15:13731–44. 51. Hardarson SH, Harris A, Karlsson RA, et al. Automatic retinal oximetry.
17. Miller DT, Williams DR, Morris GM, et al. Images of cone photoreceptors in Invest Ophthalmol Vis Sci 2006;47:5011–6.
the living human eye. Vision Res 1996;36:1067–79. 52. Mordant DJ, Al-Abboud I, Muyo G, et al. Validation of human whole
18. Roorda A, Williams DR. The arrangement of the three cone classes in the living blood oximetry, using a hyperspectral fundus camera with a model eye.
human eye. Nature 1999;397:520–2. Invest Ophthalmol Vis Sci 2011;52:2851–9.
19. Chui TY, Song H, Burns SA. Individual variations in human cone photorecep- 53. Schweitzer D, Hammer M, Kraft J, et al. In vivo measurement of the oxygen
tor packing density: variations with refractive error. Invest Ophthalmol Vis Sci saturation of retinal vessels in healthy volunteers. IEEE Trans Biomed Eng
2008;49:4679–87. 1999;46:1454–65.
20. Doble N, Choi SS, Codona JL, et al. In vivo imaging of the human rod photo- 54. Hardarson SH, Stefánsson E. Oxygen saturation in central retinal vein
receptor mosaic. Optics lett 2011;36:31–3. occlusion. Am J Ophthalmol 2010;150:871–5.
55. Hardarson SH, Stefánsson E. Oxygen saturation in branch retinal vein 66. Zhang Y, Wey HY, Nateras OS, et al. Anatomical, blood oxygenation
occlusion. Acta Ophthalmol 2011. Epub ahead of print April 21, 2011; doi: level-dependent, and blood flow MRI of nonhuman primate (baboon) retina.
10.1111/j.1755-3768.2011.02109.x. Magn Resonance Med 2011;66:546–54.
150 56. Hardarson SH, Gottfredsdottir MS, Halldorsson GH, et al. Glaucoma filtration 67. Zhang Y, Peng Q, Kiel JW, et al. Magnetic resonance imaging of vascular
surgery and retinal oxygen saturation. Invest Ophthalmol Vis Sci 2009;50: oxygenation changes during hyperoxia and carbogen challenges in the human
5247–50. retina. Invest Ophthalmol Vis Sci 2011;52:286–91.
57. Hammer M, Vilser W, Riemer T, et al. Diabetic patients with retinopathy 68. Zarbin MA, Montemagno C, Leary JF, et al. Nanomedicine in ophthalmology:
show increased retinal venous oxygen saturation. Graefe’s Arch Clin Exp the new frontier. Am J Ophthalmol 2010;150:144–62.
Section 1

Ophthalmol 2009;247:1025–30. 69. Nune SK, Gunda P, Thallapally PK, et al. Nanoparticles for biomedical imaging.
58. Tiedeman JS, Kirk SE, Srinivas S, et al. Retinal oxygen consumption during Expert Opin Drug Deliv 2009;6:1175–94.
hyperglycemia in patients with diabetes without retinopathy. Ophthalmology 70. Arvizo R, Bhattacharya R, Mukherjee P. Gold nanoparticles: opportunities and
1998;105:31–6. challenges in nanomedicine. Expert Opin Drug Deliv 2010;7:753–63.
59. Jiao S, Jiang M, Hu J, et al. Photoacoustic ophthalmoscopy for in vivo retinal 71. Faulk WP, Taylor GM. An immunocolloid method for the electron microscope.
imaging. Optics Express 2010;18:3967–72. Immunochemistry 1971;8:1081–3.
60. Jiao S, Xie Z, Zhang HF, et al. Simultaneous multimodal imaging with 72. Roth J. The silver anniversary of gold: 25 years of the colloidal gold marker
integrated photoacoustic microscopy and optical coherence tomography. system for immunocytochemistry and histochemistry. Histochem Cell Biol
Optical Imaging Technologies

Optics Lett 2009;34:2961–3. 1996;106:1–8.


Retinal Imaging and Diagnostics

61. Zhang HF, Maslov K, Stoica G, et al. Functional photoacoustic microscopy for 73. Loo C, Lin A, Hirsch L, et al. Nanoshell-enabled photonics-based imaging and
high-resolution and noninvasive in vivo imaging. Nature Biotechnol 2006;24: therapy of cancer. Technol Cancer Res Treat 2004;3:33–40.
848–51. 74. Gobin AM, Lee MH, Halas NJ, et al. Near-infrared resonant nanoshells for
62. Zhang X, Jiang M, Fawzi AA, et al. Simultaneous dual molecular contrasts combined optical imaging and photothermal cancer therapy. Nano Lett
provided by the absorbed photons in photoacoustic microscopy. Optics lett 2007;7:1929–34.
2010;35:4018–20. 75. Murphy CJ, Gole AM, Stone JW, et al. Gold nanoparticles in biology: beyond
63. Duong TQ. Magnetic resonance imaging of the retina: a brief historical and toxicity to cellular imaging. Acc Chem Res 2008;41:1721–30.
future perspective. Saudi J Ophthalmol 2011;25:137–43. 76. Betzig E, Patterson GH, Sougrat R, et al. Imaging intracellular fluorescent
64. Duong TQ, Muir ER. Magnetic resonance imaging of the retina. Jpn J proteins at nanometer resolution. Science 2006;313:1642–5.
Ophthalmol 2009;53:352–67.
65. Li Y, Cheng H, Duong TQ. Blood-flow magnetic resonance imaging of the
retina. Neuroimage 2008;39:1744–51.
Optical Imaging Technologies Section 1

Image Processing Chapter

Michael Abràmoff, Christine N. Kay


6 

INTRODUCTION
In this review, we discuss quantitative approaches to retinal
image analysis. Special emphasis is placed on familiarizing the
reader with basic concepts in imaging and image analysis.
Fundus and optical coherence tomography (OCT) image analy-
sis are reviewed as well as the use of these modalities in provid-
ing comprehensive descriptions of retinal morphology and
function. We discuss screening-motivated computer-aided
detection of retinal lesions as well as translational clinical appli-
cations in diagnosis and therapy.
After reading this chapter the reader should be able to under-
stand concepts in retinal image analysis, and critically review
the clinical impact of the research in this field.

HISTORY OF RETINAL IMAGING


The optical properties of the eye that allow image formation
prevent direct inspection of the retina. Though existence of the
red reflex has been known for centuries, special techniques are
needed to obtain a focused image of the retina. The first attempt
to image the retina, in a cat, was completed by the French physi-
cian Jean Mery, who showed that if a live cat is immersed in
water, its retinal vessels are visible from the outside.1 The Fig. 6.1 First known image of human retina, as drawn by van Trigt
in 1853. (Reproduced from Trigt AC. Dissertatio ophthalmologica
impracticality of such an approach for humans led to the inven- inauguralis de speculo oculi. Utrecht: Universiteit
tion of the principles of the ophthalmoscope in 1823 by Czech van Utrecht, 1853.)
scientist Jan Evangelista Purkynĕ (frequently spelled Purkinje)
and its reinvention in 1845 by Charles Babbage.2,3 Finally, the The initial approach to depict the three-dimensional (3D)
ophthalmoscope was reinvented yet again and reported by von shape of the retina was stereo fundus photography, as first
Helmholtz in 1851.4 Thus, inspection and evaluation of the retina described by Allen in 1964, where multiangle images of the
became routine for ophthalmologists, and the first images of the retina are combined by the human observer into a 3D shape.9
retina (Fig. 6.1) were published by the Dutch ophthalmologist Subsequently, confocal scanning laser ophthalmoscopy (SLO)
van Trigt in 1853.5 The first useful photographic images of the was developed, using the confocal aperture to obtain multiple
retina, showing blood vessels, were obtained in 1891 by the images of the retina at different confocal depths, yielding esti-
German ophthalmologist Gerloff.6 In 1910, Gullstrand devel- mates of 3D shape. However, the optics of the eye limit the depth
oped the fundus camera, a concept still used to image the retina resolution of confocal imaging to approximately 100 µm, which
today7; he later received the Nobel Prize for this invention. is poor when compared with the typical 300–500 µm thickness
Because of its safety and cost-effectiveness at documenting of the whole retina.10
retinal abnormalities, fundus imaging has remained the primary OCT, first described in 1987 as a method for time-of-flight
method of retinal imaging. measurement of the depth of mechanical structures,11,12 was later
In 1961, Novotny and Alvis published their findings on extended to a tissue-imaging technique. This method of deter-
fluorescein angiographic imaging.8 In this imaging modality, mining the position of structures in tissue, described by Huang
a fundus camera with additional narrow-band filters is used et al. in 1991,13 was termed OCT. In 1993 in vivo retinal OCT was
to image a fluorescent dye injected into the bloodstream that accomplished for the first time.14 Today, OCT has become a
binds to leukocytes. It remains widely used, because it allows prominent biomedical tissue-imaging technique, especially in
an understanding of the functional state of the retinal the eye, because it is particularly suited to ophthalmic applica-
circulation. tions and other tissue imaging requiring micrometer resolution.
HISTORY OF RETINAL IMAGE PROCESSING 6. fluorescein angiography and indocyanine angiography:
image intensities represent the amounts of emitted photons
152 Matsui et al. were the first to publish a method for retinal image from the fluorescein or indocyanine green fluorophore that
analysis, primarily focused on vessel segmentation.15 Their was injected into the subject’s circulation.
approach was based on mathematical morphology and they
used digitized slides of fluorescein angiograms of the retina. In There are several technical challenges in fundus imaging. Since
Section 1

the following years, there were several attempts to segment the retina is normally not illuminated internally, both external
other anatomical structures in the normal eye, all based on digi- illumination projected into the eye as well as the retinal image
tized slides. The first method to detect and segment abnormal projected out of the eye must traverse the pupillary plane.
structures was reported in 1984, when Baudoin et al. described Thus the size of the pupil, usually between 2 and 8  mm in
an image analysis method for detecting microaneurysms, a char- diameter, has been the primary technical challenge in fundus
acteristic lesion of diabetic retinopathy (DR).16 Their approach imaging.7 Fundus imaging is complicated by the fact that the
Optical Imaging Technologies
Retinal Imaging and Diagnostics

was also based on digitized angiographic images. They detected illumination and imaging beams cannot overlap because such
microaneurysms using a “top-hat” transform, a step-type digital overlap results in corneal and lenticular reflections diminishing
image filter.17 This method employs a mathematical morphology or eliminating image contrast. Consequently, separate paths
technique that eliminates the vasculature from a fundus image are used in the pupillary plane, resulting in optical apertures
yet leaves possible microaneurysm candidates untouched. The on the order of only a few millimeters. Because the resulting
field dramatically changed in the 1990s with the development imaging setup is technically challenging, fundus imaging his-
of digital retinal imaging and the expansion of digital filter- torically involved relatively expensive equipment and highly
based image analysis techniques. These developments resulted trained ophthalmic photographers. Over the last 10 years or
in an exponential rise in the number of publications, which so, there have been several important developments that have
continues today. made fundus imaging more accessible, resulting in less depen-
dence on such experience and expertise. There has been a shift
CURRENT STATUS OF RETINAL IMAGING from film-based to digital image acquisition, and as a conse-
Retinal imaging has developed rapidly during the last 160 years quence the importance of picture archiving and communication
and is a now a mainstay of the clinical care and management of systems (PACS) has substantially increased in clinical ophthal-
patients with retinal as well as systemic diseases. Fundus photog- mology, also allowing integration with electronic medical
raphy is widely used for population-based, large-scale detection records. Requirements for population-based early detection of
of DR, glaucoma, and age-related macular degeneration. OCT retinal diseases using fundus imaging have provided the incen-
and fluorescein angiography are widely used in the daily man- tive for effective and user-friendly imaging equipment. Opera-
agement of patients in a retina clinic setting. OCT has also become tion of fundus cameras by nonophthalmic photographers has
an increasingly helpful adjunct in preoperative planning and become possible due to nonmydriatic imaging, digital imaging
postoperative evaluation of vitreoretinal surgical patients.18 The with near-infrared focusing, and standardized imaging pro­
overview below is partially based on an earlier review paper.19 tocols to increase reproducibility.
Though standard fundus imaging is widely used, it is not suit-
able for retinal tomography, because of the mixed backscatter
FUNDUS IMAGING caused by the semitransparent retinal layers.
We define fundus imaging as the process whereby reflected light
is used to obtain a two-dimensional (2D) representation of the OPTICAL COHERENCE
3D, semitransparent, retinal tissues projected on to the imaging
plane. Thus, any process that results in a 2D image where the
TOMOGRAPHY IMAGING
image intensities represent the amount of a reflected quantity of OCT is a noninvasive optical medical diagnostic imaging modal-
light is fundus imaging. Consequently, OCT imaging is not ity which enables in vivo cross-sectional tomographic visualiza-
fundus imaging, while the following modalities/techniques all tion of the internal microstructure in biological systems. OCT is
belong to the broad category of fundus imaging: analogous to ultrasound B-mode imaging, except that it mea-
sures the echo time delay and magnitude of light rather than
1. fundus photography (including so-called red-free sound, therefore achieving unprecedented image resolutions
photography): image intensities represent the amount of (1–10 µm).20 OCT is an interferometric technique, typically
reflected light of a specific waveband employing near-infrared light. The use of relatively long-
2. color fundus photography: image intensities represent wavelength light with a very wide-spectrum range allows OCT
the amount of reflected red (R), green (G), and blue (B) to penetrate into the scattering medium and achieve micrometer
wavebands, as determined by the spectral sensitivity of the resolution.
sensor The principle of OCT is based upon low-coherence interfer-
3. stereo fundus photography: image intensities represent the ometry, where the backscatter from more outer retinal tissues
amount of reflected light from two or more different view can be differentiated from that of more inner tissues, because it
angles for depth resolution takes longer for the light to reach the sensor. Because the differ-
4. SLO: image intensities represent the amount of reflected ences between the most superficial and the deepest layers in the
single-wavelength laser light obtained in a time sequence retina are around 300–400 µm, the difference in time of arrival
5. adaptive optics SLO: image intensities represent the amount is very small and requires interferometry to measure.21
of reflected laser light optically corrected by modeling the The principle of low coherence, or low correlation, means that
aberrations in its wavefront the light coming from the light source is only correlating for
a short amount of time. In other words, the autocorrelation func- each other at a moment in time, the higher the interference;
tion of the light wave is only large for a short duration, and at remember that, after splitting, each carried the same low coher-
all other times it is essentially zero. If the light is fully coherent, ence “label.” Because the optical properties of the eye add noise 153
the autocorrelation is high forever, and it becomes impossible to and thus slightly change the reflected reference arm light wave,
create an interference pattern and determine when the light was the interference will never be perfect. Though the coherence

Chapter 6
emitted; if the light was entirely incoherent, there would be no pattern or label changes continuously over time, once they are
interference at all. A smaller coherence duration thus results in split they have the same “label” (but change rapidly over time),
a better depth resolution, but at lower intensity. so that the interference will be high as long as the reference and
Thus, the low coherence of the light essentially “labels,” with sample distances stay the same. The energy or envelope of the
its autocorrelogram, each short duration of the light wave, with interferogram is measured as intensity at the sensor and is then
the next duration having a different “label.” Though we use the displayed as the OCT signal intensity. Of course, by changing
term “label,” it is important to understand that the light wave is the position of the mirror, we can “interrogate” the amount of

Image Processing
actually continuous and not pulsed. interference at different sample tissue depths.
This label uniquely indicates when reflected light was emitted. We see the importance of the choice of a good low-coherence
The low coherent light is optically split into two bundles, called source – with either an incoherent or fully coherent source, inter-
arms, before being sent into the eye. One arm, the reference arm, ferometry is impossible. Such light can be generated by using
is aimed at a mirror with a known distance, and thereby reflected; superluminescent diodes (superbright light-emitting diodes) or
the other, the sample arm, is sent into the eye and reflects back lasers with extremely short pulses, femtosecond lasers. The
from the different tissues, at yet unknown depth. optical setup typically consists of a Michelson interferometer
If the distance to the mirror is exactly the same as the distance with a low-coherence, broad-bandwidth light source (Fig. 6.2).
to the tissue, and we optically combine the two reflected (refer- By scanning the mirror in the reference arm, as in time domain
ence and sample) arm light waves, their interference will be OCT, modulating the light source, as in swept source OCT, or
nonzero. This is because the more the two light waves resemble decomposing the signal from a broadband source into spectral

Fig. 6.2 Schematic diagram of the operation


of an optical coherence tomography
instrument, emphasizing splitting of the light
in two arms, overlapping train of bursts
“labeled” based on their autocorrelogram,
Retina and their interference after being reflected
from retinal tissue as well as from the
reference mirror (assuming the time delays
of both paths are equal).

t1

Interferometer

Beam t1 =? t2
splitter

t2

Reference Low coherent light source


mirror train of overlapping bursts
components, as in spectral domain OCT (SD-OCT), a reflectivity diffraction grating and a complementary metal oxide semicon-
profile of the sample can be obtained, as measured by the inter- ductor or charged couple device linear sensor. The Fourier trans-
154 ferogram. The reflectivity profile, called an A-scan, contains form is again applied to the spectral correlogram intensities to
information about the spatial dimensions and location of struc- determine the depth of each scatter signal.22 With SD-OCT, tens
tures within the retina. A cross-sectional tomograph (B-scan) of thousands of A-scans can be acquired each second, and thus
may be achieved by laterally combining a series of these axial true 3D imaging is routinely possible. Consequently, 3D OCT is
Section 1

depth scans (A-scan). En face imaging (C-scan) at an acquired now in wide clinical use, and has become the standard of care.
depth is possible depending on the imaging engine used.
Swept source OCT
The transverse resolution of OCT scans (x, y) depends on the
Instead of moving the reference arm, as with time domain OCT
speed and quality of the galvanic scanning mirrors and the
imaging, in swept source OCT the light source is rapidly modu-
optics of the eye, and is typically 20–40  µm. The resolution of
lated over its center wavelength, essentially attaching a second
the A-scans along the z direction depends on the coherence of
Optical Imaging Technologies
Retinal Imaging and Diagnostics

label to the light, its wavelength. A photo sensor is used to


the light source and is currently 4–8  µm in commercially avail-
measure the correlogram for each center wavelength over time.
able scanners. Isotropic (or isometric) means that the size of
A Fourier transform on the multiwavelength or spectral inter-
each imaged element, or voxel, is the same in all three dimen-
ferogram is performed to determine the depth of all tissue scat-
sions. Current commercially available OCT devices routinely
ters at the imaged location.22 With swept source OCT, hundreds
offer voxel sizes of 30 × 30 × 2  µm, achieving isometricity in
of thousands of A-scans can be obtained every second, with
the x–y plane only. Available SD-OCT scanners are never truly
additional increase in scanning density when acquiring 3D
isotropic, because the retinal tissue in each A-scan is sampled
image volumes.
at much smaller intervals in depth than are the distances
between A- and/or B-scans. The resolution in depth, or what AREAS OF ACTIVE RESEARCH IN
we call the z-dimension, is currently always higher than the
resolution in the x–y plane. The primary advantage of x–y iso-
RETINAL IMAGING
tropic imaging when quantifying properties of the retina is that Retinal imaging is rapidly evolving and newly completed
fewer assumptions have to be made about the tissue between research findings are quickly translated into clinical use.
the measured samples, thus potentially leading to more accurate
indices of retinal morphology. Portable, cost-effective fundus imaging
For early detection and screening, the optimal place for position-
Time domain OCT ing fundus cameras is at the point of care: primary care clinics,
With time domain OCT, the reference mirror is moved mechani- public venues (e.g., drug stores, shopping malls). Though the
cally to different positions, resulting in different flight time transition from film-based to digital fundus imaging has revolu-
delays for the reference arm light. Because the speed at which the tionized the art of fundus imaging and made telemedicine appli-
mirror can be moved is mechanically limited, only thousands of cations feasible, the current cameras are still too bulky, expensive,
A-scans can be obtained per second. The envelope of the inter- and may be difficult to use for untrained staff in places lacking
ferogram determines the intensity at each depth.13 The ability to ophthalmic imaging expertise. Several groups are attempting to
image the retina two-dimensionally and three-dimensionally create more cost-effective and easier-to-use handheld fundus
depends on the number of A-scans that can be acquired over cameras, employing a variety of technical approaches.23,24
time. Because of motion artifacts such as saccades, safety require-
ments limiting the amount of light that can be projected on to the
Functional imaging
retina, and patient comfort, 1–3 seconds per image or volume is For the patient as well as for the clinician, the outcome of disease
essentially the limit of acceptance. Thus, the commercially avail- management is mainly concerned with the resulting organ func-
able time domain OCT, which allowed collecting of up to 400 tion, not its structure. In ophthalmology, current functional
A-scans per second, has not yet been suitable for 3D imaging. testing is mostly subjective and patient-dependent, such as
assessing visual acuity and utilizing perimetry, which are all
Frequency domain OCT psychophysical metrics. Among more recently developed “objec-
tive” techniques, oxymetry is a hyperspectral imaging technique
In frequency domain OCT, broadband interference is acquired
in which multispectral reflectance is used to estimate the con-
with spectrally separated detectors, either by encoding the
centration of oxygenated and deoxygenated hemoglobin in the
optical frequency in time with a spectrally scanning source or
retinal tissue.25 The principle allowing the detection of such dif-
with a dispersive detector, like a grating and a linear detector
ferences is simple: deoxygenated hemoglobin reflects longer
array. The depth scan can be immediately calculated by Fourier
wavelengths better than does oxygenated hemoglobin. Never-
transform from the acquired spectra, without movement of the
theless, measuring absolute oxygenation levels with reflected
reference arm. This feature improves imaging speed dramati-
light is difficult because of the large variety in retinal reflection
cally, while the reduced losses during a single scan improve the
across individuals and the variability caused by the imaging
signal to noise proportional to the number of detection elements.
process. The retinal reflectance can be modeled by a system of
The parallel detection at multiple-wavelength ranges limits the
equations, and this system is typically underconstrained if this
scanning range, while the full spectral bandwidth sets the axial
variability is not accounted for adequately. Increasingly sophis-
resolution.
ticated reflectance models have been developed to correct for the
Spectral domain OCT underlying variability, with some reported success.26 Near-
A broadband light source is used, broader than in time domain infrared fundus reflectance in response to visual stimuli is
OCT, and the interferogram is decomposed spectrally using a another way to determine the retinal function in vivo and has
been successful in cats. Initial progress has also been demon- to an office visit for assessing DR and have been suggested as
strated in humans.27 an approach to make the dilated eye exam available to unserved
and underserved populations that do not receive regular exams 155
Adaptive optics by eye care providers.38,39 If all of these underserved popula-
The optical properties of the normal eye result in a point spread tions were to be provided with digital imaging, the annual
function width approximately the size of a photoreceptor. It is

Chapter 6
number of retinal images requiring evaluation would exceed
therefore impossible to image individual cells or cell structure 32 million in the USA alone (approximately 40% of people
using standard fundus cameras because of aberrations in the with diabetes with at least two photographs per eye).39,40 In
human optical system. Adaptive optics uses mechanically acti- the next decade, projections for the USA are that the average
vated mirrors to correct the wavefront aberrations of the light age will increase, the number of people with diabetes in each
reflected from the retina, and thus has allowed individual pho- age category will increase, and there will be an undersupply
toreceptors to be imaged in vivo.28 Imaging other cells, especially of qualified eye care providers, at least in the near term. Several

Image Processing
the clinically highly important ganglion cells, has thus far been European countries have successfully instigated in their health-
unsuccessful in humans. care systems early detection programs for DR using digital
photography with reading of the images by human experts. In
Longer-wavelength OCT imaging the UK, 1.7 million people with diabetes were screened for DR
3D OCT imaging is now the clinical standard of care for several in 2007–2008. In the Netherlands, over 30 000 people with dia-
eye diseases. The wavelengths around 840 µm used in currently betes were screened since 2001 in the same period, through an
available devices are optimized for imaging of the retina. Deeper early-detection project called EyeCheck.41 The US Department
structures, such as the choroidal vessels, which are important for of Veterans Affairs has deployed a successful photo screening
AMD and other choroidal diseases, and the lamina cribrosa, program through which more than 120 000 veterans were
relevant for glaucomatous damage, are not as well depicted. screened in 2008. While the remote imaging followed by human
Because longer wavelengths penetrate deeper into the tissue, a expert diagnosis approach was shown to be successful for a
major research effort has been undertaken to develop low- limited number of participants, the current challenge is to make
coherence swept source lasers with center wavelengths of the early detection more accessible by reducing the cost and
1000–1300 µm. Prototypes of these devices are already able to staffing levels required, while maintaining or improving DR
resolve detail in the choroid and lamina cribrosa.29 detection performance. This challenge can be met by utilizing
computer-assisted or fully automated methods for detection of
CLINICAL APPLICATIONS OF DR in retinal images.42–44
RETINAL IMAGING Early detection of systemic disease from
The most obvious example of a retinal screening application is fundus photography
retinal disease detection, in which the patient’s retinas are
In addition to detecting DR and age-related macular degenera-
imaged in a remote telemedicine approach. This scenario typi-
tion, it also deserves mention that fundus photography allows
cally utilizes easy-to-use, relatively low-cost fundus cameras,
certain cardiovascular risk factors to be determined. Such metrics
automated analyses of the images, and focused reporting of the
are primarily based on measurement of retinal vessel properties,
results. This screening application has spread rapidly over the
such as the arterial to venous diameter ratio, and indicate the
last few years, and, with the exception of the automated analysis
risk for stroke, hypertension, or myocardial infarct.45,46
functionality, is one of the most successful examples of telemedi-
cine.30 While screening programs exist for detection of glaucoma,
age-related macular degeneration, and retinopathy of prematu-
Image-guided therapy for retinal diseases
rity, the most important screening application focuses on early with 3D OCT
detection of DR. With the introduction of 3D OCT imaging, the wealth of new
information about retinal morphology has enabled its usage for
Early detection of diabetic retinopathy close monitoring of retinal disease status and guidance of retinal
Early detection of DR via population screening associated with therapies. The most obvious example of successful image-guided
timely treatment has been shown to prevent visual loss and management in ophthalmology is its use in diabetic macular
blindness in patients with retinal complications of diabetes.31,32 edema. Currently, OCT imaging is widely used to determine the
Almost 50% of people with diabetes in the USA currently do extent and amount of retinal thickening. More detailed analyses
not undergo any form of regular documented dilated eye exam, of retinal layer morphology and texture from OCT will allow
in spite of guidelines published by the American Diabetes direct image-based treatment to be guided by computer-
Association, the American Academy of Ophthalmology, and supported or automated quantitative analysis. This can be
the American Optometric Association.33 In the UK, a smaller subsequently optimized, allowing a personalized approach to
proportion or approximately 20% of diabetics are not regularly retinal disease treatment to become a reality.
evaluated, as a result of an aggressive effort to increase screen- Another highly relevant example of a disease that will benefit
ing for people with diabetes. Blindness and visual loss can be from image-guided therapy is exudative age-related macular
prevented through early detection and timely management. degeneration. With the advent of the anti-vascular endothelial
There is widespread consensus that regular early detection of growth factor (VEGF) agents ranibizumab and bevacizumab, it
DR via screening is necessary and cost-effective in patients has become clear that outer retinal and subretinal fluid is the
with diabetes.34–37 Remote digital imaging and ophthalmologist main indicator of a need for anti-VEGF retreatment.47–51 Several
expert reading have been shown to be comparable or superior studies are under way to determine whether OCT-based
quantification of fluid parameters and affected retinal tissue can second one, the repeated intensity, 128 (requiring only two bytes
help improve the management of patients with anti-VEGF of storage). To restore the original image area, an uncompression
156 agents. algorithm takes the two elements and reconstitutes the 50 pixels
each having 128 as intensity.
IMAGE ANALYSIS CONCEPTS Because no image information is lost, and the uncompression
algorithm can reconstitute the image perfectly, this is loss-less
Section 1

FOR CLINICIANS
compression.
Image analysis is a field that relies heavily on mathematics and
physics. The goal of this section is to explain the major clini- Lossy image compression
cally relevant concepts and challenges in image analysis, with To improve image compression rates even more, lossy compres-
no use of mathematics or equations. For a detailed explanation sion algorithms make use of the fact that the human visual
of the underlying mathematics, the reader is referred to the system does not notice small intensity changes in the image. A
Optical Imaging Technologies
Retinal Imaging and Diagnostics

appropriate textbooks.52 lossy compression algorithm would compress the image in the
example above in exactly the same manner. However, if we take
The retinal image an image where the 50 pixels in the area did not have exactly the
Definition of a retinal image same value, but varied slightly around the value 128, the image
As interpreted by a computer, an image is a set of elements with compression algorithm would compress the image differently.
values that are organized. The elements, called pixels, each have For the human visual system, this area would be hard to dif-
a single value, the intensity, when the image is a monochrome ferentiate from the same area where all 50 pixels had intensity
or an OCT image; and multiple values, when the image is a color values of 128. The simple loss-less algorithm above would not
image. For example, in an angiogram or OCT image, the inten- be able to compress this area, because the pixels in the area have
sity value of each pixel is the amount of reflected respectively different intensities, and would store the 50 pixels as 50 ele-
interfered light that was measured at that pixel position. In a ments. The lossy algorithm is “smarter” and “knows” the limits
color image, there are usually three intensity values (for red, of human visual perception, and will assign all pixels varying
blue, and green) assigned to a pixel, which combined make up only “a little” from 128 the intensity value of 128, and store the
the color of that pixel. repeat value, and the repeated intensity. The uncompression
algorithm would assign all 50 pixels the same 128 as intensity.
Retinal image quantities Thus the original information in the image is lost, though typi-
Computers use a binary system (1s and 0s) to store and process cally this is not noticeable to the human visual system.
information. Because they do not use the decimal system, image
intensities typically have values ranging between 0 and 255, Legal issues with lossy image compression
0–65 536, or –32 767 to +32 767, instead of the 0–1000 or 100 Lossy compression is widely used in ophthalmic imaging, espe-
000 that one might expect if computers used the decimal system. cially for storing acquired images in image databases (see PACS
This can be explained by the fact that, typically, 1, 2, or 3 bytes section, below). In theory, but so far not in practice, a medicole-
are used to store the intensity values for a pixel, as combina- gal situation could arise as a result of lossy compression artifact.
tions of 1s and 0s. Though more bytes take up more space, the In a hypothetical case where the diagnosis of a clinician is dis-
precision of the intensity values becomes greater. Psychophysi- puted, that clinician may have seen an abnormality on an image
cal research has shown that the human visual system can dif- immediately after acquisition, which subsequently underwent
ferentiate at most 500 different levels of gray, and at most 10 lossy compression, was stored, and thus became part of the
million different colors, so that increasing the precision of the medical record. Because lossy compression causes irreversible
intensity values beyond these levels will not increase the visual loss of information, that abnormality may no longer be visible
perception of quality of an image. However, there may be some on the archived image after uncompression, making it impos-
value in increasing the precision despite this fact, since image sible to view the same image that the clinician originally saw
analysis algorithms can discern a higher number of levels than and upon which his/her diagnosis was based. One can certainly
humans can. envision the legal implications and liability of this scenario.
Examples of loss-less compression image formats are com-
Retinal image compression
pressed TIFF, GIF, and PNG file formats, as well as the “raw”
Image compression is useful because it decreases the amount of formats that are generated directly by the imaging device.
memory required to store images digitally or communicate these Common lossy compression-based image formats are JPEG and
images over a network such as the internet. Image compression MPEG.
can be “loss-less” or “lossy,” and makes use of the fact that
images are always somewhat repetitive. If the intensity value of Storing and accessing retinal images:
a pixel has a certain value, the values of the pixels in its surround ophthalmology picture-archiving systems
usually have similar values. After an image is acquired on a fundus camera or OCT device,
In order to explain the concept of an image compression algo- it becomes part of the medical record. It therefore should be
rithm, let us proceed with an example. We start with an image stored in some form, so that it can be communicated to other
in which 50 pixels in an area all have the same intensity value. clinicians and providers, or consulted at a later date.
We will pick the value 128. Instead of storing 50 memory ele- Images can be stored directly on the imaging device, but PACS
ments, all having the value 50 (typically requiring 50 bytes total), are available that make image storage more practical, allowing
the simple image compression algorithm counts the number of images from a variety of imaging devices to be stored and
repetitions of an intensity value, reducing this number to two reviewed. PACS may be standalone, or may be integrated into
memory elements: the first one, the repeat value 50, and the an electronic health record. PACS do not need to be separate,
and some are an integral part of an electronic medical record one algorithm and form a combined step in another, different
system. Most PACS offer manufacturer independence: the algorithm, but the steps described below are typical.
images are stored in such a manner that they can still be viewed 157
even if the device on which they were recorded is no longer Common image-processing steps
available, and are not lost when the “old” device is retired. • Preprocessing: remove variability without losing essential

Chapter 6
With the advent of SD-OCT technology and dense OCT scan- information
ning, which can result in image sizes of a gigabyte per exam, • Detection: locate specific structures of interest, or features
deciding how clinical images are stored, and whether all data • Segmentation: determine precise boundaries of objects
acquired is stored or just the clinically relevant images, is becom- • Registration: find similar regions in two or more images
ing more and more important for the practitioner, as is choosing • Interpretation: output clinically relevant information.
the level and type of image compression.
For small practices, keeping images stored on the device can Preprocessing

Image Processing
still be a cost-effective solution. For larger practices, storage in a The purpose of preprocessing is to remove as much variation
PACS computer network accessible over the clinic allows a as possible from the image without losing essential information.
patient’s images to be accessible in the patient area during clinic. There are many sources of variation during image acquisition.
Typically, PACS takes care of compression and uncompression Image device manufacturer and type, different sizes of field of
calculations “behind the scenes.” view, variations in flash illumination, exposure duration, patient
movement, variability in retinal pigmentation or in cornea/
Different strategies for storing ophthalmic images
lens/vitreous opacities are all examples of variation between
• Slides and computer printouts stored in the paper chart or images taken for the same purpose. These variations do not
photo archive contribute to the understanding of the image, but they may alter
• Slides and paper printouts scanned and stored in a PACS further image analysis steps.
• Clinically relevant views stored in a PACS Preprocessing attempts to eliminate some or all of these
• All raw data and clinically relevant views stored in a PACS sources of variation, as much as possible. A simple example is
• Standard for storage and communication of ophthalmology field of view: by scaling the image, and subtracting unexposed
images. areas of the image, images from different cameras are normal-
Digital exchange of retinal images and DICOM ized to a “standard fundus image.” Another example is illumina-
DICOM stands for Digital Imaging and Communications in tion correction, where the pixel intensity values of underexposed
Medicine and is an organization founded in 1983 to create a areas are increased, and those of overexposed intensities reduced,
standard method for the transmission of medical images and so that the pixel intensities fall into a narrower and more predict-
their associated information across all fields of medicine. For able range.
ophthalmology, Working Group 9 (WG-9) of DICOM is a formal There are many parallels between image preprocessing using
part of the American Academy of Ophthalmology. Until recently, computers and human retinal image processing in ganglion
the work of WG-9 has focused on creating standards for fundus, cells.19
anterior-segment, and external ophthalmic photography, result-
Detection
ing in DICOM Supplement 91 Ophthalmic Photography Image
The purpose of detection is to locate, typically in a preprocessed
SOP Classes, and on OCT imaging in DICOM Supplement
image, the specific structures of interest, or features, without yet
110: Ophthalmic Tomography Image Storage SOP53,54 (http://
determining their exact boundaries. Examples of such features
medical.nema.org).
can be edges, dark or bright spots, oriented lines, and dark–
DICOM standards build as much as possible upon other stan-
bright transitions in OCT images. Other terms in use for the
dards. For example, DICOM does not prescribe an image com-
concept “structure of interest” are wavelets, textures, or filters.
pression standard. Images stored as DICOM images can contain
Typically, each individual pixel in the image is examined for the
the actual image data. A typical example of this is a JPEG image.
presence of one feature or more, and usually the surrounding
DICOM 91 and 110 standardize how metadata for an image,
area, or context, of each pixel is included in this examination.
such as patient and visit data, acquisition modes and camera
The examination itself usually involves a mathematical compu-
settings, compression settings and data formats, and clinical
tation of the similarity between prototypes of the feature and
interpretation, is stored as an integral part of the image.
each pixel and its surround. Conceptually similar terms used in
the image analysis literature resembling similarity computation
Retinal image analysis are “correlation,” “convolution,” “lifting,” “matching,” and
Image analysis is a process by which meaningful information or “comparison.” Usually a nonlinearity is utilized to convert the
measurements can be extracted from digital images, typically by similarity estimate into a discrete value, for example, “present”
computer algorithms. In ophthalmology, image analysis is pri- versus “nonpresent.”
marily used to extract clinically relevant measurements from The output of the matching process indicates if and where the
images of the eye, but also to estimate retinal biomarkers, most features were detected in the image. In some image analysis
commonly from fundus color images and from OCT images. The systems, this output is interpreted directly, while in others, a
purpose of this section is to familiarize the reader with the main segmentation step (see below) is used to determine the exact
concepts used in the ophthalmic image analysis literature. Image boundaries of the object represented by the features.
analysis is best understood as a process consisting of a combina- There are many parallels between the features and the convo-
tion of steps. Not all steps are performed in all image analysis lution process in digital image analysis, and the filters in the
algorithms, and some steps may be explicit as multiple steps in human visual cortex.55
Segmentation method functionality, training data and performance testing
The purpose of segmentation is to determine the precise bound- data sets must be completely separate.52
158 aries of objects in the image, when the presence of specific Pixel feature classification
object features has been determined in the detection step. Pixel feature classification is a machine learning technique that
For example, if the ganglion cell layer in an OCT image is assigns one or more classes to the pixels in an image.55,57 Pixel
Section 1

detected but still has disjoint boundaries, the segmentation classification uses multiple pixel features including numeric
step connects these into a connected boundary. Commonly properties of a pixel and the surroundings of a pixel. Originally,
used segmentation techniques are graph search and dynamic pixel intensity was used as a single feature. More recently,
programming, both of which try to find the mathematically n-dimensional multifeature vectors are utilized, including pixel
best-fitting boundary, given the specific detection output(s). contrast with the surrounding region and information regarding
The output of the segmentation step can be used directly for the pixel’s proximity to an edge. The image is transformed into
Optical Imaging Technologies

assessment, for example when showing the different layers


Retinal Imaging and Diagnostics

an n-dimensional feature space and pixels are classified accord-


on a macular OCT scan, or can be the input for an inter­ ing to their position in space. The resulting hard (categorical)
pretation step. or soft (probabilistic) classification is then used either to assign
Registration labels to each pixel (for example “vessel” or “nonvessel” in the
The purpose of registration is to find similar regions in two or case of hard classification), or to construct class-specific likeli-
more images so they can be colocalized. Registration is often hood maps (e.g., a vesselness map for soft classification). The
used to overlay an angiogram on an OCT image, compare number of potential features in the multifeature vector that can
images from the same patient from two different visits, to detect be associated with each pixel is essentially infinite. One or more
improvement or worsening of the patient’s condition between subsets of this infinite set can be considered optimal for classify-
visits, or mosaicing, where several fundus images are stitched ing the image according to some reference standard. Hundreds
together into one image covering a larger area of the retina. The of features for a pixel can be calculated in the training stage to
registration step often utilizes similar functions as the detection cast as wide a net as possible, with algorithmic feature selection
step. steps used to determine the most distinguishing set of features.
Extensions of this approach include different approaches to clas-
Interpretation sifying groups of neighboring pixels subsequently by utilizing
Usually, when the preceding steps have been completed an group properties in some manner, for example cluster feature
interpretation step is used to output clinically relevant informa- classification, where the size, shape, and average intensity of
tion. If the boundaries of the macular retinal layers have been the cluster may be used.
segmented, interpretation involves calculating the distance
between the boundaries, so the user can see the thickness of the
different layers at specific locations. These thicknesses can even
Measuring performance of image
be compared to a database of normal thicknesses at that same analysis algorithms
location, so that the output represents how likely it is that the Crucial for the acceptance of image analysis algorithms are eval-
retina is thickened at a specific location. Or, after microaneu- uations of its performance. Most often performance is compared
rysms and exudates have been detected and segmented in mul- to human experts, though this raises its own set of issues, as
tiple images from the same patient, these outputs are combined explained below. The agreement between an automatic system
into the clinically relevant information determining whether the and an expert reader may be affected by many influences –
patient has more than minimal DR or not. system performance may become impaired due to the algorith-
mic limitations, the imaging protocol, properties of the camera
Unsupervised and supervised image analysis
used to acquire the fundus images, and a number of other causes.
The design and development of a retinal image analysis system
For example, an imaging protocol that does not allow small
usually involve the combination of some of the steps as
lesions to be depicted and thus detected will lead to an artifi-
explained above, with specific sizes of features and specific
cially overestimated system performance if such small lesions
operations used to map the input image into the desired inter-
might have been detected with an improved camera or better
pretation output. The term “unsupervised” is used to indicate
imaging protocol. Such a system then appears to be performing
such systems. The term “supervised” is used when the algor­
better than it truly is if human experts and the algorithm both
ithm is improved in stepwise fashion by testing whether addi-
overlook true lesions.
tional steps or a choice of different parameters can improve
performance. This procedure is also called training. The theo- Sensitivity and specificity
retical disadvantage of using a supervised system with a training The performance of a lesion detection system can be measured
set is that the provenance of the different settings may not be by its sensitivity, which is the number of true positives divided
clear. However, because all retinal image analysis algorithms by the sum of the total number of (incorrectly missed) false nega-
undergo some optimization of parameters, by the designer or tives plus the number of (correctly identified) true positives.52
programmer, before clinical use, this is only a relative, not System specificity is determined as the number of true negatives
absolute, difference. Two distinct stages are required for a divided by the sum of the total number of false positives (incor-
supervised learning/classification algorithm to function: a train- rectly identified as disease) and true negatives. Sensitivity and
ing stage, in which the algorithm “statistically learns” to classify specificity assessment both require ground truth, which is rep-
correctly from known classifications, and a testing or classifica- resented by location-specific discrete values (0 or 1) of disease
tion stage in which the algorithm classifies pre­viously unseen presence or absence for each subject in the evaluation set. The
images. For proper assessment of supervised classification location-specific output of an algorithm can also be represented
by a discrete number (0 or 1). However, the output of the assess- be a better estimate. Because of the added procedure it is
ment algorithm is often a continuous value determining the less patient-friendly, and has higher cost associated with it.
likelihood p of local disease presence, with an associated prob- 3. Doing a biopsy. Often this may be ethically unacceptable. It 159
ability value between 0 and 1. Consequently, the algorithm can also displaces the problem, because the biopsy would
be made more specific or more sensitive by setting an operating necessarily be interpreted by human expert(s), for example a

Chapter 6
threshold on this probability value, p. pathologist, with intra- and interobserver variability. It is
more unequivocal, but also more invasive and has higher
Receiver operator characteristics
cost.
If an algorithm outputs a continuous value, as explained above,
4. Outcome-based. If the clinically relevant question is not so
multiple sensitivity/specificity pairs for different operating
much whether a microaneurysm is present or absent, but
thresholds can be calculated. These can be plotted in a graph,
instead whether the patient is at risk of going blind from
which yields a curve, the so-called receiver operator character-
proliferative disease, we can wait for that outcome to occur.

Image Processing
istics or ROC curve.52,56 The area under this ROC curve (AUC,
However, the true state of disease at this moment would still
represented by its value Az) is determined by setting a number
not be known, only the true state at some time in the past.
of different thresholds for the likelihood p. Sensitivity and
Clinical outcome is maximally unequivocal and minimally
specificity pairs of the algorithm are then obtained at each of
subjective.
these thresholds. The ground truth is kept constant. The
5. True state of disease, which is an unknowable quantity, as
maximum AUC is 1, denoting a perfect diagnostic procedure,
explained above.
with some threshold at which both sensitivity and specificity
are 1 (100%).
As we have seen, in practice the reference standard therefore
Repeatability and variability almost never represents the true state of the disease of a patient.
In addition to the above measures, the performance of an algo- Clinical safety relevant performance measurement
rithm is also measured by its test–retest variability. With all Performance of a system that has been developed for screening
other variables such as disease state, patient factors, imaging should not be evaluated based solely on its sensitivity and speci-
device, and operator held constant while obtaining multiple ficity for detection of that disease. Such metrics do not accurately
images, this measure determines how much the algorithm’s reflect the complete performance in a screening setup. Rare,
output remains constant on the “same” input. For an algorithm, irregular, or atypical lesions often do not occur frequently
test–retest variability is not comparable to intraobserver vari- enough in standard data sets to affect sensitivity and specificity
ability. Almost all image analysis algorithms are deterministic, but can have dramatic health and safety implications. To maxi-
and if the input image is exactly the same, the output will always mize screening relevance, the system must therefore include a
be exactly the same. mechanism to detect rare, atypical, or irregular abnormalities,
The reference standard or gold standard for example in DR detection algorithms.43 For proper perfor-
mance assessment, the types of potential false negatives – lesions
Typically these performance measurements are made by com-
that can be expected or shown to be incorrectly missed by the
paring the output of the image analysis system to some stan-
automated system – must be determined. While detection of red
dard, usually called the reference standard or gold standard.
lesions and bright lesions is widely covered in the literature,
Because the performance of some image analysis systems, for
detection of rare or irregular lesions, such as hemorrhages, neo-
example for detection of DR, is starting to exceed that of indi-
vascularization, geographic atrophy, scars, and ocular neo-
vidual clinicians or groups of clinicians, creating the reference
plasms has received much less attention, despite the fact that
standard is an area of active research.42
they all can occur in combination with DR and other retinal
The problem is that the true disease state of the patient is very
diseases, as well as in isolation. For example, presence of such
difficult and in fact, impossible, to measure. For example, at the
lesions in isolated forms and without any co-occurrence of small
limit of retinal specialists’ detection performance, one specialist
red lesions is rare in DR and thus missing these does not affect
may see a microaneurysm in the macula on clinical exam of a
standard metrics of performance to a measurable degree.41 One
patient suspected of having DR, while another only sees some
suitable approach for detecting such lesions is to use a retinal
pigmentary variation. In most cases it is impossible to state that
atlas, where the image is routinely compared to a generic normal
one of these clinicians is right and the other is wrong.
retina. After building a retinal atlas by registering the fundus
Given that determining the true state of disease necessary to
images according to a disc, fovea, and a vessel-based coordinate
create the reference standard is so challenging, the following
system, image properties at each atlas location from a previously
options have been developed and are in wide use42:
unseen image can be compared to the atlas-based image proper-
ties. Consequently, locations can be identified as abnormal if
1. Using the modality under study. The images are read and
groups of pixels have values outside the normal atlas range.
adjudicated by multiple trained readers according to a
standardized protocol. This is less biased and a better
estimate than a single clinician, but has higher cost. This
FUNDUS IMAGE ANALYSIS
method is often used, but the true disease is not known this Planar fundus imaging is the most established method of
way. retinal imaging. Until recently, fundus image analysis was
2. Using a different modality. In the case of a microaneurysm, the only source of quantitative indices reflecting retinal mor-
an angiogram would be a suitable modality. It requires phology. Retinal structures that lend themselves for fundus
expert interpretation, and preferably multiple experts. It is image analysis include: retinal vessels, hemorrhages, micro-
less biased towards the imaging modality and may therefore aneurysms, pigment epithelial abnormalities (scars, laser
spots), drusen, hyperpigmentation or hypopigmentation, choroid- Detection of fovea and optic disc
related abnormalities or lesions, and segmentation of retinal
Location of the optic disc and fovea benefits retinal image analy-
160 layers. In this section we will discuss retinal vessel detection,
sis (Fig. 6.4). It is often necessary to mask out the normal anatomy
retinal lesion detection, construction of fundus imaging-
before finding abnormal structures. For instance, the optic disc
based retinal atlases, and assessment of image analysis algo-
might be mistaken for a bright lesion if not detected. Secondly,
rithms. The previous section on image analysis concepts for
Section 1

the distribution of the abnormalities is not uniform on fundus


clinicians will be helpful in understanding the concepts in
photographs. Specific abnormalities occur more often in specific
this overview. The next section will explain image analysis
areas on the retina. Most optic disc detection methods are based
of OCT images.
on the fact that the optic disc is the convergence point of blood
vessels and it is normally the brightest structure on a fundus
Detection of retinal vessels image. Most fovea detection methods depend partially on the
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Automated segmentation of retinal vessels has been highly suc- result of optic disc detection.
cessful in the detection of large and medium vessels57–59 (Fig. 6.3). Hoover et al. proposed a method for optic disc detection based
Because retinal vessel diameter and especially the relative diam- on the combination of vessel structure and pixel brightness.65 If
eters of arteries and veins are known to signal the risk of sys- a strong vessel convergence point is found in the image, it is
temic disease, including stroke, accurate determination of retinal regarded as the optic disc. Otherwise the brightest region is
vessel diameters, as well as the ability to differentiate veins from detected. Foracchia et al. proposed an optic disc detection
arteries, has become more important. Several semiautomated method based on vessel directions.66 A parabolic model of the
and automated approaches to determining vessel diameter have main vascular arches is established and the model parameters
now been published.60–62 Other active areas of research include are the directions associated with different locations on the para-
separation of arteries and veins, detection of small vessels with bolic model. The point with a minimum sum of square error is
diameters of less than a pixel, and analysis of complete vessel reported as the optic disc location. Lowell et al., by matching an
trees using graphs. optic disc model using the Pearson correlation, determined an
Vessel detection approaches can be divided into region-based initial optic disc location, and then traced the optic disc bound-
and edge-based approaches. Region-based segmentation ary using a deformable contour model.67
methods label each pixel as either inside or outside a blood Most fovea detection methods use the fact that the fovea is a
vessel. Niemeijer et al. proposed a pixel-based retinal vessel dark region in the image and that it normally lies in a fixed
detection method using a gaussian derivative filter bank and orientation and location relative to the optic disc and the main
k-nearest-neighbor classification.57,59 Staal et al.59 proposed a vascular arch. In a study by Fleming et al., approximate locations
pixel feature-based method that additionally analyzed the of the optic disc and fovea are obtained using the elliptical form
vessels as elongated structures. Edge-based methods can be of the main vascular arch.68 Then, the locations are refined based
further classified into two categories: window-based methods on the circular edge of the optic disc and the local darkness at
and tracking-based methods. Window-based methods estimate the fovea. Li and Chutatape also proposed a method to select the
a match at each pixel against the pixel’s surrounding window. brightest 1% pixels in a gray-level image.69 The pixels are clus-
The tracking approach exploits local image properties to trace tered and principal component analysis based on a trained
the vessels from an initial point. A tracking approach can better system is applied to extract a single point as the estimated loca-
maintain the connectivity of vessel structure. Lalonde et al.63 tion of the optic disc. A fovea candidate region is then selected
proposed a vessel-tracking method by following an edge line based on the optic disc location and the main vascular arch
while monitoring the connectivity of its twin border on a vessel shape. Within the candidate region, the centroid of the cluster
map computed using a Canny edge operator. Breaks in the con- with the lowest mean intensity and pixel number greater than
nectivity will trigger the creation of seeds that serve as extra one-sixth disc area is regarded as the foveal location. In a paper
starting points for further tracking. Gang et al. proposed a retinal by Sinthanayothin et al., the optic disc was located as the area
vessel detection using a second-order gaussian filter with with the highest variation in intensity of adjacent pixels, while
adaptive filter width and adaptive threshold.64 the fovea was extracted using intensity information and a

A B C D

Fig. 6.3 Automated vessel analysis. (A) Fundus image; (B) retinal specialist annotation; (C) vesselness map from Staal algorithm (staal);
(D) vesselness map from direct pixel classification. (Reproduced with permission from Niemeijer N, Staal JJ, van Ginneken B, et al. Comparative
study of retinal vessel segmentation methods on a new publicly available database. In: Fitzpatrick JM, Sonka (Eds) SPIE medical imaging, vol.
5370. SPIE, 2004, p. 648–56.)
relative distance to the optic disc.70 Tobin et al. proposed a The possible locations for the fovea are stored in a separate
method to detect the optic disc based on blood vessel features, image and the highest-probability location is detected as the
such as density, average thickness, and orientation.71 Then the fovea location. 161
fovea location was determined based on the location of the optic
disc and a geometry model of the main blood vessel. Niemeijer Detection of retinal lesions

Chapter 6
et al. proposed a method to localize automatically both the optic In this section we will primarily focus on detection of lesions
disc and fovea in 2006.72,73 For the optic disc detection, a set of in DR. DR has the longest history as a research subject in
features are extracted from the color fundus image. A k-nearest- retinal image analysis. Figure 6.4 shows examples of a fundus
neighbor classification is used to give a soft label to each pixel photograph with the typical lesions automatically detected.
on the test image. The probability image is blurred and the pixel After preprocessing, most approaches detect candidate lesions,
with the highest probability is detected as optic disc. Relative after which a mathematical morphology template is utilized
position information between the optic disc and the fovea is used to segment and characterize the candidates (Fig. 6.5). This

Image Processing
to limit the search of fovea into a certain region. For each possible approach or a modification thereof is in use in many algorithms
location of the optic disc, a possible location of the fovea is given. for detecting DR and age-related macular degeneration.74

A B C D E

Fig. 6.4 Typical steps necessary for analysis of fundus images, in this case for early diabetic retinopathy. Top row: large sequence of image
sets from multiple patients; second row: image set of four retinal images for a single patient; third row: (A) original image; (B) vesselness map;
(C) automatically detected red lesions in white; (D) automatically detected bright lesions in white; and (E) detection of fovea (black) and optic
disc (yellow) as well as automatically detected red lesions indicated in shades of green; bright lesions in shades of blue superimposed on
original image.

A B C D

Fig. 6.5 Red lesion pixel feature classification. (A) Part of green color plane of a fundus image. Shown are pieces of vasculature and several red
lesions. Circles mark the location of some of the red lesions in the image. (B) After subtracting median filtered version of the green plane, large
background gradients are removed. (C) All pixels with a positive value are set to zero to eliminate bright lesions in the image. Note that exudates
often partially occlude red lesions. Nonoccluded parts of red lesions show up clearly in this image. An example of this is marked with a
rectangle. (D) Pixel classification result produced by contrast enhancement step. Nonoccluded parts of hemorrhages are visible together with the
vasculature and a number of red lesions. (Reproduced with permission from Niemeijer M, van Ginneken B, Staal J, et al. Automatic detection of
red lesions in digital color fundus photographs. IEEE Trans Med Imaging 2005;24:584–92.)
Additional enhancements include the contributions of Spencer lesions. Subsequently, a combination of one or more filtering
et al.75 and Frame et al.76 They added additional preprocessing operations combined with mathematical morphology is
162 steps, such as shade correction and matched filter postprocess- employed to detect red lesion suspects. In some cases, suspect
ing, to this basic framework to improve algorithm performance. red lesions are further classified into individual lesion types
Algorithms of this kind function by detecting candidate micro- and refined algorithms are capable of detecting specific retinal
aneurysms of various shapes, based on their response to specific structures and abnormalities.
Section 1

image filters. A supervised classifier is typically developed to Initially, red lesions were detected in fluorescein angiograms
separate valid microaneurysms from spurious or false responses. because their contrast against the background is much higher
However, these algorithms were originally developed to detect than that of microaneurysms in color fundus photography
the high-contrast signatures of microaneurysms in fluorescein images.75,76,80 Hemorrhages mask out fluorescence and present as
angiogram images. An important development was the addition dark spots in the angiograms. These methods employed a math-
of a more sophisticated filter, a modified version of the top-hat ematical morphology technique that eliminated the vasculature
Optical Imaging Technologies
Retinal Imaging and Diagnostics

filter, so called because of its cross-section, to red-free fundus from a fundus image but left possible microaneurysm candi-
photographs rather than angiogram images, as was first dates untouched, as first described in 1984.16 Later, this method
described by Hipwell et al.77 They tested their algorithm on a was extended to high-resolution red-free fundus photographs
large set of >3500 images and found a sensitivity/specificity by Hipwell et al.77 Instead of using morphology operations, a
operating point of 0.85/0.76. Once this filter-based approach neural network was used, as demonstrated by Gardner et al.81
had been established, development accelerated. The next step In their work, images are divided into 20 × 20 pixel grids and
was broadening the candidate detection step, originally devel- the grids are individually classified. Sinthanayothin et al. used
oped by Baudoin16 to detect candidate pixels, to a multifilter a detection step to find blood-like regions and to segment both
filter-bank approach.57,78 The responses of the filters are used vessels and red lesions in a fundus image.82 A neural network
to identify pixel candidates using a classification scheme. Math- was used to detect the vessels exclusively, and the remaining
ematical morphology and additional classification steps are objects were labeled as microaneurysms. Niemeijer et al. pre-
applied to these candidates to decide whether they indeed sented a hybrid scheme that used a supervised pixel classification-
represent microaneurysms and hemorrhages (Fig. 6.6). A similar based method to detect and segment the microaneurysm
approach was also successful in detecting other types of DR candidates in color fundus photographs.78 This method allowed
lesions, including exudates and cotton-wool spots, as well as for the detection of larger red lesions (i.e., hemorrhages) in addi-
drusen in AMD.79 tion to the microaneurysms using the same system. A large set
Small red retinal lesions, namely microaneurysms and small of additional features, including color, was added to those that
retinal hemorrhages, are typical for multiple retinal disorders, had been previously described.76,80 Using the features in a super-
including DR, hypertensive retinopathy, venous occlusive vised classifier distinguished between real and spurious candi-
disease, and other less common retinal disorders such as idio- date lesions. These algorithms can usually distinguish between
pathic juxtafoveal telangiectasia. The primary importance of overlapping microaneurysms because they give multiple candi-
small red lesions is that they are the leading indicators of DR. date responses.
Because they are difficult to differentiate for clinicians on stan- Other recent algorithms only detect microaneurysms and
dard fundus images from nonmydriatic cameras, hemorrhages forgo a phase of detecting normal retinal structures like the optic
and microaneurysms are usually detected together and associ- disc, fovea, and retinal vessels, which can act as confounders for
ated with a single combined label. Historically, red lesion detec- abnormal lesions. Instead, the recent approaches find the micro-
tion algorithms focused on detection of normal anatomical aneurysms directly using template matching in wavelet sub-
objects, especially the vessels, because they can locally mimic red bands.83 In this approach, the optimal adapted wavelet transform

A B C

Fig. 6.6 Red lesion detection. (A) Thresholded probability map. (B) Remaining objects after connected component analysis and removal of large
vasculature. (C) Shape and size of extracted objects in panel (B) do not correspond well with actual shape and size of objects in original image.
Final region growing procedure is used to grow back actual objects in original image, which are shown here. In (B) and (C), the same red lesions
as in Figure 6.6A are indicated with a circle. (Reproduced with permission from Niemeijer M, van Ginneken B, Staal J, et al. Automatic detection
of red lesions in digital color fundus photographs. IEEE Trans MedImaging 2005;24:584–92.)
is found using a lifting scheme framework. By applying a thresh- recently published.90 This method detects the location of the
old on the matching result of the wavelet template, the microan- optic disc, determines an appropriate region of interest, classifies
eurysms are labeled. This approach has meanwhile been vessels as arteries or veins, estimates vessel widths, and calcu- 163
extended explicitly to account for false negatives and false posi- lates the arteriovenous ratio. The system eliminates all vessels
tives.42 Because it avoids detection of the normal structures, such outside the arteriovenous ratio measurement region of interest.

Chapter 6
algorithms can be very fast, on the order of less than a second A skeletonization operation is then applied to the remaining
per image. vessels after which vessel crossings and bifurcation points are
Bright lesions, defined as lesions brighter than the retinal back- removed, leaving a set of vessel segments consisting of only
ground, can be found in the presence of retinal and systemic vessel centerline pixels. Features are extracted from each center-
disease. Some examples of such bright lesions of clinical interest line pixel in order to assign these a soft label indicating the likeli-
include drusen, cotton-wool spots, and lipoprotein exudates. To hood that the pixel is part of a vein. As all centerline pixels in a
complicate the analysis, flash artifacts can be present as false connected vessel segment should be the same type, the median

Image Processing
positives for bright lesions. If the lipoprotein exudates only soft label is assigned to each centerline pixel in the segment.
appear in combination with red lesions, they would only be Next, artery vein pairs are matched using an iterative algorithm,
useful for grading DR. The exudates can, however, in some cases and finally, the widths of the vessels are used to calculate the
appear as isolated signs of DR in the absence of any other lesion. average arteriovenous ratio.
Several computer-based systems to detect exudates have been
proposed (Fig. 6.7).74,79,81,82,84 Retinal atlas
Because the different types of bright lesion have different diag- The retina has a relatively small number of key anatomic struc-
nostic importance, algorithms should be capable not only of tures (landmarks) visible using planar fundus camera imaging.
detecting bright lesions, but also of differentiating among the Additionally, the expected shape, size, and color variations
bright lesion types. One example algorithm capable of detection across a population are expected to be high. While there have
and differentiation of bright lesions was reported by Niemeijer been a few reports on estimating retinal anatomic structure
et al. in 2007.79 This algorithm is based on an earlier red lesion using a single retinal image,71 we are not aware of any published
algorithm presented by Hipwell et al. in 200077 and includes the work demonstrating the construction of a statistical retinal atlas
following traditional steps, which are illustrated in Figure 6.6: using data from a large number of subjects. The choice of atlas
landmarks in retinal images may vary depending on the view of
1. lesion candidate cluster detection, where pixels are clustered interest. Regardless, the atlas should represent most retinal
into highly probable lesion regions image properties in a concise and intuitive way. Three land-
2. true bright lesion detection, where each candidate cluster is marks can be used as the retinal atlas key features: the optic disc
classified as a true lesion based on cluster features, including center, the fovea, and the main vessel arch defined as the location
surface area, elongatedness, pixel intensity gradient, of the largest vein/artery pairs. The disc and fovea provide
standard deviation of pixel values, pixel contrast, and local landmark points, while the arch is a more complicated two-part
“vesselness” (as derived from a vessel segmentation map) curved structure that can be represented by its central axis. The
3. differentiation of lesions into drusen, exudates, and cotton- atlas coordinate system then defines an intrinsic, anatomically
wool spots where a third classifier determines the likelihood meaningful framework within which anatomic size, shape,
for the true bright lesion to represent specific lesion types. color, and other characteristics can be objectively measured and
compared. Choosing either the disc center or fovea alone to
Vessel analysis define the atlas coordinate system would allow each image from
Vessel measures, such as the average width of arterioles and the population to be translated so pinpoint alignment can be
venules, the ratio of arteriolar to venular widths, and the branch- achieved. Choosing both disc and fovea allows corrections for
ing ratio, have been established to be predictive of systemic translation, scale, and rotational differences across the popula-
diseases, especially hypertension, and also have potential value tion. However, nonlinear shape variations across the population
in degenerative retinal diseases such as retinitis pigmentosa. The would not be considered – this can be accomplished when the
methods in the section on detection of retinal vessels locate the vascular arch information is utilized. The end of the arches can
vessels, but cannot determine vessel width. Additional tech- be defined as the first major bifurcations of the arch branches.
niques are needed to measure the vessel width accurately. The arch shape and orientation vary from individual to indi-
Al-Diri et al. proposed an algorithm for segmentation and mea- vidual and influence the structure of the remaining vessel
surement of retinal blood vessels by growing a “ribbon of twins” network. Establishing an atlas coordinate system that incorpo-
active contour model. Their approach uses an extraction of rates the disc, fovea, and arches allows for translation, rotation,
segment profiles algorithm, which uses two pairs of contours to scaling, and nonlinear shape variations to be accommodated
capture each vessel edge.85 The half-height full-width algorithm across a population.
defines the width as the distance between the points on the An isotropic coordinate system is a system in which the size
intensity curve at which the function reaches half its maximum of each imaged element is the same in all three dimensions. This
value to either side of the estimated center point.86–88 The Gregson is desirable for a retinal atlas so images can refer to the atlas
algorithm fits a rectangle to the profile, setting the width so that independent of spatial pixel location by a linear one-to-one
the area under the rectangle is equal to the area under the mapping. The radial distortion correction (RADIC) model
profile.33 Xu et al. recently published a method based on graph attempts to register images in a distortion-free coordinate system
search showing less variability than human experts (Fig. 6.8).89 using a planar-to-spherical transformation, so the registered
A fully automated method from the Abramoff group to image is isotropic under a perfect registration, and places the
measure the arteriovenous ratio in disc center retinal images was registered image in an isotropic coordinate system (Fig. 6.9).91
164
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

Exudates Cotton-wools Drusen

Fig. 6.7 Bright lesion detection algorithm steps performed to detect and differentiate “bright lesions.” From left to right: exudates, cotton-wool
spots, and drusen. From top to bottom: relevant regions in the retinal color image (all at same scale); a posteriori probability maps after first
classification step; pixel clusters labeled as probable bright lesions (potential lesions); bottom row shows final labeling of objects as true bright
lesions, overlaid on original image. (Reproduced with permission from Niemeijer M, van Ginneken B, Russell SR, et al. Automated detection and
differentiation of drusen, exudates, and cotton-wool spots in digital color fundus photographs for diabetic retinopathy diagnosis. Invest
Ophthalmol Vis Sci 2007;48:2260–7.)
165

Chapter 6
Image Processing
A B

Fig. 6.8 Automated vessel width measurement. (A) Automated measurement of vessel width (black lines); (B) three human experts marking the
widths of the vessel manually (in green, blue, and yellow).

A B

Fig. 6.9 Registration of fundus image pair using (A) quadratic model and (B) radial distortion correction model. Vessel center lines are overlaid
for visual assessment of registration accuracy. This registration is performed to disc-centered and macula-centered images to provide an
increased anatomic field of view. (Reproduced with permission from Lee S, Abramoff MD, Reinhardt JM. Retinal image mosaicing using the
radial distortion correction model. In: Joseph MR, Josien PWP, eds. Proceedings of the SPIE. SPIE, 2008. p 691435.)

An isotropic atlas makes it independent of spatial location to (Fig. 6.10).93 Rigid coordinate alignment is done for each fundus
map correspondences between the atlas and test image. The image to register the disc center and the fovea. The control points
intensities in overlapping area are determined by a distance- are determined by sampling points from equidistant locations in
weighted blending scheme.92 radial directions from the disc center. Usually, the sampling uses
Retinal images in clinical practice are acquired under diverse smoothed trace lines utilizing third-order polynomial curve
fundus camera settings subjected to saccadic eye movement, and fitting to eliminate locally high tortuosity of vascular tracings
with variable properties, including focal center, zoom, and tilt. which may cause large geometric distortions (Fig. 6.11).
Thus, atlas landmarks from training data need to be aligned to A retinal atlas can be used as a reference to quantitatively
derive any meaningful statistical properties from the atlas. Since assess the level of deviation from normality. An analyzed image
the projective distortion within an image is corrected in registra- can be compared with the retinal atlas directly in the atlas coor-
tion, the interimage variations in the registered images appear dinate space. The normality can thus be defined in several ways
as the difference in the rigid coordinate transformation param- depending on the application purpose – using local or global
eters of translation, scale, and rotation. chromatic distribution, degree of vessel tortuosity, presence of
The atlas landmarks serve as the reference set so each color pathological features, or presence of artifacts (Fig. 6.12). Other
fundus image can be mapped to the coordinate system defined uses for a retinal atlas include image quality detection and
by the landmarks. As the last step of atlas generation, color disease severity assessment. Retinal atlases can also be employed
fundus images are warped to the atlas coordinate system so in content-based image retrieval leading to abnormality detec-
that the arch of each image is aligned to the atlas vascular arch tion in retinal images.94
166
Section 1
Optical Imaging Technologies
Retinal Imaging and Diagnostics

A B

Fig. 6.10 Atlas coordinate mapping by thin plate spline (A) before and (B) after mapping. Naive main arch traces obtained by Dijkstra’s line
detection algorithm are drawn as yellow lines that undergo polynomial curve fitting to result in blue lines. Atlas landmarks (disc center, fovea, and
vascular arch) are drawn in green, and equidistant radial sampling points marked with dots. (Reproduced with permission from Abramoff MD,
Garvin M, Sonka M. Retinal imaging and image analysis. IEEE Rev Biomed Eng 2010;3:169–208.)

A B C

Fig. 6.11 Registration of anatomic structures according to increasing complexity of registration transform – 500 retinal vessel images are overlaid
and marked with one foveal point landmark each (red spots). Rigid coordinate alignment by (A) translation, (B) translation and scale, and
(C) translation, scale, and rotation. (Reproduced with permission from Abramoff MD, Garvin M, Sonka M. Retinal imaging and image analysis.
IEEE Rev Biomed Eng 2010;3:169–208.)

To drive the development of progressively better fundus


Performance of DR detection algorithms image analysis methods, research groups have established pub-
Several groups have studied the performance of detection algo- licly available, annotated image databases in various fields.
rithms in a real-world setting. The main goal of such a system is Fundus imaging examples are represented by the STARE,96
to decide whether the patient should be evaluated by a human DRIVE,57 REVIEW,97 and MESSIDOR databases,98 with large
expert or can return for routine follow-up, based solely on auto- numbers of annotated retinal fundus images, with expert anno-
mated analysis of retinal images.43,44 tations for vessel segmentation, vessel width measurements, and
DR detection algorithms appear to be mature and competitive DR detection. Image analysis competitions such as the Retinopa-
algorithms and have now reached the human intrareader vari- thy Online Challenge95 have also been initiated.
ability limit.41,42,95 Additional validation studies on larger, well- The Digital Retinal Images for Vessel Evaluation (DRIVE)
defined, but more diverse populations of patients with diabetes database was established to enable comparative studies on seg-
are urgently needed, anticipating cost-effective early detection mentation of retinal blood vessels in retinal fundus images. It
of DR in millions of people with diabetes to triage those patients contains 40 fundus images from subjects with diabetes, both
who need further care at a time when they have early rather than with and without retinopathy, as well as retinal vessel segmenta-
advanced DR. Validation trials are currently under way in the tions performed by two human observers. Starting in 2005,
USA, UK, and the Netherlands. researchers have been invited to test their algorithms on this
167

Chapter 6
Image Processing
A B

C D

Fig. 6.12 Example application of employing retinal atlas to detect imaging artifacts. (A, C) Color fundus images with artifacts. (B, D) Euclidean
distance maps in atlas space using atlas coordinate system. Note that distances are evaluated within atlas image. Consequently, field of view of
distance map is not identical to that of fundus image. (Reproduced with permission from Abramoff MD, Garvin M, Sonka M. Retinal imaging and
image analysis. IEEE Rev Biomed Eng 2010;3:169–208.)

database and share their results with other researchers through the advent of the Retinopathy Online Challenge competition in
the DRIVE website.89 At the same web location, results of various 2009, comparing the performance of retinal image analysis algo-
methods can be found and compared. Currently, retinal vessel rithms was difficult.95 This competition focused on detection of
segmentation research is primarily focusing on improved seg- microaneurysms. Twenty-six groups participated in the compe-
mentation of small vessels, as well as on segmenting vessels in tition, out of which six groups submitted their results on time.
images with substantial abnormalities. The results from each of the methods in this competition are
The DRIVE database was a great success, allowing compari- summarized in Table 6.1.
sons of algorithms on a common data set. In retinal image analy- A logical next step was to provide publicly available annotated
sis, it represented a substantial improvement over method data sets for use in the context of online, standardized asyn-
evaluations on unknown data sets. However, different groups chronous competitions. In an asynchronous competition, a
of researchers tend to use different metrics to compare the algo- subset of images is made available with annotations, while the
rithm performance, making truly meaningful comparisons dif- remainder of the images are available with annotations with-
ficult or impossible. Additionally, even when using the same held. This allows researchers to optimize their algorithm per-
evaluation measures, implementation specifics of the perfor- formance on the population from which the images were drawn
mance metrics may influence final results. Consequently, until (assuming the subset with annotated images is representative
Table 6.1 Sensitivities of the different methods at the average false-positive (FP) rate of the human expert (1.08 FP/image) for various
categories of microaneurysm. The ranking of the methods for a particular category is given between brackets after the sensitivities
168
Lesion category All Subtle Regular Obvious Close to vessel

Method 1152 0.31 (3) 0.03 (4) 0.33 (3) 0.66 (4) 0.22 (2)
Section 1

Method 2153 0.21 (5) 0.03 (4) 0.21 (5) 0.51 (5) 0.22 (2)

Method 3154 0.40 (1) 0.13 (1) 0.41 (1) 0.80 (1) 0.30 (1)

Method 4155 0.36 (2) 0.10 (2) 0.38 (2) 0.69 (3) 0.19 (3)

Method 5156 0.29 (4) 0.05 (3) 0.28 (4) 0.71 (2) 0.30 (1)
Optical Imaging Technologies
Retinal Imaging and Diagnostics

NFL
GCL Surface 1
IPL
Surface 2
INL
OPL Surface 3
ONL Surface 4
OLM Surface 5
Surface 6 N T
ISL
CL Surface 7
OSL Surface 8
VM Surface 9
Surface 10
RPE
Surface 11

A B C

Fig. 6.13 Segmentation results of 11 retinal surfaces (10 layers). (A) X-Z image of optical coherence tomography volume. (B) Segmentation
results: nerve fiber layer (NFL), ganglion cell layer (GCL), inner plexiform layer (IPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer
nuclear layer (ONL), outer limiting membrane (OLM), plus inner-segment layer (ISL), outer-segment layer (OSL), and retinal pigment epithelium
complex (RPE). CL, Inner–outer segment interface complex surface; VM, Verhoeff’s membrane. (C) Three-dimensional rendering of segmented
surfaces. N, nasal; T, temporal. (Reproduced with permission from Abramoff MD, Garvin M, Sonka M. Retinal imaging and image analysis. IEEE
Rev Biomed Eng 2010;3:169–208.)

of the entire population), but they are unable to test–retest on techniques for processing OCT images has a shorter history.
the evaluation images, because those annotations are withheld. Nevertheless, it is a rapidly growing and important area, espe-
All results are subsequently evaluated using the same evalua- cially as spectral domain (SD).
tion software and research groups are allowed to submit results SD-OCT technology has enabled true 3D volumetric scans of
continuously over time. the retina to be acquired. Thus, the importance of developing
advanced image analysis techniques to maximize the extraction
Areas of active research in fundus of clinically relevant information is especially important. Never-
image analysis theless, the development of such advanced techniques can be
Major progress has been accomplished in fundus image analysis. challenging as OCT images are inherently noisy, thus often
Current challenges, on which multiple research groups world- requiring the utilization of 3D contextual information (Fig. 6.13).
wide are actively working, include the following areas: differen- Furthermore, the structure of the retina can drastically change
tiating arteries from veins, assessing accurate vessel diameter during disease. Here, we review some of the important image
(particularly in vessels only a few pixels in diameter) and vessel analysis steps for processing OCT images. We start with the
tortuosity, vessel tree analysis including tree branching patterns, segmentation of retinal layers, one of the earliest, yet still
detection of irregularly shaped hemorrhages, detection of lesion extremely important, OCT image analysis areas. We then discuss
distribution patterns (i.e., drusen), and segmentation of atrophy. techniques for flattening OCT images in order to correct scan-
Finally, integration of fundus image-based quantification with ning artifacts. Building upon the ability to extract layers, we
other metrics of disease risk, such as serum glucose level or discuss use of thickness information and of texture information.
patient history, is an area of active research with immediate This is followed by the segmentation of retinal vessels, which
clinical application. currently has its technical basis in many of the techniques used
for segmenting vessels in fundus photography, but is beginning
OPTICAL COHERENCE TOMOGRAPHY to take advantage of the 3D information only available in
SD-OCT. The use of both layer-based and texture-based proper-
IMAGE ANALYSIS ties to detect the locations of retinal lesions is then described.
Because of OCT’s relatively recent presence in ophthalmic care The ability to segment layers in the presence of lesions is
compared to fundus photography, the use of image analysis described. This section is based on a review paper. 19
Retinal layer analysis from 3D OCT OCT image flattening
Retinal layer detection SD-OCT volumes frequently demonstrate motion artifacts and
other artifacts may also be present, such as the tilting due to an 169
The segmentation of retinal layers in OCT scans has been an
important goal, because thickness changes in the layers are one off-axis placement of the pupil. Approaches for reducing these
indication of disease status. Previous-generation time domain artifacts include 1D and 2D methods that use cross-correlation

Chapter 6
scanning systems (such as the Stratus OCT by Carl Zeiss Meditec) of either A-scans100 or B-scans.116,117 In some cases, a complete
offered the ability to segment and provide thickness measure- flattening of the volume is desired based on a surface segmenta-
ments for a single layer of the retina. In particular, the retinal tion to ensure a consistent shape for segmentation and visualiza-
nerve fiber layer thickness measurements of peripapillary circu- tion. Flattening the volumes makes it possible to truncate the
lar scans and total retinal thickness measurements were avail- image substantially in the axial direction (z-direction), thereby
able and used clinically. It can be assumed that commercialized reducing the memory and time requirements of an intraretinal
layer segmentation approach. Flattening an image involves first

Image Processing
methods utilized an inherently 2D approach (i.e., if multiple 2D
slices are available in a particular scanning sequence they are segmenting the retinal pigment epithelial surface in a lower
segmented independently). Indeed, most of the early approaches resolution, fitting a thin-plate spline to this surface, and then
that have been reported in the literature99–104 for the segmenta- vertically realigning the columns of the volume to make this
tion of time domain scans are also 2D in nature. surface completely flat.111
While variations to each of the early 2D approaches exist for Retinal layer thickness analysis
the segmentation of retinal boundaries, a typical 2D approach After flattening and segmentation, the properties of the macular
proceeds as follows: preprocess the image,99–101,103 perform a 1D tissues in each layer can be extracted and analyzed. In addition
peak detection (detection) on each A-scan of the processed image to layer thickness, textural properties can also be quantified, as
to find points of interest, and (in some methods) correct for pos- explained in the next paragraph. Measuring the thickening of
sible discontinuities in the 1D border detection.99 Other 2D time specific layers is crucial in the management of diabetic macular
domain approaches include the use of 2D dynamic program- edema and other retinal disorders.118 Typically, it is useful to
ming by Baroni et al.105 compare the obtained thickness values to a normative database
Haeker et al.106–108 and Garvin et al.109 reported the first true or atlas, as is available in commercial machines for the total
3D segmentation approach for the segmentation of retinal layers macular thickness and the retinal nerve fiber layer thickness.
on OCT scans, taking advantage of 3D contextual information. However, a normative atlas for all the layers in 3D currently only
Their approach was unique in that the layers were segmented exists within individual research groups.119 Nevertheless, work
simultaneously.110 For time domain macular scans, they seg- has been done to demonstrate previously unknown changes in
mented 6–7 surfaces (5–6 layers), obtaining an accuracy and the ganglion cell layer in patients with diabetes.114,115
reproducibility similar to that of retinal specialists. By extending
the approach to SD-OCT volumes,111 utilization of 3D contextual Retinal texture analysis
information had more of an advantage (Fig. 6.14). By employing Texture, defined as measures of the spatial distribution of image
a multiscale approach, the processing time was subsequently intensities, can be used to characterize tissue properties and
decreased from hours to a few minutes while enabling segment- tissue differences. Textural properties may be important for
ing additional layers.112 A similar approach for segmenting the assessing changes in the structural or tissue composition of
intraretinal layers in optic nerve head-centered SD-OCT volumes layers that cannot be measured by changes in thickness alone.
was reported with an accuracy similar to that of the inter­ Texture can be determined in each of the identified layers three-
observer variability of two human experts.113 A preliminary dimensionally.83,120,121 3D formulations of texture descriptors
layer thickness atlas was built from a small set of normal sub- were developed for pulmonary parenchymal analysis122 and
jects. Thickness loss of the macular ganglion cell layer in people have been directly employed for OCT texture analysis.123 The
with diabetes without retinopathy was thus demonstrated, wavelet transform, a form of feature detection, has been used in
showing that diabetes also leads to retinal neuropathy.114,115 OCT analysis for de-noising and de-speckling124–126 as well as for

Fig. 6.14 An illustration of why using the complete three-dimensional (3D) contextual information in the intraretinal layer segmentation process is
superior. (Top) Sequence of two-dimensional (2D) results on three adjacent slices within a spectral domain volume obtained using a slice-by-slice
2D graph-based approach. Note the “jump” in the segmentation result for third and fourth surfaces in middle slice. (Bottom) Sequence of 3D results
on same three adjacent slices using same graph-based approach, but with the addition of 3D contextual information. 3D contextual information
prevented third and fourth surface segmentation from failing.
texture analysis.127 Early work on 3D wavelet analysis of OCT kinds of possible retinal lesions, symptomatic exudate-associated
images was based on a computationally efficient yet flexible derangements (SEADs), a general term for fluid-related abnor-
170 nonseparable lifting scheme in arbitrary dimensions.123,128 malities in the retina, are of great interest in assessing the sever-
Texture characteristics can be computed for each segmented ity of choroidal neovascularization, diabetic macular edema, and
layer, several adjacent layers, or in layer combinations (Fig. 6.12). other diseases. Detection of drusen, cotton-wool spots, areas of
pigment epithelial atrophy, or pockets of fluid under epiretinal
Section 1

Detection of retinal vessels from 3D OCT membranes may be attempted in a similar fashion.
Segmenting the retinal vasculature in 3D SD-OCT volumes129,130 The deviation of local retinal tissue from normal can be com-
allows OCT-to-fundus and OCT-to-OCT image registration. The puted by determining the local deviations from the normal
absorption of light by the blood vessel walls causes vessel silhou- appearance at each location (x; y) in each layer and selecting the
ettes to appear below the position of vessels, which thus causes areas where the absolute deviation is greater than a predefined
the projected vessel positions to appear dark on either a full pro- cutoff. More generally, in order to build an abnormality-specific
Optical Imaging Technologies
Retinal Imaging and Diagnostics

jection image of the entire volume130 or a projection image from a detector, a classifier can be trained, the inputs of which may be
segmented layer for which the contrast between the vascular the z-scores computed for relevant features. Comprehensive
silhouettes and background is highest, as proposed by Niemeijer z-scores are appropriate since an abnormality may affect several
et al.129,131 In particular, this approach used the layer near the layers in the neighborhood of a given location (x; y). The
retinal pigment epithelium to create the projection image. classifier-determined label associated with each column may be
Vessels were segmented using feature detection with gaussian selected on relevant features by one of the many available cross-
filter-banks and a k-NN pixel classification approach. The per- validation and/or feature selection methods,135–137 thus forming
formance of the automated method was evaluated for both optic a SEADness or probabilistic abnormality map.
nerve head-centered as well as macula-centered scans. The
retinal vessels were successfully identified in a set of 16 3D OCT Fluid detection and segmentation
volumes (8 optic nerve head and 8 macula-centered), with high In choroidal neovascularization, diabetic macular edema, and
sensitivity and specificity as determined using ROC analysis, other retinal diseases, intraretinal or subretinal fluid is reflective
AUC = 0.96. Xu et al. reported an approach for segmenting the of disease status and changes in fluid are an indicator of disease
projected locations of the vasculature by utilizing pixel classifica- progression or regression. With the availability of anti-VEGF
tion of A-scans.132 The features used in the pixel classification therapy, assessment of the extent and morphology of fluid is
were based on a projection image of the entire volume in combi- expected to contribute to patient-specific therapy. While regions
nation with features of individual A-scans. Both of these reported of fluid are inherently 3D, determining their 2D retinal footprint
prior approaches focused on segmenting the vessels in the region is highly relevant. Following the above-described analysis, fluid
outside the optic disc region because of difficulties in the seg- detection employs generalization of properties derived from
mentation inside this region. The neural canal opening shares expert-defined examples. Utilizing the differences between
similar features with vessels, thus causing false positives.133 Hu normal regional appearance of retinal layers as described by
et al.127 proposed a modified 2D pixel classification algorithm to texture descriptors and other morphologic indices, a classifier
segment the blood vessels in SD-OCT volumes centered at the can be trained to identify abnormal retinal appearance. The
optic nerve head, with a special focus on better identifying fluid detection starts with 3D OCT layer segmentation, result-
vessels near the neural canal opening. Given an initial 2D seg- ing in 10 intraretinal layers, plus an additional artificial layer
mentation of the projected vasculature, Lee et al. presented an below the deepest intraretinal layer so that subretinal abnor-
approach for segmenting the 3D vasculature in the volumetric malities can also be detected.123 Texture-based and morphologic
scans134 by utilizing a graph-theoretic approach (Fig. 6.15). One descriptors are calculated regionally in rectangular subvolumes,
of the current limitations of that approach is the inability to the most discriminative descriptors are identified, and these
resolve the depth information of crossing vessels properly. descriptors are used for training a probabilistic classifier. The
performance of a (set of) feature(s) is assessed by calculating
Detection of retinal lesions the area under the ROC of the fluid classifier. Once the proba-
Calculated texture and layer-based properties can be used to bilistic classifier is trained, fluid-related probability is deter-
detect retinal lesions as a 2D footprint123 or in 3D. Out of many mined for each retinal location. In order to obtain a binary

A B C D

Fig. 6.15 Example of spectral three-dimensional (3D) optical coherence tomography (OCT) vessel segmentation. (A) Vessel silhouettes indicate
position of vasculature. Also indicated in red are slice intersections of two surfaces that delineate the subvolume in which vessels are segmented
(superficial retinal layers toward vitreous are at the bottom). (B) Two-dimensional projection image extracted from projected subvolume of
spectral 3D OCT volume. (C) Automatic vessel segmentation. (D) 3D rendering of vessel around the optic nerve head. (Reproduced with
permission from Lee K, Abràmoff M, Niemeijer MK, et al. 3D segmentation of retinal blood vessels in spectral-domain OCT volumes of the optic
nerve head. Proc SPIE Med Imaging 2010;7626:76260 V.)
171

Chapter 6
Image Processing
Fig. 6.16 Symptomatic exudate-associated derangement (SEAD) segmentation from three-dimensional optical coherence tomography and SEAD
development over time. Top row: 0, 28, and 77 days after first imaging visit. Middle row: 0 and 42 days after first imaging visit. Bottom row: 0,
14, and 28 days after first imaging visit. Three-dimensional visualization in right column shows data from week 0. Each imaging session was
associated with anti-vascular endothelial growth factor reinjection.

footprint for fluid in an image input to the system, the prob- mesh, which can be interactively edited to maximize fluid region
abilities are thresholded and the footprint of the fluid region segmentation accuracy in difficult or ambiguous cases.
in this image is defined as the set of all pixels with a probability
Intraretinal layer segmentation in the presence
greater than a threshold. Useful 3D textural information can
of SEADs
be extracted from SD-OCT scans and, together with an ana-
Another area of active research is layer segmentation in retina
tomical atlas of normal retinas, can be used for clinically impor-
that contains symptomatic exudate-associated derangements
tant applications.
(SEADs). Most likely, a two-step approach is necessary in which
Fluid segmentation in 3D layers are initially segmented disregarding the SEAD presence,
Complete volumetric segmentation of fluid from 3D OCT is the then SEADs are segmented, and used to constrain the second
subject of active research. A promising approach is based on stage of layer segmentation. This process yields well-segmented
identification of a seed point in the OCT data set that is “inside” retinal layers when fluid occupies a single intraretinal layer as
and “outside” a fluid region. These points can be identified well as in situations when the fluid resides in several adjacent
automatically using a 3D variant of the probabilistic classifica- retinal layers.
tion approach outlined in the previous paragraphs. Once these
two points are identified, an automated segmentation procedure
MULTIMODALITY RETINAL IMAGING
that is based on regional graph-cut method138,139 may be employed Multimodality imaging, defined as images from the same organ
to detect the fluid volumetric region. The cost function utilized acquired using different techniques using different physical tech-
in a preliminary study was designed to identify dark 3D regions niques, is becoming increasingly common in ophthalmology. For
with somewhat homogeneous appearance. The desired proper- image information from multiple modalities to be usable in
ties of the fluid region are automatically learned from the vicin- mutual context, images must be registered so that the indepen-
ity of the identified fluid region seed point. This adaptive dent information that was acquired by different methods can be
behavior allows the same graph-cut segmentation method concatenated and form a multimodality description vector. Thus,
driven by the same cost function to segment fluid of different because of its importance in enabling multimodal analysis, retinal
appearance reliably. Figure 6.16 gives an example of 3D fluid image registration reflects another active area of research. The
region segmentations obtained using this approach. Note that several clinically used methods to image the retina were intro-
the figure depicts the same locations in the 3D data sets imaged duced above and include fundus photography, SLO, fluorescence
several times during the course of anti-VEGF treatment. The imaging, and OCT. Additional retinal imaging techniques, such
surfaces of the segmented fluid regions are represented by a 3D as hyperspectral imaging, oximetry, and adaptive optics SLO,
will bring higher resolution and additional image information. to any retinal imaging modality for which a 2D vessel segmenta-
To achieve a comprehensive description of retinal morphology tion is available. In registering poor-quality multimodal fundus
172 and eventually function, diverse retinal images acquired by dif- image pairs, which may not have sufficient vessel-based features
ferent or the same modalities at different time instants must be available, Chen et al. proposed the detection of corner points
mutually registered to combine all available local information using a Harris detector followed by use of a partial intensity
spatially. The following sections provide a brief overview of invariant feature descriptor.148 They reported obtaining 89.9%
Section 1

fundus photography and OCT registration approaches in both “acceptable” registrations (defined as registrations with a
2D and 3D; a more detailed review is available.19 Registration of median error of 1.5 pixels and a maximum error of 10 pixels
retinal images from other existing and future imaging devices when compared with ground truth correspondences) when
can be performed in a similar or generally identical manner. tested on 168 pairs of multimodal retinal images.

Registration of fundus retinal photographs Registration of OCT with fundus


Optical Imaging Technologies
Retinal Imaging and Diagnostics

Registration of fundus photographs taken either at different retinal photographs


regions of the retina, or of the same area of the retina but at dif- Registration of 2D fundus images with inherently 3D OCT
ferent times, is useful to expand the effective field of view of a images requires that the dimensionality of OCT be reduced to
retinal image, determine what part of the retina is being viewed, 2D via z-axis projection. Building on the ability to obtain vascu-
or aid in analyzing changes over time.140 We have previously lar segmentation from 3D OCT projection images, the problem
discussed some other uses for fundus–fundus registration in of fundus–OCT registration becomes virtually identical to that
regard to retinal atlases. To register 2D or planar fundus images, of fundus–fundus registration that was described in the previous
most existing registration approaches utilize identification and section. Using the same general method, high-quality OCT–
extraction of features derived from retinal vasculature seg- fundus registration can be achieved. Figure 6.17 presents the
mented separately from the individual fundus images. The main steps of the registration process and shows the registration
choice of a specific image registration algorithm to align retinal performance achieved.
images into a montage depends on the image characteristics and
the application. Images acquired with only a small overlap may Mutual registration of 3D OCT images
be optimally aligned using feature-based registration approaches, Temporal changes of retinal layers leading to assessment of
while images acquired with larger overlaps may be satisfactorily disease progression or regression can be accessed from longitu-
aligned using intensity-based approaches. Examples of feature- dinal OCT images. Comparison of morphology or function over
based registration are global-to-local matching,141 hierarchical time requires that the respective OCT image data sets be regis-
model refinement,142 and dual-bootstrap.143 Local intensity fea- tered. Since OCT is a 3D imaging modality, such registration
tures144 are particularly useful when an insufficient number of needs to be performed in 3D. For follow-up studies, image reg-
vascular features are available. Following a step of vascular skel- istration is a vital tool to enable more precise, quantitative com-
etonization, vascular branching points can be easily used as parison of disease status. Another important aspect of OCT–OCT
stable landmarks for determining image-to-image correspon- registration is the ability to enlarge retinal coverage by register-
dence. As an example, the RADIC model145 parameters are esti- ing OCT data resulting from imaging different portions of the
mated during an optimization step that uses Powell’s method146 retina. A fully 3D scale-invariant feature transform (SIFT)-based
and is driven by the vessel centerline distance. The approach approach was introduced in 2009.149 The SIFT feature extractor
presented by Lee et al. in 2010 reported registration accuracy of locates minima and maxima in the difference of gaussian scale
1.72 pixels (25–30 µm, depending on resolution) when tested in space to identify salient feature points. Using calculated histo-
462 pairs of green channel fundus images.147 The registration grams of local gradient directions around each found extremum
accuracy was assessed as the vessel line error. The method only in 3D, the matching points are found by comparing the distances
needed two correspondence points to be reliably identified and between feature vectors. An application of this approach to rigid
was therefore applicable even to cases when only a very small registration of peripapillary (ONH-centered) and macula-
overlap between the retinal image pairs existed. Based on the centered 3D OCT scans of the same patient for which the macular
identified vascular features, the general approach can be applied and peripapillary OCT scans had only a limited overlap has been

A B C

Fig. 6.17 Registration of fundus images to two-dimensional optical coherence tomography (OCT) projection data. (A) Fundus camera image.
(B) Two-dimensional projection (through depth dimension) of three-dimensional OCT data. (C) Registered and blended fundus OCT images via
application of affine transformation model with three identified vascular landmarks.
reported.148 The work built on a number of analysis steps intro- needs of retinal disease management, and that the quantified
duced earlier, including segmentation of the main retinal layers retinal exam will become broadly used in systemic disease
and 3D flattening of each of the two volumes to be registered. assessment both for patient-specific care and for population 173
3D SIFT feature points were subsequently determined.150,151 studies.
Using the customary terminology for image registration, when Retinal imaging and image analysis have developed rapidly

Chapter 6
one of the registered images is called the “source” (say the over the past 10 years, and image analysis now plays a crucial
macular image) and the other the “target” (say the peripapillary role in the care of patients with retinal diseases, as well as
image), the feature point detection is performed in both source diseases that manifest in the retina. So far, image analysis has
and target images. After feature point extraction, those which mostly operated reactively, i.e., waiting for what the newest
are in corresponding positions in both images are identified. In imaging devices have as output, and then trying to find
a typical pair of two OCT scans, about 70 matching pairs can be approaches to analyze and quantify the image data. Moving
found with a high level of certainty. The primary deformations forward, we expect that imaging device development and image

Image Processing
that need to be resolved are translation and limited rotation, so analysis research will start to operate more in concert and
simple rigid or affine transform is appropriate to achieve the become closely integrated, so that retinal image analysis suc-
desired image registration. The transform parameters are esti- cesses and difficulties can directly influence device developers
mated from the identified correspondence points. Niemeijer et to focus on details that will help analyze the images reliably,
al. demonstrated the functionality of such an approach to OCT– and vice versa. Ultimately, research and development in retinal
OCT registration of macular and peripapillary OCT scans.149 3D imaging and analysis are driven by the overarching goal of
registration achieved 3D accuracy of 2.0 ± 3.3 voxels, assessed as preventing visual loss and suffering from retinal and systemic
an average voxel distance error in 1572 matched locations. Quali- disease. We expect that this integrated development, in which
tative evaluation of performance demonstrated the utility of this a number of high-profile groups participate worldwide, will
approach to clinical-quality images. Temporal registration of recognize the needs of the developed as well as the develop-
longitudinally acquired OCT images from the same subjects can ing world.
be obtained in an identical manner.
REFERENCES
1. Zhang X, Saaddine JB, Chou CF, et al. Prevalence of diabetic retinopathy in
FUTURE OF RETINAL IMAGING AND the United States, 2005–2008. JAMA 2010;304:649–56.
2. Flick CS. Centenary of Babbage’s ophthalmoscope. Optician 1947;113:246.
IMAGE ANALYSIS 3. Keeler CR. 150 years since Babbage’s ophthalmoscope. Arch Ophthalmol
1997;115:1456–7.
Translation of research in imaging and image analysis into 4. von Helmholtz H. Beschreibung eines Augens-Spiegels zur Untersuchung der
the clinic has been relatively rapid in the past, and can be Netzhaut im lebenden Auge. Berlin: Foerstner; 1851.
5. Trigt AC. Dissertatio ophthalmologica inauguralis de speculo oculi. Utrecht:
expected to accelerate in the future. This is partially explained Universiteit van Utrecht; 1853.
by the lower capital expenditure for ophthalmic imaging devices 6. Gerloff O. Über die Photographie des Augenhintergrundes. Klin Monbl
Augenheilkd 1891;29:397ff.
compared to radiologic imaging devices – the latter can often 7. Gullstrand A. Neue methoden der reflexlosen Ophthalmoskopie. Berichte
be 10–100 times more expensive – and also because retinal Dtsch Ophthalmologische Gesellschaft 1910;36.
8. Novotny HR, Alvis DL. A method of photographing fluorescence in
specialists manage patients directly and are directly involved circulating blood in the human retina. Circulation 1961;24:82–6.
in the ordering and interpreting of images, while radiologists 9. Allen L. Ocular fundus photography: suggestions for achieving consistently
good pictures and instructions for stereoscopic photography. Am J
typically do not directly manage patients. This subtle differ- Ophthalmol 1964;57:13–28.
ence in the physician–patient relationship leads to a more direct 10. Webb RH, Hughes GW. Scanning laser ophthalmoscope. IEEE Trans Biomed
coupling between imaging innovation and clinical impact that Eng 1981;28:488–92.
11. Youngquist RC, Carr S, Davies DE. Optical coherence-domain reflectometry:
is so visible in retinal imaging and analysis. Given the above, a new optical evaluation technique. Optics Lett 1987;12:158–60.
it can be expected that translation of fundamental research 12. Youngquist RC, Wentworth RH, Fesler KA. Selective interferometric sensing
by the use of coherence synthesis. Optics Lett 1987;12:944–6.
findings in retinal imaging will remain rapid in the future. 13. Huang D, Swanson EA, Lin CP, et al. Optical coherence tomography. Science
The need to computerize and automate image interpretation 1991;254:1178–81.
14. Swanson EA, Izatt JA, Hee MR, et al. In vivo retinal imaging by optical
is correspondingly high. A global push toward cost-effective coherence tomography. Optics Lett 1993;18:1864–6.
imaging and image analysis for wide-scale retinal and/or sys- 15. Matsui M, Tashiro T, Matsumoto K, et al. [A study on automatic and quantita-
temic disease detection in a population-screening setting will tive diagnosis of fundus photographs. I. Detection of contour line of retinal
blood vessel images on color fundus photographs (author’s transl).] Nippon
mandate continued efforts in perfecting automated image Ganka Gakkai Zasshi 1973;77:907–18.
analysis.19 16. Baudoin CE, Lay BJ, Klein JC. Automatic detection of microaneurysms in
diabetic fluorescein angiography. Rev Epidemiol Sante Publique 1984;32:
We expect that retinal image analysis and interpretation will 254–61.
be coupled to genetic and other assessment indices, allowing 17. Wild S, Roglic G, Green A, et al. Global prevalence of diabetes: estimates for
the year 2000 and projections for 2030. Diabetes Care 2004;27:1047–53.
truly personalized approaches to complex analyses of broad sets 18. Germain N, Galusca B, Deb-Joardar N, et al. No loss of chance of diabetic
of patient-specific data. On the technological side, it will require retinopathy screening by endocrinologists with a digital fundus camera.
Diabetes Care 2011;34:580–5.
development and wide utilization of highly automated tech- 19. Abramoff MD, Garvin M, Sonka M. Retinal imaging and image analysis. IEEE
niques for combined analysis of retinal image data in 2D, 3D, Rev Biomed Eng 2010:169–208.
and 4D (3D + time), quantification of temporal changes, includ- 20. Fujimoto JG. Optical coherence tomography for ultrahigh resolution in vivo
imaging. Nat Biotechnol 2003;21:1361–7.
ing the assessment of local and/or systemic severity of the find- 21. Fercher AF, Mengedoht K, Werner W. Eye-length measurement by
ings. On the patient management side, it will therefore lead to interferometry with partially coherent light. Optics Lett 1988;13:186–8.
22. Choma M, Sarunic M, Yang C, et al. Sensitivity advantage of swept source
broad utilization of semiautomated, clinician-supervised man- and Fourier domain optical coherence tomography. Optics Express
agement of retinal diseases, especially DR, and choroidal neo- 2003;11:2183–9.
23. Abramoff MD, Kardon RH, Vermeer KA, et al. A portable, patient friendly
vascularization. Overall, we envision that, in the next decade, scanning laser ophthalmoscope for diabetic retinopathy imaging: exudates
the utilization of retinal imaging will go far beyond the direct and hemorrhages. Invest Ophthalm Vis Sci 2007;48 E-Abstract 2592.
24. Hammer DX, Ferguson RD, Ustun TE, et al. Line-scanning laser ophthalmo- 53. Supplement 91: Ophthalmic Photography Image SOP Classes. Available
scope. J Biomed Opt 2006;11:041126. online at: ftp://medical.nema.org/medical/dicom/final/sup91_ft2.pdf.
25. Suansilpong A, Rawdaree P. Accuracy of single-field nonmydriatic digital 54. DICOM. Supplement 110: Ophthalmic Tomography Image Storage SOP Class.
174 fundus image in screening for diabetic retinopathy. J Med Assoc Thai Available online at: ftp://medical.nema.org/medical/dicom/final/sup110_
2008;91:1397–403. ft4.pdf.
26. Delori FC, Pflibsen KP. Spectral reflectance of the human ocular fundus. 55. Abramoff MD, Alward WL, Greenlee EC, et al. Automated segmentation of
Appl Opt 1989;28:1061–77. the optic nerve head from stereo color photographs using physiologically
27. Abramoff MD, Kwon YH, Ts’o D, et al. Visual stimulus-induced changes in plausible feature detectors. Invest Ophthalm Vis Sci 2007;48:1665–73.
Section 1

human near-infrared fundus reflectance. Invest Ophthalmol Vis Sci 2006;47: 56. Tobin KW, Abramoff MD, Chaum E, et al. Using a patient image archive to
715–21. diagnose retinopathy. Conf Proc IEEE Eng Med Biol Soc 2008;2008:5441–4.
28. Whited JD. Accuracy and reliability of teleophthalmology for diagnosing 57. Niemeijer M, Staal JS, van Ginneken B, et al. Comparative study of retinal
diabetic retinopathy and macular edema: a review of the literature. Diabetes vessel segmentation on a new publicly available database. Proc SPIE 2004:
Technol Ther 2006;8:102–11. 5370–9.
29. Lin DY, Blumenkranz MS, Brothers R. The role of digital fundus photography 58. Soares JV, Leandro JJ, Cesar Junior RM, et al. Retinal vessel segmentation
in diabetic retinopathy screening. Digital Diabetic Screening Group (DDSG). using the 2-D Gabor wavelet and supervised classification. IEEE Trans Med
Diabetes Technol Ther 1999;1:477–87. Imaging 2006;25:1214–22.
Optical Imaging Technologies

30. Standards and Guidelines in Telehealth for Diabetic Retinopathy: American 59. Staal J, Abramoff MD, Niemeijer M, et al. Ridge-based vessel segmentation in
Retinal Imaging and Diagnostics

Telemedicine Association position statement. Available online at: http:// color images of the retina. IEEE Trans Med Imaging 2004;23:501–9.
www.americantelemed.org/ICOT/drpositionpaper.paper.technical.htm) 60. Hubbard LD, Brothers RJ, King WN, et al. Methods for evaluation of retinal
(accessed 1 October, 2004). microvascular abnormalities associated with hypertension/sclerosis in the
31. Bresnick GH, Mukamel DB, Dickinson JC, et al. A screening approach to the Atherosclerosis Risk in Communities Study. Ophthalmology 1999;106:
surveillance of patients with diabetes for the presence of vision-threatening 2269–80.
retinopathy. Ophthalmology 2000;107:19–24. 61. Owens DR, Gibbins RL, Lewis PA, et al. Screening for diabetic retinopathy by
32. Kinyoun JL, Martin DC, Fujimoto WY, et al. Ophthalmoscopy versus fundus general practitioners: ophthalmoscopy or retinal photography as 35 mm
photographs for detecting and grading diabetic retinopathy. Invest Ophthal- colour transparencies? Diabet Med 1998;15:170–5.
mol Vis Sci 1992;33:1888–93. 62. Niemeijer M, Xu X, Dumitrescu AV, et al. Automated measurement of the
33. Preferred Practice Pattern: Diabetic Retinopathy. Available online at: www. arteriolar-to-venular width ratio in digital color fundus photographs. IEEE
aao.org/ppp (accessed October 10, 2006). Trans Med Imaging 2011;30:1941–50
34. Fong DS, Aiello L, Gardner TW, et al. Diabetic retinopathy. Diabetes Care 63. Lalonde M, Gagnon L, Boucher M-C. Non-recursive paired tracking for vessel
2003;26:226–9. extraction from retinal images. Proc Conf Vis Interface. 2000:61–8.
35. Fong DS, Sharza M, Chen W, et al. Vision loss among diabetics in a group 64. Gang L, Chutatape O, Krishnan SM. Detection and measurement of retinal
model Health Maintenance Organization (HMO). Am J Ophthalmol 2002;133: vessels in fundus images using amplitude modified second-order Gaussian
236–41. filter. IEEE Trans BioMed Eng 2002;49:168–72.
36. Wilson C, Horton M, Cavallerano J, et al. Addition of primary care-based 65. Hoover A, Goldbaum M. Locating the optic nerve in a retinal image using the
retinal imaging technology to an existing eye care professional referral fuzzy convergence of the blood vessels. IEEE Trans Med Imaging 2003;22:
program increased the rate of surveillance and treatment of diabetic retinopa- 951–8.
thy. Diabetes Care 2005;28:318–22. 66. Foracchia M, Grisan E, Ruggeri A. Detection of optic disc in retinal images by
37. Klonoff DC, Schwartz DM. An economic analysis of interventions for means of a geometrical model of vessel structure. IEEE Trans Med Imaging
diabetes. Diabetes Care 2000;23:390–404. 2004;23:1189–95.
38. Lin DY, Blumenkranz MS, Brothers RJ, et al. The sensitivity and specificity of 67. Lowell J, Hunter A, Steel D, et al. Optic nerve head segmentation. IEEE Trans
single-field nonmydriatic monochromatic digital fundus photography with Med Imaging 2004;23:256–64.
remote image interpretation for diabetic retinopathy screening: a comparison 68. Fleming AD, Goatman KA, Philip S, et al. Automatic detection of retinal
with ophthalmoscopy and standardized mydriatic color photography. Am J anatomy to assist diabetic retinopathy screening. Phys Med Biol 2007;52:
Ophthalmol 2002;134:204–13. 331–45.
39. Williams GA, Scott IU, Haller JA, et al. Single-field fundus photography 69. Li H, Chutatape O. Automated feature extraction in color retinal images by a
for diabetic retinopathy screening: a report by the American Academy of model based approach. IEEE Trans Biomed Eng 2004;51:246–54.
Ophthalmology. Ophthalmology 2004;111:1055–62. 70. Sinthanayothin C, Boyce JF, Cook HL, et al. Automated localisation of the
40. Lawrence MG. The accuracy of digital-video retinal imaging to screen for optic disc, fovea, and retinal blood vessels from digital colour fundus images.
diabetic retinopathy: an analysis of two digital-video retinal imaging systems Br J Ophthalmol 1999;83:902–10.
using standard stereoscopic seven-field photography and dilated clinical 71. Tobin KW, Chaum E, Govindasamy VP, et al. Detection of anatomic structures
examination as reference standards. Trans Am Ophthalmol Soc 2004;102: in human retinal imagery. IEEE Trans Med Imaging 2007;26:1729–39.
321–40. 72. Abramoff MD, Niemeijer M. The automatic detection of the optic disc location
41. Abramoff MD, Suttorp-Schulten MS. Web-based screening for diabetic reti- in retinal images using optic disc location regression. Conf Proc IEEE Eng
nopathy in a primary care population: the EyeCheck project. Telemed J E Med Biol Soc 2006;1:4432–5.
Health 2005;11:668–74. 73. Niemeijer M, Abramoff MD, van Ginneken B. Segmentation of the optic disc,
42. Abramoff MD, Reinhardt JM, Russell SR, et al. Automated early detection of macula and vascular arch in fundus photographs. IEEE Trans Med Imaging
diabetic retinopathy. Ophthalmology 2010;117:1147–54. 2007;26:116–27.
43. Abramoff MD, Niemeijer M, Suttorp-Schulten MS, et al. Evaluation of a 74. Walter T, Klein JC, Massin P, et al. A contribution of image processing to the
system for automatic detection of diabetic retinopathy from color fundus diagnosis of diabetic retinopathy – detection of exudates in color fundus
photographs in a large population of patients with diabetes. Diabetes Care images of the human retina. IEEE Trans Med Imaging 2002;21:1236–43.
2008;31:193–8. 75. Spencer T, Olson JA, McHardy KC, et al. An image-processing strategy for
44. Philip S, Fleming AD, Goatman KA, et al. The efficacy of automated “disease/ the segmentation and quantification of microaneurysms in fluorescein angio-
no disease” grading for diabetic retinopathy in a systematic screening pro- grams of the ocular fundus. Comput Biomed Res 1996;29:284–302.
gramme. Br J Ophthalmol 2007;91:1512–7. 76. Frame AJ, Undrill PE, Cree MJ, et al. A comparison of computer based clas-
45. Cheung N, Rogers S, Couper DJ, et al. Is diabetic retinopathy an independent sification methods applied to the detection of microaneurysms in ophthalmic
risk factor for ischemic stroke? Stroke 2007;38:398–401. fluorescein angiograms. Comput Biol Med 1998;28:225–38.
46. Photocoagulation treatment of proliferative diabetic retinopathy: the second 77. Hipwell JH, Strachan F, Olson JA, et al. Automated detection of microaneu-
report of diabetic retinopathy study findings. Ophthalmology 1978;85:82–106. rysms in digital red-free photographs: a diabetic retinopathy screening tool.
47. Rosenfeld PJ, Moshfeghi AA, Puliafito CA. Optical coherence tomography Diabet Med 2000;17:588–94.
findings after an intravitreal injection of bevacizumab (Avastin) for neovas- 78. Niemeijer M, van Ginneken B, Staal J, et al. Automatic detection of red lesions
cular age-related macular degeneration. Ophthalm Surg Lasers Imaging in digital color fundus photographs. IEEE Trans Med Imaging 2005;24:584–92.
2005;36:331–5. 79. Niemeijer M, van Ginneken B, Russell SR, et al. Automated detection and
48. Heier JS, Antoszyk AN, Pavan PR, et al. Ranibizumab for treatment of differentiation of drusen, exudates, and cotton-wool spots in digital color
neovascular age-related macular degeneration: a phase I/II multicenter, fundus photographs for diabetic retinopathy diagnosis. Invest Ophthalmol
controlled, multidose study. Ophthalmology 2006;113:633.e1–4. Vis Sci 2007;48:2260–7.
49. Preliminary report on effects of photocoagulation therapy. The Diabetic 80. Cree MJ, Olson JA, McHardy KC, et al. A fully automated comparative
Retinopathy Study Research Group. Am J Ophthalmol 1976;81:383–96. microaneurysm digital detection system. Eye (Lond) 1997;11:622–8.
50. Grading diabetic retinopathy from stereoscopic color fundus photographs – 81. Gardner GG, Keating D, Williamson TH, et al. Automatic detection of diabetic
an extension of the modified Airlie House classification. ETDRS report retinopathy using an artificial neural network: a screening tool [see
number 10. Early Treatment Diabetic Retinopathy Study Research Group. comments]. Br J Ophthalmol 1996;80:940–4.
Ophthalmology 1991;98:786–806. 82. Sinthanayothin C, Boyce JF, Williamson TH, et al. Automated detection of
51. Harvey JN, Craney L, Nagendran S, et al. Towards comprehensive population- diabetic retinopathy on digital fundus images. Diabet Med 2002;19:105–12.
based screening for diabetic retinopathy: operation of the North Wales dia- 83. Quellec G, Lamard M, Cazuguel G, et al. Adaptive nonseparable wavelet
betic retinopathy screening programme using a central patient register and transform via lifting and its application to content-based image retrieval. IEEE
various screening methods. J Med Screen 2006;13:87–92. Trans Image Process 2010;19:25–35
52. Sonka M, Fitzpatrick JM. Handbook of medical imaging, vol. 2. Medical image 84. Osareh A, Mirmehdi M, Thomas B, et al. Automated identification of dia-
processing and analysis. Wellingham, WA: International Society for Optical betic retinal exudates in digital colour images. Br J Ophthalmol 2003;87:
Engineering Press; 2000. 1220–3.
85. Al-Diri B, Hunter A, Steel D. An active contour model for segmenting and 115. van Dijk HW, Verbraak FD, Kok PH, et al. Decreased retinal ganglion cell
measuring retinal vessels. IEEE Trans Med Imaging 2009;28:1488–97. layer thickness in patients with type 1 diabetes. Invest Ophthalmol Vis Sci
86. Brinchmann-Hansen O. The light reflex on retinal arteries and veins. A theo- 2010;51:3660–5.
retical study and a new technique for measuring width and intensity profiles 116. Longmuir SQ, Longmuir R, Matthews K, et al. Retinal arterial but not venous 175
across retinal vessels. Acta Ophthalmol Suppl 1986;179:1–53. tortuosity correlates with facioscapulohumeral muscular dystrophy (FSHD)
87. Brinchmann-Hansen O, Sandvik L. The intensity of the light reflex on retinal severity. ARVO Meeting Abstracts 2009;50:5419.
arteries and veins. Acta Ophthalmol 1986;64:547–52. 117. Quellec G, Lamard M, Cazuguel G, et al. Multimodal information retrieval
88. Brinchmann-Hansen O, Sandvik L. The width of the light reflex on retinal to assist diabetic retinopathy diagnosis. ARVO Meeting Abstracts 2009;50:

Chapter 6
arteries and veins. Acta Ophthalmol 1986;64:433–8. 1363.
89. Xu X, Niemeijer M, Song Q, et al. Vessel boundary delineation on fundus 118. Agurto Rios C, Pattichis MS, Murillo S, et al. Detection of structures in the
images using graph-based approach. IEEE Trans Med Imaging 2011;30: retina using AM-FM for diabetic retinopathy classification. ARVO Meeting
1184–91. Abstracts 2009;50:313.
90. Niemeijer M, Xu X, Dumitrescu A, et al. Automated measurement of the 119. Tso DY, Schallek JB, Kardon R, et al. Hemodynamic components contribute
arteriolar-to-venular width ratio in digital color fundus photographs. IEEE to intrinsic signals of the retina and optic disc. ARVO Meeting Abstracts
Trans Med Imaging 2011. 2009;50:4322.
91. Lee S, Reinhardt JM, Cattin PC, et al. Objective and expert-independent 120. Garvin MK, Niemeijer M, Kardon RH, et al. automatically correcting for the
validation of retinal image registration algorithms by a projective imaging presence of retinal vessels on spectral-domain optical coherence tomography

Image Processing
distortion model. Med Image Anal 2010;14:539–49. images decreases variability of the segmented retinal layers. ARVO Meeting
92. Lee S, Abràmoff MD, Reinhardt JM. Retinal image mosaicing using the radial Abstracts 2009;50:1099.
distortion correction model. In: Reinhardt JM, Pluim JP (eds) Medical imaging 121. Lee K, Niemeijer M, Garvin MK, et al. Automated optic disc segmentation
2008: image processing. Proc SPIE 2008;6914:691435–9. from 3D SD-OCT of the optic nerve head (ONH). ARVO Meeting Abstracts
93. Karnowski TP, Aykac D, Chaum E, et al. Practical considerations for optic 2009;50:1102.
nerve location in telemedicine. Conf Proc IEEE Eng Med Biol Soc 2009; 122. Xu Y, Sonka M, McLennan G, et al. MDCT-based 3-D texture classification of
2009:6205–9. emphysema and early smoking related lung pathologies. IEEE Trans Med
94. Lamard M, Cazuguel G, Quellec G, et al. Content based image retrieval based Imaging 2006;25:464–75.
on wavelet transform coefficients distribution. Conf Proc IEEE Eng Med Biol 123. Quellec G, Lee K, Dolejsi M, et al. Three-dimensional analysis of retinal
Soc 2007;2007:4532–5. layer texture: identification of fluid-filled regions in SD-OCT of the macula.
95. Niemeijer M, van Ginneken B, Cree MJ, et al. Retinopathy online challenge: IEEE Trans Med Imaging 2010;29:1321–30.
automatic detection of microaneurysms in digital color fundus photographs. 124. Van Dijk HW, Kok PHB, Garvin M, et al. Selective loss of inner retinal
IEEE Trans Med Imaging 2010;29:185–95. layer thickness in type 1 diabetic patients with minimal diabetic retinopathy.
96. Hoover A, Kouznetsova V, Goldbaum M. Locating blood vessels in retinal ARVO Meeting Abstracts 2009;50:3244.
images by piecewise threshold probing of a matched filter response. IEEE 125. Abramoff MD, Russell SR, Mahajan V, et al. Performance of automated
Trans Med Imaging 2000;19:203–10. detection of diabetic retinopathy does not improve by using the distance of
97. Giancardo L, Abramoff MD, Chaum E, et al. Elliptical local vessel density: a each lesion to the fovea. ARVO Meeting Abstracts 2009;50:3268.
fast and robust quality metric for retinal images. Conf Proc IEEE Eng Med 126. Barriga ES, Russell SR, Pattichis MS, et al. Relationship between visual fea-
Biol Soc 2008;2008:3534–7. tures and analytically derived features in non-exudated AMD phenotypes:
98. Messidor. 2005. Available online at: http://latim.univ-brest.fr/index.php? closing the sematic gap. ARVO Meeting Abstracts 2009;50:3274.
option=com_content&view=article&id=61&Itemid=100034&lang=en) 127. Hu Z, Niemeijer M, Lee K, et al. Automated segmentation of the optic canal
99. Tang L, Niemeijer M, Abramoff MD. Splat feature classification: detection of in 3D spectral-domain OCT of the optic nerve head (ONH) using retinal vessel
large retinal hemorrhages. 2011 IEEE International Symposium on Biomedical suppression. ARVO Meeting Abstracts 2009;50:3334.
Imaging: From Nano to Macro. Chicago, IL. 128. Niemeijer M, Garvin MK, Lee K, et al. Automated segmentation of the retinal
100. Lee K, Garvin MK, Russell S, et al. Automated intraretinal layer segmentation vasculature silhouettes in isotropic 3D optical coherence tomography scans.
of 3-D macular OCT scans using a multiscale graph search. ARVO Meeting ARVO Meeting Abstracts 2009;50:1103.
Abstracts 2010;51:1767. 129. Niemeijer M, Garvin MK, van Ginneken B, et al. Vessel segmentation in 3D
101. Tang L, Kwon YH, Alward WLM, et al. Automated measurement of optic spectral OCT scans of the retina. Medical Imaging 2008: Image Processing
nerve head shape from stereo color photographs of the optic disc: validation 2008;6914:69141R–8.
with SD-OCT. ARVO Meeting Abstracts 2010;51:1774. 130. Niemeijer M, Sonka M, Garvin MK, et al. Automated segmentation of
102. Antony BJ, Tang L, Abramoff M, et al. Automated method for the flattening the retinal vasculature in 3D optical coherence tomography images.
of optical coherence tomography images. ARVO Meeting Abstracts ARVO Meeting Abstracts 2008;49:1832.
2010;51:1781. 131. Davis B, Russell SR, Abramoff MD, et al. Application of independent compo-
103. Mahajan VB, Folk JC, Russell SR, et al. Iowa membrane maps: SD OCT nent analysis to classify non-exudative amd phenotypes quantitatively from
guided therapy for epiretinal membrane. ARVO Meeting Abstracts 2010;51: vision-based derived features in retinal images. ARVO Meeting Abstracts
3604. 2008;49:887.
104. Verbraak FD, Van Dijk HW, Kok PH, et al. Reduced retinal thickness 132. Xu J, Ishikawa H, Wollstein G, et al. Retinal vessel segmentation on SLO
in patients with type 2 diabetes mellitus. ARVO Meeting Abstracts 2010;51: image. Conf Proc IEEE Eng Med Biol Soc 2008;2008:2258–61.
4671. 133. Nelson D, Abramoff MD, Kwon YH, et al. Correlation of progression between
105. Baroni M, Barletta G. Contour definition and tracking in cardiac imaging structure and function in ocular hypertensive patients from the SAFE study.
through the integration of knowledge and image evidence. Ann Biomed Eng ARVO Meeting Abstracts 2008;49:738.
2004;32:688–95. 134. Lee K, Abramoff MD, Niemeijer M, et al. 3-D segmentation of retinal
106. Haeker M, Abràmoff MD, Kardon R, et al. Segmentation of the surfaces of the blood vessels in spectral-domain OCT volumes of the optic nerve head.
retinal layer from OCT images. Lecture Notes Computer Sci 2006;4190: In: Molthen RC, Weaver JB, editors. Medical Imaging 2010: Biomedical
800–7. Applications in Molecular, Structural, and Functional Imaging. Proc SPIE
107. Haeker M, Abramoff MD, Wu X, et al. Use of varying constraints in optimal 2010;7626:76260V.
3-D graph search for segmentation of macular optical coherence tomography 135. Duda RA, Hart PE, Stork DG. Pattern classification. New York: Wiley-
images. Med Image Comput Comput Assist Interv 2007;10:244–51. Interscience; 2001.
108. Haeker M, Wu X, Abramoff M, et al. Incorporation of regional information in 136. Lee S, Reinhardt JM, Niemeijer M, et al. Comparing the performance of retinal
optimal 3-D graph search with application for intraretinal layer segmentation image registration algorithms using the centerline error measurement metric.
of optical coherence tomography images. Inf Process Med Imaging 2007;20: ARVO Meeting Abstracts 2008;49:1833.
607–18. 137. Garvin MK, Sonka M, Kardon RH, et al. Three-dimensional analysis of SD
109. Garvin MK, Abramoff MD, Kardon R, et al. Intraretinal layer segmentation OCT: thickness assessment of six macular layers in normal subjects. ARVO
of macular optical coherence tomography images using optimal 3-D graph Meeting Abstracts 2008;49:1879.
search. IEEE Trans Med Imaging 2008;27:1495–505. 138. Boykov Y, Kolmogorov V. An experimental comparison of min-cut/max-flow
110. Galler KE, Folk JC, Russell SR, et al. Patient preference and safety of bilateral algorithms for energy minimization in vision. IEEE Trans Pattern Anal Mach
intravitreal injection of anti-VEGF therapy. ARVO Meeting Abstracts 2009; Intell 2004;26:1124–37.
50:247. 139. Yang EB, Jin Jones Y, Alward WLM, et al. Comparing resident and fellow
111. Garvin MK, Abramoff MD, Wu X, et al. Automated 3-D intraretinal layer performance on evaluation of stereoscopic optic disc images. ARVO Meeting
segmentation of macular spectral-domain optical coherence tomography Abstracts 2008;49:3626.
images. IEEE Trans Med Imaging 2009;28:1436–47. 140. Tobin KW, Chaum E, Abramoff MD, et al. Automated diagnosis of
112. Reinhardt JM, Lee S, Xu X, et al. Retina atlas mapping from color fundus retinal disease in a large diabetic population. ARVO Meeting Abstracts
images. ARVO Meeting Abstracts 2009;50:3811. 2008;49:3225.
113. Antony BJ, Abramoff MD, Lee K, et al. Automated 3D segmentation of intra- 141. Graff JM, Abramoff MD, Russell SR. Stereo video indirect ophthalmoscopy:
retinal layers from optic nerve head optical coherence tomography images. a novel educational and research tool. ARVO Meeting Abstracts 2008;49:3228.
In: Molthen RC, Weaver JB, editors. Medical Imaging 2010: Biomedical Appli- 142. Abramoff MD, Kardon RH, Vermeer KA, et al. A portable, patient friendly
cations in Molecular, Structural, and Functional Imaging. Proc SPIE 2010; scanning laser ophthalmoscope for diabetic retinopathy imaging: exudates
7626:76260U. and hemorrhages. ARVO Meeting Abstracts 2007;48 E-Abstract:2592.
114. Van Dijk HW, Kok PH, Garvin M, et al. Selective loss of inner retinal 143. Russell SR, Abramoff MD, Radosevich MD, et al. Quantitative assessment of
layer thickness in type 1 diabetic patients with minimal diabetic retinopathy. retinal image quality compared to subjective determination. ARVO Meeting
Invest Ophthalmol Vis Sci 2009;50:3404–9. Abstracts 2007;48:2607.
144. Piette SD, Adix ML, Abramoff MD, et al. Comparison of computer aided 152. Sánchez CI, Hornero R, Mayo A, et al. Mixture model-based clustering and
planimetry between simultaneous and non-simultaneous stereo optic disc logistic regression for automatic detection of microaneurysms in retinal
photographs. ARVO Meeting Abstracts 2007;48:1184. images. In: Karssemeijer N, Giger ML, editors. Medical Imaging 2009:
176 145. Lee S, Abramoff MD, Reinhardt J. Feature-based pairwise retinal image reg- Computer-Aided Diagnosis. Proc SPIE 2009;7260:72601M.
istration by radial distortion correction. Med Imaging: Image Anal Proc SPIE 153. Cree MJ. The Waikato Microaneurysm Detector Univ. Waikato, Tech.
Med Imaging 2007;6512. Rep., 2008. Available online at: http://roc.healthcare.uiowa.edu/results/
146. Tso DY, Schallek J, Kwon Y, et al. Blood flow dynamics contribute to func- documentation/waikato.pdf
tional intrinsic optical signals in the cat retina in vivo. ARVO Meeting 154. Quellec G, Lamard M, Josselin PM, et al. Optimal wavelet transform for the
Section 1

Abstracts 2007;48:1951. detection of microaneurysms in retina photographs. IEEE Trans Med Imag
147. Lee S, Abramoff MD, Reinhardt JM. Retinal atlas statistics from color fundus 2008;27:1230–41.
images. In: Dawant BM, Haynor DR, editors. Medical Imaging 2010: Image 155. Zhang B, Wu X, You J, et al. Hierarchical detection of red lesions in retinal
Processing. Proc SPIE 2010;7623:762310–9. images by multiscale correlation filtering. In: Karssemeijer N, Giger ML,
148. Vermeer KA, Mensink MH, Kardon RH, et al. Super resolution in retinal editors. Medical Imaging 2009: Computer-Aided Diagnosis. Proc SPIE 2009;
imaging. ARVO Meeting Abstracts 2007;48:2766. 7260:72601L.
149. Niemeijer M, Garvin MK, Lee K, et al. Registration of 3D spectral OCT 156. Mizutani A, Muramatsu C, Hatanaka Y, et al. Automated microaneurysm
volumes using 3D SIFT feature point matching. Med Imaging 2009: Image detection method based on double ring filter in retinal fundus images. In:
Optical Imaging Technologies

Processing 2009;7259:72591I–8. Karssemeijer N, Giger ML, editors. Medical Imaging 2009: Computer-Aided
Retinal Imaging and Diagnostics

150. Lowe DG. Distinctive image features from scale-invariant keypoints. Diagnosis. Proc SPIE 2009;7260:72601N.
Int J Comput Vision 2004;60:91–110.
151. Klein R, Chou CF, Klein BE, et al. Prevalence of age-related macular degenera-
tion in the US population. Arch Ophthalmol 2011;129:75–80.
Retinal Diagnostics Section 2

Electrogenesis of the Electroretinogram Chapter

Laura J. Frishman
7 
component waves, progressing from distal retina to proximal
INTRODUCTION retina. As will be described, the ERG is generated by radial cur-
The electroretinogram (ERG) is an electrical potential generated rents arising either directly from retinal neurons, or as a result of
by the retina in response to a change in illumination. It is an the effect on retinal glia of changes in extracellular potassium
excellent tool for evaluating retinal function in both the clinic concentration ([K+]o) brought about by retinal neuronal activity.
and the laboratory because it can be recorded noninvasively Our understanding of the electrogenesis of the ERG was initially
from the corneal surface in vivo under physiological or nearly based on studies in a variety of cold-blooded vertebrate and as
physiological (anesthetized) conditions. However, the ERG well as some mammalian species, described in more detail in
response to a flash of light is complex. It is the summed activity previous reviews.1,2 Studies in a nonhuman primate model
of all retinal cells, and consists of overlapping positive and nega- (macaque monkey) whose retina and ERG are very similar to that
tive component potentials that originate from different stages of of humans (Fig. 7.1),3 and particularly those over the past two
retinal processing. For the ERG to be an effective tool in assess- decades, have improved our understanding of the electrogenesis
ing normal and pathologic retinal activity, it is important that of the ERG in humans. This work will be highlighted wherever
the contributions of the various retinal cell types be distin- possible in this chapter. Clinical applications of the ERG will be
guished and characterized. described in the following chapter (Chapter 8, Clinical electro-
This chapter will review current knowledge of the cellular physiology). Another recent review has examined the electrogen-
origins and mechanisms of generation of the various ERG esis of a common animal model for retinal disease, the mouse.3

Fig. 7.1 Dark- and light-adapted full-field


flash electroretinograms (ERGs) of human
Dark-adapted ERG subjects and macaque monkeys. (A) Top:
(scotopic) Dark-adapted (scotopic) ERGs in response
to brief high-energy flashes from darkness
b occurring at time zero for a normal human
Human subject (left) and an anesthetized macaque
Macaque
monkey (right). The stimulus energy was
~400 sc td/s. Bottom: Light-adapted
100 µV (photopic) flash ERGs in response to
a longer-duration flashes on a rod-saturating
background for a normal human subject (left)
and a macaque monkey (right). For the
human subject the stimulus was a 150-ms
white full-field flash of 4.0 log ph td presented
0 250 500 0 250 500 on a steady background of 3.3 log sc td. For
the macaque, the same stimulus was used,
Time after flash (ms) but the flashes were 200 ms in duration.
(Adapted from Sieving PA, Murayama K,
Naarendorp F. Push–pull model of the
Light-adapted ERG primate photopic electroretinogram: a role
(photopic) for hyperpolarizing neurons in shaping the
b-wave. Vis Neurosci 1994;11:519–32.)
Continued
b d

a 50 µV
40 µV

150 ms 200 ms
A
Stimulator
Rod Cone
178 Ganzfeld RPE
Subretinal
space
Section 2

OS

IS
OLM
Retinal Diagnostics
Retinal Imaging and Diagnostics

ONL
Electrodes
OPL
HC

INL RBC DCB HCB


AII
A17

IPL
OFF
ON GC
GCL
GC

NFL
ILM Müller
Amplifier Vitreous humor

Fig. 7.2 Schematic of rod and cone pathways of the mammalian


retina. The blue-filled cells indicate the cone pathways; the
Computer yellow-filled cells indicate the primary rod pathway. RPE, retinal
pigment epithelium; OS, outer segment; IS, inner segment; HC,
horizontal cell; RBC, rod bipolar cell; DCB, depolarizing (ON) cone
bipolar cell; HCB, hyperpolarizing (OFF) bipolar cell; A, amacrine cell;
GC, ganglion cell: OLM, outer limiting membrane; ONL, outer nuclear
layer; OPL, outer plexiform layer; INL, inner nuclear layer; IPL, inner
plexiform layer; GCL, ganglion cell layer; NFL, nerve fiber layer; ILM,
inner limiting membrane. Scale bar = 20 µm.

Fig. 7.1 Cont’d (B) ERG recording setup: recordings are made of the ERG. In the retina, because all neurons generate light-
using a traditional ganzfeld bowl (left) or more modern light-emitting evoked currents, in principle they should all contribute to the
diode-based full-field stimulator. Burian-Allen and DTL fiber electrodes4 retinal field potentials. However, depending upon various
are illustrated. ERG recordings are amplified and sent to a computer
for averaging, display, and analysis. factors, considered below, the contribution from any particular
cell type could be quite large, or not discernible in the response.
An important factor affecting a given cell’s contribution to the
ERG is its orientation in the retina. Radially oriented neurons in
GENERATION OF EXTRACELLULAR the retina (photoreceptors and bipolar cells) and glial cells
(Müller cells and retinal pigment epithelial (RPE) cells) make
POTENTIALS: GENERAL CONCEPTS larger contributions to the ERG than cells that are oriented more
Extracellular potentials that can be recorded noninvasively, such irregularly or laterally (e.g., horizontal and amacrine cells) (Fig.
as the ERG (and visual evoked potential of the cortex), are the 7.2). The currents around cells that underlie the ERG enter the
result of localized conductance changes in the membranes of ECS at one retinal depth (the current source), and return into the
activated cells that give rise to inward or outward currents. cell at another depth (the current sink), creating a current dipole.
These currents also flow in the extracellular space (ECS) and Although most of the extracellular current flowing from source
create extracellular potentials. The current flowing through the to sink traverses the ECS within the retina, some travels extra-
conductive fluid surrounding a cell whose activation has given retinally – through the vitreous humor, extraocular tissues,
rise to a local current is directed mainly toward the relatively sclera, choroid, the high resistance of the RPE (R-membrane),
less activated parts of the cell. Thus, when neurons are arranged and back into the neural retina.
so that the extracellular currents of many synchronously acti- The polarity and amplitude of the recorded ERG will depend
vated cells all flow in the same direction, the resulting extracel- upon the location of the active and reference electrodes. In non-
lular potential change, called a “field potential,” may be large invasive studies, a common location for the active electrode is
enough to be recorded at a distance, e.g., at the cornea in the case on the cornea via a contact lens electrode, e.g., as illustrated in
Fig. 7.1B, a Burian-Allen electrode, or jet electrode, or another
type of surface conductive electrode (e.g., a DTL fiber electrode,4 Subretinal
H–K loop,5 or gold foil electrode). For comfort, skin electrodes, space R R 179
OLM
which yield smaller signals, are sometimes used instead. In inva-
sive studies of ERG components in animals, the active electrode

Chapter 7
may be positioned anywhere in the current path, including at i2
different retinal depths, near particular cell types. The reference
electrode also can be positioned anywhere in the path, but is
often placed behind the RPE in studies of isolated retina, or INL
retrobulbar in intact eyes. In noninvasive and clinical applica- r2
tions, the reference is positioned either under the eyelids, such
as the speculum of the Burian-Allen electrode for bipolar record-

Electrogenesis of the Electroretinogram


ing between the contact lens electrode and the reference on the
same eye, or remote from the eye (e.g., on the temple) for mono-
polar recording. The exact position of the remote reference is of
minor consequence except for possible contamination of the IIPL
retinal signal by other sources.
Other factors that influence the magnitude of the contribution
to the ERG of a particular cell type include stimulus conditions, r1
i1
such as the strength of the stimulus and its wavelength (spec-
trum), the background illumination (that determines adaptation ILM
level of the retina), the duration and spatial extent of the stimu-
lus, and the location of the stimulus within the visual field, as Vitreous body
these stimulus parameters have different effects on the responses
of the different cells. For example, the relative contributions of Fig. 7.3 Model of K+-induced flow of current through Müller cells and
various cell types are different under dark-adapted (scotopic) extracellular space. The Müller cell K+ currents (grey arrows) are
and light-adapted (photopic) conditions when rod and cone induced by [K+]o increases in the inner plexiform layer or [K+]o
pathways, respectively, are involved in generating responses. decreases in the subretinal space (SRS); the return currents in the
extracellular space are carried by Na+ and Cl– ions. K+ enters the
Spatially extended diffuse stimuli, i.e., full-field (ganzfeld) Müller cell via strongly rectifying Kir 2.1 channels in and around the
flashes that fill the retina evenly, using stimulators such as those synaptic layers (short arrows on the left) and leave the cell via weakly
illustrated in Fig. 7.1B, are commonly used to elicit the major rectifying Kir 4.1 channels near the vitreous body, blood vessels, and
subretinal space (bidirectional arrows on the bottom). The I2 current
ERG waves (a-, b-, and d-waves) from photoreceptors and
generates slow PIII. (Adapted from Frishman LJ, Steinberg RH.
bipolar cells. Contributions from these cells generally increase Light-evoked increases in [K+]o in proximal portion of the dark-adapted
with the area of the retina stimulated as the number of cells, and cat retina. J Neurophysiol 1989;61:1233–43 and Kofuji P, Biedermann
hence the total extracellular current, is increased. In contrast, B, Siddharthan V, et al. Kir potassium channel subunit expression in
retinal glial cells: implications for spatial potassium buffering. Glia
contributions of retinal ganglion cells (and other cells with 2002;39:292–3.)
antagonistic regions within their receptive fields) to the full-field
flash ERG will be limited by the strength of surround antago-
nism. For photopic ERGs, particularly from subjects with trichro-
matic color vision, such as the macaque and human, stimulus from the ECS enters the Müller cells via inwardly rectifying K+
wavelength also affects contributions from cells whose responses channels and is carried radially as an intracellular (spatial buffer)
are dependent upon spectral antagonism.6,7 current to regions of lower [K+]o. Thus a current loop is set up:
the current inside the Müller cell is carried by K+ and, to com-
Spatial buffering by glial cells plete the circuit, the dominant extracellular ions, Na+ and Cl–,
Spatial buffering of [K+]o by radially oriented Müller cells, RPE carry the extracellular return current. Because the magnitude of
cells, and perhaps astrocytes in the optic nerve head is an impor- the [K+]o changes depends upon the integral of K+ flow rate into
tant mechanism for generating the currents underlying several the ECS, ERG components that reflect this glial current will be
ERG components, to be described later in this chapter, e.g., slower than components that reflect the currents around neurons.
c-wave, slow PIII, tail of the b-wave, scotopic threshold response This “slowing” would be equivalent to low-pass filtering of the
(STR), M-wave, and photopic negative response (PhNR). Due to neuronal signal.
its importance in generating ERGs, an overview of [K+]o spatial The electrical properties of the Müller cell membrane are
buffering in Müller cells will be presented here. important for the creation of spatial buffer currents. The mem-
[K+]o spatial buffering is important for maintaining the electro- brane is selectively permeable to K+,8,9 but the K+ conductance is
chemical gradients across cell membranes necessary for normal not distributed evenly over the cell surface. Instead, it is concen-
neuronal activity and for minimizing the changes in local [K+]o trated in the vicinity of extracellular sinks (i.e., the vitreous body,
that occur as a consequence of neuronal activation. Membrane subretinal space, and blood vessels). This regional distribution
depolarization leads to the leak of K+ from neurons, causing [K+]o facilitates “K+ siphoning” from synaptic areas where [K+]o is
to be elevated, particularly in synaptic layers of the retina (Fig. high, to those regions of high K+ conductance where [K+]o is
7.3); membrane hyperpolarization leads to reduced [K+]o as the lower.10,11 In mouse retina, as indicated in Fig. 7.3, strongly
leak conductance is reduced, but the Na+-K+ ATPase in the inward rectifying Kir 2.1 channels have been localized to synap-
membranes continues to pump K+ into (and Na+ out of) cells. K+ tic layers (small arrows on the left) where K+ moves from the ECS
into Müller cells, whereas less strongly rectifying Kir 4.1 chan- photoreceptor that generate the scotopic a-wave,29 or rod bipolar
nels at the extracellular sinks allow K+ to leave the Müller cell.12 cells and the scotopic b-wave in mammals.30,31 Correlation also
180 may be useful for identifying the origin of a response property,
APPROACHES FOR DETERMINING THE such as oscillatory potentials (OPs) in the ERG, and the light-
ORIGINS OF THE ELECTRORETINOGRAM evoked oscillatory behavior in amacrine cells as a possible source
for the potentials. However, if currents from several cell types
Section 2

Historically, several different approaches have been used to contribute to a local field potential, the relationship between
determine the neuronal origins and cellular mechanisms of field potential and local cellular responses may be difficult to
generation of the ERG. determine without using other tools, such as pharmacologic
agents.
Intraretinal depth recordings
A microelectrode positioned at some locus in the retinal ECS Pharmacologic dissection
Retinal Diagnostics
Retinal Imaging and Diagnostics

records a field potential called the “local” or “intraretinal” ERG.13 The use of pharmacologic agents that have specific effects on
The recorded potential reflects electrical activity of the cells cellular functions has been very helpful in determining origins
located near the microelectrode tip, and when a local stimulus, of ERG components. In Granit’s classical pharmacologic study
such as a small spot of light, is used, the local activity will be the of the dark-adapted ERG of the cat, he observed that compo-
entire signal. However, when full-field diffuse flashes are used, nents disappeared sequentially during induction of ether anes-
currents can be sufficiently large to produce a corneal ERG thesia.32 He called the components “processes” and numbered
simultaneously with the local ERG. Local potentials with a them in the order of disappearance: PI, the positive c-wave, was
similar timecourse to that of the corneal ERG components can first to leave, then PII, the positive b-wave, disappeared, and
be helpful in locating the cells of origin. However, this type of finally, PIII, the negative a-wave. We now know that these pro-
analysis has some complications: cesses correspond roughly to RPE, bipolar, and photoreceptor
cell contributions to the ERG respectively. The terms PII and PIII
1. Field potentials that spread over long distances will are still used.
superimpose in space and time, making it difficult to locate In recent years, much has been learned about retinal microcir-
the cells of origin with certainty. cuitry and biophysics, including information at cellular and
2. Retinal resistivity varies between and within retinal molecular levels about retinal neurotransmitters (their identity,
layers,14,15 which causes currents passing through layers of release mechanisms, and receptors), signal transduction cas-
different resistance to set up complex voltages. cades, ion channels, and other cellular proteins. This knowledge
has allowed better use of pharmacologic tools in isolating ERG
Both of these problems occur in the intact eye when a scleral components and in interpreting experimental observations. For
reference is used, and the local signal recorded with a microelec- example, specific knowledge about glutamatergic neurotrans-
trode is contaminated by the diffuse ERG, due to the high resis- mission in the retina and appropriate agonists and antagonists
tance of the RPE and sclera. Some of this contamination can be for specific receptors has improved our understanding of the
eliminated by using a vitreal reference.16 major waves of the ERG, including the ERG in primates.33–35 Use
Current source density (CSD) or “source-sink” analysis can of the voltage-gated Na+ channel blocker tetrodotoxin (TTX) has
provide a solution to these problems. Local field potentials are made it possible to identify ERG components resulting from the
measured and analyzed, but in addition, radial resistance is Na+-dependent spiking activity of inner retinal cells.
taken into account to obtain direct estimates of radial current.17
The result is a spatiotemporal profile of relatively well-localized Site-specific lesions/pathology or
current sources and sinks that can be compared with the retinal
structure (layers) and physiology. CSD analysis has elucidated
targeted mutations
the origins of particular ERG components (e.g., for the bipolar Removal of a cell type or types or circuits allows assessment of
cell origin of the b-wave).18,19 their role in the electrogenesis of the ERG. A specific cell type
For ERG components believed to depend specifically on glial can be lesioned selectively (e.g., retinal ganglion cells as a con-
K+ spatial buffer currents, intraretinal depth recordings with sequence of optic nerve section) or lost due to pathologic changes
ion-selective microelectrodes have been used to locate the retinal (e.g., ganglion cells in glaucoma), or inherited degenerations
layer(s) where neurally induced changes of [K+]o were largest (e.g., rod and/or cone dystrophy). Cellular functions, such as
and most similar in timecourse to a particular component.20–25 light responses or synaptic transmission, can be abnormal or
Application of barium (Ba2+) to block Kir channels in glial cell eliminated due to inherited or acquired conditions, or targeted
membranes,26,27 and the ERG components dependent upon the genetic manipulation, most commonly done in mice.
spatial buffer currents, or genetic inactivation of Kir4.1 channels
in mice,28 have been used to provide evidence for the role of Modeling of cellular responses and
Müller cells in generating certain slow waves of the ERG. ERG components
As our understanding of the function of retinal cell types has
Correlation of ERG with improved, it has been possible to develop quantitative models
single-cell recordings that predict the light responses of those cells, and to apply the
Correlations of the ERG with single-cell electrophysiology are models to the analysis of the ERG. Models based on suction
most useful when the light-evoked currents from a particular cell electrode recordings from single photoreceptor outer segments
type are the primary determinant of an ERG component, as is have been used to predict the leading edge of the a-wave,36–38
the case for rod photoreceptors and the currents around the although more proximal portions of the photoreceptor cell will
Dark-adapted 0.01 ERG Dark-adapted 3.0 ERG Dark-adapted 3.0
(rod response) (combined rod-cone response) oscillatory potentials
181
Peak time

Chapter 7
b
b

Electrogenesis of the Electroretinogram


Light-adapted 3.0 ERG Light-adapted 3.0 flicker
(rod response)

b Rod/Cone Oscillatory
t
ERGs potentials
100 30 µV

a 20 10 ms

Fig. 7.4 Five standard electroretinogram (ERG) tests with full-field stimulation recommended by the International Society for the Clinical
Electrophysiology of Vision (ISCEV) for use worldwide in clinical electrodiagnostic facilities. This figure shows examples, but not norms, of ERG
responses in the recommended tests in normal humans. Light calibrations in candela seconds per meter squared (cd s/m2) for each test are
indicated above the ERGs. Large arrowheads indicate time at which the stimulus flash occurred. Dotted arrows show common ways to measure
the time-to-peak (t, implicit time), and a- and b-wave amplitude. (Reproduced with permission from Marmor MF, Fulton AB, Holder GE, et al.
ISCEV standard for full-field clinical electroretinography (2008 update). Doc Ophthalmol 2009;118:69–77.)

also participate in its generation.39 The models have been Box 7.1 Standard and more specialized electroretinogram
extended to predict the leading edge of the scotopic b-wave.30 (ERG) tests
Models of stimulus–response relations of specific retinal cells
can be used to analyze amplitude versus energy curves obtained Standard ERG tests described by ISCEV standard for full-field
clinical electroretinography (2008 update);42 all numbers are
from ERG measurements into components related to the differ- stimulus calibrations in cd/s/m2
ent cell types.40,41 Dark-adapted 0.01 ERG (“rod response”)
Dark-adapted 3.0 ERG (“maximal or standard combined
STANDARD ERG TESTS IN THE CLINIC rod–cone response”)
Dark-adapted 3.0 oscillatory potentials (“oscillatory potentials”)
Standard and more specialized tests for use in the clinic that Light-adapted 3.0 ERG (“single-flash cone response”)
examine key aspects of light- and dark-adapted retinal (and Light-adapted 3.0 flicker ERG (“30-Hz flicker”)
Recommended additional response: either dark-adapted 10.0
more central visual) function have been described in various
ERG or dark-adapted 30.0 ERG
publications by the International Society for the Clinical Electro-
Specialized types of ERG and recording procedures
physiology of Vision (ISCEV), with the most recent update for Macular or focal ERG
the flash and flicker ERG in 2008.42 The “standard” tests advo- Multifocal ERG*
cated by ISCEV for basic ERG testing are listed in Box 7.1, and Pattern ERG*
Early receptor potential (ERP)
typical responses to these tests are illustrated in Figure 7.4. The Scotopic threshold response (STR), negative and positive
standards were developed so that ERGs recorded in clinics Photopic negative response (PhNR)
around the world would be comparable. The ISCEV publica- Direct-current (dc) ERG
tions, which now cover several other ERG tests as well, i.e., the Electro-oculogram*
Long-duration light-adapted ERG (ON–OFF responses)
ones with an asterisk in Box 7.1, describe basic technology and Paired-flash ERG
clinical protocols. Chromatic stimulus ERG (including S-cone ERG)
Dark and light adaptation of the ERG
DISTAL RETINAL COMPONENTS: SLOW Dark-adapted and light-adapted luminance response analyses
Saturated a-wave slope analysis
PIII, C-WAVE, FAST OSCILLATION Specialized procedures for young and premature infants*
TROUGH, AND LIGHT PEAK
*See relevant standard or guideline published by International Society for the
After the onset of a step of light, the early waves of the Clinical Electrophysiology of Vision (ISCEV).
dark-adapted ERG, the a- and b-waves, are followed by the
c-wave and then by a succession of slower responses that steady long enough for them to develop. Therefore, in the
include the fast oscillation trough (FOT), which is a negative clinic, these slower responses are generally recorded by using
deflection, and the light peak, which is a large slow positive electro-oculography.
deflection (Fig. 7.5A). Because these responses are so slow, The electro-oculogram (EOG) is an eye movement-dependent
lasting seconds to minutes, patients cannot keep their eyes voltage recorded between electrodes placed near the eye at the
inner and outer canthus. The patient is asked to look back and RPE have been recorded in anesthetized animals by placing a
forth between a pair of fixation lights separated by 30° of visual microelectrode in the subretinal space, and simultaneously
182 angle, situated in a ganzfeld bowl. The source of the voltage is recording the potentials across neural retina and the RPE, as
a corneofundal potential, also called the “standing potential” illustrated in the schematic in Fig. 7.5B. Such experiments have
that renders the cornea positive with respect to the back of the provided a good understanding of the origins and mechanisms
eye. Light-evoked changes in the EOG reflect changes in the of generation of the c-wave and other slow potentials from
Section 2

transepithelial potential (TEP) of the RPE. These changes have distal retina.
been studied experimentally in human and animal preparations
using direct current electroretinography (dc-ERG43), and these c-Wave
studies will be reviewed briefly here. The cornea-positive c-wave that follows the b-wave is the sum
Electrogenesis of the c-wave, FOT, and light peak of the of two major (sub)component voltages: a cornea-negative
dc-ERG involves ion concentration changes in the subretinal voltage, generated by the neural retina, and a cornea-positive
Retinal Diagnostics
Retinal Imaging and Diagnostics

space between photoreceptors and the RPE that in turn produce voltage of similar latency and timecourse, generated by the RPE
slow membrane responses in the Müller and RPE cells that (Figs 7.5 and 7.6). The c-wave is cornea-positive when the RPE
face the space. The Müller and/or RPE component voltages component is larger than the neural retinal component. If the
overlap in time and sum to produce the recorded dc-ERG com- two components are equal in amplitude, the c-wave will be
ponents. The (sub)component voltages from Müller cells and absent, as observed in some monkeys.44

A B
RPE
+
C-wave Vitreal
ERG
Transretinal
+
Transepithelial
Transepithelial
+

Vitreous

Neural retina
Transretinal Subretinal space
RPE
Choride
Sclera
Slow PIII Retrobulbar

2 mV
Light peak

C-wave
2nd C-wave

Vitreal
Fast oscillation trough

5 min

Fig. 7.5 Subretinal recordings from the intact cat eye. (A) The vitreal, transretinal, and transepithelial potentials were recorded simultaneously in
response to a 5-minute period of illumination. The a- and b-waves cannot be seen using this compressed timescale. In the vitreal electroretinogram
(ERG), the c-wave is followed by the fast oscillation trough (FOT) and then the light peak. The intraretinal recordings show that the c-wave is
composed of two (sub)components: the larger cornea-positive retinal pigment epithelial (RPE) transepithelial response, and the slightly smaller
cornea-negative transretinal component, the Müller cell-generated slow PIII response. For the light peak, only an RPE component is present.
(Reproduced with permission from Steinberg RH, Linsenmeier RA, Griff ER. Retinal pigment epithelial cell contributions to the electroretinogram
and electrooculogram. Prog Retin Res 1985;4:33–66.) (B) Schematic showing the recording arrangement for transretinal and transepithelial
recordings in the intact eye. The transretinal ERG is recorded between a vitreal reference and a retrobulbar reference. The microelectrode is
referenced to the vitreal reference for the transretinal recording and to the retrobulbar reference for the transepithelial recording. Double-barreled
microelectrodes were used to measure field potentials and changes in [K+]o. (Reproduced with permission from Frishman LJ, Steinberg RH.
Light-evoked increases in [K+]o in proximal portion of the dark-adapted cat retina. J Neurophysiol 1989;61:1233–43.)
Fig. 7.6 The components of c-wave of the
dark-adapted cat (DC) electroretinogram
A B (ERG): the (sub)components, and correlation
b c
Vitreal of recorded [K+]o and the retinal pigment 183
epithelial (RPE) apical membrane
hyperpolarization. Stimuli were 4-second
flashes at 8.3 log q deg2/s2. (A) The vitreal

Chapter 7
c-wave consists of a transepithelial
component (TEP c-wave) and a transretinal
component (slow PIII). B-wave deflections
Vap can be seen in both recordings; the b-wave
current generated in neural retina creates a
passive voltage drop across the large
10 mV resistance of the RPE and sclera. (B) RPE
Transepithelial apical membrane and subretinal [K+]o in

Electrogenesis of the Electroretinogram


response to the same stimulus as in part A,
recorded in a separate experiment. The
Vk+ apical membrane potential was derived by
Transretinal subtracting an intracellular recording of the
5.5 basal membrane potential from the
transepithelial potential. (Reproduced with
4.5 mM permission from Steinberg RH, Linsenmeier
RA, Griff ER. Retinal pigment epithelial cell
2 mV 3.5 contributions to the electroretinogram and
electrooculogram. Prog Retin Res 1985;4:
33–66.)
4 sec 4 sec

There is long-standing evidence that two components of oppo- to both the subretinal [K+]o decrease and to slow PIII.20,21,53 Further,
site polarity form the ERG c-wave. For example, intravenous when Ba2+ was used to block Müller cell Kir channel conduc-
injection of sodium iodate in rabbit, which poisons primarily the tances, slow PIII was suppressed but there was little effect on the
RPE, abolishes the cornea-positive c-wave and leaves a cornea- light-evoked subretinal K+ decrease.22,27,54 Finally, slow PIII was
negative potential,45 as occurs in vitro when recording from an not present in ERGs of mice with Kir 4.1 channels, the dominant
isolated neural retina preparation.46 Microelectrode recordings Kir channels in Müller cells, genetically inactivated.28
in retinas of several species,43 including monkey,47 have con-
firmed the presence of the two components. An example of such Distal versus proximal PIII
recordings in intact cat eye is shown in Fig. 7.6. The component Intraretinal depth recordings in isolated rabbit retina have iden-
from the neural retina is commonly termed slow PIII, to distin- tified a component of similar timecourse and polarity to slow
guish it from fast PIII, the photoreceptor current. The component PIII that is eliminated by aspartate, and therefore, unlike slow
from the RPE is the RPE c-wave. PIII, is generated by cells proximal to the photoreceptors.55 Proxi-
Both slow PIII and the RPE c-wave are responses to the light- mal PIII is now thought to originate from Müller cell K+ currents
evoked decrease in [K+]o in the subretinal space that occurs in that flow in the same direction in the retina as slow PIII currents.
response to intense light stimulation of the dark-adapted retina. However, the proximal PIII currents are initiated by an increase
When measurements of [K+]o were made with ion-selective in [K+]o due to neuronal activation in proximal retina, rather than
microelectrodes, either in intact eyes or in vitro preparations, the the decrease in [K+]o in the subretinal space. The term “proximal
timecourse of the [K+]o decrease was found to predict that of the PIII” is not commonly used now that responses have been identi-
ERG c-wave and its component parts (Fig. 7.6). Blocking K+ fied that are Müller cell, or perhaps astrocyte-mediated responses
conductance (via Kir channels) with various agents eliminated to [K+]o changes in proximal retina, e.g., STR and PhNR (described
both the slow PIII48 and the RPE c-wave.49 in later sections).
Müller cell contribution (slow PIll)
Intraretinal recording at various depths50 have shown that slow Retinal pigment epithelial component
PIII is generated by a radially oriented current across the neural The RPE c-wave is a cornea-positive potential that reflects an
retina. A Müller cell generator, rather than a neuronal generator, increase in the TEP of the RPE, a major component of the stand-
was suggested because slow PIII persisted after treatment with ing potential of the eye. The TEP exists because the apical and
aspartate, a nonselective glutamate agonist, to suppress all basal membranes of RPE cells are electrically separated by high-
responses of postreceptoral neurons.51 resistance tight junctions that encircle the monolayer of cells (the
Studies in amphibia and mammals have shown that slow PIII “R membrane”). The TEP is equal to the difference between the
is initiated when the distal ends of the Müller cells are passively apical (Vap) and basal (Vba) membrane potentials.43 Vap is gener-
hyperpolarized by a photoreceptor-dependent decrease in sub- ally more hyperpolarized than Vba, making the TEP cornea-
retinal [K+]o. This sets up a transretinal “K+ spatial buffer” positive. During c-wave generation in response to an increase in
current,25 and the current drop across the extracellular resistance light, the TEP increases (becomes even more positive). This is
produces the slow PIII voltage.50,52,53 The slow hyperpolarization initiated by a hyperpolarization of the apical membrane, and
recorded in Müller cells was observed to be similar in timecourse passive shunting of current to the basal membrane, resulting in
a (smaller) hyperpolarization of basal membrane, and a greater support its being the “light peak substance.”61 Epinephrine also
difference in potential between the two membranes.43 has been proposed as a candidate, and a role for ligands binding
184 As was observed for Müller cells, the slow hyperpolarization to adrenergic alpha-1 receptors on the apical membrane is
of the apical membrane, with its large K+ conductance,56 and the likely.59 Cyclic AMP has been investigated as a second messen-
RPE c-wave have a timecourse that is very similar to the subreti- ger in light peak generation but, as described above, it may be
nal [K+]o decrease, as illustrated in Fig. 7.6. In an isolated RPE
Section 2

involved in generation of the FOT, rather than the light peak.59


preparation (where only the apical bath [K+] was altered), Oakley
et al.57 demonstrated that the RPE c-wave was due solely to the ORIGIN OF THE A-WAVE
[K+]o decrease.
Fig. 7.7 shows the dark- and light-adapted flash ERG of macaque
The fast oscillation trough monkey, a good animal model for studying the origins of
The FOT (usually measured by EOG) is a change in the corneo- the human response, as illustrated in Fig. 7.1. The a-wave
Retinal Diagnostics
Retinal Imaging and Diagnostics

retinal potential – it decreases and increases in synchrony with in the dark-adapted ERG is the initial negative wave that occurs
an alternating light/dark stimulus. The response in the dc-ERG in response to strong stimuli from darkness (Fig. 7.7, top left
that corresponds to the EOG decrease (trough) also is termed the column) and it is primarily rod-driven (scotopic), but contains a
FOT. The FOT response to maintained illumination follows the cone contribution when flashes are very strong. When the back-
c-wave peak, and, when a light peak occurs, it appears as a dip ground is rod-saturating the a-wave is cone-driven (photopic;
between the c-wave and the light peak (Fig. 7.5A). right column). Under both dark- and light-adapted conditions,
The FOT originates from both neural retina and RPE. It the a-wave is truncated by the rise of the positive-going b-wave
involves recovery of Müller and RPE cells from their peak polar- that originates primarily from ON bipolar cells,30 as reviewed
izations as subretinal K+ reaccumulates following the reduction below. The slow negative wave in the dark-adapted ERG in
in concentration caused by light. This recovery may be greater response to the weakest stimuli, called the STR, is not the a-wave,
than predicted by the reaccumulation, particularly for the RPE but instead is initiated by amacrine and/or ganglion cell activity.
component. Light initially elicits a hyperpolarization of the This is known because STR is eliminated when postreceptoral
apical membrane that increases the TEP and then produces a activity is blocked pharmacologically.63,64
delayed basal hyperpolarization that decreases the TEP. This
extra decrease in TEP underlies most of the cornea-negative
The a-wave as a reflection of rod and cone
potential of the FOT.43 receptor photocurrent
The ionic mechanisms of the basal membrane hyperpolariza- Early intraretinal depth studies in intact cat eyes13,65 found that
tion involve Cl– conductances.58 In intact sheets of RPE/choroid the signal at the timecourse of the a-wave was largest in the vicin-
from human fetal eyes, the Miller lab distinguished two types of ity of the photoreceptors. The case for a receptoral origin for PIII
basal membrane Cl– channels: a 4,4’-diisothiocyanostilbene-2, was strengthened by microelectrode recordings in macaque
2’-disulfonate (DIDS)-inhibitable Ca2+-sensitive Cl– channel, and monkey retina, with the inner retinal circulation clamped to
a cyclic AMP-dependent channel that is inhibited by DIDS as suppress retinal activity proximal to the photoreceptors.66
well as by 5-nitro-2-(3phenylpropylamino) benzoate (NPPB), Penn and Hagins67 in CSD analyses of the isolated rat retina
which identifies it as a cystic fibrosis transmembrane regulator demonstrated that light suppressed the circulating (dark) current
(CFTR) channel.59 In CF patients, the FOT, but not the light of the photoreceptors, and proposed that this suppression was
peak (see below) is reduced, implicating CFTR in generation of seen in the ERG as the a-wave. Figure 7.8 provides a schematic
the FOT. of the photoreceptor layer current from a review by Pugh et al.68
The figure shows that, in the dark, cation channels (Na+, Ca2+,
The light peak and Mg2+) in the receptor outer segment (ROS) are open, current
Maintained illumination causes a slow increase in the standing flows into the ROS (a current sink with respect to the ECS), and
potential in the dc-ERG called the light peak that can be recorded K+ leaks out of the inner segment (a current source), creating
as a slow oscillation of the EOG (Fig. 7.6). Intraretinal recordings dipole current. This dipole current produces a corneal (and
in several species,43,58 including monkey,47 have shown that this vitreal) potential that is positive with respect to the scleral side
cornea-positive potential originates solely from an increase in of the retina. Suppression of the dark current reduces the cornea-
the TEP (Fig. 7.5A). Intracellular RPE recordings localized the positive potential, creating the negative-going a-wave. Consis-
origin of the increase to a slow depolarization of the basal mem- tent with this view, as illustrated in Fig. 7.9, intraretinal
brane caused by an increase in basal Cl– conductance. In both recordings and CSD analyses in the intact macaque retina have
chick RPE and human RPE cell sheets the Cl– conductance localized current sources and sinks for a local potential, corre-
increase was suppressed by DIDS.58,59 In mouse, the light peak sponding to the a-wave, to the distal third of the retina.18
is also dependent on a Cl– conductance, and it is regulated by Hood and Birch38 sought to relate the timecourse of the leading
voltage-dependent Ca2+ channel CaV1.3 subunits.60 edge of the ERG a-wave directly to the leading edge of the pho-
Although the light peak voltage originates from the RPE basal toreceptor outer-segment response to light. They demonstrated
membrane, it is then initiated in neural retina via the photore- that both the linear and the nonlinear (i.e., saturating) behavior
ceptors.61 Light stimulation leads to a change in concentration of of the leading edge of the a-wave in the human ERG could be
a “light peak substance” which then affects the basal membrane predicted by a model of photoreceptor function derived from in
via a second-messenger system. The identities of the “light peak vitro suction electrode recordings of currents around the outer
substance” and the second messenger(s) involved in producing segments of primate rod photoreceptors.69 Subsequently a sim-
the light peak are unresolved. Although dopamine affected the plified kinetic model of the leading edge of the photoreceptor
light peak in the perfused cat eye,62 studies in chick did not response (in vitro current recordings) that took into account the
Dark-adapted Light-adapted 185
ND

Chapter 7
Sc Tds
0
396 100

µV
0.3
0

Electrogenesis of the Electroretinogram


0.7
0.27

20 1.0
1.12 x 10–3

µV
0
1.5 x 10–4 1.3

0 250 500 220 ms flash


Time after flash (ms)

Fig. 7.7 Dark- and light-adapted electroretinograms (ERGs) of the macaque retina. (Left) Dark-adapted full-field ERGs of anesthetized macaque
monkeys were measured in response to brief (<5 ms) flashes from darkness generated by computer-controlled light-emitting diodes (LEDs).
Responses were recorded differentially between DTL fibers on the two eyes. Flash strength was increased over a 6-log-unit range. Responses
to the weakest stimuli were rod-driven (scotopic), whereas for the strongest stimuli there were mixed rod–cone responses. (Reproduced with
permission from Robson JG, Frishman LJ. Dissecting the dark-adapted electroretinogram. Doc Ophthalmol 1998;95:187–215.) (Right) Light-
adapted full-field ERGs of anesthetized macaques were measured using Burian-Allen electrodes. Stimulus strength was controlled with neutral-
density (ND) filters. Stimulus strength increased over a 1.3-log-unit range to a maximum at 0 ND of 4.0 log ph td for 220-ms flashes. Stimuli
were presented on a steady rod-saturating background of 3.3 log sc td. The 20 µV calibration bar applies both to the dark-adapted ERG
responses to weak stimuli and to the light-adapted ERGs. (Reproduced with permission from Sieving PA, Murayama K, Naarendorp F. Push–pull
model of the primate photopic electroretinogram: a role for hyperpolarizing neurons in shaping the b-wave. Vis Neurosci 1994;11:519–32.)

Fig. 7.8 Schematic of a longitudinal section


of the mammalian retina illustrating how
circulating currents in the photoreceptor layer
create an extracellular field potential, in which
the vitreal side of the retina has a more
positive potential than the scleral side. A
AII very bright flash will suppress the circulating
current around the photoreceptor, leading
to a vitreal or corneal response which is
negative relative to the resting preflash
Rod baseline. (Reproduced with permission from
Pugh EN Jr, Falsini B, Lyubarsky A. The
RB origin of the major rod- and cone-driven
Rod components of the rodent electroretinogram
and the effects of age and light-rearing
history on the magnitude of these
CB components. Photostasis and related
Cone phenomena. New York: Plenum Press; 1998.
p 93–128.)

stages of the biochemical phototransduction cascade was devel- photoreceptor cell that form a sharp “nose” on the leading
oped by Lamb and Pugh.70 This model could be fit to the leading edge of the response that quickly relaxes to the level of the
edge of the human a-wave generated by strong stimuli and it photocurrent.39
has been used in noninvasive studies of human rod36,71 and cone
photoreceptor72 function. Recent analyses suggest that the Postreceptoral contributions to the a-wave
a-wave generated in response to strong stimuli includes addi- The receptoral origins of the a-wave (Granit’s PIII),32 as well
tional currents associated with more proximal regions of the as postreceptoral contributions of similar timecourse, have been
Fig. 7.9 Depth profiles and current source
density results in anesthetized macaque
monkey retina for the a-wave and the b-wave.
186 Monopolar depth profiles Bipolar depth profiles Left: Monopolar depth profiles using an
intraretinal microelectrode referenced to the
forehead. Right: bipolar depth profiles using
a coaxial electrode with a distance of 25 µm
Section 2

Receptor between the tips to measure field potential


component amplitudes at the peak time of the receptor
- +
- + component, the b-wave and the dc component
of the macaque light-adapted
0 100 0 100 electroretinogram. The pluses and minuses
Relative amplitude

indicate the current sources (+) and sinks (–)


for the components, as calculated from
+ - + - current source density analyses based on the
Retinal Diagnostics
Retinal Imaging and Diagnostics

0 100 0 100
coaxial electrode recordings and resistance
B-wave measurements. The a-wave source and sink
are in the distal quarter of the retina. The
b-wave has a large sink near the outer nuclear
layer and a distributed source extending to the
vitreal surface of the retina. Plots represent
means of 26 penetrations. (Adapted from
Heynen H, van Norren D. Origin of the
0 100 0 100 electroretinogram in the intact macaque eye
Retinal depth (%) – II. Current source-density analysis. Vision
Res 1985;25:709–15.)

clarified in pharmacological studies. In early in vitro studies PDA-sensitive postreceptoral neurons, rather than photorecep-
of the isolated retina (amphibian and human), the nonspecific tors, or APB-sensitive contributions from ON pathway, were
glutamate agonist aspartate in the perfusate suppressed all found to generate the leading edge for the first 1.5 log units of
postreceptoral responses, and isolated the a-wave, revealing its flash strengths that elicited a measurable a-wave (Fig. 7.10B).
presence under the b-wave.51,73 Our understanding of the recep- Postreceptoral cells in the OFF pathway and inner retina contin-
toral and postreceptoral contributions to the light-adapted ued to contribute 10–15 µV to the total a-wave amplitude when
a-wave, as well as other waves of the macaque monkey phot- photoreceptoral contributions also were present. Postreceptoral
opic ERG, was greatly advanced by studies of Sieving and contributions to the a-wave in alert humans have also been
colleagues in which they utilized more specific glutamate demonstrated, by analysis of ERGs.75
analogs than aspartate to dissect retinal circuits, and particularly Postreceptoral contributions to the dark-adapted a-wave have
ON versus OFF circuits set up by depolarizing and hyperpolar- been observed as well in the monkey.34,76 For weak to moderate
ing bipolar cells, respectively (Fig. 7.2).33,35 They made intravit- stimuli from darkness, a-waves are dominated by rod signals,
real injections in anesthetized macaques of an mGluR6 receptor but a strong flash elicits a mixed rod–cone ERG like the macaque
agonist, 2-amino-4-phosphonobutyric acid (APB, L isomer, also ERG illustrated in Fig. 7.11. To study purely rod-driven
called L-AP4) to block metabotropic glutamatergic neurotrans- responses it is therefore necessary to separate the rod- and
mission to depolarizing (ON) bipolar cells. This eliminated cone-driven responses to strong stimuli when investigating their
light-evoked responses of ON bipolar cells as well as contribu- relative contributions to the ERG.
tions from more proximal cells of the retinal ON pathway. The rod-driven response can be extracted by subtracting the
Alternatively, they injected cis-2,3-piperidine dicarboxylic isolated cone-driven response to the same stimulus from the
acid (PDA) (or kynurenic acid) to block major ionotropic glu- mixed rod–cone response. Isolated cone-driven responses in
tamate receptors in the retina (α-amino-3-hydroxy-5-methyl-4- Fig. 7.11 (triangles) were obtained by briefly suppressing the rod
isoxazolepropionic acid (AMPA) and kainate receptors).74 These response with an adapting flash, and then measuring the
ionotropic receptors, blocked by PDA, mediate signal transmis- response to the original test stimulus presented a few hundred
sion to hyperpolarizing (OFF) bipolar cells and horizontal (Hz) milliseconds (300 ms) after offset of the adapting flash. Cone-
cells, as well as to amacrine and ganglion cells of both OFF driven responses in primates recover to full amplitude within
and ON pathways. Results of such experiments are illustrated about 300 ms whereas rod-driven responses take at least a
in Fig. 7.10A for responses to fairly weak stimuli presented on second, making it possible to isolate the cone-driven response.34
a rod-suppressing background; functions relating a-wave ampli- The effect of PDA on the rod-isolated a-wave of the macaque
tude and stimulus strength over a wider range of stimuli in is shown in Fig. 7.12 for a range of stimulus strengths.34 Most
the same study are shown in Fig. 7.10B. The a-wave was reduced of the leading edge of the a-wave in response to the weakest
in amplitude by PDA (or kynurenic acid, not shown), but not stimulus was eliminated (Fig. 7.12A), and much of the leading
by APB. Fig. 7.10 also shows that PDA had a similar effect to edge that occurred later than 15 ms after the flash was removed
that of aspartate, or to cobalt (Co2+), which was used to block in response to stronger stimuli (Fig. 7.12B), and even for stimuli
the voltage-gated Ca2+ channels which are essential for vesicular that saturated the response (not shown). When APB was injected
release of glutamate. along with PDA, to eliminate remaining ON bipolar cell con-
When the effects of PDA versus APB on the photopic a- tributions as well (Fig. 7.13B), the shape of the early portion of
wave amplitude were evaluated over a wider range of stimuli, PIII could be seen to form a nose, generally obscured by
Fig. 7.10 Postreceptoral contributions to the a-wave of the macaque
electroretinogram (ERG). (A) Comparison of the effects of 2-amino-4-
A phosphonobutyric acid (APB:1 mM vitreal concentration) and APB +
cis-2,3-piperidine dicarboxylic acid (PDA: 5 mM) with those of 187
Control aspartate (50 mM) and cobalt (10 mm, CO2+) on the photopic ERG
a-wave of three different eyes of two anesthetized monkeys. The inset
at the top shows the response of eye number 1 to the 200-ms stimulus

Chapter 7
of 3.76 log td (2.01 log cd/m2) on a steady background of 3.3 log td
(1.55 cd/m2). The a-waves for eyes 1, 2, and 3 were all in response to
APB APB+ the same stimulus. For this stimulus, most of the small a-wave (10 µV)
Aspartate that was elicited was postreceptoral in origin. In the clinic the a-wave
elicited by brief flashes often is larger, 20 µV or more, and therefore
200 ms also will include several microvolts of photoreceptor contribution, as
shown in part B. (B) Stimulus response function (V log I plot) of the
photopic a-wave of the macaque measured at times corresponding to

Electrogenesis of the Electroretinogram


the a-wave peak in the control responses (filled circles). Amplitudes
Control after APB (open circles) and after APB + PDA (triangles) were
Eye 1 measured at the same latency as the trough of the control a-waves
APB+ measured at the same stimulus intensity. The points are connected
Aspartate by solid lines. In this figure, as in part A, APB had no effect on the
a-wave amplitude. In contrast, PDA reduced the amplitude, and the
APB postreceptoral contribution was maximally between 10 and 15 µV,
about 50% of a 20 µV a-wave, but less than 25% of a saturated
Control a-wave of about 65 µV. (Reproduced with permission from Bush RA,
Eye 2 Sieving PA. A proximal retinal component in the primate photopic ERG
a-wave. Invest Ophthalmol Vis Sci 1994;35:635–45.)

Co2+

Control
Eye 3

APB + PDA
20 µV

10 ms APB

B
70
Control
60
APB
Response amplitude (µV)

50
APB + PDA
40
30
20
10
0
–10
2 3 4 5 6
Log intensity (td)

negative-going signals removed by PDA, and the b-wave postreceptoral contributions to the dark-adapted a-wave, rather
removed by APB (Fig. 7.12C). than a contribution from OFF bipolar cells.77
PDA blocks signal transmission not only to hyperpolarizing
bipolar and horizontal cells, but also to amacrine and ganglion
The timecourse of the
cells of the inner retina. In order to test whether the effect of PDA photoreceptor response
on the a-wave was due to OFF bipolar cells or more proximal The leading edge of the a-wave is the only visible portion of the
cells, it was necessary selectively to suppress amacrine and gan- photoreceptor response in the normal ERG. In order to see the
glion cell activity. This was done using N-methyl-D-aspartate entire timecourse of the photoreceptor response it is necessary
(NMDA), as only the proximal retinal neurons have functional to remove postreceptoral contributions, as was done pharmaco-
NMDA (ionotropic glutamate) receptors. Results (not shown) logically for the ERGs in Figs 7.11 and 7.12. However, such a
were similar in the dark-adapted ERG to those after PDA alone, manipulation is invasive, and does not eliminate slow PIII and
indicating a proximal retinal origin for most of the rod-driven the c-wave, should they be present. Another approach is to use
188 0 0
µV
Section 2

µV
–200 –100

A
A –200

0
Retinal Diagnostics
Retinal Imaging and Diagnostics

Cone receptor Post PDA


0

µV
–200
µV

–100
Rod receptor
–400

Mixed rod–cone response B


0 10 20 30 40

Post APB+PDA
0 5 10 15 20
0
B Time after flash (ms)

Fig. 7.11 Dark-adapted electroretinogram (ERG) of a macaque µV –200


showing the mixed rod–cone a-wave and separate components
measured (symbols) and modeled (solid lines fit to the leading edge
of the a-wave) for two stimulus strengths. (A) Responses of an
anesthetized macaque to a brief blue light-emitting diode (LED) flash
of 188 sc td/s (57 ph td/s) and (B) responses to a xenon white flash –400
of 59 000 sc td/s, (34 000 ph td/s). In both parts, the largest response 0 10 20 30 40
(open circles) is the entire mixed rod–cone a-wave; the green solid line C
restricted to the leading edge shows the modeled mixed rod–cone
Time after flash (ms)
response. The second largest response is the (isolated) rod-driven
response (filled circles); the red solid line through the leading edge is
the modeled rod photoreceptor contribution. The second to smallest Fig. 7.12 Postreceptoral contribution to rod-driven dark-adapted
response (open triangles) is the (isolated) cone-driven response, macaque electroretinogram (ERG). Rod-driven responses from an
including the postreceptoral contribution; the cyan solid line through anesthetized macaque were obtained after subtracting isolated
the leading edge shows the modeled cone-driven response. The cone-driven response from the mixed rod–cone ERG. Rod-driven
smallest response (purple line) is the modeled cone photoreceptor responses are shown to a range of stimulus energies (15.8–509 sc
contribution, based on post cis-2,3-piperidine dicarboxylic acid (PDA) td/s) before (A) and after (B) intravitreal injection of cis-2,3-piperidine
findings for the cone-driven response. Given the animal’s 8.5-mm dicarboxylic acid (PDA) (4 mM). For comparison, the ERG remaining
pupil, the stimulus for part A was about 1 cd s/m2, i.e., about three after adding PDA and 2-amino-4-phosphonobutyric acid (APB) is
times less intense (for cones) than International Society for the Clinical shown in part C. The stimulus was a xenon white flash of 59 000 sc
Electrophysiology of Vision (ISCEV) standard flash of 3 cd s/m2, td/s, and cone signals were not removed in this recording. The control
whereas the stimulus for part B is about 2000 times stronger. ISCEV response is shown by the green line, the post PDA+APB by the
also suggests flashes of 10 and 30 cd s/m2 for mixed rod–cone ERG. dashed black line. (A and B, adapted from Robson JG, Saszik SM,
(Adapted from Robson JG, Saszik SM, Ahmed J, et al. Rod and cone Ahmed J, et al. Rod and cone contributions to the a-wave of the
contributions to the a-wave of the electroretinogram of the macaque. electroretinogram of the macaque. J Physiol 2003;547:509–30; C,
J Physiol 2003;547:509–30.) from unpublished data.)

the paired-flash technique developed by Pepperberg and col- currents from more proximal regions of the photoreceptor in
leagues78 to derive, in vivo, the photoreceptor response. The addition to outer-segment currents.39
derived photoreceptor response (underneath) and the full ERG
responses to three stimuli of the same strength are shown for a
ORIGIN OF THE B-WAVE
macaque monkey in Fig. 7.13. The timecourse of the derived The cornea-positive b-wave, Granit’s PII, is the largest compo-
response is similar to that of current recordings in vitro from nent of the dark-adapted diffuse-flash ERG. There is general
individual rod photoreceptor outer segments in the macaque, consensus that the neuronal generator of the b-wave is primarily
although it reaches its peak a little earlier in vivo in the ERG than the depolarizing (ON) bipolar cells.30,31,79,80 APB (L-AP4), which
in vitro. However, the amplitude of the derived response is binds to mGluR6 receptors of ON bipolar cells and removed the
rather large, perhaps twice that expected for the photocurrent. cone b-wave in experiments of Sieving et al., also removed the
The large amplitude of the derived response may occur because dark-adapted b-wave in several species, including primates.30,80
the method for deriving responses uses measurements in the The b-wave is also absent, causing a negative ERG (Fig. 7.12C).
“nose” portion of saturated a-wave (Fig. 7.12C), which reflects Negative ERGs also occur in patients and murine models with
Dark-adapted Macaque ERG
200 Monkey 189
300 Saturated b-wave Mouse
Cat

Chapter 7
Response magnitude (percent)
200 Test flashes: 0.28, 1.24, 6.86 sc td.s Rabbit
ERG amplitude (µV)

150
Guinea pig
100
Salamander

0
100

Electrogenesis of the Electroretinogram


0

–100 50
Derived rod response

0 200 400 600


Time after flash 0

A B C D E F G H
Fig. 7.13 Dark-adapted electroretinogram response of the anesthetized Endfoot Soma Distal end
macaque monkey to three different test stimulus strengths (0.28, 1.24,
Ejection site
and 6.86 sc td/s) and the derived rod photoreceptor response for each
test stimulus. The photoreceptor response was derived using the
paired-flash approach of Pepperberg et al.,78 in which a rod-saturating
probe flash follows a test flash at fixed intervals after its onset, and the Fig. 7.14 Distribution of K+ conductance over the surface of
residual response of the probe is subtracted from the probe alone to enzymatically dissociated Müller cells. The magnitude of
derive the rod receptor response at each time point (data points). The depolarizations in response to focal K+ ejections is plotted as a
model lines used modifications of equations from Robson et al.34 function of ejection location along the Müller cell surface. Responses
(Frishman LJ, Robson JG, unpublished observations.) are normalized to response magnitude at the endfoot. In species with
avascular retinas (salamander, rabbit, guinea pig), K+ conductance is
largest at the endfoot. In vascularized species, conductance is largest
near the cell soma (mouse, monkey) or at the distal end of the cell
(cat). (Reproduced with permission from Newman EA. Distribution
of potassium conductance in mammalian Müller (glial) cells: a
complete congenital stationary night blindness (CSNB) who comparative study. J Neurosci 1987;7:2423–32.)
have mutations that cause a breakdown in the signal transduc-
tion cascade in ON bipolar cells.81,82 However, experimental
support also exists for a Müller cell contribution, likely due to
K+ currents as a consequence of bipolar cell depolarization, par-
ticularly at times past the leading edge of the response, as Measurements using intraretinal K+-selective microelectrodes
described below. of local changes in light-evoked [K+]o skate,85 , amphibian,20,86 and
Resolving the role of bipolar cells and Müller cells in generat- rabbit21 retinas demonstrated only small transient [K+]o increases
ing the b-wave has been difficult. Early intraretinal depth record- in the OPL at light onset, presumably from dendrites of ON
ings demonstrated a negative-going intraretinal b-wave with a bipolar cells. The Müller cell hypothesis required a large sink
(negative) peak amplitude in distal retina, near the outer plexi- activity in the OPL. In contrast, a large sustained inner plexiform
form layer (OPL), and the largest change in amplitude across layer (IPL) [K+]o increase was recorded in the proximal retina of
the inner nuclear layer (INL).13,65 These results suggested that several species at the onset, or at onset and offset of a light
the b-wave was generated by current flowing through a radially stimulus.87 The large proximal [K+]o increase reflected activation
oriented cell that acted as a dipole. Bipolar cells, the only radi- of amacrine and ganglion cells as well, and is now understood
ally oriented neurons that span the INL, and Müller cells, the to contribute strongly to Müller cell or astrocyte-mediated ERG
radially oriented glial cells, were implicated. components of proximal retinal origin, e.g., M-wave, STR, or
PhNR.
Müller cell hypothesis As described in the section on spatial buffering, above, the
CSD profiles for the b-wave from intraretinal recordings in frog selective permeability of the Müller cell membrane to K+ and the
retina83 showed an OPL current sink and a current source extend- nonuniform distribution of K+ conductances (Fig. 7.3) provide
ing from the OPL almost all the way to the vitreal surface, and the opportunity for K+ siphoning from synaptic layers where
this also was found in monkey retina (Fig. 7.9).18 Due to the [K+]o is high to regions of lower [K+]o. In species with avascular
extensive radial spread of sinks and sources, it was hypothesized retinas, e.g., amphibian and rabbit, the Müller cell endfoot adja-
that Müller cells generate b-wave currents, and supporting this, cent to the vitreous humor has more than 90% of the K+ con­
Miller and Dowling84 found that intracellularly recorded Müller ductance in the cell, as shown in the Newman lab study of
cell responses in amphibian resemble b-wave responses to the enzymatically isolated Müller cells subjected to puffs of K+ (Fig.
same stimuli. However studies of local changes in [K+]o and 7.14). K+ entering the Müller cell via strongly inward rectifying
Müller K+ conductances were not fully consistent with the Müller Kir channels in synaptic regions will exit predominantly from
cell hypothesis. the vitreal endfoot where the K+ conductance is only weakly
rectifying (Fig. 7.3).28 The OPL [K+]o increase could establish a sink was retained, and the IPL source involved in generating the
current loop that extends all the way to the vitreal surface. The b-wave was enhanced. These results indicated that the major
190 IPL [K+]o increase would contribute to the same current loop, b-wave generator is the bipolar cell itself.
creating a vitreal positive proximally generated potential in
Scotopic b-wave (PII) in mammals
addition to the bipolar cell-dependent b-wave. However, as
In the mammalian retina, the scotopic ERG response to weak
noted above, the OPL increases in [K+]o were small, and because
Section 2

stimuli reflects activity of the sensitive primary rod circuit,


of this, did not predict the large b-wave recorded in the ERG.
including rods, the main interneuron, rod bipolar cells, AII ama-
In contrast, in species with vascularized retinas such as mouse
crine cells, cone bipolar cell terminals, and ganglion cells (Fig.
and monkey, K+ conductance, although high at the endfoot, is
7.2). Given the simplicity of the circuit, with contributions from
greatest in the INL, near capillaries. In cat, K+ conductance is
amacrine and ganglion cells pharmacologically suppressed, the
greatest near the subretinal space (Fig. 7.14). In these species
isolated b-wave (Granit’s PII) should reflect the rod bipolar cell
with vascularized retinas the Müller cell current that arises as a
Retinal Diagnostics
Retinal Imaging and Diagnostics

response, either directly as the bipolar cell current, or via Müller


consequence of the proximal [K+]o increase contributes to a dis-
cell K+ currents resulting from K+ outflow from bipolar cells.
tally directed current loop (like slow PIII), thereby producing a
Only for strong stimuli would the underlying photoreceptor PIII
cornea-negative potential (e.g., m-wave or negative STR), rather
be a significant factor in the ERG. Figure 7.15A shows phar­
than positive b-wave.
macologically isolated PII from ERGs, recorded in vivo, of four
normal C57BL/6 mice, in response to a weak stimulus. An intra-
ON bipolar cells as the generator of vitreal injection of the inhibitory neurotransmitter γ-aminobutyric
the b-wave acid (GABA) was used to suppress inner retinal activity.31,90 Also
Experiments using Ba2+ to block Kir channels have not supported included in the figure is PII isolated from human ERG using
the Müller cell hypothesis for generation of the b-wave. Although weak adapting backgrounds to suppress the very sensitive
Ba2+ blocks slow PIII48 as well other responses associated with STRs.91 The isolated PII responses have been superimposed on
Müller cell K+ currents, it is far less effective in blocking the an average of several patch electrode current recordings in a
b-wave.23,54,88,89 In CSD studies in frog88 and rabbit,89 only the mouse retinal slice preparation of rod bipolar cell responses to
proximal sink source activity associated with the inner retinal a stimulus of similar effect on the rods. The timecourse of the
M-wave was removed by Ba2+. At least two-thirds of the OPL ERG and the single cell currents is very similar, supporting the

A B
Mouse
Average rbc current Cat Pre-Ba++
Isolated-PII
for dim flash (Rieke) Isolated-PII
Post-Ba++
1.0 GABA-isolated PII Pre–post-Ba++
(1–1.2 R8/rod)
0.8 Cx36 (–/–) with
optic nerve crush
Normalized response

Modeled
0.6 0 300
components: Time after flash (ms)

0.4 Fast PII

0.2 Slow PII

0.0

0 100 200 300 400 0 200 300 300


Time after flash (ms) Time after flash (ms)

Fig. 7.15 Rod bipolar cell component (PII) of the dark-adapted electroretinogram (ERG) of mouse, cat, and human. (A) (left) Comparison of rod
bipolar cell current from patch recordings in mouse retinal slice (courtesy of F. Rieke) with isolated PII (by weak light adaptation) from humans,
isolated PII from ERGs of six C57BL/6 mice by intravitreal injection of γ-aminobutyric acid (GABA: 32–46 mM) and additionally from a Cx36(–/–)
mouse lacking ganglion cells that lacked scotopic threshold responses. (Reproduced with permission from Cameron AM, Mahroo OA, Lamb TD.
Dark adaptation of human rod bipolar cells measured from the b-wave of the scotopic electroretinogram. J Physiol 2006;575:507–26, with
permission.) (B) (right) Pharmacologically isolated cat PII (by inner retinal blockade). The response has been analyzed into a fast component,
proposed to be a direct reflection of the postsynaptic current, and a slow component, that is a low-pass filtered version of the faster component,
believed to be the Müller cell response, contributing mainly to the tail of the response. Identification of the Müller cell component is supported by
the observation (shown in the inset) that intravitreal injection of Ba2+, used to block Kir channels in Müller cells removed a similar portion of the
total PII response. For stronger stimuli, in order to isolate the bipolar cell response, it would be necessary also to remove the underlying negative
(photoreceptor) PIII signal. (Reproduced with permission from Robson JG, Frishman LJ. Dissecting the dark-adapted electroretinogram. Doc
Ophthalmol 1998;95:187–215, and from LJ Frishman, JG Robson, unpublished observations.)
view that isolated PII reflects rod bipolar cell activity. If Müller
cell currents had generated PII, the signal would have been Eye 1 Eye 2
delayed relative to the bipolar cell responses recorded in the Control Control 191
b d
slice, due to the time necessary for accumulation of K+ ions in
the ECS.
a

Chapter 7
Pharmacologically isolated PII of cat (Fig. 7.15B) revealed a Plateau
response similar to that in mouse in its rising phase, but recov- APB PDA
ered more slowly to baseline. The cat PII response to a brief flash
can be analyzed into a fast component and a slow component
that is a low-pass filtered version of the fast component, as
20 µV
indicated by the lines in Fig. 7.15B. The inset to Fig. 7.15B shows
that intravitreal Ba2+ eliminated a slow portion of the response

Electrogenesis of the Electroretinogram


that was very similar in timecourse to the modeled slow com-
APB+PDA
ponent, but did not alter the fast component, suggesting that PDA+APB
bipolar cell current provides the leading edge of PII in the cat,
and the Müller cell current contributes at later times. Although 200 ms 200 ms
the slow component (in response to a brief flash) was lower in
amplitude than the fast component, the area under the two Fig. 7.16 Effects of 2-amino-4-phosphonobutyric acid (APB) and
curves was similar. With longer stimulus durations such as those cis-2,3-piperidine dicarboxylic acid (PDA) on the light-adapted photopic
used in early studies of the b-wave origins, the contribution to electroretinogram of two monkey eyes. Drugs were given sequentially,
the ERG of the two sources would be about equal.85 Pharmaco- APB followed by PDA for eye 1, and PDA followed by APB for eye 2.
The vertical line shows the time of the a-wave trough in the control
logic isolation of macaque PII showed a similar waveform to that response. The 200-ms stimulus was 3.76 log td (2.01 log cd/m2)
in cat.3 on a steady rod-saturating background of 3.3 log td (1.55 cd/m2).
(Reproduced with permission from Bush RA, Sieving PA. A proximal
Cone-driven b-wave retinal component in the primate photopic ERG a-wave. Invest.
The photopic ERG is commonly measured in the presence of a Ophthalmol Vis Sci 1994;35:635–45.)
rod-suppressing background. Depth profiles of the rod b-wave
measured under dark-adapted conditions and the cone b-wave,
under light-adapted conditions, are similar in cat92,93 and monkey
retinas, suggesting a similar origin for the two responses, but in B-wave
the case of the photopic ERG, depolarizing cone rather than rod D-wave
bipolar cells are involved.
Sieving and colleagues33,35 studied the origins of the photopic
b-wave in monkeys using glutamate analogs in the same series
of experiments as those in which the a-wave was studied. Figure
7.16 shows, on the right, the macaque photopic ERG response A-wave 0.5 mV
to long-duration flashes, before and after APB was injected into
the vitreal cavity (eye 1, left). APB removed the transient b-wave 1.0 sec
supporting ON bipolar cells as the generators (although the pos-
sibility of glial mediation was not eliminated in these studies).
However, when PDA was injected first in another eye (eye 2, Fig. 7.17 Electroretinogram from the all-cone retina of the ground
squirrel in response to a 1-second step. Recordings were made under
right), to remove the OFF pathway (and horizontal cell inhibi- light-adapted conditions between a contact lens electrode on the
tory feedback), a much larger and more sustained b-wave was cornea and an electrode on the forehead. (Reproduced with
revealed. These findings indicate that the normally transient permission from Arden GB, Tansley K. The spectral sensitivity of the
pure-cone retina of the grey squirrel (Sciurus carolinensis leucotis).
nature of the cone b-wave is due to truncation of a more pro- J Physiol 1955;127:592–602.)
longed depolarizing bipolar cell response. This truncation is due
to some combination of (PDA-sensitive) OFF pathway response
of opposite polarity to the ON bipolar cell contribution to the
prominent, is easily identified when stimulus duration is
ERG (a push–pull effect28), and inhibition via (PDA-sensitive)
extended, e.g., Figs 7.1 and 7.7. Although d-waves have been
horizontal cells of ON bipolar cell signals. The isolated photore-
described in scotopic ERG of mammals, they are probably not
ceptor response remaining after all postrecep­toral activity was
“true” d-waves that, as described below, include a strong con-
eliminated either by APB followed by PDA, or PDA followed
tribution from the offset of the cone photoreceptor, and the depo-
by APB, was similar.
larization at light offset of OFF cone bipolar cells. In the
predominantly rod-driven cat ERG, the small positive deflection
ORIGIN OF THE D-WAVE that Granit32 called a “d-wave” occurred only in response to offset
The d-wave is a positive-going deflection at light offset that of very intense and long-duration stimuli for which the decay of
occurs in the photopic ERG. It is best seen when the light step is the rod receptor potential appeared as a small positive deflection
prolonged. In mammals, the d-wave is very prominent in the that followed, at light offset, the negative-going offset of PII.
all-cone retina of the ground squirrel, as seen in the response to Intraretinal analysis in monkey retina indicated that the
a long-duration stimulus in Fig. 7.17, and in the monkey retina, corneal d-wave represents a combination of the rapid positive-
with its mixture of rods and cones, the d-wave, although less going offset of the cone receptor potential followed by the
negative-going offset of the b-wave.16,66 Although the early work ORIGIN OF THE MULTIFOCAL ERG
on primate ERG cited above did not identify OFF bipolar cells as
192 major contributors to the d-wave, more recent studies using Multifocal electroretinography (mfERG) was developed by
intravitreal injection of glutamate analogs have demonstrated Sutter and Tran103 as a means of recording many focal retinal
that these cells provide a significant portion of the response.35,94 responses simultaneously in a brief time period (e.g., 7 minutes)
Figs 7.10 and 7.16 both show that PDA, which blocks OFF bipolar to achieve a topographical array of little ERGs. Recording ERGs
Section 2

cell responses, reduced the d-wave, eliminating it entirely when from many different focal regions of the retina otherwise would
a large b-wave was present. The effect of PDA was not replicated be impractical, due to the time involved in averaging of the small
by NMDA blockade of inner retinal cells, that also would have signals. The common mfERG stimulus is an array of hexagons
been affected by PDA, confirming a role of the OFF bipolar cells (e.g., 64 or 103 hexagons over 30–40° of central visual field). In
themselves. PDA, but not the blockade of inner retinal cells, also the standard mode for stimulation each hexagon reverses in
eliminated a small positive wave that occurs in the falling phase contrast, according to a predetermined random “m-sequence,”
Retinal Diagnostics
Retinal Imaging and Diagnostics

of the b-wave, or just after it, in the brief flash ERG in monkeys at the frame rate of the visual display monitor, often 75 Hz for
and humans, called the “i-wave.”95 CRTs and 60 Hz for LCD displays. At any given time, each
hexagon has a 50% chance of being light or dark. The m-sequence
for all hexagons is the same, except for a specific and unique lag
Photopic hill (number of frames) in initiation of the sequence for each hexagon,
The stimulus response function acquired by measuring b-wave to allow extraction of the locally generated response by a correla-
peak amplitudes in response to brief flashes over a range of tion approach. The resulting “first-order” focal ERGs to the
increasing stimulus strengths has a characteristic inverted “U” rapidly changing stimulation look similar but not identical to
shape. More specifically, Peachey et al.96 observed that the full-field flash ERGs that integrate responses over a longer
b-wave amplitude increased with increasing stimulus strength period of time. The mfERG is generally recorded under photopic
until it reached a peak and then decreased for stronger stimuli. conditions, for which the foveal response is large, and is useful
This function was later named the “photopic hill” by Wali and for detecting local changes in function that may not be detected
Leguire.97 A model of the stimulus response function has indi- in a full-field ERG, such as those that occur in macular
cated that the positive peak of the function is formed by addition dystrophies.
of a saturating b-wave from ON pathway,98 and an early d-wave Experiments using intravitreal injections of glutamate
from OFF pathway, and this has been confirmed in experiments analogs, APB, and PDA in macaques showed that, despite
using glutamate analogs in macaques.99 differences in the mfERG and full-field flash ERG, the cellular
origins of the major negative and positive waves are essen-
tially the same in two types of ERG.104 The outcome of such
ORIGIN OF THE PHOTOPIC experiments is shown in Fig. 7.18, where the findings from
FAST-FLICKER ERG the experiments on macaque mfERG were applied to the
The fast-flicker ERG (nominally 30 Hz flicker) is used to examine human response. When blank frames were interposed so that
cone-driven responses in humans because rod-driven responses flashes occurred every 100 or 200 ms, a complete ERG includ-
generally do not respond to fast flicker. For many years the ing OPs could form before the next m-frame. For this slow-
human (or macaque) photopic ERG response to fast-flickering sequence ERG, experiments in macaques again yielded origins
stimuli was believed to reflect primarily the response of cone similar to full-field flash ERG, and provided some evidence
photoreceptors. However pharmacologic dissection studies for spike-dependent oscillatory activity related to the optic
have shown that most of the fast-flicker response seen clinically nerve head component in the mfERG, proposed by Sutter
with a full-field stimulus (or focal stimulus) is generated and Bearse.105–107
postreceptorally.
Bush and Sieving100 used glutamate analogs to remove selec-
tively postreceptoral ON and OFF pathway responses, as was ERG WAVES FROM PROXIMAL RETINA
done in studies of a-, b-, and d-waves. Intravitreal injection of
APB to remove the b-wave left a delayed flicker response, reflect-
Origin of the proximal negative response
ing OFF bipolar contribution. When both APB and PDA were and the M-wave
used, the flicker response was practically eliminated. From The proximal negative response (PNR) is a light-evoked field
experiments of this type they concluded that postreceptoral cells potential recorded intraretinally in proximal retina, named and
that normally produce the b- and d-waves are strong contribu- most fully described by Burkhardt.108 The PNR consists of a
tors to the fast-flicker response. Further experiments in macaques sharp, negative-going transient at onset and again at offset of a
have more thoroughly investigated the interaction of the ON small light spot centered on the microelectrode tip placed in the
and OFF pathways over a wide range of temporal frequencies IPL. It can be recorded in a range of vertebrate retinas, including
and stimulus conditions.101 The amplitude in both humans and the cat109 and primate.110 A PNR contribution to the transretinal
macaques dips around 12 Hz, as ON and OFF inputs cancel, and ERG is necessarily small because the intraretinal response, which
it reaches a peak around 50 Hz. Other studies have demon- is largest in response to small spots, will be shunted through
strated a small inner retinal contribution to the flicker response, adjacent low-resistance retinal regions that are not activated by
and particularly to the second harmonic component of the the light.
response, which has a strong input from TTX-sensitive sodium- The M-wave, like the PNR, is a light-evoked field potential,
dependent spiking activity of ganglion and perhaps amacrine recorded in proximal retina with microelectrodes in response to
cells.102 the onset and offset of a small well-centered spot, but it has a
neurons.23,87 As illustrated in Fig. 7.19, for cat, changes in [K+]o
recorded in proximal retina show a similar timecourse to the
simultaneously recorded field potentials. In isolated amphibian 193
retina it was shown that the Kir channel blocker Ba2+ had only
minor effects on light-evoked neural activity (PNR) and the
proximal retinal [K+]o increase, but it blocked the M-wave and

Chapter 7
K+ spatial buffer currents.25 Similar findings in intact cat retina
are illustrated in Fig. 7.19. Note that after intravitreal Ba2+ injec-
tion, the proximal [K+]o increase was larger, but the more distal
increase seen in control records was absent. This is consistent
with blockade by Ba2+ of spatial buffer currents that normally
would carry the K+ away from proximal retina to the distal

Electrogenesis of the Electroretinogram


extracellular sinks where [K+]o was lower.
The local M-wave’s contribution to the transretinal ERG
would be small. The contribution of the M-wave may be greater
when periodic stimuli such as grating patterns that stimulate
large regions of retina are used. Sieving and Steinberg111 showed
that the M-wave is tuned to a spot diameter similar to the
bar width of the optimal spatial frequency for the intraretinal
pattern ERG (PERG) in the cat. The contribution of the proxi-
mal retina to the ERG can also be enhanced by using full-field
stimulation.

Origin of the photopic negative response


In contrast to the PNR and M-wave, which provide small con-
500 nV tributions at most to the transretinal ERG, negative-going
responses from proximal retina to full-field stimulation are
0 100 ms
present in the corneal ERGs of several species, including cats,
OFF-bipolars monkeys, and humans.40,63,92,93,112–114 These responses, called the
ON-bipolars peak depolarization PhNR, and STR are generated by mechanisms similar to those
peak
ON-bipolars documented above for the M-wave.
ON-bipolars recover The PhNR is a negative-going wave in the photopic ERG that
depolarize occurs after the b-wave in response to a brief flash, and again
after the d-wave in response to a long flash. It is more prominent
in primates than in rodents. In humans and monkeys, the PhNR
is thought to reflect spiking activity of retinal ganglion cells.113,114
As shown in Fig. 7.20, the PhNR is reduced in macaque eyes
OFF-bipolars OFF-bipolars
with experimental glaucoma (also after TTX injection)113 and in
and receptors and receptors
hyperpolarize recover humans with primary open angle glaucoma. It is reduced in
several other disorders affecting inner retina and optic nerve
0 20 40 60ms head as well. The slow timecourse of the PhNR suggests glial
involvement, perhaps via K+ currents in astrocytes in the optic
Fig. 7.18 Major components of the multifocal electroretinogram nerve head or Müller cells set up by increased [K+]o due to
(mfERG). (A) mfERG trace array (field view) using 103 hexagons spiking of ganglion cells. In intraretinal microelectrode record-
scaled with eccentricity and covering about 35° of visual angle, as ings in cats, local signals of the same timecourse as the PhNR
illustrated above the traces. (B) Model for the retinal contributions
were largest in and around the optic nerve head and the PhNR
to the human mfERG based on results from the macular region
of a macaque after pharmacological separation of components. was disrupted in cats by Ba2+, indicating glial involvement in
(Reproduced with permission from Hood DC, Frishman LJ, Saszik S, generation of the response (Viswanathan and Frishman, unpub-
et al. Retinal origins of the primate multifocal ERG: implications for the lished observations).
human response. Invest Ophthalmol Vis Sci 2002;43:1673–85.)
PhNRs can be evoked using white flashes on a white back-
ground, if the flash is strong enough. However a red LED flash
on a blue background, as was used for ERGs shown in Fig.
7.20, elicits PhNRs over a wider range of stimulus strengths.
slower timecourse than the PNR. The M-wave was initially The red flash may minimize spectral opponency that would
described in studies of amphibians,87 but also has been identified reduce ganglion cell responses, and use of a blue background
in the cat,23,92 as illustrated in Fig. 7.19. suppresses rods while minimizing light adaptation of L-cone
In retinas in which both an M-wave and a PNR can be recorded signals.7 Blue stimuli on a yellow background which minimize
(amphibia and cats), the PNR is thought to be the transient neu- spectral antagonism are good stimuli as well. The PhNR can be
ronally generated portion of the proximal retinal response, while enhanced relative to other major ERG components, and can
the slower M-wave reflects spatial buffer currents in Müller cells, dominate the ERG when using focal stimuli in the region of the
generated as a consequence of K+ release from the same proximal macula.115–117
Before Ba2+ Before Ba2+
194
Intraretinal ERG VK+ Intraretinal ERG VK+
% depth % depth
0 0
Section 2

15

16
Retinal Diagnostics
Retinal Imaging and Diagnostics

43
43

0.25 mV 0.5 mV 0.25 mV 0.5 mV

4 sec 4 sec

Fig. 7.19 Effect of Ba2+ on the M-wave of the light-adapted cat retina. Effect of intravitreal barium chloride (BaCl2+, 3 mM vitreal concentration) on
the depth distribution of field potentials and [K+]o changes in light-adapted retina. Recordings were made before (A) and 30–60 minutes after
(B) BaCl2+ was injected. The stimulus was a small spot (0.8° in diameter); steady background illumination was 10.5 log q deg2/s; flash illumination
was 11.6 log q deg2/s. (Adapted from Frishman LJ, Yamamoto F, Bogucka J, et al. Light-evoked changes in [K+]o in proximal portion of light-
adapted cat retina. J Neurophysiol 1992;67:1201–12.)

Fig. 7.20 Photopic negative response (PhNR)


in macaque monkey and human in normal
A and glaucomatous eyes. (A) Full-field flash
Control eye Experimental eye Difference electroretinogram (ERG) showing the PhNR
Log phot td of a macaque in response to long (top) and
brief (middle) red light-emitting diode (LED)
20 µV flashes on a rod-saturating blue background
(3.7 log sc td) from the control (left) and
3.9 “experimental” (middle) fellow eye with
laser-induced glaucoma, and the difference
between control and experimental records
(right). Arrows mark the amplitude of the
PhNR. The mean deviation (MD, static
perimetry, C24–2 full threshold program)
200 ms 200 ms 200 ms
0 500 ms 0 500 ms 0 500 ms for the experimental eye was −2.65 dB.
Log phot td.s (Adapted from Viswanathan S, Frishman LJ,
Robson JG, et al. The photopic negative
20 µV response of the macaque electroretinogram:
reduction by experimental glaucoma. Invest
Ophthalmol Vis Sci 1999;40:1124-36, with
1.9 permission.) (B) Full-field flash ERGs of an
age-matched 63-year-old normal human
subject and a patient with primary open angle
glaucoma (POAG) under similar stimulus
<5 ms <5 ms <5 ms conditions to those used above for monkeys.
0 300 ms 0 300 ms 0 300 ms The MD (static perimetry, C24–2 full
threshold program) for the patient’s eye was
−16.2 dB. (Adapted from Viswanathan S,
B Frishman LJ, Robson JG, et al. The photopic
negative response of the flash
Log phot td.s electroretinogram in primary open angle
glaucoma. Invest Ophthalmol Vis Sci
Age-matched control POAG patient 2001;42:514–22.)
(MD= -16.2)
20 µV
i-wave

1.7
PhNR

<5 ms <5 ms
0 300 ms 0 300 ms
Relation to the pattern ERG
The PERG has been the most commonly used noninvasive retinal STR
measure of ganglion cell activity. It is a small response of a few 195
Surface ERG Surface ERG and
microvolts elicited by contrast reversal of a bar grating or scaled proximal
checkerboard pattern that shows some spatial tuning, consistent
0.01 mV

Chapter 7
with a ganglion cell origin. In response to pattern stimulus, in
which mean luminance does not change, the linear signals that Proximal
produce a- and b-waves cancel, leaving only the nonlinear (mainly
second harmonic) signals in the ERG. The PERG is eliminated by
0.25 mV
loss of ganglion cells as a consequence of optic nerve section.118 It
has been used widely in clinical studies assessing ganglion cell Mid-retinal
function in eyes with glaucoma and other diseases of inner retina

Electrogenesis of the Electroretinogram


(see reviews by Holder117 and by Bach and Hoffmann116).
PERGs can be recorded as a transient response to low reversal
frequencies (1–2 Hz) or as a steady-state response to higher fre- 0.3 sec
quencies, i.e., 8 Hz. For 1–2-Hz reversals of the pattern stimulus,
a positive wave occurs within about 50 ms of each reversal (P50) Fig. 7.21 The scotopic threshold response (STR) dominates
and a negative wave, N95, is maximal at about 95 ms. The N95 is intraretinal recordings and the electroretinogram (ERG) at low stimulus
often reduced in glaucomatous eyes (the effects on the smaller intensities below the threshold for the b-wave (PII). On the left, the top
P50 response are less consistent). Because the PhNR has a similar trace is the surface ERG recorded about 25 µm from the retinal
surface, and the bottom two traces are recordings of the STR in the
implicit time to the N95, Viswanathan et al.119 compared effects proximal retina (about 6% retinal depth) and the inverted STR around
of experimental glaucoma and of TTX on the two responses in 50% retinal depth. On the right, the scaled STR recorded in the
macaques, and found similar effects on both, indicating common proximal retina is superimposed on the surface ERG to show the
similarity of the responses. For the surface ERG, a microelectrode was
retinal origins. Furthermore, the PERG waveform for a given
referenced to a wire in the vitreous in order to reduce the effects of
stimulation frequency (2 or 8 Hz) could be simulated by adding stray light. This minimized contributions to the ERG of retinal regions
together the ERGs at onset and offset of a uniform field, with distant from the recording site of the intraretinal signals (spot diameter,
strong contributions of PhNRs in generation of N95. More recent 9.9°; spot illumination, 4.8 log q deg2/s). (Adapted from Sieving PA,
Frishman LJ, Steinberg RH. Scotopic threshold response of proximal
work using glutamate analogs and simulations has found that retina in cat. J Neurophysiol 1986;56:1049–61.)
both ON and OFF pathways contribute equally to the transient
PERG, but the ON pathway dominates the 8 Hz PERG.120
Given the similar origins of PhNR and PERG, the PhNR may
have some advantages over PERG as a clinical indicator of proxi- PII, the nSTR reversed polarity in midretina and became a
mal retinal dysfunction. With appropriate stimulus conditions, positive-going signal in the mid- and distal retina. This reversal
the PhNR is a much larger response, it does not require refractive suggests a source proximal to, and a sink distal to, the reversal
correction, and will be less affected by opacities in the ocular point (see description of the Müller cell mechanism, below). For
media than the PERG. stronger stimuli, the reversed nSTR in mid- and distal retina
was replaced by PII, which then dominated the ERG. The simi-
larity of the onset times and timecourse of the proximal retinal
Origin of the scotopic threshold response STR and the negative STR in the cat vitreal ERG can be seen
For very weak flashes from darkness, near psychophysical in Fig. 7.21.
threshold in humans,33,90 small negative (n) and positive (p) STR The nSTR also can be separated from PII using pharmacologic
dominate the ERG of most mammals that have been studied. agents (GABA, glycine, or NMDA30,90,121) to suppress responses
This response, which is more sensitive than the b-wave (or of the amacrine and ganglion cells proximal to bipolar cells.
a-wave) and saturates at a lower light level than either compo- These agents remove the STR, but not PII (Fig. 7.15). In contrast,
nent, was thus named because of its sensitivity.63 As shown in APB eliminates both the scotopic b-wave (PII) and STR, indicat-
Fig. 7.7, for the monkey and human, the nSTR at stimulus onset ing that the STR will not be generated if the primary rod circuit
dominates the dark-adapted diffuse flash ERG in response to the is no longer mediated by rod bipolar cells (Fig. 7.2).
weakest stimuli. The nSTR is distinct from the scotopic a-wave, There is a similarly sensitive positive STR in the scotopic
although it can appear as a “pseudo a-wave” at light levels ERG of animals that have a negative STR.40,90 Because the pSTR
where it can be removed by suppressing inner retinal activity is small and of opposite polarity to the nSTR, it can easily be
pharmacologically.121,122 The STRs occur in response to stimuli cancelled out in the ERG. An instance of this can be seen in
much weaker than those that elicit more distally generated the dark-adapted macaque ERG in Fig. 7.7. The delayed onset
waves of the ERG because convergence of the rod signal in the of nSTR for the weakest stimulus is due to the presence of a
retinal circuitry increases the gain of responses generated by pSTR that is slightly larger than the nSTR at early times. In
inner retinal neurons. response to a stimulus about 1 log unit higher (just above), the
The nSTR was initially observed to be generated more proxi- pSTR rides on the emerging PII as an early positive potential.
mally (IPL) than PII (INL) in intraretinal depth analysis studies Most pharmacologic agents that eliminate the nSTR also elimi-
in cats.63 As shown in Fig. 7.21, the field potential recorded in nate the pSTR.40,90 For example, NMDA eliminated both the
proximal retina in response to a weak diffuse stimulus was nSTR and pSTR in the macaque ERG for responses such as
negative-going for the duration of the stimulus, and returned those seen for the two weakest stimuli in Fig. 7.7 (Frishman,
slowly to baseline after light offset. For stimuli too weak to elicit unpublished observations).
A linear model of the contributions of pSTR, nSTR, and PII to the light-evoked proximal [K+]o increase was similar to that of
the dark-adapted cat ERG is shown in Fig. 7.22. The model the field potentials, and the retinal depth maxima for the two
196 assumes that each ERG component initially rises in proportion responses were the same.22,54 A causative role for the [K+]o
to stimulus strength, and then saturates in a characteristic increase in generating the nSTR (and a slow negative response
manner, as has been demonstrated in single-cell recordings in in the vitreal ERG following the initial STR) was supported by
mammalian retinas, as well as for ERG a- and b-waves in numer- the finding (Fig. 7.23) that Ba2+ removed the proximal retinal
Section 2

ous studies. Only with the inclusion of a small pSTR does the field potential and the nSTR in the ERG but did not, initially,
model accurately predict the whole ERG at a given “fixed” time eliminate the light-evoked increase in [K+]o. The cornea-negative
in response to a weak stimulus. The model was fit in Fig. 7.22 to polarity of the nSTR suggests a distally directed Müller cell K+
responses measured at 140 ms after a brief full-field flash current (similar to M-wave and PIII currents in the vascularized
(<5 ms), which was the peak of the nSTR in the cat scotopic ERG. cat retina). As noted above for the light-adapted M-wave, in the
Similar models have been applied to mouse90 and human ERG.40 dark-adapted retina Ba2+ also appears to block K+ siphoning by
Retinal Diagnostics
Retinal Imaging and Diagnostics

the Müller cells. Whereas the proximal [K+]o increase remained


K+ Müller cell mechanism for generation of the STR
intact when related field potentials were abolished, the distal
The STR, like the M-wave, is associated with Müller cell responses
[K+]o increase was eliminated by Ba2+.
to [K+]o released by proximal retinal neurons. In intraretinal
studies in cat, a proximal increase in [K+]o was observed that had Neuronal origins of the STR
clear similarities to the local STR that was simultaneously Whether the neurons involved in the genesis of the nSTR and
recorded: the dynamic range from “threshold” to saturation of pSTR are amacrine or ganglion cells is species-dependent. In
monkeys it is likely that the nSTR arises predominantly from
ganglion cells. It was absent in eyes in which the ganglion cells
were eliminated as a consequence of experimental glaucoma112
Dark-adapted and by intravitreal injection of TTX to block Na+-dependent
spiking activity of those neurons; the pSTR remained intact. In
400 contrast, in cats and humans122 as well as in rodents,3 the nSTR
10
is not eliminated by ganglion cell loss, and thus may be more
0 amacrine cell-based. In rodents the pSTR relies upon the integ-
ERG amplitude (µV) at 140 msec

300 rity of ganglion cells. A characteristic of Müller/glial cell-


–10
mediated ERG components is their slow timecourse. Glial cell
–20 mediation of the nSTR was demonstrated most directly in cat,
200 but the timecourse of the nSTR is slow in all species. Glial media-
–30
tion may explain the similarity in timecourse of nSTR across
–40 species regardless of the particular type of neuron producing the
100 106 local changes in proximal [K+]o that generate the response.
100
PII
pSTR
Origin of oscillatory potentials
0 The OPs of the ERG consist of a series of high-frequency, low-
nSTR amplitude wavelets, mainly superimposed on the b-wave, that
occur in response to strong stimulation. OPs are present under
–100 light- and dark-adapted conditions, with contributions from
100 101 102 103 104 105 both rod- and cone-driven signals.123 The mixed rod–cone ERG
Flash intensity (quanta.deg–2) in humans in the ISCEV standards in Fig. 7.4 shows at least four
OPs that can be extracted with a filter with a low-frequency
Fig. 7.22 Amplitude of cat dark-adapted electroretinogram responses cutoff of 75 Hz. OPs are of postreceptoral origin, and occur in
measured 140 ms after a brief flash at the maximum negativity of the isolated retinas in the absence of the RPE.73 The number of OPs
negative scotopic threshold response (nSTR). Dashed lines show induced by a flash of light varies between about four and 10
model curves for the positive scotopic threshold response (pSTR),
nSTR, and PII. Explicitly, the pSTR rises as a linear function that
depending upon species and stimulus conditions; the temporal
saturates abruptly at Vmax, while the exponential saturation of the nSTR frequency of the OPs varies as well. Figure 7.24 shows the photo­
is defined by: pic flash ERG of a macaque monkey (top); at least five OPs at
frequency of about 150 Hz can be seen by filtering the response
V = Vmax (1 − exp(−I /I o )
to extract the high-frequency signals.
where Vmax is the maximum saturated amplitude, and Io is the intensity There is consensus that OPs are generated in proximal retina.
for an amplitude of (1 – 1/e)Vmax, while the hyperbolic relation used for Three important and unresolved questions regarding their
PII is defined by: origins are: (1) Do all the OPs have the same origin? (2) Which
V = VmaxI /(I + I o ) cells generate the OPs? (3) What mechanisms are involved in
generating OPs?
where Vmax has the same meaning but Io is the intensity at which the
amplitude is Vmax/2. The inset shows the pSTR and nSTR at the lowest Do all the OPs have the same origin?
light levels that were used. (1 q deg2 is equivalent to ~–7.5 log sc cd In amphibia, where early studies were done, the various OPs do
m2/s). (Adapted from Frishman LJ, Robson JG. Processing, adaptation
not all have the same origin. Depth profiles from isolated frog
to environmental light. In: Archer SN, Djamgoz MBA, Loew ER, et al.,
editors. Adaptive mechanisms in the ecology of vision. Dordrecht: retina showed that the earliest OPs in the response arose near
Kluwer Academic.; 1999. p. 383–412, with permission.) the IPL whereas the later OPs arose more distally, perhaps in the
Fig. 7.23 Effect of Ba2+ on the scotopic
Ba2+ VK+ threshold response (STR) and b-wave (PII) in
A Before Proximal
scotopic electroretinogram (ERG) of the cat.
retina 197
(A) Effect of intravitreal Ba2+ (BaCl2 , 3.9 mM
Intraretinal ERG vitreal concentration) on the intraretinal STR
and slower K+-related negative response in
response to a 4-second stimulus and the

Chapter 7
0.5 mV simultaneously recorded light-evoked
0.25 mV decrease in [K+]o (VK+) in the proximal retina
measured at 10% retinal depth, proximal to
After Ba2+ the peak K+ change at 17% depth. Top
response was measured before Ba2+ and the
bottom response 56 minutes after Ba2+
injection. (B) Effect of intravitreal BaCl2
4 sec (3.9 mM vitreal concentration) on the STR

Electrogenesis of the Electroretinogram


and slow negative response in the vitreal
ERG (left) and the simultaneously recorded
B change in [K+]o in the subretinal space.
Measurements of the amplitude of dark-
Before Ba2+ adapted responses in the proximal retina and
Subretinal the light-evoked increase in [K+]o in proximal
Vitreal ERG space VK+ and distal retina before and after intravitreal
injection of Ba2+ were made with double-
7.0 barreled K+-sensitive microelectrodes.
(Reproduced with permission from Frishman
5 mV LJ, Steinberg RH. Light-evoked increases in
0.25 mV [K+]o in proximal portion of the dark-adapted
cat retina. J Neurophysiol 1989;61:1233–43.)
4.0
0.5 mV
0.1 mV

After Ba2+ P Illumination


(log q deg –2 sec –1)

7.0

4.0

4 sec

INL. Studies of Wachmeister124 in amphibian retina also found the other hand, genetic deletion of a major GABA receptor in the
that earlier OPs were depressed by GABA antagonists, the dopa- IPL, the GABAC receptor, enhances the OPs.127
mine antagonist haloperidol, β-alanine, and substance P, whereas
later OPs were depressed by the glycine antagonist strychnine Which cells generate the OPs?
and by ethanol. The observations described above indicate that amacrine or
In primates, intraretinal studies using stimuli that would elicit perhaps, in some instances, retinal ganglion cells are involved in
responses from both rod and cone systems did not find differ- generating OPs. A study of rabbit ERG indicates a distal to
ences in the depth profiles of the different OPs.44,125 Considering proximal progression in origin, with the late but not early OPs
the complexity of inner retinal circuitry, the similarity in depth having input from spiking cells.126 The role of ganglion cells in
profiles does not necessarily mean that the same neurons were the generation of OPs has been controversial, with inconsistent
involved in all cases. For example, in the photopic flash ERG reports on the effects of ganglion cell loss on OPs. In Ogden’s
response to brief stimuli, the major OPs are APB-sensitive in studies in primates, optic nerve section resulted in ganglion cell
primates and other mammals, indicating an origin in the ON degeneration and disappearance of the OPs, and antidromic
pathways, but later OPs and at light offset originate in the OFF stimulation of the optic nerve reduced OP amplitudes.125 Further,
pathway.105,126 Such a difference could lead to differences in the in the primate photopic ERG elicited using a multifocal para-
depth distribution in the IPL which is stratified in outer OFF digm with a slow sequence to allow formation of OPs, both TTX
sublamina and inner ON sublamina. and experimental glaucoma removed or reduced a high-
Pharmacologic agents that block inner retinal activity, such as frequency band of OPs, while affecting a lower-frequency
glycine and GABA, remove OPs in mammals, as does PDA,105 band less.105,106 It is possible in primates that high-frequency
but the effects were not reported to be specific for given OPs. On OPs (centered around 150 Hz, compared to >100 Hz for the
Monkey full-field flash ERG
198
50µV
40 mV
Section 2

+1.5 nA
0 20 40 60 80
Time after flash (ms)
Retinal Diagnostics
Retinal Imaging and Diagnostics

+0.5 nA
15µV

+0.4 nA

+0.3 nA

0 20 40 60 80 +0.2 nA
Time after flash (ms)

+0.1 nA
Fig. 7.24 Full-field flash photopic electroretinogram of a macaque
–70 mV
monkey (top) and the oscillatory potentials (OPs) extracted from these
records. Filtering was between 90 and 300 Hz for OPs that occurred
0 100 200 300
between 100 and 200 Hz. The stimulus was a xenon flash presented
on a rod-saturating blue background. (Adapted from Rangaswamy NV, Time (ms)

current
Hood DC, Frishman LJ. Regional variations in local contributions to the
primate photopic flash ERG: revealed using the slow-sequence
mfERG. Invest Ophthalmol Vis Sci 2003;44:3233–47).

Fig. 7.25 Oscillatory potentials of isolated wide-field amacrine cells in


the white perch retina. Oscillatory membrane potentials (OMPs) were
lower-frequency band) under fully photopic conditions reflect elicited by depolarizing current steps of increasing amplitude obtained in
whole-cell recordings from isolated amacrine cells that were maintained
activity in ganglion cell axons, and the manifestation of an optic
in culture. The duration and frequency of the OMPs increased with
nerve head component of the ERG. Under conditions where rod depolarization. Voltage traces are shifted vertically for visibility. Bottom:
signals are also involved, the ganglion cell contribution may be application of depolarizing pulse. The magnitude of depolarizing current
less prominent. In other mammals, TTX-sensitive OPs may be is indicated with each trace. Holding potential was –70 mV. (Reproduced
with permission from Vigh J, Solessio E, Morgans CW, et al. Ionic
among the late ones. mechanisms mediating oscillatory membrane potentials in wide-field
retinal amacrine cells. J Neurophysiol 2003;90:431–43.)
What mechanisms are involved in generating OPs?
Neuronal interaction; inhibitory feedback circuits
Two mechanisms are commonly proposed for generating OPs:
neuronal interactions/feedback circuits, and the intrinsic mem- in these isolated cells indicated that they arose from “a complex
brane properties of cells, both of which implicate amacrine cells. interplay between voltage-dependent Ca2+ currents and voltage-
The case for an inhibitory feedback mechanism is supported by and Ca2+-dependent K+ currents.”128
involvement of receptors for GABA and glycine, which are In summary, there is consensus that high-frequency OPs origi-
prominent in feedback circuits in the inner retina. Gap junctions nate from the inner retina. The exact cellular origins may depend
between inner retinal neurons can also be involved in feedfor- upon species and stimulus conditions. The mechanisms by
ward mechanism. which they are generated are unresolved, with evidence for both
OPs in intracellular responses from neurons involvement of feedback circuitry, and for intrinsic membrane
In retinal intracellular recordings from many different species, mechanisms in amacrine cells.
high-frequency oscillations have rarely been observed in
responses of photoreceptors, horizontal, or bipolar cells. In con-
CLOSING REMARKS
trast, membrane oscillations have been observed in recordings This chapter has reviewed the increasingly specific information
from amacrine cells, especially in turtle and fish retina. For about the electrogenesis of the ERG that has become available as
example, GABAergic wide-field ACs isolated from white bass our understanding of retinal microcircuitry and cellular and syn-
retina generated oscillatory membrane potentials in response to aptic mechanisms has increased. Due to the extensive research
extrinsic depolarization, as illustrated in Fig. 7.25, that reached on ERG, it has become feasible to use it to assess retinal function
more than 100 Hz for strong depolarization. Analysis of the at every level, from the RPE to the optic nerve head; also see
mechanism of generation of the oscillatory membrane potentials other recent reviews on this subject.3,129–131 Tables 7.1 and 7.2
2. Frishman LJ. Electrogenesis of the ERG. In: Ryan SJ, editor. Retina. 4th ed.
Table 7.1 Retinal cells contributing to the flash and flicker St Louis, MO: Elsevier/Mosby; 2005. p. 103–35.
electroretinogram in standard clinical testing 3. Frishman LJ, Wang MH. Electroretinogram of human, monkey and mouse.
In: Kaufman PL, Alm A, editors. Adler’s physiology of the eye. 11th ed. 199
a-wave Photoreceptors Edinburgh: Elsevier; 2011. p. 480–501.
Dark-adapted a-wave: rods Light-adapted: 4. Dawson WW, Trick GL, Litzkow CA. Improved electrode for electroretino­
cones graphy. Invest Ophthalmol Vis Sci 1979;18:988–91.
5. Hawlina M, Konec B. New noncorneal HK-loop electrode for clinical

Chapter 7
Late postreceptoral contributions from OFF
electroretinography. Doc Ophthalmol 1992;81:253–9.
cone (hyperpolarizing) bipolar cells (HCB)
6. Evers HU, Gouras P. Three cone mechanisms in the primate electroretino-
and more proximal cells in the OFF gram: two with, one without off-center bipolar responses. Vision res 1986;26:
pathway 245–54.
7. Rangaswamy NV, Shirato S, Kaneko M, et al. Effects of spectral characteristics
b-wave Bipolar cells of Ganzfeld stimuli on the photopic negative response (PhNR) of the ERG.
Dark-adapted b-wave, rod bipolar cells Invest Ophthalmol Vis Sci 2007;48:4818–28.
(RBC) 8. Newman EA. Membrane physiology of retinal glial (Müller) cells. J Neurosci
1985;5:2225–39.

Electrogenesis of the Electroretinogram


Light-adapted b-wave, ON cone
9. Connors NC, Kofuji P. Potassium channel Kir4.1 macromolecular complex in
(depolarizing bipolar cells: DCB), OFF retinal glial cells. Glia 2006;53:124–31.
cone bipolar cells, and horizontal (Hz) 10. Newman EA, Frambach DA, Odette LL. Control of extracellular potassium
cell feedback via the cones levels by retinal glial cell K+ siphoning. Science 1984;225:1174–5.
11. Newman E, Reichenbach A. The Müller cell: a functional element of the retina.
d-wave Bipolar cells Trends Neurosci 1996;19:307–12.
Mainly a light-adapted response: OFF cone 12. Kofuji P, Biedermann B, Siddharthan V, et al. Kir potassium channel subunit
expression in retinal glial cells: implications for spatial potassium buffering.
bipolar cells, offset of cone
Glia 2002;39:292–303.
photoreceptors, and offset of ON cone 13. Brown KT, Wiesel TN. Localization of origins of electroretinogram compo-
bipolar cells nents by intraretinal recording in the intact cat eye. J Physiol 1961;158:
The d-wave is not present in mice and rats 257–80.
14. Ogden TE, Ito H. Avian retina. II. An evaluation of retinal electrical aniso­
Oscillatory Amacrine and ganglion cells, bipolar cell tropy. J Neurophysiol 1971;34:367–73.
potentials terminals? 15. Karwoski CJ, Frambach DA, Proenza LM. Laminar profile of resistivity in frog
retina. J Neurophysiol 1985;54:1607–19.
(OPs)
16. Brown KT. The electroretinogram: its components and their origins.
Vision res 1968;8:633–77.
“30 Hz” fast Bipolar cells 17. Freeman JA, Nicholson C. Experimental optimization of current source-
flicker ON and OFF cone bipolar cells, and a small density technique for anuran cerebellum. J Neurophysiol 1975;38:369–82.
cone photoreceptor contribution 18. Heynen H, van Norren D. Origin of the electroretinogram in the intact
macaque eye–II. Current source-density analysis. Vision Res 1985;25:709–15.
19. Xu X, Karwoski CJ. Current source density (CSD) analysis of retinal field poten-
tials. I. Methodological considerations and depth profiles. J Neurophysiol
1994;72:84–95.
Table 7.2 Retinal cells contributing to the electroretinogram
20. Dick E, Miller RF. Extracellular K+ activity changes related to electroretino-
(ERG) in specialized testing gram components. I. Amphibian (I-type) retinas. J Gen Physiol 1985;85:
885–909.
Scotopic threshold Inner retinal neurons 21. Dick E, Miller RF, Bloomfield S. Extracellular K+ activity changes related to
response electroretinogram components. II. Rabbit (E-type) retinas. J Gen Physiol
1985;85:911–31.
pSTR Amacrine cells (monkey) 22. Frishman LJ, Steinberg RH. Light-evoked increases in [K+]o in proximal
Retinal ganglion cells (rodents) portion of the dark-adapted cat retina. J Neurophysiol 1989;61:1233–43.
23. Frishman LJ, Yamamoto F, Bogucka J, et al. Light-evoked changes in [K+]o in
proximal portion of light-adapted cat retina. J Neurophysiol 1992;67:
nSTR Retinal ganglion cells (monkey) 1201–12.
Partially retinal ganglion cells (rats, 24. Oakley 2nd B, Green DG. Correlation of light-induced changes in retinal
human?) extracellular potassium concentration with c-wave of the electroretinogram.
Amacrine cells (AII) (mice) J Neurophysiol 1976;39:1117–33.
Partially amacrine cells (rats, human) 25. Karwoski CJ, Lu HK, Newman EA. Spatial buffering of light-evoked
potassium increases by retinal Müller (glial) cells. Science 1989;244:578–80.
Glial currents
26. Halgrimson CG, Wilson CB, Dixon FJ, et al. Goodpasture’s syndrome. Treat-
ment with nephrectomy and renal transplantation. Arch Surg 1971;103:
Photopic negative Retinal ganglion cells (human, monkey) 283–9.
response Amacrine cells (rodents) 27. Newman EA. Potassium conductance block by barium in amphibian Müller
(PhNR) Glial currents cells. Brain Res 1989;498:308–14.
28. Kofuji P, Ceelen P, Zahs KR, et al. Genetic inactivation of an inwardly rectify-
ing potassium channel (Kir4.1 subunit) in mice: phenotypic impact in retina.
Pattern ERG Retinal ganglion cells (human, monkey,
J Neurosci 2000 ;20:5733–40.
(PERG) rodent) 29. Penn RD, Hagins WA. Signal transmission along retinal rods and the origin
Glial currents (transient PERG: N95, N2?) of the electroretinographic a-wave. Nature 1969;223:201–4.
30. Robson JG, Frishman LJ. Response linearity and kinetics of the cat retina: the
Multifocal ERG Initial negative and positive waves have bipolar cell component of the dark-adapted electroretinogram. Vis Neurosci
similar origins to a- and b-waves in 1995;12:837–50.
photopic ERG (monkey, and 31. Robson JG, Maeda H, Saszik SM, et al. In vivo studies of signaling in rod
pathways of the mouse using the electroretinogram. Vision Res 2004;44:
presumably human)
3253–68.
32. Granit R. The components of the retinal action potential in mammals and their
relation to the discharge in the optic nerve. J Physiol 1933;77:207–39.
33. Bush RA, Sieving PA. A proximal retinal component in the primate photopic
summarize our current understanding of the origins of ERGs ERG a-wave. Invest Ophthalmol Vis Sci 1994;35:635–45.
recorded in standard and specialized tests. Continued refine- 34. Robson JG, Saszik SM, Ahmed J, et al. Rod and cone contributions to
ment of stimuli, protocols, and analysis will further enhance the the a-wave of the electroretinogram of the macaque. J Physiol 2003;547:
509–30.
power of this noninvasive tool in the laboratory and the clinic. 35. Sieving PA, Murayama K, Naarendorp F. Push-pull model of the primate
photopic electroretinogram: a role for hyperpolarizing neurons in shaping the
REFERENCES b-wave. Vis Neurosci 1994;11:519–32.
1. Frishman LJ. Origins of the ERG. In: Heckenlively J, Arden GB, editors. Prin- 36. Breton ME, Schueller AW, Lamb TD, et al. Analysis of ERG a-wave amplifica-
ciples and practice of clinical electrophysiology of vision. 2nd ed. Cambridge, tion and kinetics in terms of the G-protein cascade of phototransduction.
MA: MIT Press; 2006. p. 139–83. Invest Ophthalmol Vis Sci 1994;35:295–309.
37. Hood DC, Birch DG. A quantitative measure of the electrical activity of 72. Hood DC, Birch DG. Human cone receptor activity: the leading edge of the
human rod photoreceptors using electroretinography. Vis Neurosci 1990;5: a-wave and models of receptor activity. Vis Neurosci 1993;10:857–71.
379–87. 73. Yonemura D, Kawasaki K, Shibata N. The electroretinographic PIII compo-
200 38. Hood DC, Birch DG. The A-wave of the human electroretinogram and rod nent of the human excised retina. Jpn J Ophthalmol 1974;18:322–33.
receptor function. Invest Ophthalmol Vis Sci 1990;31:2070–81. 74. Slaughter MM, Miller RF. An excitatory amino acid antagonist blocks cone
39. Robson JG, Frishman LJ. The a-wave of the electroretinogram: importance of input to sign-conserving second-order retinal neurons. Science 1983;
axonal currents. ARVO E-abstracts 2011:692. 219:1230–2.
40. Frishman LJ, Reddy MG, Robson JG. Effects of background light on 75. Friedburg C, Allen CP, Mason PJ, et al. Contribution of cone photoreceptors
Section 2

the human dark-adapted electroretinogram and psychophysical threshold. and post-receptoral mechanisms to the human photopic electroretinogram.
J Opt Soc Am A Opt Image Sci Vis 1996;13:601–12. J Physiol 2004;556:819–34.
41. Saszik S, Alexander A, Lawrence T, et al. APB differentially affects the cone 76. Jamison JA, Bush RA, Lei B, et al. Characterization of the rod photoresponse
contributions to the zebrafish ERG. Vis Neurosci 2002;19:521–9. isolated from the dark-adapted primate ERG. Vis Neurosci 2001;18:
42. Marmor MF, Fulton AB, Holder GE, et al. ISCEV standard for full-field clinical 445–55.
electroretinography (2008 update). Doc Ophthalmol 2009;118:69–77. 77. Robson JG, Frishman LJ. Dissecting the dark-adapted electroretinogram.
43. Steinberg RH, Linsenmeier RA, Griff ER. Retinal pigment epithelial cell con- Doc Ophthalmol 1998;95:187–215.
tributions to the electroretinogram and electrooculogram. Prog Retin Res 78. Pepperberg DR, Birch DG, Hood DC. Photoresponses of human rods in
Retinal Diagnostics

1985;4:33–66. vivo derived from paired-flash electroretinograms. Vis Neurosci 1997;14:


Retinal Imaging and Diagnostics

44. Heynen H, Wachtmeister L, van Norren D. Origin of the oscillatory potentials 73–82.
in the primate retina. Vision Res 1985;25:1365–73. 79. Stockton RA, Slaughter MM. B-wave of the electroretinogram. A reflection of
45. Noell W. Studies on the electrophysiology and the metabolism of the retina. ON bipolar cell activity. J Gen Physiol 1989;93:101–22.
USAF SAM project no 21-2101-004. United States Airforce: Randolph Field, 80. Knapp AG, Schiller PH. The contribution of on-bipolar cells to the electroret-
TX; 1953. p. 1–122. inogram of rabbits and monkeys. A study using 2-amino-4-phosphonobutyrate
46. Sillman AJ, Ito H, Tomita T. Studies on the mass receptor potential of the (APB). Vision Res 1984;24:1841–6.
isolated frog retina. I. General properties of the response. Vision Res 81. Bech-Hansen NT, Naylor MJ, Maybaum TA, et al. Mutations in NYX, encod-
1969;9:1435–42. ing the leucine-rich proteoglycan nyctalopin, cause X-linked complete
47. Valeton JM, van Norren D. Intraretinal recordings of slow electrical responses congenital stationary night blindness. Nature genet 2000;26:319–23.
to steady illumination in monkey: isolation of receptor responses and the 82. McCall MA, Gregg RG. Comparisons of structural and functional abnormali-
origin of the light peak. Vision Res 1982;22:393–9. ties in mouse b-wave mutants. J Physiol 2008;586:4385–92.
48. Bolnick DA, Walter AE, Sillman AJ. Barium suppresses slow PIII in perfused 83. Newman EA, Odette LL. Model of electroretinogram b-wave generation:
bullfrog retina. Vision Res 1979;19:1117–9. a test of the K+ hypothesis. J Neurophysiol 1984;51:164–82.
49. Hu KG, Marmor MF. Selective actions of barium on the c-wave and slow 84. Miller RF, Dowling JE. Intracellular responses of the Müller (glial) cells
negative potential of the rabbit eye. Vision Res 1984;24:1153–6. of mudpuppy retina: their relation to b-wave of the electroretinogram.
50. Witkovsky P, Dudek FE, Ripps H. Slow PIII component of the carp electro- J Neurophysiol 1970;33:323–41.
retinogram. J Gen Physiol 1975;65:119–34. 85. Kline RP, Ripps H, Dowling JE. Light-induced potassium fluxes in the skate
51. Sillman AJ, Ito H, Tomita T. Studies on the mass receptor potential of the retina. Neuroscience 1985;14:225–35.
isolated frog retina. II. On the basis of the ionic mechanism. Vision Res 86. Wen R, Oakley 2nd B. K(+)-evoked Müller cell depolarization generates
1969;9:1443–51. b-wave of electroretinogram in toad retina. Proc Natl Acad Sci U S A 1990;87:
52. Faber D. Analysis of slow transretinal potentials in response to light. PhD 2117–21.
thesis. Buffalo: University of New York; 1969. 87. Karwoski CJ, Proenza LM. Neurons, potassium, and glia in proximal retina
53. Karwoski CJ, Proenza LM. Relationship between Müller cell responses, a local of Necturus. J Gen Physiol 1980;75:141–62.
transretinal potential, and potassium flux. J Neurophysiol 1977;40:244–59. 88. Xu X, Karwoski CJ. Current source density analysis of retinal field potentials.
54. Frishman LJ, Steinberg RH. Intraretinal analysis of the threshold dark-adapted II. Pharmacological analysis of the b-wave and M-wave. J Neurophysiol
ERG of cat retina. J Neurophysiol 1989;61:1221–32. 1994;72:96–105.
55. Hanitzsch R. Intraretinal isolation of P3 subcomponents in the isolated rabbit 89. Karwoski CJ, Xu X. Current source-density analysis of light-evoked field
retina after treatment with sodium aspartate. Vision Res 1973;13:2093–102. potentials in rabbit retina. Vis Neurosci 1999;16:369–77.
56. Miller SS, Steinberg RH. Passive ionic properties of frog retinal pigment 90. Saszik SM, Robson JG, Frishman LJ. The scotopic threshold response of
epithelium. J Membr Biol 1977;36:337–72. the dark-adapted electroretinogram of the mouse. J Physiol 2002;543:
57. Oakley 2nd B, Steinberg RH, Miller SS, et al. The in vitro frog pigment epi- 899–916.
thelial cell hyperpolarization in response to light. Invest Ophthalmol Vis Sci 91. Cameron AM, Mahroo OA, Lamb TD. Dark adaptation of human rod bipolar
1977;16:771–4. cells measured from the b-wave of the scotopic electroretinogram. J Physiol
58. Gallemore RP, Steinberg RH. Light-evoked modulation of basolateral mem- 2006;575:507–26.
brane Cl– conductance in chick retinal pigment epithelium: the light peak and 92. Sieving PA, Frishman LJ, Steinberg RH. M-wave of proximal retina in cat.
fast oscillation. J Neurophysiol 1993;70:1669–80. J Neurophysiol 1986;56:1039–48.
59. Quinn RH, Quong JN, Miller SS. Adrenergic receptor activated ion transport 93. Sieving PA, Nino C. Scotopic threshold response (STR) of the human electro-
in human fetal retinal pigment epithelium. Invest Ophthalmol Vis Sci retinogram. Invest Ophthalmol Vis Sci 1988;29:1608–14.
2001;42:255–64. 94. Ueno S, Kondo M, Ueno M, et al. Contribution of retinal neurons to d-wave
60. Wu J, Marmorstein AD, Striessnig J, et al. Voltage-dependent calcium of primate photopic electroretinograms. Vision Res 2006;46:658–64.
channel CaV1.3 subunits regulate the light peak of the electroretinogram. 95. Rangaswamy NV, Frishman LJ, Dorotheo EU, et al. Photopic ERGs in patients
J Neurophysiol 2007;97:3731–5. with optic neuropathies: comparison with primate ERGs after pharmacologic
61. Gallemore RP, Hughes BA, Miller SS, editors. Light-induced responses of the blockade of inner retina. Invest Ophthalmol Vis Sci 2004;45:3827–37.
retinal pigment epithelium. New York: Oxford University Press; 1998. 96. Peachey NS, Alexander KR, Fishman GA, et al. Properties of the human cone
62. Dawis SM, Niemeyer G. Dopamine influences the light peak in the perfused system electroretinogram during light adaptation. Appl Opt 1989;28:
mammalian eye. Invest Ophthalmol Vis Sci 1986;27:330–5. 1145–50.
63. Sieving PA, Frishman LJ, Steinberg RH. Scotopic threshold response of 97. Wali N, Leguire LE. The photopic hill: a new phenomenon of the light adapted
proximal retina in cat. J Neurophysiol 1986;56:1049–61. electroretinogram. Doc Ophthalmol 1992;80:335–45.
64. Wakabayashi K, Gieser J, Sieving PA. Aspartate separation of the scotopic 98. Hamilton R, Bees MA, Chaplin CA, et al. The luminance-response function of
threshold response (STR) from the photoreceptor a-wave of the cat and the human photopic electroretinogram: a mathematical model. Vision Res
monkey ERG. Invest Ophthalmol Vis Sci 1988;29:1615–22. 2007;47:2968–72.
65. Brown KT, Wiesel TN. Analysis of the intraretinal electroretinogram in the 99. Ueno S, Kondo M, Niwa Y, et al. Luminance dependence of neural
intact cat eye. J Physiol 1961;158:229–56. components that underlies the primate photopic electroretinogram. Invest
66. Brown KT, Watanabe K, Murakami M. The early and late receptor potentials Ophthalmol Vis Sci 2004;45:1033–40.
of monkey cones and rods. Cold Spring Harbor Symposia Quant Biol 100. Bush RA, Sieving PA. Inner retinal contributions to the primate photopic fast
1965;30:457–82. flicker electroretinogram. J Opt Soc Am A Opt Image Sci Vis 1996;13:557–65.
67. Penn RD, Hagins WA. Kinetics of the photocurrent of retinal rods. Biophys J 101. Kondo M, Sieving PA. Primate photopic sine-wave flicker ERG: vector model-
1972;12:1073–94. ing analysis of component origins using glutamate analogs. Invest Ophthal-
68. Pugh Jr EN, Falsini B, Lyubarsky A. The origin of the major rod- and cone- mol Vis Sci 2001;42:305–12.
driven components of the rodent electroretinogram and the effects of age and 102. Viswanathan S, Frishman LJ, Robson JG. Inner-retinal contributions to the
light-rearing history on the magnitude of these components. Photostasis and photopic sinusoidal flicker electroretinogram of macaques. Macaque photopic
related phenomena. New York: Plenum Press; 1998. p. 93–128. sinusoidal flicker ERG. Doc Ophthalmol 2002;105:223–42.
69. Baylor DA, Nunn BJ, Schnapf JL. The photocurrent, noise and spectral sensi- 103. Sutter EE, Tran D. The field topography of ERG components in man–I. The
tivity of rods of the monkey Macaca fascicularis. J Physiol 1984;357:575–607. photopic luminance response. Vision Res 1992;32:433–46.
70. Lamb TD, Pugh Jr EN. A quantitative account of the activation steps involved 104. Hood DC, Frishman LJ, Saszik S, et al. Retinal origins of the primate multifocal
in phototransduction in amphibian photoreceptors. J Physiol 1992;449: ERG: implications for the human response. Invest Ophthalmol Vis Sci 2002;
719–58. 43:1673–85.
71. Hood DC, Birch DG. Light adaptation of human rod receptors: the leading 105. Rangaswamy NV, Hood DC, Frishman LJ. Regional variations in local contri-
edge of the human a-wave and models of rod receptor activity. Vision Res butions to the primate photopic flash ERG: revealed using the slow-sequence
1993;33:1605–18. mfERG. Invest Ophthalmol Vis Sci 2003;44:3233–47.
106. Rangaswamy NV, Zhou W, Harwerth RS, et al. Effect of experimental glau- 119. Viswanathan S, Frishman LJ, Robson JG. The uniform field and pattern
coma in primates on oscillatory potentials of the slow-sequence mfERG. ERG in macaques with experimental glaucoma: removal of spiking activity.
Invest Ophthalmol Vis Sci 2006;47:753–67. Invest Ophthalmol Vis Sci 2000;41:2797–810.
107. Sutter EE, Bearse MA, Jr. The optic nerve head component of the human ERG. 120. Luo X, Frishman LJ. Retinal pathway origins of the pattern electroretinogram 201
Vision Res 1999;39:419–436. (PERG). Invest Ophthalmol Vis Sci 2011;52:8571–84.
108. Burkhardt DA. Proximal negative response of frog retina. J Neurophysiol 121. Naarendorp F, Sieving PA. The scotopic threshold response of the cat ERG is
1970;33:405–20. suppressed selectively by GABA and glycine. Vision Res 1991;31:1–15.
109. Frishman LJ, Steinberg RH. Origin of negative potentials in the light-adapted 122. Sieving PA. Retinal ganglion cell loss does not abolish the scotopic threshold

Chapter 7
ERG of cat retina. J Neurophysiol 1990;63:1333–46. response (STR) of the cat and human ERG. Clin Vis Sci 1991;2:149–58.
110. Ogden TE. The proximal negative response of the primate retina. Vision Res 123. Peachey NS, Alexander KR, Fishman GA. Rod and cone system contributions
1973;13:797–807. to oscillatory potentials: an explanation for the conditioning flash effect.
111. Sieving PA, Steinberg RH. Proximal retinal contribution to the intraretinal Vision Res 1987;27:859–66.
8-Hz pattern ERG of cat. J Neurophysiol 1987;57:104–20. 124. Wachtmeister L. Oscillatory potentials in the retina: what do they reveal.
112. Frishman LJ, Shen FF, Du L, et al. The scotopic electroretinogram of macaque Prog retinal eye res 1998;17:485–521.
after retinal ganglion cell loss from experimental glaucoma. Invest Ophthal- 125. Ogden TE. The oscillatory waves of the primate electroretinogram. Vision Res
mol Vis Sci 1996;37:125–41. 1973;13:1059–74.
113. Viswanathan S, Frishman LJ, Robson JG, et al. The photopic negative response 126. Dong CJ, Agey P, Hare WA. Origins of the electroretinogram oscillatory

Electrogenesis of the Electroretinogram


of the macaque electroretinogram: reduction by experimental glaucoma. potentials in the rabbit retina. Vis Neurosci 2004;21:533–43.
Invest Ophthalmol Vis Sci 1999;40:1124–36. 127. McCall MA, Lukasiewicz PD, Gregg RG, et al. Elimination of the rho1 subunit
114. Viswanathan S, Frishman LJ, Robson JG, et al. The photopic negative response abolishes GABA(C) receptor expression and alters visual processing in the
of the flash electroretinogram in primary open angle glaucoma. Invest mouse retina. J Neurosci 2002;22:4163–74.
Ophthalmol Vis Sci 2001;42:514–22. 128. Vigh J, Solessio E, Morgans CW, et al. Ionic mechanisms mediating oscillatory
115. Kondo M, Kurimoto Y, Sakai T, et al. Recording focal macular photopic nega- membrane potentials in wide-field retinal amacrine cells. J Neurophysiol
tive response (PhNR) from monkeys. Invest Ophthalmol Vis Sci 2008;49: 2003;90:431–43.
3544–50. 129. Heckenlively J, Arden GB. Principles and practice of clinical electrophysiology
116. Bach M, Hoffmann MB. Update on the pattern electroretinogram in glaucoma. of vision. 2nd ed. Cambridge, MA: MIT Press; 2006.
Optom Vis Sci 2008;85:386–95. 130. Fishman GA, Birch DG, Holder GE, et al. Electrophysiological testing in dis-
117. Holder GE. Pattern electroretinography (PERG) and an integrated approach orders of the retina, optic nerve, and visual pathways. 2nd ed. Singapore:
to visual pathway diagnosis. Prog Retin Eye Res 2001;20:531–61. American Academy of Ophthalmology; 2001.
118. Maffei L, Fiorentini A, Bisti S, et al. Pattern ERG in the monkey after section 131. Lam BL. Electrophysiology of vision; clinical testing and applications.
of the optic nerve. Exp Brain Res 1985;59:423–5. Boca Raton, FL: Taylor and Francis Group; 2005.
Section 2 Retinal Diagnostics

Chapter Clinical Electrophysiology


8  Yozo Miyake, Kei Shinoda

In this chapter, we describe the method and value of various


clinical electrophysiological tests and how they play a role in
the diagnosis, analysis of pathogenesis, prognostic evaluation,
and understanding of the underlying genetics of retinal dis­
orders, using several examples of clinical cases. We also dem­
onstrate the role of the focal macular electroretinogram (ERG)
and multifocal ERG (mfERG) for particular diseases where the
affected region is limited to a certain region of the retina. During
the recent history of clinical electrophysiology, several new
clinical entities have been defined by analyzing electrophysi­
ological results. These new entities will also be discussed in
this chapter.

STANDARD FULL-FIELD ERG


Stimulus and recording devices
The human ERG recorded at the cornea and elicited by a full-
field stimulus is a mass response generated by cells across the Fig. 8.1 Representative electroretinogram recording electrodes. Top,
entire retina. To obtain reproducible amplitudes and implicit Burian–Allen electrode. A contact lens-type electrode with a lid
speculum to minimize the effect of blinking and eyelid closure. Bottom,
times in the response, the stimulus and background light should Dawson–Trick–Litzkow (DTL) electrode – a conductive Mylar thread
be homogeneous and cover the entire retina, so all of the recep­ usually placed in the lower fornix, it contacts the inferior bulbar
tors are stimulated or adapted in a relatively homogeneous conjunctiva or the corneal limbus. Dots show 10-mm distance.
manner. The full-field, or Ganzfeld, stimulator represents such
a stimulus. It consists of a large-diameter (40-cm) hemispheric
dome (see Chapter 7, Electrogenesis of the electroretinogram)
with a xenon stroboscopic light bulb placed at the top of the At the left, the scotopic threshold response (STR),3 a cornea-
dome. This stimulus system has been recommended by the negative wave, is first recorded at –8.2 log units, approximately
International Society of Clinical Electrophysiology for Vision 0.6 log units higher than psychophysical threshold. The
(ISCEV) Standards Committee1 for use when obtaining clinical maximum amplitude of STR is approximately 20 µV before it is
ERG recordings internationally. masked by the developing b-wave. The implicit time of the STR
The ERG is recorded using corneal electrodes, usually referred near threshold is approximately 160 ms, and the implicit time
to surface reference electrodes at the ipsilateral outer canthi or decreases as the stimulus intensity increases. The STR originates
zygomatic fossae. Electrodes in common use2 include the Burian– from retinal neurons that are postsynaptic to the photoreceptors
Allen and the ERG jet, both of which are contact lens electrodes, (see Chapter 7, Electrogenesis of the electroretinogram).
the gold foil, Dawson–Trick–Litzkow (DTL), and H–K loop elec­ The b-wave is first seen at an intensity of –5.8 log units; the
trodes, which are noncontact lens. The representative electrodes amplitude increases and the implicit time shortens as the stimu­
are shown in Fig. 8.1. lus intensity increases. The amplitude of b-wave essentially
saturates at –3.4 log units; and at intensities higher than –0.8
Stimulus intensity versus ERG responses log unit, the oscillatory potentials (OPs) become clearly visible
and components on the ascending limb of the b-wave. The a-wave is first seen
Scotopic condition at –1.7 log units and increases progressively as the stimulus
Figure 8.2 shows the full-field ERGs elicited by increasing stimu­ intensity increases.
lus intensities from a normal subject after 1 hour of dark adapta­ As shown fully in Chapter 7, Electrogenesis of the ERG, many
tion. The ERGs elicited by relatively weak stimulus intensities studies have shown that the a-wave of the full-field ERGs
are shown at the left and those by stronger stimulus are shown recorded in the dark is the leading edge of the photoreceptor
at the right. The calibrations for the amplitude and time are dif­ potential.4 The b-wave originates indirectly from bipolar and
ferent for the weak and strong stimulus ERGs. The maximum Müller cells in the middle layers of the retina.5 The OPs are seen
stimulus luminance (0 log unit) is 44.2 cd/m2/s. as a series of three or four rhythmic wavelets having almost
Fig. 8.2 The full-field electroretinogram
–5.4 (ERG) elicited by increasing stimulus
–8.8 intensities recorded from a normal subject
after 1 hour of dark adaptation. The left 203
–8.2 –4.4 column shows responses elicited by relative
STR low intensity and the right by relative high
–7.6 intensity. Note that the calibration differs for

Chapter 8
–3.9
the ERGs in the two columns. Arrowheads
–7.1 indicate the stimulus onset. STR, scotopic
–3.4 threshold response; b, b-wave; a, a-wave;
–6.8 Op, oscillatory potentials. (Reproduced with
log relative intensity

–1.7 permission from Miyake Y, Horiguchi M,


Terasaki H, et al. Invest Ophthalmol Vis Sci
–6.2 1994;35:3770–5.)
–1.4

Clinical Electrophysiology
b a

–5.8 –0.8
Op

–0.3

0.0
–5.4

50 µV
100 mse 200 µV
50 msec

equal intervals of about 6.5 ms in humans.6 The best experimen­


tal evidence indicates that the OPs reflect the activity of feedback 0.6
synaptic circuits within the retina and represent an inhibitory or 1.0
modulating effect of amacrine cells on the b-wave.7
Photopic condition 1.5
The photopic, short-flash ERGs elicited by increasing stimulus
intensities in a normal subject are shown in Fig. 8.3.8 At
Log flash intensity (cd/m2)

1.9
lower stimulus intensities, the amplitude of the b-wave increases b
with increasing stimulus until it reaches a maximum at a i
2.3
stimulus intensity of 3.0 log cd/m2. Further increases in the
a
stimulus intensity result in a progressive decrease in the
amplitude of the b-wave. Because a plot of the b-wave ampli­ 2.7
tude as a function of the stimulus intensity has an inverted
U shape, this phenomenon has been termed the photopic hill
phenomenon.9. 3.0

Bright flash mixed rod–cone ERG 3.3


ERG recorded with a bright flash of light after dark adaptation
for 30 minutes or longer (0 log unit in Fig. 8.2) shows mixed
3.5
rod–cone response, which can provide variable information
about retinal pathology and is of significant diagnostic value.
We have an impression that about 70% of ERG information 3.9
can be obtained by the evaluation of only the mixed rod–cone
ERG. The five different types of mixed rod–cone ERG are shown 100 µV
in Fig. 8.4. 5 ms 25 ms

Normal
The normal type shows a-wave, b-wave, and OPs. The ampli­
tude of b-wave is always larger than that of a-wave in the Fig. 8.3 Photopic short-flash electroretinograms elicited by various
regular stimulus intensity range. The normal ERG can be stimulus intensities from a normal subject. Stimulus duration is 5 ms
and the constant background illumination is 40 cd/m2. The vertical
seen in patients with localized macular dysfunction, optic dashed line indicates 30 ms. The b-wave amplitude increases with
nerve diseases, and central nervous system disease such as increasing stimulus intensity until 3.0 log cd/m2. It decreases with
amblyopia. Even when the entire retina is ophthalmoscopically further increases in stimulus intensity. When the b-wave amplitude is
abnormal, such as rubella retinopathy or female carrier of plotted against stimulus intensity, it shows an inverted U shape; this
phenomenon has been termed the photopic hill phenomenon.
ocular albino and choroideremia (Fig. 8.5), the ERG can be (Reproduced with permission from Kondo M, Piao CH, Tanikawa A,
essentially normal. et al. Japanese Journal of Ophthalmology 2000;44:20–8.)
Selectively abnormal oscillatory potentials
Mass ERG An OP abnormality means either reduction of amplitude or
204 delay of implicit time, or both. A selective OP abnormality is
observed in the early stage of diabetic retinopathy10,11 (Fig. 8.6)
normal or mild circulatory disturbance of retina such as central retinal
Section 2

vein occlusion.
Subnormal
The amplitudes of all components are reduced approximately to
OP (–)
the same degree. A reduced a-wave indicates abnormal photo­
receptor function. This pattern is seen in patients with localized
damage of the photoreceptors, such as partial retinal detachment
Retinal Diagnostics
Retinal Imaging and Diagnostics

or sectoral retinal degeneration. The amplitude of the full-field


ERG is proportional to the area of functioning retina. This rule
subnormal
is shown when the extent of retinal detachment is compared
with the ERG (Fig. 8.7).
negative This rule is also shown in eyes with panretinal photocoagula­
tion (PRP) in diabetic retinopathy. Following PRP, the ampli­
tudes of ERG components are reduced by 40–45%, but the
200 µV b-wave : a-wave (b/a) ratio is not changed significantly.11 When
extinct 25 msec the media is hazy due to vitreous hemorrhage and the fundus is
invisible, the presence or absence of retinal detachment is an
important evaluation preoperatively. By combining ERG and
Fig. 8.4 The five different types of mixed rod–cone electroretinogram ultrasonography, the differentiation between totally detached
(ERG). OP(–), selective reduction of oscillatory potentials; subnormal,
both a- and b-waves are attenuated approximately to the same degree; retina and dense vitreous membrane may be possible, as shown
negative, the amplitude of the b-wave is smaller than that of the in Fig. 8.8. When the ERG is recordable, even if the amplitude
a-wave; extinct, no discernible a- or b-wave.

A B

Fig. 8.5 Fundus photograph (A) and fluorescein angiogram (B) obtained
from a 20-year-old man with rubella retinitis. Fundus photograph from a
60-year-old female carrier of choroideremia (C). The electroretinograms
in these two patients were normal. (Reproduced with permission from
C Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag;
2006.)
is small, the thick membrane in the vitreous cavity is not totally
O1 O2 detached retina, but vitreous membrane.
O3
O4 205
Normal Negative
The negative ERG indicates that the amplitude of the b-wave is
smaller than that of the a-wave (b/a ratio <1.0). As mentioned

Chapter 8
above, the amplitude of the b-wave is always larger than that of
DR the a-wave in normal subjects. A normal a-wave with a reduced
Case 1
b-wave loca­lizes the defect to postphototransduction processes.
The negative ERG can be of useful prognostic or diagnostic value
in retinal diseases.
Case 2 Prognostic value

Clinical Electrophysiology
Among the acquired retinal diseases, the negative ERG may be
100 µV seen in severe retinal circulatory disturbance such as central
50 ms retinal arterial occlusion or proliferative diabetic retinopathy. In
central retinal vein occlusion, the ischemic type shows negative
ERG more frequently than nonischemic type, indicating that the
Fig. 8.6 Oscillatory potentials (OPs) of full-field electroretinograms b/a ratio can be an important index for evaluating the prognosis
recorded from a normal subject (top) and two patients with diabetic of central retinal vein occlusion.12,13 Figure 8.9 shows a patient
retinopathy (cases 1 and 2). The oscillatory potentials were found to
have delayed implicit time (case 1) or reduced amplitude (case 2).
with an initially normal, but later lower, b/a ratio which resulted
DR, diabetic retinopathy. (Reproduced with permission from Miyake Y. in negative configuration in ERG.11 The fluorescein angiogram
Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.) changed from the nonischemic pattern to the ischemic pattern,
showing an extensive nonperfusion area accordingly.
When massive vitreous hemorrhage prevents ophthalmo­
scopic examination of the fundus in patients with proliferative
diabetic retinopathy, it makes it difficult to predict the surgical
Normal
and visual outcome after vitrectomy. In these eyes, the ampli­
tudes of the ERGs may be markedly reduced by various factors:
pathological changes induced by the diabetic retinopathy, earlier
PRP, and vitreous hemorrhage. As mentioned above, the PRP
reduces the ERG amplitude without changing the b/a ratio.11
Retinal detachment
Because most diabetic patients with vitreous hemorrhage have
undergone PRP, it is difficult to arrive at a prognosis of the
Case 1 outcome after vitrectomy using only the amplitudes. The b/a
ratio provides more useful information about the visual progno­
sis after vitrectomy.14 The preoperative mixed rod–cone ERGs
were classified into three groups in patients with diabetic
retinopathy associated with significant vitreous hemorrhage
(Fig. 8.10, left). Group A indicates those with a b/a ratio>1.0 and
the OPs are clearly recordable. Group B includes those with a
b/a ratio >1.0 but the OPs are absent. Group C comprises those
with a b/a ratio <1.0 with absent OPs. Thick proliferative tissues
Case 2 were found at the disc (Fig. 8.11) intraoperatively in 36% of the
eyes in group A, 67% in group B, and 90% in group C.14 It was
suggested that the fibrous proliferation at the disc may restrict
retinal circulation by compressing the central retinal artery.
The distribution of the postoperative visual acuity for each
group is shown in Fig. 8.10, right.14 The postoperative visual
acuity for group C was significantly worse than for group A or
group B. The low b/a ratio may indicate more severe ischemic
retina, which in turn may account for the relatively good correla­
Case 3 tion with visual acuity. However, among the patients in group
C, there were some whose postoperative visual acuity was good,
indicating that a b/a ratio <1.0 is not necessarily a contraindica­
tion for vitrectomy. Another important finding is that most
patients who have distinct OPs preoperatively have favorable
visual acuity after vitrectomy. This observation is important
when we discuss the visual prognosis with patients before
Fig. 8.7 Mixed rod–cone (bright flash) electroretinograms (left) and
surgery. The light-filtering effect of a dense vitreous hemorrhage
fundus drawings of three patients with rhegmatogenous retinal
detachment (right). The reduction of the electroretinogram amplitude should also be considered when evaluating the preoperative
corresponds proportionally to the extent of retinal detachment. ERG in diabetic patients. Severe vitreous hemorrhage reduces
Fig. 8.8 Ultrasonographic (US) image (top)
and mixed rod–cone electroretinogram (ERG)
CASE 1 CASE 2 (bottom) from eyes with vitreous hemorrhage.
206 Ultrasonography shows thick membrane-like
reflex in the vitreous cavity in both eyes.
When the ERG is recordable, even if the
amplitude is small, the thick membrane in the
Section 2

vitreous cavity is not totally detached retina,


but vitreous membrane (case 1). In contrast,
when the ERG is unrecordable, the thick
US membrane is most likely totally detached
retina (case 2).
Retinal Diagnostics
Retinal Imaging and Diagnostics

ERG

10 µV
20 msec

100 µV
10 msec

Fig. 8.9 Top panel, A 39-year-old woman had a central retinal vein occlusion in the right eye (top left). Fluorescein angiogram (top center) and
electroretinogram (ERG) (top right) showed nonischemic pattern at her initial visit. Bottom panel, One month later, the retinal hemorrhage
increased (bottom left), the fluorescein angiogram showed extensive areas of nonperfusion (bottom center), and the waveform of the ERG
became negative (right). (Reproduced with permission from Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.)
Fig. 8.10 Preoperative full-field
* electroretinograms (ERGs) recorded from
Normal a normal control and three diabetic patients
* with vitreous hemorrhage (HM) who were 207
classified into three groups (left).
1.0
Postoperative visual acuity in the three

Postoperative visual acuity


groups classified according to the ERG

Chapter 8
Group A
waveform (right), showing that the
postoperative visual acuity for group C was
significantly worse than that for group A or
group B. (Reproduced with permission from
0.1 Kondo M, Piao CH, Tanikawa A, et al.
Group B
Japanese Journal of Ophthalmology
2000;44:20–8.)

Clinical Electrophysiology
Group C 0.01
HM
100 µV
10 msec A B C
Group

On the other hand, when endophthalmitis develops a relatively


long time after surgery and the ERG b/a ratio is >1.0, the timing
of the vitrectomy is not as critical. Representative examples15 are
shown in Fig. 8.13.
Diagnostic value
The negative ERG is seen in some hereditary retinal diseases,
which provides diagnostic information, particularly when the
a-wave amplitude is normal. The representative diseases, where
the negative ERG shows the diagnostic value, include complete-
type congenital stationary night blindness16 (CSNB: see Fig. 8.20,
below), incomplete-type CSNB16 (see Fig. 8.20, below), X-linked
juvenile retinoschisis11 (XLRS: see Fig. 8.20), juvenile-onset neu­
ronal ceroid lipofuscinosis,17 and infantile Refsum disease. Since
both complete and incomplete CSNB show essentially normal
fundi and most patients with CSNB have moderately low visual
acuity,16 the negative ERG finding is extremely important to pick
Fig. 8.11 Proliferative tissue on the optic disc in a patient with up these disorders, differentiating them from other diseases with
diabetic retinopathy. (Reproduced with permission from Miyake Y. normal fundi, low visual acuity, and normal ERG, such as psy­
Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.) chological eye problems, amblyopia, optic nerve disease, central
nervous system disease, or occult macular dystrophy (OMD).
the intensity of the stimulus light reaching the retina, which can The detailed findings separating rod and cone components will
increase the b/a ratio (Fig. 8.2). When the vitreous hemorrhage be treated later.
is extremely dense, the intensity of stimulus light may be When the a-wave amplitude is normal and the b/a ratio is less
decreased and the effective stimulus light to evoke ERG may not than 1.0, the selective abnormality of the second-order neuron is
reach the retina. In such situations, we need a much brighter indicated. On the other hand, when the amplitude of the a-wave
stimulus than the regular maximum stimulus to evoke ERG. is smaller than normal with the b/a ratio <1.0, there are two
Such an example is shown in Fig. 8.12. In such cases, we have interpretations. One is the combined dysfunction of photorecep­
an impression that ERG often has a negative configuration, as tor and middle retinal layer. This situation is often observed in
shown in this patient. patients with retinitis pigmentosa. The other is the ERG showing
In the prognostic evaluation of eyes that develop endophthal­ the photopic hill phenomenon8 (see above). When the rod func­
mitis after intraocular lens implantation, the b/a ratio is also tion is completely gone and the cone function is well preserved,
valuable.15 Eyes with early (within 1 week) endophthalmitis the ERG shows cone ERG even in the dark. In this condition,
associated with a b/a ratio of <1.0 have a worse postoperative when the stimulus light intensity is strong, the ERG reveals the
prognosis than eyes with late-onset endophthalmitis and/or a photopic hill phenomenon (Fig. 8.3), showing negative configu­
b/a ratio of >1.0. These observations are quite important when ration with a small a-wave. Such examples are seen in bright
deciding on the appropriate time to perform vitrectomy for treat­ flash mixed rod–cone ERG in the dark in Oguchi disease or
ment. For example, a patient with endophthalmitis that was fundus albipunctatus (FA) (Fig. 8.16).
detected within 1 week of intraocular lens implantation and with In acquired diseases, negative ERG may be seen in melanoma-
an ERG b/a ratio of <1.0 should undergo vitrectomy urgently. associated retinopathy,18,19 birdshot choroidopathy,20 ocular
Isolation of rod and cone components in
standardized ERG
208
Although the rods outnumber the cones 13 to 1 in the normal
human retina, the cone ERG response accounts for 20–25% of the
ERG response amplitude. For the purposes of diagnosis, it often
Section 2

becomes necessary for the examiner to evaluate rod and cone


activity separately. The full-field ERGs using the ISCEV Stan­
dard (see Chapter 7, Electrogenesis of the electroretinogram) in
a normal subject are shown in Fig. 8.14.
After 30 minutes of dark adaptation, a rod (scotopic) ERG is
recorded with a dim flash of light at approximately –3.9 log units
Retinal Diagnostics
Retinal Imaging and Diagnostics

in Fig. 8.2. A bright flash (mixed rod–cone) ERG is elicited by a


single flash of white light at maximum intensity of log 0 units in
ERG Fig. 8.2. Cone and 30-Hz flicker ERG are recorded with a stimu­
lus intensity of 3.3 log units in Fig. 8.3 under the background
illumination of 40 cd/m2, which is sufficient to suppress all rod
activity. The photopic recordings (cone and 30-Hz flicker ERG)
regular are made after 10 minutes of light adaptation to 40 cd/m2,
because the maximum photopic ERG can be obtained when
recorded after light adaptation.11 In addition to the conventional
ERG components, the photopic negative response (PhNR) was
introduced21: this originates from retinal ganglion cells and will
be treated more fully later.
The representative patients diagnosed by the isolation of rod
and cone components of full-field ERG are shown below in
relation to the abnormal cells of the retina.

Cone photoreceptor dysfunction


bright The congenital stationary disorder of cone dysfunction is rep­
flash
resented by rod monochromacy which is inherited in an auto­
somal recessive mode. This disorder is characterized in the
complete form by complete absence or severely depressed color
vision, reduced visual acuity, nystagmus, and photophobia11
50 µV
40 msec (see Chapter 44, Abnormalities of cone and rod function). There
is also an incomplete form of this disorder, where color vision
and/or visual acuity is not severely affected11 (see Chapter 44).
Fig. 8.12 Ultrasonographic image (top) and mixed rod–cone In both forms, the fundus and fluorescein angiograms are
electroretinograms (ERGs) (bottom) with various stimulus intensities normal, and the most characteristic feature in terms of the
from eyes with extremely dense vitreous hemorrhage. As the intensity diagnosis is selective reduction or absence of the photopic com­
of the stimulus light is decreased, a sufficiently bright stimulus to
evoke the ERG may not reach the retina. In this situation, a much ponents while preserving normal scotopic components of the
brighter stimulus than the regular maximum stimulus may evoke the full-field ERG even in incomplete form (Fig. 8.15). Molecular
ERG response. In such cases, it appears that the ERG frequently genetic studies have shown that mutations in the CNGB3 gene
shows negative configuration.
encoding the β-subunit of the cone photoreceptor cGMP-gated
channel are responsible for rod monochromacy.22
Blue cone monochromacy shares many characteristics with
siderosis, quinine retinopathy, and methanol toxicity. The
rod monochromacy, except that the hereditary mode is X-linked
negative configuration of ERG provides the diagnostic value
recessive11 (see Chapter 44, Abnormalities of cone and rod func­
in these disorders (see Chapters 76, White spot syndromes
tion). The visual acuity is approximately 0.2–0.3, which is slightly
and related diseases; 77, Autoimmune retinopathies; 89, Drug
better than that of the complete form of rod monochromacy.
toxicity of the posterior segment; and 134, Remote effects of
Unlike rod monochromacy, the blue cone function is selectively
cancer of the retina).
preserved. Panel D-15 test shows several crossing lines perpen­
Extinct dicular to the tritan axis (Fig. 8.15). The fundus is essentially
The extinct ERG is often seen in the advanced stage of rod– normal, although in the late stage some atrophic changes may
cone dystrophy, including retinitis pigmentosa, gyrate atrophy develop in the macula. The molecular genetics study indicated
or choroideremia, and total retinal detachment. In retinitis that mutations exist in the red and green opsin in the blue cone
pigmentosa, gyrate atrophy, or choroideremia, even when the monochromacy. The full-field ERGs are similar to those of rod
macular area is preserved, ERG may become undetectable. monochromacy, showing nearly normal rod ERGs with absence
Cancer-associated retinopathy,19 an autoimmune retinopathy, of the photopic ERG (Fig. 8.15). Although the blue cone ERG is
may often show extinct ERG and should be differentiated from normally present, the amplitude of the normal blue cone ERG is
retinitis pigmentosa. too small to be detected in the regular full-field cone ERG, and
Fig. 8.13 Left, Preoperative mixed rod–cone
(bright flash) electroretinograms (ERGs)
recorded from two patients with
endophthalmitis after intraocular lens 209
implantation. The negative configuration of
the ERG (case 1) suggests poorer visual
prognosis after vitrectomy than for the patient

Chapter 8
with normal-shaped ERGs (case 2). Right,
Postoperative fundus. Case 1 showed
extensive retinal vascular occlusions
(arrows), with poor postoperative visual
function, as expected. Case 2 showed an
essentially normal fundus with good
postoperative visual function. (Reproduced
with permission from Horio N, Terasaki H,

Clinical Electrophysiology
Yamamoto E, et al. Am J Ophthalmol
2001;132:258–9.)

100 µV
100 msec

is characterized by a peculiar grayish white discoloration of the


fundus. This unusual fundus coloration disappears after a long
period of dark adaptation, which is called the Mizuo–Nakamura
Rod 100 µV
phenomenon24 (see Chapter 44, Disorders of rod and cone func­
50 ms tion). Only the rod function is abnormal and is absent after 30
minutes of dark adaptation, but the subjective and electroretino­
graphic rod function may increase after 2–3 hours of dark adap­
Bright 200 µV tation.25 Mutations in the gene of arrestin26 or the rhodopsin
(mixed rod and cone)
25 ms kinase,27 both of which are rod phototransduction, cause the
recessive form of Oguchi disease.
Full-field ERGs (Fig. 8.16) recorded after 30 minutes of dark
adaptation show absent rod ERG and essentially normal cone-
100 µV
Cone mediated ERG.25 The mixed rod–cone ERG has a negative con­
25 ms figuration with relatively well-preserved OPs. The a-wave
amplitude in Oguchi disease is reduced compared to that of
normal controls. As mentioned above, this ERG indicates the
30-Hz 25 µV cone ERG in spite of the fact that the ERG is recorded in the dark
flicker
25 ms and after 30 minutes of dark adaptation, because rod function is
absent even under this condition. When the cone ERG is recorded
Fig. 8.14 Standard full-field electroretinograms with isolation of the rod with a bright flash of light, it shows the photopic hill phenom­
and cone components. Arrowheads indicate the stimulus onset. The enon, and the ERG configuration is negative with a small a-wave.
arrow indicates photopic negative response.
After 3 hours of dark adaptation, the amplitudes of the a-wave
and b-wave of the mixed rod–cone ERGs are larger, but they are
the implicit time is too long to follow 30-Hz flicker ERG stimuli
still negative in configuration. As the mutated genes indicate, the
(see section on S-cone ERG, below).
pathogenesis that only the rod itself is impaired in Oguchi disease
Rod photoreceptor dysfunction is comparable to the normal photopic ERG and negative configu­
The model diseases with congenital rod photoreceptor dysfunc­ ration with small a-wave in mixed rod–cone ERG, but the nega­
tion include Oguchi disease and FA, both of which are catego­ tive configuration with normal a-wave after a long time of dark
rized as CSNB. adaptation may suggest some additional abnormalities of the
Oguchi disease, first reported by Oguchi23 in 1907, is an bipolar cell function. Electro-oculogram (EOG) is abnormal with
unusual form of CSNB with autosomal recessive inheritance. It low light to dark ratio in most patients with Oguchi disease.25
Rod monochromacy Blue cone monochromacy
210 Normal
13y, F 18y, M Carrier Case 1 Panel D–15 test in Case 1
Section 2

Rod 100 µV 3 4 5
2
50 msec REFERENCE 1 6

PROTAN
CAP

DEUTAN
Cone 100 µV 7
25 msec
AN
TRIT
30-Hz 8
25 µV
Retinal Diagnostics
Retinal Imaging and Diagnostics

flicker
25 msec
15
9
Bright 200 µV 14
25 msec
13 10
12 11

Fig. 8.15 Full-field electroretinograms (ERGs) and Farnsworth dischromous panel D-15 test from patients with cone photoreceptor dysfunction.
Second and third columns (from left), full-field ERGs recorded from two siblings with rod monochromacy showing selective absence of the
photopic components. During 10-year follow-up, their visual function remained stable and their fundi remained normal. Visual acuity was 0.1/0.4 in
a 13-year-old sister and 1.0/1.0 in an 18-year-old brother. The sister showed mild acquired red–green deficiency and the brother had normal color
vision due to functional cones preserved only in the fovea.
Fourth and fifth (columns from left), full-field ERGs recorded from a family with blue cone monochromacy (carrier mother and son) showing normal
rod components and nearly absent cone components. Although the blue cone ERG is normally present, the amplitude of the normal blue cone ERG is
too small to be detected in the regular full-field cone ERG, and the implicit time is too long to follow 30-Hz flicker ERG stimuli.
Rightmost column, Farnsworth dischromous panel D-15 test from case 1 showing that several crossing lines were perpendicular to the tritan
axis. (Revised and reproduced with permission from Kondo M, Piao CH, Tanikawa A, et al. Japanese Journal of Ophthalmology 2000;44:20–8,
with permission.)

The pathogenesis of Oguchi disease has long been believed Although FA has been believed to be a stationary condition,
to exist in the rod bipolar function, as seen in complete CSNB, our study indicated that about one-third of patients with FA are
because reports published a long time ago28 indicated normal progressive, and associated with cone dystrophy.33 Such patients
a-wave with reduced b-wave (negative ERG) as well as normal often have bull’s-eye maculopathy (Fig. 8.17). In addition to the
EOG. In addition, normal rhodopsin kinetics were shown by characteristic ERG findings of FA, the photopic ERGs are
rhodopsin densitometry.29 We demonstrated that many patients extremely abnormal (Fig. 8.16). All of these patients also showed
with Oguchi disease show smaller a-wave than normal and RDH5 gene mutation.34 The ERG results have changed the
abnormal EOG, suggesting that there is a dysfunction of pho­ disease concept of FA, which had been believed to be a subtype
totransduction.25 Our hypothesis, obtained from electrophysi­ of CSNB.
ological results, was proven true by mutated gene of Oguchi
disease.26,27 Rod–cone or cone–rod photoreceptor dystrophy
FA has been considered to be a type of CSNB with autosomal Patients with cone–rod or rod–cone dystrophy belong clinically
recessive inheritance. The fundus has a characteristic appearance and genetically to a heterogeneous group of patients with inher­
of a large number of discrete, small, round or elliptical yellowish ited retinal dystrophies and the disease process is progressive.
white regions at the level of the retinal pigment epithelium (see They are characterized by widespread degeneration of predomi­
Chapter 44, Abnormalities of cone and rod function). The most nantly the cone (cone dystrophy) or the rod (rod dystrophy)
characteristic property of their visual function is a delay in dark photoreceptors in the early stage and, at the advanced stage,
adaptation, which can be detected by the psychologically deter­ patients also have remaining rod (cone–rod dystrophy) or cone
mined dark adaptation curve, ERGs and EOGs. It requires 2–3 (rod–cone dystrophy) degeneration. The fundus in patients with
hours to attain the final dark adaptation threshold, the maximum cone or cone–rod dystrophy may be within normal limits or may
scotopic ERG response, and the normal EOG light rise.11,29–31 have subtle changes in the early stage. In such cases, patients
Examples of full-field ERGs after 30 minutes of dark adaptation may be misdiagnosed as having optic nerve disease, central
and after 3 hours of dark adaptation in a typical patient with FA nervous system disease, amblyopia, or OMD (see below). These
are shown in Fig. 8.16. The scotopic (rod) ERG is absent after 30 changes may progress to bull’s-eye maculopathy and diffuse
minutes of dark adaptation but becomes normal after 3 hours of atrophy of the RPE in the far-advanced stage. Patients with rod–
dark adaptation. The mixed rod–cone ERG after 30 minutes of cone dystrophy show abnormal scotopic vision first, followed by
dark adaptation shows a negative configuration with a small abnormal photopic vision. The diseases include retinitis pigmen­
a-wave, just as is seen in Oguchi disease. However, unlike tosa, choroideremia, gyrate atrophy, and others. Full-field ERG
Oguchi disease, it becomes normal after 3 hours of dark adapta­ is important to differentiate between cone–rod and rod–cone
tion. EOG is abnormal when measured in the regular method dystrophy, particularly in their early stage.
of 15 minutes of dark adaptation; however it becomes normal Full-field ERGs in a typical patient with cone dystrophy and
when the dark adaptation is prolonged.31 Mutations in the gene rod–cone dystrophy are shown in Fig. 8.18. Selective abnormali­
encoding 11-cis retinol dehydrogenase (RDH5) cause delayed ties of the photopic components (cone and 30-Hz flicker ERG)
dark adaptation and FA.32 are seen in cone dystrophy and a more severe abnormality in rod
Normal Oguchi Fundus Fundus 211
disease albipunctatus albipunctatus
with
cone dystrophy

Chapter 8
Rod 100 µV

50 ms

Clinical Electrophysiology
Bright 200 µV
flash
25 ms
DA 30 min

DA 3 hours 200 µV

25 ms

100 µV
Cone
25 ms

30-Hz 25 µV
flicker
25 ms

Fig. 8.16 Full-field electroretinograms (ERGs) recorded from normal control, patients with Oguchi disease, fundus albipunctatus (FA), and FA
with cone dystrophy. Bright flash ERGs were recorded after 30 minutes and after 3 hours’ dark adaptation.
In Oguchi disease, full-field ERGs recorded after 30 minutes of dark adaptation show absent rod ERG and essentially normal cone-mediated
ERG. The mixed rod–cone ERG has a negative configuration with relatively well-preserved oscillatory potentials. Because the rod function is
absent, it shows photopic hill phenomenon, and the ERG configuration is negative with a small a-wave. But the negative configuration with
normal a-wave after 3 hours’ dark adaptation may suggest some additional abnormalities of the bipolar cell function.
In FA, the rod ERG is absent after 30 minutes of dark adaptation but becomes normal after 3 hours of dark adaptation. The mixed rod–cone
ERG after 30 minutes of dark adaptation shows a negative configuration with a small a-wave, just as is seen in the photopic phenomenon of
Oguchi disease. However, unlike Oguchi disease, it becomes normal after 3 hours of dark adaptation. About one-third of patients with FA have
associated cone dystrophy, often showing bull’s-eye maculopathy (Fig. 8.17). Such patients show extremely abnormal photopic ERGs in addition
to the characteristic ERG findings of FA. (Revised and reproduced with permission from Kondo M, Piao CH, Tanikawa A, et al. Japanese Journal
of Ophthalmology 2000;44:20–8.)

than cone function is shown in retinitis pigmentosa as a repre­ pathology lies mainly in the rod itself, complete and incomplete
sentative disease of rod–cone dystrophy. At the advanced stage, CSNB are caused by the dysfunction of ON or ON–OFF bipolar
most patients with cone dystrophy also have abnormal scotopic cells, respectively.35,37 They are the model disorders of bipolar
vision (cone–rod dystrophy) and may sometimes be difficult cell dysfunction. EOGs in both diseases are normal.36 It should
to differentiate from rod–cone dystrophy, such as retinitis be noted that all this new information in terms of the classifica­
pigmentosa. tion and pathology of both complete and incomplete CSNB was
obtained from detailed analysis of ERG,36,37 and molecular genet­
Second-order neuron dysfunction ics confirmed these ERG findings later.
The fundamental differences between rod and cone connections The hereditary mode of complete CSNB is X-linked recessive
to the bipolar cells are shown in Fig. 8.19.35 The photoreceptors or autosomal recessive.36 X-linked complete CSNB has a muta­
transmit visual information to the bipolar cells, which are the tion of the leucine-rich repeat proteoglycan (NYX) gene,38 and
second-order neurons. Rods contact only depolarizing (ON) autosomal recessive complete CSNB has a mutation of GRM6
bipolar cells (DBCs), creating ON visual pathways. On the other gene39 encoding the metabotropic glutamate receptor mGluR6
hand, cones have more extensive postsynaptic connections. They and transient receptor potential cation channel subfa­mily
synapse on to depolarizing DBCs and hyperpolarizing OFF member 1 (TRPM1).40 All these proteins are distributed on the
bipolar cells. postsynaptic ON bipolar cells and are required for the depolar­
Complete and incomplete CSNB were classified as indepen­ ization of the cell. The visual functions and ERGs are essentially
dent clinical entities based mainly on the analysis of full-field the same in patients with these three different gene mutations,
ERGs.36 Unlike Oguchi disease and FA, where the responsible which have an almost complete block of ON synaptic
Rod Cone
212
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

_ _ +

Fig. 8.17 Fundus of a patient with fundus albipunctatus associated


with bull’s-eye maculopathy. (Reproduced with permission from Miyake
Y, Shiroyama N, Sugita S, et al. Fundus albipunctatus associated with
cone dystrophy. Br J Ophthalmol 1992;76:375–9, with permission from
BMJ Publishing Group.) ON-pathway ON-pathway OFF-pathway

Fig. 8.19 Simplified schema showing retinal wiring of the rod and cone
pathway .The photoreceptors transmit visual information to the bipolar
cells, which are the second-order neurons. Rods contact only
depolarizing bipolar cells (DBCs: ), creating ON visual pathways. On
the other hand, cones have more extensive postsynaptic connections.
They synapse on to depolarizing DBCs and hyperpolarizing OFF
bipolar cells (HBCs: ).

Rod Cone Bright flash 30-Hz flicker

Normal

Retinitis
pigmentosa

Cone
dystrophy

100 µV 100 µV 200 µV 25 µV


50 ms 25 ms 25 ms 25 ms

Fig. 8.18 Full-field electroretinograms (ERGs) recorded from a normal control (top) and a patient with retinitis pigmentosa at an early stage
(middle) and a patient with cone dystrophy (bottom). The more severe abnormality in rod than cone function is shown in retinitis pigmentosa as
a representative disease of rod–cone dystrophy and selective abnormalities of the photopic components (cone and 30-Hz flicker ERG) are seen
in cone dystrophy. At the advanced stage, most patients with cone dystrophy also have abnormal scotopic vision (cone–rod dystrophy) and may
sometimes be difficult to differentiate from a rod–cone dystrophy, such as retinitis pigmentosa. (Reproduced with permission from Miyake Y.
Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.)
transmission from the photoreceptors to the bipolar cells in both ERG has a plateau-like bottom (Fig. 8.20). In contrast, the cone
rod and cone visual pathways, preserving the OFF pathway and 30-Hz flicker ERGs are extremely reduced in incomplete
intact.11 CSNB, which is highly characteristic and extremely important 213
X-linked incomplete CSNB has a mutation of the calcium for the differential diagnosis.
channel (CACNA1F) gene.41 Loss of the functional channel In spite of the complete defect of ON visual pathway, cone and

Chapter 8
impairs the calcium influx into rods and cones that is needed to 30-Hz flicker ERG of complete CSNB appear nearly normal. This
sustain the tonic release of neurotransmitters from the presyn­ mechanism can be explained by the analysis of photopic long-
aptic terminals. Therefore it is conceivable that patients with flash ERG35,37 as shown in Fig. 8.21. Using photopic ERGs elicited
incomplete CSNB have an incomplete defect of the synapses in by long-duration square-wave stimuli, the cone ON response
the ON and OFF bipolar cells in both rod and cone visual generated by depolarizing ON bipolar cells is selectively and
pathways.11 severely depressed in patients with complete CSNB; moreover,
The comparison of full-field ERGs between complete and the waveform is similar to that of monkeys after 2-amino-4-

Clinical Electrophysiology
incomplete CSNB is shown in Fig. 8.20. The mixed rod–cone phosphonobutyric acid (APB) is injected into the vitreous to
ERG shows a negative configuration with a normal a-wave in block the synapse between photoreceptors and ON bipolar
both types, but OPs can be better recorded in the incomplete cells.35 The OFF response, on the other hand, which is generated
type than the complete type. The normal a-wave with reduced by hyperpolarizing bipolar cells, is intact in patients with com­
b-wave suggests that both types of CSNB have a defect not in plete CSNB, leading us to hypothesize that the ON function of
the rod photoreceptors but in the second-order neurons or their both the rod and cone visual pathway is completely blocked in
synapses in the rod visual pathway. These findings are compa­ eyes with complete CSNB.35,37 The mechanism as to why normal-
rable to molecular genetics.36–41 Rod ERG is absent in the com­ looking brief-flash cone ERG can be obtained under this condi­
plete type but present with subnormal amplitude in the tion is shown in Figure 8.22. With long-duration stimuli, the
incomplete type. Absent rod ERG in complete CSNB and sub­ a-waves, b-waves, and d-waves are clearly separated. As the
normal rod ERG in incomplete CSNB are comparable to the stimulus duration is shortened (brief-flash stimuli), the positive
pathology of complete defect (complete CSNB) and incomplete component of the photopic ERG consists mainly of the d-wave.
defect (incomplete CSNB) of rod bipolar cell transmission. On Therefore even when the b-wave, a component of the ON
the other hand, cone and 30-Hz flicker ERGs appear nearly response, is absent (as in complete CSNB), the d-wave replaces
normal in the complete type except that the a-wave of the cone the b-wave, and a positive wave is recorded with brief-flash

Normal Complete Incomplete XLRS


CSNB CSNB

Bright flash
200 µV
25 ms

100 µV
Rod
50 ms

Cone 100 µV
25 ms

30-Hz
flicker 25 µV
25 ms

Fig. 8.20 Full-field electroretinograms (ERGs) recorded from a normal control, patient with complete or incomplete congenital stationary night
blindness (CSNB), and patient with X-linked retinoschisis (XLRS). In CSNB, the mixed rod–cone ERG shows negative configuration with a
normal a-wave in both types, suggesting a defect not in the rod photoreceptors but in the second-order neurons or their synapses in the rod
visual pathway. However, oscillatory potentials can be recorded better in the incomplete type than the complete type. Rod ERG is absent in the
complete type but present with subnormal amplitude in the incomplete type. On the other hand, cone and 30-Hz flicker ERGs appear nearly
normal in the complete type except that the a-wave of the cone ERG has a plateau-like bottom. In contrast, the cone and 30-Hz flicker ERGs are
extremely reduced in incomplete CSNB, which is highly characteristic and extremely important for the differential diagnosis. In XLRS, the mixed
rod–cone ERG shows a negative configuration, which is observed even when the retinoschisis is confined to the fovea ophthalmoscopically. The
full-field ERG findings similar to those of incomplete CSNB suggest that both ON and OFF bipolar cell function is mainly impaired in the rod and
cone visual pathways. (Reproduced with permission from Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.)
Monkey Human Normal Complete CSNB
(sieving PA, 1993)
214 d
d b d
b b
a a
d d
Normal
Section 2

d
a
a
d
d

APB Complete
d d
CSNB
Retinal Diagnostics
Retinal Imaging and Diagnostics

25 µV Incomplete
CSNB
ON OFF ON OFF 25 µV
300 ms 125 ms

Fig. 8.21 Comparison of photopic long-duration electroretinograms


100 µV
(ERGs) recorded from a monkey and a human. Left, Normal control
50 msec
ERG for the monkey eye and after being treated by 2-amino-4-
phosphonobutyric acid (APB). Right, ERGs recorded from a normal
human control, from a patient with complete congenital stationary night
blindness (CSNB), and from a patient with incomplete CSNB. The Fig. 8.22 Photopic electroretinograms (ERGs) elicited by square-wave
cone ON response generated by depolarizing ON bipolar cells is stimuli of various durations from a normal control and a patient with
selectively depressed whereas the OFF response, which is generated complete congenital stationary night blindness (CSNB), explaining why
by hyperpolarizing bipolar cells, is intact in patients with complete complete CSNB shows normal-looking brief-flash cone ERG. With
CSNB; moreover, the waveform is similar to that of monkeys treated long-duration stimuli, the a-waves, b-waves, and d-waves are clearly
with APB. (Reproduced with permission from Kondo M, Piao CH, separated. As the stimulus duration is shortened (brief-flash stimuli),
Tanikawa A, et al. Japanese Journal of Ophthalmology 2000;44:20–8, the positive component of the photopic ERG consists mainly of the
with permission.) d-wave. Therefore even when the b-wave, a component of the ON
response, is absent (as in complete CSNB), the d-wave replaces the
b-wave, and a positive wave is recorded with brief-flash stimuli. Thick
lines underneath the responses represent the stimulus duration.
stimuli.11 With incomplete CSNB, on the other hand, the ON and (Reproduced with permission from Miyake Y. Nippon Ganka Gakkai
OFF response are both subnormal, suggesting that the ON and Zasshi 2002;106:737–56.)
OFF systems are incompletely disturbed at the level of the
bipolar cells.11,37
XLRS is a vitreoretinal dystrophy that manifests early in life. protein is heavily concentrated in the inner segments of both
XLRS is one of the more common causes of juvenile macular rods and cones, electroretinographic studies suggest that it does
degeneration in males. Intraretinal cysts form in the macula and not inherently affect the photoreceptor function of either cell
splitting of the retinal layers occurs in the peripheral retina (see type. And from the above ERG and EOG results, it currently is
Chapter 41, Hereditary vitreoretinal degenerations). Most reasonable to propose that both ON and OFF pathways are
patients, including young ones, show moderately poor visual defective, although precise subcellular localization has not yet
acuity that gradually decreases with increasing age. Hyperme­ determined whether both depolarizing and hyperpolarizing
tropia has been shown to be a frequent accompaniment of this bipolar cells are involved.44
disorder.42 In fact, many patients with XLRS are first diagnosed
with hypermetropic amblyopia or heterotropia during infancy.
As mentioned above, mixed rod–cone ERG is of significant
FOCAL ERG
diagnostic value because of the negative configuration. The neg­ In order to record the ERG responses with focal stimuli, there
ative ERG is observed even when the retinoschisis is confined to are two methods which have been reported in the past. The
the fovea ophthalmoscopically. The full-field ERG findings conventional focal macular ERG can show a similar waveform
(Fig. 8.20) are similar to those of incomplete CSNB, suggesting as a conventional photopic ERG from the limited area of
that both ON and OFF bipolar cell function is mainly impaired macula.45 The advantage of this method is that the analysis of
in the rod and cone visual pathways. EOG in XLRS is normal. ERG components can be done using the same concept as that
The schisis occurs in the plane of the nerve fiber and ganglion of full-field photopic ERG. By analyzing several components,
cell layers of the retina. It has long been suggested that degen­ layer-by-layer macular function can be evaluated.
erating Müller cells or inner retinal cells may be the primary Another method is the mfERG technique, which was deve­
cause of the pathological changes in XLRS. The XLRS gene was loped in 1992.46 With this method, focal ERGs can be recorded
cloned in 1997 and was designated RS1.43 However, the RS1 simultaneously from multiple retinal locations during a single
protein is heavily expressed in inner segments of both rod and recording session using cross-correlation techniques. Unlike
cone photoreceptors and is also seen in cells of the inner nuclear conventional focal macular ERGs, there are still questions about
layer. There is some discrepancy between the results of full-field how this method works and what it measures because the
ERG and genetic findings. Although the expression of RS1 technique is relatively new. We have an impression that the
layer-by-layer analysis of the macula can be done more precisely sensitivity of the area surrounding the stimulus. By combining
using focal macular ERG than with mfERG. the focal stimulus with background illumination properly, focal
responses can be recorded. It is also essential to monitor the 215
Principle, method, and characteristics location of the stimulus on the fundus during the recordings,
The focal macular ERG is primarily used to evaluate macular particularly in eyes with a central scotoma, to be certain that only
function. The full-field ERG is unable to detect small focal lesions

Chapter 8
the fovea is stimulated. An example of a recording system of
or pathogenesis in the retina, and is normal in the presence of focal macular ERG is shown in Fig. 8.23. The examiner records
macular diseases. In contrast, the full-field ERG may be unde­ the ERGs while monitoring the fundus by the infrared television
tectable when only macular function is preserved, such as in fundus camera. The optical system of adequate combination of
patients with retinitis pigmentosa.11 stimulus light and background illumination for focal stimulus is
The principle of recording of focal macular ERG includes pre­ installed in the fundus camera, and the focal macular ERGs can
senting a small stimulus to the macula and recording the be recorded under the fundus monitor by summating the

Clinical Electrophysiology
response from the stimulated area by summating the responses responses with a computer. Focal macular ERGs recorded from
using computer. To eliminate contaminating stray light a normal subject demonstrating the various components are
responses, background illumination must be used to depress the shown in Fig. 8.24. All components of photopic ERG can be

Fig. 8.23 Overall view of the observation


and stimulation systems for focal macular
electroretinogram (ERG) and visually evoked
response (VER) recordings. The examiner
records the ERGs while monitoring the
stimulus on the fundus by the infrared
television fundus camera (A). A plastic
hemisphere with miniature lamps is attached
to the top of the camera to obtain
background illumination for the peripheral
retina (B). A Burian–Allen bipolar contact lens
is used to record the ERGs (C). (Revised
with permission from Miyake Y, Yanagida K,
Kondo T, et al. Nippon Ganka Gakkai Zasshi
1981;85:1521–33.)

B C
The visual acuity of eye with CME was 0.6. Six months later,
OFF the CME resolved spontaneously, and fluorescein angiogram
216 disclosed a normal pattern; the visual acuity improved to 1.2.
1 Hz
The focal macular ERGs returned to normal levels, with the
amplitude of the OPs comparable to those from the normal
ON OFF 1 µV
fellow eye.51
Section 2

200 ms
Occult macular dystrophy (OMD) is one of the most represen­
tative disorders where the focal macular ERG or multifocal ERG
T.C. T.C.
0.03 sec 0.003 sec is a key for diagnosis. OMD was discovered by focal macular
ERG in 1989.52 The clinical findings of OMD are progressive
decrease of visual acuity, normal fundus and fluorescein angio­
grams, normal full-field ERGs, but abnormal focal macular ERG
Retinal Diagnostics
Retinal Imaging and Diagnostics

5 Hz
and multifocal ERG (Fig. 8.27). Although the fundus appearance
and fluorescein angiogram show normal findings in OMD,
optical coherence tomography (OCT) may reveal some mild
ON ON abnormality from its early stage. Whether focal macular ERG or
OCT is more sensitive to detect early abnormality is an interest­
ing point of argument. The point is that OMD may not be a rare
30 Hz disease and that many patients with OMD may be misdiagnosed
1 µV as having several other diseases, such as a psychological eye
ON OFF 50 ms problem, optic nerve problem, central nervous system problem,
or amblyopia.53
Fig. 8.24 Components of the focal macular electroretinogram recorded The hereditary mode of OMD is autosomal dominant, although
from a normal subject. ON and OFF responses recorded with 1-Hz some patients show sporadic mode.52,53 Our genetic studies have
stimulus frequency (top); a-wave, b-wave, and oscillatory potentials detected mutations in retinitis pigmentosa 1-like 1 (RP1L1) gene
recorded with 5-Hz stimulus frequency (middle); and 30-Hz flicker in autosomal dominant OMD.54 It was demonstrated that RP1L1
responses (bottom) are shown. Arrows indicate photopic negative
response. (Reproduced with permission from Miyake Y. plays an essential role in cone function in humans and that dis­
Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.) ruption of RP1L1 function leads to OMD.
Acute zonal occult outer retinopathy is characterized by an
acute zonal loss of one or more large zones of central retinal
function in one or both eyes, predominantly in young women.55
recorded: they are a-waves, b-waves, OPs, PhNR, ON and OFF Other ocular findings include initially minimum ophthalmo­
components, and 30-Hz flicker responses.11 scopic changes, photophobia, and permanent visual field loss,
Several important characteristics of focal macular ERG in often associated with late development of retinal pigmentary
human were detected, particularly in macular OPs.45 An changes and narrowing of the retinal vessels in the affected
example is shown in Fig. 8.25, demonstrating nasotemporal zones (see Chapter 76, White-spot syndromes and related
asymmetry.47 Semicircular stimuli were used to compare the diseases). Since the full-field ERG may be normal or only
ERGs elicited by stimulating the temporal and nasal macula. slightly abnormal, it is not informative. The responses of
The amplitudes and implicit times of the a-waves and b-waves mfERG become abnormal in the limited area where visual
in the nasal retina are almost identical to those from the tem­ field loss is present11 (Fig. 8.28).
poral retina, whereas the amplitudes of OPs are much larger
than in the temporal retina than those in the nasal retina. The OTHER SPECIAL RESPONSES OR
amplitude of the focal ERGs recorded with circular stimulus is TECHNIQUES IN ERG
approximately the same sum as the amplitudes of the temporal
and nasal ERGs. Pattern ERG
The principle, recording method, and clinical applications of Pattern ERG (PERG) is the retinal response to a structured stimu­
mfERG are described in an ISCEV guideline.48 Readers are also lus, such as a reversing black-and-white checkerboard or grating.
referred to Chapter 7, Electrogenesis of the ERG, where the The standard for PERG recording has been established by the
origin of the mfERG is described in detail. ISCEV.56 While the luminance ERG is evoked by changes in
stimulus luminance, the PERG is evoked by changes of stimulus
Clinical applications contrast. The PERG is therefore a retinal response that is absent
The examples of value of focal ERG are shown here. In focal of a net change of stimulus luminance. The generators of the
macular ERG, the macular OPs are the most sensitive indicator responses are mainly the retinal ganglion cells.57 Thus ganglion
in variable macular diseases. The selective reduction of macular cell function can be evaluated by PERG.
OP amplitude is observed in the early stage of macular edema,49 The waveform consists of an initial cornea-positive response,
epimacular membrane,50 and convalescent stage of central referred to as P50, followed by a cornea-negative response,
serous chorioretinopathy.51 The fluorescein angiograms and referred to as N95 (Fig. 8.29). Holder58 reported in his review on
focal macular ERGs in an eye with pseudophakic cystoid PERG in 520 eyes that P50 was reduced with preganglion cell
macular edema (CME) and after resolution of CME are illus­ dysfunction but usually spared with optic nerve diseases. On the
trated in Fig. 8.26. The OPs of the focal macular ERGs are other hand, N95 loss was the most common abnormality and
selectively reduced compared with that of a normal fellow eye. was reliably associated with optic nerve diseases.
Fig. 8.25 Comparison of focal
electroretinograms using semicircular stimuli
with the edge of the semicircle passing
through the vertical axis (top) on the nasal 217
and temporal macular areas and a circular
stimulus (15°). The oscillatory potentials in
the temporal macula are significantly larger

Chapter 8
than those in the nasal macula, and only the
oscillatory potentials show this significant
asymmetry. (Reproduced with permission
from Miyake Y. Electrodiagnosis of retinal
diseases. Tokyo: Springer-Verlag; 2006, and
Miyake Y, Shiroyama N, Hiroguchi M, et al.
Invest Ophthalmol Vis Sci 1989;30:1743–9.)

Clinical Electrophysiology
T.C. T.C.
0.03 sec 0.003 sec

Temporal

Nasal

Circular
stimulus O2

O1 O3
O4

a
1 µV
ON ON 50 msec
Fig. 8.26 Focal macular electroretinograms
(ERGs) (left) and fluorescein angiograms
(right) in a 51-year-old man with
218 Focal macular ERG pseudophakic cystoid macular edema (CME:
top) and after the resolution of CME (bottom).
The oscillatory potentials of the focal macular
Affected eye Fellow eye ERGs are selectively reduced compared with
Section 2

that of a normal fellow eye. The visual acuity


of the eye with CME was 0.6 (20/30). After
spontaneous resolution of the CME, the focal
macular ERGs returned to normal levels, with
the amplitude of the oscillatory potentials
T.C. comparable to those from the normal fellow
0.03 sec eye. The visual acuity improved to 1.2.
0.003 sec (Reproduced with permission from Miyake Y,
Retinal Diagnostics
Retinal Imaging and Diagnostics

Miyake K, Shiroyama N, et al. Am J


Ophthalmol 1993;116:576–83)

1 µV
50 msec
Normal Patient
219
Full-field ERG
100 µV

Chapter 8
Rod 50 msec

200 µV
Cone 25 msec

30Hz 100 µV

Clinical Electrophysiology
Normal Patient
flicker 50 msec

Bright 25 µV
flash 25 msec

Multifocal ERG Focal macular ERG

10°
1 µV 1 µV
100 ms 100 ms

15°

1 µV

25 ms ON ON

Fig. 8.27 The clinical findings of occult macular dystrophy. Left top, normal fundus and fluorescein angiograms; left middle, optical coherence
tomography images from a normal person and a patient with occult macular dystrophy. The horizontal section through the foveal center
demonstrates an abnormality, especially in the outer retinal bands at the fovea in the patient. Left bottom, wave three-dimensional topography
and multifocal electroretinogram (ERG) trace array from a normal person and a patient, showing markedly reduced response density in the
central 7° of the retina. Right top, Full-field ERGs showing normal responses; right bottom, focal macular ERG showing abnormal responses in
the patient.
Multifocal ERG Response arrays Static visual field (Humphey 30-2) Deviation plot
220 Right Left Right Left
Section 2

1 µV
500 nV
100 ms 100 ms

3 dimensional plots
Right Left < 5%
< 2%
< 1%
Retinal Diagnostics
Retinal Imaging and Diagnostics

< 0.5%

OCT Right Left

s
nV/deg2 nV/deg2 s N
1
T
1
0 10 20 0 10 20 T N I

Fig. 8.28 Clinical findings of a patient with acute zonal occult outer retinopathy. A 22-year-old female developed acute central visual loss in
the right eye. Visual acuity was 0.5 (20/40) OD and 1.2 (25/20) OS. The top and bottom left panels are 61 response arrays and three-
dimensional plots of the multifocal electroretinograms, respectively, showing abnormal responses in the limited area, where visual field loss is
present in the right eye. The top right panels are the deviation plot of the static visual field, showing reduced sensitivity in the central area in the
right eye. The bottom right panels are Fourier-domain optical coherence tomographic (FD-OCT) images from the affected right eye and intact left
eye, respectively, showing that both the border of the photoreceptor inner- and outer-segment line (IS/OS line) and the cone outer-segment tip
(COST) line between the IS/OS line and the retinal pigment epithelium (RPE) are absent in the macular area of the right eye. The IS/OS line,
COST line, and RPE/Bruch membrane are intact in the left eye.

ERG (Fig. 8.24) and mfERG has enabled objective assessment of


4 retinal ganglion cell damage in glaucoma, optic nerve disease,
and retinal vascular diseases.59,60 Other studies showed that
P50 PhNR can be correlated with retinal sensitivity as well as retinal
2 microstructure such as nerve fiber thickness.61
Amplitude (µV)

ERG recordings by LED


0 Light-emitting diodes (LEDs) (Fig. 8.30) have been in the lime­
light recently as light sources to elicit and record full-field ERG.62
N35 The LEDs are small and inexpensive, and they require low cur­
–2 rents to drive them. They can be controlled by a simple electronic
N95 circuit to give either a continuous light output or extremely brief
flashes over a large range of intensities. The stimulus and record­
ing system using LED can provide not only routine full-field
–4
ERG but also some other special usages for clinical ERG, as
0 50 100 shown below.
Time (msec) ERG recording under general anesthesia11
ERG recordings with LED are useful for recording standard
Fig. 8.29 Pattern electroretinogram responses recorded from a normal ERGs from pediatric patients under general anesthesia. The
control (gray), patient with macular dysfunction (green), and patient equipment needed to obtain recordings that correspond to
with optic nerve dysfunction (blue). ISCEV standard ERGs is compact and easily portable.63 The
ERGs recorded using an LED system on a 3-month-old baby
under general anesthesia and a normal adult are compared in
Fig. 8.31.
Photopic negative response
PhNR has already been described and is a negative wave in ERG monitoring during eye surgery
photopic ERG that is evoked following the b-wave (Figs 8.14 and As vitreoretinal surgery continues to advance, close monitoring
8.24). The origin of the PhNR is the retinal ganglion cells,21 and of retinal function during these procedures has become impor­
this is fully described in Chapter 7, Electrogenesis of the ERG. tant. Although ERGs directly reflect retinal function, monitoring
Significant reduction in PhNR was reported in patients with during surgery has proven difficult. Each recording must be
open angle glaucoma and several other optic neuropathies.21 made quickly under aseptic conditions, and the instruments and
Moreover, recent analysis of PhNR recorded in focal macular electrodes must be such that they do not cause interference for
221

Chapter 8
1.0
Relative luminous intensity

B
0.8

Clinical Electrophysiology
0.6

Diffuser lens LED cable


0.4
Three white LEDs

0.2

400 500 600 700 800


Wavelength (nm)
A Electrode (gold)
Electrode cable
C Contact lens (PMMA)

Fig. 8.30 Structure of the white light-emitting diode (LED) contact lens electrode. (A) Relative spectral emission of the LED. (B) Output that
appears as visible white. (C) Structure of the contact lens electrode with three built-in white LEDs. PMMA, polymethylmethacrylate. (Reproduced
with permission from Kondo M, Piao CH, Tanikawa A, et al. Doc Ophthalmol 2001;102:1–9.)

Full–field ERG from normal subjects

Adult 3-month-old baby

200 µV
Rod
50 ms

200 µV
Bright
flash 20 ms

Cone 100 µV
20 ms

30-Hz
flicker 100 µV
20 ms

Fig. 8.31 Full-field electroretinograms (ERGs) recorded with white light-emitting diode (LED) contact lens electrodes from a normal adult subject
(left) and a normal 3-month-old baby (right). The pictures show standard full-field ERG recording from a baby using this system. Top, LED contact
lenses are placed in both eyes under general anesthesia. Bottom, Background illumination from the contact lens is used during the recording of
the photopic ERGs in the dark. (Reproduced with permission from Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006.)
ERG monitor during vitrectomy
222
Start
Section 2

Infusion

Vitrectomy

Fluid–air
Retinal Diagnostics
Retinal Imaging and Diagnostics

exchange

5 days
after
surgery 100 µV
on 50 ms

Fig. 8.32 Full-field electroretinogram (ERG) recording during vitrectomy. Left, a light-emitting diode (LED) electrode is sterilized and placed on the
cornea undergoing surgery. Right, 30-Hz flicker ERGs recorded during vitrectomy in a patient with epimacular membrane. Start indicates the time
when local anesthesia was completed and Infusion denotes the time the infusion needle was introduced into the vitreous cavity. (Reproduced with
permission from Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag; 2006, and Horiguchi M, Miyake Y. Arch Ophthalmol
1991;109:1127–9.)

the retinal surgeon. Furthermore, the eye undergoing surgery is The components that reflect the “OFF” visual system (a-waves
intensively light-adapted. and d-waves) are essentially absent in S-cone ERGs because,
The LED contact lens electrode has been found to be highly unlike the LM cone system, the S-cone is mainly connected to
suitable for this purpose.64 It is easily sterilized and is used as the “ON” visual system.67
both a stimulus source and a recording electrode for 30-Hz
flicker ERGs during vitreoretinal surgery (Fig. 8.32). An example ELECTRO-OCULOGRAM
of ERG monitor during vitrectomy from a patient with epimacu­
In 1849, Du Bois Reymond68 reported that in the normal eye
lar membrane is shown in Fig. 8.32. The ERGs recorded after
there is a flow of electrical current, because the cornea is posi­
local anesthesia (start), and after the introduction of the infusion
tive with respect to the back of the eye. The source of the voltage
needle into the vitreous cavity (infusion) were not significantly
is the corneofundal potential. This potential difference is
different in regard to amplitude and peak time. However, after
referred to as the standing potential or resting potential of the
vitrectomy, which required 10 minutes, the peak time was
eye. The EOG is an indirect measure of the amplitude of the
delayed and the amplitude decreased (vitrectomy). Additional
standing potential, which changes during dark and light adapta­
studies have demonstrated that lowering the intravitreal tem­
tion. To obtain an EOG in humans, electrodes are placed at the
perature by applying an infusion solution kept at room tempera­
inner and outer canthi of the eyes and the patient is asked to
ture can alter the ERG during vitrectomy. Filling the whole
look back and forth between a pair of fixation lights (Fig. 8.34).
vitreous cavity with air after the epimacular membrane was
When the cornea moves closer to one of the electrodes, it
peeled off resulted in a markedly reduced amplitude and
becomes more positive and the other electrode becomes more
delayed peak time (fluid–air exchange). This extreme reduction
negative. The opposite happens when the eyes move to the
of ERG following fluid–air or fluid–silicon oil exchange in the
other side.
vitreous cavity results from reduced electrical conductivity in
The principle and practical use of EOG are described in the
the vitreous cavity. Five days after surgery, when the air was
ISCEV standard.69 The changes in the amplitude of the EOG in
resolved from the vitreous cavity, the ERG recovered to the
the dark-adapted and light-adapted state of a normal subject are
postoperative amplitude and peak time.
shown in Fig. 8.35. The smaller amplitudes are recorded when
S-Cone ERG the eyes make the saccadic eye movements in the dark (dark
Recording short-wavelength cone (S-cone) ERGs is valuable trough); the peak amplitude is recorded against a steady light
clinically because it allows us to evaluate the S-cone visual background (light peak). The light peak/dark trough (L/D) ratio
system. S-cone ERGs have been recorded using stimulation with is an index (Arden index)70 used to assess retinal function. Gen­
strong blue stimuli on a bright yellow background, which sup­ erally, a ratio of 1.80 is the lower limit of normal.
presses the middle- and long-wavelength (LM) cone system.65 The origin of the retinal standing potential is thought to be in
LEDs emitting blue light can be used in the LED built-in contact the retinal pigment epithelium. However, the light rise is gener­
lens electrode66 (Fig. 8.33). By using bright yellow background ated by light stimulation of the photoreceptor–retinal pigment
illumination, the S-cone ERGs are recordable and are compared epithelium complex; and it is not detected when certain struc­
with LM cone ERGs (Fig. 8.33). The amplitude is much smaller tures of the middle retinal layer is affected, such as in central
and the implicit time is longer than those of the LM cone ERG. retinal arterial occlusion.71
Fig. 8.33 S-cone electroretinogram (ERG) recording with blue-emitting
light-emitting diode (LED) built-in contact lens electrode. Top, LED
built-in contact lens electrode with blue-emitting LEDs. Bottom,
comparison of long-wavelength (LM) cone and S-cone ERGs with 223
long-duration stimuli in a normal subject. The a-wave and d-wave that
reflect the “OFF” visual system are essentially absent in the S-cone
ERGs, because, unlike the LM cone system, the S-cone is mainly

Chapter 8
connected to the “ON” visual system. (Reproduced with permission from
Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer-Verlag;
2006, and Horiguchi M, Miyake Y, Kondo M, et al. Invest Ophthalmol
Vis Sci 1995;36:1730–2.)

Clinical Electrophysiology
LM-cone 20 µV

50 ms

S-cone

5 µV

50 ms
ON OFF

flash VEP pattern VEP pattern onset/offset VEP

P2 P3 P100 C3
C1

P1

N1 N2
N75 C2
N135
N3

Fig. 8.34 Three standard responses in a clinical visual evoked potential (VEP) test. Top, VEP stimulation. Left, flash stimulus is delivered with
monitor or full-field dome. Such a dome is used for recording full-field electroretinogram as well and a pair of fixation lights in the dome is used
for an electro-oculogram test. Middle, reversing black-and-white checkerboard stimulus provided using cathode ray tube or light-emitting diode
(LED) monitor. Right, reversing black-and-white checkerboard stimulus with diffuse blank screen at regular intervals is presented.
Fig. 8.35 Electro-oculogram (EOG) recordings. Top, diagram
Dark Light adaptation illustrating the EOG test and the determination of the light peak/dark
trough (L/D) ratio (Arden ratio), which is 2.0 in this case. The smaller
amplitudes are recorded when the eyes make saccadic eye
224 movements in the dark (dark trough); the peak amplitude is recorded
against a steady light background (light peak). Bottom, plottings of the
amplitude of EOG in a normal subject and a patient with Best disease.
The Arden ratio is 2.0 for the normal subject and 1.3 for the patient.
Section 2

Dark Light
trough peak

Dark Light
Retinal Diagnostics
Retinal Imaging and Diagnostics

Relative amplitude

Normal

Abnormal

0 15 30
Time (minutes)

Normal

VEP ERG

9.0
8.0
7.0
6.0

5.0 Optic atrophy


Log neutral density filter

4.0 VEP ERG

3.0
3.0

2.0 2.0

1.0
1.0

0.0 0.0 5 µV
100 msec

Fig. 8.36 Simultaneous recording of flash visual evoked potential (VEP) and full-field electroretinogram (ERG) with different stimulus intensities
in a normal subject and in a patient with optic atrophy. For the recording, a silver disc electrode was placed on the scalp over the visual cortex
at Oz according to the International 10/20 system74 for VEP and a Beckman’s electrode was attached to the center of the inferior eyelid. In a
normal subject, stimulus threshold for VEP is much lower than that for ERG. In contrast, the stimulus threshold for VEP is higher than that for
ERG in a patient with optic atrophy. This can be used to differentiate whether retina or optic nerve is the pathological site in eyes with opaque
media. The arrow indicates stimulus onset.
The EOG is generally abnormal in any condition in which 10. Yonemura D, Aoki T, Tsuzuki K. Electroretinogram in diabetic retinopathy.
Arch Ophthalmol 1962;68:19–24.
the flash ERG is abnormal, except complete or incomplete 11. Miyake Y. Electrodiagnosis of retinal diseases. Tokyo: Springer; 2006.
CSNB.36 The reverse, however, is not true. An abnormal EOG 12. Karpe G, Uchermann A. The clinical electroretinogram. VII. The electroretino­ 225
gram in circulatory disturbances of the retina. Acta Ophthalmol 1955;33:
with a normal ERG may be seen in Best disease, pattern dys­ 493–516.
trophy of the RPE, and dominant drusen. Detailed clinical 13. Sabates R, Hirose T, McMeel JW. Electroretinography in the prognosis and
classification of central retinal vein occlusion. Arch Ophthalmol 1983;101:

Chapter 8
values of EOG in each disease are described in Chapter 42,
232–5.
Macular dystrophies. 14. Hiraiwa T, Horio N, Terasaki H, et al. Preoperative electroretinogram and
postoperative visual outcome in patients with diabetic vitreous hemorrhage.
VISUAL EVOKED POTENTIAL Jpn J Ophthalmol 2003;47:307–11.
15. Horio N, Terasaki H, Yamamoto E, et al. Electroretinogram in the diagnosis of
The visual evoked potential (VEP) is the signal of the brain endophthalmitis after intraocular lens implantation. Am J Ophthalmol 2001;
132:258–9.
evoked by the visual stimulus. It is recorded, like the electro­ 16. Miyake Y, Yagasaki K, Horiguchi M, et al. Congenital stationary night blind­
ness with negative electroretinogram: a new classification. Arch Ophthalmol

Clinical Electrophysiology
encephalogram (EEG), at the scalp in the occipital region by
1986;104:1013–20.
surface electrodes. The principle and practical use of clinical VEP 17. Horiguchi M, Miyake Y. Batten disease: deteriorating course of ocular findings.
are shown in the ISCEV standard.72 Jpn J Ophthalmol 1992;36:91–6.
18. Berson EL, Lessell S. Paraneoplastic night blindness with malignant melanoma.
The cellular sources of the VEP are poorly understood. VEPs Am J Ophthalmol 1988;106:307–11.
can be recorded under either transient or steady-state stimulus 19. Heckenlively JR, Aptsiauri N, Holder GE. Autoimmune retinopathy, CAR and
MAR syndromes. In: Heckenlively JR, Arden GB, editors. Principles and prac­
conditions. The transient VEP includes flash VEP, pattern rever­ tice of clinical electrophysiology of vision. 2nd edn. Cambridge, MA: MIT
sal VEP, and pattern onset/offset VEP (Fig. 8.34). Press; 2006. p. 691–8.
Several important features of the VEP can be stated: 20. Hirose T, Katsumi O, Pruett RC, et al. Retinal function in birdshot retinocho­
roidopathy. Acta Ophthalmol 1991;69:327–37.
21. Viswanathan S, Frishiman LJ, Robson JG, et al. The photopic negative response
1. The VEP is dominated by the central 20° of retina owing to of the macaque electroretinogram: reduction by experimental glaucoma. Invest
Ophthalmol Vis Sci 1999;40:1124–36.
“cortical magnification.” 22. Kohl S, Baumann B, Broghammer M, et al. Mutatons in the CNGB3 gene encod­
2. The VEP is highly variable among individuals, but varies ing the beta-subunit of the cone photoreceptor cGMP-gated channel are
responsible for achromatopsia (ACHM3) linked to chromosome 8q21.
less than 10% bilaterally when responses from the two eyes Hum Mol Genet 2000;9:2107–16.
of single person are compared. 23. Oguchi C. Über die eigenartige Hemeralopie mit diffuser weissgr ä ulicher
3. VEP abnormality can result from abnormality in the retina, Verfärbung des Augenhintergrundes. Graefes Arch Ophthalmol 1912;81:
109–17.
optic nerve, optic tracts, optic radiations, or visual cortex. 24. Mizuo A. On a new discovery in the dark adaptation in Oguchi’s disease.
Acta Soc Ophthalmol Jpn 1913;17:1854–9.
25. Miyake Y, Horiguchi M, Suzuki S, et al. Electrophysiological findings in
Flash VEP is easy to record, so is performed for evaluation of patients with Oguchi’s disease. Jpn J Ophthalmol 1996;40:511–9.
gross visual function in infants, small children, or in persons 26. Fucks S, Nakazawa M, Maw M et al. A homozygous 1-base pair deletion in the
arrestin gene is a frequent cause of Oguchi disease in Japanese. Nat Genet
who are unable to perform PERG because of poor fixation due 1995;10:360–2.
to poor visual function, nystagmus, or incorporation. Since flash 27. Yamamoto S, Sipple KC, Berson EL, et al. Defects in the rhodopsin kinase gene
in the Oguchi form of stationary night blindness. Nat Genet 1997;15:175–8.
VEP may still be recordable in patients with undetectable ERG, 28. Carr RE, Gouras P. Oguchi’s disease. Arch Ophthalmol 1965;73:646–56.
such as retinitis pigmentosa or dense vitreous hemorrhage, 29. Carr RE. Congenital stationary night blindness. Trans Am Ophthalmol Soc
simultaneous recordings of flash VEP and ERG may provide 1974;72:448–87.
30. Carr RE, Ripps H, Siegel IM, et al. Rhodopsin and the electrical activity of the
useful information73 (Fig. 8.36). retina in congenital night blindness. Invest Ophthalmol 1966;5:497–507.
Pattern VEP is elicited by a checkerboard pattern of alternating 31. Yamamoto H, Simon A, Eriksson U, et al. Mutations in the gene encoding 11-cis
retinol dehydrogenase cause delayed dark adaptation and fundus albipuncta­
black-and-white checks that reverse in a regular phase fre­ tus. Nat Genet 1999;22:188–91.
quency. It is a robust response and relatively consistent among 32. Miyake Y, Watanabe I, Asano T, et al. Further studies on EOG in retinitis
punctata albescens: effects of change of dark adaptation time on EOG.
individuals and in most cases is the clinical study of choice. Folia Ophthalmol Jpn 1974;25:518–27.
Clinically, pattern VEP is a test of choice mainly for patients with 33. Miyake Y, Shiroyama N, Sugita S, et al. Fundus albipunctatus associated with
optic nerve disease or more distal lesion and nonorganic disease, cone dystrophy. Br J Ophthalmol 1992;76:375–9.
34. Nakamura M, Hotta Y, Tanikawa A, et al. A high association with cone
such as malingering or hysteria. It is also useful for the estima­ dystrophy in fundus albipunctatus caused by mutations of the RDH5 gene.
tion of visual acuity and monitoring developmental changes in Invest Ophthalmol Vis Sci 2000;41:3925–32.
35. Sieving PA. Photopic ON- and OFF-pathway abnormalities in retinal dytro­
small children. phies. Trans Am Ophthalmol Soc 1993;91:701–73.
36. Miyake Y, Yagasaki K, Horiguchi M, et al. Congenital stationary night blind­
REFERENCES ness with negative electroretinogram. A new classification. Arch Ophthalmol
1. Marmor MF, Fulton AB, Holder GE, et al. For the International Society for 1986;104:1013–20.
Clinical Electrophysiology of Vision. ISCEV Standard for full-field clinical 37. Miyake Y, Yagasaki K, Horiguchi M, et al. On- and off-responses in photopic
electroretinography (2008 update). Doc Ophthalmol 2009;118:69–77. electroretinogram in complete and incomplete types of congenital stationary
2. Gjötterberg M. Electrodes for electroretinography: A comparison of four night blindness. Jpn J Ophthalmol 1987;31:81–7.
different types. Arch Ophthalmol 1986;104:569–70. 38. Bech-Hansen NT, Naylor MJ, Maybaum TA, et al. Mutation in NYX, encoding
3. Sieving PA, Frishman LJ, Steinberg RH. Scotopic threshold response of the leucine-rich proteoglycan nyctalopin, cause X-linked complete congenital
proximal retina in cat. J Neurophysiol 1986;56:1049–61. stationary night blindness. Nat Genet 2000;26:319–23.
4. Brown KT. The electroretinogram: its components and their origins. Vision Res 39. Dryja TP, McGee TL, Berson EL, et al. Night blindness and abnormal cone
1968;8:633–77. electroretinogram ON responses in patients with mutations in the GRM6 gene
5. Newman EA, Odette LL. Model of electroretinogram b-wave generation: a test encoding mGluR6. Proc Natl Acad Sci USA 2005;102:4884–9.
of the K+ hypothesis. J Neurophysiol. 1984;51:164–82. 40. Audo I, Kohl S, Leroy BP, et al. TRPM1 is mutated in patients with autosomal-
6. Cobb WA, Morton HB. A new component of the human electroretinogram. recessive complete congenital stationary night blindness. Am J Hum Genet
J Physiol 1954;123:36–7. 2009;85:720–9.
7. Wachtmeister L, Dowling JE. The oscillatory potentials of the mudpuppy 41. Strom TM, Nyakatura G, Apfelstedt-Sylla E, et al. An L-type calcium-channel
retina. Invest Ophthalmol Vis Sci 1978;17:1176–88. gene mutated in incomplete X-linked congenital stationary night blindness.
8. Kondo M, Piao CH, Tanikawa A, et al. Amplitude decrease of photopic ERG Nat Genet 1998;19:260–3.
b-wave at higher stimulus intensities in humans. Jpn J Ophthalmol 2000;44: 42. Kato K, Miyake Y, Kachi S, et al. Axial length and refractive error in X-linked
20–8. retinoschisis. Am J Ophthalmol 2001;131:812–4.
9. Peachey NS, Alexander KR, Fishman GA, et al. Properties of the human cone 43. Sauer CG, Gehrig A, Warneke-Wittstock R, et al. Positional cloning of the
system electroretinogram during light adaptation. Appl Optics 1989;28: gene associated with X-linked juvenile retinoschisis. Nat Genet 1997;17:
1145–50. 164–70.
44. Sieving PA. Juvenile X-linked retinoschisis. In: Heckenlively JR, Arden GB, 60. Nakamura H, Miyamoto K, Yokota S, et al. Focal macular photopic negative
editors. Principles and practice of clinical electrophysiology of vision, 2nd edn. response in patients with optic neuritis. Eye (Lond) 2011;25:358–64.
Cambridge, MA: MIT Press; 2006. p. 823–7. 61. Machida S, Gotoh Y, Toba Y, et al. Correlation between photopic negative
226 45. Miyake Y, Shiroyama N, Ota I, et al: Oscillatory potentials in electroretino­ response and retinal nerve fiber layer thickness and optic disc topography in
grams of the human macular region. Invest Ophthalmol Vis Sci 1988;29: glaucomatous eyes. Invest Ophthalmol Vis Sci 2008;49:2201–7.
1631–5. 62. Kooijman AC, Damhof A. ERG lens with built-in ganzfeld light source for
46. Sutter EE, Tan D. The field topography of ERG components in man-I. The stimulation and adaptation. Invest Ophthalmol Vis Sci 1980;19:315–8.
photopic luminance response. Vision Res 1992;32:433–46. 63. Kondo M, Piao CH, Tanikawa A, et al. A contact lens electrode with built-in
Section 2

47. Miyake Y, Shiroyama N, Horiguchi M, et al. Asymmetry of focal ERG in human high intensity white light-emitting diodes. A contact lens electrode with
macular region. Invest Ophthalmol Vis Sci 1989;30:1743–9. built-in white LEDs. Doc Ophthalmol 2001;102:1–9.
48. Hood DC, Bach M, Brigell M, et al. For the International Society for Clinical 64. Miyake Y, Yagasaki K, Horiguchi M, et al. Electroretinographic monitoring of
Electrophysiology of Vision. ISCEV guidelines for clinical multifocal electro­ retinal function during eye surgery. Arch Ophthalmol 1991;109:1123–6.
retinography (2007 edition). Doc Ophthalmol 2008;116:1–11. 65. Miyake Y, Yagasaki K, Ichikawa H. Differential diagnosis of congenital tritan­
49. Miyake Y, Miyake K, Shiroyama N. Classification of aphakic cystoid macular opia and dominantly inherited juvenile optic atrophy. Arch Ophthalmol
edema with focal macular electroretinograms. Am J Ophthalmol 1993;116: 1985:103:1496–501.
576–83. 66. Horiguchi M, Miyake Y, Kondo M, et al. Blue light-emitting diode built-in
Retinal Diagnostics

50. Tanikawa A, Horiguchi M, Kondo M, et al. Abnormal focal macular electro­ contact lens electrode can record human S-cone electroretinogram. Invest
Retinal Imaging and Diagnostics

retinograms in eyes with idiopathic epimacular membrane. Am J Ophthalmol Ophthalmol Vis Sci 1995;36:1730–2.
1999;127:559–64. 67. Kolb H, Lipets LE. The anatomical basis for color vision in the vertebrate retina.
51. Miyake Y, Shiroyama N, Ota I, et al. Local macular electroretinographic In: Gouras P, editor. The perception of colour. London: Macmillan; 1991.
responses in idiopathic central serous chorioretinopathy. Am J Ophthalmol p. 128–45.
1988;106:546–50. 68. Du Bois Reymond EH. Chapter 3. Von dem ruhen Nervenstrome. Unter­
52. Miyake Y, Ichikawa K, Shiose Y, et al. Hereditary macular dystrophy without suchungen Über Thierische Electricität. Vol. 2. Berlin: G Reimer; 1849;
visible fundus abnormality. Am J Ophthalmol 1989;292–9. p. 251–288.
53. Miyake Y, Horiguchi M, Tomita N, et al. Occult macular dystrophy. Am J 69. Marmor MF, Brigell MG, McCulloch DL, et al. For the International Society for
Ophthalmol 1996;122:644–53. Clinical Electrophysiology of Vision. ISCEV standard for clinical electro-
54. Akahori M, Tsunoda K, Miyake Y, et al. Dominant mutations in RP1L1 are oculography (2010 update). Doc Ophthalmol 2011;122:1–7.
responsible for occult macular dystrophy. Am J Hum Genet 2010;87:424–9. 70. Arden GB, Barrada A, Kelsey JH. New clinical test of retinal function
55. Gass JD. Acute zonal occult outer retinopathy. J Clin Neurol Ophthalmol based upon the standing potential of the eye. Br J Ophthalmol 1962;46:
1993;13:79–97. 449–67.
56. Holder GE, Brigell MG, Hawlina M, et al. For the International Society for 71. Arden GB. Origin and significance of the electro-oculogram. In: Heckenlively
Clinical Electrophysiology of Vision. ISCEV standard for clinical pattern JR, Arden GB, editors. Principles and practice of clinical electrophysiology of
electroretinography. (2007 update). Doc Ophthalmol 2007;114:111–6. vision. 2nd edn. Cambridge, MA: MIT Press; 2006. p. 123–138.
57. Maffei L, Fiorentini A. Electroretinographic responses to alternating gratings 72. Odom JV, Bach M, Brigell M, et al. ISCEV standard for clinical visual evoked
before and after section of the optic nerve. Science 1981;211:953–5. potentials (2009 update). Doc Ophthalmol. 2010;120:111–9.
58. Holder GE. The pattern electroretinogram. In: Heckenlively JR, Arden GB, 73. Miyake Y, Hirose T, Hara A. Electrophysiologic testing of visual functions for
editors. Principles and practice of clinical electrophysiology of vision, 2nd edn. vitrectomy candidates. I. Results in eyes with known fundus diseases. Retina
Cambridge, MA: MIT Press; 2006. p. 341–51. 1983;3:86–94.
59. Ogino K, Tsujikawa A, Nakamura H, et al. Focal macular electroretinogram in 74. American Clinical Neurophysiology Society. Guideline 5: guidelines for stan­
macular edema secondary to central retinal vein occlusion. Invest Ophthalmol dard electrode position nomenclature. J Clin Neurophysiol 2006;23:107–110.
Vis Sci. 2011;52:3514–20. Available at https://www.acns.org/.
Retinal Diagnostics Section 2
For additional online content visit http://www.expertconsult.com

Diagnostic Ophthalmic Ultrasound Chapter

Rudolf F. Guthoff, Leanne T. Labriola, Oliver Stachs


9 
INTRODUCTION ultrasound appeared in the medical literature in 1956.9 By the
mid-1970s, ophthalmologists were using ultrasound to deter-
Ultrasonography is a pervasive diagnostic tool within medicine. mine axial length in a clinical setting. These measurements facili-
It is used to obtain noninvasive images that can aid in the man- tated calculations of intraocular lens power which led to a
agement of patients in almost all fields of medicine. Ultrasound revolution in cataract surgery.10 Further innovations came when
technology has a unique role in ophthalmology since it can Baum and Greenwood introduced their two-dimensional
provide quantitative and qualitative assessments of the globe B-mode image to ophthalmology.11 Soon afterwards, Bronson
and orbit. Ultrasound images are formed by capturing the et al.12 developed a hand-held contact transducer for this type of
reflected acoustic signal from different tissues. These images can image acquisition which led to the rapid dissemination of ultra-
provide important details of almost every part of the eye from sound devices within ophthalmology clinics. The B-mode images
the anterior cornea and ciliary body to the posterior retina and could be used to delineate accurately retinal detachments, vitre-
choroid. This chapter will explain the principles involved in ous membranes, and choroidal tumors. In the early 1990s, new
ophthalmic ultrasound as well as provide examples of its use technology made it possible to image the anterior segment of the
within ophthalmology. eye with devices that captured images at higher frequencies of
35–50 MHz. This improved image resolution four- to fivefold
ULTRASOUND – PAST AND PRESENT
and is still the gold standard for analysis of certain anterior-
In 1880, the Curie brothers first demonstrated that a difference segment disease such as ciliary body effusions, infiltrates, and
in electric potential could be created by mechanically pressing tumors.
opposing surfaces of a tourmaline crystal.1–7 This phenomenon
is called the piezoelectric effect. This effect is the basis for ultra-
sound technology and was first applied in underwater sonar
EXAMINATION TECHNIQUES
systems during World War II.8 During that same era, the medical The ultrasound examination is performed with the patient in a
community also adopted the use of ultrasound technology. Sci- reclined position. The frequency of the ultrasound cannot pass
entists realized the diagnostic potential of this technology when through air; therefore, a coupling medium is needed to transmit
they were able to use acoustic wavelengths to study the consis- the sound waves from the transducer to the ocular tissues. A
tency of a material without damaging the material itself. common coupling agent is methylcellulose (Fig. 9.1). The cou-
In 1949, Ludwig used ultrasound to detect gallstones in pling agent is applied to the tip of the transducer probe, which
patients. The first publication on the use of ophthalmologic is then placed on the patient’s anesthetized cornea.

A B C

Fig. 9.1 Ultrasound images simulating the effect of transducer (A) without tissue-coupling agent, (B) with partial tissue-coupling agent, and
(C) with a complete coupling agent, such as a gel-like contact substance.
Fig. 9.2 (A) Cross-sectional echogram
through a normal globe. (B) Schematic
drawing of the eye: the arrow marks the
228 entrance of the optic nerve.
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

A B

Fig. 9.3 The ultrasound biomicroscopy images of the anterior segment in (A) a phakic and (B) a pseudophakic eye with multiple echo of implant
surface.

A-mode technique turning the probe 90°, orienting the transducer tip parallel to the
limbus. The images obtained are the acoustic reflections from the
The A-scan is a linear representation of echo amplitude that
opposing inner surface of the globe.
is acquired along a single line of sight. Measurements of the
The reflected sound waves are recorded by the device and can
ocular tissue can be achieved using the time interval between
be viewed as a two-dimensional image on the screen (Fig. 9.2).
emission of the acoustic pulse and echo return to calculate
The ocular structures can be examined individually. The cornea
the distance of the tissue being imaged. A-scan images should
is characterized ultrasonographically by two separate acoustic
only be obtained through open lids since the eyelid tissue can
interfaces. The anterior chamber appears planoconvex in cross-
attenuate the sound waves and decrease the resolution. The
section. The iris diaphragm cannot be satisfactorily imaged
pressure applied by the examiner to the transducer tip is
because of the limited lateral resolution power of the normal
especially important when obtaining A-scan measurements.
B-mode. A clear lens is acoustically empty and appears as an
Excessive pressure can deform the cornea, and lead to inac-
ellipsoid structure in axial sections. Similarly, normal vitreous
curate measurements.
does not give an acoustic signal; however, the presence of a
detached posterior vitreous membrane presents an interface that
B-mode technique can be imaged by increasing the amplification of the echo signal.
The B-scan is a two-dimensional cross-section image formed by The sclera is the most strongly reflecting structure on ocular
mechanically sweeping the transducer over an angle of 50–60° ultrasonography.
with the probe oriented in a specific axis. A systematic approach
should be used to acquire all images. One method is first to
obtain axial scans of the globe by placing the probe in the center High-frequency ultrasound technique
of the cornea with the transducer tip oriented toward 12 o’clock High-frequency echograms can be used for ultrasound biomi-
in order to image the posterior pole and optic nerve. Next, the croscopy (UBM). The shorter wavelengths provide better resolu-
transducer can be turned temporally 90° to obtain images tion of the anterior structures of the eye, including the cornea,
through the macula. Finally, radial and transverse images of the lens, aqueous (Fig. 9.3), and ciliary body (Fig. 9.4).13 High-
globe can be obtained by placing the probe at each clock-hour frequency probes range from 50 to 100 MHz.14–16 The 50-MHz
around the limbus. Radial scans are acquired when the probe is probe provides the best balance between depth and resolution
placed perpendicular to the limbus and the transducer tip is for UBM technique. One limitation of this technique is that the
oriented toward the cornea. Transverse scans are obtained by shorter wavelengths, from the higher frequency, have poor
depth of penetration. UBM cannot visualize structures deeper distinguish between higher and lower flow states, which aids in
than 4 mm from the surface. the interpretation of the final result (Fig. 9.5).
UBM requires immersion of the transducer in a medium
Ultrasound biometry 229
to transmit the higher-frequency wavelengths. Saline or methyl-
Basic physics formulae can be used to calculate the speed of
cellulose can be used as the coupling agent and is held in place
sound as it passes through various ocular tissues. This number

Chapter 9
over the eye with the use of a custom cup during the examina-
can then be used to calculate distance measurements within the
tion. UBM is performed through open eyelids in order to obtain
eye (Fig. 9.6). In order to obtain accurate measurements, the
a good reflection signal. Images produced by UBM have a reso-
specific speed of sound of the different intraocular media, such
lution of 30–40 µm, which is similar to that seen with a low-
as the lens, aqueous, and vitreous, must be known.18 These for-
power microscope.17
mulae provide precise measurements that can be used to measure
The cornea is the first structure seen on UBM. The anterior-
intraocular tumors or to deduce the axial length of the globe for
chamber depth can be measured from the posterior surface of

Diagnostic Ophthalmic Ultrasound


intraocular lens power calculations.
the cornea to the anterior lens pole. The posterior lens pole
cannot be imaged by UBM due to its distance from the anterior Three-dimensional reconstructions
surface. The iris is seen as a flat uniform echogenic area. The Real-time three-dimensional (3D) and four-dimensional (4D)
iris and ciliary body converge in the iris recess and insert into images are currently used in some medical specialties, including
the scleral spur. The area under the peripheral iris and above gynecology, obstetrics, and cardiology, but their use in ophthal-
the ciliary processes is defined as the ciliary sulcus. The angle mology is limited. 3D ultrasonic images can be produced from
of the eye can be studied in cross-section by orienting the probe a series of scan planes.19–22 Silverman et al.23 characterized the
in a radial fashion at the limbus. The scleral spur is the most ciliary bodies in rabbits and human subjects using 3D high-
important landmark in the angle on UBM. resolution ultrasound. In the authors’ laboratory a simple exten-
sion of the Ultrasound Biomicroscope Model 840 (Humphrey
Doppler ultrasound Instruments, Carl Zeiss Group) and VuMax UBM 35/50
Doppler images are obtained by using frequency shifts from
acoustic reflections to measure movements within a tissue and
flow conditions within vessels. These frequency shifts can be
observed in tissue volumes of less than 10 mm. False color can
be added to the images based on ultrasound frequency to

Fig. 9.4 Ultrasound biomicroscopy of the ciliary body.The shorter Fig. 9.5 Cross-sectional ultrasonogram through the posterior pole of
wavelengths are able to obtain high-resolution images of the anterior the eye: color-coded signals from the central retinal artery (red) and
structures. the central retinal vein (blue) are displayed inside the optic nerve.

A B

Fig. 9.6 (A) Immersion A-scan biometry showing peak amplitudes of the signal reflection. (B) B-scan image with distance measurement captured
on a frozen scan. Measurements are obtained by placing the cursor marks on the image and reading the distance between points.
Fig. 9.7 Prototype (Sonomed) into a user-friendly 3D ultrasonic imaging system
of a handpiece for was developed (Figs 9.7 and 9.8).24–28
three-dimensional
230 high-frequency
scanning using the ULTRASOUND IN INTRAOCULAR
VuMax UBM 35/50
(Sonomed). PATHOLOGY
Section 2

Changes in the shape of the globe


Staphyloma
A staphyloma is an abnormal ectasia of the globe that involves
uveal tissue. The ectasia typically has a smaller radius of curva-
ture then the normal sclera of the globe. It can be identified on
Retinal Diagnostics
Retinal Imaging and Diagnostics

ultrasound by taking axial cross-sectional scans with the trans-


ducer probe (Fig. 9.9).
Scleral buckle
A scleral buckle can create a posterior scleral deformity that
looks similar to a staphyloma. It can be distinguished from
a true staphyloma by a careful history or identification of the
encircling band around the anterior sclera. Also, if silicone oil
was used for repair, the higher index of refraction within the
silicone oil can alter the reflectance of the ultrasound wave-
lengths, which might provide a false impression of globe
deformation (Fig. 9.10).

A B

Fig. 9.8 Clinical pictures of the ciliary body with a pigment cyst (A) through a dilated pupil and (B) with retroillumination. (C) The same eye
shown with three-dimensional reconstructed volume with oblique sections.
231

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.9 Staphyloma in a highly myopic eye (axial length 34.8 mm). (A) An axial section of the eccentric entrance of the optic nerve can be seen.
(B) The misshapen globe is especially well demonstrated on a sectional plane which lies outside the optical axis. (C) Schematic drawing.

NO
G

P
S

A
B

Fig. 9.10 (A) Scleral buckle produced by a silicone sponge explant. After placement of a scleral buckle, the deformity of the globe can be seen in an
acoustic cross-section. Silicone explants almost completely reflect the ultrasound. They also cast an acoustic shadow. (B) Schematic drawing.
NO, optic nerve; P, explant; S, sound shadow; G, syneretic, densified vitreous.
Microphthalmos Vitreous
Congenital microphthalmos is an abnormally small eye that Echographic examination can provide information on vitreous
232 can be associated with other ocular abnormalities. The main structure which is particularly useful when visualization of the
finding in microphthalmos is axial shortening. This can be posterior pole is poor due to anterior media opacities. Ultraso-
identified with A-scan measurements. The B-scan mode can nographic findings allow the examiner to differentiate dot-,
Section 2

be used to obtain radial and transverse scans to identify abnor- strand-, and membrane-like reflections (Fig. 9.13). Table 9.1 sum-
malities in the vitreous and posterior segment of the eye, which marizes the most frequent conditions associated with pathologic
can also be associated features of microphthalmos. These fea- changes in the vitreous.
tures include the presence of a coloboma of the retina or optic
nerve head, orbital cysts (Fig. 9.11), or persistent hyperplastic Vitreous degeneration
vitreous.
Vitreous syneresis can appear as dot-like reflections which can
Retinal Diagnostics
Retinal Imaging and Diagnostics

Phthisis be more pronounced in myopia or senile vitreous. During a


Phthisis is defined as severe atrophy of the globe associated with symptomatic posterior vitreous detachment, the B-mode echo
hypotony. Phthisis is characterized ultrasonographically by a may demonstrate various stages of vitreous syneresis and may
thickened outer scleral wall. Occasionally, calcification or ossifi- reveal the remaining adhesions of the hyaloid membrane to the
cation may be observed (Fig. 9.12). This may be due to degenera- retinal surface (Fig. 9.14).
tive processes and from metaplasia of the retinal pigment
epithelium (RPE). In advanced cases of phthisis, the sclera and Asteroid hyalosis
choroid can represent up to 70% of the total volume of the globe. The calcium-containing lipids of asteroid hyalosis are suspended
The degree of thickening of the globe in chronic hypotony can in the vitreous framework and act as distinctive sound reflectors
be an indication of impending phthisis, but the precise thickness (Fig. 9.15). They can demonstrate the dynamics of vitreous
threshold for phthisis formation is unknown. movements.

A B

Fig. 9.11 Microphthalmos with orbital cyst. (A) Cystoid space is seen on the B-scan within the muscle cone, which is interpreted as a “completely
separated coloboma.” (B) A-scan image. (C) Schematic drawing.
233

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.12 Circumscribed calcification in the ocular wall in advanced phthisis bulbi. (A) Echographically we find highly reflective changes in the
ocular wall from calcification or ossification of the choroid which casts a shadow on the soft tissue located posteriorly. (B) These strong echoes
can be selectively imaged by reducing the amplification. (C) Schematic drawing.

Table 9.1 Clinical conditions with ultrasonographically


demonstrable vitreous changes

Changes in the shape of the globe


Vitreous opacities
Asteroid hyalosis
Synchysis scintillans
Acute posterior vitreous detachment
Vitreous hemorrhage
Uveitis (idiopathic)
Uveitis (infectious)
Intraocular foreign body
Persistence and hyperplasia of the primary vitreous

center of the vitreous body. This clinical picture may resemble


Fig. 9.13 Vitreous opacities. In maximal amplification, small asteroid hyalosis, but after a few seconds the cholesterol crystals
heterogeneous spots can be seen echographically, even though the will sink toward the bottom of the cavity. A-mode images
vitreous appears optically clear (left). They act as dot-like reflectors.
display characteristic flickering spikes from the reflections of
these crystals.
Synchysis scintillans
In synchysis scintillans the vitreous is filled with cholesterol Persistent and hyperplastic primary vitreous
crystals. Unlike asteroid hyalosis, these crystals are not sus- The primary vitreous contains the tunica vasculosa lentis, which
pended within the vitreous but instead float freely in the vitre- is part of the fetal vasculature system. During development,
ous space. When the globe moves, the crystals appear in the the tunica vasculosa lentis emanates from the optic nerve head
234
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

Fig. 9.14 (A) Detached posterior hyaloid membrane imaged as a floating structure of low reflectivity. (B) Schematic drawing.

A B

C D

Fig. 9.15 Asteroid hyalosis. (A) Slit-lamp microscopic photograph of the anterior vitreous space. (B) Histological image of asteroid hyalosis
shows the calcium crystals adherent to the vitreous scaffold. (C) B-scan cross-section of the crystals, which represent good reflectors for the
ultrasound. There is always an echo-free retrovitreal space seen near the ocular walls. (D) Schematic drawing.
235

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

C D

Fig. 9.16 (A–D) Posterior polar cataract in persistent hyperplastic primary vitreous (PHPV). An ultrasonographically demonstrable strand
attached to the posterior lens pole points is suspicious for PHPV (A). Frontal plane taken temporally with maximal adduction of the globe (C).
(B,D) Schematic drawings. The dark, hatched parts correspond to the opaque area of the posterior cortex and posterior capsule in a child with
severe PHPV. (E) Histological section of PHPV as seen at optic nerve head with loupe magnification.

and supplies the posterior lens. This structure should involute clear at birth but may become cataractous when the posterior
prior to birth. Failure of the primary vitreous to regress fully lens capsule ruptures.
is termed persistent hyperplastic primary vitreous. As men-
tioned earlier, this can be associated with microphthalmos and Vitreous hemorrhages
cataract formation in the newborn. The condition persistent An acute vitreous hemorrhage is an important indication for
hyperplastic primary vitreous can be ultrasonographically char- ultrasonography. Acute hemorrhages can fill the vitreous cavity
acterized by two features. The first is a strand of membrane with small opacities from the particles of the red blood cells.
that extends between the posterior surface of the lens and the These opacities usually accumulate after a few hours in the
area of the optic nerve head. The second is the reduced axial lower circumference of the vitreous base (Fig. 9.17).
length of the globe from microphthalmos on ultrasound biom- If a detachment of the posterior hyaloid membrane pre-
etry (Fig. 9.16). If the anomaly is only mild, the lens may be cedes a vitreous hemorrhage, the erythrocytes frequently
236
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C
B

Fig. 9.17 (A) Conspicuous bleeding into syneretic vitreous; erythrocytes within the vitreous create reflective opacities. A static picture may give the
impression of a solid lesion. (B) After a few hours the opacities usually accumulate in the lower aspect of the vitreous cavity. (C) Schematic drawing.

operculum (Fig. 9.20). This area should be localized echo-


graphically and then carefully scrutinized with ophthalmos-
copy if possible.
In larger hemorrhages, the blood can also disseminate into
multiple pre-existing vitreous compartments. In the early phase
of this process, the erythrocytes will collect in the retrovitreal
space (Fig. 9.21). The retrovitreal space may completely clear
after a few days or weeks due to its high fluid exchange rate;
however, blood on the vitreous framework absorbs much more
slowly (Fig. 9.22).
Vitreous hemorrhage from neovascularization
Hemorrhages that develop from proliferative changes in
patients with diabetic retinopathy and retinal neovasculariza-
tion will always be accompanied by pathologic changes in
the vitreous. Vitreous membranes tent rectilinearly between
Fig. 9.18 Fresh vitreous hemorrhage. In a cross-sectional echogram the adhesions to the retina. The normal aftermovements that
the vitreous framework converges towards the ocular wall. Blood should occur in the vitreous after eye movements are extin-
precipitates increase the acoustic reflectivity of the vitreous. Traction guished in the presence of peripheral neovascular tufts. The
has to be assumed where the vitreous is in contact with the ocular wall.
vitreous tufts create adhesions that encircle the posterior pole.
precipitate on to a vitreous strand (Fig. 9.18). This strand This is an ominous sign, which is indicative of early retinal
may be responsible for the development of a retinal tear, and tractional detachment from these circular adhesions (Fig. 9.23).
its traction can be demonstrated directly in acoustic section- Choroidal neovascularization from age-related macular degen-
ing (Fig. 9.19). A circumscribed thickening of the ocular wall eration will have hemorrhage in multiple layers of the eye
in cross-section may indicate the presence of a retinal (Fig. 9.24).
237

Chapter 9
Diagnostic Ophthalmic Ultrasound
A
B

Fig. 9.19 (A) Recent vitreous hemorrhage. The low reflecting membranes float freely with ocular movement. A newly formed horseshoe tear may
be present at their connection point to the wall. (B) Schematic drawing.

A B

B
C

Fig. 9.20 (A,B) Recent vitreous hemorrhage. Erythrocytes have precipitated on to the partly detached posterior hyaloid membrane, increasing its
acoustic reflectivity. (C) Schematic drawing.
238
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C
B

Fig. 9.21 (A) Vitreous hemorrhage with posterior vitreous detachment emphasizing the retrovitreal space. (B) In the early stage after the
hemorrhage, erythrocytes accumulate in the retrovitreal space. They may ensheath the part of the vitreous that was free of blood and had a
normal structure. (C) Schematic drawing.

A B

Fig. 9.22 The retrovitreal space may completely clear after a few days or weeks due to its high fluid exchange rate; however, blood on the
vitreous framework or located subretinally absorbs much more slowly (A). (B) Schematic drawing.
239

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.23 (A) Beginning traction detachment at the posterior pole with vitreous contraction and proliferative diabetic retinopathy, which is hidden
behind a diffuse vitreous hemorrhage. A springboard-like, taut, detached hyaloid membrane is still adherent to the retina at the posterior pole
and has led to a traction detachment in several places. (B) Schematic drawing.

A
B

Fig. 9.24 (A) Extensive vitreous hemorrhage from disciform macular degeneration. The blood dissipates into the preretinal or intrachoroidal
space, into the area of the macular lesion and into the detached vitreous. The retrovitreal space is echo-free because of its high fluid exchange.
(B) Schematic drawing.

Terson syndrome body (Fig. 9.27). If panophthalmitis follows a perforating injury,


Terson’s sign is a multilayered, intraocular hemorrhage at the ultrasound evaluation can detect a local reaction at the entrance
posterior pole that typically occurs after blunt trauma to the point of the infection (Figs 9.28 and 9.29).
head. This is usually accompanied by a subarachnoid hemor-
Vitreous inflammation
rhage. If the posterior hyaloid membrane is still attached, the
preretinal bleeding will slowly diffuse into the formed vitreous Inflammatory and hemorrhagic vitreous changes cannot be dif-
(Fig. 9.25). This can damage the underlying retina and may be ferentiated on the basis of ultrasonographic findings alone. Both
an indication for an early vitrectomy. conditions may cause densification of pre-existing vitreous struc-
tures with subsequent shrinkage of the vitreous; tractional
Intraocular infections detachment of the retina can occur, especially if there are postin-
Ocular infection that extends toward the anterior segment or flammatory adhesions between the vitreous and retina. In chronic
results in a hypopyon formation will have changes within the uveitis, an early and complete posterior vitreous detachment can
anterior vitreous space that are demonstrable on ultrasound. A occur and cause the formed vitreous to shrink and form a frontal
thickening of the retina or choroid can be seen if the inflamma- membrane that extends across the vitreous base (Fig. 9.30). If this
tion penetrates to the outer layers of the globe (Fig. 9.26). After membrane adheres to the ciliary body, it may detach the ciliary
only a few hours, these changes may involve the entire vitreous body and produce subsequent ocular hypotony.29
240
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C
B

Fig. 9.25 (A,B) Retrovitreal bleeding associated with a subarachnoid hemorrhage (Terson syndrome). The bleeding originates from the area of
the optic nerve head. (C) Schematic drawing.
241

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.26 (A,B) Panophthalmitis after an intraocular operation. Widening of the ocular wall to about 2.1 mm indicates inflammatory choroidal
infiltrates. (C) Schematic drawing.
242
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C D

Fig. 9.27 Vitreous abscess that caused a bacterial orbital inflammation. (A) B-scan ultrasonography demonstrates a highly reflective vitreous
body which is partly detached from the retina. (B) Schematic drawing of this image. (C) Posterior pole shows infiltration of Tenon’s space and
widening of the optic nerve sheath. (D) Schematic drawing.

A
B

Fig. 9.28 Posttraumatic intraocular infection starting from the side of perforation. (A) B-scan ultrasonography demonstrates localized thickening of
the ocular wall indicating the side of the perforation. The vitreous is filled with inflammatory cells, the posterior hyaloid is thickened, and there is
a localized retinal detachment. (B) Schematic drawing.
243

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

C D

Fig. 9.29 Chronic inflammation with massive vitreous infiltration after pars plana vitrectomy and scleral buckle. (A) Remnants of the infiltrated
vitreous can be seen at the vitreous base anterior to the scleral buckle. (B,C) Cross-sections through the vitreous base. (D) Schematic drawing.

Intraocular foreign bodies visualization. It is important to have a strong fundamental


Intraocular foreign bodies induce a change in echo reflectivity knowledge of ocular anatomy as well as a good technical
which is based on the composition of the material (Figs 9.31–9.33). approach for acquiring ultrasound images in order to make
The change in the reflectivity on the image should be a helpful accurate assessments of retinal disease on ultrasound. This
clue in the localization of the foreign body within the globe; section will review the anatomical features of common retinal
however, this is not always the case since the foreign bodies can conditions as well as the special techniques needed to examine
also create signal artifact on echograms that can make identify- the retina with ultrasound.
ing their exact location difficult. For example, large metallic Acute retinal detachment
foreign bodies have significant artifacts from strong reflected In a retinal detachment, the neurosensory retina separates from
signals that can distort their true location. In addition, foreign the RPE layer. This development allows fluid to collect in the
bodies from trauma can be associated with air bubbles within potential space between these two layers. The detached neuro-
the vitreous that can mask the presence of the nearby foreign sensory retina appears as a membrane in the vitreous space on
body within the acoustic shadow (Fig. 9.34). ultrasound. Partial retinal detachments may still maintain con-
nections to the optic nerve or ora serrata since these areas have
Retina the strongest connections to the retina. Identification of these
Ultrasound can be used to assess the structure of the retina in connections on ultrasound can distinguish a partial retinal
order to discern anatomical changes such as retinal tears and detachment from a vitreous or choroidal detachment, which
detachments. It can also be used to identify changes in retinal would have different anatomical connections (Fig. 9.35). A com-
thickness from infiltrative or exudative ocular diseases. The plete retinal detachment can form a funnel shape due to the
sound reflections in ultrasound images can delineate these path- retina folding in the center of the globe.
ological retinal changes even in eyes with opaque anterior Complicated retinal detachments with severe pathology can
media. This is a particularly useful tool since many diseases that make it difficult to identify all the structures on ultrasound (Fig.
affect the retina can also lead to vitreous changes that limit direct 9.36). For example, in severe trauma cases that are associated
244
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

2.9
mm

C D

Fig. 9.30 Phthisis oculi in chronic panuveitis. (A) The vitreous has shrunk to form a frontal membrane of high acoustic reflectivity. The ocular wall is
widened to 2.9 mm. (B) Using linear amplification, the various layers of the ocular wall have become clearly outlined. (C) Isolated areas of the ocular
wall show high acoustic reflectivity, which indicates calcification of the choroid or in the sclera. (D) Schematic drawing highlighting wall thickness.

A B

Fig. 9.31 (A) Metallic foreign body in the vitreous with total retinal
detachment. (B) The foreign-body spike is characterized by its high
C amplitude and repetitive echoes. (C) With reduced sensitivity the
foreign body can almost be imaged as an isolated echo.
245

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.32 Acute perforating injury with intraocular metallic foreign body. (A) Ultrasonographically, there is a typical foreign-body echo with
repetitive spikes in the vitreous about 2 mm in front of the ocular wall. (B) Decreasing the amplification displays the foreign-body echo as the
only intraocular structure remaining visible on ultrasonography. (C) Schematic drawing.
246
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

B
C

C
B

C D

Fig. 9.33 (A) Intraocular foreign body of a piece of wire. (B) The anterior end of the wire remained visible in the lens. Cross-sectional image
taken temporally with maximal adduction of the globe. (C) Frontal section through iris and lens. (D) Schematic drawing.

• What is the spatial extent of the membrane? At which point


is there contact with the ocular wall?
• What is the shape of a cross-section of the membrane,
especially in the optic nerve head area?
• Which aftermovements occur?
• How great is the difference of the spike from the membrane
in question to the scleral standard or to the echo from a
standard reflector?

These questions should be clarified in the following way:

1. What is the spatial extent of the membrane? At which point


is there contact with the ocular wall? A recent
rhegmatogenous retinal detachment can be characterized in
cross-section by a membranous structure of high reflectivity
that converges in an acute angle toward the ocular wall. If
the imaged acoustic section is centered on the optic nerve
Fig. 9.34 Computed tomography image of a metallic foreign body head, then the border of the detached retina will be captured
wedged into the ocular wall with an air bubble adjacent to it. as it connects to the nerve head (Fig. 9.37). If the membrane
passes over the optic nerve head instead of connecting to the
with proliferative vitreoretinopathy or in advanced diabetic optic nerve in the echo image, it is not a retinal detachment
disease associated with proliferative retinopathy, the mem- (Fig. 9.38). This feature can help identify retinal membranes
branes formed within the vitreous can appear similar to a true form vitreous membranes.
retinal detachment. The following questions can guide the ultra- 2. What is the shape of a cross-section of the membrane,
sound examination of the retina in order to differentiate these especially at the optic nerve head? In order to appreciate the
common causes of vitreous membranes: shape of a retinal detachment, the performance of
247

Chapter 9
Diagnostic Ophthalmic Ultrasound
A
B

Fig. 9.35 (A) Complete retinal detachment extending between the optic nerve head and the ora serrata. Heterogeneous material in the anterior
vitreous is a sign of vitreous reaction. (B) Schematic drawing.

A B

C B

Fig. 9.36 Development of a traction detachment with massive vitreous retraction. (A) Flat subtotal retinal detachment with folds; rigid vitreous
strands. (B) In a frontal section, multiple contacts of the retina to the ocular wall can be demonstrated. (C) Schematic drawing.
248
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A
B

Fig. 9.37 (A) Sectoral retinal detachment. In acute retinal detachment, short aftermovements appear when the globe moves. These
aftermovements extend like a whiplash from the area of the contact to the ocular wall. (B) Schematic drawing.

A B

Fig. 9.38 (A,B) Detached posterior hyaloid membrane. Vitreous membranes may reach an acoustic reflectivity similar to that of the retina
seen on A-scan. A retinal detachment can be excluded if the membrane tents over the posterior pole without reaching the optic nerve head.
(C) Schematic drawing.
sonographic examinations in various planes is indicated. temporal part of the lid fissure when the globe is maximally
First, in a sagittal section, a total detachment looks like an adducted. In these images, the conical shape of the
isosceles triangle which is open toward the anterior segment detachment will appear oval to nearly circular in the various 249
(the sides of which may be unevenly tented: Fig. 9.35). Next, sections (Fig. 9.39).
frontal-plane sections should be examined with the disc 3. Which aftermovements occur? Dynamic ultrasound can be

Chapter 9
centered in the image. These sectional plans are obtained at obtained with patient participation. The quality of tissue
a right angle to the sagittal. The frontal planes can be movement at the end of the ocular saccade, or the
examined best with the transducer probe placed in the aftermovement of the tissue, can be used to distinguish

Diagnostic Ophthalmic Ultrasound


A

B C
B

D
E

Fig. 9.39 (A) Clinical fundus photography of recurrent retinal detachment. (B–E) Recurrent retinal detachment with massive vitreous retraction.
(B) Axial cross-sectional echogram. The amplitudes of the aftermovements decrease. High-frequency flicker of the taut membrane appears after
the eye changes position. (D) Frontal section toward 6 o’clock. An epiretinal membrane of the retinal surface appears echographically as an
apparent widening of the retina. (C,E) Schematic drawings.
vitreous tissue from retina tissue. An acute rhegmatogenous A few weeks after a retinal detachment develops, changes in
retinal detachment shows aftermovements of short duration the proliferation of Müller cells and astrocytes will lead to altera-
250 that extend with a whiplash effect from the area where the tions in the mechanical properties of the detached retina. The
retina is still attached, which is usually the optic nerve head. massive periretinal proliferation of these cells creates a decrease
The amplitudes of these aftermovements are smaller and less in the aftermovement amplitude of the retina (Fig. 9.40). The
extensive than those seen in the sinusoidal movements of a large excursions are replaced by a high-frequency flicker and the
Section 2

vitreous hemorrhage or in asteroid hyalosis.30 retina may appear thickened. After a long-standing retinal
4. How great is the difference of the spike from the membrane detachment, cyst-like cavities within the detached retina can be
in question to the scleral standard or to the echo from a seen on ultrasound (Fig. 9.41). A long-standing detachment can
standard reflector? Quantitative ultrasonography can detect a also result in a funnel-shaped retinal detachment. The first step
difference in the echo of the retina compared to that of the in this process is the proliferation of the epiretinal connective
sclera, extending from 8 to 15 db.31 Unfortunately, these tissue that causes the vitreous to contract. This stage can be
Retinal Diagnostics
Retinal Imaging and Diagnostics

measurements provide only guidelines. Well-developed identified on ultrasound by the presence of new acoustic inter-
connective tissue membranes may show reflection properties faces, which are seen as increased vitreous signals that surround
quite similar to those of a detached retina. In complicated the detached retina (Fig. 9.42). The vitreous shrinks, which
cases it may be difficult to correlate an isolated A-spike to the creates a narrowing of the retinal cavity, which typically begins
multiple membrane structures as they appear on B-mode. to narrow anterior to the optic nerve head and continues to the
posterior lens. Frontal “cyclitic membranes” can often be seen
Chronic retinal detachment early, extending from the vitreous base (Fig. 9.43). Next, the
The duration of a retinal detachment can affect the thickness and peripheral retina is pulled closer to this membrane until the
mobility of the retina, the shape and mechanical properties of retinal detachment does not show any recognizable cavity of
the vitreous base, and the contents of the subretinal space. Iden- the original funnel. In frontal sections the shrunken retina
tifying and understanding these changes can help with the man- appears as a strand, only a few millimeters in diameter
agement of these conditions. (Fig. 9.44). Occasionally, a few retinal folds can be observed.

A B

Fig. 9.40 Massive periretinal proliferation with large retinal folds. (A) Ophthalmoscopic picture of large horseshoe tear. (B) The rigid, fixed retinal
folds are echographically demonstrable. (C) Schematic drawing.
251

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.41 Long-standing total retinal detachment with macrocyst. Intraretinal cyst in the axial area seen on cross-sectional ultrasound (A) and
frontal plan (B). The cysts form from coalescing microcystoid degeneration of the retina in chronic detachments. These cavities will not be
involved in secondary vitreous changes, such as vitreous hemorrhages or cholesterol deposits. (C) Schematic drawing.

In addition to the above findings, the consistency of the in retinoschisis are less conspicuous than in a retinal detach-
subretinal space can change with increased duration of the ment. This is because in retinoschisis the retina is attached and
detachment. The protein content will increase and may pre- there are no vitreous adhesions.
cipitate out of the subretinal fluid. This can be seen as free- Coats disease
floating opacities on ultrasound (Fig. 9.45). If these opacities Coats disease is an exudative retinopathy most commonly seen
appear as static particles and not free-floating, then intraocular unilaterally. It mainly affects males in the first decade of life. A
tumor has to be excluded. Under these circumstances, ultra- clinical diagnosis can be made if aneurysmal malformations of
sonography provides a vital role in obtaining a diagnosis by retinal vessels and yellowish subretinal plaques are associated
distinguishing the freely moving protein precipitation in long- with an exudative detachment. Coats disease should be differ-
standing detachments from the fixed hyperechoic particles of entiated from retinoblastoma, which can have a similar clinical
malignant tumors. appearance in this patient group. Ultrasound can differentiate
Retinoschisis these conditions with careful evaluation of the subretinal space.
Retinoschisis is a splitting within the neurosensory layer of the Retinoblastoma typically shows calcifications with high reflec-
retina. This condition frequently occurs in the inferotemporal tivity. The exudative detachment that is seen in Coats disease
quadrant. A cross-section echogram can display a membranous has different echo quality due to the subretinal cholesterol
structure in the far periphery that has a convex border facing the deposits (Fig. 9.47). In Coats disease, the crystals floating in the
vitreous (Fig. 9.46). In this situation, clinical correlation is impor- subretinal space are seen as floating opacities similar to the
tant in order to distinguish the retinoschisis from a retinal appearance of crystals in synchysis scintillans. They appear in
detachment since these two entities can appear identical on static an A-mode echogram as flickering spikes.32 On B-mode images
ultrasound. However, use of dynamic ultrasound and evalua- these high-frequency motions of the echo spikes produce a
tion of the aftermovements can show that the aftermovements blurred pattern in the subretinal space.
252
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C B

Fig. 9.42 (A,B) Long-standing retinal detachment with increased connective tissue in the vitreous tunnel. Free-floating opacities accumulate in
the subretinal space (possibly representing hemorrhage or cholesterol crystals). (C) Schematic drawing.

A
B

Fig. 9.43 (A) Long-standing total retinal detachment. A tube-like remnant of the vitreous within the shrunken retinal tunnel can be seen. (B) Schematic
drawing.
253

Chapter 9
Diagnostic Ophthalmic Ultrasound
A
B

Fig. 9.44 (A) Long-standing retinal detachment with completely obliterated vitreous space. In sagittal sections the extensive adhesions of the
retinal leaves produce various acoustic sections. (B) Schematic drawing.

A B

Fig. 9.45 (A,B) Long-standing retinal detachment with floating opacities in the subretinal space. The densifications in the vitreous space indicate
a tendency for shrinkage. (C) Schematic drawing.
254
Section 2

A B
Retinal Diagnostics
Retinal Imaging and Diagnostics

Fig. 9.46 (A) Senile retinoschisis in the inferotemporal quadrant. The typical biconvex cross-sectional image in association with slight aftermovements
supports a tentative diagnosis of retinoschisis. (B) The acoustic reflectivity corresponds to that of a detached retina. (C) Schematic drawing.

A
C

Fig. 9.47 (A) Fundoscopic clinical photo of Coats disease. Circumscribed, strongly reflecting retinal detachment in Coats disease. (B) The A-scan
shows the high-intensity signal from the strongly reflecting membrane of the retinal detachment depictred in the image. (C) The B-scan shows
the retinal detachment in association with floating opacities in the subretinal space that corroborate the diagnosis of Coats disease.
Retinoblastoma extension outside the orbit, optic nerve invasion, or pinealoma
Retinoblastoma is a life-threatening tumor that can present (Fig. 9.50).
in children as isolated leukocoria or with a constellation of Retinopathy of prematurity 255
other ocular findings. Some findings can mimic benign ocular Early stages of retinopathy of prematurity can be diagnosed by
conditions. Since treatment of retinoblastoma can include ocular indirect ophthalmoscopy. However, later stages with partial or

Chapter 9
enucleation, making the appropriate diagnosis is critical. Oph- complete tractional retinal detachments from extensive fibrovas-
thalmoscopy should be performed but sometimes is limited cular tissue require ultrasonography for diagnosis and surgical
if there is cataract, hypopyon, vitreous seeding, or opacity. planning. Machemer and Aalberg33 emphasized the importance
Ultrasound plays a critical role in these patients, especially of information obtained by cross-section ultrasonography in
since certain features on echography can be pathognomonic these stages, such as the diameter of the retinal funnel in the
for retinoblastoma. The presence of calcium deposits in reti- retrolental space (Fig. 9.51).
noblastoma lesions produces a high sound reflection that

Diagnostic Ophthalmic Ultrasound


creates an acoustic shadow on more distant structures such Optic nerve
as the sclera (Fig. 9.48). The calcium deposits can be selec- Coloboma of the ocular fundus
tively demonstrated on the image by decreasing the ampli-
Colobomas can occur in the iris, lens, retina, and optic nerve.
fication in the cross-section echogram until they are the only
Some are spontaneous and some occur in association with
remaining tissue structure visible on the screen (Fig. 9.49). In
systemic syndromes. Colobomas are also in the differential
some retinoblastomas the calcium appears only in few areas,
diagnosis of retinoblastoma.34 Colobomas can be seen clearly
but any calcification is indicative of tumor burden and is an
by ultrasonography even if the fundus cannot be visualized
important feature for the diagnosis. In some cases, ultrasound
optically (Fig. 9.52).
can also define extraocular tumor extension in retinoblastoma
and is of great prognostic and therapeutic importance. Addi- Assessment of optic nerve cupping
tional imaging to supplement ultrasonography is important Evaluation of optic nerve cupping is important for treatment of
in retinoblastoma. Computed tomography can show calcifica- glaucomatous changes. This is difficult with anterior media
tion and magnetic resonance imaging can show tumor opacity. Recently, with improvement of the lateral resolution of

Fig. 9.48 Extensive retinoblastoma with varying appearance. As


demonstrated in the cross-sectional echogram (A), the upper tumor
portions show high acoustic reflectivity due to massive calcifications.
The lower part of the tumor has less acoustic reflectivity than the
adjacent sclera. A-scans of the eye (the scleral spikes in the acoustic
scans (B,C) are marked by arrows).

B C
256
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C D

Fig. 9.49 Large retinoblastoma with calcifications and acoustic shadow cast on the adjacent sclera and orbit. (A) The calcifications light up
in cross-sectional echograms. (B) In A-mode echograms, they can be quantified as the strongest reflecting structures in the examined area.
(C) With reduced amplification, the calcified areas are the only visible signals. (D) Schematic drawing.
257

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

C
D

NO

E S

Fig. 9.50 Extensive retinoblastoma. (A) A cross-sectional echographic image. The tumor is characterized by areas of high acoustic reflectivity
alternating with areas of low reflectivity. (B) The differences in reflection can be quantified on A-mode echography: calcified parts of the tumor
have markedly higher amplitudes as seen at point K as compared to the sclera reflection as seen at point S. (C) Schematic drawing of the
ultrasonographical image in (A). (D) Computed tomography scan displays the calcified parts of the retinoblastoma that can be identified with
greater certainty than in a plain X-ray picture. (E) A histological sample of the infiltration into the optic nerve. NO, optic nerve; R, retinoblastoma.
The calcium deposits explain the nearly pathognomonic echogram.
258
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

Fig. 9.51 Stage V of retinopathy of prematurity. (A) In cross-sectional images we see a complete retinal detachment with a widening of the
avascular peripheral retina toward the subretinal space. (B) Schematic drawing.

B-scan images, optic nerve head cupping can be reliably mea- and ossification of ocular tissue that is easily identified on ultra-
sured and reproduced on ultrasound images.35 sound because of the total reflection of all sound waves noted
on the acoustic image.
Choroid
Choroidal neovascularization
Changes in the ocular layers due to hypotony
Choroidal neovascularization is a complication seen in many
In acute hypotony, exudation into the suprachoroidal space
eye diseases, most commonly in macular degeneration. The
creates an increase in the choroidal vascular pressure. This can
histological findings include a vascularized collection of con-
lead to a choroidal detachment which will then exacerbate the
nective tissue that extends from the choriocapillaris through
globe hypotony (Fig. 9.53). This situation is ultrasonographically
defects in Bruch’s membrane and then into the subretinal
characterized by a convex border on a cross-section image that
pigment epithelial or subretinal space. This heterogeneous
denotes the location of the choroidal detachment. This convex
structure produces mixed ultrasound features, including the
shape is formed between the pars plana and location of the
strong acoustic reflections from the fibrovascular membranes
vortex veins, both of which have strong choroidal attachments.
and dense connective tissue septa and the acoustically silent
Knowledge of the choroidal anatomic structure and its attach-
areas of exudate and subretinal fluid (Figs 9.24 and 9.57).
ments can aid in ultrasound diagnosis. In addition to the pars
Peripheral choroidal neovascularization membranes can appear
plana and vortex veins, the choroid is firmly attached to the
similar to choroidal melanomas; therefore, familiarity with the
optic nerve. The choroid inserts at the optic nerve at a blunt
ultrasonographic features of these lesions is important, espe-
angle, as compared to the retina, which has a steeper insertion.
cially if vitreous hemorrhage prevents direct visualization.
This feature can be used to help differentiate choroidal and
retinal detachments of the posterior pole on ultrasound images. Choroidal melanoma
In addition, a choroidal detachment would begin at the ciliary Evaluation of choroidal melanomas includes a comprehensive
body and not the ora serrate, as in a retinal detachment (Fig. examination with ophthalmoscopy combined with ultrasound
9.54). Occasionally, the detached ciliary body can compress the imaging. In choroidal melanomas, the signals produced within
lens. In severe cases, choroidal detachments may meet in the the tumor are complicated by overlapping and dampening echo
center of the globe. This feature has been termed “kissing cho- processes. Connective tissue septa and vessels vary in their
roidals” and needs to be surgically corrected (Fig. 9.55). In prominence and interfere with the signals from the tumor tissue.
typical cases of serous choroidal detachments the subchoroidal Furthermore, the resolution of the image is limited because the
space is acoustically silent. In contrast, an expulsive choroidal densely packed tumor cells are separated by a considerably
hemorrhage can be identified by the hyperechoic signal on shorter distance than the ultrasound wavelength. A thorough
ultrasound that the blood clots create within the detachment understanding of the principles of ultrasonography and a strong
(Fig. 9.56). knowledge base of the tissue complexity in metastatic processes
In chronic hypotony with long-standing choroidal detach- combined with an extensive case reference aids in the image
ments, the outer layer of the globe can appear concentrically interpretation of choroidal melanomas. The interpretation of
widened on ultrasound. In advanced stages, the appearance on echograms in A- and B-mode is based on reports on this topic
ultrasound changes, the outer walls appear thickened, and the which were published by Oksala,36 Baum,37 Buschmann and
vitreous cavity becomes substantially reduced. In these eyes, Trier,38 Till and Ossoinig,32 Trier,39 Coleman et al.,40 and
metaplasia of the pigment epithelium can result in calcification Text continued on page 263
259

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

C D

Fig. 9.52 (A) Echogram of the small optic nerve coloboma depicted in (E). In comparision, B-scan section showing normal physiological cupping
of an optic nerve head (C). To obtain this vertical section a probe is placed temporal to the limbus, thereby avoiding any artifacts, caused by the
lens. (B,D) Schematic drawings. (E) Small coloboma of the optic nerve with a diameter of about 2.5 mm. Fundus photograph showing excavation
within the nerve.
260
Section 2

A B
Retinal Diagnostics
Retinal Imaging and Diagnostics

Fig. 9.53 (A) Detachment of the ciliary body and the peripheral choroid due to hypotony. (B) The thickening of the ocular walls begins in the area
of the ciliary body and extends to the equator, as seen on the B-scan. (C) Schematic drawing.
261

Chapter 9
A B

Diagnostic Ophthalmic Ultrasound


B

C
B

Fig. 9.54 Subtotal exudative choroidal detachment. (A) In contrast to a retinal detachment, the choroidal detachment extends beyond the ora
serrata; the detachment extends from the iris diaphragm to the posterior pole without reaching the optic nerve head. (B) The frontal sections
depict indentations which are caused by large vessels. (C) Schematic drawing.

A
B

Fig. 9.55 Total exudative choroidal detachment in persistent hypotony after penetrating glaucoma surgery with external fistulation. (A) The apices
of the choroidal detachment touch each other in the vitreous and are seen on the echogram. This finding is commonly termed “kissing choroidals.”
Strand-like structures (possibly taut vortex veins) course through the intrachoroidal space. (B) Schematic drawing.
262
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C
B

Fig. 9.56 Extensive choroidal detachment in an eye with expulsive hemorrhage. (A) The intrachoroidal space has acquired acoustic reflectivity
due to the accumulation of blood. There may be anatomical healing after the absorption of the blood, but we cannot expect visual function to
improve. (B) Frontal image of the hemorrhagic choroidal detachments. (C) Schematic drawing.
263

Chapter 9
A B

Diagnostic Ophthalmic Ultrasound


NH
S

D
C

Fig. 9.57 Disciform macular degeneration with exudative retinal detachment. (A) In the cross-sectional echogram, the thickened ocular walls in
the macular area appear as a strongly reflecting, layered structure. (B) The acoustic interfaces of the macular degeneration produce high
signals. (C) Histologic section of an eye with disciform macular degeneration under loupe magnification. Vascularized connective tissue
scars with exudation without acoustic interfaces produce the substrate for the heterogeneous nature of the echogram. NH, retina; S, sclera.
(D) Schematic drawing.

Silverman et al.,41 which include data from the original scans of likely due to more than just an attenuation phenomenon. The
ocular tissues. The next section will review the principles used features of the collar button can be explained on the basis of
to image choroidal tumors and the features of a choroidal mela- echographic–histopathologic correlations (Fig. 9.61). Usually, a
noma on B-mode and A-mode imaging. tumor lying immediately beneath the sensory retina reflects
The characteristics of a choroidal melanoma on ultrasound more than the portion of the tumor beneath Bruch’s
B-mode echography membrane. In a collar button, the part of the tumor growing
In B-mode echograms, a melanoma appears as a biconvex lesion. in front of Bruch’s membrane will be drained less well, leading
The internal structure is relatively homogeneous so produces to a dilatation of the vessels in this part of the tumor.31,42 This
markedly fewer signals than the tumor surface or the sclera creates new acoustic interfaces with higher signal intensity,
(Fig. 9.58). If the tumor has broken through Bruch’s membrane, resulting in brighter dots in B-mode and higher spikes in
then a mushroom-shaped lesion, or a collar button, can be A-mode. In the area of the tumor base, a “choroidal excava-
demonstrated in cross-section and serves as a pathognomonic tion” can be seen (Figs 9.62 and 9.63). This excavation in the
sign (Fig. 9.59). If present, this perforation does not necessarily acoustic section develops at the edge of the tumor where mela-
occur at the peak of the tumor; careful ultrasound examination noma tissue intersects the adjacent strongly reflecting intact
of the entire lesion is needed (Fig. 9.60). In some sections, the choroid.37,43
collar button of a tumor may even simulate a tumor mass lying The characteristics of a choroidal melanoma on
free in the vitreous (Fig. 9.59). In addition, the collar button A-mode ultrasonography
portion of the lesion has unique ultrasound features due to The ultrasonographic internal structure of a tumor is quantita-
the presence of the mass above Bruch’s membrane in some tively best described in the A-mode echogram. An unfocused
places and below Bruch’s membrane in other places. Previously A-mode transducer can obtain a summation signal from the
this feature was attributed to sound attenuation, which is the large tissue area,31 which typically demonstrates spikes of rela-
continuous decay of spike altitudes in the A-mode and was tively uniform amplitudes within the large biconvex tumor if the
called the angle kappa. However, the change in signal is most lesion has not extensively broken through Bruch’s membrane
264
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

S
A

NO

B C

Fig. 9.58 Typical findings of a choroidal melanoma. (A) In cross-section the tumor is biconvex. The diameter of the base is 14 × 14 mm;
elevation, 8.5 mm; calculated tumor volume, 750 mm3. (B) With standardized A-mode, the spike amplitude within the tumor amounts to about
20% of the scleral spike. (C) Histological section visualized under loupe magnification of a peripapillary choroidal melanoma. The reason for the
low homogeneous acoustic reflectivity of a choroidal melanoma is the densely packed tumor cells, the margins of which are separated by a
distance much smaller than the wavelength of ultrasound. R, retina; S, sclera; NO, optic nerve.
265

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

C B

Fig. 9.59 Large choroidal melanoma, mushroom-shaped after perforating Bruch’s membrane. (A) Sagittal section. (B) In the frontal plane, one
part of the tumor does not seem to have contact with the ocular outer walls, which demonstrates the principle of the necessity to image tumors
in multiple planes to define the full extent of invasion. (C) Schematic drawing.
266
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C D

E
F

Fig. 9.60 (A) Large mushroom-shaped choroidal melanoma with collar button seen on fundus photography. (B) Sagittal echogram cross-section
with A-mode echogram. There is markedly increased reflection in the area of the tumor peak. The part of the tumor in front of Bruch’s membrane
often shows dilated vessels, which act as interfaces to increase the acoustic reflectivity. (C) Histological section through the tumor. (D) Schematic
drawing. (E) Echogram from the tumor base. This image was obtained with maximal adduction of the globe, and made possible because of the far
temporal location of the tumor base. Also in this direction of examination, the part of the tumor anterior to Bruch’s membrane demonstrates high
reflectivity. (F) Schematic drawing.
267

Chapter 9
Diagnostic Ophthalmic Ultrasound
A
B

Fig. 9.61 (A) Choroidal melanoma with eccentric perforation of Bruch’s membrane. Echogram shows a mushroom-like shape of the lesion, which is
nearly pathognomonic for a melanoma and therefore of great differential diagnostic value. (B) Schematic drawing.

A
B

Fig. 9.62 (A) Small choroidal melanoma. Elevation, 2.5 mm; base, 8 × 8 mm; calculated tumor volume: 66 mm3. The choroid at the tumor base
has been replaced by tumor, which has less acoustic reflectivity and provokes a choroidal excavation echographically. (B) Schematic drawing.

(Fig. 9.58). This corresponds to the uniform histological structure 50 eyes with melanoma, Doppler shifts could be detected in all
of a melanoma (Fig. 9.58). Exceptions occur when features of patients except one. The tumor size varied between 80 and
hemorrhage or necrosis within the tumor develop.44,45 1500 mm3. Doppler imaging has also been used to study an eye
Most melanomas consist of solid tissue; therefore, no after- with secondary glaucoma and significantly increased intraocular
movements occur as might be expected with a large subretinal pressure. In this patient the tumor perforated the sclera and
or choroidal hemorrhage. In addition, flickering spike complexes invaded the muscle cone (Fig. 9.65). An intraocular pressure of
may be seen on the A-mode echogram within the tumor itself. 45 mmHg compressed the intraocular tumor, which influenced
Various authors interpret them differently. It is assumed that the maximal flow velocities measured by Doppler sonography
they are due to blood circulating in large tumor vessels. Although (Fig. 9.65).
the features of the ultrasound combined with a thorough clinical The use of Doppler ultrasonography in studying ocular blood
examination can establish the diagnosis of choroidal melanoma flow in disease states is increasing; however, at the present time
with enough certainty to recommend treatment, a preoperative this method does not allow exact measurements of maximal flow
correlation between the sonographic pattern and the histopatho- velocities.
logical findings is currently still in debate.40,41 Determining the volume of a choroidal melanoma
B-mode Doppler devices can obtain signals from the interior by ultrasonography
of the tumor by using a high-resolution power and a frequency Ultrasonography produces spikes in the recorded signal when
between 7 and 20 MHz. This method is more sensitive than the interfaces with different media are encountered. In a normal eye,
visual evaluation of a time amplitude echogram46 (Fig. 9.64). In the first acoustic interface after the signal passes through the
some patients it was possible to determine the blood flow within vitreous is the retina. In the case of a choroidal tumor the apex
the tumor using Doppler color-coding technology. In a series of is adjacent to the retina spike and the base is anterior to the
268
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

Fig. 9.63 Choroidal melanoma. (A) Elevation, 6 mm; diameter at the base, 11 × 12 mm; calculated tumor volume, 330 mm3. The highly reflecting
choroid has been replaced by the poorly reflecting melanoma, producing a choroidal excavation in the cross-section. (B) Histologic section
through the margin of a choroidal melanoma. At the base, normal choroid is replaced by tumor. (C) Schematic drawing.

scleral spike. Identifying these positions, however, can be chal-


lenging in some settings. For example, if the retina is attached
to the tumor surface, no separate discrimination of the two inter-
faces is possible (Fig. 9.58). On the other hand, if the accompany-
ing retinal detachment is also present over the peak of the tumor,
then the retina will be imaged as a separate membranous struc-
ture lying in front of the lesion (Fig. 9.66). Then the transition
between tumor and scleral must be identified. In most cases, the
scleral spike is still the strongest signal even in the presence of
a choroidal tumor. If the tumor breaks through the scleral barrier,
the continuity of this strong signal is interrupted at the site of
perforation. An exact measurement of the volume of the tumor
is important when measuring the lesion height, planning a treat-
ment, or analyzing the effect of therapy.
Ultrasonography is capable of providing precise measure-
ments of tumor size. Ultrasound devices have measurement
tools that can be applied in the window of the image. The exam-
iner can use this feature to measure the height of the largest and
Fig. 9.64 B-mode Doppler sonography (duplex technique) of a smallest diameter of the tumor. In biconvex tumors, volume data
choroidal melanoma of average size. In A-mode the occasionally and tumor can be calculated from these linear measurements or
observed flickering amplitudes are produced by the blood flow. They
can be quantified when using a B-mode Doppler instrument of high it can be evaluated using special equipment designed for 3D
resolution. evaluation (Fig. 9.67).
269

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

D E

Fig. 9.65 Choroidal melanoma with orbital invasion in a 65-year-old woman. Referral diagnosis was orbital cellulitis. Echographic findings include
a large intraocular tumor with wide invasion of the orbital tissues (A). Remnants of the sclera can be demonstrated within the tumor in both
A- and B-mode echograms because of their strong acoustic reflectivity (B). Additional findings include a total retinal detachment as seen in both
images. (C) Schematic drawing of the echogram. (D,E) Doppler signal of the choroidal tumor. (D) Frequency shift from the intraocular tumor,
maximum velocity: 10 cm/s. (E) Frequency shift from the extraocular tumor, maximum velocity: 2 cm/s. With Doppler technique the intraocular
part of the tumor shows reduced blood flow compared to the extraocular part.
Fig. 9.66 Choroidal melanoma with associated retinal detachment over
0 5 10 15 20 25 30 the peak of the tumor. Tumor size: base, 9 × 9 mm; elevation, 3.5 mm.
The detached retina lies about 2 mm in front of the tumor. The
270 calculated tumor volume is about 150 mm3. If the retinal detachment
were erroneously included, the volume would be about 230 mm3.
10
5
Section 2

0 0
Retinal Diagnostics
Retinal Imaging and Diagnostics

5
10

0 5 10 15 20 25 30

A B

3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
0
150
50
100
100
col
50
150
0

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5


Apex height scale (mm)
C Surface plot

Fig. 9.67 Example of three-dimensional rendering of the posterior segment tumor. (A) Area of interest marked. (B) Contour-finding procedure
with outlining of the space-occupying lesion. (C) Surface plot for volume determination.
The role of ultrasonography for planning the treatment of Serial examination with ultrasonography posttreatment can
choroidal melanomas be used to monitor the effect of the radiation plaque by mea-
After a choroidal melanoma is diagnosed by careful clinical suring the tumor elevation, tumor base, and reflectivity of 271
examination and ultrasound interpretation, the next step for the internal tumor structure as well as by identifying the
the physician is to formulate a treatment plan. Radiation therapy presence of tumor vessels. Figure 9.71 illustrates the successful
is successfully used for the treatment of medium or small ocular

Chapter 9
melanomas. Scleral contact radiotherapy using ruthenium-l06,
cobalt-90, or iodine-125 applicators shows a relatively steep
decay of radiation dosage. At a distance of 8  mm from the
radiation surface, only 10% of the energy is still available. In Depth in tissue (mm)
order to achieve complete tumor necrosis in this area, an appli-

7
6
cation of 10 times the duration of radiation is needed. Figure

Diagnostic Ophthalmic Ultrasound


5
10%

4
9.68 shows the isodose curves for ruthenium-106 applicators.

3
The aim of any radiation therapy is to destroy the tumor tissue

2
1
30%
and to spare the adjacent normal ocular tissues as much as
possible. The radiation pellets are placed on custom scleral 50%

plaques that are designed to cover the full extent of the tumor. 70%

It is therefore important to have an exact measurement of the 90%

size of the tumor and have precise placement of the scleral


plaque over the tumor (Figs 9.69 and 9.70). Ultrasonography
provides the most accurate measurements for this purpose.
Dynamic imaging using the ultrasound B-mode can also provide Fig. 9.68 Measuring protocol for a ruthenium applicator type CCC;
information on adjacent pathology such as overlying retinal diameter of the emitting plane, 21 mm. At 5 mm from the applicator
surface, only 25% of the scleral contact dose is still available. Because
detachments, which may overestimate the total height of the of this steep decay of effective radiation, it is critical to plan the
tumor if not properly identified. treatment exactly.

A B

Fig. 9.69 (A,B) Echogram of an eye with the ruthenium-beta applicator in place. The slit between the posterior scleral surface and the applicator
is filled with fluid and is less than 0.5 mm wide. The lateral extent of the applicator is marked by the sound shadow. (C) Schematic drawing.
272
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

Fig. 9.70 (A) Ruthenium-beta applicator in place with a wide slit between the sclera and the applicator surface. On the basis of this echographic
finding, the position of the applicator should be changed or the duration of the treatment has to be prolonged. Distance of the measuring marks:
1.9 mm. (B) Schematic drawing.

A B C D

Fig. 9.71 A-mode (bottom) and B-mode (top) echograms during follow-up assessments of a successfully treated choroidal melanoma. The tumor
volume was reduced from 380 to 0 mm3 within 10 months. After 8 months the spikes within the tumor increased from about 40% of the scleral
spike to about 90% (white arrows). (A) Before radiation, 380 mm3, reflection 40%; (B) after 4 months, 230 mm3, reflection 80%; (C) after 8
months, 100 mm3, reflection 90%; (D) after 10 months, 0 mm3, no reflection.
treatment of a choroidal melanoma that was documented Determining blood flow by B-mode Doppler sonography is an
echographically. In this case, after 4 months there was a additional parameter that could be used to monitor the treat-
marked increase in reflectivity in the internal tumor structures. ment response in highly vascularized choroidal tumors, such as 273
Within 10 months the volume was reduced from 380 mm3 choroidal melanomas. Doppler can detect a decrease in blood
to 0 mm3. flow in the necrotic mass that is left after radiation therapy. The

Chapter 9
In some cases of successfully treated tumors, the increased necrotic tissue from treated tumors can be absorbed by the adja-
reflectivity within the tumor is the only ultrasonographic cent blood vessels, but radiation may damage these vessels and
parameter that changes after radiotherapy (Fig. 9.72). prevent the clearing of these products, which explains the resid-
Increased reflectivity may demonstrate tumor necrosis; ual mass on ultrasound seen after treatment with radiation: this
however, this may not always be the case, as some tumors mass is called tumefaction. This may explain the increased
remain viable even with the increase in reflectivity noted on reflectivity of treated tumors. Figure 9.73 shows an elevated
ultrasound. tumor before radiation.

Diagnostic Ophthalmic Ultrasound


Fig. 9.72 A-mode (bottom) and B-mode (top)
echograms demonstrating the effect of
brachyradiotherapy on a choroidal
melanoma. The external shape of the tumor
also remains unchanged after a second
series of radiation; the reflectivity of the
tumor increases from about 20% of the
scleral spike before radiation to about 80%
after radiation (white arrows). (A) Before
radiation, volume 250 mm3, reflection 20%;
(B) after 8 months, 250 mm3, reflection 70%;
(C) after 36 months and 22 months after
second radiation, volume 250 mm3, reflection
80%.

A B C

A B

Fig. 9.73 (A) Choroidal melanoma with an elevation of 5 mm. Cross-sectional echogram obtained with the duplex instrument ATL mark 8. The
sample volume is identified by the marks on the aiming beam. In this area Doppler spectra are obtained, indicating a high blood flow velocity
(represented in the lower part of the illustration). (B) Same patient as in panel A, 4 months after radiation treatment of the choroidal melanoma
with ruthenium. There is still a considerable volume of tumor remaining (maximal elevation, about 3.5 mm). No frequency shift can be obtained
from the interior of the tumor when using duplex examination. The tissue lying in the guiding beam produces only a biphasic noise (illustrated in
the lower part of the image).
Metastatic choroidal tumors Typically carcinoma metastasis presents as a large, highly
Choroidal metastasis is a known complication of several cancers, reflective thickening of the ocular outer layers. The internal
274 including all types of carcinomas and sarcomas. The most acoustic properties can resemble a disciform macular degen-
frequent primary tumors are breast (40%) and lungs (29%). eration or a choroidal hemangioma. A metastatic adenocarci-
One study examined 230 eyes from deceased patients with noma typically presents with high reflectivity from the strong
acoustic interfaces from its adenoid-like histological structure
Section 2

known systemic carcinoma and showed that 12% of the eyes


had choroidal metastasis on pathological specimens.47 Many (Fig. 9.74).
of these tumors remained undetected at the time of death. In contrast, there are several atypical presentations of choroi-
The detection of such lesions can be a poor prognostic factor. dal metastasis that have also been reported.49,50 Pathological
The average survival time after treatment for choroidal metas- examination of an eye that was enucleated because of suspicion
tasis is 7.4 months.48 Therefore, many of these patients are for choroidal melanoma from ultrasound examination revealed
a choroidal metastasis from small-cell bronchial carcinoma
Retinal Diagnostics

not followed for extended periods of time by their


Retinal Imaging and Diagnostics

ophthalmologist. (Fig. 9.75).49

NH S

C B

Fig. 9.74 Extensive choroidal metastatic tumor in the lower nasal quadrant. (A) On the echogram the choroid is widened to about 2.5 mm; the
retina is partly detached by an exudate. The tissue inside the metastatic tumor shows high acoustic reflectivity. (B) Histologic section of a
metastatic adenocarcinoma in the choroid. The gland-like structure provides good acoustic interfaces. NH, retina; S, sclera. (C) Schematic
drawing.
Fig. 9.75 Histological metastatic tumor from a small-cell bronchial
carcinoma. The echogram (not shown) resembled that of a typical
choroidal melanoma. Similar findings have been seen in choroidal
metastases from cutaneous melanoma. NH, retina; S, sclera. 275

Chapter 9
Diagnostic Ophthalmic Ultrasound
NH

Choroidal hemangioma Choroidal osteoma – metastatic calcifications


Choroidal hemangioma may be an isolated lesion or may be Choroidal osteoma is a rare condition which is characterized
associated with Sturge–Weber syndrome. Isolated hemangio- ultrasonographically by a localized area of high reflectivity at
mas, described as circumscribed choroidal hemangiomas, the outer wall (Fig. 9.78). Several conditions can mimic the
usually occur at the posterior pole. The lesions are typically appearance of a choroidal osteoma. As mentioned above, a
diffuse, slightly elevated, and may have indistinct margins. chronic circumscribed hemangioma can ossify and resemble a
These features make ophthalmic interpretation of the lesion choroidal osteoma.53–55 In addition, several choroidal lesions can
difficult and often they can be missed. However, associated contain calcium, including ocular metastasases of the choroid
features of these circumscribed lesions can include retinal and the sclera51 or metabolic disorders of calcium metabolism
detachment and secondary changes of the RPE, which makes that create a localized calcium deposition within the choroid
their presence more conspicuous on exam. Ultrasonographi- known as an osseous choristoma (Fig. 9.79).54
cally, choroidal hemangiomas appear as a strongly reflecting,
nearly concentric widening of the outer layers of the eye (Fig. Choroidal tuberculoma
9.76). In spite of the abundant vascularization, ultrasound Choroidal tuberculomas are another extremely rare choroidal
images of choroidal hemangiomas do not show the circulating lesion (Fig. 9.80).56 Tuberculomas can also appear similar to
blood as in choroidal melanomas. Instead, the echogram of a choroid melanoma (Figs 9.81). Images that are suggestive of
choroidal hemangioma is similar to that of metastatic adeno- choroidal tuberculomas exemplify the importance of placing the
carcinoma or disciform macular degeneration, which may be ultrasound image in context with the rest of examination of the
indicative of a slower circulation more consistent with laminar patient in order to make the proper diagnosis. Tuberculomas are
flow in the cavities of dilated blood vessels shown by Doppler found in patients with disseminated tuberculosis and a complete
ultrasonography (Fig. 9.77). In long-standing cases the epicho- examination can identify other tuberculomas to confirm the
roidal layer may ossify and produce sound shadows. In this diagnosis. Laboratory confirmation of mycobacterium by blood
stage, a hemangioma may appear identical to an osteoma or a or sputum sampling is also helpful. In addition, these lesions
metastatic calcification of the choroid.51,52 should respond to antituberculous treatment.
276
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C D

Fig. 9.76 Extensive choroidal hemangioma in Sturge–Weber syndrome (additional finding: total retinal detachment). (A) Cross-sectional
echogram through the area of the optic nerve head. In the lower quadrant, the ocular walls are markedly thickened. (In spite of the associated
retinal detachment, the intraocular pressure was 50 mmHg by applanation.) (B) With reduced sensitivity, the scleral surface can be delineated in
spite of the high acoustic reflectivity within the tumor (distance of the measuring marks, 3.1 mm). (C) Histologic section through the tumor
illustrated in (A) and (B): the septa of the cavities are thin and lined with endothelium. They produce strong reflectivity within the lesion.
(D) Schematic drawing.
277

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.77 (A) Cross-sectional echogram of a circumscribed choroidal


hemangioma in a 12-year-old boy. The tumor has an elevation of
4 mm. (B) In an A-mode echogram with S-shaped amplification the
cross-sectional picture corroborates the high reflectivity of the tumor.
C (C) On Doppler sonography, we see blood circulating within the tumor,
synchronous with the pulse. The illustrated velocity profile speaks for
a relatively low resistance by the blood vessels.

A B

11mm

C D

Fig. 9.78 Cross-sectional echogram of the choroidal osteoma. (A) The echogram of the lesion is characterized by total reflection of the
ultrasound, and shadow formation. An elevation cannot be unequivocally documented. (B) With reduced amplification it is possible to image the
ossification as an isolated signal. The horizontal diameter is 11.0 mm. (C,D) Schematic drawings.
278
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

B
C

Fig. 9.79 Metastatic calcification of the ocular walls of unknown etiology. Differential diagnosis includes choroidal osteoma. (A) A computed
tomography scan was obtained because of nonophthalmological problems. A circumscribed calcified structure within the ocular walls was found
incidentally. (B) In cross-sectional echograms, a lesion of the ocular walls above the optic nerve head showed total reflection of the ultrasound
signal. The lateral extension was about 7.5 mm. There was no corresponding lesion seen on fundus examination that would be typical of an
osteoma. The etiology of this lesion is unknown. (C) Schematic drawing.

Fig. 9.80 Fundus picture of a 52-year-old


woman with active pulmonary tuberculosis.
This lesion resolved with antituberculosis
treatment.
279

Chapter 9
Diagnostic Ophthalmic Ultrasound
A B

Fig. 9.81 (A,B) Echogram of the fundus lesion shown in Fig. 9.80.
(C) The lesion is characterized by low acoustic reflectivity. Tenon’s
space can be well demonstrated. The reflectivity corresponds to that of
C a choroidal melanoma. The infiltration of Tenon’s space points toward
inflammatory etiology.

The uveal effusion syndrome ULTRASOUND IMAGING USED TO


Patients with uveal effusion syndrome can have partial or circu-
lar choroidal detachment combined with an exudative retinal
DIFFERENTIATE OCULAR DISEASE
detachment (Fig. 9.82). Ultrasonography is very important in Ultrasonography is invaluable in the diagnosis of certain ocular
this condition. It can reveal fluid in the suprachoroidal space or and orbital conditions. Proper imaging with ultrasound can
differentiate choroidal hemorrhage or tumor from the serous narrow the differential diagnosis in certain conditions and guide
fluid seen on imaging from choroidal effusion syndrome.49,57 further workup and management. These conditions include cho-
roidal folds (Tables 9.2–9.4 and see Fig. 9.85), leukocoria (Table
Sclera 9.5 and see Fig. 9.86), and vitreous hemorrhage (Table 9.6 and
Posterior scleritis see Fig. 9.87). First, choroidal folds can be from orbital masses,
Ultrasonography is the diagnostic method of choice for posterior ocular inflammation, disc edema in an atypical presentation, or
scleritis.52,58,59 The clinical picture is characterized by an acute from idiopathic causes.49 Next, leukocoria is a condition of a
loss of vision associated with folds at the posterior pole. The white pupil that blocks ophthalmic examination, typically diag-
normal high reflectivity of the sclera will be decreased by nosed in infancy. This finding can be from retinoblastoma. Dif-
changes in the tissue from inflammatory swelling. The result is ferentiating this disease from other benign causes of leukocoria
a low reflective signal from a thickened choroid, which is is critical for providing appropriate treatment of the patient.
suggestive of posterior scleritis. Choroid and vitreous may Finally, vitreous hemorrhage may manifest from a variety of
appear normal. Occasionally, there is a slight exudation into changes in the posterior globe; the urgency of treatment is
the subretinal space with accompanying disc edema. In 50% dependent on the diagnosis of a retinal detachment. All of these
of the patients, fluid accumulates in the Tenon’s space.60 This conditions require ultrasound technology to establish the proper
signal can be similar to a diffuse choroidal melanoma. There is diagnosis. The tables listed above provide information on the
one case report of an eye that was enucleated because of sus- differential diagnosis of each condition and ultrasound features
picion of an ocular melanoma based on the finding of brawny that will help guide image interpretation.
scleral thickening (Figs 9.83 and 9.84). This case highlights the Furthermore, ultrasonography can aid in the preoperative
importance of proper ultrasound interpretation in the clinical planning and patient counselling for surgery. Table 9.7 lists
context of each patient.61 some of the possible ultrasonographic information and
280
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

C
B

Fig. 9.82 Uveal effusion syndrome in a 55-year-old woman. (A) In the axial cross-section, the retina is attached to the optic nerve head. There is
an associated, strongly reflecting choroidal detachment. (B) Cross-section of image in (A). (C) Schematic drawing.

A
B

Fig. 9.83 (A) Brawny scleritis with inflammatory infiltration of Tenon’s space. This disease entity can be clinically and ultrasonographically
identical to a choroidal melanoma. T, Tenon’s space. (B) Schematic drawing. M, muscle.
Table 9.4 Choroidal folds: ultrasonographic findings helpful for
differential diagnosis
281
Diagnosis Ultrasonographic findings

Myositis Thickened extraocular muscles

Chapter 9
Graves
orbitopathy 1

Periorbital Change in the relief of the orbital wall,


space-occupying sound propagation into perinasal
lesions 2 sinuses

Orbital neoplasm 3 Directly evident (it may be difficult to

Diagnostic Ophthalmic Ultrasound


demonstrate a small cavernous
hemangioma because of its high
acoustic reflectivity)

Inflammatory Widening of normal orbital structures,


Fig. 9.84 Macroscopic photo of a brawny scleritis. On the basis of the orbital low acoustic reflectivity, Tenon’s space
ultrasonographic findings and a positive P32 test, the eye was pseudotumor 4 may be demonstrated
enucleated because a melanoma was suspected.
Disc edema 5 Widened dural diameter of the optic
nerve

Table 9.2 Orbital causes of orbital folds49 Axial hyperopia 6 Axial length below 22 mm, ocular walls
concentrically thickened
Diagnosis Number of patients
Ocular Ocular walls concentrically thickened
Graves orbitopathy 11 hypotony 7

Sinusitis 5 Macular Thickening of the ocular walls in the


degeneration area of the macula, high acoustic
Mucocele 2 reflectivity

Hemangioma 6 Scleritis 8 Circumscribed widening of the ocular


walls
Orbital pseudotumor 5 Tenon’s space apparent
For companion schematic drawing, see Fig. 9.85.
Various orbital tumors 8

Unexplained 6

Total 43 1

Table 9.3 Ocular causes of choroidal folds


4
Diagnosis Number of patients

Hyperopic eye 17
3
Macular degeneration 12

Ocular hypotony 12 6

Posterior scleritis 9

Buckling operation 9
7 5
Trauma 6

Intraocular tumor 3 8 2

Miscellaneous 10

Total 78 Fig. 9.85 Choroidal folds: ultrasonographic contribution to the


differential diagnosis (for detail, see Table 9.4).
Table 9.5 Leukocoria: ultrasonographic findings helpful for Table 9.6 Vitreous hemorrhage: ultrasonographic findings helpful
differential diagnosis for establishing the etiology
282
Diagnosis Ultrasonographic findings Diagnosis Ultrasonographic findings

Normal axial length Symptomatic posterior Thickened detached posterior


Section 2

for the patient’s age vitreous detachment 1 hyaloid membrane, occasionally


early retinal detachment
Retinoblastoma 1 Widening of the ocular walls,
extremely high acoustic reflectivity, Recently formed retinal Blood-covered vitreous strands
shadowing effect, atypical findings break with torn vessel 2 converge toward the retinal break;
possible occasionally a high-floating
operculum may be detected
Congenital cataract 2 Increased reflectivity from the
Retinal Diagnostics
Retinal Imaging and Diagnostics

posterior lens surface, vitreous Proliferative retinopathy 3 Strands or membranes extending


space empty, ocular walls normal from the optic nerve head or the
posterior pole, high acoustic
Shortened axial reflectivity
length
Terson syndrome Vitreous opacities in front of the
Retinopathy of In stages IV and V, beginning or (vitreous hemorrhage optic nerve head or behind the
prematurity 3 complete traction detachment after subchoroidal detached vitreous
(normal findings in stages I–III) bleeding 4

Persistent hyperplastic Dense strand of tissue between optic Disciform macular Widening of the ocular walls in the
primary vitreous nerve head and posterior lens pole; degeneration 5 macular area, high acoustic
(PHPV) 4 formes frustes may occur (posterior reflectivity, vitreous strands
or anterior PHPV) extending from the macula

Retinal anomalies Membranes in the vitreous, atypical Choroidal melanoma 6 Biconvex thickening of the ocular
detachment, which in part appears wall, low acoustic reflectivity,
solid (no typical echogram) sometimes mushroom-shaped;
accompanying retinal
Fundus coloboma 5 Directly demonstrable protrusion of detachment distant from the
ocular wall, sometimes with orbital tumor
cyst (microphthalmos with cyst) For companion schematic drawing, see Fig. 9.87.

Coats disease 6 Floating crystals in the vitreous and


subretinal space (fast-flickering
spikes on A-mode)
For companion schematic drawing, see Fig. 9.86.

2
3
1 3
4

6
2
5

1
6

Fig. 9.86 Leukocoria: echographic contribution to the differential Fig. 9.87 Vitreous hemorrhage: echographic contribution to determine
diagnosis (for detail, see Table 9.5). the pathogenesis (for detail, see Table 9.6).
Table 9.7 Examination before a vitrectomy: ultrasonographic 1
findings helpful for planning the operation 3
283
Questions on the Consequences for planning
ultrasonographic examination the operation 7

Chapter 9
Is the choroidal detachment Avoiding this area when
including the pars plana? 1 inserting the ports

Is the posttraumatic vitreous If retina is detached, strong


hemorrhage associated with a indication for vitrectomy 5
detached retina?
8
Is an intraocular foreign body Choice of surgical approach,

Diagnostic Ophthalmic Ultrasound


demonstrable? If so, where is magnet extraction possible?
it in regard to ocular outer
wall? 2
4
Estimating the prognosis

Is there a choroidal Extremely poor prognosis 6


detachment caused by blood? 3 that may not benefit from
surgical intervention 2
9
Is there a free-floating retinal Reattachment probable, can Fig. 9.88 Examination before a vitrectomy: echographic contribution to
detachment? 4 be achieved without the planning of the operation (for detail, see Table 9.7).
tamponade from the inside

Is there a rigid retinal Removal of preretinal


detachment? Is there a membranes necessary In the field of ophthalmology new Doppler modes for analyzing
thickened retina? 5 retinal blood flow are currently being utilized in research initia-
tives. A new technique is elastographic imaging. Recently, this
Are there vitreous strands with Consider intraoperatively
traction effect demonstrable? 6 cutting the strands technique has been used to image ocular and periocular tissues.
Patterns of elastic imaging in the vitreous cavity could be attrib-
Is there retinal detachment Special care indicated when uted to posterior vitreous detachment, whereas that of medial
that is displaced anteriorly to entering the vitreous space and lateral rectus muscles may be related to the level of muscle
be near the lens? 7
fiber strain. With improved resolution, higher frequencies, and
Are there thickened ocular Poor prognosis for central spatial images on ultrasound, new frontiers in ophthalmic
walls in the macula that may vision imaging are being reached and further research is still under
indicate a disciform lesion way. These advancements as well as the safety in image acquisi-
from macular degeneration? 8
tion maintain the utility of ultrasound imaging within all fields
Are there indications of a Additional diagnostic of medicine.
secondary, e.g., solid retinal examinations; enucleation For online acknowledgments visit http://www.
detachment? 9 may be necessary expertconsult.com
Is there attached retina with Reconsider indication for REFERENCES
minimal remaining vision? operation, prognosis for 1. Mundt G, Hughes W. Ultrasonics in ocular diagnosis. Am J Ophthalmol
vision extremely poor 1956;41:488.
2. Baum G, Greenwood J. The application of ultrasonic locating techniques to
For companion schematic drawing, see Fig. 9.88. ophthalmology, part 1. Am J Ophthalmol 1958;46:319.
3. Oksala A, Lehtinen A. Diagnostic of detachment of the retina by means of
ultrasound. Acta Ophthalmol 1957;35:461–7.
4. Oksala A, Lehtinen A. Diagnostic value of ultrasonics in ophthalmology.
subsequent conclusions that can be obtained for operative plan- Ophthalmologica 1957;134:387–95.
5. Oksala A, Lehtinen A. Diagnostic of rupture of the sclera by means of
ning (and see Fig. 9.88). ultrasound. Acta Ophthalmol 1958;36:37–42.
6. Oksala A, Lehtinen A. Measurement of the velocity of sound in some parts of
FUTURE DEVELOPMENTS the eye. Acta Ophthalmol 1958;36:633–9.
7. Ossoinig K. Standardized echography: basic principles, clinical applications
Many medical fields are using advances in technology to improve and results. Int Ophthalmol Clin 1979;19:127.
8. Buschmann W, Haigis W. Influence of equipment parameters on results in
ultrasound images. These improvements involve the use of new ophthalmic ultrasonography. Doc Ophthalmol Proc Ser 1981;29:487.
piezoelectric materials and broadband transducers.62 Current 9. Ossoinig K, Till P. 10 years’ study on clinical echography in orbital disease.
Bibl Ophthalmol 1975;83:200.
and future developments can be focused on the following trends: 10. Gernet H. Ultrasonic biometry of the eye. Klin Monatsbl Augenheilkd 1967;
151:853–71.
• Advances in the signal transmission process 11. Baum G, Greenwood J. Ultrasound in ophthalmology. Am J Ophthalmol
1960;49:249–61.
• Advances in the signal reception process 12. Bronson N, Fisher Y, Pickering N, et al. Ophthalmic contact B-scan ultrasonog-
• Compounding, equalization, and extended-field acquisitions raphy. Westport, CT: Intercontinental; 1976.
13. Pavlin C, Foster F. Ultrasound biomicroscopy of the eye. New York: Springer;
• 3D and 4D imaging 1995.
• Elastographic imaging 14. Sherar M, Starkowski B, Taylor W, et al. A 100 Mhz B-scan ultrasound backscat-
ter microscope. Ultrasound Imaging 1989;11:95–105.
• Miniaturization of units and probes, transfer, and archiving 15. Pavlin C, Harasiwicz K, Sherar M, et al. Clinical use of ultrasound biomicros-
ultrasonic data. copy. Ophthalmology 1991;98:287–95.
ACKNOWLEDGMENTS
A special thank you to a number of colleagues who have been 283.e1
of great assistance in creating this chapter: Sönke Langner,
Kirsten Franke, Beate Stroteich, Angrit Stachs, and Enrique
Pfeiffer.

Chapter 9
Diagnostic Ophthalmic Ultrasound
16. Sherar M, Foster F. The design and fabrication of high frequency transducer. 38. Buschmann W, Trier H. Ophthalmologische Ultraschalldiagnostik, mit Atlas,
Ultrasound Imaging 1989;11:75–94. Standardisierung und Einordnung in den ophthalmologischen Untersuchun-
17. Pavlin C, Harasiwicz K, Foster F. Ultrasound biomicroscopy of anterior gsbefund. Berlin: Springer; 1989.
284 segment structures in normal and glaucomatous eyes. Am J Ophthalmol 39. Trier H. Gewebsdifferenzierung mit Ultraschall. Bibl Ophthalmol 1977;86:92.
1992;113:381–9. 40. Coleman DJ, Silverman R, Rondeau MJ, et al. Explaining the current role of
18. Atta HR. New applications in ultrasound technology. Br J Ophthalmol 1999; high frequency ultrasound in ophthalmic diagnosis (ophthalmic ultrasound).
83:1246–9. Expert Rev Ophthalmol 2006;1:63–76.
19. Iezzi R, Rosen R, Tello C, et al. Personal computer-based 3-dimensional ultra- 41. Silverman R, Kong F, Chen Y, et al. High-resolution photoacoustic imaging of
Section 2

sound biomicroscopy of the anterior segment. Arch Ophthalmol 1996;114: ocular tissues. Ultrasound Med Biol 2010;36:733–42.
520–4. 42. Guthoff R. Ultraschall in der ophthalmologischen Diagnostik. In: Naumann
20. Cusumano A, Coleman D, Silverman R, et al. Three dimensional ultrasound GOH, Hollwich F, Gloor B, editors. Ein Leitfaden für die Praxis. Stuttgart:
imaging - clinical applications. Ophthalmology 1998;105:300–6. Enke-Verlag; 1988.
21. Coleman D, Silverman R, Daly S. Advances in ophthalmic ultrasound. Radiol 43. Wolter J. Parallel horizontal choroidal folds secondary to an orbital tumor.
Clin North Am 1998;36:1073–82. Am J Ophthalmol 1974;77:669.
22. Reinstein D, Raevsky T, Coleman D. Improved system for ultrasonic imaging 44. Coleman D, Lizzi S, Jack R. Ultrasonography of the eye and orbit. Philadelphia:
and biometry. J Ultrasound Med 1997;16:117–24. Lea and Febiger; 1977.
Retinal Diagnostics

23. Silverman R, Lizzi F, Ursea B, et al. High-resolution ultrasonic imaging 45. Bujara K, von Domarus D, Guthoff R. Necrotic malignant melanoma of the
Retinal Imaging and Diagnostics

and characterization of the ciliary body. Invest Ophthalmol Vis Sci 2001;42: choroid with unusual clinical, echographical and histological case study.
885–94. Ophthalmologica 1980;180:222–7.
24. Stachs O, Martin H, Behrend D, et al. Three-dimensional ultrasound biomicros- 46. Guthoff R, Berger R, Helmke K, et al. Doppersonographische Befunde bei
copy, environmental and conventional scanning electron microscopy investi- intraokulären Tumoren Forstschr Ophthalmol 1989;86:239.
gations of the human zonula ciliaris for numerical modelling of accommodation. 47. Bloch RS, Gartner S. The incidence of ocular metastatic carcinoma. Arch
Graefes Arch Clin Exp Ophthalmol 2006;244:836–44. Ophthalmol 1971;85:673.
25. Stachs O, Martin H, Kirchhoff A, et al. Monitoring accommodative ciliary 48. Ferry AP, Font RI. Carcinoma metastatic to the eye and the orbit: a clinical
muscle function using three-dimensional ultrasound. Graefes Arch Clin Exp pathological study of 227 cases. Arch Ophthalmol 1974;92:276.
Ophthalmol 2002;240:906–12. 49. Verbeek A. Echographic findings in 36 patients with choroidal folds. Doc
26. Kirchhoff A, Stachs O, Guthoff R. Three-dimensional ultrasound findings Ophthalmol Proc Ser 1981;29.
of the posterior iris region. Graefes Arch Clin Exp Ophthalmol 2001;239: 50. Manschot WA. The relation between histopathological and ultrasonogra-
968–71. phyin intraocular tumors. Doc Ophthalmol Proc Ser 1981;29:71.
27. Stachs O, Schneider H, Stave J, et al. Potentially accommodating intraocular 51. Shields CL, Shields JA, Augsburger JJ, editors. Update on choroidal osteomas.
lenses – an in vitro and in vivo study using three-dimensional high-frequency Proceedings of the 2nd international meeting on the diagnosis and treatment
ultrasound. J Refract Surg 2005;21:37–45. of intraocular tumors; Lyons; 1987.
28. Schneider H, Stachs O, Göbel K, et al. Changes of the accommodative ampli- 52. Wende S, Aulich A, Nover A, et al. Computer tomography of orbital lesions:
tude and the anterior chamber depth after implantation of an accommodative a cooperative study of 210 cases. Neuroradiology 1978;13:123.
intraocular lens. Graefes Arch Clin Exp Ophthalmol 2006;244:322–9. 53. Gass JDM, Guerry RK, Jack RI, et al. Choroidal osteoma. Arch Ophthalmol
29. Jaffe NS. Complications of acute posterior vitreous detachment. Arch 1978;96:428.
Ophthalmol 1968;79:568–71. 54. Gass JDM. New observations concerning choroidal osteomas. Int Ophthalmol
30. McLeod D. (chair) Round table discussion on vitreous pathology. Doc 1979;71.
Ophthalmol Proc 1981;Ser. 547. 55. Wing GI, Schepenz CL, Trempe CL, et al. Serous choroidal detachment and the
31. Ossoinig KC. Advances in diagnostic ultrasound. Acta 14th Int Congr thickened choroidal sign detected by ultrasonography. Am J Ophthalmol
Ophthalmol 1983:89. 1982;94:499.
32. Till P, Ossoinig KC. 10 years’ study on clinical echography in intraocular 56. Blodi FC. Ein Tuberkulom der Aderhaut, ein melanom vortäuschend. Klin
disease. Bibl Ophthalmol 1975;83:49. Monatsbl Augenheilkd 1977;170:845.
33. Machemer R, Aalberg TM, editors. Glaskörperchirurgie, Vitrektomie. 57. Wirold J, Orlowski-Szczypinski J. Ultrasonography of the subretinal space in
Indikationen und Technik. Berne: Huber Verlag; 1981. rhegmatogeneous retinal detachment. Z Mod Probl Ophthalmol 1976;18:40.
34. Shapiro DR, Stone RD. Ultrasonic characteristics of retinopathy of prematurity 58. Cappaert W, Purnell E, Frank K. Use of B-scan ultrasound in the diagnosis of
presenting with leucokoria. Arch Ophthalmol 1885;103:1690. benign choroidal folds. Am J Ophthalmol 1977;84:375.
35. Darnley-Fisch DD, Frazier-Byrne S, Hughes JR, et al. Contact B-scan echogra- 59. Singh G, Guthoff RF, Foster S. Observations on long-term follow-up of
phy in the assessment of optic nerve cupping. Am J Ophthalmol 1990; posterior scleritis. Am J Ophthalmol 1986;101:570.
109:55. 60. Guthoff RF. Die differentialdiagnostische Bedeutung des Tenon’schen Raumes.
36. Oksala A. Echogram in melanoma of the choroid. Br J Ophthalmol 1959; Fortschr Ophthalmol 1974;81:388.
43:408. 61. Feldon S, Sigelman J, Albert D, et al. Clinical manifestations of brawny scleritis.
37. Baum G. Use of ultrasonography in the differential diagnosis of ocular tumors. Am J Ophthalmol 1978;85:781.
In: Boniuk M, editor. Ocular and adnexal tumors. St Louis: Mosby; 1964. 62. Claudon M, Tranquart F, Evans DH, et al. Advances in ultrasound. Eur Radiol
p. 308. 2002;12:7–18.
Retinal Diagnostics Section 2
For additional online content visit http://www.expertconsult.com

Color Vision and Night Vision Chapter

Dingcai Cao
10 
OVERVIEW ROD AND CONE FUNCTIONS
Day vision and night vision are two separate modes of visual Differences in the anatomy and physiology (see Chapters 4,
perception and the visual system shifts from one mode to the Autofluorescence imaging, and 9, Diagnostic ophthalmic ultra-
other based on ambient light levels. Each mode is primarily sound) of the rod and cone systems underlie different visual
mediated by one of two photoreceptor classes in the retina, i.e., functions and modes of visual perception. The rod photorecep-
cones and rods. In day vision, visual perception is primarily tors are responsible for our exquisite sensitivity to light, operat-
cone-mediated and perceptions are chromatic. In other words, ing over a 108 (100 millionfold) range of illumination from near
color vision is present in the light levels of daytime. In night total darkness to daylight. Cones operate over a 1011 range of
vision, visual perception is rod-mediated and perceptions are illumination, from moonlit night light levels to light levels that
principally achromatic. Under dim illuminations, there is no are so high they bleach virtually all photopigments in the cones.
obvious color vision and visual perceptions are graded varia- Together the rods and cones function over a 1014 range of illu-
tions of light and dark. Historically, color vision has been studied mination. Depending on the relative activity of rods and cones,
as the salient feature of day vision and there has been emphasis a light level can be characterized as photopic (cones alone
on analysis of cone activities in color vision. Night vision has mediate vision), mesopic (both rods and cones are active), or
historically been studied in terms of rod activity and consider- scotopic (rods alone mediate vision).1 In the literature, the terms
ations of the shift from day vision to night vision. photopic vision and scotopic vision are used to reflect cone and
This chapter will review basic aspects of rods and cones rod vision, respectively. Table 10.1 shows this overlapping range
and neural pathways that process rod and cone information. of activity.
Measurement of sensitivity during dark adaptation is discussed The distribution of rods and cones in the retina (see Chapter
as the established measure of the shift between day vision 4, Autofluorescence imaging) is also reflected in visual function.
(cone vision) and night vision (rod vision). Clinical assessment The greatest sensitivity to light occurs in the midperiphery of
of rod and cone sensitivities using dark adaptation function the visual field, which has a predominance of rods, while high-
as a means of assessing retinal disease is also discussed. Color acuity and good color vision are mediated by the fovea, which
vision is discussed in terms of experimental paradigms and has a predominance of cones. Nonetheless, the entire retina,
theoretical considerations and variations in human color vision with the exception of a very small area within the fovea, is
are described. Evaluation of color vision can be helpful in capable of mediating night vision, and color vision is present
understanding the underlying mechanisms of some retinal throughout the visual field with daylight stimulation of the
diseases and suggestions for clinical evaluation of color vision entire retina. The following will introduce rod and cone dif-
are offered. In the last section of this chapter, new develop- ferences in light adaptation, spectral sensitivity, and spatial/
ments in color vision research are discussed. temporal sensitivity.

Table 10.1 The dynamic range of the human visual system

Visual Starlight Moonlight Indoor lighting Sunlight


environments

Photopic luminance -6 -4 -2 0 2 4 6 8
(log cd/m2)

Light category Scotopic Mesopic Photopic

Photoreceptors Rods only Rods and cones Cones only

Visual function Rod absolute threshold Cone absolute threshold Rod saturation begins Damage possible

No or poor color vision Good color vision


Modified from Hood DC, Finkelstein MA. Sensitivity to light. In: Boff KR, Kaufman L, Thomas JP, editors. The handbook of perception and human performance,
vol. 1. Sensory processes and perception. New York: John Wiley; 1986.
Light adaptation
Photoreceptors, whether they are rods or cones, respond well
286 to only a small range of variations in illumination within a

Log increment threshold (scotopic trolands)


4 Rod mechanism
steady adapting background.2 However, adaptation mecha-
9 degree extra- Rods saturated
nisms adjust photoreceptor sensitivity so that this small range 3 foveal vision
Section 2

of responses is always centered near the current adaptation


level, even though adaptation levels can vary over a wide range. 2
1.00

Fraction of pigment bleached


This behavior forms the basis for the large operating range of
1
the visual system.
Π
It is possible to measure a threshold for the perception of ∞ 0.75
0
an increment in light on a large, steady background field. As
Retinal Diagnostics
Retinal Imaging and Diagnostics

the background light level is increased, the increment threshold –1 0.50


starts to increase. Rods and cones behave somewhat differently Scale
in this regard. For the rod system, as shown in Fig. 10.1A, the –2
Scale 0.25
increment threshold increases steadily over almost a thousand-
–3
fold range. With further increases in background adaptation 0
levels, an increment is not detected, no matter how much addi- –4 –2 0 2 4 6 8 10
tional test light is presented as an increment, due to rod satu-
Log adapting field retinal illuminance
ration. In comparison, the cone system, as shown in Fig. 10.1B, A (scotopic trolands)
shows a continuous steady increase in the increment threshold
with increases in background illumination, even at levels that
bleach almost the entire amount of available photopigment.
The portion of the curve that rises linearly with illumination
levels is called the Weber region (Fig. 10.1). In the Weber region,
5
an incremental light can be detected when it is a constant t.v.r.
proportion (i.e., the Weber fraction) of the background light Fovea
level. Different photoreceptor systems have a characteristic 15m 578nm
Weber fraction. Cones have lower Weber fractions than rods 4
Relative log increment threshold

Fraction bleached
and the M and L cones have lower Weber fractions than S
cones. Under optimal conditions, the cone system can detect
a light level difference of 1%, while rods need a light change 3
of 20%.
In addition to photoreceptor adaptational properties, other
factors, including pupil size, the temporal and spatial summa- 2 1.00
tion characteristics, and photopigment depletion, can also con-
tribute to extend the operating range of the visual system over 0.75
a large luminance range. While some adaptation operates within
1 0.50
the photoreceptors themselves, other properties of adaptation
may reflect the effects of the complex neural circuitry of the 0.25
retina.2
0 2 4 6 8
Spectral sensitivity Log background retinal illuminance
Day vision is primarily mediated by three types of cone photo- (photopic trolands)
receptors with different but overlapping spectral sensitivities.
B
Each is identified by the relative position of the peak in spectral
sensitivity. The three cone types are called the long-, middle-, Fig. 10.1 The increment threshold functions for scotopic (rod) and
photopic (cone) vision as a function of background illuminance. (A) Rod
and short-wavelength-sensitive (L, M, and S) cones. When increment thresholds measured at 9° in the parafovea. The dashed line
overall sensitivity to light is measured at the light-adapted fovea, has a slope of 1. The portion where the curve has unit slope (in parallel
a broad sensitivity spectrum peaking near 555 nm is found. This to the dashed line) is the Weber region, followed at higher levels by the
sensitivity spectrum represents the combined activity of the L region of rod saturation. To the right is shown the fraction of rod
photopigment bleached. The rods are saturated before there is
and M cones and is called the V(λ) function. When sensitivity to substantial photopigment bleaching. (B) Cone increment thresholds
light is measured in the dark-adapted peripheral retina, where measured at the fovea. To the right is shown the fraction of
rods dominate, a broad-sensitivity spectrum is found with a photopigment bleached. The Weber region extends to luminances
(6 million tds) that bleach virtually all the cone photopigment.
peak sensitivity at 507 nm. This rod spectral sensitivity function
(Reproduced with permission from Enoch JM. The two-color threshold
is called V’(λ) (Fig. 10.2A). Both V(λ) and V’(λ) functions have technique of Stiles and derived component color mechanisms. In:
practical significance and have been accepted by the Interna- Jameson D, Hurvich L, editors. Visual psychophysics: handbook of
tional Commission of Illumination as representative of human sensory physiology, vol. VII/4. Berlin: Springer-Verlag; 1972.)
vision relative luminous efficiency at photopic and scotopic
levels. They are also used to relate luminous (perceived energy
of light) to radiant (emitted light) energy.
Photopic and scotopic luminosity functions Cone spectral sensitivity functions 287
Scotopic: V’(λ) Photopic: V(λ) S M L
1.00 1.00

Chapter 10
0.75 0.75
Relative sensitivity

Relative sensitivity
0.50 0.50

Color Vision and Night Vision


0.25 0.25

0.00 0.00
400 450 500 550 600 650 700 400 450 500 550 600 650 700
Wavelength (nm) Wavelength (nm)
A B

Fig. 10.2 Spectral sensitivities of cones and rods. (A) The relative spectral luminous efficiency functions for scotopic and photopic vision adopted
by the Commission International d’Eclairage (CIE), V ’(λ) and V(λ) respectively. (Data from Wyszecki G, Stiles WS. Color science – concepts and
methods, quantitative data, and formulae, 2nd edn. New York: John Wiley; 1982). (B) Spectral sensitivities of the S, M, and L cones derived from
color-matching function.14

Estimates of the spectral sensitivities of the three cone VISUAL PATHWAYS FOR ROD AND
types have been obtained from a variety of psychophysical pro-
CONE FUNCTIONS
cedures. One approach was to derive the spectral sensitivities of
cones from analysis of color-matching data. Another approach Retinal pathways
used spectral bleaching lights to depress the responses of two Rod signals are conveyed by two primary neural pathways that
cone types relative to the third so that measurements of the spec- are dependent on the illumination level.5 One pathway is via ON
tral sensitivity of the third cone type could be isolated. Figure rod bipolars, AII amacrine cells, and ON and OFF cone bipolars,
10.2B shows the relative spectral sensitivities of the three cone which are all cells in the retina. This is a temporally sluggish
types. The S cones are most sensitive to light near 445 nm, with pathway that mediates rod vision at low scotopic light levels.
sensitivity declining rapidly at longer wavelengths. At 555 nm The second pathway transmits rod information via rod–cone
and longer wavelengths, the S cones are virtually unresponsive gap junctions and ON and OFF cone bipolar cells in the retina.
to light. The M and L cones have overlapping spectral sensitivi- This is a fast pathway that mediates vision at higher scotopic
ties that span the entire visible spectrum. The M cones peak in and mesopic light levels. A third insensitive rod pathway
sensitivity near 543 nm, while the L cones peak near 566 nm. The between rods and OFF cone bipolars has been identified in
differential spectral sensitivity functions of the L, M, and S cones rodents but, thus far, not in primates. The significant point here
provide the foundation of early spectral processing. is that rods and cones share neural pathways and have joint
Spatial and temporal resolution inputs to retinal ganglion cells.

Compared with the cone system, the rod system has poorer
spatial resolution (acuity). For an observer with 20/20 photopic Retinogeniculate pathways
acuity, scotopic acuity would be about 20/200 (10 times worse There are three major neural retinogeniculate pathways in pri-
than photopic acuity). The rod system also has poorer temporal mates that convey retinal information to the visual cortex.6,7 The
resolution, which refers to the ability to perceive a physically pathways are named after the layers of the lateral geniculate
alternating light as steady or flickering in time. The transitional nucleus (LGN) that receive input from distinct types of ganglion
temporal frequency at which the light appears from flickering cells and project to different areas of the primary visual cortex.
to steady is called the critical fusion frequency (CFF). CFFs The MC layer of the LGN receives inputs from parasol ganglion
increase with light adaptation level, reaching a maximum of cells. The MC pathway processes the summed output of the L
20  Hz for rods and 55–60  Hz for cones. This means that and M cones to signal luminance information. The parvocellular
flickering lights can be perceived at higher frequencies in (PC) layer of the LGN receives input from midget ganglion cells.
brighter light conditions. Interestingly, dark-adapted rods can The PC pathway mediates spectral opponency of L and M cones
suppress cone-mediated flicker detection3 and this suppression (discussed later) to signal chromatic information. The koniocel-
mainly occurs in the magnocellular (MC) pathway (explained lular (KC) layer of the LGN, which receives input from small
below).4 bistratified as well as other ganglion cells, detects changes in
Rod bipolar Rod–cone
288 pathway pathway
–1
Rods Cones

Log luminance (cd/m2)


–2
Section 2

Rod bipolars
–3

All amacrine –4

–5
Retinal Diagnostics
Retinal Imaging and Diagnostics

Cone bipolars
–6

MC-, PC-, KC- ganglion cells


0 5 10 15 20 25 30
Time in dark (min)
LGN

Fig. 10.4 The time course of dark adaptation. The range of threshold
Cortex sensitivities of 110 normal observers is shown. The circular points
represent data of the most and least sensitive individuals. The area
enclosed by the dashed lines represents 80% of this population.
Visual perception The cone absolute thresolds are obtained during the cone and rod
plateaus, respectively. (Data from Hecht S, Mandlebaum J.
The relationship between vitamin A and dark adaptation.
JAMA 1939;112:1910–6.)
Fig. 10.3 The schematic diagram of visual pathways carrying rod
and cone inputs for visual perception. MC, magnocellular; PC,
parvocellular; KC, koniocellular; LGN, lateral geniculate nucleus.
minutes (cone plateau, reflecting cone absolute thresholds).
Then, there is a second rapid decrease in thresholds due to sen-
sitivity recovery of the rods, referred to as the rod–cone break,
S-cone signals compared to the sum of the L- and M-cone signals.
to a new plateau that is reached in 40–50 minutes (rod plateau,
These three pathways mediate different aspects of vision, with
reflecting rod absolute thresholds).
the MC pathway mainly carrying out luminance and motion
The shape of the dark adaptation function depends on testing
processing, the PC pathway mainly processing red–green color,
parameters, including retinal location, wavelength, and temp­
acuity, and shape information, and the KC pathway mainly han-
oral and spatial characteristics.1,11 The effects of these parameters
dling blue–yellow color processing.
on dark adaptation curves can be understood by the differences
The sharing of neural pathways between rods and cones
between rods and cones in terms of their distributions, spectral
implies that rods should have input to the MC, PC, and KC
sensitivity, and spatial/temporal resolution characteristics. For
pathways. Indeed, physiological studies have shown that there
instance, because there are only cones in the fovea, dark adapta-
is strong rod input to the MC pathway, but weak input to the
tion measured at the fovea using a small test light reveals a rapid
PC pathway.8 Demonstration of rod input to the KC pathways
monophasic branch attributable to the cones. On the other hand,
is less clear. An earlier study8 did not find rod input to the bi­
because the rod and cone systems have similar absolute sensi-
stratified ganglion cells in the parafovea, while two recent
tivities at long wavelengths, dark adaptation measured with
studies demonstrated strong rod input in the peripheral retina.9,10
long-wavelength lights is monophasic, resembling the cone
Figure 10.3 shows a schematic diagram of the visual pathways
function. As the test wavelength is changed to shorter wave-
conveying rod and cone inputs to the MC, PC, and KC ganglion
lengths, a biphasic curve emerges because the rods show greater
cells, which would then produce signals that are projected into
absolute sensitivity than cones at shorter wavelengths.12
the cortex to mediate different aspects of visual perception.
Clinical evaluation using dark
DARK ADAPTATION FUNCTIONS:
adaptation functions
ASSESSMENT OF THE SHIFT FROM DAY
It is known that certain retinal disorders may selectively affect
VISION TO NIGHT VISION rods (e.g., retinitis pigmentosa) or cones (e.g., cone dystrophies).
Measurements of sensitivity thresholds during adaptation to Clinically, rod and cone functions can be evaluated electrophysi-
darkness have produced a characteristic biphasic function with ologically by measuring rod and cone electroretinograms (ERG:
an initial segment that is attributed to cone responses and see Chapter 7, Electrogenesis of the electroretinogram) or, psy-
a subsequent segment attributed to rod responses. Figure 10.4 chophysically, by measuring dark adaptation functions. Dark
shows a characteristic dark adaptation function measured in the adaptation functions quantify the ability of the rod and cone
peripheral retina. Thresholds decrease quickly initially and this systems to recover sensitivity (i.e., regenerate photopigment)
rapid recovery is attributed to cones. Thresholds then reach a after exposure to light. The recovery is faster for cones, but the
plateau in about 5 minutes and remain invariant for another 5 absolute level of sensitivity is greatest for rods. Variations in
sensitivity and sensitivity recovery times can be used to charac- When all three primaries are added together, they can be set to
terize retinal disorders. match a neutral white.
Clinical evaluation using dark adaptation functions involves Procedurally, due to the mechanics of testing optical equip- 289
a measure of the cone and rod absolute thresholds and the time ment, the spectral test color and one primary are made to appear
of the rod–cone break. Specifically, rod absolute thresholds have in one field and the other two primaries appear in a second field

Chapter 10
been used as a psychophysical supplement to ERG measurement with the two fields usually appearing as two halves of a circle.
for night-blindness evaluation. The instrument for dark adapta- The observer’s task in a color-matching experiment is to make
tion rod absolute threshold measurement is called a dark adap- the two fields appear identical in color. Color-matching data are
tometer and the most widely used is a Goldmann–Weekers dark usually represented in one of two ways. In the first, the amounts
adaptometer (Haag–Streit). This instrument is old, however, and of energy of each primary can be plotted as a function of the test
finding replacement lamps is difficult. A new light-emitting wavelength. These are called color-matching or color mixture
diode-based dark adaptometer has recently become commer- functions. In such plots, the primary that is added to the test

Color Vision and Night Vision


cially available (LKC Technologies scotopic sensitivity tester-1: color is given a negative value, and the other two primaries are
SST-1). The SST-1 dark adaptometer can determine a full dark given positive values. The second plotting method is to normal-
adaptation curve as well as full-field scotopic sensitivities. ize the value of each matching primary relative to the sum of the
three primaries, leading to the sum of the normalized primaries
COLOR VISION being equal to 1. Therefore, a plot of one primary value against
a second primary value is sufficient to display all the information
Color vision refers to our ability to perceive colors based on
in the color-matching data. This kind of plot is called a chroma-
spectral variations in light absorbed by the photoreceptors.
ticity diagram, which shows the relative contributions of each
Color vision includes both chromatic discrimination and color
spectral primary needed to match any spectral light.
appearance appreciation. Color matching and color discrimina-
tion experimental tasks are two fundamental psychophysical
The CIE colorimetric system
procedures that have provided theoretical insights into the
This spectral primary-based chromaticity diagram has proven
nature of color vision and have also been developed for clinical
to be very useful as a generalized color specification system.
diagnosis of color vision. Color matching and color discrimina-
However, linear transformation is needed to compare data col-
tion results, however, do not address questions of color appear-
lected with different choices of primaries. In 1931, the Com-
ance, for example, why an object appears red. Color appearance
mission Internationale d’Eclairage (CIE) standardized the
is far more complex because it depends on not only the chro-
spectral-primary-based chromaticity system by adopting three
matic properties of an object but also the spatial, temporal, and
imaginary primaries (X, Y, Z primaries) that are out of the
spectral characteristics of the neighboring objects.13 Neural pro-
spectral locus and therefore do not exist physically. The choices
cessing beyond the retina is required for color appearance
of the imaginary primaries were based on two important con-
perceptions.
siderations: first, to ensure that the chromaticities of all physical
Color matching lights have positive values and, second, to relate colorimetric
functions to the previously adopted luminosity function, V(λ).
Color matching as the foundation for the theory
of trichromacy In the system, the color-matching function for the Y primary
is identical to V(λ) of the photometric system as designed.
The psychophysical procedure in which an observer sets a
Figure 10.5A shows the 1931 CIE chromaticity chart, in which
mixture of three primary lights to match the color of a test stimu-
the coordinates of the Y primary [y = Y/(X + Y + Z)] are plotted
lus is called color matching. It has been known since the 19th
against those of the X primary [x = X/(X + Y + Z)]. The spectrum
century that different colors perceived by humans can be speci-
loci (their wavelengths are indicated on the graph) form the
fied by an economical three-variable (trichromatic) system. In
horseshoe-shaped curve. Equal-energy-spectrum (EES) light is
the 1800s, Thomas Young and Hermann von Helmholtz pro-
plotted in the center, with the coordinates of x = 0.3333 and y =
posed that there must be three kinds of physiological entities in
0.3333. A straight line connects 400 nm to 700 nm for purples,
the eye accounting for this trichromacy and their theory is called
which result from mixtures of short- and long-wavelength light.
the Young–Helmholtz trichromatic theory of color vision. We
All possible lights occur within the boundaries of the spectrum
now know the basis for trichromacy is the existence of three cone
locus and the purple line. Highly saturated colors occur near the
types in the retina. Color matching was the means to uncover
locus and desaturated (pale) colors occur near the white point.
the spectral sensitivity functions of the three different cone types
The 1931 CIE system was based on 2° field color-matching
(Fig. 10.1B).14
functions that were derived from color matches of many observ-
Color-matching experimental techniques and data ers. The averaged color-matching function from these observers
Theoretically, a color match occurs when the photoreceptor was treated as the standard. Therefore, an observer with the
quantal catch for the test stimulus and the quantal catch for the standard color-matching function is referred to as the standard
stimulus that is a combination of the three primary lights are the observer. Since the color-matching data are affected by the
same. Color matches are unperturbed by changes in luminance size of the stimulus field presented to the observer, in 1964,
levels, as long as no significant photopigment bleaching occurs, the CIE also adopted a large-field XYZ system that was based
which means that the basic nature of trichromacy exists under on the 10° field standard observer color-matching functions.
wide variations in light levels. In a classical color-matching For large stimuli, such as those generated by Ganzfeld for
experiment, three spectral lights are chosen as the three prima- ERG measurements, it is recommended to use the 1964 CIE
ries and the precise setting of the relative percentages of the chromaticities to reflect more accurately the large field color-
three primaries to match a test light is the data for that match. matching functions.
CIE 1931 Chromaticity chart Macleod and Boynton cone space
290

1.0 1.0
420 400
Section 2

520
0.8 0.8
510 540
440
560
0.6 0.6
Retinal Diagnostics
Retinal Imaging and Diagnostics

S/(L+M)
500
580
y

0.4 0.4
600
460
490
700
0.2 0.2
480
480
400 520 EES 600 700
400
0.0 0.0
0.0 0.2 0.4 0.6 0.8 1.0 0.0 0.2 0.4 0.6 0.8 1.0
A x B L/(L+M)

Fig. 10.5 Chromaticity spaces. (A) The Commission International d’Eclairage (CIE) 1931 x, y chromaticity diagram. An equal-energy-spectrum
(EES) light has a coordinate of x = 0.3333 and y = 0.3333. Spectral wavelengths are represented on the horseshoe-shaped spectrum locus.
All lights can be represented in this diagram. (Data from Wyszecki G, Stiles WS. Color science – concepts and methods, quantitative data,
and formulae, 2nd edn. New York: John Wiley; 1982.) (B) The MacLeod and Boynton cone space, which plots S-cone excitation versus relative
L/M-cone excitations. In this space, the S/(L + M) chromaticity of 400 nm spectral light is normalized to be 1. In such a normalization, an EES
light has a chromaticity of L/(L + M) = 0.665 and S/(L + M) = 0.016. In a relative cone troland space, the S/(L + M) for an EES light is normalized
to be 1. The chromaticities of the spectral loci form an “L” shape in this space.

Cone chromaticity space Chromatic discrimination


The CIE colorimetric system is valuable for light specifications; Chromatic discrimination refers to the ability of an observer to
however, psychophysical experiments using the CIE system discriminate two colors. Chromatic discrimination has been
cannot yield results that allow easy interpretation of the underly- investigated using three approaches: wavelength discrimina-
ing physiological mechanisms. When the physiological mecha- tion, purity discrimination, and chromaticity discrimination
nisms of color vision are of interest, a cone chromaticity space (reviewed by Pokorny and Smith19). Chromatic discrimination is
that can represent cone stimulations, as well as the postrecep­ usually measured at a constant luminance level to avoid poten-
toral pathways, is preferred. tial interactions between the luminance pathway (MC pathway)
The concept of cone chromaticity space appeared in the early and the chromatic pathway (PC or KC pathway).
20th century. It was not until 1979 that MacLeod and Boynton15
published a cone chromaticity space based on modern estimates Wavelength discrimination
of the cone spectral sensitivities.14 In the MacLeod and Boynton In a typical wavelength discrimination experiment, the stimulus
cone chromaticity space (Fig. 10.5B), the horizontal axis [L/(L + consists of two equiluminant semicircular fields, one filled with
M)] represents the variation of relative L- versus M-cone stimu- a narrow band of spectral light to serve as the standard field and
lation at equiluminance, while the vertical axis [S/(L + M)] rep- the other as the comparison field. The observer is instructed to
resents the variation of S-cone stimulation. The space normalizes change the wavelength of the comparison field to achieve a just
in that S/(L + M) = 1 for spectral light of 400 nm. Later, a relative noticeable difference (JND) from the standard wavelength.
cone troland space that normalizes S/(L + M) for EES light to be Typical wavelength discrimination thresholds, as a function of
1 was proposed to link cone excitations with retinal illuminance, standard wavelengths, form a skewed “W” shape with two
which is measured in trolands.16 Another spectral opponency minima, one at 490 nm and the other at 580 nm.20
space normalizes the cone chromaticity based at the EES-white
to reflect both cone contrasts and postreceptoral opponency Purity discrimination
signals.17 These cone chromaticity spaces are a major break- There are two ways to measure purity discrimination. The first
through for vision research because neurons in the PC and KC method measures the minimum amount of spectral light the
pathways show preferred responses to stimuli along the two observer adds into a white field to achieve a JND from the same
axes of the cone chromaticity spaces.17,18 Therefore, psychophysi- white in another juxtaposed field. Discrimination thresholds
cal experiments can be designed to infer the functions of the measured in this way are the largest at 570 nm and the smallest
postreceptoral pathways by generating stimuli along the two at 400 nm. The second method measures the minimum amount
theoretical axes. of white light added into a spectral light to achieve a JND from
the spectral light. Purity discrimination thresholds measured in
1.0
this way do not vary much with variations in the wavelength of Achromatic
the spectral light. 291
Chromaticity discrimination
Early attempts to measure chromaticity discrimination included

Chapter 10
0.5
measurement of the minimum variation needed in chromatic-
ity to achieve a JND from any point in the CIE diagram.21
Another measure of chromatic discrimination involved derived

Valence
discrimination ellipses using the standard deviations of
0.0
repeated color matches at a set of chromaticities in the CIE
diagram.22

Color Vision and Night Vision


In modern chromaticity discrimination experiments, discrimi-
nation thresholds are obtained for test field chromaticities varied
along the two cardinal axes of a cone chromaticity space in a –0.5
steady adaptation field. A cone chromaticity space allows that (Y-B) (R-G)
discrimination can be mediated by the L/M cones only (L/M
cone discrimination) or by the S cones only (S-cone discrimina-
tion).23 Chromatic discrimination was found to be the best at the –1.0
adaptation chromaticity and then deteriorated with increasing
350 400 450 500 550 600 650 700 750
chromatic contrast between the test and adapting fields. Chro-
Wavelength (nm)
matic discrimination data can be explained adequately by a
model based on primate ganglion cell responses in the PC and Fig. 10.6 Color opponency. Theoretical curves for the first stage of
opponent coding for normal 2° foveal color vision. The (all-positive)
KC pathways.24 achromatic function represents the whiteness response. Two
(opponent) chromatic functions, (R-G) and (Y-B), represent redness–
Color appearance greenness and blueness–yellowness, respectively. (Reproduced with
permission from Hurvich LM, Jameson D. Some quantitative aspects
In color-matching and color discrimination experiments, observ- of an opponent-colors theory. II. Brightness, saturation and hue in
ers determine whether two colors appear the same or different normal and dichromatic vision. J Opt Soc Am 1955;45:602–16.)
but they do not determine which color is perceived. Color
appearance has three perceptual dimensions: hue, saturation,
and brightness. Hue is the perceptual dimension that differs
from white, such as red, orange, green, blue, yellow, purple, and In a complex visual scene, the perceived color of a light
pink. Saturation indicates how different the hue is from white. (emitted from a source or reflected from an object) cannot be
For instance, colors on the spectral locus are highly saturated predicted from its spectral power distribution because the
compared with desaturated colors near white (Fig. 10.5A). context of other nearby light affects color appearance. For
Brightness is the perceptual dimension related to luminance instance, chromatic induction occurs when perception changes
variance. because of the presence of other lights nearby in space or time.
An important feature of color appearance is that colors do not Chromatic induction includes both chromatic contrast and chro-
simultaneously appear red and green, nor do they simultane- matic assimilation. Chromatic contrast occurs when the color
ously appear blue and yellow. However, colors do appear as appearance of a test light shifts away (in terms of color oppo-
mixtures of red and yellow or red and blue and they also appear nency) from the color appearance of a nearby light. Chromatic
as mixtures of green and yellow or green and blue. Further, assimilation occurs when the color appearance of a test light
human observers can separately abstract the qualities of redness– shifts toward the color appearance of inducing nearby light.
greenness or blueness–yellowness in an arbitrary test color. Therefore, the color of a light seen in isolation (surrounded by
These facts have led to the concept that color appearance can be darkness) is called an unrelated color while the color perceived
represented in a double-opponent system, with red and green in the presence of a complex context is called a related
placed in opposite directions on one dimension and blue and color; that is, its percept is related to the surrounding colors.
yellow placed in opposite directions in the other dimension (Fig. Cortical mechanisms are likely involved in color perception in a
10.6). Red and green are said to be opponent sensations, as are complex scene.26
blue and yellow. This observation triggered Ewald Hering, a
German physiologist, to propose the “opponent-color” theory in
VARIATIONS IN HUMAN COLOR VISION
the late 19th century. Abnormal color vision that is either inherited or acquired is
The theories of opponency and trichromacy were two compet- present in about 4.5% of the population. Congenital color vision
ing theories in the history of color vision research; however, the defects are stationary over the lifespan and do not result from
two theories have been reconciled in that chromatic processing other visual problems. These color vision defects have been
starts with three types of cones, supporting trichromacy, and studied extensively and their classification is well established
spectral responses from the cones are transmitted in bipolar and based on psychophysical and genetic works. The most common
ganglion cells that have antagonistic receptive field structures, are the congenital X-chromosome-linked red–green color vision
consistent with opponency. This interpretation of color vision, defects, which have been associated with alterations in the
first proposed by von Kries in 1905,25 is referred to as the two- gene sequences encoding the opsins on the X chromosome.27,28
stage or two-process model of color vision. Acquired color vision defects refer to abnormalities that
accompany eye diseases or drug toxicity. Acquired color vision have a high homology to each other (about 96%), suggesting a
defects are more variable and their classification is more difficult more recent evolutionary appearance. An unexpected finding
292 and less satisfactory. Color vision is often tested clinically with was that of multiple genes in a tandem array on the X chromo-
screening tests that allow identification of abnormalities and some, which has been proposed to range from 2 to 6. Nathans
most screening tests are based on color discrimination and et al.28 postulated that the multiple genes in the tandem array on
color-matching abilities.29 the X chromosome arose as a result of unequal homologous
Section 2

recombination. Subsequent study has suggested there are poly-


Color vision classifications morphisms among these genes in color-normal and color-
Color vision classifications are based on both the number of defective individuals.27
functioning cone types and the presence of abnormal cones. The initial study of the opsin genes on the X chromosome
An observer with three functioning cone types is called a tri- was based on the long-standing conventional ideas about
chromat, an observer with two functioning cone types is a X-linked congenital color vision defects28; that is, protanopes
Retinal Diagnostics
Retinal Imaging and Diagnostics

dichromat, and an observer with one functioning cone type is are missing L cones and deuteranopes are missing M cones,
a monochromat (monochromacy sometimes is also termed and that the genetics of these types of defective color vision
achromatopsia in the literature since it is believed that vision may be based on missing genes for either L or M cones. Spe-
based on a single cone type cannot produce color perception, cifically, protanopes were thought to be dichromats who lacked
assuming rods are not involved). An observer with normal an L-cone gene and deuteranopes were dichromats who lacked
color vision has three normal cone types, in terms of spectral an M-cone gene.
sensitivity, and is called a normal trichromat. Observers who The initial work with the X-chromosome opsin genes, in
are said to have defective color vision have at least one abnor- attempting to link color vision defects with these genes, was
mal cone type or are missing at least a cone type with con- carried out using restriction fragment length polymorphisms
ventional color-matching techniques. Observers with rods only (RFLPs), an early molecular genetic technique that simplified the
(lack of any cones) are called rod monochromat or complete study of DNA sequences. The initial RFLP analysis of the
achromatopsia.30 X-chromosome genes was done on DNA from one observer with
X-linked color vision defects have been recognized since the normal color vision and a few protanopes and deuteranopes.
18th century and were subdivided into two qualitatively dif- Comparisons were carried out to determine which of the frag-
ferent types: protan (“red-blind”) and deutan (“green-blind”). ments found in the normal observer’s RFLPs were missing in the
The term “protanope” is used for a dichromat who is thought protanopes and deuteranopes. A fragment found in the normal
to be missing L cones and the term “deuteranope” is used for observer that was missing in the protanope was said to be part
a dichromat who is thought to be missing M cones, based on of the assumed missing L-cone gene. Similarly, a fragment found
color-matching characteristics using a 2° visual field in the fovea. in the normal observer that was missing in the deuteranope was
Anomalous trichromacy is a variation in color vision that is said to be part of the assumed missing M-cone gene. In the initial
attributed to the presence of a cone type that is shifted in study, observers with dichromatic as well as anomalous trichro-
spectral sensitivity. A protanomalous observer has trichromatic matic color vision defects showed the presence of hybrid genes
color vision but the L cones have spectral sensitivity that is (genes comprising the head of one type of gene and the tail of
shifted to shorter wavelengths compared to normal L cones. the other type of gene). The hybrid genes were said to be the
A deuteranomalous observer also has trichromatic color vision basis for the altered spectral sensitivity of anomalous L or M
but the M cones have spectral sensitivity that is shifted to cones associated with X-linked anomalous trichromacy (dis-
longer wavelengths compared to normal M cones. A third cussed previously).
qualitively different type of color vision is tritan (“blue-blind”). Subsequent studies that have been carried out on both normal
Tritan color vision defects are thought to arise from variations and X-linked defective color vision have their roots in the initial
in S cones. RFLP analysis based on the conventional idea of missing cone
types and missing genes using one normal observer and a few
The genes encoding the color-defectives. The variations that have been found in subse-
human photopigments quent studies on the visual pigment genes have correlated
It has been known for many years that the spectral sensitivities largely, but not absolutely, with phenotypes established by color
of rods and cones reflect the absorption spectra of the visual vision testing. The current view is that the X-chromosome
photopigments. In a major advance, the genes encoding the tandem array consists of one or more opsin genes encoding
opsins of the human visual photopigments were cloned and L-cone photopigment, followed by one or more opsin genes
mapped in the human genome in 1983.27,31 The gene for rhodop- encoding the M-cone photopigment. The expression of the genes
sin was found on chromosome 3 and the human rhodopsin gene in the tandem array is believed to be governed by a stochastic
showed high homology (93.4%) to that of bovine rhodopsin.31 A process.27
visual photopigment gene for the opsin of S cones (OPN1SW There have been advances in molecular genetic technology
gene) was found on chromosome 7. A tandem array of visual and in the understanding of the human genome since the
photopigment genes for the opsins of the M and L cones initial RFLP studies of the opsin genes. Knowledge derived
(OPN1MW and OPN1LW genes) was found on the X chromo- from the Human Genome Project, which has spurred an
some. The human opsin genes show about 45% homology understanding of the scarcity of genes in the human genome,
between rhodopsin and any of the three cone photopigment and the developing knowledge of epigenetics, such as the
genes and between the chromosome 7 and the X-chromosome editing processes of microRNAs, may contribute to future
pigment genes. This similarity among the photopigment genes studies and understanding of the genetics of color vision
suggests a common ancestor. The genes on the X chromosome variations.
CLINICAL EVALUATION OF COLOR VISION ordering, manual dexterity, and patience. As a result, they are
rarely suitable for children under 10 years of age.
Screening tests The best known of the arrangement tests is the Farnsworth– 293
Screening tests are rapid tests (requiring 2–3 minutes) and color- Munsell 100-hue test. The test includes 85 black plastic caps
defective observers are identified due to their inability to see the with inserted papers that vary in hue but have constant light-

Chapter 10
difference between certain colors that are easily discriminated ness and saturation. The caps are divided into four boxes with
by normal observers. Screening tests can be administered to both each box covering one-quarter of the color circle. Observers
children and adults. being tested arrange the caps in a natural color order according
to their unique perception. An error occurs if the caps are
Pseudoisochromatic plate tests misplaced from the ideal color order. A numeric score can be
The most commonly used screening tests are the pseudoiso- calculated and displayed on a polar graph. Age norms have
chromatic plate tests. First introduced by Stilling, a pseudoiso- been prepared giving the range of the expected total error

Color Vision and Night Vision


chromatic plate presents a figure composed of colored dots in scores for an unselected population as well as the expected
a background of differently colored dots. Usually, the colors intereye variability.
are chosen so that an X-linked color-defective observer does Observers with congenital color vision deficiencies make char-
not see the figure that is easily seen by normal observers. The acteristic errors on arrangement tests because their chromatic
cleverest designs use four sets of colors, chosen so that the discrimination ability is weakened or lost on particular axes in
normal observer sees one figure and the defective observer chromaticity space. Discrimination loss in acquired color vision
sees a different figure. defects is more variable. However, following the idea that dis-
The majority of pseudoisochromatic plate tests (such as the crimination of blueness content is, to a first order, independent
Ishihara) were designed to identify observers with X-linked of discrimination of redness–greenness, it is possible to partition
congenital color defects (i.e., protan or deutan color anomalies). the caps into those where correct ordering depends on normal
The choice of colors was optimized to take advantage of the function of the S-cone system (caps 1 through 12, 34 through 54,
particular discrimination losses found in X-linked color vision and 76 through 84) and those where correct ordering depends
defects and the tests are successful in detecting 90–95% of on normal function of the M- and L-cone opponent system (caps
color-defective observers. Pseudoisochromatic plate tests 13 through 33 and 55 through 75). The partitioned scores can be
cannot be used to identify acquired color vision defects, which examined to determine whether an acquired color vision defect
are most likely to affect blue/yellow color vision. More causes a particular type of discrimination loss; i.e., the S or L/M
recently developed pseudoisochromatic plates (such as the systems.32
HRR and SPP2 plates) have been designed specifically for
acquired color vision deficiencies, including tritan (blue/ Importance of the test illuminant for plate
yellow) anomalies.
and discrimination color vision tests
Other rapid tests of color vision The plate and discrimination tests described above use reflective
Other rapid tests of color vision involve sorting colored pieces. materials as colored test objects and the perceived color pre-
In principle, this approach can be used successfully with acquired sented to an observer depends on the illuminating light as well
color vision abnormalities since it does not involve the choice of as the reflective properties of the test materials. The original
a particular pair of colors that has a prediction based on common pseudoisochromatic plate tests were designed to be viewed
X-linked color vision defects. under afternoon daylight in the northern hemisphere, and more
recent tests have followed this design convention. Standardized
Chromatic discrimination ability tests illuminants (called illuminant C or illuminant D65) that closely
Clinical assessment of color discrimination ability involves simulate the spectra of afternoon daylight are more preferable
arrangement tests that require the observer to arrange a set of to natural daylight, which may vary substantially in both spec-
colored samples according to their similarity in color. If the trum and radiance with time of day and weather. Light sources
samples are closely spaced in chromaticity (e.g., the Farnsworth– that approximate these standard illuminants are commercially
Munsell 100-hue test: Fig. 10.7), the task becomes one of fine available and are suitable for use in illuminating clinical color
chromatic discrimination. Tests involving fine chromatic dis- vision tests. Most fluorescent light sources, however, do not
crimination are usually relatively time-consuming. If the samples accurately mimic the colors produced in natural light and, there-
are widely spaced in chromaticity (e.g., the Farnsworth panel fore, they are not appropriate illuminants for color vision tests.
D-15), the test evaluates color confusions that occur with defec-
tive color vision that would not be perceived by a normal Color-matching tests
observer. Tests with widely spaced colors are conducted rapidly Adjusting three primaries to match a test color is not intui-
and can even be used for screening. An arrangement test may tive for many observers and research experimental methods
use samples that differ only in chromaticity to test hue discrimi- are therefore not appropriate for clinical purposes. To adapt
nation (e.g., the Farnsworth–Munsell 100-hue test, Farnsworth color-matching procedures for clinical evaluation, simplified
panel D-15, Farnsworth desaturated panel D-15, and Lanthony methods have been developed by using two-variable matches
new color test), may vary in luminance only to test lightness and rapid, less complicated tasks. The instruments that allow
discrimination (Verriest’s lightness discrimination test), or may these matches to be carried out are called anomaloscopes and
vary only in grayness to test saturation discrimination (Sahl- the color-matching paradigms are called equations. The equa-
gren’s saturation test, Lanthony new color test). Arrangement tions are named after the researchers who first proposed or
tests are easy to administer but require the concept of abstract used them.
610 633 600 610 633
Normal 595
600
nm Normal 595 nm
294 590
14
590
14
13 13
5 5
58 12 58 12
11 11
10 10
9
Section 2

9
8 8

0
0

58
58

7 7
6 6
5 5
4 4
3 3

575
2 2
575

21 21
6 4

84
6 4

84

82
82
8 8

80
80
10 10

78
RED

78
12 RED
76 12 76

4
4 74 74

61
61

W
RP RP

PUR
OW
72

PUR
72

181
YR
181

YR

LLO
18 70 18 70
L

445 460

PLE

445 460
PLE
YEL
Retinal Diagnostics
Retinal Imaging and Diagnostics

YE
22 68 22 68

570
570

24 66 24 66

GY
GY

PB
PB
64 26 64
26

nm

nm
62 62
28 BG
60 28 BG
60
30
BL BL
30
N
58
N
5 EE UE
56 8
EE
32 4
UE

470
32 4

470
GR GR
3 3

56 4
5
36
54
36

52
38
52
38

565

40

50
40

50
565

42

48
42

48

44 46
44 46

475

475
560
560

5
5

55
55

0
0

55
55

48

48
5
0
5

0
54
54

0
54
0
54
35 35
05 05
53 48 53 48
0 5 0 5
51 51
505 505
500 490 500 490
495 495

Panel 1 Panel 2

600 610 633


Deuteranope 595
600 610 633
nm Protanope 595 nm
590
590 14
14
13 13
5
5 12 58 12
58 11
11
10 10
9 9
8
0

8
58
0

7
58

7
6 6
5 5
4 4
3 3
575

2 2
575

21
6 4

84
21
6 4

82
84

8
82

80
8 10
80

10

78
12 RED
76
78

12 RED
76 4
74 74
61
4
61

W
RP

PUR
72
181

RP YR
W

PUR

72
181

LLO
YR
LO

18 70
18 70

PLE

445 460
445 460
PLE
YEL

YE

22 68
570

22 68
570

66 24 66
GY

PB
24
GY

64
PB

64 26

nm
26
nm

62 62
28 BG
60
28 60 30
BG N BL
58
BL
30 EE
N UE
5 32 4

470
56 8
EE UE
470

32 4 GR 3
GR
56 4
3 5
36
54

52
36

38
565

40
52

50
38

42

48
44 46
40

50
565

42

48
44 46

475
475

560
560

5
55
55
5

0
55
0

48
55

48

0
54
0
5
54

0
54

40
55 35
53 05
0 53 48
53 5
48 0 5
0 51
51 505
505 500
500 490 495 490
495

Panel 3 Panel 4

Fig. 10.7 Examples of Farnsworth–Munsell 100-hue test error patterns in normal subjects (panels 1 and 2) and in observers with congenital
(panels 3–6) and later onset or acquired (panels 7 and 8) color vision defects. Panel 1, Normal with good discrimination. Panel 2, Normal with
poor discrimination. Panel 3, Deuteranope. Panel 4, Protanope.

of the yellow to make the two fields appear identical. Observers


Anomaloscope color matching test using the with normal color vision accept a narrow range of ratios near
Rayleigh equation the middle of the red–green range, but color-deficient observers
In a Rayleigh match, a spectral “yellow” test field (589 nm) is will pick ratios shifted away from this range, depending on the
presented in one-half of a circular field with an adjustable bright- specific deficiency. Matching abnormalities accompanying oph-
ness. In the half field, a mixture of two “green” and “red” spec- thalmic disease or X-linked color vision variations may include
tral primaries (545 and 670 nm) is presented. The mixture field a widened matching range or acceptance of an abnormal match.33
can appear green (545 nm), green–yellow, yellow, orange, or red The Rayleigh equation assesses the normality of M- and
(670 nm) as the ratio of the primaries is varied. The task of the L-cone functions, since S cones are not contributing to the match
observer is to adjust the primary red–green ratio and brightness because they are unresponsive to the primary wavelengths.
610 633 600 610 633
Tritanope 595
600
nm Achromat 595 nm

590 590 295


14 14
13 13
85 12 585 12
5
11 11
10 10

Chapter 10
9 9
8 8

0
0

58
58

7 7
6 6
5 5
4 4
3 3

575
2 2
575

21 21
6 4

84
6 4

84

82
82
8 8

80
80
10 10

78
RED

78
12 RED
76 12 76

4
74 74
4

61
61

W
RP RP

PUR
W
72

PUR
72

181
YR
181

YR

LLO
LO

18 70 18 70

445 460

PLE

445 460
PLE

Color Vision and Night Vision


YEL

YE
22 68 22 68

570
570

24 66 24 66

GY
GY

PB
PB
64 26 64
26

nm

nm
62 62
28 BG
60 28 BG
60
30
BL BL
30
N
58
N
5 EE UE
56 8
EE
32 4
UE

470
32 4

470
GR GR
3 3

56 4
5
36
54
36

52
38
52
38

565

40

50
40

50
565

42

48
42

48

44 46
44 46

475

475
560
560

5
5

55
55

0
0

55
55

48

48
5
0
5

0
54
54

0
54
40
55 35
53 0 05
53 48 53 48
0 5 0 5
51 51
505 505
500 490 500 490
495 495

Panel 5 Panel 6

600 610 633


Retinitis pigmentosa 595
600 610 633
nm Optic neuropathy 595 nm
590
590 14
14
13 13
5
85 12 58 12
5 11
11
10 10
9 9
8
0

8
58
0

7
58

7
6 6
5 5
4 4
3 3
575

2 2
575

21
6 4

84
21
6 4

82
84

8
82

80
8 10
80

10

78
12 RED
76
78

12 RED
76
4

74 74
61
4
61

W
RP

PUR
72
181

RP YR
W

PUR

72
181

LLO

YR
LO

18 70
18 70

PLE

445 460
445 460
PLE
YEL

YE

22 68
570

22 68
570

66 24 66
GY

PB
24
GY

64
PB

64 26

nm
26
nm

62 62
28 BG
60
28 60 30
BG N BL
58
BL
30 EE
N UE
5 32 4

470
56 8
EE UE
470

32 4 GR 3
GR
56 4
3
5
36
54

52
36

38
565

40
52

50
38

42

48
44 46
40

50
565

42

48
44 46

475
475

560
560

5
55
5
55

0
55
0

48
55

48

0
54
0
5
54

40 5

0
54 35
35 05
05 53 48
53 5
48 0 5
0 51
51 505
505 500
500 490 495 490
495

Panel 7 Panel 8

Fig. 10.7 Cont’d Panel 5, Tritanope. Panel 6, Achromat. Panel 7, Retinitis pigmentosa with an acquired violet–yellow defect. Panel 8, Optic
neuritis with an acquired red–green defect.

A graphic display of the cone photopigment excitations allows labeled Rod) is also shown. The normal match occurs at or near
the tester to evaluate which photopigments participated in the intersection of the L- and M-cone settings and the matching
an observer’s match. Figure 10.8A shows the expected yellow range is narrow.
setting, based on the L- and M- photopigments, as a function Deuteranopes can make satisfactory matches throughout the
of the red–green primary ratios. For each photopigment, the L line. A deuteranope is thought to lack functional M cones in
quantal catch in cones may be characterized by a straight line the retinal area responding to the stimuli. Studies on the
in a chromaticity diagram as the red–green primary ratio is X-chromosome opsin genes in deuteranopes show that many of
changed. Two lines show the predicted responses of the L cones these observers have only a single gene that is thought to encode
alone (dashed and labeled L) and the M cones alone (dotted the L photopigment. Protanopes can make matches throughout
and labeled M). The rod photopigment is responsive at these the extent of the M line. It has been speculated that the protanope
wavelengths as well and the predicted rod response (solid and lacks functional L cones in the retinal area responding to the
stimuli and genetic studies have suggested that these observers
80 usually have only a single gene on the X chromosome that has
296 been described as a hybrid gene composed of a piece of the
normal L- and a piece of the normal M-opsin genes.
Rod
Other X-linked color-defective observers, called anomalous
trichromats, have a wider range of acceptable matches that are
Section 2

60
Amount of yellow test field

displaced from the normal match. Such observers are trichro-


matic but have one cone photopigment with an abnormal spec-
tral sensitivity. Deuteranomalous matches occur along the L line
40 (indicating a normal L-cone photopigment) but are shifted to
low red–green primary ratios. The psychophysical interpretation
is that the deuteranomalous trichromat has an abnormal M-cone
Retinal Diagnostics
Retinal Imaging and Diagnostics

MWS
photopigment that is shifted so that its spectral sensitivity closely
20
LWS overlaps that of the normal L-cone photopigment. Protanoma-
lous matches occur along the M line (indicating a normal M-cone
N
photopigment) but are shifted to high red–green primary ratios.
0 The interpretation of these psychophysical studies is that the
10 20 30 40 50 60 70 protanomalous trichromat has an abnormal L-cone photopig-
Amount of red primary
ment, shifted so that its spectral sensitivity closely overlaps that
A of the normal M-cone photopigment. Anomaloscope color
matching using the Rayleigh equation is recognized as the only
clinical method that allows definitive classification of the
X-linked color vision defects.
100
Anomaloscope analysis is important in ophthalmic disease
assessment since it can be used to recognize a number of clinical
entities, such as rod-dominated vision characteristic of cone
80
SWS degenerations and incomplete and complete achromatopsias.
Amount of cyan in test field

LWS/MWS These diseases are characterized by Rayleigh matches extending


60
along the rod line (solid line in Fig. 10.7A). Matching abnormali-
ties that are characteristic of choroidal disorders affecting the
Rod fovea lead to Rayleigh matches that extend along or just below
40
the L line but are shifted to higher red primary ratios (pseudo-
protanomaly). The discrimination loss characteristic of optic
nerve disorders leads to matches that widen around the normal
20 match on the L line.
N

Anomaloscope color-matching test using the


0
Moreland equation
0.2 0.4 0.6 0.8 1.0 The Moreland equation34 is a match of a bicolor test field (480
and 580 nm) to a mixture of two primaries (440 and 500 nm).
B Proportion of blue primary The test field appears blue–green to a normal observer, while
appearance of the mixture field appearance ranges from violet,
Fig. 10.8 Graphic analysis of the Rayleigh equation and Moreland to blue, to blue–green, to green–blue, to green as the primary
equation. (A) The Rayleigh equation. The relative light in the “yellow” ratio is changed from mostly 440 nm to mostly 500 nm. The
(589 nm) test is plotted as a function of the proportion of the “red” primary ratio is expressed in terms of the amount of the 440 nm
(670 nm) primary. The lines are calculated from the cone and rod
photopigment excitations. The line marked LWS represents the L-cone (“violet”) primary required for the match. As with the Rayleigh
response of Figure 10.2B and predicts the matching behavior of equation, an observer with normal color vision can find a
deuteranopes. The line marked MWS represents the middle- primary ratio and test field luminance at which both fields
wavelength cone response of Figure 10.2B and predicts the matching
appear identical.
behavior of protanopes. The normal match (N) occurs at the
intersection of the L and M lines. The line marked Rod represents the The Moreland equation assesses the normality of S-cone func-
rod response of Figure 10.2A and predicts the matching behavior of tion. Figure 10.8B shows the predicted function for a single
achromats. (B) The Moreland equation. The relative light in the bicolor photopigment to the bicolor test field as a function of the violet–
(480 nm + 580 nm) test is shown as a function of the proportion of
the 440-nm primary. The lines are calculated from the cone and rod green primary ratio. The L and M cones share the same line
photopigment excitations. The line marked LWS/MWS represents (dotted and labeled as L/M). The S-cone response crosses this
the L- and M-cone response and predicts the matching behavior of on the diagonal (dashed and labeled as S). The predicted rod
tritanopes. The line marked SWS represents the short-wavelength cone (solid line labeled as Rod) line has the same direction as the L-
response. The normal match (N) occurs at the intersection of the L/M
line with the S line. The line marked Rod represents the rod response and M-cone lines. The normal match occurs at the intersection
and predicts the matching behavior of achromats. of the S-cone line with the L/M line. Both congenital and
acquired color vision defects can be recognized using the More-
land equation.35 The tritanope (an observer lacking S-cone func-
tion) has matches extending along the L/M line, as do observers
with acquired defects that affect S-cone function. Point muta- protan, deutan, and tritan lines of a chromaticity diagram. The
tions in the S-cone opsin gene on chromosome 7 have been task is to indicate the position of the gap in the target C. A stair-
reported in affected individuals in tritan pedigrees.36 case procedure is used to measure discrimination thresholds 297
Patients with X-linked achromatopsia (i.e., S-cone monochro- along any of the three lines. The results are plotted as discrimina-
macy) have matches that extend along the S-cone line. The tion ellipses in a CIE space. Individuals with color vision defi-

Chapter 10
majority of X-linked achromat pedigrees show major deletions ciencies will have elongated discrimination ellipses along a
of the region just preceding the cone opsin tandem gene array protan, deutan, or tritan line and, therefore, this test can be used
on the X chromosome in affected individuals.37 This region is for all color vision deficiencies.
thought to be a regulatory area controlling expression of the L-
The portal color sort test (PCST)
and M- cone opsin genes. Additionally, some X-linked achromat
The PCST is based on the FM-100 hue test but uses only 36
pedigrees show only a single abnormal gene instead of the
colored “chips,” which significantly reduces the testing time.
normal tandem array. Patients with complete achromatopsia

Color Vision and Night Vision


The chips are representative of the original 85 chips in the
have matches extending along the rod line.
FM-100 hue test. The observer arranges the order of the chips
Considerations in the use of anomaloscopes according to color similarities and the computer provides auto-
Even though the color-matching task on an anomaloscope is matic scores. The correlation between the PCST and FM-100 hue
simplified compared with usual research color-matching proce- test is high for testing congenital color vision defects but is
dures, anomaloscope testing procedures are not easily explained unknown for testing acquired defects.41
and an observer may require some practice before being able to
Smartphone/tablet applications for color
complete a match. Correct use of an anomaloscope requires
vision screening
extensive operator training and these instruments are therefore
With the popularity of mobile communication devices (e.g.,
usually found only in research centers. However, if properly
smartphones or tablets) increasing in medical settings, numer-
used, the anomaloscope is a diagnostic instrument of great
ous applications have developed for these devices as tools for
power.
clinical testing/screening, patient education, or physician educa-
tion and reference. Several applications use the pseudoisochro-
Computerized color vision tests matic plate principle for color vision screening. These applications
Computer-controlled color vision tests have been developed might be useful for a quick screening of color vision; however,
using a similar principle as that used for pseudoisochromatic these tools have not been validated and their sensitivity and
plate tests or discrimination ability tests (such as the FM-100 hue specificity for color vision defect screening are not known.
test). Computerized tests are automated and therefore easy to Further, there are many factors that can affect the test results.
use. Further, computerized tests can avoid the illuminating light The most important is the display characteristics of the mobile
issue that exists for reflective materials. However, the sensitivity device. These displays are not calibrated and they may not be
and specificity of the computerized tests largely rely on the homogeneous; therefore the tests for color vision screening are
accurate presentation of color on computer monitors, including not necessarily “pseudoisochromatic.” Finally, the illumination
cathodes ray tubes (CRTs) or liquid crystal displays (LCDs). A in the office may impair the test reliability. Therefore, results
CRT is typically preferred over a LCD because CRTs have reli- from these applications need to be confirmed by comparison
able temporal characteristics. To present color accurately, the with other established tools for color vision screening.
displays have to be calibrated, including spectral distribution
measurements and linearization. Caution must be taken when Which test to use in a clinical setting?
using a computerized color vision test that does not incorporate Many clinicians want to have some means of testing color vision
specific display calibration. without necessarily acquiring expensive instruments and exper-
Color assessment and diagnosis (CAD) test tise needed for professional evaluation and diagnosis. Tests
using colored papers (pigment tests) and the proper illuminant
The CAD test was developed by Dr Barbur and coworkers at
offer office clinicians the possibility of some color vision
City University, London.38 The web-based CAD uses a color
evaluation.
square that moves against a flickering luminance contrast noise.
A screening plate test with the proper illuminant would be
The color of the square changes along different chromaticity
minimal equipment for testing color vision. Approximately
directions. Color-defective observers have difficulty seeing the
8–10% of American males have one of the X-linked color vision
square moving, whereas normal observers easily see the square
defects. Identification of color vision defects in children before
move. The web-based CAD test requires a monitor to be
they enter grade school allows the clinician to provide counsel-
balanced at around 9000 K and the ambient illumination must
ing to the parents. Many children with color vision defects have
be kept at a minimum. It is reported that this web-based test
memories of being teased or ridiculed in the early grades and
has good sensitivity and specificity for red–green color
early testing allows appropriate counseling. Also, many males
deficiencies.39
with color vision defects may inadvertently choose careers, for
Cambridge color test (CCT) example, as pilots or firefighters, from which they will be barred
The CCT is a computer-controlled, easy-to-use color vision test. due to their color vision status. It should be noted, however, that
The CCT was developed by Drs Mollon, Reffin, and Regan the screening plate tests that may be very useful for the common
at the University of Cambridge, England.40 The CCT system genetic color vision defects are less successful at identifying
provides 14-bit color and luminance control on a calibrated CRT acquired color vision defects.
display. The stimulus is a Landolt C on an achromatic back- A more ambitious plan to test color vision in an office
ground. The chromaticity of the target C is varied along the would be to combine a screening plate test with a test of color
discrimination. A discrimination test (e.g., the Farnsworth– can provide high resolution of the cone mosaic and it can
Munsell 100-hue test) allows the clinician to follow changes in show whether a particular type of cone (e.g., L cones) is missing
298 color vision over time, such as might occur with optic neuritis. in the retina. Combining this information with genetic studies
Occasionally, a clinician will see a patient who has an extremely is potentially informative because AO imaging can provide
rare form of color defect and color vision testing may be informa- insights about cone distributions that can be related to the
tive. For example, cerebral achromatopsia is a fascinating case cone opsin genes.48 (See Chapter 5, Advanced imaging tech-
Section 2

that likely arises from damage to higher-order visual processes. nologies, for more details about this topic.)
In these cases, a more complete color vision testing (such as
spectral sensitivity and saturation discrimination evaluation) Rod and cone interactions in color vision
should be considered and case reports would be of wide interest Duplex theory states that rods and cones independently contrib-
in the medical community. Patients should be referred to a psy- ute to different aspect of visual perception. However, rods and
chophysics laboratory for more visual function testing, such as cones share common neural pathways from the retina to the
Retinal Diagnostics
Retinal Imaging and Diagnostics

contrast discrimination evaluation. brain and this provides a neural basis for rod–cone interactions
in visual function, including color vision.49 Conventionally, rod
vision has been considered to be achromatic. However, numer-
NEW DEVELOPMENTS IN COLOR
ous psychophysical studies have indicated that rods contribute
VISION RESEARCH to color vision at either mesopic50 or even scotopic light levels.51
Color vision has been studied considering genetics, evolution, Psychophysical evidence for rod contributions to color vision
physiology, and psychophysics. A few newer research areas comes from measurements of scotopic color contrast,52 photo-
related to color vision are provided here. chromatic intervals during the course of dark adaptation follow-
ing a light bleach,53 chromatic discrimination,54 and color-matching
Gene therapy for color vision defects or color appearance methods using unique hue measurement or
Drs Jay and Maureen Neitz and coworkers have carried out hue-scaling methods.55,56
pioneering studies on “curing” red–green color deficiency in Recently, rod contributions to color vision were studied using
dichromatic adult squirrel monkeys that were missing the L-cone a four-primary Maxwellian-view photostimulator57 that allowed
opsin gene at birth.42,43 As gene therapy, the human L-cone opsin independent control of rod and cone excitations at the same
gene was delivered into the photoreceptor layers of the retinas chromaticity, retinal locus, and light level. This new method has
of the monkeys. A few months after the introduction of the new yielded new insights into rod contributions to color vision. Spe-
opsin gene, these monkeys exhibited trichromatic color vision cifically, rods contribute to color percepts in a manner analogous
behavior with spectral sensitivity shifted, chromatic discrimina- to M-cone signals at all mesopic light levels and analogous to
tion improved, and color perception enriched. An experiment S-cone signals only at low mesopic light levels near cone thresh-
involving gene therapy in humans would require approval from olds.50 Also, the strength of rod contributions is linearly related
the National Institutes of Health Office of Recombinant DNA to rod contrasts.58
Activities (ORDA)/Recombinant DNA Advisory Committee For online acknowledgments visit http://www.
(RAC) and the Food and Drug Administration and this would expertconsult.com
not be expected to be granted without a long and thorough
REFERENCES
approval process. However, the fundamental question of 1. Hood DC, Finkelstein MA. Sensitivity to light. In: Boff KR, Kaufman L, Thomas
whether it is necessary to “cure” color deficiencies is currently JP, editors. Handbook of perception and human performance, vol I: Sensory
the point of debate since the majority of dichromats live a normal processes and perception. New York: John Wiley; 1986. p. 5-1–5-66.
2. Smith VC, Pokorny J, Lee BB, et al. Sequential processing in vision: The interac-
life and most are not significantly affected by having the color tion of sensitivity regulation and temporal dynamics. Vision Res 2008;48:
vision deficiency. 2649–56.
3. Goldberg SH, Frumkes TE, Nygaard RW. Inhibitory influence of unstimulated
rods in the human retina: evidence provided by examining cone flicker. Science
Adaptive optics (AO) retinal imaging system 1983;221:180–2.
4. Cao D, Zele AJ, Pokorny J. Dark-adapted rod suppression of cone flicker detec-
AO was initially used in astronomy to remove the effects of tion: evaluation of receptoral and postreceptoral interactions. Vis Neurosci
atmospheric distortion to improve the performance of teles­ 2006;23:531–7.
copes and laser communication systems.44 An AO system has 5. Sharpe LT, Stockman A. Rod pathways: the importance of seeing nothing.
Trends Neurosci 1999;22:497–504.
three components: (1) a wavefront sensor for ocular aberration 6. Dacey DM. Parallel pathways for spectral coding in primate retina. Annu Rev
measurement; (2) a deformable mirror for aberration correc- Neurosci 2000;23:743–75.
7. Lee BB. Visual pathways and psychophysical channels in the primate. J Physiol
tion; and (3) a control system that compares the sensor output 2011;589.1:41–7.
and adjusts the deforming mirrors to achieve optimal resolu- 8. Lee BB, Smith VC, Pokorny J, et al. Rod inputs to macaque ganglion cells.
Vision Res 1997;37:2813–28.
tion. This technology was first adopted for retinal imaging in 9. Crook JD, Davenport CM, Peterson BB, et al. Parallel ON and OFF cone bipolar
the 1990s by Dr David Williams to reduce ocular aberrations inputs establish spatially coextensive receptive field structure of blue-yellow
ganglion cells in primate retina. J Neurosci 2009;29:8372–87.
in the eyes.45 After its initial introduction, the AO imaging 10. Field GD, Greschner M, Gauthier JL, et al. High-sensitivity rod photoreceptor
system was considered to have great scientific and clinical input to the blue-yellow color opponent pathway in macaque retina.
application potential because it had the capability of imaging Nat Neurosci 2009;12:1159–64.
11. Bartlett NR. Dark adaptation and light adaptation. In: Graham CH, editor.
photoreceptors, the retinal pigment epithelium, retinal blood Vision and visual perception. New York: John Wiley; 1965. p. 185–207.
vessels and, potentially, ganglion cells at a high magnification. 12. Graham CH. Vision and visual perception. New York: John Wiley; 1965.
13. Shevell SK, Kingdom FA. Color in complex scenes. Annu Rev Psychol
For instance, AO made it possible to measure color percep- 2008;59:143–66.
tion46 or cell responses in the postreceptoral pathways47 associ- 14. Smith VC, Pokorny J. Spectral sensitivity of the foveal cone photopigments
between 400 and 500 nm. Vision Res 1975;15:161–71.
ated with tiny flashes of light stimulating a single cone in the 15. MacLeod DIA, Boynton RM. Chromaticity diagram showing cone excitation
eye. For color vision screening, in particular, the AO system by stimuli of equal luminance. J Opt Soc Am 1979;69:1183–5.
ACKNOWLEDGMENTS
The preparation of this chapter is partially supported by a 298.e1
National Institutes of Health grant (R01 EY019651). I express my
great appreciation to Drs Vivianne Smith and Joel Pokorny for
allowing me access to the materials they prepared for an earlier

Chapter 10
version of the chapter in a previous edition of Retina. I thank Dr
Margaret Lutze for her assistance in preparing this chapter.

Color Vision and Night Vision


16. Boynton RM, Kambe N. Chromatic difference steps of moderate size measured 38. Barbur J, Harlow A, Plant G. Insights into the different exploits of colour in the
along theoretically critical axes. Color Res Appl 1980;5:13–23. visual cortex. Proc Biol Sci 1994;258:327–34.
17. Derrington AM, Krauskopf J, Lennie P. Chromatic mechanisms in lateral 39. Seshadri J, Christensen J, Lakshminarayanan V, et al. Evaluation of the new
geniculate nucleus of macaque. J Physiol (Lond) 1984;357:241–65. web-based “Colour Assessment and Diagnosis” test. Optom Vision Sci 299
18. Lee BB, Martin PR, Valberg A. Sensitivity of macaque retinal ganglion cells to 2005;82:882–5.
chromatic and luminance flicker. J Physiol (Lond) 1989;414:223–43. 40. Regan BC, Reffin J, Mollon JD. Luminance noise and the rapid determina-
19. Pokorny J, Smith VC. Chromatic discrimination. In: Chalupa LM, Werner JS, tion of discrimination ellipses in colour deficiency. Vision Res 1994;34:

Chapter 10
editors. The visual neuroscience. Cambridge, MA: MIT Press; 2004. 1279–99.
pp. 908–23. 41. Melamud A, Simpson E, Traboulsi EI. Introducing a new computer-based test
20. Pokorny J, Smith VC. Wavelength discrimination in the presence of added for the clinical evaluation of color discrimination. Am J Ophthalmol
chromatic fields. J Opt Soc Am 1970;69:562–9. 2006;142:953–60.
21. Wright WD. The sensitivity of the eye to small colour differences. Proc Phys 42. Mancuso K, Mauck MC, Kuchenbecker JA, et al. A multi-stage color model
Soc (Lond) 1941;53:93–112. revisited: implications for a gene therapy cure for red-green colorblindness.
22. MacAdam DL. Visual sensitivities to color differences in daylight. J Opt Soc Adv Exp Med Biol 2010;664:631–8.
Am 1942;32:247–74. 43. Mancuso K, Hauswirth WW, Li Q, et al. Gene therapy for red-green colour
23. Cao D, Zele AJ, Smith VC, et al. S-cone discrimination for stimuli with spatial blindness in adult primates. Nature 2009;461:784–7.
and temporal chromatic contrast. Vis Neurosci 2008;25:349–54. 44. Tyson RK. Principles of adaptive optics. San Diego, CA: Academic Press;

Color Vision and Night Vision


24. Krauskopf J, Gegenfurtner K. Color discrimination and adaptation. Vision Res 1991.
1992;32:2165–75. 45. Liang J, Williams DR, Miller DT. Supernormal vision and high-resolution
25. von Kries J. Influence of adaptation on the effects produced by luminous retinal imaging through adaptive optics. J Opt Soc A 1997;14:2884–92.
stimuli. In: MacAdam DL, editor. Sources of color science. Cambridge, MA: 46. Hofer H, Singer B, Williams DR. Different sensations from cones with the same
MIT Press; 1905. photopigment. J Vis 2005;5:444–54.
26. Shevell SK, Cao D, editors. Chromatic assimilation: evidence for a neural 47. Sincich LC, Zhang Y, Tiruveedhula P, et al. Resolving single cone inputs to
mechanism. Oxford: Oxford University Press; 2003. visual receptive fields. Nat Neurosci 2009;12:967–9.
27. Neitz J, Neitz M. The genetics of normal and defective color vision. Vision Res 48. Williams DR. Imaging single cells in the living retina. Vision Res 2011;51:
2011;51:633–51. 1379–96.
28. Nathans J, Piantanida TP, Eddy RL, et al. Molecular genetics of inherited 49. Buck SL. Rod–cone interaction in human vision. In: Chalupa LM,
variation in human color vision. Science 1986;232:203–10. Werner JS, editors. The visual neuroscience. Cambridge, MA: MIT Press;
29. Working-Group-41. Procedures for testing color vision, NAS-NRC Committee 2004. p. 863–78.
on Vision. Washington, DC: National Academy Press; 1981. 50. Cao D, Pokorny J, Smith VC. Matching rod percepts with cone stimuli.
30. Pokorny J, Smith VC, Pinckers AJLG, et al. Classification of complete and Vision Res 2005;45:2119–28.
incomplete autosomal recessive achromatopsia. Graefes Arch Clin Exp 51. Pokorny J, Lutze M, Cao D, et al. The color of night: surface color perception
Ophthalmol 1982;219:121–30. under dim illuminations. Vis Neurosci 2006;23:525–30.
31. Nathans J, Hogness DS. Isolation and nucleotide sequence of the gene 52. Willmer EN. Low threshold rods and the perception of blue. J Physiol (Lond)
encoding human rhodopsin. Proc Natl Acad Sci U S A 1984;81:4851–5. 1949;111:17P.
32. Smith VC, Pokorny J, Pass AS. Color-axis determination on the Farnsworth- 53. Lie I. Dark adaptation and the photochromatic interval. Doc Ophthalmol
Munsell 100-hue Test. Am J Ophthalmol 1985;100:176–82. 1963;17:411–510.
33. Pokorny J, Smith VC, Verriest G, et al., editors. Congenital and acquired color 54. Stabell U, Stabell B. Wavelength discrimination of peripheral cones and its
vision defects. New York: Grune and Stratton; 1979. change with rod intrusion. Vision Res 1977;17:423–6.
34. Moreland JD, Kerr J. Optimization of stimuli for trit-anomaloscopy. Mod Probl 55. Buck SL, Knight RF, Bechtold J. Opponent-color models and the influence of
Ophthalmol 1978;19:162–6. rod signals on the loci of unique hues. Vision Res 2000;40:3333–44.
35. Pokorny J, Smith VC, Went LN. Color matching in autosomal dominant tritan 56. Volbrecht VJ, Nerger JL, Imhoff SM, et al. Effect of the short-wavelength-
defect. J Opt Soc Am 1981;71:1327–34. sensitive-cone mosaic and rods on the locus of unique green. J Opt Soc Am A
36. Weitz CJ, Miyake Y, Shinzato K, et al. Human tritanopia associated with two 2000;17:628–34.
amino acid substitutions in the blue-sensitive opsin. Am J Hum Genet 1992;50: 57. Pokorny J, Smithson H, Quinlan J. Photostimulator allowing independent
156. control of rods and the three cone types. Vis Neurosci 2004;21:263–7.
37. Nathans J, Davenport CM, Maumenee IH, et al. Molecular genetics of human 58. Cao D, Pokorny J, Smith VC, et al. Rod contributions to color perception: linear
blue cone monochromacy. Science 1989;245:831–8. with rod contrast. Vision Res 2008;48:2586–92.
Section 2 Retinal Diagnostics

Chapter Visual Acuity and Contrast Sensitivity


11  Gary S. Rubin

VISUAL ACUITY TESTS suitability for use with illiterate subjects. However, the Landolt
C test is not widely used because it has a guessing rate of 25%,
Introduction so an alternative is to standardize another optotype set by com-
Visual acuity is the most widely used measure of visual function. paring acuities obtained with it to Landolt C test acuities. The
In fact, visual function is often equated with visual acuity, Sloan letters,4 a set of 10 upper-case sans serif letters, are the most
thereby ignoring other important dimensions of visual stimuli, popular substitute. An acuity chart based on Sloan letters was
such as color and contrast. Visual acuity tests have proven to be developed for the Early Treatment Diabetic Retinopathy Study
useful for assessment of refractive error, screening for ocular (ETDRS)5 and is illustrated in Fig. 11.1. The original ETDRS chart
health, following the course of eye disease, evaluating the effec- has been replaced by the 2000 series revised ETDRS chart that
tiveness of medical and surgical treatment, prescribing aids for more accurately equates the difficulty of letters on all lines. The
the visually impaired, and setting vision standards for employ- ETDRS charts are the most widely used acuity charts for clinical
ment and driving. research.
Given the variety of its applications, it is not surprising that Chart layout
many different types of visual acuity tests have evolved. Gener- Careful attention must be paid to the layout of the acuity chart.
ally, these tests were developed with little concern for standard- The chart should follow a uniform progression of letter sizes,
ization. Since the 1980s, several attempts have been made to typically a 0.1 log unit (or 26%) reduction in size from line to
formulate standards for test design and administration. The line. The uniform progression ensures that a one-line loss will
Committee on Vision of the National Academy of Sciences- have the same meaning at any point on the chart and at any
National Research Council (NAS-NRC)1 has published stan- viewing distance. The same number of letters should appear
dards for clinical testing of visual acuity that are widely adopted on each line, and the spacing should be uniform, both within
in the USA, and the British Standards Institution2 has published and between lines. The spacing requirement results in a large
similar standards for the UK. The NAS-NRC standards are used chart, with the letters forming an inverted triangle. The
as the basis of this chapter. NAS-NRC recommends 8–10 letters per line, but studies6
suggest that as few as three letters are required for an accurate
Chart design estimate of visual function. The ETDRS chart uses five letters
Optotypes per line.
Most familiar acuity tests require the subject to identify letters Concern about uncontrolled “crowding” effects has led to
arrayed in rows of decreasing size. The so-called Snellen acuity further modifications of the ETDRS chart. Crowding refers to the
test is the prime example, although Snellen acuity now usually reduced visibility of letters when they are surrounded by other
refers to a way of reporting test results rather than to any par- letters. Crowding has a larger effect in some types of visual
ticular type of chart. To facilitate testing of young children and dysfunction, notably amblyopia7 and macular degeneration.8 For
people unfamiliar with the Latin alphabet, other optotype tests most acuity charts, letters at either the end of a line or at the top
based on the tumbling E, Landolt C, numerals, or simple pictures or bottom of the chart are less subject to crowding than are
of familiar objects are also used. Visual acuity can also be internal letters. To equalize crowding effects, contour interaction
assessed with grating patterns, but grating acuity often overes- bars may be added around the perimeter of the chart.9
timates Snellen acuity in patients with age-related maculopathy,3
typically by a factor of 2 or more. Testing procedure
The NAS-NRC recommends that the Landolt C test be used Acuity test distance
as the standard by which other acuity tests are compared. The ETDRS charts are available for a range of test distances from 4
Landolt C is a broken circle in which the width of the break meters (13 feet) to 2 meters (6.5 feet) and when used at the des-
and the stroke width are both equal to one-fifth the height of ignated distance can measure acuities from 20/10 to 20/200.
the C. The ring is shown with the break at one of four loca- However, given the logarithmic progression of letter sizes, they
tions, and the subject responds by saying “right,” “down,” “up,” can be used at any distance with an appropriate correction of the
or “left,” or by pointing in the appropriate direction. reported results. For patients with very poor acuity, the clinician
There are several advantages to the Landolt C test, including may resort to finger counting or hand motion. This strategy is
equal difficulty of all targets (unlike letters that vary in degree strongly discouraged by low-vision practitioners because it can
of difficulty), sensitivity to astigmatic refractive error, and be demeaning and depressing for the patient to be left with the
Fig. 11.1 The Early Treatment Diabetic
Retinopathy Study (ETDRS) acuity chart.
(Reproduced with permission from Ferris FL
III, Kassof PA, Bresnick GH, et al. New visual 301
acuity charts for clinical research. Am J
Ophthalmol 1982;94:91–96.)

Chapter 11
Visual Acuity and Contrast Sensitivity
impression that their vision is so poor it cannot even be mea- The relationship of visual acuity to letter contrast follows a
sured with an eye chart. Instead, it is recommended that the square-root law.14 For example, decreasing contrast by a factor
patient be moved closer to the chart. Using a test distance of of 2 would decrease acuity by roughly a factor of 1.4. The
50 cm and appropriate refractive correction it is possible to NAS-NRC recommends that letter contrast be at least 0.85.
reliably measure “counting finger” acuity with an ETDRS chart Transilluminated, projection, and reflective charts (wall charts)
(approximately 20/1460 ± 10%).10 “Hand motions” can be mea- can all meet these standards, but some transilluminated charts
sured with some electronic acuity tests (see below). Distance are deficient in luminance, and some projection systems lack
itself should have little effect on visual acuity, provided that the sufficient luminance and contrast. Accurate calibration requires
subject’s accommodative state and pupil size are controlled. a spot photometer, for which procedures are described in the
However, one study11 found that acuity changed by as much as NAS-NRC document.1
seven letters (more than one ETDRS line) when the test distance
Test administration
was reduced to less than 2 meters. The reason for this discrep-
Administration of visual acuity tests is simple and straightfor-
ancy remains unexplained.
ward. However, one detail often overlooked in clinical testing is
Luminance and contrast that the test must be administered in a “forced-choice” manner.
Whatever the test distance, the chart must be adequately illu- Rather than allowing the patient to decide when the letters
minated and of high contrast. Illumination standards vary from become indistinguishable, the patient should be required to
100 cd/m2 in the USA to 300 cd/m2 in Germany. Increasing guess the identity of each letter until a sufficient number of
chart luminance improves visual acuity in normal subjects, but errors are made to justify terminating the test. People differ in
reaches a plateau at about 200 cd/m2.12 Various types of visual their willingness to respond to questions when they are not
dysfunction can change the effects of luminance on acuity. For confident about the answers. A person with a conservative cri-
example, patients with retinitis pigmentosa may show a terion answers only when absolutely certain about the identity
decrease in acuity at higher luminance, whereas patients with of the letter, whereas a person with a liberal criterion ventures
age-related macular degeneration often continue to improve a guess for any letter that is even barely discernible. These two
at luminances well above the normal plateau.13 A luminance people may receive different acuity scores because of differences
standard of 100 cd/m2 can be justified because it represents in their criteria rather than because of variations in visual func-
good room illumination for ordinary reading material. Fur- tion. This is not merely a theoretical concern. Several studies15,16
thermore, most of the currently proposed standards would have shown that criterion-dependent test procedures lead to
yield the same acuity scores, plus or minus one letter (assum- inaccurate and unreliable test results. Forced-choice procedures
ing five letters per line and a 0.1 log unit size progression) in are criterion-free because the examiner, rather than the observer,
normal subjects. determines whether the letter is correctly identified.
Scoring is the Jaeger J notation. Jaeger notation is based on a numeric
Until recently, visual acuity tests were usually scored line by line scale (J1, J2, and so on) that follows no logical progression except
302 with the patient being given credit for a line when a criterion that larger numbers correspond to larger print sizes. Further-
number of letters were identified correctly. The NAS-NRC more, print with the same J specification can vary by as much as
recommends that at least two-thirds of the letters on a line be 90% from one test manufacturer to another.20
Alternatives to the Jaeger notation are the typesetter’s point
Section 2

correctly identified to qualify for passing. Allowing a small pro-


portion of errors improves test reliability.17 For tests that follow system, the British N system, and the M notation introduced by
the recommended format of an equal number of letters on each Sloan. The typesetter’s point is commonly used to specify letter
line and a constant progression of letter sizes, it is preferable to size for printed text and is equal to 0.32 mm (1/72 inch).
give partial credit for each letter correctly identified. This is com- However, the measurement refers to the size of the metal slug
monly done by counting the number of letters read correctly on that contains the letter and varies from one font to another. A
study21 of the effect of font on reading speed showed that the
Retinal Diagnostics

the entire chart and converting this to an acuity score by means


Retinal Imaging and Diagnostics

of a simple formula that values each letter as L/N, where L = nominal sizes of printed text can be very misleading. Of four
difference in acuity between adjacent lines and N = number of fonts, all labeled as 12 point, one was much more legible than
letters per line. So for a chart with five letters per line and a 0.1 the other three. But it turned out that the more legible font was
logMAR (see below) progression from line to line (such as the actually larger than the others and when equated for real size
standard ETDRS chart) each correct letter is worth 0.1/5 = 0.02 there was no difference in legibility.
logMAR. Although differences between scoring methods are The British N system standardized the point size specification
usually small, it has been shown2,6,18 that letter-by-letter scoring by adopting the Times Roman font. Sloan’s M notation,4 widely
is more reproducible than line-by-line scoring. used in the USA, is standardized according to the height of a
The most familiar method for reporting visual acuities is the lower-case “x.” A lower-case 1M letter subtends 5 minarc at a
Snellen fraction. The numerator of the Snellen fraction indicates 1-meter viewing distance and corresponds roughly to the size of
the test distance, and the denominator indicates the relative size ordinary newsprint. None of the print size specifications can be
of the letter, usually in terms of the distance at which the stroke used for quantitative comparisons unless viewing distance is
width would subtend a visual angle of 1 minute. Thus “20/40” also specified. For example, 1M print read at a distance of 40 cm
indicates that the actual test distance was 20 feet and that the would be recorded as 0.40/1.00M.
strokes of the letters would subtend 1 minute of arc at 40 feet. Words versus continuous text
To simplify comparison of acuities measured at different dis- One issue that remains unresolved is whether near acuity tests
tances, the minimum angle of resolution (MAR) should be used. should be based on unrelated words or meaningful text. An
The MAR is the visual angle corresponding to stroke width in argument in favor of unrelated words is that contextual informa-
minutes of arc and is equal to the reciprocal of the Snellen frac- tion promotes guessing and may lead to an overestimate of near
tion. Visual acuities are frequently converted to log10 and acuity.22 In addition, the presence of semantic context introduces
reported as “logMAR.” For example, 20/20 acuity corresponds variability because of cognitive intellectual factors that may
to a MAR of 1 minute of arc, or a logMAR of 0, and 20/100 acuity mask the visual factors of primary concern.23 On the other side
corresponds to a MAR of 5 minutes of arc, or a logMAR of 0.7 it is argued that the main reason for measuring near acuity is to
(as does an acuity of 2/10 or 6/30). The MAR and logMAR scales gain information about reading performance. Since context is
increase with worsening acuity. One often sees visual acuities normally available to the reader, a reading test that includes
reported as the decimal equivalent of the Snellen fraction. meaningful text will be a better indicator of everyday perfor-
However acuities are more normally distributed when con- mance. Fortunately, reading speed for meaningful text is highly
verted to logMAR rather than decimal values. Moreover, deci- correlated with reading speed for unrelated words.24
mals give the false impression that acuity scores can be equated The MNREAD test25 uses meaningful text to evaluate near
with overall loss of visual function. For example, an acuity of 0.1 acuity. The test is composed of 19 standardized sentences in a
(the decimal equivalent of 20/200) may misleadingly suggest logarithmic progression of sizes. Each sentence is 55 characters
that the patient has retained 10% of residual visual function. and the words are drawn from a controlled vocabulary. The test
can be used to measure reading acuity (the smallest print size
Near and reading acuity tests that can be read), maximum reading speed, and critical print size
Near acuity is usually tested to evaluate reading vision. These (the smallest print size for maximum reading speed). The test–
tests are particularly important for prescribing visual aids for retest variability for the MNREAD test has been assessed in
persons with low vision. Near acuity has been shown to be a patients with macular disease26 and the coefficient of repeatabil-
better predictor of the optimal magnification needed by visually ity was found to be greater than 65 words/minute. The high
impaired readers than traditional distance acuity.19 level of variability may be due, in part, to the short sentences.
Although some near acuity tests are simply reduced versions The International Reading Speed Test (IReST)27 uses 150-word
of distance acuity charts, most tests consist of printed text, either paragraphs of continuous text to measure reading speed, which
unrelated words or complete sentences or paragraphs, covering should yield more reproducible measures. The IReST is available
a range of sizes. Near acuity tests are even less standardized than in 17 languages.
distance acuity tests.
Specifying letter size Electronic acuity tests
As with all acuity tests, the most critical parameter is the visual Video-based acuity tests have been available since the 1980s, but
angle subtended by the optotype. Many systems have been did not become popular until the introduction of inexpensive
devised for specification of print size. One of the most common LCD monitors in recent years. These electronic tests include
computer software that can be used with the experimenter’s own that contrast sensitivity tests are more sensitive to early eye
computer hardware (such as the Freiburg Acuity and Contrast disease than visual acuity. While this may be true, much of the
Test (FrACT)), devices that display optotypes under operator apparent difference in sensitivity is due to careless measurement 303
control (such as the Test Chart 2000) and systems that adminis- of visual acuity (poorly designed charts and test procedures).
ter, score, and store the results of the acuity measurement (such Even if the test were more sensitive, its lack of specificity for

Chapter 11
as the E-ETDRS and COMPlog systems). distinguishing between ocular and retinal/neural disorders
Electronic tests offer several potential advantages over paper- limits its usefulness as a screening test.35
based tests: The real value of clinical contrast sensitivity testing is to gain
a better understanding of the impact of visual impairment on
1. multiple types of test, such as acuity, contrast sensitivity, functional ability. Several studies have demonstrated that con-
and stereoacuity, with one instrument trast sensitivity is useful for understanding the difficulties in
2. better randomization of optotypes. Each test administration performing everyday visual tasks faced by older people with

Visual Acuity and Contrast Sensitivity


can use a different arrangement of letters instead of being essentially normal vision36 and by patients with retinal disease.37,38
constrained to two or three printed charts Studies have shown that contrast sensitivity loss leads to mobil-
3. easier standardization of luminance and contrast, although if ity problems and difficulty recognizing signs or faces,39 even
calibration instructions are ignored, luminance and contrast when adjusted for loss of acuity.40
errors can be greater than for paper tests The association of contrast sensitivity with functional ability
4. the promise of advanced testing algorithms that reduce argues in favor of including contrast sensitivity measurements
testing time and/or increase measurment accuracy and in clinical trials. Although visual acuity is the most common
precision. So far the electronic systems have not lived up to primary visual outcome measure, several studies have included
this promise with test times as long as or longer and contrast sensitivity as a secondary outcome. Considering both
measurement accuracy and precision no better than visual acuity and contrast sensitivity when assessing the out-
conventional chart tests.28 One test, the E-ETDRS system comes of clinical trials may provide a more complete picture of
used in clinical trials for treatment of central retinal vein the effects of treatment on vision than either measure alone.
occlusion,29 is reported to take longer without any increase Examples where contrast sensitivity has been used as a second-
in reliability.28 ary outcome include the Optic Neuritis Treatment Trial,41 the
TAP study of photodynamic therapy for age-related macular
CONTRAST SENSITIVITY TESTS degeneration (AMD),42 and the ABC trial of bevacizumab
for AMD.43
Introduction
Visual acuity has been and will probably continue to be the Methods
most often used clinical measure of visual function. However, Common contrast sensitivity tests
contrast sensitivity testing has been widely promoted as an
Traditional methods for measuring contrast sensitivity require
important adjunct or even replacement for visual acuity testing.
relatively expensive and sophisticated equipment – typically
Acuity measures the eye’s ability to resolve fine detail but
a computer-controlled video monitor – and employ time-
may not adequately describe a person’s ability to see large
consuming psychophysical procedures. However, several
low-contrast objects such as faces. Contrast sensitivity testing
simpler contrast sensitivity tests have been developed primarily
was originally developed as a research tool by engineers and
for clinical use. These include the Functional Acuity Contrast
vision scientists interested in characterizing normal visual func-
Test44 (FACT, replacement for the popular Vistech VCTS chart)
tion. For theoretical reasons, most investigators have used
and the CSV-1000,45 sine-wave grating tests in chart form, and
sine-wave grating stimuli, patterns consisting of alternating
various low-contrast optotype tests such as the Lea test,46 the
light and dark bars, which have a sinusoidal luminance profile.
Pelli–Robson letter chart,17 the Melbourne Edge Test47 and the
Sine-wave gratings vary in spatial frequency (bar width) and
Mars Letter Contrast Sensitivity Test.48 Examples of three of
contrast. A contrast sensitivity function (CSF) is derived by
the most commonly used clinical contrast sensitivity tests are
measuring the lowest detectable contrast across a range of
illustrated in Fig. 11.2.
spatial frequencies.
Gratings versus optotypes
Utility of contrast sensitivity tests A thorough discussion of the relative merits of the various
For people with normal vision, contrast sensitivity and visual tests is beyond the scope of this chapter; however, a few
acuity are correlated. However, various types of visual dysfunc- salient points are worth noting. Various investigators disagree
tion, including cerebral lesions,30 optic neuritis related to mul- about whether measurement of an entire CSF is necessary or
tiple sclerosis,31 glaucoma,32 diabetic retinopathy,33 and cataract,34 whether a single measure of contrast sensitivity is adequate
may cause a reduction in contrast sensitivity despite near-normal for clinical purposes. Proponents of the sine-wave grating tests
visual acuity. This led to the suggestion that contrast sensitivity argue that visual dysfunction can cause reductions in contrast
might serve as a tool for differential diagnosis and screening. sensitivity over a limited range of spatial frequencies, which
However, there is no pattern of CSF loss that is unique to any would be missed by more global measures of contrast sensi-
particular vision disorder. The types of CSF measured in patients tivity. On the other hand, advocates of global measures note
with macular disease or glaucoma can be similar to the CSFs that contrast sensitivities at specific spatial frequencies tend
measured in cataract patients, although detailed analyses with to be highly correlated with one another, and they maintain
targets of different size or at different retinal eccentricities may that overall changes in contrast sensitivity are clinically more
help distinguish between various causes of the loss. It is argued important than subtle bumps and wiggles in the CSF. Data
Fig. 11.2 Commonly used clinical contrast
sensitivity tests. (A) Vistech VCTS 6500.
(Courtesy of Vistech Consultants, Dayton,
304 OH.) (B) Lea numbers low-contrast acuity
test. (Courtesy of Good-Lite, Streamwood,
IL.)
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

B
Interpretation of clinical versus statistical
significance: an example from the literature
One of the vexing problems with contrast sensitivity testing is 305
how to interpret the clinical significance of test scores. After
many decades of acuity testing, we have arrived at a consensus

Chapter 11
that a doubling of the MAR (increase of 0.3 logMAR or 15 ETDRS
letters) represents a meaningful change in acuity. Recent data
from large population-based studies suggest that a doubling of
contrast threshold (reducing sensitivity by 0.3 logCS units or six
letters on the Pelli–Robson chart) has a comparable impact on
task performance and quality of life.36,39
After several decades of laboratory investigation, contrast

Visual Acuity and Contrast Sensitivity


sensitivity testing is taking its place alongside visual acuity in
clinical vision research. While early claims that contrast sensi-
tivity would replace visual acuity proved to be exaggerated,
this additional test does have an important role to play. Contrast
sensitivity may not be particularly useful for differential diag-
nosis and screening, but its close association with everyday
task difficulty has made it an important outcome measure for
assessing treatment safety and efficacy.

REFERENCES
1. NAS-NRC. Recommended standard procedures for the clinical measurement
and specification of visual acuity. Report of working group 39. Adv
Ophthalmol 1980;41:103–48.
2. British Standards Institution. Specification for test charts determining distance
visual acuity. London: British Standards House.
3. White JM, Loshin DS. Grating acuity overestimates Snellen acuity in patients
with age-related maculopathy. Optom Vis Sci 1989;66:751–5.
4. Sloan LL, Rowland WM, Altman A. Comparison of three types of test target
C for the measurement of visual acuity. Q Rev Opthalmol 1952;8:4–16.
5. Ferris FL, Kassoff A, Bresnick GH, et al. New visual acuity charts for clinical
research. Am J Ophthalmol 1982;94:91–6.
Fig. 11.2 Cont’d (C) Pelli–Robson letter sensitivity chart. (Reproduced 6. Raasch T, Bailey I, Bullimore M. Repeatability of visual acuity measurement.
with permission from Pelli DG, Robson JG, Wilkins AJ. The design of Optom Vis Sci 1998;75:342–8.
a new letter chart for measuring contrast sensitivity. Clin Vis Sci 7. Flom MC, Bedell HE. Identifying amblyopia using associated conditions,
1988;2:187–200.) acuity, and nonacuity features. Am J Optom Physiol Opt 1985;62:153–60.
8. Kitchin JE, Bailey I. Task complexity and visual acuity in senile macular
degeneration. Aust J Optom 1981;64:235–42.
9. Strong G, Woo GC. A distance visual acuity chart incorporating some new
design features. Arch Ophthalmol 1985;102:44.
10. Schulze-Bonsel K, Feltgen N, Burau H, et al. Visual acuities “hand motion”
and “counting fingers” can be quantified with the Freiburg visual acuity test.
from large-scale studies of vision impairment indicate that Invest Ophthalmol Vis Sci 2006;47:1236–40.
11. Dong LM, Hawkins BS, Marsh MJ. Consistency between visual acuity scores
global measures of contrast sensitivity are valuable predictors obtained at different test distances: theory vs observations in multiple studies.
of difficulty in everyday life.39 Arch Ophthalmol 2002;120:1523–33.
12. Sheedy JE, Bailey IL, Raasch TW. Visual acuity and chart luminance. Am J
Test design and procedure Optom Physiol Opt 1984;61:595–600.
13. Sloan LL, Habel A, Feiock K. High illumination as an auxiliary reading aid in
In order to be useful, contrast sensitivity must be measured diseases of the macula. Am J Ophthalmol 1973;76:745–57.
accurately and reliably. Many of the same principles of test 14. Legge GE, Rubin GS, Luebker A. Psychophysics of reading. V. The role of
contrast in normal vision. Vision Res 1987;27:1165–77.
design previously discussed for visual acuity tests apply to con- 15. Higgins KE, Jaffe MJ, Coletta NJ, et al. Spatial contrast sensitivity. Importance
trast sensitivity tests. Most important, the test should employ a of controlling the patient’s visibility criterion. Arch Ophthalmol 1984;102:
criterion-free procedure, a uniform progression of contrasts, and 1035–41.
16. Rubin GS. Reliability and sensitivity of clinical contrast sensitivity tests.
an adequate number of trials at each contrast to make a reliable Clin Vis Sci 1988 1988;2:169–77.
estimate of sensitivity. Most of the optotype tests conform to 17. Pelli DG, Robson JG, Wilkins AJ. The design of a new letter chart for measuring
contrast sensitivity. Clin Vis Sci 1988;2:187–99.
good design principles and produce reliable results, with the 18. Arditi A, Cagenello R. On the statistical reliability of letter-chart visual acuity
Mars test outperforming the very popular Pelli–Robson test.49 measurements. Invest Ophthalmol Vis Sci 1993;34:120–9.
19. Lovie-Kitchin JE, Whittaker SG. Prescribing near magnification for low vision
Sine-wave grating charts tend to be less reliable because they patients. Clin Exp Optom 1999;82:214–24.
have a limited number of trials to make measurements at several 20. Jose RT, Atcherson RM. Type-size variability for near-point acuity tests.
Am J Optom Physiol Opt 1977;54:634–8.
spatial frequencies.50 21. Rubin GS, Feely M, Perera S, et al. The effect of font and line width on reading
Some of the electronic acuity testing systems can also measure speed in people with mild to moderate vision loss. Ophthalm Physiol Opt
contrast sensitivity, including the Vision Test 2000 and FrACT. 2006;26:545–54.
22. Sloan LL, Brown DJ. Reading cards for selection of optical aids for the partially
However, it is difficult to display a wide-enough range of con- sighted. Am J Ophthalmol 1963;55:1187–99.
trasts to measure normal thresholds, and accurate calibration 23. Baldasare J, Watson GR, Whittaker SG, et al. The development and evaluation
of a reading test for low vision individuals with macular loss. J Visual Impair-
of the display monitor is critical. One study found that the ment Blindness 1986;1986:785–9.
computer-based test was less reliable than paper charts,49 pre- 24. Legge GE, Ross JA, Luebker A, et al. Psychophysics of reading. VIII. The
Minnesota Low-Vision Reading Test. Optom Vis Sci 1989;66:843–53.
sumably due to problems generating low-contrast patterns with 25. Mansfield JS, Legge GE, Bane MC. Psychophysics of reading. XV: Font effects
LCD displays. in normal and low vision. Invest Ophthalmol Vis Sci 1996;37:1492–501.
26. Patel PJ, Chen FK, Da Cruz L, et al. Test–retest variability of reading 38. Rubin GS, Legge GE. Psychophysics of reading. VI. The role of contrast in low
performance metrics using MNREAD in patients with age-related macular vision. Vision Res 1989;29:79–91.
degeneration. Invest Ophthalmol Visual Sci 2011;52:3854–9. 39. Rubin GS, Bandeen-Roche K, Huang GH, et al. The association of multiple
306 27. Hahn GA, Penka D, Gehrlich C, et al. New standardised texts for assessing visual impairments with self-reported visual disability: SEE project.
reading performance in four European languages. Br J Ophthalmol Invest Ophthalmol Vis Sci 2001;42:64–72.
2006;90:480–4. 40. Rubin GS, Bandeen-Roche K, Prasada-Rao P, et al. Visual impairment and
28. Laidlaw DA, Tailor V, Shah N, et al. Validation of a computerised logMAR disability in older adults. Optom Visual Sci 1994;71:750–60.
visual acuity measurement system (COMPlog): comparison with ETDRS 41. Beck RW, Cleary PA, Anderson Jr MM, et al,. A randomized, controlled trial
Section 2

and the electronic ETDRS testing algorithm in adults and amblyopic children. of corticosteroids in the treatment of acute optic neuritis. N Engl J Med
Br J Ophthalmol 2008;92:241–4. 1992;326:581–8.
29. Scott IU, Ip MS, VanVeldhuisen PC, et al. A randomized trial comparing the 42. Rubin GS, Bressler NM. Effects of verteporfin therapy on contrast on sensitiv-
efficacy and safety of intravitreal triamcinolone with standard care to treat ity: results from the Treatment of Age-related macular degeneration with Pho-
vision loss associated with macular edema secondary to branch retinal vein todynamic therapy (TAP) investigation – TAP report no 4. Retina 2002;22:
occlusion: the Standard care vs Corticosteroid for Retinal Vein Occlusion 536–44.
(SCORE) study report 6. Arch Ophthalmol 2009;127:1115–28. 43. Patel PJ, Chen FK, Da Cruz L, et al. Contrast sensitivity outcomes in the ABC
30. Bodis-Wollner I. Visual acuity and contrast sensitivity in patients with cerebral trial: a randomized trial of bevacizumab for neovascular age-related macular
Retinal Diagnostics

lesions. Science 1972;178:769–71. degeneration. Invest Ophthalmol Vis Sci 2011;52:3089–93.


Retinal Imaging and Diagnostics

31. Regan D, Silver R, Murray TJ. Visual acuity and contrast sensitivity in multiple 44. Ginsburg A. Next generation contrast sensitivity testing. In: Rosenthal BP, Cole
sclerosis-hidden visual loss: an auxiliary diagnostic test. Brain 1977;100: R, editors. Functional assessment of low vision. St Louis: Mosby Year Book;
563–79. 1996. p. 77–88.
32. Bron AJ. Contrast sensitivity changes in ocular hypertension and early 45. Pomerance GN, Evans DW. Test–retest reliability of the CSV-1000 contrast test
glaucoma. Surv Ophthalmol 1989;33(Suppl):405–6; discussion 9–11. and its relationship to glaucoma therapy. Invest Ophthalmol Vis Sci 1994;35:
33. Howes SC, Caelli T, Mitchell P. Contrast sensitivity in diabetics with retinopa- 3357–61.
thy and cataract. Aust J Ophthalmol 1982;10:173–8. 46. Jarvinen P, Hyvarinen L. Contrast sensitivity measurement in evaluations of
34. Rubin GS, Adamsons IA, Stark WJ. Comparison of acuity, contrast sensitivity, visual symptoms caused by exposure to triethylamine. Occup Environ Med
and disability glare before and after cataract surgery. Arch Ophthalmol 1997;54:483–6.
1993;111:56–61. 47. Verbaken JH, Johnston AW. Population norms for edge contrast sensitivity.
35. Legge GE, Rubin GS. The contrast sensitivity function as a screening test: Am J Optom Physiol Opt 1986;63:724–32.
A critique. Am J Optom Physiol Opt 1986;63:265–70. 48. Arditi A. Improving the design of the letter contrast sensitivity test.
36. West SK, Rubin GS, Broman AT, et al. How does visual impairment Invest Ophthalmol Vis Sci 2005;46:2225–9.
affect performance on tasks of everyday life? The SEE project. Salisbury Eye 49. Thayaparan K, Crossland MD, Rubin GS. Clinical assessment of two new
Evaluation. Arch Ophthalmol 2002;120:774–80. contrast sensitivity charts. Br J Ophthalmol 2007;91:749–52.
37. Lennerstrand G, Ahlström CO. Contrast sensitivity in macular degeneration 50. Pesudovs K, Hazel CA, Doran RM, et al. The usefulness of Vistech and FACT
and the relation to subjective visual impairment. Acta Ophthalmol (Copenh) contrast sensitivity charts for cataract and refractive surgery outcomes research.
1989;67:225–33. Br J Ophthalmol 2004;88:11–6.
Retinal Diagnostics Section 2

Visual Fields in Retinal Disease Chapter

Rajeev S. Ramchandran, Steven E. Feldon


12 
One need only review the classic atlas by Dr J. Donald M. Gass1 represented by a hole in the “hill” about 15° temporal to the
to recognize that most retinal diseases are readily diagnosed by foveal “peak.”
careful ophthalmoscopic examination. However, for the oph- Perimetric tests have been developed that systematically
thalmologist to understand and document visual consequences measure the level of light sensitivity in the visual field. Two basic
of retinal disease, qualitative and quantitative assessment is criti- types exist: kinetic perimetry and static perimetry. In kinetic
cal. In particular, visual field testing helps with the correlation perimetry a target is moved from outside the potentially seeing
of structural changes in the retina or elsewhere in the visual area toward the seeing area until it is detected. In static perim-
pathway with deficits in function. For example, AM is a 70-year- etry a target located within a potentially seeing area can be
old woman diagnosed as having an old central retinal vein increased in size or intensity until it is detected. With either
occlusion (CRVO) in the left eye and surface wrinkling retino­ method a region of vision can be defined in relation to a test
pathy in both eyes. She also had been treated for elevated intra- stimulus of given size, hue, brightness, and uniform level of
ocular pressure. Fluorescein angiography of the left eye showed background illumination. For instance, under standardized con-
shunting vessels around the optic nerve head, thought to be ditions, the central visual field of about 30° can be mapped with
consistent with the old vein occlusion. However, progressive a moving white target of 3 mm at a distance of 2 meters.3
bilateral visual field loss (Fig. 12.1) over 2 years suggested the
need for further investigation. A computed tomography (CT) METHODS OF VISUAL FIELD TESTING
scan demonstrated a large subfrontal meningioma compressing
There is no single best test for the evaluation of visual fields
both optic nerves (Fig. 12.2).
in retinal disease. Depending on the location and extent of the
Visual field tests can be qualitative or quantitative. They span
disease, the examiner must choose from an ever-increasing
multiple formats from the simplest confrontation field or Amsler
selection of sensitive qualitative and quantitative perimetric
grid to the most advanced quantitative kinetic, static, and micro-
techniques.
perimetry. For all visual field tests, reliability and reproduc-
ibility are important. There are currently three main uses for Qualitative techniques
visual field tests in patients with retinal pathology: (1) to deter-
Vision may be extremely poor in many patients with widespread
mine the current level of visual field loss and monitor progres-
visual dysfunction resulting from retinal disease. In such
sion; (2) to evaluate the effects of treating retinal pathology on
instances visual fields may be obtained only by confrontation
visual function; and (3) to correlate functional and structural
testing. However, this technique also may be used to detect even
changes in the retina.
subtle relative field defects and scotomata. Confrontation testing
This chapter reviews the use of visual fields and the principles
is performed with the examiner facing the patient, usually at
of perimetry relevant to retinal disease. Visual field defects cor-
about arm’s length. The peripheral vision of the patient is evalu-
responding to common retinal diseases are described, with an
ated in relation to that of the examiner’s. To test the patient’s
emphasis on clinical situations for which perimetry is useful
right eye, the examiner occludes the right eye and aligns the left
diagnostically. The value of specialized visual field testing in
eye so that both patient and examiner share a common visual
clinical research is also reviewed.
axis. The examiner then moves the hand or test object from
outside the visual field toward fixation. Relative preservation of
PRINCIPLES OF PERIMETRY peripheral vision can be established if the examiner places both
hands or two test objects in different seeing quadrants of the
The island of Traquair patient and asking the patient to identify the clearer object.
The single most important concept in understanding the visual Central visual function is estimated by having the patient look
field is depicted by the island of Traquair, which is defined as toward the examiner’s nose or, if vision is very poor, toward the
a “hill of vision” surrounded by a “sea of blindness.”2 The shape direction of the examiner’s voice. The patient describes which
of the normal visual field is oval. There is decreasing sensitivity parts of the examiner’s face are missing or most clearly seen.
with increasing eccentricity in the field, as the elevation cor- Results are recorded either descriptively or with a simple
responds to the sensitivity of the field, and the flat plane cor- drawing.
responds to location within the field (Fig. 12.3). From the Although the tangent screen can be used quantitatively, it is
point of fixation, the field extends 60° superiorly, 60° nasally, most effective as a qualitative tool for estimation of visual loss
70–75° inferiorly, and 100–110° temporally. The blind spot is in the central field. A 1-meter screen can be used to evaluate
308
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

B C

Fig. 12.1 (A, B) Fluorescein angiograms of a 70-year-old woman who has had progressive loss of vision in both eyes over 2 years. She has
controlled ocular hypertension and surface-wrinkling retinopathy. The history of an old central retinal vein occlusion in the left eye is supported by
the slightly dilated and tortuous retinal veins and the multiple shunting capillaries on the surface of the disc (B). (C) Progressive field loss,
documented in both eyes over 3 years, is inconsistent with the ophthalmoscopic appearance. A computed tomographic scan was ordered, the
results of which are shown in Figure 12.2.

Kinetic
perimetry

Static
perimetry

Fig. 12.3 The “island,” or “hill of vision,” proposed by Traquair is


surrounded by a “sea of blindness.” The height of the island
represents increasing sensitivity. Using kinetic perimetry, the island
is intercepted by a moving target of fixed size. Using static perimetry,
a target’s visibility is increased in size or luminosity until it descends
on to the island. The blind spot located 15° temporal to fixation is
absolute, creating a small “well” in the sensitivity contour. (Courtesy of
Steven Newman, MD.)

Fig. 12.2 Computed tomographic scan from the patient described in


Figure 12.1 demonstrates a large subfrontal meningioma with bilateral
optic nerve compression. Surgical resection improved sensorium but
not vision.
suspected midperipheral field loss, but a 2-meter screen is essen- drug therapy is becoming an important aspect of patient care.
tial if the intent is to map central scotomata or the physiologic Also, quantitative records are invaluable for achieving new
blind spot. The sensitivity of the test can be increased with the insights into the types of visual disturbances found in various 309
use of smaller test objects and by seeking subjective responses retinal diseases.
about the quality (e.g., blurry, dim, flickering, faded) of seen Both Goldmann and Tübinger perimetry require examiners

Chapter 12
targets. Most important, conical visual field constriction of with a high degree of training. The Goldmann visual field (GVF)
retinal origin can be readily distinguished from tubular fields of perimeter uses a kinetic strategy, although static perimetry is
nonorganic visual loss by using large test objects at varying possible (Fig. 12.7). It is especially useful for monitoring periph-
distances. With a doubling of the distance to the screen, there eral visual field loss and large scotomata. It has limited capabili-
must be a doubling in size of the test object used. Although not ties for the evaluation of small central scotomata. The Tübinger
routinely available or performed in a retinal specialist’s office, perimeter uses a static strategy. It is a sensitive but time-
no other form of perimetric evaluation is as effective in discrimi- consuming technique for identifying small or large relative field

Visual Fields in Retinal Disease


nating real from fictitious visual field constriction (Fig. 12.4). defects once the approximate region of the field loss has been
The Amsler grid is another extremely important qualitative
tool for the evaluation of central vision. It is routinely used in
regular self-monitoring of vision between eye exams by patients Fig. 12.5 The
with macular degeneration or other progressive macular pathol- Amsler grid is
ogy to detect early signs of disease progression. In its original viewed at 35.5 cm
(14 inches), using
form, the patient fixates on the center dot of a white grid on a A B
the central dot for
black background from a distance of 35.5 cm (14 inches) (Fig. fixation. Regions of
12.5). The patient describes the appearance of the grid pattern, metamorphopsia (C),
especially in regard to missing areas (scotomata) and distortion scotoma (B), and
blur (A) are noted.
(metamorphopsia). The sensitivity for detection of macular
lesions can be increased with the use of red grids with or without
the use of crossed polarizing lenses (Fig. 12.6) to decrease the C
light entering the patient’s eye.4

Quantitative techniques
Quantitative techniques of perimetry are utilized for both diag-
nosis of disease and following the course of disease. Documenta-
tion of visual recovery in eyes with retinal pathology after
treatment, such as retinal detachment surgery, laser therapy,
anti-inflammatory and antivascular endothelial growth factor

90°
30°
25°
135° 45°
20°

15°
10°

180° 0°

225° 315°

270°
10/2000 mm 5/2000 mm
Fig. 12.6 The sensitivity of the Amsler grid test can be increased by
using red lines on a black background rather than black or white lines
Fig. 12.4 A tangent screen field from a hysterical or malingering on contrasting backgrounds. Further sensitivity may be achieved by
patient fails to show expansion of the visual field with doubling of both using crossed-polarizing glasses that decrease the amount of light
test object size and distance. entering the eye. (Courtesy of Alfredo Sadun, MD.)
Fig. 12.7 The Goldmann visual field strategy
70 developed by Armaly and Drance uses
60 kinetic targets (arrows) to map the peripheral
310 isopters and suprathreshold static targets
(dots) to check central visual field function.
50
(Reproduced with permission from Rock WJ,
Drance SM, Morgan RW. Visual field
Section 2

40
screening in glaucoma. An evaluation of the
Armaly technique for screening glaucomatous
30
visual fields. Arch Ophthalmol 1973;89:
287–90.)
20

10
Retinal Diagnostics
Retinal Imaging and Diagnostics

90 80 70 60 50 40 30 20 20 30 40 50 6 0 7 0 8 0 90

10

20

30

40

50

60

70

quantitative threshold perimetry is ideal for the evaluation of


asb retinal disease. Randomized sequences for stimulus presentation
40 0.1 within the visual field enhance the possibility of detecting small
35 0.32 or irregular field defects. Density of points can be varied to
characterize fully field losses that are either focal or diffuse.
30 1 Although the HVF has the ability to test 60° of visual field, this
25 3.2 protocol is very time-consuming. The standard protocols used
are the 30-2, 24-2, and 10-2, which measure the central 30, 24,
20 10 and 10° respectively. The choice of field is based on the region
15 32 of pathology in the visual field. The dash 2 (-2) refers to protocol
2, the analysis which points on either side of the vertical and
10 100 horizontal meridians rather than just on the vertical and hori-
05 320 zontal axis as performed in protocol 1. Thus, both the 24-2 and
30-2 protocols test points 3° from the horizontal and vertical axis
0 1000 as well as points at 6° intervals beyond this region in the central
30° 20° 10° 0° 10° 20° 30°
24 or 30° respectively. A 10-2 protocol tests points 1° from the
horizontal and vertical meridian as well as points at 2° beyond
Fig. 12.8 The Tübinger perimeter produces a static profile of visual
sensitivity through any chosen meridian. (Reproduced with permission this region in the central 10° and thus is more sensitive for detect-
from Harrington DO. The visual fields: a textbook and atlas of clinical ing subtle field losses in the macular region.6
perimetry. St Louis: Mosby, 1981.) Newer algorithms such as the Swedish interactive threshold
algorithm (SITA) have been developed to increase efficiency and
decrease variability in the standard Humphrey strategy. This
determined. Laborious Tübinger profiles are usually made only strategy uses normative and real-time data from patients during
across a single meridian of the visual field, limiting its screening the test to update estimates of thresholds and adjust presentation
capabilities (Fig. 12.8). times of stimuli continually. Short-wavelength automated
Quantitative perimetry received a tremendous boost with the perimetry (SWAP) is a variant on the usual white light stimulus
advent of automated, computerized methodology. Suprathresh- used in standard threshold testing. SWAP isolates the S-cone
old automated visual screeners, such as the Fieldmaster 101 and system (blue–yellow pathway), using a blue stimulus on a
the Dicon units, did much to popularize visual field testing in yellow background. Normal, age-corrected retinal sensitivity
the ophthalmic community. Pseudothreshold algorithms have values have been established, increasing the utility in clinical
been developed for these machines to enhance their quantitative testing.7,8 Care must be taken when using this technique to
capabilities.5 However, threshold perimeters, such as the Octopus correct for chromatic filtering effects from the crystalline lenses.9
and the Humphrey visual field (HVF) analyzer, have surpassed Longer test duration, increased variability, and learning effects
the capabilities of even the Tübinger instrument. Automated, make SWAP most applicable to conditions for which early
detection has important therapeutic implications and requires Perimetric tests are being developed to screen for early
careful supervision of the patient by the tester during visual field changes in retinal diseases affecting the macula. Rarebit testing,
testing.10 In each case, the ability to map statically determined or the matching of test targets to receptive field sizes, has been 311
threshold retinal sensitivity directly on to the retina with auto- used in select studies in the last decade to assess central func-
mated quantitative threshold perimetry with a high degree of tional vision in a variety of retinal diseases. A compact rarebit

Chapter 12
accuracy is important in the evaluation of retinal disease.11 This screening test has been developed and tested in patients with
ability has further been refined by techniques such as micrope- age-related macular degeneration (AMD).16 Preferential hyper-
rimetry. However, as with any repeated test used to follow acuity perimetry (PHP) is another screening device for detecting
changes over time, consistency in choice of testing method is early visual field changes in retinal disease. PHP uses the phe-
important in accurately characterizing and following the pro- nomenon of hyperacuity (Vernier acuity), the ability to discern
gression of retinal diseases. differences in the spatial location of two or more stimuli, to test
one’s ability to identify local distortion of a series of dotted

Visual Fields in Retinal Disease


vertical or horizontal signals. The responses are correlated to a
Other methodologies of visual field testing normative database and scored to generate a probability of
in retinal disease deviation from normal. PHP has been found to be more sensi-
Macular diseases may be degenerative, hereditary, traumatic, tive than Amsler grid testing for the early detection of neo­
toxic, or inflammatory. All are characterized by central visual vascular AMD in patients with intermediate AMD. Home
field defects or distortion, which may be small enough to be monitoring devices using PHP, which allow for convenient and
undetected without detailed perimetry or Amsler grid testing. timely early detection of progression of AMD, have been devel-
Ancillary psychophysical tests of visual acuity, contrast sensitiv- oped and are currently being evaluated for more widespread
ity, and color vision may be helpful in determining visual clinical use.17
function.
Microperimetry is a relatively new methodology for assessing Reliability and reproducibility of visual
visual fields that is especially helpful for evaluating and follow- field tests
ing macular disease. It is used to correlate anatomic pathology Anatomic, physiologic, and psychological factors unrelated to
with function of the visual system by integrating fundus imaging the pathology in question can significantly affect measurement
and computerized threshold perimetry at specific locations in of the visual field. The nose, brow, and lid may constrict the
the fundus. One technique of microperimetry uses a scanning nasal and superior fields artifactually. Numerous variables per-
laser ophthalmoscope (SLO) (e.g., Rodenstock, Ottobrunn, taining to both the patient and the environment must be con-
Germany) to plot field defects within geographically defined trolled during testing if meaningful results are to be obtained.
regions of the retina. A modulated helium-neon laser beam of Even so, considerable short-term variability must be taken into
variable intensity (0–21 dB) at 633 nm projects stimuli on to the account. Box 12.1 outlines a list of factors that must be controlled
retina during ophthalmoscopy performed with an infrared for optimal determination of the visual field. Perimetric testing
diode laser at 780 nm.7,12 Another type of microperimetry is has a degree of subjectivity and therefore ultimate reliability and
Micro Perimeter 1 (MP 1, Nidek Instruments, Padova, Italy) repeatability of the testing rely on the patient and the test giver.18
which uses an infrared fundus camera that provides a 45° view Patients with retinal disease may be more prone to variability in
and performs perimetry using a liquid crystal display with a testing over time, which can influence the validity of test results.
special software. The MP-1 allows for eye tracking and real-color Seiple and colleagues19 demonstrated that for patients with reti-
fundus image acquisition. Images from other tests, such as fluo- nitis pigmentosa (RP) the results of repeated HVF testing per-
rescein angiogram, can be overlaid on to the microperimetry, formed at different time points were two times more variable
which is not possible in SLO microperimetry. Moreover, as SLO when compared to similar fields performed in normal controls
microperimetry is also restricted to red laser light, comparison despite controlling for disease progression. Microperimetry has
with standard perimetry or MP-1 is difficult.12 the added advantage of real-time retinal surface monitoring with
Both the SLO and MP-1 microperimetry devices allow for
kinetic and static perimetric testing of the macula and permit
simultaneous observation of the retina during perimetric testing.
Similar to conventional perimetry, stimulus sizes range from
Box 12.1 Variables affecting measurement of the visual field
Goldmann size I to V and the central 15–20° of visual field can
be tested in both devices, although the MP-1 allows for a slightly • Environmental
larger field for testing. Rohrschneider and colleagues deter- • Illumination
• Equipment
mined that for patients with retinal disease both the SLO and
• Examiner
MP-1 microperimetry devices provided comparable results, • Technique
with the SLO devices providing better-resolution fundus images • Ocular
and the MP-1 providing better fixation analysis with more accu- • Retinal adaptation
• Refractive state
rate real-time image alignment.13 As efforts to correlate better • Media
anatomic and functional changes in the retina have advanced, • Pupil size
more refined devices are being developed that combine tech- • Global
niques of visualizing the ultrastructure of the retina, such as • Age
• Fixation
three-dimensional spectral optical coherence tomography (OCT) • Reaction time
and adaptive optics, with corresponding perimetric functional • Fatigue
assessment.14,15
eye tracking to correct for eye movements during testing. Wein- Retinal dystrophies
gessel and colleagues have demonstrated good interexaminer
Visual fields have traditionally been used to characterize and
312 and intraexaminer reliability of microperimetry using the MP-1
monitor the progression of visual field loss in RP. The earliest
device in eyes with and without retinal disease.20
field defect in RP is reported to be a group of isolated scotomata
PERIMETRY IN SPECIFIC 20–25° from fixation.3 Eventually these isolated defects coalesce
Section 2

into a “ring scotoma” affecting the midperiphery of the visual


RETINAL DISEASES field. Usually the peripheral field loss progresses and leaves a
The visual fields tests described are used in patients with retinal small, central island of vision. Eventually, complete visual loss
disease to assess and catalog progression of functional visual may occur (Fig. 12.9).
field loss, to determine the outcomes of treatment on visual Characterization and comparison of specific patterns of visual
function, and to correlate functional and structural changes in field loss in genotypically and phenotypically different forms of
Retinal Diagnostics
Retinal Imaging and Diagnostics

the retina. RP such as X-linked, dominant, pericentral, and Usher syndrome

Fig. 12.9 Classic development of a ring


scotoma with focal sparing of a peripheral
A 135
120 105 90 75 60 120 105 90 75 60
45 temporal island seen in retinitis pigmentosa
60 60
50 50
and cone–rod dystrophies. (A) Progressive
150 30 depression of peripheral visual field in a
40 40
patient with cone–rod degeneration at
30 30
presentation. (B) Same patient after 1 year.
165 20 20 15
(C) Same patient during fourth year of
10 10 follow-up. (Reproduced with permission from
180 80 70 60 50 40 30 20 20 30 40 50 6060 50 40 30 20 20 30 40 50 60 70 80 0 Krauss HR, Heckenlively JR. Visual field
13 changes in cone–rod degenerations. Arch
13
10 14 IV4 10 Ophthalmol 1982;100:1784–90.)
195 20 IV4 20 345
14
30 30
40 40
210 330
50 50

225 60 60 315
70 70
240 255 270 285 300 240 255 270 285 300
Left Right
B 135
120 105 90 75 60 120 105 90 75 60
45
60 60
50 50
150 30
40 40
30 30
165 20 20 15
10 10

180 80 70 60 50 40 30 20 20 30 40 50 6060 50 40 30 20 20 30 40 50 60 70 80 0
13 14
10 10
IV4 13
195 20 20 345
14
30 IV4 30
40 40
210 330
50 50

225 60 60 315
70 70
240 255 270 285 300 240 255 270 285 300
Left Right
C 135
120 105 90 75 60 120 105 90 75 60
45
60 60
50 50
150 30
40 40
30 30
165 20 20 15
10 10

180 80 70 60 50 40 30 20 20 30 40 50 6060 50 40 30 20 20 30 40 50 60 70 80 0

IV4 10 10
20 20 IV4
195 345
30 30
40 40
210 330
50 50

225 60 60 315
70 70
240 255 270 285 300 240 255 270 285 300
Left Right
using GVF perimetry have been attempted to differentiate better crystalline dystrophy.37 Correlations between HVF sensitivity
subgroups of RP based on visual prognosis and genotype.21–23 and multifocal electroretinogram (mERG) have been docu-
Association of degree of visual field loss with different geno- mented for eyes with central areolar choroidal dystrophy and 313
types in syndromic associations of RP, including Bardet–Biedl North Carolina macular dystrophy.38,39 Functional changes as
syndrome 1, has also been studied.24 In addition, HVF measure- evidenced by perimetry and mERG have also been correlated

Chapter 12
ments have been correlated with contrast sensitivity in RP and with morphological changes documented by fundus autofluo-
were found to be a sensitive predictor for central visual function rescence in adult vitelliform macular dystrophy.40
in advanced RP.25,26 Visual field perimetry has been used to test
outcomes of treatments for varieties of RP, including fundus Diabetic retinopathy
albipunctatus.27 The diffuse retinal ischemia associated with diabetic retinopathy
Visual fields have also been used in correlating photoreceptor would seem to make visual field assessment an ideal method for
anatomy and visual function in RP. Decreased retinal sensitivity follow-up of the disease. However, visual fields are not routinely

Visual Fields in Retinal Disease


on electroretinogram has been linked to visual field loss demon- used to evaluate this retinal disease, even though field defects
strated by perimetry in RP.28 Recently, changes in GVF, HVF, may exist in the absence of observable retinopathy. Roth41 found
and microperimetry have been correlated with anatomical central field defects in about 40% of eyes without visible reti-
changes at the cellular level using OCT and autofluorescence to nopathy and in all diabetic patients with retinopathy. SWAP
identify abnormal photoreceptor morphology and damage to studies on patients with early diabetic maculopathy demon-
retinal pigment epithelial (RPE) cells.29–35 strated a correlation between the decrease in mean thresholds
Perimetric changes occur in a variety of retinal dystrophies. and the increase in size of the foveal avascular zone and the
Cone dystrophy produces progressive symmetric to slightly perifoveal intercapillary area. These changes were not observed
asymmetric central visual loss. This is associated with a macular with standard white-on-white perimetry.42
pigmentary disturbance. The visual fields demonstrate central In patients with clinically significant diabetic macular edema,
scotomata with relative sparing of the fovea (Fig. 12.10). A Hudson and colleagues43 found all patients had abnormal SWAP
central or paracentral scotoma can also be seen in Stargardt 10–2 fields, but only one-third had abnormal standard perimetric
disease. However, microperimetry identifies two types of fields. Further, the area of abnormal sensitivities was greater
scotoma in patients with Stargardt disease. In one type there than that expected by clinical assessment. Using microperimetry,
is a dense ring scotoma associated with stable fixation. In the macular scotomata were also found in 74% of 19 patients with
second type there is a dense central scotoma associated with clinically significant macular edema.44 Furthermore, SWAP sen-
fixation shift. The second type is also correlated with poorer sitivity in the central 10° of visual field of diabetic patients
acuity.36 In contrast, there is constriction of the visual field without macular edema was significantly reduced compared to
corresponding to the progressive peripheral retinal degenera- standard white-on-white perimetry.45 A study of visual field
tion with scalloped margins in gyrate atrophy of the retina defects in diabetic children without retinopathy by Mastro-
and choroid, a rare tapetoretinal degeneration caused by an pasqua and colleagues46 suggested that light sensitivity was
inborn error of ornithine aminotransferase activity (Figs 12.11 impaired in the midperiphery of the visual field proportional to
and 12.12). Constricted GVF tests are also seen in Bietti the degree of microalbuminuria.

90/30

180/30 0/30

A B 270/30

Fig. 12.10 (A) Fluorescein angiogram of the right eye of a 54-year-old man with a 5-year history of slowly progressive loss of central vision in
each eye shows marked pigmentary disturbance of the macula. The left fundus showed similar changes. Electrophysiologic testing was
diagnostic of cone dystrophy. (B) Octopus perimetry (program 31) demonstrates a dense central scotoma corresponding exactly to the region of
macular pigmentary change. Note the relative sparing of foveal sensitivity.
314
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

Fig. 12.11 Central (A) and peripheral (B) fundus appearance of a patient with gyrate atrophy of the retina. The visual field of this patient is
shown in Fig. 12.12.

Fig. 12.12 Thirty-degree Octopus visual


field (program 31) demonstrates marked
peripheral constriction, with relative sparing
in the centrocecal region. In addition, there
are a few more peripheral islands of reduced
sensitivity. (Reproduced with permission from
Feldon SE. Computerized perimetry in
selected disorders of the retina. In: Whalen
WR, Spaeth GL, editors. Computerized visual
fields: what they are and how to use them.
Thorofare, NJ: Slack; 1985.)
Fig. 12.13 Fluorescein angiogram of a
patient with preproliferative diabetic
retinopathy. Static sensitivities from Octopus
perimetry are superimposed. Areas of good 315
perfusion (G) have normal visual function.
Areas of intermediate perfusion (I) have a
moderate decrease of visual function, and

Chapter 12
nonperfused areas (P) have complete loss of
visual function. (Reproduced with permission
from Bell JA, Feldon SE. Retinal
microangiopathy: correlation of Octopus
perimetry with fluorescein angiography. Arch
Ophthalmol 1984;102:1294–8.)

Visual Fields in Retinal Disease


Once diabetic retinopathy is present, visual field loss is readily
documented. Gandolfo and colleagues47 studied 85 eyes with 30 Diabetic retinopathy
preproliferative diabetic retinopathy using the Goldmann perim- Branch retinal vein occlusion
eter. They were able to identify retinal hemorrhages and exu- 25
dates of at least 3–4° in diameter as localized depressions in the
Average sensitivity (dB)

visual field. Macular exudation and edema caused an irregular 20


depression and flattening of static profiles of the central field.
Wisznia and colleagues48 tried to correlate the degree of reti- 15
nopathy with the amount of visual field loss. They hypothesized
that a correlation might exist between retinal capillary perfusion
10
and field loss. Bell and Feldon11 used Octopus static perimetry
to show that visual sensitivity is quantitatively correlated with
retinal perfusion in preproliferative diabetic retinopathy (Figs 5
12.13 and 12.14). Utilizing standard perimetry, Federman and
Lloyd49 found the degree of perfusion to be more important in 0
predicting field loss than the amount of proliferative retinopa- Good Intermediate Poor
perfusion
thy. This relationship between nonperfusion and regional field
loss has been confirmed in other studies as well.47,50
Lutze and Bresnick51 demonstrated a correlation between the Fig. 12.14 Average sensitivity of the retina decreases with decreasing
perfusion, both for diabetic retinopathy (dots) and for branch retinal
degree of retinopathy in type I diabetic patients and visual field vein occlusion (diamonds). (Reproduced with permission from Bell JA,
loss using SWAP. These findings were consistent with those of Feldon SE. Retinal microangiopathy: correlation of Octopus perimetry
Zwas and coworkers and have been confirmed in more recent with fluorescein angiography. Arch Ophthalmol 1984;102:1294–8.)
studies.9,52 In addition, automated perimetry was found to cor-
relate better with severity of diabetic retinopathy than visual In instances of vitreous hemorrhage and tractional retinal
acuity.53A study by Agardh and colleagues demonstrated that detachment due to high-risk proliferative diabetic retinopathy,
SWAP sensitivity of visual field loss was correlated to ischemic visual acuity is often dramatically improved by vitrectomy;
changes in areas of macular edema, rather than to the severity however, severely impaired visual fields due to extensive retinal
of macular edema.54 SWAP analysis also demonstrated a greater ischemia may still preclude driving.57 Moreover, the treatment
decrease in mean sensitivity in menstruating diabetic women of diabetic retinopathy with either panretinal or focal photoco-
who were in the luteal phase, which was not seen in menstru- agulation may produce visual field defects, a fact that should be
ating control patients.55 In a study by Stavrou and Wood,56 considered in overall patient management.58,59 In a study by Zin-
flicker perimetry appeared to be more sensitive than static girian and colleauges,60 isolated photocoagulation of diabetic
perimetry in documenting early visual field changes in diabetic retinopathy results in small scotomata that are difficult to isolate
retinopathy, especially in a region of clinically significant by kinetic perimetry. Confluent lesions measuring one to two
diabetic macular edema. disc diameters cause correspondingly sized scotomata with
sloping margins. Panretinal photocoagulation produces a defects corresponded to retinal vascular occlusions in sickle-cell
marked concentric contraction of the visual field. Yoon et al.61 disease.71 GVF perimetry showed a slight constriction of periph-
316 demonstrate preservation of retinal sensitivity in central visual eral visual fields, thought to be visually insignificant, a decade
field after panretinal photocoagulation in diabetic patients. At 1 after diode laser retinal ablative therapy for retinopathy of pre-
week after treatment, there is significant depression, but recov- maturity. This field construction was similar to peripheral field
ery of up to 95% occurred within the ensuing 3 months. They changes observed years after cryotherapy for retinopathy of pre-
Section 2

attribute these encouraging findings to the use of burn sizes of maturity.72 Mean deviation improvement recorded with auto-
200 µm or less, as recommended by Hulbert and Vernon.62 mated static Octopus 500 perimetry was noted following carotid
Using automated perimetry, an initial loss of sensitivity seen endarterectomy for clinically significant carotid stenosis.73
after grid laser for diabetic macular edema was seen followed by Microperimetry has been used to document functional
improvement.63 Hudson and colleagues64 followed 24 diabetic improvement with resolution of absolute scotomata and
patients with macular edema before grid laser treatment and up improvement in vision to baseline in the setting of Purtscher’s
Retinal Diagnostics
Retinal Imaging and Diagnostics

to 12 weeks following treatment with microperimetry. They retinopathy after treatment with oral steroids.74 Recovery of
found correlation between the edema index and visual function visual field loss was also seen using HVF and microperimetry
in some, but not all, patients. In another study of 30 patients, 8 after central and branch artery occlusions.75,76 In addition, greater
eyes remained stable, 15 improved mean deviation after treat- decrease in scotopic macular sensitivity was shown with micro-
ment, and laser scars corresponded to marked loss of function.65 perimetry fine matrix mapping of the macula in eyes with
Thus, retinal sensitivity tested by microperimetry appears to type 2 idiopathic macular telangiectasia. In similar eyes, Wong
increase after micropulse diode laser, but to decrease after modi- and colleagues77 demonstrated correlations between micro­
fied Early Treatment Diabetic Retinopathy Study focal laser in perimetry sensitivities, OCT retinal morphology, and fundus
eyes with clinically significant diabetic macular edema. These autofluorescence.
perimetric changes are observed even though there is no differ- After injection of intravitreal triamcinolone for treatment of
ence in visual acuity or retinal thickness after either treatment.66 branch retinal vein occlusion, improvement in macular sensitiv-
Recent studies highlight associations between morphological ity by microperimetry correlates with improvement in macular
and functional alterations in diabetic macular edema using edema.78 According to the Branch Vein Occlusion Study guide-
microperimetry. Microperimetry sensitivities are reduced in lines, microperimetry is useful in assessing the benefit of laser
eyes with diabetic macular edema, and direct correlations have treatment. Regression of the scotoma from the foveal avascular
been made between decreased microperimetry sensitivity and zone is observed in one-third of patients, but in one-half of
increased cystoid edema, as evidenced by OCT and increased treated patients an increase in total scotoma size occurs.79 In
fundus autofluorescence.67–70 branch vein occlusions, Bell and Feldon11 show good correlation
between residual capillary perfusion and threshold retinal
Other vascular diseases and nondiabetic sensitivity (Figs 12.14 and 12.15).
macular edema Visual fields may effectively document the effects of treat-
Many other vascular abnormalities of the choroid and retina ments for CRVO.80 For instance, microperimetry improves
have been evaluated with perimetry. For example, visual field central fixation and retinal sensitivity following resolution of

Fig. 12.15 Fluorescein angiogram from a


patient with a superior branch retinal vein
occlusion. Retinal sensitivities from Octopus
perimetry are superimposed to show
depressed function in the area of
intermediate retinal perfusion (I). G, Area of
good perfusion. (Reproduced with permission
from Bell JA, Feldon SE. Retinal
microangiopathy: correlation of Octopus
perimetry with fluorescein angiography. Arch
Ophthalmol 1984;102:1294–8.)
macular edema due to CRVO after treatment with intravitreal is used to assess retinal sensitivities for a variety of other dis-
triamcinolone acetonide. Though microperimetry shows a eases affecting retinal function in the macula, including X-linked
benefit in macular function and field after radial optic neurot- retinoschisis, S-cone syndrome, retinopathy of membranoprolif- 317
omy for CRVO, according to Tsujikawa and colleages,81 persis- erative glomerulonephritis type II, and atrophic maculopathy
tent peripheral field defects are documented by full-field associated with spinocerebellar ataxia type 7.94–97

Chapter 12
perimetry corresponding to the incision site on the optic nerve AMD of the pigmentary type usually causes irregularly shaped
head in similar eyes with CRVO that underwent the same central scotomata with sloping margins and variable density.
treatment.82 Field defects may be bilateral but often are asymmetric. Micro-
Metamorphopsia on Amsler grid testing is characteristic of perimetry retinal sensitivity correlates with alterations in fundus
central serous retinopathy, but there is an accompanying mild autofluorescence even in early stages of AMD.98 Perimetry is
central depression which varies in size from 2 to 5° (Fig. 12.16). also used to evaluate macular retinal sensitivity after novel
The scotoma is usually substantially larger using SWAP relative treatments for nonexudative AMD.99–101

Visual Fields in Retinal Disease


to that detected with white-on-white perimetry.83 HVF and Disciform subfoveal scarring from subretinal neovasculariza-
microperimetry central retinal sensitivity is reduced as subreti- tion, hemorrhage, and gliosis due to exudative AMD causes
nal fluid in central serous retinopathy increases, documented by a dense central scotoma,3 as shown in Figure 12.18. Detailed
OCT.84,85 Even after resolution of edema, the majority of patients studies of subfoveal choroidal neovascularization in exudative
have residual Amsler and perimetric defects86–88 (Fig. 12.17). AMD have been performed by microperimetry (Fig. 12.19). Of
Similar changes may result from other causes of fluid accumula- 179 eyes evaluated by Fujii and colleagues,102 135 (75%) had
tion in the macula, such as diabetic retinopathy, Irvine–Gass central fixation, 42% had stable fixation, and in 28%, there was
syndrome, trauma (Berlin’s edema), and retinal vasculitis.3,89 a dense central scotoma. The authors found that both central and
stable fixation deteriorated over time. These fixational patterns
Age-related macular degeneration and were felt to be important in the selection of patients for macular
other maculopathies translocation surgery.103 When microperimetry was utilized to
Macular drusen are not usually associated with any reduction
of retinal sensitivity using standard techniques. However, in a
prospective study using SWAP, the mean sensitivity of patients
with soft drusen and early AMD is significantly lower compared
to patients without drusen.90,91 In this study the presence or
absence of focal hyperpigmentation does not affect mean sensi-
tivity. Microperimetry of macular drusen shows decreased over-
lying sensitivity in some, but not all, studies.92,93 Microperimetry

Fig. 12.16 An Amsler grid from a patient with long-standing Fig. 12.17 A set of Tübinger static perimetric profiles showing pattern
metamorphopsia caused by central serous retinopathy. (Reproduced of recovery over 7 months in a patient with central serous retinopathy.
with permission from Natsikos VE, Hart JCD. Static perimetric and (Reproduced with permission from Natsikos VE, Hart JCD. Static
Amsler chart changes in patients with idiopathic central serous perimetric and Amsler chart changes in patients with idiopathic central
retinopathy. Acta Ophthalmol 1980;58:908–17.) serous retinopathy. Acta Ophthalmol 1980;58:908–17.)
318
Section 2

30°
Retinal Diagnostics
Retinal Imaging and Diagnostics

A B

Fig. 12.18 (A) Fluorescein angiogram of a 60-year-old patient referred for evaluation of transient right homonymous hemianopia documents
disciform macular degeneration of the left eye. (B) Dense central scotoma corresponding to the fundus lesion is shown by gray-scale printout
from Humphrey perimeter (program 30–2).

evaluate the anatomic abnormalities associated with an absolute beginning of this chapter, the psychophysical property of hyper-
scotoma in subfoveal choroidal neovascularization, Tezel and acuity has been used to develop a device which detects progres-
associates found that the relative risk (RR) was highest in areas sive maculopathy and the early onset of exudative disease in
of chorioretinal scar (RR = 107.61) compared to areas of RPE AMD.114,115 The device, PreView PHP (Carl Zeiss Meditec,
atrophy (RR = 9.97), subretinal hemorrhage (RR = 2.88), and Dublin, CA), was evaluated as a home-based device in a multi-
neovascular membrane (RR = 1.86).104 The majority of patients center trial and found to have a sensitivity and specificity of 85%
with stable fixation preferred an area of RPE hyperplasia. Using to detect alterations in hyperacuity corresponding to exudative
the HVF macular threshold protocol, improvement in macular and intermediate nonexudative AMD.17 Other home-based peri-
visual field sensitivity occurred after treatment with intravitreal metric devices for patients to monitor their vision routinely are
bevacizumab for exudative AMD, even in cases where visual expected in the near future. With the development of better
acuity has not improved.105 Microperimetry has shown similar algorithms to detect early disease progression, these home
improvement in central retinal sensitivity after intravitreal devices will hopefully ensure timely sight-saving treatment.
ranibizumab treatment for exudative AMD.106,107 HVF 10–2 and
microperimetry have also demonstrated improvements in Macular holes and epiretinal membrane
macular visual fields after photodynamic therapy for exudative Cysts may develop in the macula without producing appreciable
AMD and subfoveal polypoidal choroidal vasculopathy.108–110 scotomata. Macular holes, however, result in dense scotomata
Microperimetry has also been used to assess retinal sensitivity with steep margins (Figs 12.20 and 12.21). Microperimetry may
after autologous RPE choroid graft for exudative AMD.111 be helpful in predicting the outcome of macular hole surgery. In
Patients with AMD or other macular pathology are routinely a study by Amari and associates, visual outcome correlated with
instructed to monitor their visual field in each eye with an the maximum sensitivity adjacent to the hole.116 In another study,
Amsler grid regularly. Comparison of the original white lines on absolute scotomata disappeared completely in 18 of 28 eyes that
a black background Amsler grid proved to be superior at detect- achieved complete closure, became relative in five of six eyes
ing metamorphopsia and central vision changes than the modi- with partial closure, and remained absolute in four eyes with
fied grid, which displays black lines on a white background.112 atrophic closure.117 Ozedemir and colleagues118 suggested that
This is likely because of the increased contrast provided by the MP-1 microperimetry may be more sensitive than visual acuity
white lines on black background. However, due to ease in pho- in measuring retinal function following closure of a macular hole
tocopying, the grid with black lines on a white background is with pars plana vitrectomy and internal limiting membrane
most commonly used in the office setting and provided to (ILM) peeling. Increases in retinal sensitivity by microperimetry
patients for home use. have also been correlated with the degree of fundus autofluores-
With the increasing prevalence of AMD and the ability to treat cence after macular hole closure.119 Perimetry has also been used
early neovascular AMD effectively, new perimetric tests that to evaluate visual fields in eyes with epiretinal membrane (ERM).
allow for patient self-monitoring of visual fields are being Binocular correspondence perimetry, a method akin to PHP, but
devised to detect early deficits. Nazemi and colleagues113 devel- which uses the principle of retinal correspondence and requires
oped a three-dimensional automated computer-based threshold binocular testing, was used to quantify metamorphopsia in eyes
Amsler grid test that maps the visual field and records steep with ERM.120 This study demonstrated focal areas of abnormal
slopes in areas of nonexudative AMD and shallow slopes in retinal correspondence in eyes with ERM compared to the
regions corresponding to exudative AMD. As discussed at the normal fellow eye. Increased microperimetry retinal sensitivity
A B
319

Chapter 12
Visual Fields in Retinal Disease
C D

E F

Fig. 12.19 A sequence of scanning laser ophthalmoscope (SLO) microperimetry shows the progressive functional deterioration in one eye with
subfoveal choroidal neovascularization (CNV) secondary to age-related macular degeneration (AMD). The SLO testing demonstrated that eyes
with subfoveal CNV secondary to AMD experienced a predictable and progressive loss of fixation stability, decreased central retinal sensitivity,
and loss of central fixation location. A 65-year-old man presented with 20/150 vision and a 1-month history of decreased vision due to a
predominantly classic subfoveal CNV secondary to AMD. (A) The SLO testing performed at presentation disclosed a pattern of predominantly
central and stable fixation. (B) The balls indicate the areas where the patient could perceive the stimulus; the triangles indicate the areas where
the patient could not perceive the stimulus. Each ball and triangle is color-coded to indicate the intensity of the stimulus. The SLO microperimetry
also showed a mild decrease in central retinal sensitivity. The patient elected not to receive any treatment and had a follow-up visit 4 months
after initial visual symptoms. (C) An SLO test was performed and demonstrated that the fixation pattern became poor central and relatively
unstable. (D) The microperimetry also showed that retinal sensitivity was markedly affected with some central areas of dense scotoma. Best-
corrected visual acuity at this visit was 20/200. Twelve months after onset of initial visual symptoms and no treatment, SLO microperimetry was
performed and disclosed further functional deterioration. (E) The fixation became predominantly eccentric and unstable. (F) Retinal sensitivity
testing demonstrated a large central area of dense central scotoma. (Reproduced with permission from Wong WT, Kam W, Cunnigham D, et al.
Treatment of geographic atrophy by the topical administration of OT-551: results of a phase II clinical trial. Invest Ophthalmol Vis Sci
2010;51:6131–9.)
human retinal cell lines.130 The postsurgical visual defects are
probably due to: (1) toxic effects of ICG; (2) alterations in the
320 retina, such as damage to the peripapillary nerve fibers, during
the pars plana vitrectomy; or (3) mechanical damage incurred
with peeling of the ILM. Damage to the nerve fiber layer from
intraocular gas tamponade in cases of macular hole repair must
Section 2

also be considered.121 Further studies that systematically and


more accurately compare pre- and postoperative visual fields in
eyes undergoing retinal surgery are needed to understand better
the effects of the procedure and the adjuvant dyes or agents used
to assist in the procedure on visual function.
Retinal Diagnostics
Retinal Imaging and Diagnostics

Toxic retinopathies
Toxic maculopathy is epitomized by chloroquine and hydroxy-
chloroquine retinopathy. The most characteristic field defect
caused by macular involvement is a ring-like central scotoma
with a small island of slightly less visual loss in its center, com-
monly referred to as a “bull’s eye” (Fig. 12.23).131 GVF and HVF
perimetry document visual field changes that can persist and
worsen for decades even after stopping the medicines.132–134
Threshold Amsler grid testing, which uses variable light trans-
mission through two cross-polarizing filters, and PHP hyperacu-
ity may be useful in screening for early functional changes due
Fig. 12.20 Fundus photograph of an eye with a full-thickness macular to chloroquine and hydroxychloroquine.135,136
hole. The visual field defect is shown in Figure 12.21. To screen for chloroquine and hydroxychloroquine toxicity
HVF, 10–2 white-on-white protocol is recommended. mERG is
a helpful adjunct when early perimetric changes are seen, but
A B may not be readily available in all retina practices. Recent screen-
Z
ing guidelines advocate the addition of high-resolution imaging,
32.00 such as spectral domain OCT and fundus autofluorescence, to
correlate functional changes with structural alterations in the
photoreceptor and RPE layers.137 In concert with other testing
20.48 modalities, early perimetric changes that may be nonspecific for
macular toxicity can be validated to allow for timely changes in
Y medication with ultimate preservation of vision.
8.88 4.00 A number of medications are associated with retinal toxicity
1.33 resulting in visual field loss. Thioridazine, a phenothiazine
-1.33 derivative, is a cause of pigmented maculopathy with associated
-2.52 -4.00 central scotoma. Even in the absence of clinical tamoxifen reti-
14.1 7.4 0.7 -8.0
nopathy, SWAP fields show depressed mean deviations which
correlated with duration of therapy.138,139 Peripheral visual field
Fig. 12.21 Three-dimensional reconstruction of high-density macular loss is detected in about 30% of patients using the antiepileptic
grid (1° spacing) documenting steep absolute central scotoma of the
left eye (A) and unaffected right eye (B). Retinal sensitivity is noted on drug vigabatrin.140–143 Sildenafil (Viagra) has been associated
the vertical axis and position is noted on the horizontal axis. (Copyright with nonarteritic ischemic optic neuropathy in individuals with
1983, Wesley K. Herman, MD and Joseph M. DeFaller, Alcon pre-existing cardiovascular disease.144 Although visual field
Laboratories, Inc. Courtesy of Dr Herman.)
defects can occur in these cases, sildenafil has not been proven
to cause retinal toxicity.145
without change in metamorphopsia as quantified by PHP fol-
lowing vitreoretinal surgery for ERM and macular hole was Infectious and inflammatory retinopathies
reported by Richter-Mueksch and colleagues.121 Infectious and inflammatory retinopathies exhibit visual field
After uncomplicated repair of macular holes and ERM and defects corresponding to the area of pathology. Toxoplasmosis
using pars plana vitrectomy and ILM peeling, several investiga- commonly produces focal chorioretinal destruction that causes
tors reported a high incidence of peripheral field defects infring- corresponding dense, irregular, steep-margined, isolated scoto-
ing on the central visual field122–128 (Fig. 12.22). A majority of mata.146 More disseminated types of inflammation, such as syph-
studies documented postoperative field defects only after ilitic choroiditis, produce a more diffuse depression of visual
patients complained of perceived field loss. Tsuiki and col- field function. In a patient with idiopathic retinal vasculitis, the
leagues129 specifically compared pre- and postoperative GVF inferior arcuate field defect corresponds to leakage from the
tests and found new peripheral field defects in 17 of 140 eyes superior temporal branch vein (Fig. 12.24). One unusual case
postoperatively. In the majority of eyes, indocyanine green (ICG) report describes the use of microperimetry to determine the
was used to enhance visualization of the ILM during peeling. In focal visual loss associated with nematode-induced unilateral
vitro studies demonstrated the toxicity of ICG exposure to subacute neuroretinitis.147
120 105 90
70
75 60
321
135 60 45

50
150 30

Chapter 12
40

30

165 20 15

10

90 80 70 60 50 40 30 20 10 10 20 30 40 50 60 70 80 90
180 0 30

1-3 10

195 20 345

Visual Fields in Retinal Disease


1-4 30

40
210 330
V-4 50

60
225 315

70
240 255 270 285 300

120 105 90 75 60
70
135 60 45

50
150 30
40

30

165 20 15

10

90 80 70 60 50 40 30 20 10 10 20 30 40 50 60 70 80 90
180 0 30

10

195 20 345

30 1-3
40
210 1-4 330
50

225
60
V-4 315

70
240 255 270 285 300

120 105 90 75 60 120 105 90 75 60


70 70
135 60 45 135 60 45

50 50
150 30 150 30
40 40
V-4
30 30

165 15 165
1-4 15
20 20

10 10 1-3
90 80 70 60 50 40 30 20 10 10 20 30 40 50 60 70 80 90 90 80 70 60 50 40 30 20 10 10 20 30 40 50 60 70 80 90
180 0 180 0

10 10

20 20
195
1-3 345 195 345

30 30

1-4 40
330
40
330
210 210

V-4 50 50

60 60
225 315 225 315

70 70
240 255 270 285 300 240 255 270 285 300

Fig. 12.22 Goldmann and Humphrey 30–2 visual field perimetry depicting peripheral wedge-like visual field loss encroaching on the central visual
field in the eyes of 3 patients after pars plana vitrectomy, internal limiting membrane (ILM) peeling assisted by indocyanine green staining of the
ILM for external limiting membrane. (Reproduced with permission from von Jagow B, Hoing A, Gandorfer A, et al. Functional outcome of
indocyanine green-assisted macular surgery: 7-year follow-up. Retina 2009;29:1249–56.)
0.0 4.3 13.5 11.2 2.5 5.5
322
6.5 3.8 8.5 2.5 5.5 3.0
6.5 11.5 8.5
Section 2

3.5 9.2 20.5 19.5 19.5 16.8


11.5 4.9 17.5 17.5 3.1
12.5 20.5 13.5
Retinal Diagnostics
Retinal Imaging and Diagnostics

16.5 18.8 14.5 17.8 22.5 23.8


19.5 1.7 16.5 1.7 23.5 1.5
19.5 22.5 25.5

Fluctuations (R.M.S.): 3.1 DB Lum interval: 2


A B

Fig. 12.23 (A) Fluorescein angiogram of a patient with perifoveal atrophy of the pigment epithelium corresponding to a bull’s eye maculopathy,
typical of chloroquine retinopathy. (B) Central retinal sensitivities (Octopus program 11) at 3° spacing show depression superiorly and temporally,
corresponding to areas of fluorescein angiogram with more marked depigmentation of the macula. Each cluster of five numbers represents a
single test location. The three numbers at the left of each cluster are independently determined thresholds. The number at the upper right in
each cluster is the mean of the three thresholds. The number at the lower right in each cluster is the standard deviation of the mean threshold
value. (Reproduced with permission from Feldon SE. Computerized perimetry in selected disorders of the retina. In: Whalen WR, Spaeth GL,
editors. Computerized visual fields: what they are and how to use them. Thorofare, NJ: Slack, 1985.)

30

1/ e
4
15

30 40 50 60 70 80 0
1/
2e
1/
4c
1/
1e

345
1/
1/ 4c
3e

1/
4c
330

315
Alt. resp. to 1/
4e
A B

Fig. 12.24 (A) Fluorescein angiogram from a patient with idiopathic retinal vasculitis demonstrates leakage from the superotemporal retinal
branch vein of the right eye. (B) Goldmann perimetry shows an inferior arcuate field defect that corresponds to the affected region of the retina.

Even in patients with no clinically evident infectious retinopa- may also be related to HIV-related brain dysfunction.150 More
thy, human immunodeficiency virus (HIV)-positive patients marked changes in retinal sensitivity have been found in HIV-
demonstrated significant localized as well as mean defects. positive patients with low CD4 counts.150
These defects were more apparent in SWAP than in white-on- In a small case series of patients with multiple evanescent
white automated perimetry.148 Correlation of HVF perimetry white-dot syndrome, areas of decreased microperimetric sensi-
with mERG in similar eyes demonstrated involvement of the tivity were shown to correlate with focal regions of inner and
inner retina and sparing of the outer retina.149 Perimetric changes outer photoreceptor segment disruption documented by spectral
domain OCT. Both the microperimetry and OCT findings high percentage of retinal detachment surgeries, especially if
changed location in keeping with the resolution and presence of retinotomy for drainage of subretinal fluid (12 of 14, 86%) is
new lesions during the disease course, and returned to normal performed. In this study, risk of visual field loss is more fre- 323
by the end of clinical recovery.151,152 Similar correlation between quent if the retinotomy is relatively posterior (less than 5 disc
function and structure was seen in eyes with acute posterior diameters from fixation).159 Although visual field loss may

Chapter 12
multifocal placoid pigment epitheliopathy.153 Microperimetry recover initially after retinal detachment repair, GVF perimetry
retinal sensitivity was also found to correlate with visual acuity shows persistent decreased visual field following scleral buckle
in both serpiginous choroiditis and birdshot chorioretinopathy. repair of retinal detachment despite continued improvement
Gordon and colleagues154 documented and analyzed HVF in in visual acuity.160 In a recent report, microperimetry used in
patients with birdshot chorioretinopathy, in an attempt to deter- conjunction with spectral domain OCT and fundus autofluo-
mine characteristic patterns of field loss. The visual fields of this rescence demonstrates good correlation of visual function with
small series of patients exhibited diffuse loss, central sparing, retinal morphology after rhegmatogenous retinal detachment

Visual Fields in Retinal Disease


and blind-spot enlargement. In addition, assessment of out- repair.161
comes after valacyclovir treatment for acute zonal occult outer
retinopathy using GVF demonstrated improved peripheral Tumors
visual function.155 Retinoblastoma and choroidal melanoma are the most common
malignant intraocular tumors in children and adults respec-
Retinal detachment tively. Long-term visual field defects that correspond to tumor
Typically, rhegmatogenous retinal detachments have sloping size, location, and treatment modality have been documented
isopters on kinetic perimetry. Occasionally, this feature helps to in eyes with retinoblastoma.162 Anterior melanomas produce
differentiate a retinal detachment from a retinoschisis, which is localized constriction of the visual field, whereas posterior
characterized by dense defects with steep margins, usually melanomas produce dense scotomata with steep borders. Only
located supranasally. However, long-standing retinal detach- a subtle field defect, if any, is associated with choroidal nevus.
ments may develop steep isopters. In the case of a shallow Thus fields may be important in distinguishing between these
detachment, assessment of the visual field may be more accurate entities.
than ophthalmoscopy in identifying the border of detached In Fig. 12.25 the Octopus visual field is superimposed on the
retina.3 fundus photograph of a patient with a small melanoma of the
If the detachment is long-standing, recovery of visual field posterior pole. In this instance, loss of sensitivity is found only
sensitivity is incomplete.156 Performing visual fields under con- in the center of the mass. Use of a finer grid pattern would have
ditions of both light and dark adaptation, Alexandridis and facilitated a better correlation between the pathologic process
Janzarik157 report that cone function returns before rod func- and the degree of visual loss.
tion after successful surgical reattachment of the retina. Assess- Like melanomas, choroidal metastases also produce dense
ment of visual fields using SWAP is a sensitive measure of scotomata with steep borders. However, in a study by Rahhal
functional visual improvement in the macula following surgical and colleagues,163 HVF depression does not consistently corre-
repair for macula involving retinal detachment.158 One study spond with tumor size or location. The visual fields shown in
suggests that central visual field defects occur following a Fig. 12.26 are obtained from a patient with breast carcinoma

Fig. 12.25 A fundus photograph of a patient


with a small malignant melanoma of the
posterior pole has been superimposed on the
visual field obtained by Octopus perimetry.
Because of the relatively coarse (6°) grid
pattern of the perimeter, sensitivity is
markedly reduced only at the center of the
lesion. (Reproduced with permission from
Feldon SE. Computerized perimetry in
selected disorders of the retina. In: Whalen
WR, Spaeth GL, editors. Computerized visual
fields: what they are and how to use them.
Thorofare, NJ: Slack, 1985.)
324
Section 2
Retinal Diagnostics
Retinal Imaging and Diagnostics

Left eye Right eye

Fig. 12.26 Octopus perimetry (program 32) on the same patient as in Figure 12.27. An inferonasal field defect in the right eye corresponds to the
choroidal metastasis. There is also an unexpected dense bitemporal hemianopsia. A computed tomographic scan was requested for further
evaluation. (Reproduced with permission from Feldon SE. Computerized perimetry in selected disorders of the retina. In: Whalen WR, Spaeth
GL, editors. Computerized visual fields: what they are and how to use them. Thorofare, NJ: Slack, 1985.)

Fig. 12.27 A 50-year-old patient with known breast carcinoma is referred


for evaluation of a mass in the posterior pole of the right eye. The visual
fields are shown in Figure 12.26. (Reproduced with permission from
Feldon SE. Computerized perimetry in selected disorders of the retina.
In: Whalen WR, Spaeth GL, editors. Computerized visual fields: what
they are and how to use them. Thorofare, NJ: Slack, 1985.)

whose fundus photograph is shown in Figure 12.27. This patient Fig. 12.28 Computed tomographic scan from the patient whose visual
demonstrates not only a dense inferonasal field defect corre- field and fundus photograph are shown in Figures 12.26 and 12.27. A
sponding to the choroidal metastasis, but also a bitemporal suprasellar mass is shown that is consistent with either pituitary tumor
field defect. Subsequent CT scan demonstrated a suprasellar or metastasis. (Reproduced with permission from Feldon SE.
Computerized perimetry in selected disorders of the retina. In: Whalen
mass consistent with either a pituitary adenoma or metastasis WR, Spaeth GL, editors. Computerized visual fields: what they are and
(Fig. 12.28). how to use them. Thorofare, NJ: Slack, 1985.)
FUTURE OF PERIMETRY IN disease, including AMD and cystoid macular edema. Continued
refinement of this functional imaging modality is expected to
RETINAL DISEASE
allow for better detection and monitoring of retinal disease. 325
Layer-by-layer perimetry Adaptive optics imaging of cones and other ultrastructural
Enoch, along with his collaborators Lawrence, Fitzgerald, and elements of the retina combined with ultrafine microperimetry

Chapter 12
Campos,164–166 developed a clinical perimetric technique empha- shows promise in pinpointing finite areas of decreased retinal
sizing the detection of a small luminous spot in a stationary sensitivity. The ability to resolve cones and rods through adap-
(sustained) or flashing (transient) surround, based on analogies tive optics allows for the reduction in the size of retinal image
with the neural processing of the inner versus outer retinal and, hence, the number of photoreceptors stimulated by adap-
layers. Graded or “sustained-like” electrical responses are tive optics microperimetry. Using microflashes in a patient with
obtained from the retinal receptor, bipolar, and horizontal cells. a genetic mutation for a cone photopigment and no clinical
visual deficits, Williams and colleagues at the University of

Visual Fields in Retinal Disease


Transient or “spike” impulses are recorded from amacrine and
ganglion cells. Enoch and colleagues investigated layer-by-layer Rochester have detected retinal microscotomata representing
perimetry in diabetic retinopathy, macular drusen, macular early cone photoreceptor loss through adaptive optics micrope-
degeneration, and angioid streaks. However, layer-by-layer rimetry.169 In addition, Roorda and colleagues15 have demon-
perimetry has not gained clinical acceptance. The relationship strated correlations between healthy and unhealthy RPE and
between retinal function and the testing parameters of this microperimetry retinal sensitivity in patients with maculopathy
modality remains unconfirmed. and normal visual acuity. The precision of testing cellular func-
tion in the retina with this modality highlights its ability to detect
Color perimetry early loss of function of a few photoreceptors. As this focal loss
of retinal function is clinically compensated by the redundancy
Assessment of color vision may be helpful in screening for
in the visual system, it is not noticed by patients or captured
retinal disease and in separating neural from retinal causes of
by standard perimetry and visual acuity tests. Detecting very
visual loss. Acquired diseases of the retina and choroid typically
early functional and structural pathology at the photoreceptor
produce a tritan-like pattern of color confusion, and neural dis-
level before its clinical manifestation would allow for a better
eases typically produce a protan or deutan-like pattern of color
understanding of early pathophysiology and disease course.
confusion on Farnsworth–Munsell 100-hue discrimination
With this knowledge, refined screening algorithms and timely
testing.167 Thus color perimetry might be used to advantage in
interventions for vision-threatening retinal disease might be
the identification and quantitative description of certain retinal
developed.
and choroidal diseases. Although color contrast might be more
sensitive in finding visual field defects, Hart and Burde168 dem- CONCLUSIONS
onstrate that colored test objects used in standard techniques of
perimetry may be of little clinical value over small or low- Achromatic and static SWAP provides a cost-efficient, standard-
contrast white test objects for detecting scotomata in a number ized, and reliable way to evaluate and follow visual fields in
of optic nerve and retinal disorders. Currently, there is no com- patients with retinal disease. Microperimetry seems to be a valu-
mercially available, standardized color perimeter. able clinical research tool that has been used more frequently to
quantify macular function in a variety of retinal diseases. The
High-resolution OCT and adaptive optics clinical value of determining very localized retinal sensitivity
and correlating it with corresponding retinal microstructure
with microperimetry remains uncertain. Nonetheless, a detailed knowledge of how
Although innovative, both layer-by-layer and color perimetry do the underlying retinal pathology and morphology impact on
not appear to add new basic or clinical information relevant to vision is crucial in developing and delivering effective treat-
the etiology, localization, or treatment of retinal diseases beyond ments for retinal disease to preserve and restore eyesight.
that already available from more standard clinical tests, such as
REFERENCES
electroretinography, electro-oculography, contrast sensitivity, 1. Gass JDM. Steroscopic atlas of macular diseases: diagnosis and treatment.
and fluorescein angiography. Anatomical imaging of the retina 4th ed. St Louis: Mosby; 1997.
2. Traquair HM. Introduction to clinical perimetry. London: Kimpton; 1927.
at the ultrastructural level has advanced to a high degree of 3. Harrington DO. The visual fields: a textbook and atlas of clinical perimetry.
sophistication, allowing correlations of structure and function at 5th ed. St Louis: Mosby; 1981.
the microscopic level. Hopefully, these new research strategies 4. Wall M, Sadun AA. Threshold Amsler grid testing: cross-polarizing lenses
enhance yield. Arch Ophthalmol 1986;104:520–3.
will result in clinical applications that will allow earlier detection 5. Keltner JL, Johnson CA, Balestrery FG. Suprathreshold static perimetry: initial
and treatment of potentially debilitating and widely prevalent clinical trials with the Fieldmaster automated perimeter. Arch Ophthalmol
1979;97:260–72.
retinal diseases such as AMD. 6. Wiggins MN, Dersu I. Understanding visual fields, part III: Which field
The combined spectral domain OCT and SLO, introduced in should be performed? J Ophthalm Med Techno [Internet] 2007;3 [cited 2011
Jun 18]. Available from: www.JOMTonline.com.
2006 (Spectral OCT/SLO; OPKO/OTI, Miami, FL, USA) is a 7. Rohrschneider K, Becker M, Schumacher N, et al. Normal values for fundus
significant achievement in advancing retinal imaging. The perimetry with the scanning laser ophthalmoscope. Am J Ophthalmol 1998;
enhancement in accurate and real-time image capture with this 126:52–8.
8. Mojon DS, Zulauf M. Normal values of short-wavelength automated
system allows for integration with microperimetry. Thus, a func- perimetry. Ophthalmologica 2003;217:260–4.
tional spectral OCT/SLO allows assessment of retinal morphol- 9. Zwas F, Weiss H, McKinnon P. Spectral sensitivity measurements in early
diabetic retinopathy. Ophthalm Res 1980;12:87–96.
ogy and function at specific retinal loci. Landa and colleagues14 10. Wild JM, Cubbidge RP, Pacey IE, et al. Statistical aspects of the normal visual
demonstrate the effectiveness of a three-dimensional spectral field in short-wavelength automated perimetry. Invest Ophthalmol Vis Sci
1998;39:54–63.
OCT/SLO topography and microperimetry in assessing func- 11. Bell JA, Feldon SE. Retinal microangiopathy: correlation of Octopus perimetry
tional and structural changes of the macula in a variety of retinal with fluorescein angiography. Arch Ophthalmol 1984;102:1294–8.
12. Rohrschneider K, Bultmann S, Springer C. Use of fundus perimetry (micro- 44. Mori F, Ishiko S, Kitaya N, et al. Use of scanning laser ophthalmoscope micro-
perimetry) to quantify macular sensitivity. Prog Retinal Eye Res 2008;27: perimetry in clinically significant macular edema in type 2 diabetes mellitus.
536–48. Jpn J Ophthalmol 2002;46:650–5.
326 13. Rohrschneider K, Springer C, Bultmann S, et al. Microperimetry – comparison 45. Remky A, Weber A, Hendricks S, et al. Short-wavelength automated perim-
between micro perimeter 1 and scanning laser ophthalmoscope – fundus etry in patients with diabetes mellitus without macular edema. Graefes Arch
perimetry. Am J Ophthalmol 2005;139:125–34. Clin Exp Ophthalmol 2003;241:468–71.
14. Landa G, Rosen RB, Garcia PM, et al. Combined three-dimensional 46. Mastropasqua L, Verrotti A, Lobefalo L, et al. Visual field defects in
spectral OCT/SLO topography and microperimetry: steps toward achieving diabetic children without retinopathy: relation between visual function and
Section 2

functional spectral OCT/SLO. Ophthalm Res 2010;43:92–8. microalbuminuria. Acta Ophthalmol Scand 1995;73:125–8.
15. Roorda A, Zhang Y, Duncan JL. High-resolution in vivo imaging of the RPE 47. Gandolfo E, Zingirian M, Corrallo G, et al. Diabetic retinopathy: perimetric
mosaic in eyes with retinal disease. Invest Ophthalmol Vis Sci 2007;48: findings. In: Greve EL, Heijl A, editors. Fifth International Visual Field
2297–303. Symposium. The Hague: Dr W Junk; 1983.
16. Winther C, Frisen L. A compact rarebit test for macular diseases. Br J 48. Wisznia KI, Lieberman TW, Leopold IH. Visual fields in diabetic retinopathy.
Ophthalmol 2010;94:324–7. Br J Ophthalmol 1971;55:183–8.
17. Lowenstein A, Ferencz JR, Lang Y, et al. Toward earlier detection of choroidal 49. Federman JL, Lloyd J. Automated static perimetry to evaluate diabetic
neovascularization secondary to age-related macular degeneration. Retina retinopathy. Trans Am Ophthalmol Soc 1984;82:358–70.
Retinal Diagnostics

2010;30:1058–64. 50. Pahor D Automated static perimetry as a screening method for evaluation of
Retinal Imaging and Diagnostics

18. Norden LC. Reliability in perimetry. J Am Optom Assoc 1989;60:880–90. retinal perfusion in diabetic retinopathy. Int Ophthalmol 1997–1998;21:
19. Seiple W, Clemens CJ, Greenstein VC, et al. Test–retest reliability of the mul- 305–9.
tifocal electroretinogram and Humphrey visual fields in patients with retinitis 51. Lutze M, Bresnick GH. Lens-corrected visual field sensitivity and diabetes.
pigmentosa. Doc Ophthalmol 2004;109:255–72. Invest Ophthalmol Vis Sci 1994;35:649–55.
20. Weingessel B, Sacu S, Vecsei-Marlovits PV, et al. Interexaminar and intraex- 52. Remky A, Weber A, Hendricks S, et al. Short-wavelength automated perim-
aminer reliability of the microperimeter MP-1. Eye 2009;23:1052–8. etry in patients with diabetes mellitus without macular edema. Graefes Arch
21. Sandberg MA, Gaudio AR, Berson EL. Disease course of patients with Clin Exp Ophthalmol 2003;241:468–71.
pericentral retinitis pigmentosa. Am J Ophthalmol 2005;140:100–6. 53. Bengtsson B, Heijl A, Agardh E. Visual fields correlate better than
22. Sandberg MA, Rosner B, Weigel-DiFranco C, et al. Disease course of patients visual acuity to severity of diabetic retinopathy. Diabetologia 2005;48:
with X-linked retinitis pigmentosa due to RPGR gene mutations. Invest 2494–500.
Ophthalmol Vis Sci 2007;48:1298–304. 54. Agardh E, Stjernquist H, Heijl A, et al. Visual acuity and perimetry as mea-
23. Schwartz SB, Aleman TS, Cideciyan AV, et al. Disease expression in Usher sures of visual function in diabetic macular oedema. Diabetologia 2006;49:
syndrome caused by VLGR1 gene mutation (USH2C) and comparison with 200–6.
USH2A phenotype. Invest Ophthalmol Vis Sci 2005;46:734–43. 55. Apaydin KC, Akar Y, Akar ME, et al. Menstrual cycle dependent changes
24. Azari AA, Aleman TS, Cideciyan AV, et al. Retinal disease expression in in blue-on-yellow visual field analysis of young diabetic women with
Bardet–Biedl syndrome-1 (BBS1) is a spectrum from maculopathy to retina- severe non-proliferative diabetic retinopathy. Clin Exp Ophthalmol 2004;32:
wide degeneration. Invest Ophthalmol Vis Sci 2006;47:5004–10. 265–9.
25. Gerth C, Wright T, Heon E, et al. Assessment of central retinal function in 56. Stavrou EP, Wood JM. Central visual field changes using flicker perimetry in
patients with advanced retinitis pigmentosa. Invest Ophthalmol Vis Sci 2007; type 2 diabetes mellitus. Acta Ophthalmol Scand 2005;83:574–80.
48:1312–8. 57. Barsam A, Laidlaw A. Visual fields in patients who have undergone
26. Lodha N, Westall CA, Brent M, et al. A modified protocol for the assessment vitrectomy for complications of diabetic retinopathy. A prospective study.
of visual function in patients with retinitis pigmentosa. Adv Exp Med Biol BMC Ophthalmol 2006;6:5.
2003;533:49–57. 58. Frank RN. Visual fields and electroretinography following extensive photo-
27. Rotenstreich Y, Harats D, Shaish A, et al. Treatment of a retinal dystrophy, coagulation. Arch Ophthalmol 1975;93:591–8.
fundus albipunctatus, with oral 9-cis-β-carotene. Br J Ophthalmol 2010;94: 59. Wessing A, Meyer-Schwickerath G. Die Behandlung der Retinopathia
616–21. diabetica mit Lichtkoagulation. Diabetologia 1969;5:312–7.
28. Iarossi G, Falsini B, Piccardi M. Regional cone dysfunction in retinitis pigmen- 60. Zingirian M, Pisano E, Gandolfo E. Visual field damage after photocoagu-
tosa evaluated by flicker ERGs: relationship with perimetric sensitivity losses. lative treatment for diabetic retinopathy. In: Greve EL, editor. Second
Invest Ophthalmol Vis Sci 2003;44:866–74. International Visual Field Symposium. The Hague: Dr W Junk, 1977.
29. Fischer MD, Fleischhauer JC, Gillies MC, et al. A new method to monitor 61. Yoon YH, Lee J, Kim YJ. Preservation of retinal sensitivity in central visual
visual field defects caused by photoreceptor degeneration by quantitative field after panretinal photocoagulation in diabetics. Korean J Ophthalmol
optical coherence tomography. Invest Ophthalmol Vis Sci 2008;49:3617–21. 1996;10:48–54.
30. Rangaswamy NV, Patel HM, Locke KG, et al. A comparison of visual 62. Hulbert MFG, Vernon SA. Passing the DVLC field regulations following
field sensitivity to photoreceptor thickness in retinitis pigmentosa. Invest bilateral panretinal photocoagulation in diabetics. Eye 1992;6:456–60.
Ophthalmol Vis Sci 2010;51:4213–9. 63. Hudson C, Flanagan JG, Turner GS, et al. Correlation of a scanning laser
31. Jacobson SG, Roman AJ, Aleman TS, et al. Normal central retinal function and derived oedema index and visual function following grid laser treatment for
structure preserved in retinitis pigmentosa. Invest Ophthalmol Vis Sci 2010; diabetic macular oedema. Br J Ophthalmol 2003;87:455–61.
51:1079–85. 64. Hudson C, Flanagan JG, Turner GS, et al. Correlation of a scanning laser
32. Aizawa S, Mitamura Y, Hagiwara A, et al. Changes of fundus autofluores- derived oedema index and visual function following grid laser treatment for
cence, photoreceptor inner and outer segment junction line, and visual func- diabetic macular oedema. Br J Ophthalmol 2003;87:455–61.
tion in patients with retinitis pigmentosa. Clin Exp Ophthalmol 2010;38: 65. Rohrschneider K, Bultmann S, Gluck R, et al. Scanning laser ophthalmoscope
597–604. fundus perimetry before and after laser photocoagulation for clinically
33. Genead MA, Fishman GA, Stone EM, et al. The natural history of Stargardt significant diabetic macular edema. Am J Ophthalmol 2000;129:27–32.
disease with specific sequence mutation in the ABCA4 gene. Invest 66. Vujosevic S, Bottega E, Casciano M, et al. Microperimetry and fundus auto-
Ophthalmol Vis Sci 2009;50:5867–71. fluorescence in diabetic macular edema: subthreshold micropulse diode laser
34. Wakabayashi T, Sawa M, Gomi F, et al. Correlation of fundus autofluores- versus modified early treatment diabetic retinopathy study laser photocoagu-
cence with photoreceptor morphology and functional changes in eyes with lation. Retina 2010;30:908–16.
retinitis pigmentosa. Acta Ophthalmol 2010;88:e177–83. 67. Deak GG, Bolz M, Ritter M, et al. A systematic correlation between morphol-
35. Popovic P, Jarc-Vidmar M, Hawlina M. Abnormal fundus autofluorescence ogy and functional alterations in diabetic macular edema. Invest Ophthalmol
in relation to retinal function in patients with retinitis pigmentosa. Graefes Vis Sci 2010;51:6710–4.
Arch Clin Exp Ophthalmol 2005;243:1018–27. 68. Okada K, Yamamoto S, Mizunoya S, et al. Correlation of retinal sensitivity
36. Mori F, Ishiko S, Kitaya N, et al. Scotoma and fixation patterns using scanning measured with fundus-related microperimetry to visual acuity and retinal
laser ophthalmoscope microperimetry in patients with macular dystrophy. thickness in eyes with diabetic macular edema. Eye 2006;20:805–9.
Am J Ophthalmol 2001;132:897–902. 69. Vujosevic S, Midena E, Pilotto E, et al. Diabetic macular edema: correlation
37. Gaucher D, Saleh M, Sauer A, et al. Spectral OCT analysis in Bietti crystalline between microperimetry and optical coherence tomography findings. Invest
dystrophy. Eur J Ophthalmol 2010;20:612–4. Ophthalmol Vis Sci 2006;47:3044–51.
38. Gundogan FC, Dinc UA, Erdem U, et al. Multifocal electroretinogram 70. Vujosevic S, Casciano M, Pilotto E, et al. Diabetic macular edema: fundus
and central visual field testing in central areolar choroidal dystrophy. Eur J autofluorescence and functional correlations. Invest Ophthalmol Vis Sci
Ophthalmol 2010;20:919–24. 2011;52:442–8.
39. Szlyk JP, Paliga J, Seiple W, et al. Comprehensive functional vision assessment 71. Cusick M, Toma HS, Hwang TS, et al. Binasal visual field defects from simul-
of patients with North Carolina macular dystrophy (MCDR1). Retina 2005; taneous bilateral retinal infarctions in sickle cell disease. Am J Ophthalmol
25:489–97. 2007;143:893–6.
40. Renner AB, Tillack H, Kraus H, et al. Morphology and functional character- 72. McLoone E, O’Keefe M, McLoone S, et al. Effect of diode laser retinal ablative
istics in adult vitelliform macular dystrophy. Retina 2004;24:929–39. therapy for threshold retinopathy of prematurity on the visual field: results
41. Roth JA. Central visual field in diabetes. Br J Ophthalmol 1969;53:16–25. of goldmann perimetry at a mean age of 11 years. J Pediatr Ophthalmol
42. Remky A, Arend O, Hendricks S. Short-wavelength automated perimetry and Strabismus 2007;44:170–3.
capillary density in early diabetic maculopathy. Inv Ophthalmol Vis Sci 73. Kozobolis VP, Detorakis ET, Georgiadis GS, et al. Perimetric and retrobulbar
2000;41:274–81. blood flow changes following carotid endarterectomy. Graefes Arch Clin Exp
43. Hudson C, Flanagan JG, Turner GS, et al. Short-wavelength sensitive visual Ophthalmol 2007;245:1639–45.
field loss in patients with clinically significant diabetic macular oedema. 74. Meyer CH, Callizo J, Schmidt JC, et al. Functional and anatomical findings in
Diabetologia 1998;41:918–28. acute Purtscher’s retinopathy. Ophthalmologica 2006;220:343–6.
75. Imasawa M, Tsumura T, Kikuchi T, et al. Humphrey perimetry as a predictor 105. Lavinsky F, Tolentino MJ, Lavinsky J. The macular threshold protocol of the
of visual improvement after photodynamic therapy. Jpn J Ophthalmol Humphrey visual field analyzer: a superior functional outcome of intravitreal
2009;53:281–2. bevacizumab for the treatment of neovascular age-related macular degenera-
76. Chalam KV, Agarwal S, Gupta SK, et al. Recovery of retinal sensitivity after tion. Arq Bras Oftalmol 2010;73:111–5. 327
transient branch retinal artery occlusion. Ophthalm Surg Lasers Imaging 106. Squirrel DM, Mawer NP, Moody CH, et al. Visual outcome after intravitreal
2007;38:328–9. ranibizumab for wet age-related macular degeneration: a comparison between
77. Wong WT, Forooghian F, Majumdar Z, et al. Fundus autofluorescence in type best-corrected visual acuity and microperimetry. Retina 2010;30:436–42.

Chapter 12
2 idiopathic macular telangiectasia: correlation with optical coherence tomog- 107. Parravano M, Oddone F, Tedeschi M, et al. Retinal functional changes mea-
raphy and microperimetry. Am J Ophthalmol 2009;148:573–83. sured by microperimetry in neovascular age-related macular degeneration
78. Yamaike N, Kita M, Tsujikawa A, et al. Perimetric sensitivity with the micro treated with ranibizumab: 24-month results. Retina 2010;30:1017–24.
perimeter 1 and retinal thickness in patients with branch retinal vein occlu- 108. Schmidt-Erfurth UM, Elsner H, Terai N., et al. Effects of verteporin therapy
sion. Am J Ophthalmol 2007;143:342–4. on central visual field function. Ophthalmology 2004;111:931–9.
79. Barbazetto IA, Schmidt-Erfurth UM. Evaluation of functional defects in 109. Yodoi Y, Tsujikawa A, Kameda T, et al. Central retinal sensitivity measured
branch retinal vein occlusion before and after laser treatment with scanning with the micro perimeter 1 after photodynamic therapy for polypoidal cho-
laser perimetry. Ophthalmology 2000;107:1089–98 roidal vasculopathy. Am J Ophthalmol 2007;143:984–94.
80. Ageno W, Cattaneo R, Manfredi E, et al. Parnaparin versus aspirin in the 110. Imasawa M, Tsumura T, Kikuchi T, et al. Humphrey perimetry as a predictor
treatment of retinal vein occlusion. A randomized, double blind, controlled of visual improvement after photodynamic therapy. Jpn J Ophthalmol

Visual Fields in Retinal Disease


study. Thromb Res 2010;125:137–41. 2009;53:281–2.
81. Tsujikawa A, Hangai M, Kikuchi M, et al. Visual field defect after radial optic 111. Chen FK, Uppal GS, MacLaren RE, et al. Long-term visual and microperim-
neurotomy for central retinal vein occlusion. Jpn J Ophthalmol 2006;50: etry outcomes following autologous retinal pigment epithelium choroid graft
158–60. for neovascular age-related macular degeneration. Clin Exp Ophthalmol
82. Barak A, Kesler A, Gold D, et al. Visual field defects after radial optic 2009;37:275–85.
neurotomy for central retinal vein occlusion. Retina 2006;26:549–54. 112. Augustin AJ, Offermann I, Lutz J, et al. Comparison of the original Amsler
83. Afrashi F, Erakgun T, Uzunel D, et al. Comparison of achromatic and grid with the modified Amsler grid: result for patients with age-related
blue-on-yellow perimetry in patients with resolved central serous chorio- macular degeneration. Retina 2005;25:443–5.
retinopathy. Ophthalmologica 2005;219:202–5. 113. Nazemi PP, Fink W, Lim JI, et al. Scotomas of age-related macular degenera-
84. Sekine A, Imasawa M, Iijima H. Retinal thickness and perimetric sensitivity tion detected and characterized by means of a novel three-dimensional
in central serous chorioretinopathy. Jpn J Ophthalmol 2010;54:578–83. computer-automated visual field test. Retina 2005;25:446–53.
85. Dinc UA, Yenerel M, Tatlipinar S, et al. Correlation of retinal sensitivity and 114. Loewenstein A, Malac R, Goldstein M, et al. Replacing the Amsler grid: a new
retinal thickness in central serous chorioretinopathy. Ophthalmologica 2010; method for monitoring patients with age-related macular degeneration.
224:2–9. Ophthalmology 2003;110:966–70.
86. Baran NV, Gurlu VP, Esgin H. Long-term macular function in eyes with 115. Preferential Hyperacuity Perimetry Research Group. Preferential hyperacuity
central serous chorioretinopathy. Clin Experiment Ophthalmol 2005;33: perimeter (PreView PHP) for detecting choroidals neoavascularization study.
369–72. Ophthalmology 2005;112:1758–65.
87. Ozdemir H, Karacorlu SA, Senturk F, et al. Assessment of macular function 116. Amari F, Ohta K, Kojima H, et al. Predicting visual outcome after macular
by microperimetry in unilateral resolved central serous chorioretinopathy. hole surgery using scanning laser ophthalmoscope microperimetry. Br J
Eye 2008;22:204–8. Ophthalmol 2001;85:96–8.
88. Natsikos VE, Hart JCD. Static perimetric and Amsler chart changes in patients 117. Hikichi T, Ishiko S, Takamiya A, et al. Scanning laser ophthalmoscope correla-
with idiopathic central serous retinopathy. Acta Ophthalmol 1980;58:908–17. tions with biomicroscopic findings and foveal function after macular hole
89. Kiss CG, Barisani-Asenbauer T, Simader C, et al. Central visual field impair- closure. Arch Ophthalmol 2000;118:193–7.
ment during and following cystoid macular oedema. Br J Ophthalmol 118. Ozdemir H, Karacorlu M, Senturk F, et al. Retinal sensitivity and fixation
2008;92:84–8. changes 1 year after triamcinolone acetonide assisted internal limiting mem-
90. Remky A, Lichtenberg K, Elsner AE, et al. Short wavelength automated brane peeling for macular hole surgery – a MP-1 microperimetric study.
perimetry in age related maculopathy. Br J Ophthalmol 2001;85:1432–6. Acta Ophthalmol 2010;88:e222–7.
91. Remky A, Elsner AE. Blue on yellow perimetry with scanning laser ophthal- 119. Chung H, Shin CJ, Kim JG, et al. Correlation of microperimetry with fundus
moscopy in patients with age related macular disease. Br J Ophthalmol autofluorescence and spectral-domain optical coherence tomography in
2005;89:464–9. repaired macular holes. Am J Ophthalmol 2011;151:128–36.e3.
92. Takamine Y, Shiraki K, Moriwaki M, et al. Retinal sensitivity measurement 120. Kroyer K, Jensen OM, Larsen M. Objective signs of photoreceptor dis­
over drusen using scanning laser ophthalmoscope microperimetry. Graefes placement by binocular correspondence perimetry: a study of epiretinal
Arch Clin Exp Ophthalmol 1998;236:285–90. membranes. Invest Ophthalmol Vis Sci 2005;46:1017–22.
93. Iwama D, Tsujikawa A, Ojima Y, et al. Relationship between retinal sensitivity 121. Richter-Mueksch S, Vecsei-Marlovits PV, Sacu SG, et al. Functional macular
and morphologic changes in eyes with confluent soft drusen. Clin Exp mapping in patients with vitreomacular pathologic features before and after
Ophthalmol 2010;38:483–8. surgery. Am J Ophthalmol 2007;144:23–31.
94. Sohn EH, Chen FK, Rubin GS, et al. Macular function assessed by micro­ 122. Ezra E, Arden GB, Riordan-Eva P, et al. Visual field loss following vitrectomy
perimetry in patients with enhanced S-cone syndrome. Ophthalmology for stage 2 and 3 macular holes. Br J Ophthalmol 1996;80:519–25.
2010;117:1199–206.e1. 123. Hutton WL, Fuller DG, Snyder WB, et al. Visual field defects after macular
95. Biswas S, Funnell CL, Gray J, et al. Nidek MP-1 microperimetry and Fourier hole surgery: a new finding. Ophthalmology 1996;103:2152–9.
domain optical coherence tomography (FD-OCT) in X linked retinoschisis. 124. Melberg NS, Thomas MA. Visual field loss after pars plana vitrectomy with
Br J Ophthalmol 2010;94:949–50. air/fluid exchange. Am J Ophthalmol 1995;120:386–8.
96. Shenoy R, McCilvenny S. Microperimetric evaluation of macula in retino­ 125. Hillenkamp J, Saikia P, Gora F, et al. Macular function and morphology after
pathy of membranoproliferative glomerulonephritis type II: a case report. peeling of idiopathic epiretinal membrane with and without the assistance of
Eur J Ophthalmol 2006;16:634–6. indocyanine green. Br J Ophthalmol 2005;89:437–43.
97. Ahn JK, Seo JM, Chung H, et al. Anatomical and functional characteristics 126. von Jagow B, Hoing A, Gandorfer A, et al. Functional outcome of indocyanine
in atrophic maculopathy associated with spinocerebellar ataxia type 7. green-assisted macular surgery: 7-year follow-up. Retina 2009;29:1249–56.
Am J Ophthalmol 2005;139:923–5. 127. Uemura A, Kanda S, Sakamoto Y, et al. Visual field defects after uneventful
98. Midena E, Vujosevic S, Convento E, et al. Microperimetry and fundus vitrectomy for epiretinal membrane with indocyanine green-assisted internal
autofluorescence in patients with early age-related macular degeneration. limiting membrane peeling. Am J Ophthalmol 2003;136:252–7.
Br J Ophthalmol 2007;91:1499–503. 128. Tari SR, Vidne-Hay O, Greenstein VC, et al. Functional and structural
99. Remky A, Weber A, Arend O, et al. Topical dorzolamide increases pericentral measurement for the assessment of internal limiting membrane peeling in
visual function in age-related maculopathy: pilot study findings with short- idiopathic macular pucker. Retina 2007;27:567–72.
wavelength automated perimetry. Acta Ophthalmol Scand 2005;83:154–60. 129. Tsuiki E, Fujikawa A, Miyamura N, et al. Visual field defects after macular
100. Feher J, Kovacs B, Kovasc I, et al. Improvement of visual functions and hole surgery with indocyanine green-assisted internal limiting membrane
fundus alterations in early age-related macular degeneration treated with peeling. Am J Ophthalmol 2007;143:704–5.
a combination of acetyl-L-carnithine, n-3 fatty acids, and coenzyme Q10. 130. Balaiya S, Brar VS, Murthy RK, et al. Comparative in vitro safety analysis of
Ophthalmologica 2005;219:154–66. dyes for chromovitrectomy. Retina 2011;31:1128–36.
101. Wong WT, Kam W, Cunnigham D, et al. Treatment of geographic atrophy 131. Hart WM Jr, Burde RM, Johnston GP, et al. Static perimetry in chloroquine
by the topical administration of OT-551: results of a phase II clinical trial. retinopathy: perifoveal patterns of visual field depression. Arch Ophthalmol
Invest Ophthalmol Vis Sci 2010;51:6131–9. 1984;102:377–80.
102. Fujii GY, de Juan Jr E, Humayun MS, et al. Characteristics of visual loss by 132. Michaelides M, Stover NB, Francis PJ, et al. Retinal toxicity associated with
scanning laser ophthalmoscope microperimetry in eyes with subfoveal hydroxychloroquine and chloroquine: risk factors, screening, and progression
choroidal neovascularization secondary to age-related macular degeneration. despite cessation of therapy. Arch Ophthalmol 2011;129:30–9.
Am J Ophthalmol 2003;136:1067–78. 133. Salu P, Uvijls A, van den Brande P, et al. Normalization of generalized retinal
103. Fujii GY, de Juan Hr E, Sunness J, et al. Patient selection for macular transloca- function and progression of maculopathy after cessation of therapy in
tion surgery using the scanning laser ophthalmoscope. Ophthalmology 2002; a case of severe hydroxychloroquine retinopathy with 19 years follow-up.
109:1737–44. Doc Ophthalmol 2010;120:251–64.
104. Tezel TH, Del Priore LV, Flowers BE, et al. Correlation between scanning laser 134. Xiaoyun MA, Dongyi HE, Linping HE. Assessing chloroquine toxicity in RA
ophthalmoscope microperimetry and anatomic abnormalities in patients with patients using retinal nerve fibre layer thickness, multifocal electroretinogra-
subfoveal neovascularization. Ophthalmol 1996;103:1829–36. phy and visual field test. Br J Ophthalmol 2010;94:1632–6.
135. Almony A, Garg S, Peters RK, et al. Threshold Amsler grid as a screening tool 153. Souka AA, Hillenkamp J, Gora F, et al. Correlation between optical coherence
for asymptomatic patients on hydroxychloroquine therapy. Br J Ophthalmol tomography and autofluorescence in acute posterior multifocal placoid
2005;89:569–74. pigment epitheliopathy. Graefes Arch Clin Exp Ophthalmol 2006;244:
328 136. Anderson C, Pahk P, Blaha GR, et al. Preferential hyperacuity perimetry to 1219–23.
detect hydroxychloroquine retinal toxicity. Retina 2009;29:1188–92. 154. Gordon LK, Goldhardt R, Holland GN, et al. Standardized visual field assess-
137. Rodriguez-Padilla JA, Hedges 3rd TR, Monson B, et al. High-speed ultra-high- ment for patients with birdshot chorioretinopathy. Ocul Immunol Inflamm
resolution optical coherence tomography findings in hydroxychloroquine 2006;14:325–32.
retinopathy. Arch Ophthalmol 2007;125:775–80. 155. Mahajan VB, Stone EM. Patients with an acute zonal occult outer retinopathy-
Section 2

138. Eisner A, Austin DF, Samples JR. Short wavelength automated perimetry and like illness rapidly improve with valacyclovir treatment. Am J Ophthalmol
tamoxifen use. Br J Ophthalmol 2004;88:125–30. 2010;150:511–8.
139. Eisner A, Austin DF, Samples JR. Short wavelength automated perimetry and 156. Ozgur S, Esgin H. Macular function of successfully repaired macula-off retinal
tamoxifen use. Br J Ophthalmol 2004;88:125–30. detachments. Retina 2007;27:358–64.
140. Toggweiler S, Wieser HG. Concentric visual field restriction under vigabatrin 157. Alexandridis E, Janzarik BI. Restitution of the retinal sensitivity after cured
therapy: extent depends on the duration of drug intake. Seizure 2001;10: retinal detachment. In: Greve EL, editor. Second International Visual Field
420–3. Symposium. The Hague: Dr W Junk; 1977.
141. Jensen H, Sjo O, Uldall P, et al. Vigabatrin and retinal changes. Doc Ophthal- 158. Sakai T, Iida K, Tanaka Y, et al. Evaluation of S-cone sensitivity in reattached
Retinal Diagnostics

mol 2002;104:171–80. macula following macula-off retinal detachment surgery. Jpn J Ophthalmol
Retinal Imaging and Diagnostics

142. Hosking SL, Hilton EJ. Neurotoxic effects of GABA-transaminase inhibitors 2005;49:301–5.
in the treatment of epilepsy: ocular perfusion and visual performance. 159. Bourke RD, Dowler JG, Milliken AB, et al. Perimetric and angiographic effects
Ophthalm Physiol Optics 2002;22:440–7. of retinotomy. Aust NZ J Ophthalmol 1996;24:245–9.
143. Hilton EJ, Cubbidge RP, Hosking SL, et al. Patients treated with vigabatrin 160. Sasoh M, Ito Y, Wakitani Y, et al. 10-year follow-up of visual functions in
exhibit central visual function loss. Epilepsia 2002;43:1351–9. patients who underwent scleral buckling. Retina 2005;25:965–71.
144. McGwin G, Vaphiades MS, Hall TA, et al. Non-arteritic anterior ischaemic 161. Lai WW, Leung GY, Chan CW, et al. Simultaneous spectral domain OCT and
optic neuropathy and the treatment of erectile dysfunction. Br J Ophthalmol fundus autofluorescence imaging of the macula and microperimetric corre-
2006;90:154–7. spondence after successful repair of rhegmatogenous retinal detachment.
145. Jagle H, Jagle C, Serey L, et al. Visual short-term effects of Viagra: double- Br J Ophthalmol 2010;94:311–8.
blind study in healthy young subjects. Am J Ophthalmol 2004;137:842–9. 162. Abramson DH, Melson MR, Servodidio C. Visual fields in retinoblastoma
146. Stanford MR, Tomlin EA, Comyn O, et al. The visual field in toxoplasmic survivors. Arch Ophthalmol 2004;122:1324–30.
retinochoroiditis. Br J Ophthalmol 2005;89:812–4. 163. Rahhal FM, Abramson DH, Servodidio CA, et al. Automated perimetry in
147. Moraes LR, Cialdini AP, Avila MP, et al. Identifying live nematodes in diffuse patients with choroidal metastases. Br J Ophthalmol 1996;80:309–13.
unilateral subacute neuroretinitis by using the scanning laser ophthalmo- 164. Enoch JM. Quantitative layer-by-layer perimetry: Proctor lecture. Invest
scope. Arch Ophthalmol 2002;120:135–8. Ophthalmol Vis Sci 1978;17:208–57.
148. Plummer DJ, Sample PA, Arevalo JF, et al. Visual field loss in HIV-positive 165. Enoch JM, Fitzgerald CR, Campos EC. Quantitative layer-by-layer perimetry:
patients without infectious retinopathy. Am J Ophthalmol 1996;122:542–9. an extended analysis. New York: Grune & Stratton, 1980.
149. Falkenstein I, Kozak I, Kayikcioglu O, et al. Assessment of retinal function in 166. Enoch JM, Lawrence B. A perimetric technique believed to test receptive
patients with HIV without infectious retinitis by multifocal electroretinogram field properties: sequential evaluation in glaucoma and other conditions.
and automated perimetry. Retina 2006;26:928–34. Am J Ophthalmol 1975;80:734–58.
150. Plummer DJ, Marcotte TD, Sample PA, et al. Neuropsychological impairment- 167. Dubois Poulsen D. Acquired dyschromatopsias. Mod Probl Ophthalmol
associated visual field deficits in HIV infection. HNRC Group, HIV Neurobe- 1972;11:84–93.
havioral Research Center. Invest Ophthalmol Vis Sci 1999;40:435–42. 168. Hart Jr WM, Burde RM. Color contrast perimetry: the spatial distribution of
151. Hangai M, Fujimoto M, Yoshimura N. Features and function of multiple color defects in optic nerve and retinal diseases. Ophthalmology 1985;92:
evanescent white dot syndrome. Arch Ophthalmol 2009;127:1307–13. 768–76.
152. Forooghian F, Stetson PF, Gross NE, et al. Quantitative assessment of photo- 169. Makous W, Carroll J, Wolfing JI, et al. Retinal microscotomas revealed
receptor recovery in atypical multiple evanescent white dot syndrome. with adaptive-optics microflashes. Invest Ophthalmol Vis Sci 2006;47:
Ophthalmic Surg Lasers Imaging 2010;41(Suppl):S77–80. 4160–7.
Section 1 Anatomy and Physiology

Chapter The Development of the Retina


13  Thomas A. Reh

Our understanding of the mechanisms by which the complex


cellular assemblies of the retina develop has increased tremen- A
dously in the past 10 years. The growth of information in this
field has been stimulated by the increasing use of molecular
genetic methods and the identification of the genes that control
eye development in a variety of species. This chapter provides
an overview of the basic principles of retinal development that
have been known for some time and then highlights recent data
that have significantly extended these principles. It is impossible
to present a comprehensive review of this dramatically expand-
B
ing field in this short a space, but we hope the reader will come
to appreciate the complexity of the cellular and molecular inter-
actions that generate this spectacular tissue.

EMBRYOLOGY OF THE EYE


The development of the eye is an extremely complex process,
each step of which requires exquisite coordination among the
various tissues that contribute to it. As a result, examples of
inductive interactions at precise times during ocular develop-
ment are numerous. Thus it is not surprising that many terato-
gens disrupt these processes and lead to developmental C
abnormalities in eye formation. In addition, there are many con-
genital defects in eye formation that are caused by mutations in
genes critical for different stages in the process.
The retina is first recognized as the optic pit in the anterior
neuroectoderm at day 23 of gestation1; a few days later (day 25)
the optic vesicle can be recognized as an evagination from the
diencephalon of the neural tube (Fig. 13.1). The optic vesicles
initially lie immediately adjacent to the ectoderm (Fig. 13.1), but
then a layer of mesenchymal cells, derived from the neural crest,
becomes interposed between the ectoderm and the vesicles. The
neural crest cells that surround the developing optic cup, along
with the periocular mesenchyme, contribute to many ocular
tissues: the corneal endothelium, the choroid, the ciliary body
and the retinal vasculature (including the hyaloid artery).2
Fig. 13.1 The retinas develop as paired evaginations from the
During the next stage of ocular development, the optic vesicle
anterior central nervous system. (A) At stage 11 (13–20 somites in
folds in on itself, becoming the optic cup, and the lens ectoderm humans), the optic vesicles are continuous with the ectoderm near
invaginates into the vesicle (Fig. 13.1). The resulting two tissue the point of closure of the anterior neuropore. (B) By postovulatory
layers of the optic cup are the outer presumptive retinal pigment day 26, stage 12, the neuropore has closed and continued rostral
growth of the prosencephalon now results in a relative caudal
epithelium (RPE) and the inner presumptive neural retina, with displacement of the vesicles. At this time the presumptive neural
the lens vesicle filling much of the interior of the cup. In humans, retina (green) is in close contact with the lens ectoderm (red), the
these dramatic morphologic changes occur at approximately ventral part of the optic vesicle contributes to the optic stalk, and
25–35 days of gestation. The optic stalk, connecting the develop- the dorsal region of the optic vesicle gives rise to the pigmented
epithelium (blue). (C) By stage 15, 34 postovulatory days, the lens
ing retina with the diencephalon, will form the scaffold for the vesicle has completely formed, and the neural retina and pigment
growing axons of the optic nerve. The developing optic cup epithelial layers are distinct.
Table 13.1 Key stages of retinal development, genes associated with those stages, and clinical conditions resulting from their mutations

Age Stage Genes Phenotype 331


3–4 weeks Optic pit; vesicle Pax6, Sox2, Rax, Shh, Six3, Bmp7 Anophthalmia

Chapter 13
5 weeks Optic cup patterned into RPE and Vsx2, Bcor, Maf, Pax2, Bmp4 Microphthalmia
neural retina

5.5–6 Neurogenesis Zfhx1b, Vsx2, Bcor, Maf, Pax2, Vax2 Microphthalmia, coloboma
weeks Choroidal fissure closure

8–30 weeks Neural retina histogenesis Aipl1, Crx, Crb1, RPE65, Gucy2d, Leber’s congenital amaurosis
Rpgrip

The Development of the Retina


RPE, retinal pigment epithelium.

grows around the stalk from the dorsal sides, fusing to form the Pax6. Pax6 codes for a transcription factor that is a member of
ventral (or embryonic) fissure. This fissure “closes” at day 33 in the paired class of homeodomain proteins. Transcription factors
the human embryo, and incomplete closure of the fissure occurs of this class are expressed very early in the development of many
from mutations in several different genes leading to colobomas tissues and appear to be involved in controlling the identity of
(Table 13.1). The hyaloid artery, the major source of blood for the various regions of the embryo. Pax6 is expressed in the eye
the embryonic retina and lens, enters the eye through the ventral field, and continues to be expressed by both the optic vesicle and
fissure. The hyaloid artery regresses by birth, but the central the developing lens. Mutations in this gene cause a phenotype
retinal artery remains. Incomplete or delayed regression of the in mice characterized by small eyes and aniridia in humans.6 The
hyaloid artery can lead to vitreal hemorrhage and studies in homologous gene in Drosophila, called eyeless, is necessary for
mice have implicated the gene Norrin in this process.3 eye development in these animals as well.7 Remarkably, mis­
expression of the eyeless gene in the larval tissue that normally
gives rise to the leg causes ectopic eyes to form on the leg in the
THE EYE FIELD adult fly.8 This result has led to the proposal that Pax6/eyeless is
Although the eye is first apparent as it evaginates from the dien- a master control gene, responsible for activating the other genes
cephalon, this region of the embryo is already specified to give necessary for eye development.9
rise to the eye some time earlier.4 Since much of the experimental More recent data have shown that Pax6 is only one of many
embryology of the developing eye has been carried out in frog homeodomain proteins that are important for normal eye devel-
and chick embryos, most of the information on this process is opment in both vertebrates and invertebrates. The coordinated
derived from these species; however, it is likely that the process actions of these genes together contribute to the formation of the
of eye development has been largely conserved throughout ver- cells of the neural retina. These can act both as multimeric com-
tebrate evolution, and most of what has been learned from other plexes and/or as part of a hierarchical pathway (Fig. 13.2). One
species is likely to be true of human eye development as well. of the key transcription factors, Rx (RAX in humans), for example,
During gastrulation, the process known as neural induction is necessary for the very earliest stages of eye formation, and
transforms a region of ectoderm into the precursors of the central targeted deletion of this gene in mice blocks eye development
nervous system (CNS), called the neural plate. Even at these almost completely.10 Rx, then, is close to the beginning of the
early times in development, the anterior region of the neural cascade of the EFTFs. When Rx is expressed in retinal precursors,
plate is specified to give rise to the retina. This region is called it then activates the transcription of Pax6, which then turns on
the eye field. Indelibly labeling the cells in the eye field of the many of the other EFTFs. Experimental misexpression of each of
neural plate with vital dyes allowed embryologists of the 1920s the different EFTFs can produce some ectopic eye-like structure,
to track the cells as they developed into the neural retina and but coordinated misexpression of the EFTFs together (Pax6, Rx1,
pigmented epithelium (Fig. 13.2). Additional evidence that the Six3, Six6, Lhx2, Nr2e1, and Tbx3), along with the anterior pat-
eye field cells were specified to produce eye tissue came from terning gene Otx2, is sufficient to induce ectopic eye fields and
transplantation studies of these cells to ectopic locations, such as eyes in amphibia at a high frequency even in nonneural regions
the trunk or tail of other embryos; ectopic eyes formed from the of the animal like the belly. These experiments in frog embryos,
transplanted cells. In situ hybridization studies to localize these along with others in mice, have led to the current model that
genes have shown that genes that are critical to eye development EFTFs cross-regulate one another in a feedforward manner.5 The
are expressed in the eye-forming regions of the neural plate fact that similar genes are critical for eye development in both
before any clear morphologic differentiation into retinal struc- Drosophila and vertebrates has had a major influence on the
tures (see below). understanding of the evolution of the eye.9 Although the cellular
What makes the eye field cells capable of generating retina? In components of eyes in the various phyla are quite different, the
part, the eye-forming character of these cells comes from their genes that may ultimately control the expression of the photo-
expression of a group of proteins, called the eye field transcrip- transduction machinery may have been shared by a common
tion factors (EFTFs), which bind to DNA and selectively activate ancestor.
genes important for eye development.5 Among the first of these The eye field is originally continuous across the neural plate
transcription factor genes to be expressed in eye development is at its anterior end; however, this single field is soon split in
332
ET & Rx1 ET & Rx1
noggin Otx2 Pax6, Six3 Pax6, Six3
Eye
Section 1

Lhx2 Lhx2, tll


Optx2

Neural indication Fore-/Midbrain specification Eye field specification


stage 10.5 stage 11 stage 12.5
Anatomy and Physiology
Basic Science and Translation to Therapy

Six3
Otx2

noggin ET Rx1 Pax6 tll

Lhx2

Optx2

Fig. 13.2 A network of transcription factors, in addition to Pax6, regulates the formation of the eye. Upper series of drawings show the neural
plate stages of the Xenopus frog and the stages in eye field specification. Drawings in the middle panel show the complex genetic regulatory
network activated at the corresponding stages. See text for detailed descriptions of these genes. Lower panels show representative in situ
hybridization for several of the eye transcription factors at the same stages as drawn above to show their localization in Xenopus embryos. From
left to right, the panels show Otx2, Stage 12; Otx2, Stage 13; Otx2 (purple) and Rx1 (red), Stage 13; and Rx1 alone, Stage 13. (Modified with
permission from Zuber ME, Gestri G, Viczian AS, et al. Specification of the vertebrate eye by a network of eye field transcription factors.
Development 2003;130:5155–67.)

two by a factor from the underlying prechordal mesoderm that


lies immediately adjacent to the ventral midline of the neural
plate. Deletion of the prechordal mesoderm results in the devel-
opment of a single fused eye (cyclopia) at the ventral part of
the diencephalon. The factor released by the prechordal meso-
derm that suppresses eye development in the middle of the PE
field is thought to be a molecule called Sonic hedgehog (Shh),
an extracellular glycoprotein important in several other induc-
tive events throughout the embryo. Shh is released initially by
NR
the prechordal mesoderm and induces cells in the ventral dien- OS
cephalon (Fig. 13.3) to produce the same factor.11 This factor
then acts on the neighboring cells of the ventral diencephalon
to suppress eye development. Mice lacking the Shh gene die
as embryos; nevertheless, these embryos develop to a stage
where the paired optic vesicles would normally form. However,
in animals lacking Shh, the eye field is not split at the midline
and a single optic vesicle forms, resulting in cyclopia.12 The Fig. 13.3 Factors that pattern the embryonic diencephalon.
conservation of these inductive signals in human development Shh (blue) inhibits eye development in the ventral diencephalon, and
fibroblast growth factor (red), in the ectoderm overlying the optic
has been confirmed by the report that congenital holoprosen- vesicle, promotes neural retinal development. PE, pigment epithelium;
cephaly, a condition that frequently displays varying ocular NR, neural retinal differentiation; OS, optic stalk.
defects, including cyclopia, is caused by mutations in the expressed in the lens ectoderm and the developing neural retinal
Shh gene.12–14 domain.20,21 Second, experimental treatment of the developing
chick optic vesicle with exogenous FGFs or antibodies that block 333
PATTERNING THE RETINAL, RPE, AND FGFs causes perturbations in the development of the retina. The
optic vesicle of the chick embryo develops into an optic cup
ANTERIOR DOMAINS OF THE OPTIC CUP

Chapter 13
when isolated from the embryo and maintained overnight in
The next stages of eye development in vertebrates also require culture. The addition of exogenous FGF to these optic vesicle
a number of inductive interactions to coordinate the various cultures causes the presumptive pigmented epithelial layer to
tissues that ultimately contribute to the structure. Both the neural develop instead into a neural retina.21 Antibodies raised against
retina and the pigmented epithelium arise from the optic vesicle FGF cause the opposite effect and block neural retinal formation
region of the neural tube. Although both are derived from the in similar optic vesicle cultures. Although it is not clear which
optic vesicle, these two tissues are quite distinct; the neural FGFs are necessary in mammals for neural retinal specification,

The Development of the Retina


retina is a multilayered structure containing millions of neurons interruption of all FGF signaling by conditional deletion of a
and photoreceptors, whereas the pigment epithelium is a single downstream factor in the FGF signal transduction pathway,
layer of nonneural, pigmented, cuboidal cells. The appropriate Shp2, causes a loss in Vsx2 expression and a transition of the
development of both of these two very different parts of the affected region of the optic cup from neural retina to RPE.22
retina requires interactions with the adjacent tissues. If the optic These results are consistent with a model in which FGF promotes
vesicle is isolated from the surrounding epidermis and mesen- neural retinal specification in the optic cup, by promoting Vsx2
chyme, differentiation is arrested at the optic vesicle stage and expression, which represses expression or activity of Mitf. The
the eye does not form. Transplantation experiments in many outer part of the optic cup, the presumptive RPE, is then speci-
species have shown that the developmental decision to develop fied by Shh, Wnt and activin/BMP signaling, which promotes
as either a neural retinal progenitor or alternatively as a pig- Mitf expression to repress Vsx215 (Fig. 13.3).
mented epithelial cell is regulated by factors in the microenvi-
ronment surrounding the eye. For example, if an optic vesicle is HISTOGENESIS OF THE RETINAL
transplanted to a position in an embryo adjacent to the develop-
ing hindbrain, near the otic vesicle, a second neural retina is
CELL TYPES
formed from the presumptive pigmented epithelial layer. In the next phase of retinal development, the various classes of
The distinction between the RPE and the neural retina comes neurons are generated by cells that undergo repeated mitotic
about as a result of their expression of different transcription cell divisions. The mitotic progenitors have a relatively short cell
factors. The transcription factor Vsx2/Chx10 specifies the neural cycle and are able to produce the hundreds of millions of cells
retinal domain, while the basic helix–loop–helix (bHLH) tran- in the human retina in a few months, from the 10th to the 24th
scription factor Mitf defines the RPE. Loss of Mitf leads to con- week of gestation in humans. At the same time that many of
version of the RPE into a second layer of neural retina, whereas the progenitors undergo symmetric cell divisions to enable the
loss of function mutations in Vsx2/Chx10 leads to a conversion nearly exponential increases in cell numbers, some of the cell
of the neural retina into RPE (for review, see Fuhrmann15). Otx2 divisions of the progenitors result in postmitotic neurons
is also required for RPE development, and expression of Otx2, throughout this period of histogenesis. The different types of
along with the signaling molecule Wnt, can convert neural retina retinal cells are not produced by the progenitor cells all at the
to RPE.16 Along with these transcription factors, the specification same time. Rather they are generated in a sequence that has been
of RPE is also controlled in part by soluble signaling molecules, conserved in all vertebrates. The sequence was first described
which induce the outer region of the optic vesicle to adopt the using the 3H-thymidine “birthdating” technique. Following an
pigmented cell fate instead of becoming retina. In particular, this injection of 3H-thymidine, the nucleotide is incorporated into the
mesenchymal tissue upregulates RPE-specific genes like Mitf, DNA of the retinal progenitor cells during the S phase of their
while downregulating retina-specific genes. The transforming cell cycle. Those cells that withdraw from the cycle after their
growth factor-β (TGF-β) family member activin was able to next mitosis, and become postmitotic neurons/photoreceptors,
mimic the effects of the extraocular tissue on the optic vesicle, retain a high level of label in their nucleus. Since the 3H-thymidine
which suggests an activin-like signal is required for normal RPE is available for only a short period, those cells that remain in the
development.17 Experiments in developing chick embryos have cycle for additional cell divisions become progressively less
also implicated another TGF-β-like signaling molecule, bone heavily labeled. If the animal is allowed to survive to adulthood
morphogenetic protein (BMP), in the process of RPE specifica- and the retina processed for autoradiography to reveal the label
tion18 and deletion of the Bmp7 gene in mice leads to disruptions in the various retinal cells, those retinal cells that were generated
in ocular development. Another signaling molecule, Shh, intro- on the day of the thymidine injection are easily identified by the
duced earlier in this review in the context of the eye field, is also large number of silver grains over their nuclei. In a typical thy-
involved in the induction of RPE cell fate in the ventral region midine birthdating study, injections of the nucleotide are made
of the optic vesicle, since inhibition of Shh signaling with cyclo- in pregnant animals on each day of gestation, and the number
pamine in frog embryos disrupts ventral RPE development, and of labeled cells of the various retinal cell classes is calculated to
two strains of mice with altered Shh signaling do not develop produce a graph like the one shown in Fig. 13.4.23
ventral RPE.19 This type of thymidine birthdating analysis has been carried
Signaling factors are also important in the specification of the out in the retina in many different animals, and the sequence of
neural retinal domain of the optic cup. Several lines of evidence cell generation is remarkably conserved among the various ver-
support a critical role for fibroblast growth factor (FGF) in the tebrates. Figure 13.4 shows the sequence of generation of the
development of the neural retinal part of the eye. First, FGFs are retinal cell classes in the monkey,23 but a very similar graph can
334
Section 1

ROD

CON

HOR
Anatomy and Physiology
Basic Science and Translation to Therapy

PRO
MUL
AMA
BIP
RGC

100
Cumulative percent of cells generated

80

60 Ganglion
Horizontal 50%
Cones
40 Amacrine
Müller
Bipolar
20
Rods

0
0 50 100 150
B Age at injection (postconception days)

Fig. 13.4 (A, B) Each retinal cell type is generated over a slightly different time course during retinal histogenesis. Addition and integration of
new cells during the histogenesis of the retina at three different ages, corresponding to the chart of thymidine birthdating of retinal cells in the
monkey. CON, cones; HOR, horizontal; PRO, progenitor cells; RGC, retinal ganglion cell; AMA, amacrine; MUL, Müller; BIP, bipolar. (Panel B
modified with permission from La Vail MM, Rapaport DH, Rakic P. Cytogenesis in the monkey retina. J Comp Neurol 1991;309:86–114.)

be produced in other vertebrates. Overall, the cell classes can be many different types of retinal neurons up to and including
divided into two phases of generation. In the first phase, the their final mitotic cell division.26,27 Several methods have been
ganglion cells, the cones, and the horizontal cells are generated. developed for tracing the lineage of individual retinal progeni-
In the second phase of histogenesis, the rod photoreceptors, the tor cells: (1) retroviral infection of progenitors with a virus
bipolar cells, and the Müller glial cells are produced by the pro- containing a reporter gene26,27; (2) direct injection of progeni-
genitor cells.24,25 Amacrine cells are primarily generated in the tors with a cell-impermeant dye28,29; (3) genetically inducing
later phase, but many amacrine cells become postmitotic at the a lineage marker into specific types of progenitors based on
same time as ganglion cells are generated, so these cells do not their gene expression.30,31 These methods all show that many
fall as neatly into one or the other phase. Despite this seeming of the mitotic progenitor cells can produce various, somewhat
regularity in histogenesis, it should be noted that there are dis- random mixtures of the different retinal cell classes; however,
tinct central-to-peripheral gradients of histogenesis, and that they also show that in some cases progenitors can generate
peripheral retina may still be in the first “phase” at the time only a subset of the different types of retinal cell types; for
central retina is generating later cell types. example, the progenitors that express the transcription factor
Even though the retinal cell types are generated in a defined Ascl1 can produce all the different types of retinal cells except
sequence, the progenitors are multipotential, and can generate ganglion cells.30
The sequential development of the different retinal cell types transcription factor, FoxN4, produces the complementary result:
has led several investigators to propose that the production of all cells develop normally, except amacrine cells.43 The combina-
one cell class induces the progenitor cells to make the next cell tion of FoxN4 and Pax6 is therefore able to confer progenitors 335
type in the sequence. For example, the retinal ganglion cells, with competence for all retinal cell fates. Other transcription
being generated first by the progenitor cells, might secrete a factors that are expressed in progenitors or newly produced

Chapter 13
substance that prevents additional cells from differentiating into neurons play important roles in the production of cell diversity
this fate, and at the same time instructs the progenitor cells to and/or the maintenance of this diversity as the neurons acquire
begin making the next cell type, the horizontal cells; the horizon- their differentiated fates. Deletion of the proneural gene Atoh7
tal cells would then secrete a factor that instructs the progenitor (Math5) leads to a dramatic failure in ganglion cell production44;
cells to make cones, and so on until all the retinal cell types have the gene Otx2 is expressed in the newly developing photorecep-
been generated. Cell culture studies have provided some support tors and genetic deletion of this gene specifically in the retina
for this model: when progenitor cells from the retina are isolated prevents the progenitors from producing photoreceptors, and

The Development of the Retina


from early stages of retina, they predominantly differentiate into instead they produce more amacrine cells45–47; the transcription
retinal ganglion cells.32 However, if these same early progenitor factors Ptf1a, NeuroD1, and Math3 are expressed in amacrine
cells are mixed with retinal cells from later stages of develop- cells, and loss of one or more of these genes causes a loss in
ment, presumably containing factors derived from the cell types amacrine cells in the developing retina48; deletion of the Nrl
generated later, the early progenitor cells will be biased to adopt transcription factor causes all the rods to develop as short-
later cell identities.33–35 Thus the type of cell that the progenitor wavelength sensitive cones instead49; mutation in the paired-
will produce at any particular time in development can be influ- homeodomain gene Chx10/Vsx2 leads to an absence of most
enced by factors produced by the neighboring cells. There is also bipolar cells50; elimination of the transcription factor Prdm1/
in vivo evidence in mice that cell-type specific feedback mecha- Blimp causes the photoreceptors to become bipolar cells instead51;
nisms control the relative ratios of retinal cells: the signaling deletion in the Ascl1 gene, another bHLH transcription factor,
factor Shh, mentioned earlier in the development of the eye field, leads to an increase in the production of Müller glia at the
is also expressed in the retinal ganglion cells, and this factor expense of late-generated retinal neurons, like bipolar cells and
alters the rate of proliferation of the progenitors and their cell rods.52–54 Putting all these molecular interactions into a coherent
cycle exit, thereby controlling the addition of new ganglion network model has yet to be accomplished; moreover, integra-
cells.36 However, while there appear to be feedback mechanisms tion of these transcriptional regulators with the miRNAs and
within cell types for controlling their numbers, elimination of signaling factors remains a challenge.
ganglion cells does not appear to affect the development of the Several cell classes that are resident in the retina are not
other retinal cells drastically,37 and so the evidence to date sup- derived from the progenitor cells in the ventricular zone, includ-
ports regulation within cell types rather than between cell types. ing the microglia, vascular endothelial cells, and retinal astro-
An alternative mechanism for the production of the different cytes. Microglia in the retina, like microglia elsewhere in the
retinal cell types from a common precursor could be that the CNS, are derived from the blood; monocytes enter the retina
progenitors undergo a progressive change in their “internal through the vascular endothelial cells and become “ameboid” in
state,” like a clock ticking through the different cell fates.38,39 In morphology.55,56 These highly motile, phagocytic cells digest
this model the retinal progenitor cells change over development those retinal cells that undergo apoptotic cell death (see below)
in the cell types they are competent to give rise to. First, the during the course of retinal development. When they are first
default state, as discussed above, is the retinal ganglion cell. observed in development, they are concentrated in the ganglion
Next, the progenitor cells shift their competence so that they are cell layer (GCL), presumably as a result of the large number of
more likely to produce horizontal cells, then cone photorecep- degenerating cells in that layer. Later in development, when
tors, and so on. A cascade of transcription factors might be other types of retinal cells undergo apoptosis, microglia are
responsible, with the first one setting in motion the mechanism found in the inner nuclear layer (INL) and outer nuclear layer
for the production of the second, which acts to produce a third (ONL). A second type of glial cell that is present in the mature
transcription factor, and so on. Regardless of which of these retina is the astrocyte. These cells form from ventricular zone
models explains the generation of cell diversity from the multi- cells in other areas of the CNS, but they are not generated by the
potent progenitors, recent data have shown that miRNAs are retinal progenitors; instead, the retinal astrocytes migrate along
necessary in this process. Deletion of the gene that codes for the the developing optic nerve and enter the retina at the optic nerve
enzyme Dicer, needed to produce mature miRNAs, causes the head. The vascular endothelial cells emerge from the same point,
progenitors to be “stuck” in the early state, and only generate and both the astrocytes and the endothelial cells migrate across
early cell fates, like ganglion cells, cones, and horizontal cells.40,41 the retinal surface, eventually covering it completely.57,58
Although we still do not understand the mechanism by which
the progenitors change over time to control the types of cells that
are generated, several transcription factors important for the
INNER RETINAL DEVELOPMENT
development of different types of retinal cells have been identi- The further development of the inner retina follows a sequence
fied. One transcription factor expressed by progenitors that con- similar to that described in other areas of the developing CNS.
trols the types of cells that are generated is Pax6. Although Immediately after their final mitotic division at the ventricular
mutations of the Pax6 gene cause early defects in retina develop- (scleral) surface, retinal cells migrate to their appropriate lamina.
ment, it has also been possible to delete the gene specifically As they migrate, the different types of neurons begin to take on
from the retinal progenitors at later stages of development. some morphologic features of their characteristic cell type. For
This leads to a loss of competence in the progenitor cells to gen- example, ganglion cells begin to elongate an axon from their
erate anything except amacrine cells.42 Deletion of a different basal/vitreal process before their soma reaches the GCL.59,60 The
next phase in the differentiation of retinal neurons is the growth subspecificity from the outset.65 In fact, the arbors have more
of dendritic processes. In the last stages of differentiation the higher-order branches than adult ganglion cells. As the ganglion
336 retinal neurons make functionally active synapses with one cells mature, the dendritic arbor is remodeled; the dendritic pro-
another and express their transmitters and receptors. Although cesses become restricted to a single sublamina of the IPL,
the time course of these events overlaps considerably, this depending on whether the ganglion cell will become an ON or
sequence is typical of most classes of retinal cells. OFF type, and the numerous tertiary processes regress. After the
Section 1

The development of the inner retina is led by the differentia- active phase of ganglion cell dendritic growth, the total extent
tion of the retinal ganglion cells. The morphologic development of the dendritic field continues to expand, most likely as the
of retinal ganglion cells has been well characterized in several result of the passive stretching of the retina with the continued
species, including primates, and is known to proceed through a growth of the eye.
characteristic sequence.61,62 The first phase of ganglion cell mor- What factors determine the extent and shape of the ganglion
phogenesis begins as soon as the cells complete their final mitotic cell dendritic arbors? As the ganglion cells begin to send out
Anatomy and Physiology
Basic Science and Translation to Therapy

division at the ventricular surface of the developing retina. The dendrites, the amacrine cells are migrating to the inner nuclear
vitreal process begins to resemble an axon, which extends layer; the processes of these two cell types grow on one another
toward the optic disc even before the migration of the cell soma to begin the IPL. The dendritic arbors of ganglion cells are in
from the ventricular surface. The cell soma then migrates to the part regulated by the same CAMs that mediate the lamination
vitreal surface of the retina. The molecular mechanisms that of the cells themselves. Kljavin et al.66 cultured retinal ganglion
underlie ganglion cell migration are not understood; neverthe- cells on substrates of purified CAMs and found that cells grown
less, the laminar structure of the retina is likely the result of the on NCAM, L1, or N-cadherin developed distinctly different
selective migration and specific adhesions among the different morphologies from one another. Those cells grown on N-cadherin
types of retinal cell. Selective adhesive interactions are mediated developed a highly branched morphology most similar to that
by cell adhesion molecules (CAMs). It has been known for some observed in vivo, whereas the cells grown on L1 developed
time that interfering with CAM function, particularly the mol- simpler, axon-like processes. However, these generic CAMs
ecule known as N-cadherin, causes a disruption of the normal cannot provide the degree of specificity characteristic of the
lamination pattern in the retina.63 Although interfering with retina. To generate the sublaminar specificity, for example, at
CAM function leads to a disruption in the lamination of the least two other types of molecule are required. Several members
retina, it is still not well understood how these relatively nonsel­ of the immunoglobulin superfamily of adhesion molecules
ective molecules direct the particular retinal cell types to their (Dscam (Down’s syndrome CAM), Dscaml, Sidekick-1 and
appropriate laminae. In the developing cerebral cortex, newly Sidekick-2) are required in chick retina to define the sublaminae
generated neurons migrate to their destinations along radially in the IPL67; however, in the mouse retina, loss of Dscam does
arranged glial cells that span the expanding neural tube from the not interfere with sublaminar specificity. Instead, in the mouse
ventricular surface at the core of the brain to the external surface. retina, molecules known to be critical for axonal pathfinding in
Several different molecules have been identified that are critical the CNS (Sema6A and PlexinA4) are required for sublaminar
in this migration, including adhesion molecules that mediate the dendritic stratification of at least some types of ganglion cell and
selective attachment of the migrating neurons to the glial scaf- amacrine cell.68 Sema6A and PlexinA4 have complementary
fold.64 Although similar molecules may be expected to mediate expression patterns in the sublaminae of the IPL (Fig. 13.5B), and
the migration of ganglion cells in the retina, there is little evi- mice with PlexA4 knocked out have a defect in the correct sub-
dence for the presence of radial glial cells in the developing laminar specificity of the dendrites of some types of retinal
retina. Müller cells could provide such a scaffold; however, these neurons; Fig. 13.5 shows the effects of loss in PlexA4 on the
cells have not yet been generated at the time of ganglion cell dendrites of the tyrosine hydroxylase-expressing dopaminergic
migration (see above). Although some reports using ultrastruc- amacrine cells.
tural criteria have claimed that Müller cells are present in the In addition to their laminar specificity, a number of studies in
developing retina before they have been birthdated by thymi- many different species have shown that the dendritic arbor of
dine, extensive serial section reconstructions by Hinds and each subclass of ganglion cells “tiles” the retinal surface.69 Inter-
Hinds59 failed to find any evidence for radial glial cells during actions among the ganglion cells control the extent of coverage
the period of ganglion cell genesis. Therefore it is likely that the of the dendrites of the various ganglion cell classes. When a
newly generated ganglion cells use the other retinal progenitor region of the retina is experimentally depleted of ganglion cells,
cells as their scaffold for migration. In fact, more recent evidence the neighboring cells will sprout dendrites into the depleted
in the cerebral cortex indicates that the radial glial cells are in areas and thereby expand the size of their arbors considerably.
fact progenitor cells, consistent with the possibility that migrat- In addition, when the density of ganglion cells is increased by
ing neurons in the retina use the progenitors to guide their monocular enucleation before the period of normal cell death
migration. (see below), the ganglion cells have smaller dendritic fields.70
In the next phase of ganglion cell development, the cells Although these data demonstrate that size of the dendritic arbor
extend dendrites to form the inner plexiform layer (IPL). These of a ganglion cell is determined by cell–cell interactions, the
first dendrites are very simple, no more than a single primary nature of the interactions is not clear. The ganglion cells could
large filopodial process, with growth cones at their ends. Gan- be competing for some dendrite-promoting factor derived from
glion cells next elaborate their dendritic arbors. The complex the amacrine cells, so when fewer ganglion cells are present, they
arbor emerges rather quickly. For many ganglion cells, the den- can get more of the factor. Alternatively, the ganglion cells may
dritic arbors initially span the entire width of the IPL, with intrinsically extend exuberant dendrites, but their growth may
numerous secondary and higher-order branches; however, in be limited by a phenomenon known as contact inhibition, in
at least one type of ganglion cell, the cells have laminar which a direct contact between two cells leads to the cessation
Wild type Dscam -/- 337

Chapter 13
The Development of the Retina
A
PlexA4/Sema6A Wild type PlexA4 -/-

Fig. 13.5 The tiling and lamination of dendrites are regulated by specific cell adhesion molecules. (A, left) Midget ganglion cell mosaic of human
retina shows a highly regular distribution, and their dendritic arbors show no overlap. (Modified with permission from Dacey DM. The mosaic of
midget ganglion cells in the human retina. J Neurosci 1993;13:5334.) (A, right) In Dscam knockout mice the ganglion cells no longer tile evenly,
but have fasciculated dendrites, and their cell bodies (green) form large clumps. (Modified with permission from Fuerst PG, Bruce F, Tian M,
et al. DSCAM and DSCAML1 function in self-avoidance in multiple cell types in the developing mouse retina. Neuron 2009;64:484–97.) (B, left)
Sema6A (green) and PlexinA4 (red) have complementary expression patterns in the sublaminae of the inner plexiform layer. (B, right) Tyrosine
hydroxylase (TH)-expressing dopaminergic amacrine cells (green) in PlexA4 knockout mice send dendrites into the wrong sublamina. (Modified
with permission from Matsuoka RL, Nguyen-Ba-Charvet KT, Parray A, et al. Transmembrane semaphorin signalling controls laminar stratification
in the mammalian retina. Nature 2011;470:259–63.)

of process extension because of the collapse of their growth even been generated.72 Thus the flow of information in the devel-
cones. Some evidence for this latter mechanism has been pro- oping retina is initially horizontal and only later develops the
vided from an analysis of the Dscam and Dscaml mouse mutants. predominantly vertical flow of information characteristic of the
As noted above, although these molecules are not required in mature retina.
mouse for sublaminar specificity, they do appear to be critical An exception to this pattern is found in the primate fovea,
for the phenomenon of tiling of the dendrites and cell bodies via where there are virtually no rods at any time in development.
“self-avoidance.” In Dscam knockout mice (Fig. 13.5), the gan- In the fovea the first synapses observed in the IPL are the
glion cells had fasciculated dendrites, and their cell bodies ribbon synapses between bipolar cells and ganglion cells.73 In
formed large clumps, instead of being spread out evenly across the monkey fovea, ganglion cells are born as early as 30 days
the retinal surface.71 of gestation, but bipolar and amacrine cells are not generated
The further development of the plexiform layers is a reflection until approximately 1 week later, at E38. As described above,
of the dendritic growth and the synapse formation of the various ganglion cells have primitive dendrites at E55, and the first
types of retinal cell. As they mature, the bipolar cells begin to synapses in the foveal IPL can be morphologically identified
produce dendritic arborizations that can form synaptic connec- at this same time, approximately 2 weeks after the bipolar cells
tions with the ganglion cells. As noted above, the IPL develops were generated. Thus in the primate fovea the vertical flow of
before the outer plexiform layer in all species that have been information appears to be in place before the conventional syn-
studied. In most mammals, conventional synapses, between apses among ganglion cells and amacrine cells. This is not true
amacrine cells and ganglion cells, develop before the ribbon of peripheral retina in the primate, where the typical mammalian
synapses between bipolar cells and ganglion cells. This sequence pattern characteristic of rod-dominated retinas is observed.
parallels the sequence of generation of these cell types, since the Since the connections among the ganglion cells and amacrine
amacrine cells are born before the bipolar cells in vertebrate cells in immature retina are thought to contribute to the coor-
retinas. In addition, these first synapses apparently mediate dinated bursts of ganglion cell activity required for appropriate
horizontal interactions among the cells of the inner retina that retinogeniculate connections, there may be differences in the
are important for the development of the appropriate pattern of mechanisms by which the connections from foveal ganglion
connections between ganglion cells and their targets. Studies by cells are specified.
Wong and collaborators in the ferret have shown that waves of The development of functional circuits among the retinal cells
activity are spread among the retinal ganglion cells through requires synaptic transmitters and their receptors to be expressed
synapses in the inner retina before most of the bipolar cells have in the appropriate cells (for review of synaptic development in
the retina, see Bleckert and Wong74 and Yoshimatsu et al.75). In cone. At each stage, a different transcription factor seems to
fact, the major neurotransmitters of the retina – glutamate, control the competence of the protophotoreceptor in its fate
338 gamma-aminobutyric acid, and glycine – are all localized to choices. The homeodomain transcription factor otx2 is the earli-
specific retinal neurons before development of morphologically est factor biasing progenitor cells to become photoreceptor.45,46
identifiable synapses. In addition, both ligand-gated and voltage- Conditional knockout studies show that photoreceptors do not
gated channels are present in retinal cells soon after they have develop in Otx2–/– retinas, and retroviral gene transfer of otx2
Section 1

been generated by the progenitor cell. For example, ganglion into retinal progenitors biases cells to become photoreceptors.
cells in the mouse have Na+, K+, and Ca2+ channels as early as Otx2 also activates transcription of cone–rod homeobox gene
E15, only a few days after birth. The fact that these very imma- (Crx), which is also required for expression of many photoreceptor-
ture neurons have both neurotransmitters and receptors allows specific genes, including the opsins.82 Otx2-positive Crx-positive
for these molecules to act as signals to shape the development protophotoreceptors then must choose to become rods or cones,
of the circuit. Although genetic suppression of bipolar cell trans- and this decision is made by expression of neural retina leucine
Anatomy and Physiology
Basic Science and Translation to Therapy

mitter release in mice does not affect the gross dendritic mor- (Nrl) zipper transcription factor. Nrl expression promotes rod
phology of the ganglion cell dendrites,76 neurotransmitters may cell fate and inhibits cone cell fate. Nrl–/– retinas do not develop
be more important in shaping the dendrites in the outer retina rods, but have many more cones than normal.49 Photoreceptor
(see below). progenitors not expressing Nrl become cones. Interestingly, the

PHOTORECEPTOR DEVELOPMENT
When photoreceptors are first recognizable by their opsin immu-
noreactivity or mRNA expression, they have a very simple
morphology and no outer-segment formation. Figure 13.6 shows
the simple morphology of the early differentiating cones in the
monkey retina. In all vertebrates the cone photoreceptors are
generated before the rod photoreceptors (see above); however,
the rod photoreceptors express their specific opsin before the
cones do.77,78 Thus at least this aspect of rod differentiation pro-
ceeds more rapidly than that of cones. The monkey has been a
particularly favorable species to study the normal pattern of A
photoreceptor differentiation, owing to the relatively long time L/M
course of retinal development in this species and the fact that 2 mm
S
the rod-dominated retinas of most other mammals have rela- L/M + S
tively few cones. The foveal cones of the rhesus monkey are Fwk 22
born between gestational days E38 and E50 and first make syn-
apses in the outer plexiform layer by E55. Thymidine birthdating F
studies indicate that the first foveal cones are generated on fetal
day 38 (see above) but do not express their specific opsin until
several weeks later at fetal day 75, well after the cones have
differentiated to the point of making synaptic connections with
bipolar cells (E55). This delay is a general feature of photorecep-
tor development in vertebrates and suggests that the factors
that control opsin expression act at a later time than the factors
that direct the retinal progenitor cells to the photoreceptor Fwk 17.4
cell fate. F
The factors that control the mosaics of cell differentiation
alluded to above must also be at work in the development of the
cone mosaics. In the primate fovea the cones expressing the S
(short-wavelength) opsin and those expressing the long- and
middle-wavelength opsins (L/M) both are distributed in regular
F
mosaics. In both primates and mice, the S opsin-expressing cone
photoreceptors emerge first in development.77 Figure 13.6 shows
the progression of S opsin expression from the central to periph- Fwk 15
B Fwk 13
eral retina in monkey retina. Once the S opsin cones have covered
a relatively large fraction of the retinal surface, the L/M opsin
cones begin their development in central retina.79 The L/M opsin Fig. 13.6 L/M and S cones in the fetal monkey retina. (A) Double-
labeled immunohistochemical staining of a fetal day 108 parafoveal
cone wave of development then follows that of the S opsin cones retinal section; L/M cones appear green and S cones appear red.
from the central retina to the periphery.80,81 (Reproduced with permission from Burnsted K, Jasoni C, Szel, et al.
Emerging evidence suggests that photoreceptor differentia- J Comp Neurol 1997;378:117–34.) (B) The distribution of the
different types of cones across the retinal surface as a function of age.
tion is controlled by transcription factors that promote a pro­
F, fovea; Fwk, fetal week (Redrawn with permission from Xiao M,
genitor cell to become a photoreceptor progenitor, then either Hendrickson A. Spatial and temporal expression of cone opsins during
a protocone or protorod, and then further to a specific type of monkey retinal development. J Comp Neurol 2000;425:545–59.)
choice of cone subtype is determined by yet another transcrip- other cells in the nervous system. The major family of trophic
tion factor, thyroid hormone receptor-ß2 (TRß2) and RXR-gamma,83,84 molecules shown to be important for ganglion cell survival is
which promote M opsin expression while inhibiting the expres- the neurotrophins, including nerve growth factor (NGF), brain- 339
sion of S opsin. Mice deficient in the TRß2 gene have no M derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), and
opsin-expressing cones, but rather have only S opsin cones.85 NT4/5. Of the neurotrophins, the best evidence indicates that

Chapter 13
A number of studies have used in vitro techniques to identify BDNF and NT3 are the most important for ganglion cell sur-
the factors that control photoreceptor differentiation during vival. Both BDNF and NT3 are expressed by cells in the targets
retinal development. Since many of these studies have been of the ganglion cells.97 Both of these neurotrophins promote the
carried out in rodents, and since rodent retinas have relatively survival of ganglion cells when added to cell cultures of retinal
few cones, most of the in vitro studies have concentrated on rod cells or when injected in excess into the target during the normal
photoreceptor differentiation. The finding that rod photorecep- period of cell death. Ganglion cells express receptors for BDNF
tors do not differentiate in low-density cell cultures, but do so and NT3, receptor tyrosine kinases known as trkB and trkC,

The Development of the Retina


readily in high-density cultures or retinal explants, led to the respectively. These data taken together are consistent with the
development of several in vitro assays for rod differentiation following model. Over twice as many ganglion cells are gener-
factors and the identification of several signaling molecules that ated by the retinal progenitor cells than are actually needed in
promote rod photoreceptor differentiation in vitro.86 These the adult.98 These cells extend axons to the lateral geniculate
factors are now being used to promote photoreceptor develop- nucleus and the superior colliculus and other targets. At the time
ment in cultures of embryonic stem cells.87–90 the axons reach the targets, the ganglion cells express trkB and
The development of the outer plexiform layer and synapses trkC, and they now require activation of these receptors for their
between the photoreceptors, horizontal cells, and bipolar cells continued survival. Since the supply of BDNF and NT3 is limited,
lags behind that of the IPL. In addition, the molecules that regu- many of the ganglion cells will have an insufficient activation of
late the specific connections among these cells are not identified. their trk receptors, so they then initiate a program of apoptosis.
Nevertheless, there is good evidence that the dendrites of the Although this scheme is intellectually attractive and complies
cells are regulated through interactions with one another. For with much of the available data, evidence suggests that addi-
example, the dendritic branching of horizontal cells depends on tional growth factors are also important in controlling the
the ratio of rods to cones. In mice, reducing cone number during number of ganglion cells in the retina.99,100
development causes an increase in horizontal cell dendritic There is evidence that transcription factors of the Brn3 class are
branching. The formation of ribbon synapses, specialized synap- important for ganglion cell survival. The Brn3 transcription
tic structures between rod and cone pedicles and horizontal cell factors are among a class of molecules called the POU domain
and bipolar cell dendrites, is also dependent on the activity of transcriptional regulators. In the retina, the Brn3 genes are expressed
the cells: inhibition of phototransduction in cones forces the cone exclusively in the retinal ganglion cells. Their expression has
bipolar cells instead to make synapses with rods.91 Blocking been characterized in mouse, cat, and monkey retina, but there
transmitter release in rods causes rod bipolar cells and horizon- does not seem to be a simple relationship between the particular
tal cells to send aberrant processes into the ONL and synapse Brn gene expressed and one of the major morphologic subtypes
with cones.92,93 of ganglion cells. Targeted disruption of the Brn genes in mice
causes the loss of nearly all of the retinal ganglion cells.101–103
These experiments lend support to the idea that Brn genes may
GANGLION CELL DEATH be important for the expression of the neurotrophins or their
It has long been recognized that a considerable number of receptors, as well as other genes critical for their survival.101,104
neurons die during the development of the vertebrate CNS. The
death of “excess” neurons is thought to be the result of their
inability to compete effectively for a limited supply of trophic
RETINAL MATURATION
support, usually from their postsynaptic targets. Such a mecha- As the retina matures, nearly all of the events continue to proceed
nism could ensure that presynaptic and postsynaptic popula- in a central to peripheral direction. Once the process of retinal
tions of neurons are numerically matched during development. histogenesis and death is complete, considerable remodeling
In the retina the phenomenon of cell death has been most thor- continues, primarily in the form of retinal stretch, owing to the
oughly studied in the retinal ganglion cell population,94 although steadily increasing intraocular pressure. The pigmented epithe-
cell death has been documented in other cell types.95 A consider- lium and the scleral tissue also participate actively in the growth
able fraction of the ganglion cells that are initially produced are of the eye, as these tissues proliferate in response to increases
subsequently lost during the phase of cell death. The loss of in intraocular pressure. Because the retina matures later in the
ganglion cells occurs as the axons of the cells reach the lateral periphery, a disproportionate amount of expansion occurs in the
geniculate nucleus and superior colliculus. The neurons in these peripheral retina. As a result, the ganglion cell density is lower
targets presumably produce only enough trophic factor to keep in the peripheral retina than in the central retina, and the den-
less than half of the ganglion cells alive in most vertebrates and dritic arbor size of most types of retina cells increases in extent
in humans. In addition to numerically matching ganglion cells with eccentricity because of the disproportionate retinal stretch.
and their targets, it is possible that different trophic factors are The development of the fovea in primates is another important
produced by the different targets, and this ensures that only aspect of later retinal development. The fovea is a small, avas-
those ganglion cells survive that have extended their axon to the cular depression that is devoid of all cells except cone photore-
right place.96 ceptors and Müller glia in the central part of the retina of Old
The trophic factors supplied by the targets are likely to be World primates. Foveal development has been extensively char-
among the same molecules identified as survival factors for acterized by Hendrickson and her colleagues.105–108 This region
is initially one of the thickest parts of the retina, and by a process 16. Westenskow P, Piccolo S, Fuhrmann S. Beta-catenin controls differentiation
of the retinal pigment epithelium in the mouse optic cup by regulating Mitf
of cell migration is transformed into a depression or pit. The and Otx2 expression. Development 2009;136:2505–10.
340 GCL begins to thin between the 24th and 26th fetal week in 17. Fuhrmann S, Levine EM, Reh TA. Extraocular mesenchyme patterns the optic
vesicle during early eye development in the embryonic chick. Development
humans, and the number of amacrine and bipolar cells begins to 2000;127:4599–609.
decline soon after. The loss of these cells from the fovea is not 18. Müller F, Rohrer H, Vogel-Hopker A. Bone morphogenetic proteins specify
due to their death, but rather to their migration from the fovea the retinal pigment epithelium in the chick embryo. Development 2007;134:
Section 1

3483–93.
to the surrounding retina. Since these cells already have formed 19. Dakubo GD, et al. Indian hedgehog signaling from endothelial cells is
synaptic connections by this time in development, the synaptic required for sclera and retinal pigment epithelium development in the mouse
eye. Dev Biol 2008;320:242–55.
pedicles of the cones remain in contact with horizontal and 20. Vogel-Hopker A, et al. Multiple functions of fibroblast growth factor-8
bipolar cells as they migrate, which causes a dramatic elongation (FGF-8) in chick eye development. Mechanisms Dev 2000;94:25–36.
21. Pittack C, Grunwald GB, Reh TA. Fibroblast growth factors are necessary for
of the fiber of Henle. At birth, the GCL and INL are only a single neural retina but not pigmented epithelium differentiation in chick embryos.
cell layer thick; after birth, the GCL and INL continue to thin,
Anatomy and Physiology

Development 1997;124:805–16.
Basic Science and Translation to Therapy

22. Cai Z, Feng GS, Zhang X. Temporal requirement of the protein tyrosine
but now the ONL begins to increase in thickness; by age 4, the phosphatase Shp2 in establishing the neuronal fate in early retinal develop-
ONL has six to seven layers of cone photoreceptor nuclei. ment. J Neurosci 2010;30:4110–9.
Although the development of the fovea has been well described, 23. Rapaport DH, Rakic P, LaVail MM. Spatiotemporal gradients of cell genesis
in the primate retina. Perspect Dev neurobiol 1996;3:147–59.
little is known of the mechanisms by which the cells migrate. 24. Carter-Dawson LD, LaVail MM. Rods and cones in the mouse retina. II.
Autoradiographic analysis of cell generation using tritiated thymidine.
J Comp Neurol 1979;188:263–72.
CONCLUSION 25. Rapaport DH, et al. Timing and topography of cell genesis in the rat retina.
J Comp Neurol 2004;474:304–24.
The past 15 years have witnessed an explosive growth in our 26. Turner DL, Snyder EY, Cepko CL. Lineage-independent determination of cell
knowledge of retinal development. The remarkable conserva- type in the embryonic mouse retina. Neuron 1990;4:833–45.
27. Turner DL, Cepko CL. A common progenitor for neurons and glia persists in
tion of molecular mechanisms for eye development throughout rat retina late in development. Nature 1987;328:131–6.
evolution has allowed the use of the powerful molecular genet- 28. Wetts R, Fraser SE. Multipotent precursors can give rise to all major cell types
of the frog retina. Science 1988;239:1142–5.
ics of Drosophila to identify many of the key molecular com­ 29. Holt CE, et al. Cellular determination in the Xenopus retina is independent of
ponents required for retinal differentiation. However, many lineage and birth date. Neuron 1988;1:15–26.
30. Brzezinski J, et al. Ascl1 expression defines a subpopulation of lineage-
aspects of retinal development remain mysterious. A better restricted progenitors in the mammalian retina. Development 2011;138:
understanding, for example, of the mechanisms that control cell 3519–31.
fate will allow better derivation of specific retinal cell types 31. Godinho L, et al. Nonapical symmetric divisions underlie horizontal cell layer
formation in the developing retina in vivo. Neuron 2007;56:597–603.
from human embryonic stem and induced pluripotent stem cells 32. Reh TA, Kljavin IJ. Age of differentiation determines rat retinal germinal cell
for cell replacement therapies and disease modeling. Discovery phenotype: induction of differentiation by dissociation. J Neurosci
1989;9:4179–89.
of the mechanisms that control specific synaptic connections 33. Reh TA. Cellular interactions determine neuronal phenotypes in rodent
will have implications for developmental disorders in the eye retinal cultures. J Neurobiol 1992;23:1067–83.
34. Watanabe T, Raff MC. Diffusible rod-promoting signals in the developing rat
and the rest of the CNS. Elucidation of the processes of eye retina. Development 1992;114:899–906.
development will continue to inform the way we look at retinal 35. Watanabe T, Raff MC. Rod photoreceptor development in vitro: intrinsic
disease and pathology and potentially lead us to methods to properties of proliferating neuroepithelial cells change as development pro-
ceeds in the rat retina. Neuron 1990;4:461–7.
recapitulate developmental processes and provide regenerative 36. Dakubo GD, et al. Retinal ganglion cell-derived Sonic hedgehog signaling
treatments of retinal disease. is required for optic disc and stalk neuroepithelial cell development.
Development 2003;130:2967–80.
37. Brown NL, et al. Math5 encodes a murine basic helix-loop-helix transcription
REFERENCES factor expressed during early stages of retinal neurogenesis. Development
1. O’Rahilly R, Müller F. Neurulation in the normal human embryo. Ciba Foun- 1998;125:4821–33.
dation symposium, 1994;181:70–82; discussion 82–9. 38. Reh TA, Cagan RL. Intrinsic and extrinsic signals in the developing vertebrate
2. Gage PJ, et al. Fate maps of neural crest and mesoderm in the mammalian and fly eyes: viewing vertebrate and invertebrate eyes in the same light.
eye. Invest Ophthalmol Vis Sci 2005;46:4200–8. Perspect Dev Neurobiol 1994;2:183–90.
3. Luhmann UF, et al. Role of the Norrie disease pseudoglioma gene in sprouting 39. Cepko CL, et al. Cell fate determination in the vertebrate retina. Proc Natl
angiogenesis during development of the retinal vasculature. Invest Ophthal- Acad Sci U S A 1996;93:589–95.
mol Vis Sci 2005;46:3372–82. 40. Georgi SA, Reh TA. Dicer is required for the transition from early to late
4. Zuber ME. Eye field specification in Xenopus laevis. Curr Topics Dev Biol progenitor state in the developing mouse retina. J Neurosci 2010;30:
2010;93:29–60. 4048–61.
5. Zuber ME, et al. Specification of the vertebrate eye by a network of eye field 41. Georgi SA, Reh TA. Dicer is required for the maintenance of Notch signaling
transcription factors. Development 2003;130:5155–67. and gliogenic competence during mouse retinal development. Dev Neurobiol
6. Glaser T, et al. PAX6 gene dosage effect in a family with congenital cataracts, 2011;71:1153–69.
aniridia, anophthalmia and central nervous system defects. Nature Genet 42. Marquardt T, et al. Pax6 is required for the multipotent state of retinal pro-
1994;7:463–71. genitor cells. Cell 2001;105:43–55.
7. Quiring R, et al. Homology of the eyeless gene of Drosophila to the Small eye 43. Li S, et al. Foxn4 controls the genesis of amacrine and horizontal cells by
gene in mice and aniridia in humans. Science 1994;265:785–9. retinal progenitors. Neuron 2004;43:795–807.
8. Halder G, Callaerts P, Gehring WJ. Induction of ectopic eyes by targeted 44. Brown NL, et al. Math5 is required for retinal ganglion cell and optic nerve
expression of the eyeless gene in Drosophila. Science 1995;267:1788–92. formation. Development 2001;128:2497–508.
9. Gehring WJ. The master control gene for morphogenesis and evolution of the 45. Koike C, et al. Functional roles of Otx2 transcription factor in postnatal mouse
eye. Genes Cells 1996;1:11–5. retinal development. Mol Cell Biol 2007;27:8318–29.
10. Mathers PH, et al. The Rx homeobox gene is essential for vertebrate eye 46. Nishida A, et al. Otx2 homeobox gene controls retinal photoreceptor cell fate
development. Nature 1997;387:603–7. and pineal gland development. Nat Neurosci 2003;6:1255–63.
11. Pera EM, Kessel M. Patterning of the chick forebrain anlage by the prechordal 47. Omori Y, et al. Analysis of transcriptional regulatory pathways of photorecep-
plate. Development 1997;124:4153–62. tor genes by expression profiling of the Otx2-deficient retina. PloS One
12. Chiang C, et al. Cyclopia and defective axial patterning in mice lacking Sonic 2011;6:e19685.
hedgehog gene function. Nature 1996;383:407–13. 48. Akagi T, et al. Requirement of multiple basic helix-loop-helix genes for retinal
13. Belloni E, et al. Identification of Sonic hedgehog as a candidate gene respon- neuronal subtype specification. J Biol Chem 2004;279:28492–8.
sible for holoprosencephaly. Nature Genet 1996;14:353–6. 49. Mears AJ, et al. Nrl is required for rod photoreceptor development. Nature
14. Roessler E, et al. Mutations in the human Sonic hedgehog gene cause holo- Genet 2001;29:447–52.
prosencephaly. Nature Genet 1996;14:357–60. 50. Burmeister M, et al. Ocular retardation mouse caused by Chx10 homeobox
15. Fuhrmann S. Eye morphogenesis and patterning of the optic vesicle. null allele: impaired retinal progenitor proliferation and bipolar cell differen-
Curr Topics Dev Biol 2010;93:61–84. tiation. Nature Genet 1996;12:376–84.
51. Brzezinski JAT, Lamba DA, Reh TA. Blimp1 controls photoreceptor versus 82. Furukawa T, Morrow EM, Cepko CL. Crx, a novel Otx-like homeobox gene,
bipolar cell fate choice during retinal development. Development 2010;137: shows photoreceptor-specific expression and regulates photoreceptor differ-
619–29. entiation. Cell 1997;91:531–41.
52. Jasoni CL, et al. A chicken achaete-scute homolog (CASH-1) is expressed in a 83. Roberts MR, et al. Retinoid X receptor (gamma) is necessary to establish the 341
temporally and spatially discrete manner in the developing nervous system. S-opsin gradient in cone photoreceptors of the developing mouse retina.
Development 1994;120:769–83. Invest Ophthalmol Vis Sci 2005;46:2897–904.
53. Tomita K, et al. Mash1 promotes neuronal differentiation in the retina. 84. Roberts MR, et al. Making the gradient: thyroid hormone regulates cone opsin

Chapter 13
Genes Cells 1996;1:765–74. expression in the developing mouse retina. Proc Natl Acad Sci U S A 2006;
54. Nelson BR, et al. Acheate-scute like 1 (Ascl1) is required for normal delta-like 103:6218–23.
(Dll) gene expression and notch signaling during retinal development. 85. Ng L, et al. A thyroid hormone receptor that is required for the development
Dev Dynamics 2009;238:2163–78. of green cone photoreceptors. Nature Genet 2001;27:94–8.
55. Ashwell KW, et al. The appearance and distribution of microglia in the devel- 86. Levine EM, Fuhrmann S, Reh TA. Soluble factors and the development of rod
oping retina of the rat. Vis Neurosci 1989;2:437–48. photoreceptors. Cell Mol Life Sci 2000;57:224–34.
56. Schnitzer J, Scherer J. Microglial cell responses in the rabbit retina following 87. Lamba DA, Gust J, Reh TA. Transplantation of human embryonic stem cell-
transection of the optic nerve. J Comp Neurol 1990;302:779–91. derived photoreceptors restores some visual function in Crx-deficient mice.
57. Schnitzer J. The development of astrocytes and blood vessels in the postnatal Cell Stem Cell 2009;4:73–9.
rabbit retina. J Neurocytol 1988;17:433–49. 88. Lamba DA, Karl MO, Reh TA. Strategies for retinal repair: cell replacement

The Development of the Retina


58. Watanabe T, Raff MC. Retinal astrocytes are immigrants from the optic nerve. and regeneration. Prog Brain Res 2009;175:23–31.
Nature 1988;332:834–7. 89. Lamba DA, et al. Efficient generation of retinal progenitor cells from human
59. Hinds JW, Hinds PL. Early ganglion cell differentiation in the mouse retina: embryonic stem cells. Proc Natl Acad Sci U S A 2006;103:12769–74.
an electron microscopic analysis utilizing serial sections. Dev Biol 1974;37: 90. Lamba DA, et al. Generation, purification and transplantation of photorecep-
381–416. tors derived from human induced pluripotent stem cells. PloS ONE 2010;5:
60. Snow RL, Robson JA. Ganglion cell neurogenesis, migration and early dif- e8763.
ferentiation in the chick retina. Neuroscience 1994;58:399–409. 91. Haverkamp S, et al. Synaptic plasticity in CNGA3(–/–) mice: cone bipolar
61. Kirby MA, Steineke TC. Morphogenesis of retinal ganglion cells: a model of cells react on the missing cone input and form ectopic synapses with rods.
dendritic, mosaic, and foveal development. Perspect Dev Neurobiol 1996;3: J Neurosci 2006;26:5248–55.
177–94. 92. Mansergh F, et al. Mutation of the calcium channel gene Cacna1f disrupts
62. Kirby MA, Steineke TC. Early dendritic outgrowth of primate retinal ganglion calcium signaling, synaptic transmission and cellular organization in mouse
cells. Vis Neurosci 1991;7:513–30. retina. Hum Mol Genet 2005;14:3035–46.
63. Masai I, et al. N-cadherin mediates retinal lamination, maintenance of fore- 93. Dick O, et al. The presynaptic active zone protein bassoon is essential for
brain compartments and patterning of retinal neurites. Development photoreceptor ribbon synapse formation in the retina. Neuron 2003;37:
2003;130:2479–94. 775–86.
64. Rakic P. Neuronal migration and contact guidance in the primate telencepha- 94. Wong RO, Hughes A. Role of cell death in the topogenesis of neuronal distri-
lon. Postgrad Med J 1978;54(Suppl 1):25–40. butions in the developing cat retinal ganglion cell layer. J Comp Neurol
65. Kim IJ, et al. Laminar restriction of retinal ganglion cell dendrites and axons: 1987;262:496–511.
subtype-specific developmental patterns revealed with transgenic markers. 95. Valenciano AI, Boya P, de la Rosa EJ. Early neural cell death: numbers and
J Neurosci 2010;30:1452–62. cues from the developing neuroretina. Int J Dev Biol 2009;53:1515–28.
66. Kljavin IJ, et al. Cell adhesion molecules regulating neurite growth from 96. Shen S, et al. Retinal ganglion cells lose trophic responsiveness after axotomy.
amacrine and rod photoreceptor cells. J Neurosci 1994;14:5035–49. Neuron 1999;23:285–95.
67. Yamagata M, Sanes JR. Dscam and Sidekick proteins direct lamina-specific 97. Frade JM, et al. Control of early cell death by BDNF in the chick retina.
synaptic connections in vertebrate retina. Nature 2008;451:465–9. Development 1997;124:3313–20.
68. Matsuoka RL, et al. Transmembrane semaphorin signalling controls laminar 98. Farah MH. Neurogenesis and cell death in the ganglion cell layer of vertebrate
stratification in the mammalian retina. Nature 2011;470:259–63. retina. Brain Res Rev 2006;52:264–74.
69. Dacey DM. The mosaic of midget ganglion cells in the human retina. J Neuro­ 99. Isenmann S, Kretz A, Cellerino A. Molecular determinants of retinal ganglion
sci 1993;13:5334–55. cell development, survival, and regeneration. Prog Retinal Eye Res 2003;22:
70. Perry VH, Linden R. Evidence for dendritic competition in the developing 483–543.
retina. Nature 1982;297:683–5. 100. Pollock GS, et al. TrkB receptor signaling regulates developmental death
71. Fuerst PG, et al. DSCAM and DSCAML1 function in self-avoidance in dynamics, but not final number, of retinal ganglion cells. J Neurosci
multiple cell types in the developing mouse retina. Neuron 2009;64: 2003;23:10137–45.
484–97. 101. Mao CA, et al. Eomesodermin, a target gene of Pou4f2, is required for retinal
72. Wong RO, et al. Early functional neural networks in the developing retina. ganglion cell and optic nerve development in the mouse. Development
Nature 1995;374:716–8. 2008;135:271–80.
73. Hendrickson AE. Synaptic development in macaque monkey retina and its 102. Wang SW, et al. Abnormal polarization and axon outgrowth in retinal
implications for other developmental sequences. Perspect Dev Neurobiol ganglion cells lacking the POU-domain transcription factor Brn-3b. Mol Cell
1996;3:195–201. Neurosci 2000;16:141–56.
74. Bleckert A, Wong RO. Identifying roles for neurotransmission in circuit 103. Xiang M, et al. Targeted deletion of the mouse POU domain gene Brn-3a
assembly: insights gained from multiple model systems and experimental causes selective loss of neurons in the brainstem and trigeminal ganglion,
approaches. BioEssays 2011;33:61–72. uncoordinated limb movement, and impaired suckling. Proc Natl Acad Sci
75. Yoshimatsu T, Suzuki S, Wong ROL. Synapse formation in the developing U S A 1996;93:11950–5.
retina. In: Rakic P, Rubenstein JL, editors. Comprehensive developmental 104. Weishaupt JH, Klocker N, Bahr M. Axotomy-induced early down-regulation
neuroscience. Oxford: Elsevier; 2011. of POU-IV class transcription factors Brn-3a and Brn-3b in retinal ganglion
76. Kerschensteiner D, et al. Neurotransmission selectively regulates synapse cells. J Mol Neurosci 2005;26:17–25.
formation in parallel circuits in vivo. Nature 2009;460:1016–20. 105. Hendrickson A. A morphological comparison of foveal development in man
77. Bumsted K, et al. Spatial and temporal expression of cone opsins during and monkey. Eye 1992;6:136–44.
monkey retinal development. J Comp Neurol 1997;378:117–34. 106. Hendrickson A, Kupfer C. The histogenesis of the fovea in the macaque
78. Hendrickson A, et al. Rod photoreceptor differentiation in fetal and infant monkey. Invest Ophthalmol Vis Sci 1976;15:746–56.
human retina. Exp Eye Res 2008;87:415–26. 107. Hendrickson AE. Primate foveal development: a microcosm of current
79. Packer O, Hendrickson AE, Curcio CA. Development redistribution of pho- questions in neurobiology. Invest Ophthalmol Vis Sci 1994;35:3129–33.
toreceptors across the Macaca nemestrina (pigtail macaque) retina. J Comp 108. Hendrickson AE, Yuodelis C. The morphological development of the human
Neurol 1990;298:472–93. fovea. Ophthalmology, 1984;91:603–12.
80. Cornish EE, et al. The role of opsin expression and apoptosis in determination
of cone types in human retina. Exp Eye Res 2004;78:1143–54.
81. Cornish EE, Hendrickson AE, Provis JM. Distribution of short-wavelength-
sensitive cones in human fetal and postnatal retina: early development of
spatial order and density profiles. Vis Res 2004;44:2019–26.
Section 1 Anatomy and Physiology

Chapter Structure and Function of Rod


14  and Cone Photoreceptors
Jeannie Chen, Alapakkam P. Sampath

INTRODUCTION and animal models with analogous mutation(s) and/or disease


phenotype.
Our visual experience is initiated by rod and cone photorecep- Only recently has it become possible to incorporate the
tors in the retina. The human eye contains over 100 million rods insights from numerous studies into biochemical pathways and
and about 6 million cones, which are located within the outer mechanisms of photoreceptor function. Here, we review the
nuclear layer of the retina and allow our visual experience to principal proteins and pathways involved in rod photoreceptor
extend over 12 orders of magnitude in light intensity by splitting structure and function, and highlight the parallelisms in cone
this range. Rod photoreceptors mediate vision under conditions photoreceptors.
of dim illumination, and allow our visual system to reach the
limit imposed by the absorption of single photons.1,2 Cones are
less sensitive by ~100-fold, but their tremendous capacity for
PHOTORECEPTOR FUNDAMENTALS
adaptation allows them to encode light intensities in the bright- The rod and cone photoreceptors are specialized sensory neurons
est of days.3,4 Over the last decade, our understanding of struc- that contain the protein machinery necessary to convert incident
ture and function of these cells has increased dramatically. Over light into a signal that can be interpreted by the nervous system.
150 genes have been cloned or linked to retinal diseases, and, Rod photoreceptors are more numerous than cones in most
surprisingly, as many as half of these genes are specifically mammalian retina, and are highly sensitive. In the fully dark-
expressed or highly enriched in the photoreceptor cells. Discov- adapted state, rods can reliably report the absorption of single
ery of the molecular constituents of the rods and cones is pro- photons to the retinal output, and they permit our scotopic, or
gressing at an increasing rate, particularly enhanced by the night, vision. Cone photoreceptors are morphologically and
availability of “complete” genomic sequences for both human functionally distinct from rods and express several types of
and mouse. While photoreceptor genes can be identified through visual pigments, or opsins, whose spectral sensitivity varies
association with a retinal disease (linkage), information about based on the cone’s subtype. In humans, three classes of cones
their function does not accompany their identification.5 Basic confer robust color vision: S, M, and L cones (see Chapter 10,
science research must then be undertaken to explain their role Color vision and night vision). Cones are also less sensitive than
in both the normal, healthy photoreceptor as well as in photo­ rods and generate light responses that are temporally briefer.
receptor diseases. This allows cones to mediate our photopic, or day, vision with
Though much of what we know about the structure and func- improved temporal resolution. The concerted action of these two
tion of photoreceptor cells has come from studying animal types of photoreceptors, and the retinal circuitry that carries
models of inherited blindness,6,7 more recently, transgenic and their signals to the retinal output, ultimately underlie our rich
knockout animal technologies have established themselves as visual experience.
powerful tools for understanding function and studying disease. Both rod and cone photoreceptors are highly polarized elon-
After new photoreceptor genes are identified from patients, gated cells that can be described as having four subcellular com-
mutant animals can be engineered to emulate human photo­ partments: the outer segment (OS), the inner segment (IS), the
receptor pathologies. Before these molecular technologies were nucleus, and the synaptic terminal (Figs 14.1 and 14.2). The OS is
available, most data about retinal disease were gleaned from the where photons are captured and activation of the phototransduc-
rare “informative” patient or donor retina that had surviving tion cascade begins. The IS lies immediately proximal to the OS,
photoreceptors to examine.8 Other major sources of information and contains the cell’s protein synthesis (Golgi apparatus and
were studies of animal models that occurred through inbreeding endoplasmic reticulum) and metabolic (mitochondria) machin-
or random inheritance of mutations in photoreceptor genes. ery. Light-evoked signals are relayed passively down the photo-
Examples of these are the Irish Setter dog,9 the Briard dog,10 the receptor axon (up to 75 µm long) to the synaptic terminals in the
Abyssinian cat,11 the Royal College of Surgeons (RCS) rat,12 and outer plexiform layer. The structure of photoreceptor terminals is
the rd13,14 and rds15,16 mouse models. Now, the ability to engineer unique in the nervous system, as they contain a specialized struc-
transgenic animals has made the search for “informative” ture called a ribbon that facilitates the release of the excitatory
patients and naturally occurring animal models less acute, since neurotransmitter glutamate on to second-order retinal neurons
any single gene of interest can be introduced17–19 or removed20–24 (bipolar and horizontal cells). Thus, the photoreceptor cells trans-
from the photoreceptor. However, the most instructive examples duce the sensory stimulus, light, and pass on a signal to retinal
of structure–function relationships occur when there are patients circuits that carry this information to higher visual centers.
343

Chapter 14
Structure and Function of Rod and Cone Photoreceptors
A B C D

E F

Fig. 14.1 (A) Scanning electron micrograph of rods and cones from the African clawed frog, Xenopus laevis. The basic shape of the rod
photoreceptor inner (IS) and outer segments (OS) can be appreciated by this surface view. The cones in this species are exceptionally short and
cone-shaped, unlike the cylindrical appearance of cone OS in the mammalian retina. (B) Immunolocalization of opsin in the IS of the frog retina.
Arrows show location of newly synthesized opsin in the Golgi apparatus (solid arrows) and a track of labeled vesicles (open arrow) from the myoid
to the cilium. (C) Transmission electron micrograph of photoreceptors from the mouse retina. Note the connecting cilium between the IS and OS
and that a large number of mitochondria are visible in this orientation. (D) High-magnification view of frog photoreceptor OS as viewed from the top
of the disc stack. Upper left shows a single disc labeled with an antirod opsin antibody. The plasma membrane surrounding the disc can be
observed in this section. Lower left is a cone photoreceptor OS that is not labeled by the antibody. (E) High-magnification view of frog photoreceptor
OS as viewed from the side of the disc stack. Discs are labeled with an anti-rod opsin antibody. The rim region of the disc can be observed, as well
as the close packing of the individual discs within the OS. (F) Synaptic terminal of the rod photoreceptor. Directly below the nucleus is the
photoreceptor spherule. Synaptic ribbons are visible in this section. (D and E courtesy of Robert Molday, University of British Columbia.)

PHOTORECEPTOR diameter and length that ranges from 25 to 45 µm,25–28 that


depends on numerous factors, including the time of day, the
OUTER-SEGMENT STRUCTURE light intensity, their location in the retina, and the animal species.
The OS compartment contains all components necessary for The cone OS is shorter, typically half the length of rods, with a
phototransduction, which is a set of biochemical reactions that larger diameter at the base that gradually tapers towards the tip
convert photon capture to a change in a cationic current at the (Fig. 14.2). Mouse rod OS contains an average of 810 membra-
plasma membrane. Rods have cylindrical OS about 1.3 µm in nous discs, which occupy two-thirds of the volume of the OS.29
344
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

Model 1

OS

CC

BB

M
Model 2

ER

Rod Cone

Fig. 14.2 The fundamental organization of vertebrate rod and cone photoreceptors. Parts of the cell include: OS, outer segment; CC, connecting
cilium; A, axoneme; BB, basal body; M, mitochondria; G, Golgi apparatus; ER, endoplasmic reticulum; N, nucleus; and S, synaptic terminal. Insets
depict the structure of the axoneme at the level of the outer segment where microtubule structure is 9 × 1 + 0 (top), and where the inner and outer
segment join the microtubule structure is 9 × 2 + 0 (bottom). Middle panels depict two models for outer-segment disc formation. In model 1,
outer-segment discs originate from evaginations of the plasma membrane but become closed off and separate from the plasma membrane. In
model 2, newly generated outer-segment discs are already closed and separate from the plasma membrane. (Modified with permission from
Anderson DH, Fisher SK, Steinberg RH. Mammalian cones: disc shedding, phagocytosis, and renewal. Invest Ophthalmol Vis Sci 1978;17:117–33;
Steinberg RH, Fisher SK, Anderson DH. Disc morphogenesis in vertebrate photoreceptors. J Comp Neurol 1980;190:501–8; and Chuang JZ, Zhao
Y, Sung CH. SARA-regulated vesicular targeting underlies formation of the light-sensing organelle in mammalian rods. Cell 2007;130:535–47.)

The majority of the protein on these discs is rod opsin (or rho- force microscopy.32 Whether it exists as dimers in vivo is still an
dopsin when the opsin is bound to its chromophore, 11-cis area of controversy. However, it is known that rhodopsin is
retinal). The density of rhodopsin on the disc membrane has functional as monomers.33
been measured to be about 24 000 molecules/µm2.30 Despite this Other integral membrane proteins of the phototransduction
dense packing, rhodopsin freely diffuses in the membrane,31 pathway that are embedded in the discs are two isoforms of the
which facilitates its encounter with, and activation of, transducin guanylyl cyclases, RetGC1 and RetGC2, which are single-pass
molecules to amplify the light signal. Many G-protein-coupled transmembrane proteins. Other transduction proteins, such as
receptors are known to form dimers and higher-order oligomers. transducin, phosphodiesterase 6 (PDE6), recoverin, and guan­
Rhodopsin has been observed to form a paracrystalline array of ylyl cyclase-activating proteins (GCAPs) are peripheral mem-
dimers when native disc membrane is viewed under atomic brane proteins (see below).
ABCR are responsible for a large variety of retinal degenera-
Proteins that stabilize the structure of tions, including Stargardt macular dystrophy, fundus flavi-
outer-segment discs maculatus, some forms of cone–rod degeneration, and retinitis 345
Rhodopsin is not only integral to the phototransduction cascade, pigmentosa (RP). Other ABCR mutations are thought to increase
it is also required for the formation and maintenance of OS discs; the risk of developing age-related macular degeneration

Chapter 14
in the absence of rhodopsin, the OS structure is not formed.22 (AMD). ABCR is a member of the ABC superfamily. This is
Peripherin and Rom-115,34,35 are two other disc membrane com- a large family of transmembrane proteins involved in energy-
ponents that contribute to maintaining the flattened structure of dependent transport of many different substrates across mem-
the disc. Both of these proteins belong to the tetraspanin family brane “barriers.” The localization of ABCR at the disc rim
of integral membrane proteins that form large multiprotein com- suggests that it is involved in the movement of molecules from
plexes known as the tetraspanin web or tetraspanin-enriched the disc lumen into the cytosol. This hypothesis was supported
microdomains.36 Peripherin/rds is normally found within the by studies in ABCR knockout mice, which show delayed dark

Structure and Function of Rod and Cone Photoreceptors


edge or “rim” of the disc. It self-associates to form higher-order adaptation, increased all-trans retinal following light exposure,
complexes and also interacts with Rom-1.37 Peripherin/rds and and elevated phosphatidylethanolamine (PE) in the rod OS.48
Rom-1 likely function as adhesion molecules that assist in Biochemical analysis of retinas from ABCR knockout mice
keeping the discs vertically aligned, and may stabilize the disc revealed accumulation of a novel complex of all-trans retinal
stack with bridges to the overlying plasma membrane. and PE, termed N-retinylidene-PE, which is not found in normal
Peripherin/rds is also thought to function in forming the curva- retina. Once in the cytosol, the all-trans retinol moves to the
ture of the disc rim.38 retinal pigment epithelium (RPE) for recycling and chromo-
Deletion or disruption of peripherin/rds results in malforma- phore (11-cis retinal) regeneration (see Chapter 16, Cell biology
tion of discs at the OS base. The naturally occurring retinal of the retinal pigment epithelium). The primary pathologic
degeneration in the mouse named “retinal degeneration, slow” defect in Stargardt disease, and also in ABCR knockout mice,
(rds) has been shown to be due to a defect in peripherin/rds. is accumulation of toxic lipofuscin pigments, such as A2E, in
The absence of peripherin/rds in the rds/rds mouse prevents RPE cells. Thus the phenotype is remarkably similar between
normal development of the photoreceptor OS and leads to pho- mice and that observed in the fundus of Stargardt patients.
toreceptor cell death.15,39 Transgenic mice that lack peripherin/ The animal model and further biochemical investigations49
rds fail to form an OS. When the level is reduced, as in the het- suggest that ABCR functions as an outwardly directed flippase
erozygous mice, large whorls of disc membrane are formed for N-retinylidene-PE. Delayed dark adaptation is likely due to
instead of an organized stack of uniform discs.15,39 Mutations that accumulation (in discs) of the noncovalent complex between
affect the quaternary structure of peripherin/rds also lead to opsin and all-trans retinal. ABCR-mediated retinal degeneration
malformed discs. These observations underscore the importance in patients may result from “poisoning” of the RPE due to A2E
of peripherin/rds in the formation and maintenance of disc accumulation, with secondary photoreceptor degeneration due
structure in the OS. to loss of the ABCR support role.
Although Rom-1 is analogous to peripherin in structure and Though ABCR most definitely plays a role in the structure of
function, its absence in rods shows a less severe phenotype than discs, no mutations have been conclusively associated with
peripherin/rds knockout mice, suggesting that peripherin/rds abnormal structural features per se. In other words, it appears
is more critical for disc formation. Although the discs grow that mutations do not directly affect folding at the disc rim, and
larger than normal sizes, photoreceptor function appears to be more likely affect the biochemical function of the transporter. In
retained to a much higher level than when peripherin/rds is corroboration of these data, Radu and colleagues tested the
absent. Nevertheless, abnormal Rom-1 does lead to slow and effects of isotretinoin on lipofuscin accumulation in ABCR
progressive photoreceptor degeneration both in mice and in knockout mice,50 a model of recessive Stargardt disease. They
humans.40 observed by electron microscopy that isotretinoin blocked both
Functional differences of peripherin/rds and Rom-1 have the formation of A2E biochemically, and the accumulation of
been noted in rods and cones. Cones appear to have a lower ratio lipofuscin pigments. Further, no significant visual loss was
of Rom-1/peripherin,36 and different peripherin/rds mutations observed in ABCR null mice by electroretinography (when
appear to affect rods and cones differentially. For example, the treated), and isotretinoin also blocked the slower, age-dependent
C214S and N244K mutations have a greater effect on rods accumulation of lipofuscin in wild-type mice. The results suggest
whereas R172W and N244H tend to cause cone-dominant dis- that treatment with isotretinoin may inhibit lipofuscin accumu-
eases such as macular dystrophy.36 lation and delay the onset of visual loss in patients with Star-
Transmission electron microscopy has revealed structural ele- gardt disease and may be an effective treatment for other forms
ments between the lamellar discs as well as between the discs of retinal or macular degeneration associated with lipofuscin
and the plasma membrane that are mainly localized to the rim accumulation, though “normal” visual function may be some-
region and incisures of discs.41–44 A study of OS structure using what compromised by such treatment.
cryoelectron tomography of vitrified OS showed previously The presence of an OS is clearly not optional, as there are many
unobserved spacers distributed throughout the discs.30 These examples of retinal disease in which the photoreceptor cell dies
structural elements likely maintain the proper distance between shortly after loss of the OS.51–53 Why loss of the OS triggers rod
adjacent discs and between discs and the plasma membrane. The cell death is not obvious, since all of the required cellular organ-
protein identities of these spacers are not known, but peripherin/ elles inhabit the IS and cell body; the OS appears devoid of
rds and Rom-1 may be candidates. organelles. The necessity to maintain an OS is further surprising
Another component of the rod disc is ABCR (photoreceptor since photoreceptors continually replace all the OS components.
cell-specific ATP-binding cassette transporter).45–47 Mutations in In fact, the renewal of rod cell OS components is perhaps more
readily seen in the photoreceptor than in any other cell in the specialized proteins, many of which regulate the movement of
body. The disc and plasma membrane components of the OS are ions into and out of the OS. The best-characterized proteins of
346 completely replaced within 2 weeks.54,55 To support this turnover this type are the retinal cGMP-gated (CNG) cation channels. In
there is an influx of newly synthesized proteins and lipids from the dark-adapted rods and cones, Na+ and Ca2+ flow into the OS
the IS. These components are transported to the base of the OS, through these channels in the plasma membrane. Calcium com-
and from there discs gradually migrate towards the RPE for prises about 10% of the dark current carried by these channels
Section 1

phagocytosis and recycling. One idea is that the high oxygen in rods,60 and perhaps 20% or more in cones.61,62 In both rods and
tension coming from the choroidal vasculature may interfere cones Na+ is extruded from the IS through the Na+/K+ pump.
with the cellular machinery in the IS and nucleus of the photo- This flow of ions sets up the circulating dark current, of which
receptor, and the presence of the OS puts a protective distance the vast majority is carried by the Na+. The probability of the
between the choroid and the photoreceptor nuclei.56 opening of the CNG channel, which in turn determines the size
of the circulating current, depends on the amount of free [cGMP],
Anatomy and Physiology
Basic Science and Translation to Therapy

Disc morphogenesis which in the dark is estimated to be 3–4 µM.63 At this concentra-
Although it is known that new discs are formed at the base of tion, the probability of channel opening is estimated to be only
the OS, the process by which these discs are formed is not fully 0.1–0.2.64,65 This underscores the impact that elevated [cGMP] can
resolved (Fig. 14.2). One model was described in the work of have on the number of open channels in the diseased state. The
Anderson et al. (model 1).57,58 Based on morphologic studies of influx of Ca2+ through the channel is balanced by an efflux of
adult monkey rods, it was observed that the folded membrane Ca2+ by the Na+/Ca2+-K+ exchanger (NCKX) in the rod OS plasma
stacks at the base of the OS are continuous with the plasma membrane, thereby maintaining the intracellular level of Ca2+ at
membrane of the connecting cilium, whereas the older discs are a relatively constant level.66 Kaupp and colleagues67 cloned the
closed off and separated from the plasma membrane. From this cGMP channel from bovine retina, while Pittler and colleagues68
appearance a model was proposed that the newly arrived determined the primary structures of the human and mouse
rhodopsin-bearing vesicles fuse with the plasma membrane, and retinal rod cGMP-gated cation channel. These studies found that
the growing membrane evaginates to form open discs. Eventu- the sequence of the cGMP channel has significant similarity
ally these discs pinch off and form the mature closed discs. (59%) to the olfactory cAMP-gated channel. The retinal rod CNG
Another model (model 2), the vesicular targeting model, was channel is a hetero-oligomer composed of three alpha (CNGA1)
recently proposed by Chuang and colleagues.59 They provide and one beta (CNGB1) subunits,69,70 each with cytoplasmic amino
molecular evidence that rhodopsin’s carboxyl terminus interacts (N)- and carboxyl (C)-termini, six putative transmembrane
with a protein called SARA (smad anchor for receptor activa- domains, and a pore region.71,72 Mutations in CNGA1 and
tion), as well as phosphatidylinositol 3-phosphate (PI3P), and CNGB1 have been linked to disease. A point mutation in
syntaxin 3. In their model, the new discs are already closed and CNGB173 and several mutations in CNGA174 have been identi-
enveloped by the OS plasma membrane. Their evidence suggests fied and linked to recessive RP in humans. The CNG channel in
that the protein–protein and protein–lipid interactions orga- cones consists of two CNGA3 and two CNGB3 subunits.75
nized by SARA regulate the vesicular targeting of axonemal Together, mutations in either of these genes account for ~75% of
rhodopsin-bearing vesicles to the newly formed discs. Thus the complete acromatopsia.76,77
discs grow by fusing with the rhodopsin-bearing vesicles and In addition to the CNG channel, there are several other chan-
not from the evaginated plasma membrane. This model is more nels and transporters in the plasma membrane that serve to
consistent with the morphology of their experimental model regulate the intracellular contents of the OS. The best studied of
system, the mouse rods, which do not display open discs at the these is the NCKX. Rods express NCKX1 whereas cones express
base of the OS. NCKX2.78 The exchanger moves with every cycle 4 Na+ into the
The degree of translational and posttranslational control in the OS, and in exchange, moves one Ca2+ and one K+ into the sub-
processes of disc formation remains an open question. The pro- retinal space; exchange is thus electrogenic. Other transmem-
cesses necessary to manage and partition the correct ratio of brane proteins, such as the GLUT-1 glucose transporter, have
rhodopsin to the other proteins of the phototransduction cascade been shown to be present on both rod and cone OS.79
(transducin: PDE: GC: GCAP: cGMP-gated channel) in the discs
are unknown. Additionally, the insertion of the structural pro- Outer-segment lipids
teins peripherin, Rom-1, and ABCR into the forming disc rim is Docosahexaenoic acid (DHA, 22:6n-6) is the major fatty acid
likely to require precise instruction. How these processes found in the retinal rod OS, and rod photoreceptors have higher
combine to generate a functional OS will require extensive new levels of DHA than in any other membrane system examined.80
investigation and methodology. It is unlikely that these complex Numerous studies have established that the high level of DHA
morphogenic processes will be elucidated using static two- in rod OS membranes provides an optimal microenvironment
dimensional microscopy methods with fixed materials, the for rhodopsin. DHA belongs to the n-3 family of essential poly-
method that has yielded most information to date. These prob- unsaturated fatty acids. These fatty acids cannot be synthesized
lems may require the application of three-dimensional micros- by vertebrates and they, or their shorter-chain precursors, must
copy, in conjunction with immunocytochemical tags and therefore be obtained in the diet.81
fluorescent labels such as green fluorescent protein, to follow the Humans with RP and dogs with progressive rod–cone degen-
assembling proteins in preparations of living photoreceptors. eration (prcd) have lower than normal blood levels of long-chain
polyunsaturated fatty acids, including DHA. In addition, prcd-
Outer-segment plasma membrane affected dogs have lower levels of DHA in their rod OS than
It is clear that, in addition to its obvious role in phototransduc- control animals.82 The reason for the reduced level of DHA in
tion, the OS plasma membrane contains a rich array of rod OS of animals with inherited retinal degeneration is not
known. However, the fatty acid composition of the rod OS of Signal activation and amplification
these animals suggests that the synthesis of DHA-containing
A hallmark of rod phototransduction is its high sensitivity.
glycerolipids is downregulated in retinas of animals with inher-
Fully dark-adapted rods achieve sensitivity that reaches the
347
ited retinal degenerations.
theoretical maximum, the ability to detect individual quanta of
PHOTOTRANSDUCTION light.1,2 This high sensitivity is generated through several stages

Chapter 14
of amplification, resulting in a tremendous increase in the
Visual transduction is initiated by the phototransduction signal gain (Fig. 14.4). This cascade of amplification begins at
cascade, which is perhaps the best characterized of all G-protein the light-activated G-protein-coupled receptor rhodopsin (R).
receptor-coupled signaling pathways. Our knowledge about Absorption of a photon by R leads to a conformation change
phototransduction is relatively advanced because the compo- (R*), allowing it to interact with the heterotrimeric G-protein
nents of this pathway are located selectively in the photoreceptor transducin (Tα, β, and γ), thereby promoting the exchange of

Structure and Function of Rod and Cone Photoreceptors


OS and can be isolated in quantities suitable for biochemical bound GDP for GTP. The complex then dissociates into Tα-GTP
analysis. Photoreceptors can also be isolated easily for electro- and Tβγ, and R* is free to activate many other transducin mol-
physiological measurements, either as single cells using suction ecules during its catalytic lifetime.83 In the subsequent step,
electrodes (Fig. 14.3), or en masse using field potentials called Tα-GTP binds and removes the inhibitory γ-subunit of the
electoretinograms (ERG: see Chapter 7, Electrogenesis of the cGMP-PDE so that it can now hydrolyze cGMP to 5’-GMP; the
electroretinogram). The phototransduction cascade within rods enzymatic activity of PDE (PDE*) in turn degrades thousands
is so robust that single-photon absorption by rhodopsin gives of cGMP molecules. The decrease in cytoplasmic cGMP con-
rise to a change in current that can be monitored by suction centration then leads to closure of the cGMP-gated cation con-
electrode recordings. Similarly, the remarkable ability of cone ductance channel in the rod plasma membrane, resulting in a
phototransduction to adapt to increases in background light reduction in the influx of ~ 1 000 000 Na+ ions.84 This is a graded
intensity can also be monitored with suction electrodes. The effect: a slight lowering in the cGMP concentration leads to
combination of transgenic technology and electrophysiological closure of some channels, while a large decrease will eventually
analysis has greatly enhanced our understanding of signal trans- lead to closure of all channels. In this instance, the rod cell is
duction within these photoreceptor cells. In particular, trans- said to be saturated.
genic technologies have allowed the introduction of targeted In darkness, glutamate, the transmitter used by both rods and
changes into specific components of the signaling pathway. cones, is released at a steady rate because the photoreceptor’s
These genetically altered intact photoreceptor cells can then membrane potential is in a relatively depolarized state. The
be subjected to electrophysiological measurements, and their closure of CNG channels results in a graded hyperpolarization
tissues can subsequently be used in biochemical or immuno­ of the cell, leading to a decrease in glutamate release at the
cytochemical assays to pinpoint the molecular mechanism synapse. In this manner, the first sensation of light perception is
behind the physiological phenotype. In this section we transmitted from the photoreceptor cell to second-order cells of
summarize the current state of our understanding of photo­ the retina, where signals are further processed and ultimately
transduction in retinal photoreceptors. conveyed to the retinal ganglion cells (see Chapter 15, Functional
anatomy of the mammalian retina).

Signal deactivation
A reversal of the activation steps is required ultimately for the
photoreceptor to return to its resting state. First, the catalytically
active components of the phototransduction cascade, R* and
PDE*, must be quenched, then cGMP needs to be resynthesized
(Fig. 14.4). These events must be rapid and reproducible for the
photoreceptor to maintain its high sensitivity and respond to
subsequent stimuli. Our current understanding of these pro-
cesses is detailed below.
Quenching R*: phosphorylation and arrestin binding
Since the early 1980s, phosphorylation was recognized to play
an important role in deactivation of R*.85,86 To determine how
receptor deactivation occurs in vivo, transgenic mouse models
were developed to assess the contribution of receptor phosphor-
ylation to R* shutoff. In early experiments, a rhodopsin trunca-
tion mutation, S334ter, was expressed in the photoreceptors of
transgenic mice17 that removed the terminal 15 amino acid resi-
dues and thus all putative Ser and Thr phosphorylation sites.
Fig. 14.3 Suction electrode used in recording the outer-segment Single-photon responses produced by S334ter R* failed to shut
current. The cells in this clump of toad retina are being superfused off in a timely and stereotyped manner, indicating that this
with bicarbonate-buffered Ringer solution equilibrated with 5% carbon domain is important for R* quenching. In the 1990s, numerous
dioxide. A rod outer segment is carefully drawn into the glass
biochemical experiments suggested that specific Ser residues on
electrode, which makes a high-resistance seal against the cell.
Responses are recorded during the presentation of a stimulus, in this the C-terminus are crucial in R* shutoff.87–89 However, single-cell
case a slit of focused green light. (Courtesy of D. Baylor.) recordings from transgenic mice rods expressing rhodopsin
Intradiscal Activation cGMP
Open
348 cGMP
Na+
hv Rhodopsin R* Phosphodiesterase
cGMP Ca2+
cGMP
Section 1

γ Tα γ Tα Tα Tα Tα
β β β
α
γ γ
GDP GTP cGMP
Closed
Anatomy and Physiology
Basic Science and Translation to Therapy

GTP
GDP
Cytosolic GMP cGMP
A

Intradiscal R* Deactivation Closed

R*

cGMP

RK RK
P P
RV P P
P P
Ca2+ Ca2+ Arr cGMP
Closed
RV

Cytosolic Arr
B

Intradiscal PDE* Deactivation and cGMP Synthesis Open

Guanylyl
cyclase
cGMP
GCAPs
cGMP
Tα γ Tα γ β γ cGMP
Tα Tα Na+
β + α GCAPs
α β GTP Ca2+
cGMP
γ γ
cGMP Open

C Cytosolic cGMP

Fig.14.4 (A) The phototransduction cascade in the rod and cone outer segment is initiated when a photon (hν) strikes the visual pigment,
rhodopsin in rods, and promotes it to an active state (R*). R* interacts with a heterotrimeric G-protein (transducin) and promotes the exchange of
guanosine triphosphate (GTP) for guanosine diphosphate (GDP) on the α-subunit (Tα). Tα in turn disinhibits a cyclic guanosine monophosphate
(cGMP) phosphodiesterase (PDE), thereby allowing the hydrolysis of cGMP and the closure of cyclic nucleotide gated (CNG) channels. The
closure of CNG channels interrupts the flow of Na+ and Ca2+ into the photoreceptor and hyperpolarizes the membrane potential. (B) Deactivation
of R* is required to quench the phototransduction cascade. R* deactivation is initiated by the phosphorylation Ser and Thr residues at its
C-terminus by rhodopsin kinase (RK). RK is inhibited by recoverin (Rv), a process which is controlled by Ca2+. Ca2+ concentration falls as CNG
channels close during the activation of phototransduction, allowing the dissociation of RK from Rv, and subsequently the phosphorylation of R*.
The R* catalytic activity is further quenched by the binding of visual arrestin (Arr) to phosphorylated R*. (C) Deactivation of activated PDE (PDE*)
requires the hydrolysis of GTP to GDP on Tα. To reopen CNG channels, guanylyl cyclase synthesizes cGMP from GTP, a process that is
regulated by the Ca2+-binding protein, guanylyl cyclase-activating protein 1 and 2 (GCAPs).

where these phosphorylation sites removed in different combi- rhodopsin kinase is solely responsible for phosphorylating R*,
nations showed that all of the Ser and Thr sites at the C-terminus since phosphorylation of R* does not occur in mice lacking
are required for rapid and reproducible R* deactivation; the rhodopsin kinase.91
cluster of Ser and Thr sites at rhodopsin’s C-terminus functions Subsequent to C-terminus phosphorylation, the catalytic activ-
to ensure fast and reproducible R* deactivation.90 It is clear that ity of R* is quenched fully by binding of visual arrestin. Arrestins
are soluble cytoplasmic proteins that bind to G-protein-coupled cGMP and reopen CNG channels. The physiological actions of
receptors, thus switching off activation of the G-protein and GCAPs have been evaluated in physiological recordings. For
terminating the signaling pathway that triggers the cellular instance, when GCAP1 is dialyzed into gecko rods, the recovery 349
response; the most commonly studied arrestin is β-arrestin. phase of the light response is accelerated.109 In addition, suction
Visual arrestin exhibits exquisite specificity to phosphorylated electrode recordings from rods of GCAPs knockout mice show

Chapter 14
R*. Our understanding of how this specificity is conferred that the Ca2+ dependence of cGMP synthesis is required to set
emerges from extensive mutagenesis studies, and the crystal the time course of the rod’s photoresponse, and to suppress
structure of visual arrestin.92–95 These studies reveal that arrestin noise within the phototransduction cascade.110
is constrained into a latent, inactive structure by a network of
intramolecular interactions.92 According to the current model of Light adaptation
arrestin activation, these intramolecular constraints are released In a normal cycle of day and night, the illumination at the
by: (1) interaction with multiple phosphates on rhodopsin’s Earth’s surface varies over 11 orders of magnitude, making

Structure and Function of Rod and Cone Photoreceptors


C-terminus; and (2) interaction with the cytoplasmic loops of R*. photoreceptor adaptation fundamentally important to the
The physiological role of visual arrestin in controlling the rod normal functioning of the vertebrate visual system. Rods cells
single-photon response was studied by Xu and colleagues,24 who are able to adjust their sensitivity over 2–3 log units of light
created transgenic mice lacking visual arrestin. Suction electrode intensities, while cone cells exhibit no response saturation over
recordings of photoresponses from the OS of these transgenic 6–7 log units.3,4 The switch between rod and cone function
rods displayed a rapid, partial recovery, followed by a pro- from night to bright daylight covers a large portion of vision’s
longed final recovery phase. The timing of the slow phase of functional range, and downstream circuitry accounts for the
shutoff is consistent with the spontaneous decay of the R*. Thus, remainder of the range extension. While the molecular events
while R* phosphorylation is required to terminate rapidly its underlying the amplification cascade following photon capture
catalytic activity, the full quenching of R* activity requires visual by rhodopsin are well delineated, the mechanisms underlying
arrestin binding. photoreceptor adaptation are less well understood; however,
Ca2+ is known to play a feedback role in the adaptation
Deactivating PDE: control of transducin’s process.111,112
GTPase activity The role of Ca2+ feedback
Quenching of the phototransduction cascade also requires the In darkness, Na+ ions enter the OS through the CNG channels
shutoff of PDE*, which occurs through the deactivation of Tα. and are extruded in the IS through Na+/K+ ATPase pumps. This
The GTPase activity of Tα allows the reassociation of the inhibi- steady-state influx and efflux of Na+ is largely the basis for the
tory γ-subunit to the catalytic PDE subunits,96 a process which is circulating dark current in photoreceptor cells. Similarly, Ca2+
speeded through the participation of a photoreceptor-specific enters through CNG channels in the OS, but is extruded locally
RGS protein, RGS-9, and its binding partner, Gβ5.97 These pro- by NCKX exchangers. In mouse rods the steady-state influx and
teins act sequentially on Tα to facilitate the hydrolysis of bound efflux of Ca2+ hold concentration near 250 nM,113 but as light
GTP. The physiological role of all these proteins in the shutoff closes CNG channels, Ca2+ entry is blocked, Ca2+ efflux contin-
of the rod photoresponse was evaluated with transgenic mice ues, and [Ca2+]i falls. This change in [Ca2+]i is graded just like the
that had alterations in each of these components. Rod photores- dark current: the degree of lowered [Ca2+]i is proportional to the
ponses from transgenic mice expressing a PDE-γ with an intensity of background light.
impaired ability to stimulate GTP hydrolysis98 showed abnor- This decrease in [Ca2+]i triggers a feedback signal that is neces-
mally slowed recovery, demonstrating the necessary role of sary for light adaptation, since, when the [Ca2+]i decrease is
PDE-γ in Tα deactivation.99 Similarly, mice lacking RGS-9 blocked, adaptation to background illumination is greatly com-
showed profoundly slowed response recovery,100 as do mice promised.114,115 Existing evidence suggests that this feedback is
lacking Gβ5.101 RGS-9 is anchored to photoreceptor membranes rather complex, as it orchestrates several pathways directed
by the adaptor protein, R9AP.102–104 Interestingly, human patients toward different components of the transduction machinery.116–118
with mutations in R9AP have difficulties adapting to sudden Here we review the role of Ca2+ on each of these components.
changes in light levels that are mediated by cones,105 consistent
Adaptation mediated by Ca2+ feedback to retinal
with the higher concentration of RGS-9 complex in cones com-
guanylyl cyclase
pared to rods.106 Together, the current data support an important
It has long been recognized that feedback controlling acceler-
role of PDE-γ, RGS-9/Gβ5, and R9AP in Tα deactivation, and
ated recovery contributes to light adaptation.108 Recovery of the
consequently, response recovery, in both rods and cones.
light response requires the resynthesis of cGMP, which is needed
Resynthesis of cGMP: Ca2+ dependence of to reopen the CNG channels and re-establish the circulating
guanylyl cyclase current. The control of cGMP synthesis by RetGC/GCAPs is
Recovery of the dark current also requires that cGMP is resyn- central to this process. The contribution of accelerated cGMP
thesized to allow CNG channels to open. cGMP is synthesized synthesis to adaptation in rod photoreceptors was investigated
in photoreceptor OS by a retinal guanylyl cyclase (RetGC), in GCAP knockout mice.23,110 This work showed that the pres-
which is regulated by the protein GCAPs (GCAP 1 and 2).107 ence of GCAPs extends the sensitivity to background light in
GCAPs is an EF hand Ca2+-binding protein that confers on RetGC rod photoreceptors ~10-fold (Fig. 14.5), and can account for
a highly cooperative Ca2+ dependence for cGMP synthesis.108 about half of the rod’s capacity to adapt to background light.
Near the dark resting Ca2+ concentration RetGC’s activity is Recordings from cones in GCAPs knockout mice also show that
inhibited, but as OS Ca2+ falls in response to illumination as CNG GCAP contributes to background adaptation, but to a lesser
channels close, RetGC’s activity accelerates to restore the resting extent than it does in mouse rods.119
inhibition by recoverin in reconstituted biochemical assays was
1 similarly high.120 These results placed into doubt the role of
350 recoverin in mediating light adaptation. Suction electrode
recordings from the rods of transgenic mice lacking recoverin,
however, reveal a response speeding.124 This speeding effect is
exacerbated in rods that lack recoverin, but also have a reduced
Section 1

0.1 expression of rhodopsin kinase.125 Thus, in the physiological


SF/SFD

range of Ca2+ concentrations, recoverin can influence R* lifetime.


Despite this effect on R* lifetime, recoverin appears to play a
relatively minor role in rod light adaptation (Fig. 14.5).124 It
0.01 remains to be seen in cone photoreceptors what is the efficacy of
recoverin’s action. However, one might expect a greater contri-
Anatomy and Physiology
Basic Science and Translation to Therapy

bution to adaptation as experiments from salamander L cones


show that R* lifetime dominates the light response.126
10–3
100 101 102 103 104 Feedback regulation of the
IB (photons µm–2s–1) cGMP-gated channel
Fig. 14.5 Sensitivity as a function of background light was measured Another target of the Ca2+ feedback is the CNG channel itself. In
from mouse rod photoreceptors using suction electrodes (Fig. 14.3). vitro data have shown that the presence of Ca2+ calmodulin
Sensitivity was calculated from dim flash responses as the peak causes a decrease in the apparent affinity of the CNG channels
response amplitude divided by the flash strength. Flash sensitivity (SF) for cGMP.127 This effect is reversed in light when [Ca2+]i is low,
in the presence of increasing background light, background light (IB)
was normalized against the flash sensitivity in darkness (SFD) and is such that more channels tend to open despite the lowering of
plotted for the rods of several transgenic mice with particular elements cGMP levels. However, the role of CNG channel modulation in
of the phototransduction cascade altered. These mice include wild-type intact rods remains controversial, because, based upon the mea-
(), mice lacking recoverin (), mice where the calmodulin-binding site
on the cyclic nucleotide gated (CNG) channels has been eliminated
sured effect of Ca2+ on the apparent affinity of the channel for
(), mice lacking guanylyl cyclase-activating proteins 1 and 2 (), and cGMP and the intracellular range of cGMP concentration, it has
mice where both the calmodulin-binding site on the CNG channel and been predicted theoretically that the channel contributes little to
guanylyl cyclase-activating proteins 1 and 2 have been eliminated (). the overall adaptive behavior of the rod photoreceptor.117 The
The red line shows the predicted decline in sensitivity as a function
of background light intensity if rods displayed no light adaptation. site that confers Ca2+ calmodulin sensitivity has been identified
The blue line is best-fitting Weber–Fechner function for wild-type rods, in the β-subunit of the CNG channel.128,129 Transgenic mice with
and shows that light adaptation extends the sensitivity of rod this Ca2+-binding site disrupted show little influence on the light
photoreceptors to 100-fold higher background light levels. Rods from
response or in the adaptive features of rod phototransduction,130
mice lacking recoverin, or the calmodulin-binding site on the CNG
channels, show little deviation from wild-type rods, indicating that these consistent with previous conjecture (Fig. 14.5). The role of CNG
proteins do not play a significant role in light adaptation. However, channel modulation in cones, however, may be more profound
in mice lacking guanylyl cyclase-activating protein 1 and 2, light than for rods. For instance, the cones of the ground squirrel
adaptation is impaired, but not absent, even when the calmodulin-
binding site on the CNG channel is also eliminated (red dashed line). demonstrate strong Ca2+-dependent modulation of the CNG-
The remaining mechanisms that control light adaptation in rod gated current in the normal range of Ca2+ concentrations, while
photoreceptors remain unidentified. (Data replotted from Chen J, the rods of the same species do not.131 In addition, Ca2+ concen-
Woodruff ML, Wang T, et al. Channel modulation and the mechanism tration varies over a larger dynamic range in cones than rods.132
of light adaptation in mouse rods. J Neurosci 2010;30:16232–40.)
Modulation of cone CNG channels may also involve an uniden-
tified soluble factor instead of calmodulin.133 Ultimately how this
modulation influences light adaptation in cones remains to be
Recoverin and control of rhodopsin kinase seen.
The accelerated response recovery that mediates light adapta-
tion is also achieved through the speeded deactivation of R*. The
Other (Ca2+-independent) adaptation
first step in the quenching of R* activity is the phosphorylation mechanisms: protein translocation
of its C-terminus by rhodopsin kinase, a process that is believed A mechanism operating at a longer timescale (minutes rather
to be regulated in a Ca2+-dependent manner by the protein recov- than seconds) that may contribute to regulating photoreceptor
erin. Recoverin is a highly conserved Ca2+-binding protein found cell performance is the light-driven redistribution of specific
in both rod and cone photoreceptors. In vitro, recoverin binds signal transduction proteins between the compartments of the
and inhibits rhodopsin kinase when it is Ca2+-bound, and accel- photoreceptor cell. The immunoreactivity of both Tα and visual
erates rhodopsin kinase activity as Ca2+ is removed.120,121 Since arrestin is known to redistribute in rods in response to light.134–137
[Ca2+]i is high in darkness, it has been suggested that, under Visual arrestin immunostaining predominates at rod IS, as well
dark-adapted conditions, recoverin prolongs the catalytic activ- as at the outer nuclear layer and outer plexiform layers of dark-
ity of R* by inhibiting rhodopsin phosphorylation. Early evi- adapted retinas, and shifts to rod OS upon light exposure. Tα
dence in experiments that introduce recombinant recoverin into immunoreactivity shifts in the opposite direction in response to
gecko and salamander rods indicated that recoverin prolongs light.136–138 Quantitative measurements show that bright light
the light response.122,123 However, recoverin’s effect was most exposure is required to trigger translocation: for visual arrestin
prominent at Ca2+ concentrations outside the rod’s physiological the threshold is 1000 rhodopsin excitations per rod per second,
range (>1 µM). The Ca2+ requirement for rhodopsin kinase a light intensity at the upper limits of rod vision; for Tα at least
10 000 rhodopsin excitations are required.139 Sokolov and col- differential modulation of components of the phototransduction
leagues confirmed the physiological movement of Tα by com- cascade. In addition, differences in their characteristic morphol-
bining serial tangential cryosectioning of the retina with Western ogy may play a role in setting the response properties (i.e., the 351
blot analysis, and demonstrated that such movement may help continuously invaginating OS plasma membrane in cones pro-
to extend the range of light intensities in which the rods are able vides greater surface area for ion exchange and chromophore

Chapter 14
to operate.140 Similarly, visual arrestin movement may regulate recycling). Also important may be their signal transduction cas-
rod performance under varying light environments. cades: molecular cloning has revealed that phototransduction in
Our current understanding of protein translocation is that it the vertebrate rods and cones is regulated by structurally homol-
occurs by diffusion. In the case of Tα, the lipid modifications on ogous but distinct groups of signaling proteins. Therefore, it is
the α- and γ-subunits act synergistically to anchor Tα GDP on reasonable to assume that the phototransduction in rods and
the membrane. Upon activation, the subunits dissociate and cones is qualitatively similar, and that quantitative differences
become solubilized.141 In an elegant set of experiments, Lobanova in the transduction steps underlie the characteristic rod and cone

Structure and Function of Rod and Cone Photoreceptors


and colleagues showed that light threshold can be shifted to behavior. For example, R* may activate more rod Tα molecules
either a lower or higher light intensity using mutant mice whose than cone opsin R* in activating cone Tα, resulting in a higher
GTPase-activating (GAP) complex is enhanced or decreased.142 gain and a larger response amplitude.
Their data suggest that, when the GAP complex is saturated, Tα To determine the roles of rod and cone phototransduction
GTP can escape the OS compartment and diffuse to the IS. proteins in setting the response properties of the photoreceptor
Whether such a mechanism also governs cone translocation is cells, transgenic technology has recently been used to express
uncertain. It has been observed that cones contain a higher con- rod components in cones, and vice versa. For example, Kefalov
centration of GAP proteins,106,143 a reason that could explain a and colleagues148 expressed an L-cone opsin in Xenopus rods,
much higher threshold for cone Tα translocation.144 However, and rhodopsin in Xenopus cones. They surprisingly found that
others have observed that cone Tα readily translocates.145 In the the activation of rhodopsin and L-cone opsin within the same
case of rod arrestin, its interaction with microtubules restricts its cell yielded nearly identical photoresponses. In addition, similar
location to the IS in the dark. Light exposure creates its high- photoresponses were observed in mouse rods transgenically
affinity binding partner, R*, which drives its movement towards expressing an S-cone pigment.149 These results collectively rule
the OS.146 out the visual pigment from being a major contributor to rod–
It has become increasingly clear that Ca2+-dependent light cone functional differences. Similar experiments have also
adaptation results from the sum of multiple mechanisms that act addressed the role of Tα. Deng and colleagues150 showed no
at different sites in the phototransduction cascade. While bio- functional differences in mouse rods expressing cone Tα, or
chemical experiments have identified the Ca2+-sensitive steps in mouse cones expressing rod Tα. However, Chen and col-
the phototransduction cascade, a full understanding of how leagues151 found to the contrary that complete exchange of
these steps contribute to visual adaptation requires a method rod Tα with cone Tα produced rod photoresponses that were
that critically evaluates the timing, as well as the strength, of 10-fold desensitized, and thus transducin might contribute
feedback modulation under physiological conditions. Given the partially to observed rod–cone functional differences. These
complexity of Ca2+ feedback, coupled with the possibility that results, as well as homolog substitution experiments with other
other adaptation mechanisms may overlay on this feedback components of phototransduction, require further evaluation
regulation, visual adaptation, particularly in cones, remains one to determine how differences in rod and cone photoresponses
of the least understood areas in phototransduction. This com- are established.
plexity is being unraveled by the systematic removal of Ca2+
feedback pathways in vivo using mouse genetics, and by mea- INNER SEGMENT AND CONNECTING CILIA
suring the resultant change in adaptation with electrophysiolog-
ical measurements. The molecular mechanism behind the Inner segment
observed phenotype can then be confirmed by biochemical The functional role of the IS can be appreciated by examination
experiments. Using this approach, the Ca2+ feedback loops can of its unique anatomy, which is highly specialized to fuel the
be evaluated independently, as well as in sum, since the mice high energy and protein synthesis requirements of the photore-
can be bred to combine the individual components (Fig. 14.5). ceptor. The metabolic functions of the IS are constantly and
Eventually, as each and every Ca2+-dependent pathway is peeled highly active, generating most of the rod cell’s energy as well as
away, the remaining (perhaps Ca2+-independent) pathways will being the primary site for synthesizing new proteins. The proxi-
be revealed. mal portion of the IS, termed the myoid, contains the endoplas-
mic reticulum and Golgi apparatus, while the distal portion,
Differences between rod and termed the ellipsoid, is densely packed with elongated mito-
cone phototransduction chondria that are aligned circumferentially along the axis of the
What factors underlie the specialized function of rods and cones? rod cell (Fig. 14.2). Cones have a larger IS, perhaps reflecting
This remains a very important problem in phototransduction. their higher metabolic needs. This photoreceptor cell compart-
Investigation of rod phototransduction has benefited from rod- ment is structured for high O2 consumption, as well as for gly-
dominated species where biochemical and physiological assays colysis at very high rates. One major metabolic demand comes
are robust. The relatively low number of cones in most mam- from the extrusion of Na+ from the cytoplasm by the Na+/K+
malian species has made a biochemical analysis of cone photo- pump at the IS plasma membrane, which balances the influx of
transduction less fruitful, although the Nrl knockout mouse has Na+ through the cGMP-gated channels at the OS. In darkness,
proven useful for generating cone-dominant retinas.147 As these pumps operate at a very high rate to accommodate the
described above, some quantitative differences may arise for high fluxes generated by many open channels in the OS, setting
up the “dark” circulating current.152 The rate that these pumps Trafficking of membrane-associated proteins to the OS may
can operate is related to the supply of ATP153 generated by the occur through their interaction with transmembrane proteins as
352 mitochondria in the ellipsoid. This ATP synthesis rate is coupled they are delivered out of the trans-Golgi network. It was also
with the O2 delivery to the IS, which is completely supplied by recently discovered that membrane-associated phototransduc-
the choroidal circulation. tion proteins are transported by prenyl- or acyl-chain-binding
Another major metabolic need is a high rate of protein synthe- proteins. For example, rhodopsin kinase and cone PDE6α’
Section 1

sis that occurs in the myoid region of the IS to meet the demand require PrBP/δ, which contains a binding site for prenyl group,
of high levels of phototransduction proteins at the OS. In par- for transport to the OS,168 while the α-subunit of rod transducin
ticular, each rod contains ~5 × 107 rhodopsin molecules, of which requires UNC119, which binds acyl chains.169 Thus diverse
10% at the tip of the OS are phagocytosed by the RPE, beginning mechanisms are responsible for correct targeting of phototrans-
at the onset of light each day. To maintain homeostasis, ~5 × 106 duction proteins from the IS to the OS compartment.
molecules are synthesized daily and added to the base of the OS.
Anatomy and Physiology
Basic Science and Translation to Therapy

Similar events occur in cones, although they express about half The connecting cilium
the amount of visual pigment as rods. As with other cells, the A slender nonmotile connecting cilium that is 0.3 µm in diameter
soluble and peripheral membrane proteins are made in the connects the IS to the OS. The microtubule-based axoneme of the
cytosol, while the transmembrane proteins are made in the endo- connecting cilium lacks the central microtubule pair of the motile
plasmic reticulum. To facilitate the speed of molecular collisions, cilium, and therefore has a 9 × 2 + 0 microtubule structure
many of the phototransduction proteins are associated with the towards the base of the cilia. The doublet microtubules transition
membrane through lipid modifications and protein–protein to a 9 × 1 + 0 singlet microtubule arrangement more distally from
interactions. These interactions are dynamic, with functional the basal body and continue through much of the OS (Fig. 14.2).
complexes forming and dissociating on the membrane surface The axoneme arises from basal bodies which, together with the
on a rapid timescale. For example, the rod Tα is heterogeneously centrioles, act as microtubule organizing centers of the photo­
acylated at the amino-terminal glycine residue while the receptor cell. It is the site of tremendous vectorial flow of lipids
γ-subunit is carboxyl-methylated and prenylated at the cysteine and proteins from their site of synthesis in the IS to the OS. It
residue. PDE6α and rhodopsin kinase are farnesylated, while has been estimated that 2000 opsin molecules are transported
PDE6β is geranylgeranylated.154,155 In some instances, the lipid every minute from the IS to the OS.170 As post-Golgi carrier
modification is exposed as a consequence of a change in protein vesicles are delivered to the connecting cilium, proteins destined
structure. An example is recoverin that, upon binding calcium, for the OS need to be sorted between the disc lamellar region,
exposes its amino-terminal myristoyl group.156 GCAP1 and the disc rim region, and the plasma membrane in the OS. How
GCAP2 are also calcium-binding proteins that are myristoylated, the protein cargo is sorted at this point is not understood fully,
but exposure of their lipid group is not regulated by calcium but is thought to involve recognition of the specific trafficking
binding. These proteins are complexed with RetGCs and regu- motif displayed on the protein cargo by their cognate transport
late their activity in a calcium-dependent manner. complex that also includes the intraflagellar transport (IFT) par-
ticles.170 The IFT particles associate with the post-Golgi carrier
Targeting of phototransduction proteins from vesicles at the base of the cilium and transport them along the
the inner segment to the outer segment connecting cilium to the OS. IFTs are large protein complexes
How phototransduction proteins traffic from the IS, the site of that move along the axoneme by motor proteins and may be
synthesis, to the OS, the site of phototransduction, is an area of viewed as conveyer belts that deliver different cargos to their
active research. This question is made more important by the respective destination.
fact that defective trafficking can cause photoreceptor cell death. Actin and myosin, two proteins common to many cells in the
For example, a cluster of naturally occurring mutations on rho- body, are also present in photoreceptors. Both myosin and actin
dopsin’s carboxyl-terminus causes autosomal dominant RP in filaments are localized to the photoreceptor connecting cilium
human patients.157 These mutations do not affect the biochemical and IS/OS. Disruption of either of these proteins causes retinal
properties of rhodopsin with respect to its ability to bind 11-cis degeneration.171 Actin depolymerization promotes disc over-
retinal and form a visual pigment, nor its ability to activate Tα growth and photoreceptor death,172 and myosin mutants appear
following photon absorption in reconstituted systems.158–160 The to have defects in the transport of molecules from the IS to the
underlying defect remained a mystery until these mutants were OS. Myosin defects cause numerous diseases, including Usher
expressed in transgenic mice, whereupon it was observed that syndrome, that affect both hearing and vision.173–175
they are mislocalized throughout the cell and caused retinal Other proteins are receiving increased attention because of
degeneration.161–163 It is now known that the VxPx motif at the their association with the connecting cilium and human retinal
rhodopsin’s carboxyl terminus is important for sorting of rho- disease. First described nearly two decades ago, one such protein,
dopsin into transport carriers from the trans-Golgi network RP1, is responsible for 5–10% of all RP cases.176 RP1 is a large
towards the connecting cilium en route to the OS.164 This is a protein localized to the connecting cilium in both rods and cones
recognition site for transport proteins that include the small and is especially concentrated at the site of nascent OS discs. RP1
GTPase Arf4, ASAP1, Rab11, and FIP3.165 The VxPx motif is also has recently been shown to bind microtubules, suggesting that
present in cone opsins. Other integral membrane proteins this interaction may stabilize microtubules in the axoneme.177–179
contain their own trafficking motif. These motifs have been iden- An RP1 knockout mouse model shows that discs are misshapen,
tified in peripherin/rds166 and the cGMP-gated channel in overgrown, and fail to align properly.177 This phenotype corre-
rods.167 Often defects in transport of one transmembrane protein lates well with ERG abnormalities in RP1 patients.
do not affect transport of others, suggesting that transport occurs The RP GTPase regulator (RPGR) is a GTPase regulator found
independently. in rods. Mutations in RPGR180 cause X-linked RP3, a severe and
progressive retinal dystrophy. Antibodies localize RPGR and a Nucleus
related protein, RPGRIP (RPGR interacting protein) throughout
The nucleus contains the primary genome of the photoreceptor
the human rod and cone OS,181 as well as in amacrine cells. RPGR 353
and is responsible for the initiation of genetic programs in the
is found in connecting cilia of rods and cones and in the cilia of
cell. As a result, the nucleus is the target for gene therapy pro-
airway epithelia.182 An RPGR gene knockout mouse (an animal
tocols designed to correct genetic defects in the photoreceptor.

Chapter 14
model of RP3)183 demonstrated mislocalization of opsin in cones,
Rods and cones differ in their nuclear features. The nucleus of
and reduced quantities of rhodopsin in rods prior to photorecep-
the rod photoreceptor is rounder and is stained darker by
tor degeneration. These data suggest a role for RPGR in main-
nucleophilic stains due to the presence of a large clump of het-
taining the polarized distribution of proteins between the IS and
erochromatin. Nuclei of cones are larger and oval in shape, with
the OS through the connecting cilium.
one to several clumps of heterochromatin and a larger amount
Centrins are calcium-binding proteins associated with
of lightly staining euchromatin. Like most cells, photoreceptors
centrosome-related structures. The mammalian rods and cones

Structure and Function of Rod and Cone Photoreceptors


also contain a second, nonnuclear genome in the mitochondria.
express four centrin isoforms; three of these (Cen1p–3p) are in
Biochemical pathways linking photoreceptor cell death, the
the connecting cilium and three (Cen2p-4p) are in the basal
nucleus, and the mitochondria of the IS are beginning to come
body.184 These centrins may participate in the alignment of the
into focus.
photoreceptor OS. Inasmuch as centrins were shown to interact
with transducin in a calcium-dependent manner, they have been
proposed to regulate the light-dependent translocation of Tα
from the OS to the IS.184
PHOTORECEPTOR SYNAPTIC TERMINAL
The ciliary rootlet, first recognized over a century ago, is a The light response generated by the phototransduction cascade
prominent structure originating from the basal body at the in the OS is passively relayed to the synaptic terminal, called a
proximal end of a cilium. Rootlets appear to be particularly spherule for rods and a pedicle for cones, where it modulates
robust in retinal photoreceptors, extending from the basal the rate of glutamate release on to second-order retinal neurons
bodies to the synaptic terminals, anchoring endoplasmic reticu- (Fig. 14.6). Since open CNG channels leave the membrane poten-
lum membranes along their length. Recent studies indicate tial of photoreceptors relatively depolarized in darkness, these
that rootlets are composed of a structural protein, aptly named synapses support a high level of Ca2+ influx and thus a high level
rootletin. Rootletin protofilaments are bundled into variably of glutamate release. Light activates the phototransduction
shaped thick filaments within the cilium.185 cascade, leading to the closure of CNG channels, the hyper­
Additional proteins, such as TULP1,186,187 cadherin,188 and polarization of the membrane potential, and a reduction in Ca2+
myocilin,189 are associated with the connecting cilium: several influx, which in turn reduces glutamate release. This reduction
are known to be associated with rod photoreceptor disease. It in glutamate release constitutes the “light response” that is
suggests, not surprisingly, that the highly specialized connecting passed to the retinal circuitry.
cilium is a complicated structure connecting the IS to OS, and To allow high levels of glutamate release, the photoreceptor
that there will undoubtedly be additional proteins associated synaptic terminals house a specialized synaptic structure, called
with this unique subcellular structure. a ribbon, which is composed of the protein ribeye.190 The ribbon

Fig. 14.6 Glutamate release from


photoreceptor cells occurs at a specialized
synaptic terminal, called a spherule for rods,
Synaptic ribbon and a pedicle for cones. Depicted is a slice
through a rod spherule. Glutamate release is
facilitated by a specialized structure called a
synaptic ribbon, which binds glutamatergic
vesicles and brings them to the plasma
membrane. Vesicle fusion then occurs as
L-type Ca channels near the release site
allow the influx of Ca2+, allowing glutamate
(black dots) to be released into the synaptic
cleft, where it can be sensed by rod bipolar
cells (RB) and horizontal cells (HC). Both RB
and HC make an invaginating synapse with
Ca2+ the photoreceptor cells.

HC HC

RB RB
tethers glutamatergic vesicles and is believed to act like a con- PHOTORECEPTOR DYSFUNCTION
veyor belt to bring these vesicles to the active zone, where they
AND DISEASE
354 promote vesicle priming and fusion with the plasma membrane
to release glutamate into the synaptic cleft191,192 (Fig. 14.6). While Rhodopsin mutations
the spherules of rods contain only one ribbon (i.e., one release More than 100 different mutations of rhodopsin have been asso-
site) in mammalian species,193 the cone pedicles contain many
Section 1

ciated with autosomal dominant and recessive RP (adRP and


ribbons whose numbers vary by species. In mice, for instance, arRP) and autosomal dominant congenital stationary night
cone pedicles contain ~10 ribbons,194 while pedicles in other blindness.213 This phenotypic heterogeneity points to diverse
species like ground squirrels may contain ~15 or more ribbons.195 disease mechanisms arising from the same gene product. In the
Primate cone pedicles may also contain in excess of 25 case of adRP, it has been proposed that some mutations lead to
ribbons.196,197 Multiple ribbons in each cone pedicle allow the misfolding of the opsin protein, which then accumulates in
output of the cone to be relayed to many classes of cone bipolar
Anatomy and Physiology
Basic Science and Translation to Therapy

the endoplasmic reticulum and elicits the unfolded protein


cells (see Chapter 15, Functional anatomy of the mammalian response.214 The accumulation of misfolded opsin may also inter-
retina). fere with the sorting and trafficking of normal proteins destined
Compared to conventional central synapses, photoreceptor for the OS or other parts of the cell.215 This may result in a patho-
cells release glutamate in the absence of the stimulus. Such a logic mechanism where the cell cannot replenish other required
configuration places fundamental constraints on the mecha- proteins that do not harbor mutations.
nisms that regulate glutamate release. For instance, the persis- Other rhodopsin mutations appear to affect trafficking of rho-
tent depolarization of photoreceptors in darkness would cause dopsin from the IS to the OS. As mentioned previously, muta-
many types of ion channels to desensitize. To allow robust tions involving the last five carboxyl amino acids, such as P347A,
control of glutamate release with changes in membrane poten- P347S or S344ter, allow rhodopsin to appear to fold normally
tial, a Cav1.4 channel located close to the active zone in the rod and to function normally in terms of Tα activation and deactiva-
spherule and cone pedicle allows the Ca2+ influx that regulates tion by phosphorylation and visual arrestin binding. Yet, muta-
release. These L-type Ca2+ channels display little voltage or Ca2+- tions in these residues cause adRP. Several lines of evidence
dependent desensitization,198,199 and thus are ideal to support suggest that the C-terminus of opsin contains a signal for vec­
continuous glutamate release in darkness. In addition, in rod torial transport of newly synthesized opsin toward the OS. In a
spherules these channels associate with the modulatory Ca2+- retinal cell-free system, it was demonstrated that a monoclonal
binding protein, CABP4.200 The association of Cav1.4 with CABP4 antibody to the C-terminal domain of rhodopsin inhibited post-
shifts the voltage sensitivity of Cav1.4 to more hyperpolarized Golgi vesicle formation and arrested newly synthesized rhodop-
membrane potentials.200,201 This allows the gating of Cav1.4 chan- sin in the trans-Golgi network.216 A wild-type C-terminal peptide,
nels to be modulated robustly over the rod photoreceptor’s but not a mutant peptide bearing the mutation at the P347 posi-
physiological voltage range. tion, can also block this process.217 Another line of evidence
Additional control of glutamate release may also involve comes from an in vivo model where a truncation mutant, S344ter,
the interaction of other proteins with the synaptic machinery. was expressed in the photoreceptors of transgenic mice. Anti-
For instance, visual arrestin serves to quench fully phosphory- bodies that specifically recognize the truncated opsin showed
lated rhodopsin in the rod OS, but has been shown to interact strong immunoreactivity in the IS, indicating a block in the trans-
with N-ethylmaleimide-sensitive factor at the rod spherule to port to the OS.163 However, some truncated opsins are also
increase the turnover rate of SNARE complexes.202 Similarly, cotransported with the full-length protein to the OS. The role of
the protein recoverin, which serves as a modulator of rhodopsin the rhodopsin C-terminal domain in vectorial transport was also
phosphorylation in the rod OS, has a separate action in the investigated in MDCK cells.218 This cell line exhibits polarized
rod spherule that increases the magnitude of the reduction in structure when it is grown on a porous support. When trans-
glutamate release on to rod bipolar cells.203 Such modulation fected into this cell line, wild-type opsin was transported to the
may also be controlled by the light-dependent translocation apical membrane surface, whereas an opsin mutant lacking the
of these components,135,204 or in the activity of signaling cas- terminal 32 amino acids was not.
cades initiated by metabotropic glutamate receptors in the
photoreceptor terminal.205,206 Finally, the control of Ca2+ homeo-
stasis in photoreceptor terminals can also modulate glutamate
Constitutive phototransduction and
release.207–209 retinal disease
It should be noted that most of our understanding of the It is thought that constitutive phototransduction causes some
structure and function of ribbon synapses has emerged from the forms of retinal disorders. Unabated signal flow can arise from
study of rod photoreceptors in the amphibian retina, particularly different steps in the visual cascade. For example, certain muta-
the salamander. However, the characteristics of glutamate tions in rhodopsin, particularly those that affect the salt bridge
release in cones can also be studied in this same species and between Lys-296 and Glu-113 (such as A292E and G90D), can
reveal characteristics that are tuned for the faster kinetics of the lead to constitutive activity.219 The interaction between Lys-296
light response in cones (reviewed by Thoreson210). Furthermore, and Glu-113 constrains the chromophore-free opsin to an inac-
since the cone photocurrent is difficult to saturate, increased tive conformation.220 Disruption of this bond leads to an opsin
vesicle turnover and recruitment are required to maintain the conformation that can support Tα activation. This activity is
high level of glutamate release.211,212 Ultimately, the mechanisms suppressed when the mutant opsin is regenerated with the chro-
that produce these functional distinctions between rod spherules mophore 11-cis retinal. In the case of G90D and A292E muta-
and cone pedicles remain largely unidentified and are currently tions, light exposure leads to formation of opsin that can still
under investigation. activate the visual cascade. This can lead to impairment of visual
function because the transduction machinery is overwhelmed by finding is uncertain because the site of cGMP accumulation and
this constitutive activity. Patients carrying these mutations suffer action is intracellular. For example, the CNG channels open only
from night blindness because phototransduction in rods cannot when cGMP is dialyzed inside the truncated OS, not when it is 355
be fully quenched.221,222 However, in the instance of the rhodop- applied externally.232
sin K296E mutation, no evidence of constitutive activity was Although PDE6 is responsible for hydrolyzing cGMP, RetGCs

Chapter 14
observed when it was expressed in rod photoreceptors of trans- are responsible for synthesizing the cGMP that is necessary for
genic mice; instead, it was found bound to arrestin.223 It was reopening of the CNG channel leading to recovery of the dark
found subsequently that stable rhodopsin–arrestin complex is current. Two membrane forms of RetGC are expressed specifi-
toxic, and rod photoreceptor survival can be prolonged if such cally in the retina of several mammalian species, including rat,233
complex is prevented in transgenic mice that lack visual arrestin human,234,235 and bovine.236 Whereas other membrane cyclases
and Tα.224 are activated by the binding of peptide ligands to their extra­
Defective shutoff can also be caused by the lack of visual cellular domains, the activity of RetGCs is controlled by interac­

Structure and Function of Rod and Cone Photoreceptors


arrestin or rhodopsin kinase. These genes are mutated in tion with GCAPs through their intracellular domain.237,238
patients diagnosed with the recessive genetic disorder Oguchi Because the extracellular domain of RetGCs is situated in the
disease,225,226 a nonprogressive disorder characterized by early enclosed and seemingly inaccessible intradiscal space, it is ques-
onset and stationary night blindness. These mutations are tionable whether the extracellular domain serves as a receptor.
thought to cause a loss of function in the visual arrestin or The retinal cyclases are therefore termed “orphan” receptors
rhodopsin kinase gene. Because of the inability to shut off signal because no ligands have yet been identified. Although two forms
flow, the rod photoreceptor becomes saturated even at very low of RetGC are expressed in the photoreceptor layer, there is evi-
light levels, leading to night blindness. Interestingly, some dence that they are not functionally redundant. First, they appear
patients with the visual arrestin gene mutation also suffer from to be expressed at different levels in different photoreceptors.
RP, pointing to a potential causal role between constitutive For example, it was observed that RetGC1 immunoreactivity is
signal flow and photoreceptor cell death. Using the visual arres- stronger in cone OS than in rod OS239; RetGC2, on the other hand,
tin knockout mouse model, Chen et al. demonstrated that the is expressed in the rod OS,240 but evidence for cone expression
absence of visual arrestin in the rod photoreceptor does lower is lacking. Second, RetGC1 and RetGC2 behave differently bio-
the threshold for light damage.227 Therefore, protection from chemically in terms of their regulation by GCAPs: RetGC1 is
light exposure is an important preventive measure to prolong regulated by both GCAP1 and GCAP2, whereas RetGC2 is regu-
photoreceptor cell survival in those instances where constitutive lated by GCAP2 but not by GCAP1.234,237–239,241 Third, loss of func-
signal flow is the disease mechanism. tion of RetGC1 appears to affect cones more than rods; a mouse
knockout of RetGC1 lacked cone function altogether, but rod
Transducin defects and retinal disease function was only reduced.242 The importance of RetGC1 in
Transducin is composed of three subunits: α, β, and γ. Exten- cones is also reflected in the rd chicken model where a loss of
sive genetic screening has revealed mutations only in the rod RetGC1 function is thought to cause retinal degeneration in this
Tα.228 The associated disease is autosomal dominant congenital cone-dominant animal.243 These animal models will be valuable
stationary night blindness. The mutation is in the position for evaluating the pathophysiology for Leber’s congenital amau-
homologous to that affected by the oncogenic mutation in rosis, an early-onset blindness caused by a naturally occurring
p21ras, a small G-protein. It is therefore hypothesized that mutation in RetGC1.
this mutation may affect Tα deactivation, leading to the night Another naturally occurring mutation that may influence
blindness phenotype. Interestingly, mice that lack rod Tα have intracellular cGMP concentration is the Y99C mutation in
normal retinal structure, indicating that this abundant visual GCAP1, a mutation that is thought to cause cone dystrophy in
G-protein has no structural function in the photoreceptor, and an autosomal dominant manner.244 This mutation renders
that signal flow is not a prerequisite for rod photoreceptor GCAP1 capable of stimulating RetGC, independently of Ca2+
cell survival.20 concentration.245,246 Constitutive activity of RetGC may therefore
lead to an accumulation of intracellular cGMP.
cGMP and photoreceptor cell physiology It is known that a small increase in the level of free cGMP can
One of the first characterized mutations affecting components profoundly affect the open probability of CNG channels, such
in the phototransduction cascade is the null mutation in the that a greater number of channels will be open. This will in turn
β-subunit of PDE6, leading to the rapid retinal degeneration influence the ion influx, including Ca2+, which would then
phenotype in the rd mouse.13,14 Subsequently, null mutations in directly and/or indirectly influence the physiology of the pho-
PDE6β have been found in patients diagnosed with arRP.229 The toreceptor cell, leading to cell death. In support of this idea,
complete PDE6 enzyme is made up of two catalytic chains (α survival of rd1 photoreceptors, whose mutation in PDE6β leads
and β), each associated with an inhibitory chain (γ). Since both to buildup of cGMP, was enhanced when the mutation was put
α- and β-subunits are necessary for the formation of a functional into a CNG channel knockout mouse background.247 This obser-
PDE holoenzyme, the mutation in the γ-subunit renders the vation motivates effort to synthesize small molecules that spe-
holoenzyme inactive. Farber and Lolley230 showed that photo­ cifically block the CNG channel as a means of prolonging
receptor cell death is preceded by an accumulation of cGMP in photoreceptor cell survival.248 A loss of RetGC1, on the other
the retina, which is consistent with the role of this enzyme in hand, is expected to lead to lowered cGMP levels. This might be
degrading cGMP. Another study investigating the relationship functionally equivalent to constant light and may lead to cell
between cGMP and photoreceptor cell death showed that exter- death. However, further experiments will be needed to evaluate
nal application of cGMP on to cultured eyecups resulted in rapid rigorously the relationship between cGMP concentration and
photoreceptor cell death.231 However, the relevance of this retinal degeneration.
34. Arikawa K, Molday LL, Molday RS, et al. Localization of peripherin/rds in
REFERENCES the disk membranes of cone and rod photoreceptors: relationship to disk
1. Baylor DA, Lamb TD, Yau KW. Responses of retinal rods to single photons. membrane morphogenesis and retinal degeneration. J Cell Biol 1992;116:
356 J Physiol 1979;288:613–34. 659–67.
2. Hecht S, Schlaer S, Pirenne MH. Energy, quanta, and vision. J Gen Physiol 35. Travis GH, Sutcliffe JG, Bok D. The retinal degeneration slow (rds) gene
1942;25:819–40. product is a photoreceptor disc membrane-associated glycoprotein. Neuron
3. Burkhardt DA. Light adaptation and photopigment bleaching in cone photo- 1991;6:61–70.
receptors in situ in the retina of the turtle. J Neurosci 1994;14:1091–105. 36. Conley SM, Stuck MW, Naash MI. Structural and functional relationships
Section 1

4. Normann RA, Perlman I. The effects of background illumination on the pho- between photoreceptor tetraspanins and other superfamily members.
toresponses of red and green cones. J Physiol 1979;286:491–507. Cell Mol Life Sci 2012;69:1035–47.
5. Morimura H, Fishman GA, Grover SA, et al. Mutations in the RPE65 gene in 37. Bascom RA, Manara S, Collins L, et al. Cloning of the cDNA for a novel
patients with autosomal recessive retinitis pigmentosa or Leber congenital photoreceptor membrane protein (rom-1) identifies a disk rim protein family
amaurosis. Proc Natl Acad Sci U S A 1998;95:3088–93. implicated in human retinopathies. Neuron 1992;8:1171–84.
6. Daiger SP. Identifying retinal disease genes: how far have we come, how far 38. Molday RS, Warren R, Loewen C, et al. Cyclic GMP-gated channel and
do we have to go? Novartis Found Symp 2004;255:17–27; discussion 36, peripherin/rds-rom-1 complex of rod cells. Novartis Found Symp 1999;
177–8. 224:249–61; discussion 61–4.
Anatomy and Physiology

7. Sheffield VC, Fishman GA, Beck JS, et al. Identification of novel rhodopsin 39. McNally N, Kenna PF, Rancourt D, et al. Murine model of autosomal domi-
Basic Science and Translation to Therapy

mutations associated with retinitis pigmentosa by GC-clamped denaturing nant retinitis pigmentosa generated by targeted deletion at codon 307 of the
gradient gel electrophoresis. Am J Hum Genet 1991;49:699–706. rds-peripherin gene. Hum Mol Genet 2002;11:1005–16.
8. Flannery JG, Farber DB, Bird AC, et al. Degenerative changes in a retina 40. Clarke G, Goldberg AF, Vidgen D, et al. Rom-1 is required for rod photorecep-
affected with autosomal dominant retinitis pigmentosa. Invest Ophthalmol tor viability and the regulation of disk morphogenesis. Nat Genet 2000;25:
Vis Sci 1989;30:191–211. 67–73.
9. Suber ML, Pittler SJ, Qin N, et al. Irish setter dogs affected with rod/cone 41. Corless JM, Schneider TG. Patterns of interdisk connections within the lamel-
dysplasia contain a nonsense mutation in the rod cGMP phosphodiesterase lar domains of retinal rod outer segment disks: observations relevant to the
beta-subunit gene. Proc Natl Acad Sci U S A 1993;90:3968–72. axial propagation of incisures. Exp Eye Res 1987;45:883–905.
10. Wrigstad A, Narfstrom K, Nilsson SE. Slowly progressive changes of the 42. Kajimura N, Harada Y, Usukura J. High-resolution freeze-etching replica
retina and retinal pigment epithelium in Briard dogs with hereditary retinal images of the disk and the plasma membrane surfaces in purified bovine rod
dystrophy. A morphological study. Doc Ophthalmol 1994;87:337–54. outer segments. J Electron Microsc (Tokyo) 2000;49:691–7.
11. Narfstrom K. Hereditary progressive retinal atrophy in the Abyssinian cat. 43. Miyaguchi K, Kuo CH, Miki N, et al. Topography of opsin within disk
J Hered 1983;74:273–6. and plasma membranes revealed by a rapid-freeze deep-etch technique.
12. Edwards RB, Szamier RB. Defective phagocytosis of isolated rod outer seg- J Neurocytol 1992;21:807–19.
ments by RCS rat retinal pigment epithelium in culture. Science 1977;197: 44. Roof DJ, Korenbrot JI, Heuser JE. Surfaces of rod photoreceptor disk mem-
1001–3. branes: light-activated enzymes. J Cell Biol 1982;95:501–9.
13. Bowes C, Li T, Danciger M, et al. Retinal degeneration in the rd mouse is 45. Allikmets R, Gerrard B, Hutchinson A, et al. Characterization of the human
caused by a defect in the beta subunit of rod cGMP-phosphodiesterase. ABC superfamily: isolation and mapping of 21 new genes using the expressed
Nature 1990;347:677–80. sequence tags database. Hum Mol Genet 1996;5:1649–55.
14. Pittler SJ, Baehr W. Identification of a nonsense mutation in the rod photore- 46. Allikmets R, Shroyer NF, Singh N, et al. Mutation of the Stargardt disease
ceptor cGMP phosphodiesterase beta-subunit gene of the rd mouse. Proc Natl gene (ABCR) in age-related macular degeneration. Science 1997;277:1805–7.
Acad Sci U S A 1991;88:8322–6. 47. Allikmets R, Singh N, Sun H, et al. A photoreceptor cell-specific ATP-binding
15. Connell G, Bascom R, Molday L, et al. Photoreceptor peripherin is the normal transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy.
product of the gene responsible for retinal degeneration in the rds mouse. Nat Genet 1997;15:236–46.
Proc Natl Acad Sci U S A 1991;88:723–6. 48. Weng J, Mata NL, Azarian SM, et al. Insights into the function of Rim protein
16. Travis GH, Brennan MB, Danielson PE, et al. Identification of a photoreceptor- in photoreceptors and etiology of Stargardt’s disease from the phenotype in
specific mRNA encoded by the gene responsible for retinal degeneration slow abcr knockout mice. Cell 1999;98:13–23.
(rds). Nature 1989;338:70–3. 49. Molday LL, Rabin AR, Molday RS. ABCR expression in foveal cone photore-
17. Chen J, Makino CL, Peachey NS, et al. Mechanisms of rhodopsin inactivation ceptors and its role in Stargardt macular dystrophy. Nat Genet 2000;25:
in vivo as revealed by a COOH-terminal truncation mutant. Science 257–8.
1995;267:374–7. 50. Radu RA, Mata NL, Nusinowitz S, et al. Treatment with isotretinoin inhibits
18. Lem J, Flannery JG, Li T, et al. Retinal degeneration is rescued in transgenic lipofuscin accumulation in a mouse model of recessive Stargardt’s macular
rd mice by expression of the cGMP phosphodiesterase beta subunit. Proc Natl degeneration. Proc Natl Acad Sci U S A 2003;100:4742–7.
Acad Sci U S A 1992;89:4422–6. 51. Sanyal S, De Ruiter A, Hawkins RK. Development and degeneration of retina
19. Travis GH, Groshan KR, Lloyd M, et al. Complete rescue of photoreceptor in rds mutant mice: light microscopy. J Comp Neurol 1980;194:193–207.
dysplasia and degeneration in transgenic retinal degeneration slow (rds) 52. Sanyal S, Jansen HG. Absence of receptor outer segments in the retina of rds
mice. Neuron 1992;9:113–9. mutant mice. Neurosci Lett 1981;21:23–6.
20. Calvert PD, Krasnoperova NV, Lyubarsky AL, et al. Phototransduction in 53. van Nie R, Ivanyi D, Demant P. A new H-2-linked mutation, rds, causing
transgenic mice after targeted deletion of the rod transducin alpha-subunit. retinal degeneration in the mouse. Tissue Antigens 1978;12:106–8.
Proc Natl Acad Sci U S A 2000;97:13913–8. 54. Young RW. The renewal of rod and cone outer segments in the rhesus
21. Humphries MM, Rancourt D, Farrar GJ, et al. Retinopathy induced in mice monkey. J Cell Biol 1971;49:303–18.
by targeted disruption of the rhodopsin gene. Nat Genet 1997;15:216–9. 55. Young RW. Visual cells and the concept of renewal. Invest Ophthalmol Vis
22. Lem J, Krasnoperova NV, Calvert PD, et al. Morphological, physiological, and Sci 1976;15:700–25.
biochemical changes in rhodopsin knockout mice. Proc Natl Acad Sci U S A 56. Travis GH. Mechanisms of cell death in the inherited retinal degenerations.
1999;96:736–41. Am J Hum Genet 1998;62:503–8.
23. Mendez A, Burns ME, Sokal I, et al. Role of guanylate cyclase-activating 57. Anderson DH, Fisher SK, Steinberg RH. Mammalian cones: disc shedding,
proteins (GCAPs) in setting the flash sensitivity of rod photoreceptors. phagocytosis, and renewal. Invest Ophthalmol Vis Sci 1978;17:117–33.
Proc Natl Acad Sci U S A 2001;98:9948–53. 58. Steinberg RH, Fisher SK, Anderson DH. Disc morphogenesis in vertebrate
24. Xu J, Dodd RL, Makino CL, et al. Prolonged photoresponses in transgenic photoreceptors. J Comp Neurol 1980;190:501–8.
mouse rods lacking arrestin. Nature 1997;389:505–9. 59. Chuang JZ, Zhao Y, Sung CH. SARA-regulated vesicular targeting underlies
25. von Greeff R. Mikroskopische Anatomie des Sehnerven und der Netzhaut. In: formation of the light-sensing organelle in mammalian rods. Cell 2007;130:
Graefe u. Saemisch Handbuch d. Ges. Augenheilkunde, 2. Aufl., 1, Teil, bd 1, 535–47.
Kap. V. Leipzig: 1900. 60. Nakatani K, Yau KW. Calcium and magnesium fluxes across the plasma
26. von Greeff R. A guide to the microscopic examination of the eye. Philadelphia: membrane of the toad rod outer segment. J Physiol 1988;395:695–729.
Blackiston; 1902. 61. Perry RJ, McNaughton PA. Response properties of cones from the retina of
27. Hendrickson A, Drucker D. The development of parafoveal and mid- the tiger salamander. J Physiol 1991;433:561–87.
peripheral human retina. Behav Brain Res 1992;49:21–31. 62. Picones A, Korenbrot JI. Permeability and interaction of Ca2+ with cGMP-
28. Detwiler SR. Vertebrate photoreceptors. New York: Macmillan; 1943. gated ion channels differ in retinal rod and cone photoreceptors. Biophys J
29. Liang Y, Fotiadis D, Maeda T, et al. Rhodopsin signaling and organization in 1995;69:120–7.
heterozygote rhodopsin knockout mice. J Biol Chem 2004;279:48189–96. 63. Pugh Jr EN, Lamb TD. Amplification and kinetics of the activation steps in
30. Nickell S, Park PS, Baumeister W, et al. Three-dimensional architecture of phototransduction. Biochim Biophys Acta 1993;1141:111–49.
murine rod outer segments determined by cryoelectron tomography. J Cell 64. Nakatani K, Yau KW. Guanosine 3’,5’-cyclic monophosphate-activated con-
Biol 2007;177:917–25. ductance studied in a truncated rod outer segment of the toad. J Physiol
31. Poo M, Cone RA. Lateral diffusion of rhodopsin in the photoreceptor mem- 1988;395:731–53.
brane. Nature 1974;247:438–41. 65. Ruiz ML, Karpen JW. Single cyclic nucleotide-gated channels locked in dif-
32. Liang Y, Fotiadis D, Filipek S, et al. Organization of the G protein-coupled ferent ligand-bound states. Nature 1997;389:389–92.
receptors rhodopsin and opsin in native membranes. J Biol Chem 2003;278: 66. Yau KW, Nakatani K. Electrogenic Na-Ca exchange in retinal rod outer
21655–62. segment. Nature 1984;311:661–3.
33. Bayburt TH, Vishnivetskiy SA, McLean MA, et al. Monomeric rhodopsin is 67. Kaupp UB, Niidome T, Tanabe T, et al. Primary structure and functional
sufficient for normal rhodopsin kinase (GRK1) phosphorylation and arrestin-1 expression from complementary DNA of the rod photoreceptor cyclic GMP-
binding. J Biol Chem 2011;286:1420–8. gated channel. Nature 1989;342:762–6.
68. Pittler SJ, Lee AK, Altherr MR, et al. Primary structure and chromosomal 103. Hu G, Zhang Z, Wensel TG. Activation of RGS9–1GTPase acceleration by its
localization of human and mouse rod photoreceptor cGMP-gated cation membrane anchor, R9AP. J Biol Chem 2003;278:14550–4.
channel. J Biol Chem 1992;267:6257–62. 104. Lishko PV, Martemyanov KA, Hopp JA, et al. Specific binding of RGS9-Gbeta
69. Zheng J, Trudeau MC, Zagotta WN. Rod cyclic nucleotide-gated channels 5L to protein anchor in photoreceptor membranes greatly enhances its cata- 357
have a stoichiometry of three CNGA1 subunits and one CNGB1 subunit. lytic activity. J Biol Chem 2002;277:24376–81.
Neuron 2002;36:891–6. 105. Nishiguchi KM, Sandberg MA, Kooijman AC, et al. Defects in RGS9 or its
70. Zhong H, Molday LL, Molday RS, et al. The heteromeric cyclic nucleotide- anchor protein R9AP in patients with slow photoreceptor deactivation.

Chapter 14
gated channel adopts a 3A:1B stoichiometry. Nature 2002;420:193–8. Nature 2004;427:75–8.
71. Molday LL, Cook NJ, Kaupp UB, et al. The cGMP-gated cation channel of 106. Cowan CW, Fariss RN, Sokal I, et al. High expression levels in cones of RGS9,
bovine rod photoreceptor cells is associated with a 240-kDa protein exhibiting the predominant GTPase accelerating protein of rods. Proc Natl Acad Sci
immunochemical cross-reactivity with spectrin. J Biol Chem 1990;265: U S A 1998;95:5351–6.
18690–5. 107. Gorczyca WA, Gray-Keller MP, Detwiler PB, et al. Purification and physio-
72. Molday RS, Molday LL, Dose A, et al. The cGMP-gated channel of the rod logical evaluation of a guanylate-cyclase activating protein from retinal rods.
photoreceptor cell characterization and orientation of the amino terminus. Proc Natl Acad Sci U S A 1994;91:4014–8.
J Biol Chem 1991;266:21917–22. 108. Koch KW, Stryer L. Highly cooperative feedback-control of retinal rod
73. Bareil C, Hamel CP, Delague V, et al. Segregation of a mutation in CNGB1 guanylate-cyclase by calcium-ions. Nature 1988;334:64–6.
encoding the beta-subunit of the rod cGMP-gated channel in a family with 109. Gorczyca WA, Polans AS, Surgucheva IG, et al. Guanylyl cyclase-activating

Structure and Function of Rod and Cone Photoreceptors


autosomal recessive retinitis pigmentosa. Hum Genet 2001;108:328–34. protein – a calcium-sensitive regulator of phototransduction. J Biol Chem
74. Dryja TP, Finn JT, Peng YW, et al. Mutations in the gene encoding the alpha 1995;270:22029–36.
subunit of the rod cGMP-gated channel in autosomal recessive retinitis pig- 110. Burns ME, Mendez A, Chen J, et al. Dynamics of cyclic GMP synthesis in
mentosa. Proc Natl Acad Sci U S A 1995;92:10177–81. retinal rods. Neuron 2002;36:81–91.
75. Peng C, Rich ED, Varnum MD. Subunit configuration of heteromeric cone 111. Detwiler PB, Gray-Keller MP. The mechanisms of vertebrate light adaptation:
cyclic nucleotide-gated channels. Neuron 2004;42:401–10. speeded recovery versus slowed activation. Curr Opin Neurobiol 1996;6:
76. Kohl S, Marx T, Giddings I, et al. Total colourblindness is caused by mutations 440–4.
in the gene encoding the alpha-subunit of the cone photoreceptor cGMP-gated 112. Krispel CM, Chen CK, Simon MI, et al. Novel form of adaptation in mouse
cation channel. Nat Genet 1998;19:257–9. retinal rods speeds recovery of phototransduction. J Gen Physiol 2003;122:
77. Kohl S, Varsanyi B, Antunes GA, et al. CNGB3 mutations account for 50% of 703–12.
all cases with autosomal recessive achromatopsia. Eur J Hum Genet 2005;13: 113. Woodruff ML, Sampath AP, Matthews HR, et al. Measurement of cytoplasmic
302–8. calcium concentration in the rods of wild-type and transducin knock-out
78. Lytton J. Na+/Ca2+ exchangers: three mammalian gene families control Ca2+ mice. J Physiol 2002;542:843–54.
transport. Biochem J 2007;406:365–82. 114. Matthews HR, Murphy RLW, Fain GL, et al. Photoreceptor light adaptation
79. Hsu SC, Molday RS. Glycolytic enzymes and a GLUT-1 glucose transporter is mediated by cytoplasmic calcium concentration. Nature 1988;334:67–9.
in the outer segments of rod and cone photoreceptor cells. J Biol Chem 115. Nakatani K, Yau K-W. Calcium and light adaptation in retinal rods and cones.
1991;266:21745–52. Nature 1988;334:69–71.
80. Fliesler SJ, Anderson RE. Chemistry and metabolism of lipids in the vertebrate 116. Fain GL, Matthews HR, Cornwall MC, et al. Adaptation in vertebrate photo-
retina. Prog Lipid Res 1983;22:79–131. receptors. Physiol Rev 2001;81:117–51.
81. Tinoco J. Dietary requirements and functions of alpha-linolenic acid in 117. Koutalos Y, Yau K-W. Regulation of sensitivity in vertebrate rod photorecep-
animals. Prog Lipid Res 1982;21:1–45. tors by calcium. TINS 1996;19:73–81.
82. Anderson RE, Maude MB, Bok D. Low docosahexaenoic acid levels in rod 118. Pugh Jr EN, Nikonov S, Lamb TD. Molecular mechanisms of vertebrate
outer segment membranes of mice with rds/peripherin and P216L peripherin photoreceptor light adaptation. Curr Opin Neurobiol 1999;9:410–8.
mutations. Invest Ophthalmol Vis Sci 2001;42:1715–20. 119. Sakurai K, Chen J, Kefalov VJ. Role of guanylyl cyclase modulation in mouse
83. Fung BK, Hurley JB, Stryer L. Flow of information in the light-triggered cyclic cone phototransduction. J Neurosci 2011;31:7991–8000.
nucleotide cascade of vision. Proc Natl Acad Sci U S A 1981;78:152–6. 120. Chen CK, Inglese J, Lefkowitz RJ, et al. Ca(2+)-dependent interaction of recov-
84. Yee R, Liebman PA. Light-activated phosphodiesterase of the rod outer erin with rhodopsin kinase. J Biol Chem 1995;270:18060–6.
segment. Kinetics and parameters of activation and deactivation. J Biol Chem 121. Kawamura S. Rhodopsin phosphorylation as a mechanism of cyclic GMP
1978;253:8902–9. phosphodiesterase regulation by S-modulin. Nature 1993;362:855–7.
85. Wilden U, Hall SW, Kühn H. Phosphodiesterase activation by photoexcited 122. Erickson MA, Lagnado L, Zozulya S, et al. The effect of recombinant recoverin
rhodopsin is quenched when rhodopsin is phosphorylated and binds the on the photoresponse of truncated rod photoreceptors. Proc Natl Acad Sci
intrinsic 48-kDa protein of rod outer segments. Proc Natl Acad Sci USA U S A 1998;95:6474–9.
1986;83:1174–8. 123. Gray-Keller MP, Polans AS, Palczewski K, et al. The effect of recoverin-like
86. Wilden U, Kuhn H. Light-dependent phosphorylation of rhodopson: number calcium-binding proteins on the photoresponse of retinal rods. Neuron
of phosphorylation sites. Biochemistry 1982;21:3014–22. 1993;10:523–31.
87. Ohguro H, Johnson RS, Ericsson LH, et al. Control of rhodopsin multiple 124. Makino CL, Dodd RL, Chen J, et al. Recoverin regulates light-dependent
phosphorylation. Biochemistry 1994;33:1023–8. phosphodiesterase activity in retinal rods. J Gen Physiol 2004;123:729–41.
88. Ohguro H, Palczewski K, Ericsson LH, et al. Sequential phosphorylation of 125. Chen CK, Woodruff ML, Chen FS, et al. Background light produces a
rhodopsin at multiple sites. Biochemistry 1993;32:5718–24. recoverin-dependent modulation of activated-rhodopsin lifetime in mouse
89. Ohguro H, Van Hooser JP, Milam AH, et al. Rhodopsin phosphorylation and rods. J Neurosci 2010;30:1213–20.
dephosphorylation in vivo. J Biol Chem 1995;270:14259–62. 126. Matthews HR, Sampath AP. Photopigment quenching is Ca2+ dependent and
90. Mendez A, Burns ME, Roca A, et al. Rapid and reproducible deactivation of controls response duration in salamander L-cone photoreceptors. J Gen
rhodopsin requires multiple phosphorylation sites. Neuron 2000;28:153–64. Physiol 2010;135:355–66.
91. Chen CK, Burns ME, Spencer M, et al. Abnormal photoresponses and light- 127. Hsu Y-T, Molday RS. Modulation of the cGMP-gated channel of rod photo-
induced apoptosis in rods lacking rhodopsin kinase. Proc Natl Acad Sci receptor cells by calmodulin. Nature 1993;361:76–9.
U S A 1999;96:3718–22. 128. Grunwald ME, Yu W-P, Yu H-H, et al. Identification of a domain on the
92. Gurevich VV, Gurevich EV. The molecular acrobatics of arrestin activation. ß-subunit of the rod cGMP-gated cation channel that mediates inhibition by
Trends Pharmacol Sci 2004;25:105–11. calcium-calmodulin. J Biol Chem 1998;273:9148–57.
93. Hirsch JA, Schubert C, Gurevich VV, et al. The 2.8 A crystal structure of visual 129. Weitz D, Zoche M, Müller F, et al. Calmodulin controls the rod photoreceptor
arrestin: a model for arrestin’s regulation. Cell 1999;97:257–69. CNG channel through an unconventional binding site in the N-terminus of
94. Vishnivetskiy SA, Hosey MM, Benovic JL, et al. Mapping the arrestin–receptor the ß-subunit. EMBO J 1998;17:2273–84.
interface. Structural elements responsible for receptor specificity of arrestin 130. Chen J, Woodruff ML, Wang T, et al. Channel modulation and the mechanism
proteins. J Biol Chem 2004;279:1262–8. of light adaptation in mouse rods. J Neurosci 2010;30:16232–40.
95. Vishnivetskiy SA, Paz CL, Schubert C, et al. How does arrestin respond to the 131. Rebrik TI, Korenbrot JI. In intact mammalian photoreceptors, Ca2+-dependent
phosphorylated state of rhodopsin? J Biol Chem 1999;274:11451–4. modulation of cGMP-gated ion channels is detectable in cones but not in rods.
96. Heck M, Hofmann K. G-protein effector coupling – a real time light-scattering J Gen Physiol 2004;123:63–75.
assay for transducin phosphodiesterase interaction. Biochemistry 1993;32: 132. Sampath AP, Matthews HR, Cornwall MC, et al. Light-dependent changes in
8220–7. outer segment free-Ca2+ concentration in salamander cone photoreceptors.
97. He W, Cowan CW, Wensel TG. RGS9, a GTPase accelerator for phototrans- J Gen Physiol 1999;113:267–77.
duction. Neuron 1998;20:95–102. 133. Rebrik TI, Korenbrot JI. In intact cone photoreceptors, a Ca2+-dependent, dif-
98. Slepak VZ, Artemyev NO, Zhu Y, et al. An effector site that stimulates fusible factor modulates the cGMP-gated ion channels differently than in
G-protein GTPase in photoreceptors. J Biol Chem 1995;270:14319–24. rods. J Gen Physiol 1998;112:537–48.
99. Tsang SH, Burns ME, Calvert PD, et al. Role for the target enzyme in deactiva- 134. Broekhuyse RM, Janssen AP, Tolhuizen EF. Effect of light-adaptation on the
tion of photoreceptor G protein in vivo. Science 1998;282:117–21. binding of 48-kDa protein (S-antigen) to photoreceptor cell membranes.
100. Chen CK, Burns ME, He W, et al. Slowed recovery of rod photoresponse in Curr Eye Res 1987;6:607–10.
mice lacking the GTPase accelerating protein RGS9–1. Nature 2000;403: 135. Broekhuyse RM, Tolhuizen EF, Janssen AP, et al. Light induced shift and
557–60. binding of S-antigen in retinal rods. Curr Eye Res 1985;4:613–8.
101. Krispel CM, Chen CK, Simon MI, et al. Prolonged photoresponses and defec- 136. Philp NJ, Chang W, Long K. Light-stimulated protein movement in rod
tive adaptation in rods of Gbeta5-/- mice. J Neurosci 2003;23:6965–71. photoreceptor cells of the rat retina. FEBS Lett 1987;225:127–32.
102. Hu G, Wensel TG. R9AP, a membrane anchor for the photoreceptor GTPase 137. Whelan JP, McGinnis JF. Light-dependent subcellular movement of photore-
accelerating protein, RGS9–1. Proc Natl Acad Sci U S A 2002;99:9755–60. ceptor proteins. J Neurosci Res 1988;20:263–70.
138. McGinnis JF, Whelan JP, Donoso LA. Transient, cyclic changes in mouse 172. Williams DS, Linberg KA, Vaughan DK, et al. Disruption of microfilament
visual cell gene products during the light-dark cycle. J Neurosci Res 1992; organization and deregulation of disk membrane morphogenesis by cytocha-
31:584–90. lasin D in rod and cone photoreceptors. J Comp Neurol 1988;272:161–76.
358 139. Calvert PD, Strissel KJ, Schiesser WE, et al. Light-driven translocation of 173. Adato A, Weil D, Kalinski H, et al. Mutation profile of all 49 exons of the
signaling proteins in vertebrate photoreceptors. Trends Cell Biol 2006;16: human myosin VIIA gene, and haplotype analysis, in Usher 1B families from
560–8. diverse origins. Am J Hum Genet 1997;61:813–21.
140. Sokolov M, Lyubarsky AL, Strissel KJ, et al. Massive light-driven translocation 174. Weil D, Blanchard S, Kaplan J, et al. Defective myosin VIIA gene responsible
of transducin between the two major compartments of rod cells: a novel for Usher syndrome type 1B. Nature 1995;374:60–1.
Section 1

mechanism of light adaptation. Neuron 2002;34:95–106. 175. Weil D, Kussel P, Blanchard S, et al. The autosomal recessive isolated deaf-
141. Rosenzweig DH, Nair KS, Wei J, et al. Subunit dissociation and diffusion ness, DFNB2, and the Usher 1B syndrome are allelic defects of the myosin-
determine the subcellular localization of rod and cone transducins. J Neurosci VIIA gene. Nat Genet 1997;16:191–3.
2007;27:5484–94. 176. Pierce EA, Quinn T, Meehan T, et al. Mutations in a gene encoding a new
142. Lobanova ES, Finkelstein S, Song H, et al. Transducin translocation in rods is oxygen-regulated photoreceptor protein cause dominant retinitis pigmentosa.
triggered by saturation of the GTPase-activating complex. J Neurosci Nat Genet 1999;22:248–54.
2007;27:1151–60. 177. Gao J, Cheon K, Nusinowitz S, et al. Progressive photoreceptor degeneration,
143. Zhang X, Wensel TG, Kraft TW. GTPase regulators and photoresponses in outer segment dysplasia, and rhodopsin mislocalization in mice with targeted
Anatomy and Physiology

cones of the eastern chipmunk. J Neurosci 2003;23:1287–97. disruption of the retinitis pigmentosa-1 (Rp1) gene. Proc Natl Acad Sci U S A
Basic Science and Translation to Therapy

144. Lobanova ES, Herrmann R, Finkelstein S, et al. Mechanistic basis for the 2002;99:5698–703.
failure of cone transducin to translocate: why cones are never blinded by light. 178. Liu Q, Lyubarsky A, Skalet JH, et al. RP1 is required for the correct stacking
J Neurosci 2010;30:6815–24. of outer segment discs. Invest Ophthalmol Vis Sci 2003;44:4171–83.
145. Chen J, Wu M, Sezate SA, et al. Light threshold-controlled cone alpha- 179. Liu Q, Zuo J, Pierce EA. The retinitis pigmentosa 1 protein is a photoreceptor
transducin translocation. Invest Ophthalmol Vis Sci 2007;48:3350–5. microtubule-associated protein. J Neurosci 2004;24:6427–36.
146. Nair KS, Hanson SM, Mendez A, et al. Light-dependent redistribution of 180. Meindl A, Dry K, Herrmann K, et al. A gene (RPGR) with homology to the
arrestin in vertebrate rods is an energy-independent process governed by RCC1 guanine nucleotide exchange factor is mutated in X-linked retinitis
protein-protein interactions. Neuron 2005;46:555–67. pigmentosa (RP3). Nat Genet 1996;13:35–42.
147. Mears AJ, Kondo M, Swain PK, et al. Nrl is required for rod photoreceptor 181. Mavlyutov TA, Zhao H, Ferreira PA. Species-specific subcellular localization
development. Nat Genet 2001;29:447–52. of RPGR and RPGRIP isoforms: implications for the phenotypic variability of
148. Kefalov V, Fu Y, Marsh-Armstrong N, et al. Role of visual pigment properties congenital retinopathies among species. Hum Mol Genet 2002;11:1899–907.
in rod and cone phototransduction. Nature 2003;425:526–31. 182. Hong DH, Pawlyk B, Sokolov M, et al. RPGR isoforms in photoreceptor con-
149. Shi G, Yau KW, Chen J, et al. Signaling properties of a short-wave cone visual necting cilia and the transitional zone of motile cilia. Invest Ophthalmol Vis
pigment and its role in phototransduction. J Neurosci 2007;27:10084–93. Sci 2003;44:2413–21.
150. Deng WT, Sakurai K, Liu J, et al. Functional interchangeability of rod and cone 183. Hong DH, Pawlyk BS, Shang J, et al. A retinitis pigmentosa GTPase regulator
transducin alpha-subunits. Proc Natl Acad Sci U S A 2009;106:17681–6. (RPGR)-deficient mouse model for X-linked retinitis pigmentosa (RP3).
151. Chen CK, Woodruff ML, Chen FS, et al. Replacing the rod with the cone Proc Natl Acad Sci U S A 2000;97:3649–54.
transducin subunit decreases sensitivity and accelerates response decay. 184. Trojan P, Krauss N, Choe HW, et al. Centrins in retinal photoreceptor cells:
J Physiol 2010;588:3231–41. regulators in the connecting cilium. Prog Retin Eye Res 2008;27:237–59.
152. Okawa H, Sampath AP, Laughlin SB, et al. ATP consumption by mammalian 185. Yang J, Liu X, Yue G, et al. Rootletin, a novel coiled-coil protein, is a structural
rod photoreceptors in darkness and in light. Curr Biol 2008;18:1917–21. component of the ciliary rootlet. J Cell Biol 2002;159:431–40.
153. Trachtenberg MC, Packey DJ, Sweeney T. In vivo functioning of the Na+, K+- 186. Hagstrom SA, Adamian M, Scimeca M, et al. A role for the Tubby-like protein
activated ATPase. Curr Top Cell Regul 1981;19:159–217. 1 in rhodopsin transport. Invest Ophthalmol Vis Sci 2001;42:1955–62.
154. Karan S, Zhang H, Li S, et al. A model for transport of membrane-associated 187. Lewis CA, Batlle IR, Batlle KG, et al. Tubby-like protein 1 homozygous splice-
phototransduction polypeptides in rod and cone photoreceptor inner site mutation causes early-onset severe retinal degeneration. Invest Ophthal-
segments. Vision Res 2008;48:442–52. mol Vis Sci 1999;40:2106–14.
155. Wensel TG. Signal transducing membrane complexes of photoreceptor outer 188. Rattner A, Chen J, Nathans J. Proteolytic shedding of the extracellular domain
segments. Vision Res 2008;48:2052–61. of photoreceptor cadherin. Implications for outer segment assembly. J Biol
156. Ames JB, Ishima R, Tanaka T, et al. Molecular mechanics of calcium-myristoyl Chem 2004;279:42202–10.
switches. Nature 1997;389:198–202. 189. Kubota R, Noda S, Wang Y, et al. A novel myosin-like protein (myocilin)
157. Malanson KM, Lem J. Rhodopsin-mediated retinitis pigmentosa. Prog Mol expressed in the connecting cilium of the photoreceptor: molecular
Biol Transl Sci 2009;88:1–31. cloning, tissue expression, and chromosomal mapping. Genomics 1997;41:
158. Kaushal S, Khorana HG. Structure and function in rhodopsin. 7. Point muta- 360–9.
tions associated with autosomal dominant retinitis pigmentosa. Biochemistry 190. Schmitz F, Konigstorfer A, Sudhof TC. RIBEYE, a component of synaptic
1994;33:6121–8. ribbons: a protein’s journey through evolution provides insight into synaptic
159. Mendes HF, van der Spuy J, Chapple JP, et al. Mechanisms of cell death in ribbon function. Neuron 2000;28:857–72.
rhodopsin retinitis pigmentosa: implications for therapy. Trends Mol Med 191. Jackman SL, Choi SY, Thoreson WB, et al. Role of the synaptic ribbon in
2005;11:177–85. transmitting the cone light response. Nat Neurosci 2009;12:303–10.
160. Sung CH, Davenport CM, Nathans J. Rhodopsin mutations responsible for 192. Snellman J, Mehta B, Babai N, et al. Acute destruction of the synaptic ribbon
autosomal dominant retinitis pigmentosa. Clustering of functional classes reveals a role for the ribbon in vesicle priming. Nat Neurosci. 2011 14:
along the polypeptide chain. J Biol Chem 1993;268:26645–9. 1135–41.
161. Concepcion F, Mendez A, Chen J. The carboxyl-terminal domain is essential 193. Carter-Dawson LD, LaVail MM. Rods and cones in the mouse retina. I. Struc-
for rhodopsin transport in rod photoreceptors. Vision Res 2002;42:417–26. tural analysis using light and electron microscopy. J Comp Neurol 1979;
162. Li T, Snyder WK, Olsson JE, et al. Transgenic mice carrying the dominant 188:245–62.
rhodopsin mutation P347S: evidence for defective vectorial transport of rho- 194. Tsukamoto Y, Morigiwa K, Ueda M, et al. Microcircuits for night vision in
dopsin to the outer segments. Proc Natl Acad Sci U S A 1996;93:14176–81. mouse retina. J Neurosci 2001;21:8616–23.
163. Sung CH, Makino C, Baylor D, et al. A rhodopsin gene mutation responsible 195. West RW, Dowling JE. Anatomical evidence for cone and rod-like receptors
for autosomal dominant retinitis pigmentosa results in a protein that is defec- in the gray squirrel, ground squirrel, and prairie dog retinas. J Comp Neurol
tive in localization to the photoreceptor outer segment. J Neurosci 1994;14: 1975;159:439–60.
5818–33. 196. Chun MH, Grunert U, Martin PR, et al. The synaptic complex of cones in the
164. Deretic D. A role for rhodopsin in a signal transduction cascade that regulates fovea and in the periphery of the macaque monkey retina. Vision Res
membrane trafficking and photoreceptor polarity. Vision Res 2006;46: 1996;36:3383–95.
4427–33. 197. Hopkins JM, Boycott BB. The cone synapses of cone bipolar cells of primate
165. Deretic D, Williams AH, Ransom N, et al. Rhodopsin C terminus, the site of retina. J Neurocytol 1997;26:313–25.
mutations causing retinal disease, regulates trafficking by binding to ADP- 198. Koschak A, Reimer D, Walter D, et al. Cav1.4alpha1 subunits can form slowly
ribosylation factor 4 (ARF4). Proc Natl Acad Sci U S A 2005;102:3301–6. inactivating dihydropyridine-sensitive L-type Ca2+ channels lacking Ca2+-
166. Tam BM, Moritz OL, Papermaster DS. The C terminus of peripherin/rds dependent inactivation. J Neurosci 2003;23:6041–9.
participates in rod outer segment targeting and alignment of disk incisures. 199. Wahl-Schott C, Baumann L, Cuny H, et al. Switching off calcium-dependent
Mol Biol Cell 2004;15:2027–37. inactivation in L-type calcium channels by an autoinhibitory domain.
167. Huttl S, Michalakis S, Seeliger M, et al. Impaired channel targeting and retinal Proc Natl Acad Sci U S A 2006;103:15657–62.
degeneration in mice lacking the cyclic nucleotide-gated channel subunit 200. Haeseleer F, Imanishi Y, Maeda T, et al. Essential role of Ca2+-binding protein
CNGB1. J Neurosci 2005;25:130–8. 4, a Cav1.4 channel regulator, in photoreceptor synaptic function. Nat
168. Zhang H, Li S, Doan T, et al. Deletion of PrBP/delta impedes transport of Neurosci 2004;7:1079–87.
GRK1 and PDE6 catalytic subunits to photoreceptor outer segments. Proc Natl 201. Morgans CW, Bayley PR, Oesch NW, et al. Photoreceptor calcium channels:
Acad Sci U S A 2007;104:8857–62. insight from night blindness. Vis Neurosci 2005;22:561–8.
169. Zhang H, Constantine R, Vorobiev S, et al. UNC119 is required for G protein 202. Huang SP, Brown BM, Craft CM. Visual Arrestin 1 acts as a modulator for
trafficking in sensory neurons. Nat Neurosci 2011;14:874–80. N-ethylmaleimide-sensitive factor in the photoreceptor synapse. J Neurosci
170. Insinna C, Besharse JC. Intraflagellar transport and the sensory outer segment 2010;30:9381–91.
of vertebrate photoreceptors. Dev Dyn 2008;237:1982–92. 203. Sampath AP, Strissel KJ, Elias R, et al. Recoverin improves rod-mediated
171. Williams DS. Transport to the photoreceptor outer segment by myosin VIIa vision by enhancing signal transmission in the mouse retina. Neuron 2005;46:
and kinesin II. Vision Res 2002;42:455–62. 413–20.
204. Strissel KJ, Lishko PV, Trieu LH, et al. Recoverin undergoes light-dependent 227. Chen J, Simon MI, Matthes MT, et al. Increased susceptibility to light damage
intracellular translocation in rod photoreceptors. J Biol Chem 2005;280: in an arrestin knockout mouse model of Oguchi disease (stationary night
29250–5. blindness). Invest Ophthalmol Vis Sci 1999;40:2978–82.
205. Higgs MH, Lukasiewicz PD. Activation of group II metabotropic glutamate 228. Dryja TP, Hahn LB, Reboul T, et al. Missense mutation in the gene encoding 359
receptors inhibits glutamate release from salamander retinal photoreceptors. the alpha subunit of rod transducin in the Nougaret form of congenital
Vis Neurosci 2002;19:275–81. stationary night blindness. Nat Genet 1996;13:358–60.
206. Koulen P, Kuhn R, Wassle H, et al. Modulation of the intracellular calcium 229. McLaughlin ME, Sandberg MA, Berson EL, et al. Recessive mutations in the

Chapter 14
concentration in photoreceptor terminals by a presynaptic metabotropic gene encoding the beta-subunit of rod phosphodiesterase in patients with
glutamate receptor. Proc Natl Acad Sci U S A 1999;96:9909–14. retinitis pigmentosa. Nat Genet 1993;4:130–4.
207. Duncan JL, Yang H, Doan T, et al. Scotopic visual signaling in the mouse 230. Farber DB, Lolley RN. Cyclic guanosine monophosphate: elevation in degen-
retina is modulated by high-affinity plasma membrane calcium extrusion. erating photoreceptor cells of the C3H mouse retina. Science 1974;186:
J Neurosci 2006;26:7201–11. 449–51.
208. Szikra T, Barabas P, Bartoletti TM, et al. Calcium homeostasis and cone signal- 231. Lolley RN, Farber DB, Rayborn ME, et al. Cyclic GMP accumulation causes
ing are regulated by interactions between calcium stores and plasma mem- degeneration of photoreceptor cells: simulation of an inherited disease.
brane ion channels. PLoS One 2009;4:e6723. Science 1977;196:664–6.
209. Szikra T, Cusato K, Thoreson WB, et al. Depletion of calcium stores regulates 232. Fesenko EE, Kolesnikov SS, Lyubarsky AL. Induction by cyclic GMP of cat-
calcium influx and signal transmission in rod photoreceptors. J Physiol ionic conductance in plasma membrane of retinal rod outer segment. Nature

Structure and Function of Rod and Cone Photoreceptors


2008;586:4859–75. 1985;313:310–3.
210. Thoreson WB. Kinetics of synaptic transmission at ribbon synapses of rods 233. Yang RB, Foster DC, Garbers DL, et al. Two membrane forms of guanylyl
and cones. Mol Neurobiol 2007;36:205–23. cyclase found in the eye. Proc Natl Acad Sci U S A 1995;92:602–6.
211. Choi SY, Jackman S, Thoreson WB, et al. Light regulation of Ca2+ in the cone 234. Lowe DG, Dizhoor AM, Liu K, et al. Cloning and expression of a second
photoreceptor synaptic terminal. Vis Neurosci 2008;25:693–700. photoreceptor-specific membrane retina guanylyl cyclase (RetGC), RetGC-2.
212. Sheng Z, Choi SY, Dharia A, et al. Synaptic Ca2+ in darkness is lower in rods Proc Natl Acad Sci U S A 1995;92:5535–9.
than cones, causing slower tonic release of vesicles. J Neurosci 2007;27: 235. Shyjan AW, de Sauvage FJ, Gillett NA, et al. Molecular cloning of a retina-
5033–42. specific membrane guanylyl cyclase. Neuron 1992;9:727–37.
213. Gal A, Apfelstedt-Sylla E, Janecke AR, et al. Rhodopsin mutations in 236. Goraczniak RM, Duda T, Sitaramayya A, et al. Structural and functional
inherited retinal dystrophies and dysfunctions. Prog Ret Eye Res 1997;16: characterization of the rod outer segment membrane guanylate cyclase.
51–79. Biochem J 1994;302:455–61.
214. Lin JH, Li H, Yasumura D, et al. IRE1 signaling affects cell fate during the 237. Duda T, Goraczniak R, Surgucheva I, et al. Calcium modulation of bovine
unfolded protein response. Science 2007;318:944–9. photoreceptor guanylate cyclase. Biochemistry 1996;35:8478–82.
215. Colley NJ, Cassill JA, Baker EK, et al. Defective intracellular transport is the 238. Laura RP, Dizhoor AM, Hurley JB. The membrane guanylyl cyclase, retinal
molecular basis of rhodopsin-dependent dominant retinal degeneration. guanylyl cyclase-1, is activated through its intracellular domain. J Biol Chem
Proc Natl Acad Sci U S A 1995;92:3070–4. 1996;271:11646–51.
216. Deretic D, Puleo-Scheppke B, Trippe C. Cytoplasmic domain of rhodopsin 239. Dizhoor AM, Lowe DG, Olshevskaya EV, et al. The human photoreceptor
is essential for post-Golgi vesicle formation in a retinal cell-free system. membrane guanylyl cyclase, retGC, is present in outer segments and is
J Biol Chem 1996;271:2279–86. regulated by calcium and a soluble activator. Neuron 1994;12:1345–52.
217. Deretic D, Schmerl S, Hargrave PA, et al. Regulation of sorting and post-Golgi 240. Yang RB, Garbers DL. Two eye guanylyl cyclases are expressed in the
trafficking of rhodopsin by its C-terminal sequence QVS(A)PA. Proc Natl same photoreceptor cells and form homomers in preference to heteromers.
Acad Sci U S A 1998;95:10620–5. J Biol Chem 1997;272:13738–42.
218. Chuang JZ, Sung CH. The cytoplasmic tail of rhodopsin acts as a novel 241. Gorczyca WA, Polans AS, Surgucheva IG, et al. Guanylyl cyclase activating
apical sorting signal in polarized MDCK cells. J Cell Biol 1998;142: protein. A calcium-sensitive regulator of phototransduction. J Biol Chem
1245–56. 1995;270:22029–36.
219. Rao VR, Oprian DD. Activating mutations of rhodopsin and other G 242. Yang RB, Robinson SW, Xiong WH, et al. Disruption of a retinal guanylyl
protein-coupled receptors. Annu Rev Biophys Biomol Struct 1996;25: cyclase gene leads to cone-specific dystrophy and paradoxical rod behavior.
287–314. J Neurosci 1999;19:5889–97.
220. Robinson PR, Cohen GB, Zhukovsky EA, et al. Constitutively active mutants 243. Semple-Rowland SL, Lee NR, Van Hooser JP, et al. A null mutation in the
of rhodopsin. Neuron 1992;9:719–25. photoreceptor guanylate cyclase gene causes the retinal degeneration chicken
221. Rao VR, Cohen GB, Oprian DD. Rhodopsin mutation G90D and a molecular phenotype. Proc Natl Acad Sci U S A 1998;95:1271–6.
mechanism for congenital night blindness. Nature 1994;367:639–42. 244. Payne AM, Downes SM, Bessant DA, et al. A mutation in guanylate cyclase
222. Sieving PA, Richards JE, Naarendorp F, et al. Dark-light: model for night- activator 1A (GUCA1A) in an autosomal dominant cone dystrophy pedigree
blindness from the human rhodopsin Gly-90–>Asp mutation. Proc Natl Acad mapping to a new locus on chromosome 6p21.1. Hum Mol Genet
Sci U S A 1995;92:880–4. 1998;7:273–7.
223. Li T, Franson WK, Gordon JW, et al. Constitutive activation of phototransduc- 245. Dizhoor AM, Boikov SG, Olshevskaya EV. Constitutive activation of photo-
tion by K296E opsin is not a cause of photoreceptor degeneration. Proc Natl receptor guanylate cyclase by Y99C mutant of GCAP-1. Possible role in
Acad Sci U S A 1995;92:3551–5. causing human autosomal dominant cone degeneration. J Biol Chem 1998;
224. Chen J, Shi G, Concepcion FA, et al. Stable rhodopsin/arrestin complex leads 273:17311–4.
to retinal degeneration in a transgenic mouse model of autosomal dominant 246. Sokal I, Li N, Surgucheva I, et al. GCAP1 (Y99C) mutant is constitutively
retinitis pigmentosa. J Neurosci 2006;26:11929–37. active in autosomal dominant cone dystrophy. Mol Cell 1998;2:129–33.
225. Fuchs S, Nakazawa M, Maw M, et al. A homozygous 1-base pair deletion in 247. Paquet-Durand F, Beck S, Michalakis S, et al. A key role for cyclic nucleotide
the arrestin gene is a frequent cause of Oguchi disease in Japanese. Nat Genet gated (CNG) channels in cGMP-related retinitis pigmentosa. Hum Mol Genet
1995;10:360–2. 2011;20:941–7.
226. Yamamoto S, Sippel KC, Berson EL, et al. Defects in the rhodopsin kinase 248. Andrade AL, Melich K, Whatley GG, et al. Cyclic nucleotide-gated channel
gene in the Oguchi form of stationary night blindness. Nat Genet 1997;15: block by hydrolysis-resistant tetracaine derivatives. J Med Chem 2011;54:
175–8. 4904–12.
Section 1 Anatomy and Physiology
For additional online content visit http://www.expertconsult.com

Chapter Function and Anatomy of the Mammalian Retina


15  Ronald G. Gregg, Maureen A. McCall, Stephen C. Massey

The vertebrate retina forms a thin sheet of neural tissue at the of information that are transmitted simultaneously throughout
back of the eye that converts light to an electrical signal. The the visual system. It is the purpose of this chapter to describe
neural retina is approximately 100–200  µm thick, depending on the functional anatomy of the retina that leads to the formation
the species, and represents a triumph of miniaturization.1 The of some of these independent channels of visual information.
retina is a spatial information processor built upon a mosaic of
rod and cone photoreceptors, which are the light-responsive
elements that initiate signaling using graded electrical signals.2
The spatial information content is preserved through the lateral
geniculate nucleus, retinotopically mapped on to area V1 (area
17) of the primary visual cortex and from there on to a number
of higher visual processing areas. There is considerable process-
ing both within the retina as well as in higher brain structures
and we interpret these electrical signals as vision. While vision
is an analog system, a rough approximation to a digital system
would result in a resolution in excess of 500 megapixels.
Although initially conceived to be a simple model for the brain,
more probably, the retina approaches limits imposed by metabo-
lism, blood flow, and diffusion. This pressure to pack more
function into a small volume of neural tissue leads to increased
complexity.

VISUAL ILLUSIONS AND


MULTIPLE CHANNELS
Vision appears to be deceptively simple only because we are
very good at it: with few exceptions, a large portion of the mam-
malian brain is devoted to vision. A moment’s thought reveals
that vision must dovetail neatly with such high-order processes
as image storage and retrieval, forms of learning and memory
essential to comparison and recognition. However, even in the
retina, it is easy to demonstrate the presence of multiple pro-
cesses using well-known visual illusions.
At first glance, Figure 15.1 looks like a random array of large
pixels with three bright areas. Now, screw up your eyes, glance
aside or look at it from the other side of the room, and sud-
denly you may recognize da Vinci’s Mona Lisa. The bright
areas are the face, chest, and hands. But why does the image
seem to be more recognizable when visual input is distorted?
The high-spatial-frequency components have been removed by
greatly reducing the pixel number. What detail does remain is
a confusing array of boxes. However, this detailed view can
Fig. 15.1 Parallel pathways. A highly filtered image, 13 × 20 pixels, of
be blurred by squinting, which removes the high-frequency a well-known icon containing mostly low-spatial-frequency information.
components, leaving an easily recognized low-acuity version. At first, it may be hard to recognize because it is natural to focus on
So this is a direct demonstration that the visual system oper- the pixel edges. However, if you squint to blur the image, the true
identity of the picture may suddenly become more obvious. This is
ates on at least two channels of information at different spatial
a direct demonstration that the visual system carries several parallel
scales simultaneously. In fact, there is empirical evidence that streams of visual information with different spatial properties. For more
vision is composed of at least 15 parallel channels or streams details, see the text.
The retina is a piece of brain PRIMATE RETINA
Like the rest of the central nervous system (CNS), the retina is OUTER SEGMENTS
embryologically derived from the neural tube. It is formed from
361
the same components as the rest of the CNS and like all other
OUTER NUCLEAR LAYER
sensory systems has specialized structures, photoreceptors that

Chapter 15
CONES
transduce environmental energy into electrical potentials. As
OUTER PLEXIFORM LAYER
such, one can divide the retina in two parts3: (1) the sensory
retina, concerned with phototransduction of light by rod and INNER NUCLEAR LAYER CONE
PEDICLES
cone photoreceptors; and (2) the neural retina, consisting of
more typical interneurons (bipolar, horizontal, and amacrine INNER PLEXIFORM LAYER
cells) and projection neurons (ganglion cells) that carry out the GANGLION CELL LAYER

Function and Anatomy of the Mammalian Retina


first steps in processing visual information. NERVE FIBER LAYER BLOOD
The retina has been characterized by Dowling as an approach- VESSEL
able part of the brain,4 because it is a ready-made brain slice with
few barriers to the penetration of drugs or antibodies. In addi- Fig. 15.2 Primate retina. A vertical section through the macaque retina
tion, its natural stimulus, light, is easily controlled and the same taken with differential interference contrast optics. The retina is a
stimuli can be presented either to the intact animal or to the layered structure with two synaptic or plexiform layers sandwiched by
three nuclear layers: the outer nuclear layer, inner nuclear layer, and
retina removed from the eye and placed in vitro. This chapter ganglion cell layer. Although many fine details, such as cone pedicles,
focuses on processing in the mammalian retina. However, many are visible, individual neurons must be stained to see their dendritic
of the pioneering studies in retinal function were initially per- fields and specific connections. Large arrows: cones; small arrows:
formed in fish and amphibian retina. In particular, the salaman- cone pedicles. (Courtesy of SC Massey.)
der retina has been a long-standing model because its large cells
enhance the ease of electrophysiological recording.
photoreceptors, horizontal and bipolar cell dendrites interact.
NEURONAL COMMUNICATION: CHEMICAL The inner plexiform layer (IPL) separates the INL and the GCL
and this is where the bipolar cell axons, amacrine, and ganglion
AND ELECTRICAL cells interact. When we speak of the retina, outer or distal refers
In the retina, as in the rest of the CNS, the dominant form of to the scleral side of the retina and inner or proximal refers to
communication between neurons uses chemical neurotransmis- the vitreal side of the retina.
sion. Pulses of neurotransmitter (commonly the amino acids The functional stratification of the retina extends to the IPL,
glutamate, gamma-aminobutyric acid (GABA) and glycine plus which is organized according to the polarity of bipolar cell
acetylcholine (ACh), dopamine, and serotonin) are released from inputs.10 Since the time of Cajal, the IPL has been divided into
a presynaptic neuron and diffuse across the narrow synaptic five layers: 1 and 2 for sublamina a and layers 3–5 for sub-
cleft to bind to one of a variety of postsynaptic receptors. Recep- lamina b. In mammalian species, there are 9–11 morphological
tors can be ion channels themselves or can be linked via intracel- types of bipolar cell, plus the rod bipolar cell, which terminate
lular mechanisms to ion channels. Depending on the ion at different depths in the IPL.11–13 The polarity of a bipolar cell
selectivity (anion or cation), the opening or closing of the chan- response is determined by the differential expression of post-
nels produces a hyperpolarization or a depolarization of the synaptic glutamate receptors on their dendrites. The dendrites
postsynaptic cell. of OFF cone bipolar cells carry α-amino-3-hydroxy-5-methyl-
Many neurons also are directly connected via electrical syn- 4-isoxazolepropionic acid (AMPA)/kainate receptors and they
apses known as gap junctions.5–9 Gap junctions are named after ramify in sublamina a of the IPL.14 In contrast, ON cone bipolar
the narrow gap formed by docked hemichannels, or connexons cells and rod bipolar cells express mGluR6 receptors and their
aligned on either side of two cell membranes. Each hemichannel axons ramify in sublamina b.15–17 The separation of ON and
is built from six connexins that surround a central pore, forming OFF pathways appears to be a fundamental principle of retinal
an intercellular channel, which passes ions and small molecules organization, which is reflected throughout the visual system.18,19
(≤1 kDa). Gap junctions are not static pores; they are modulated There is a wealth of evidence to support the functional divi-
by light and contribute to neural processing. sion of the IPL into ON and OFF sublaminae. For example,
cholinergic amacrine cells (ChACs), also known as starburst
The retina is a layered structure amacrine cells on account of their unique morphology, are
The retina is a beautifully layered structure in which three layers present as mirror-image pairs.20 The conventionally placed
of neurons can be visualized without staining (Fig. 15.2). The ChACs have somas in the innermost layer of the INL. They have
outer nuclear layer (ONL) contains the cell bodies of photorecep- OFF responses to light stimulation and ramify in sublamina a.
tors, both rods and cones. The inner nuclear layer (INL) contains In contrast, the displaced ChACs reside in the GCL, produce
the cell bodies of horizontal, bipolar, amacrine and radial glial ON responses, and ramify in sublamina b. Likewise, alpha gan-
(or Müller) cells. The ganglion cell layer (GCL) contains dis- glion cells are present as paramorphic pairs such that the den-
placed amacrine and ganglion cells. Ganglion cells are the pro- dritic trees of OFF alpha ganglion cells are stratified in sublamina
jection neurons of the retina: their axons form the optic nerve a to receive input from OFF bipolar cells while ON alpha gan-
and project to a variety of subcortical nuclei. The three nuclear glion cells ramify in sublamina b to make contact with ON
layers are separated by two synaptic (plexiform) layers that bipolar cells.21,22 In primate retina, both midget and parasol gan-
contain the dendrites and synapses. The outer plexiform layer glion cells are present as paramorphic pairs, which conform to
(OPL) lies between the ONL and the INL. This is where the the stratification rules of the IPL. ON/OFF directionally selective
(DS) ganglion cells produce both ON and OFF responses of short known as the fovea. Here there is a depression in the retina,
latency, indicating direct input, and they are bistratified with known as the foveal pit, where cone photoreceptor density is
362 dendrites in sublaminae 2 and 4, coincident with the cholinergic highest and there are no rods. The pit results from the lack of
bands. overlying neurons (Fig. 15.3). Instead, cone axons, Henle fibers,
The ON and OFF stratification of the IPL is a fundamental run obliquely to cone pedicles away from the fovea where the
tenet of retinal organization which applies to many, if not most, bipolar and ganglion cells connected to the central cones are
Section 1

cell types. However, several exceptions to this rule have now “piled up” in an annular zone with the GCL 6–8 cells thick. The
been identified. For example, the dopaminergic amacrine cells
(DACs) stratify predominantly in sublamina a, the OFF layer,
yet they apparently produce ON responses to light.23,24 Likewise, Table 15.1 Cellular and synaptic layers of the retina
the intrinsically photosensitive retinal ganglion cells (ipRGCs),
which stratify in sublamina a of the primate retina, were all ON Layer Contains:
Anatomy and Physiology
Basic Science and Translation to Therapy

cells.25 These cell types, among others, receive input from ON


Outer nuclear layer (ONL) Photoreceptors, rods, and
bipolar axons as they traverse sublamina a of the IPL. These
cones
axonal ribbons provide a set of inputs that break the stratifica-
tion rules of the IPL. Thus, there is an additional accessory ON Inner nuclear layer (INL) Horizontal cells, bipolar cells,
sublayer in the outer portion of the IPL26,27 (see sections on DACs amacrine cells, Müller cells
and ipRGCs) (Table 15.1).
Ganglion cell layer (GCL) Ganglion cells, displaced
amacrine cells
GROSS RETINAL MORPHOLOGY
Outer plexiform layer (OPL) Photoreceptors talk to
The fovea horizontal cells and bipolar cells
When a peripheral visual stimulus gets our attention, we auto-
matically focus the retinal image in the center of our gaze. In the Inner plexiform layer (IPL) Bipolar cells talk to amacrine
cells and ganglion cells
primate retina this central region of highest visual acuity is

Fig. 15.3 The fovea and distribution of rods and cones in the human
eye. (A) This cross-section of the macaque retina shows the foveal pit,
Choroid
a specialization of the primate retina. In this region, the cones are
smallest and their packing density reaches a maximum. There are no
PE
rods. At the fovea, ganglion cells and other retinal layers are reduced
OS for maximum sensitivity and acuity. PE, pigment epithelium; OS, outer
ONL segments; ONL, outer nuclear layer; INL, inner nuclear layer; IPL,
inner plexiform layer; GCL, ganglion cell layer. (Courtesy of Louvenia
Carter-Dawson.) (B) Rod and cone density across the retina. Note how
INL
the cones peak at the fovea. Rod density falls to zero at the fovea but
IPL light reaches a maximum around 5 mm outside the fovea. There are no
GCL photoreceptors at the point where the optic nerve leaves the eye, also
known as the blind spot. (Modified with permission from Wandell BA.
Foundations of vision. Sunderland, MA: Sinauer Associates, 1995.)
A

1.8
Blind spot Cones
Rods
Number of receptors (mm-2 x 105)

1.4

1.0

–60º
60º 0.6
Blind spot
–40º
40º

–20º 0.2
20º

Fovea –60 –40 –20 0 20 40 60
B Angle relative to fovea (deg)
363

Chapter 15
Fig. 15.4 How to find the blind spot. To demonstrate the blind spot, hold the page level at normal reading distance. Look at the O in the figure,

Function and Anatomy of the Mammalian Retina


then close your right eye. The X on the left should disappear because the image of the cross falls on the optic nerve head. You can reverse this
demonstration: look at the X, then close the left eye to make the O disappear. Remember, the blind spot is nasal to the fovea. The light rays
cross over at the lens so the blind spot is lateral to the point of focus.

foveal structure is thought to maximize sensitivity because light


cannot be scattered by passing through other retinal layers. It
OPL
also optimizes acuity by packing the maximum number of cones
and reducing their size. In human retina, the peak cone density
approaches 200 000/mm2 and the ONL is slightly thicker to
accommodate these extra cells.28 There are no blood vessels in
the fovea and in the central fovea there are no blue cones. The INL
low density of blue cones lowers their acuity,29 to match the
blurring caused by chromatic aberration in the lens.30 Other
mammals have an area centralis (cat) or a visual streak (rabbit,
swine) with similar high cell density but these structures lack
the central depression. A consequence of the exclusion of rods
IPL
from the fovea is that in dark-adapted conditions, say looking
for a dim star, it is necessary to look slightly off the visual axis
to focus the image in the region of high rod density.

The blind spot and how to find it GCL

There are no photoreceptors where the optic nerve exits the eye PKC TOH
Calbindin ChAT
and so any image that falls on this region cannot be processed
by the retina. Curiously, we do not perceive a hole in the visual Fig. 15.5 Quad labeling of the rabbit retina. In this vertical section,
scene because the visual system fills in. To demonstrate the multiple antibodies were used to label different cell types. Red,
calbindin, shows horizontal cells, large somas high in the inner nuclear
blind spot, hold the page level at normal reading distance. Look layer (INL) adjacent to the outer plexiform layer (OPL). Also in red, an
at the O in Figure 15.4, and then close your right eye. The X ON cone bipolar cell with a single axon descending to sublamina 4 of
on the left should disappear because the image of the cross falls the inner plexiform layer (IPL). Green, protein kinase C, shows multiple
rod bipolar cells descending to sublamina 5 of the IPL where they
on the optic nerve head. You can reverse this demonstration: have prominent terminals. Blue, choline acetyltransferase, labels
look at the X, then close the left eye to make the O disappear. starburst amacrine cells, one in the INL and two in the ganglion cell
Remember, the blind spot is nasal to the fovea. The light rays layer. These cells make two dense bands in the IPL. Pink, tyrosine
cross over at the lens so the blind spot is lateral to the point hydroxylase, shows the plexus of dopaminergic processes, mostly in
sublamina a, adjacent to the amacrine cell layer. There are also a few
of focus. processes in the middle of the IPL. As well as staining multiple
neurons, this figure demonstrates that the IPL is highly stratified with
Painting the retina – techniques to label each cell type at a distinct depth. Ten to 12 distinct layers can be
and visualize retinal neurons found in the IPL. (Courtesy of W Li and SC Massey.)

The major landmarks of the retina, two synaptic layers sand-


wiched between three nuclear layers, are obvious even in the
unstained section of macaque retina shown in Figure 15.2. In cell types selectively (Fig. 15.5), so that we can differentiate
fact, there is an immense amount of detail present in this image. between rod and cone pathways, for example. Furthermore, it is
For example, individual cones can be seen with their descending very useful to visualize several cell types at once so that the
axons terminating in a row of cone pedicles in the OPL. However, number and position of one cell type relative to another can be
the somas of all the other retinal neurons are indistinguishable determined. This can be achieved by confocal microscopy, which
and the details of their structures cannot be visualized. One goal permits the simultaneous acquisition of three or more different
of vision scientists is to have a complete map of all the intercon- labels in addition to providing improved resolution and three-
nections between the retinal neurons so that the functional dimensional visualization (Fig. 15.5). With care, confocal micro­
outputs can be fully understood. To uncover the hidden path- scopy can be used to visualize both chemical and electrical
ways and circuits through the retina, we need to stain specific synapses in the retina.31 Two-photon confocal microscopy
permits imaging using an infrared laser, which does not stimu- Table 15.2 Major cell types in the retina
late the visual pigments. It has been used to measure changes in
364 intracellular calcium concentration of single neurons or den- Neuronal types Role Types
drites in response to stimulation with visible light.32,33 One dis-
Photoreceptors Rod and cones 2
advantage of these methods is that individual synapses cannot
easily be visualized because of the inherent resolution limits of
Section 1

Horizontal cells Lateral interneurons, OPL 2


confocal microscopy. However, high-throughput electron
microscopy methods have been developed that allow three- Bipolar cells Vertical connection 10–12
dimensional reconstruction of blocks of retina, albeit in fixed
Amacrine cells Lateral interneurons, IPL ~30
tissue. By careful analysis and multiple staining approaches it is
possible to identify all the synapses and cells to which an indi- Ganglion cells Output neurons ~20
vidual neuron makes connections.34
Anatomy and Physiology
Basic Science and Translation to Therapy

The mainstay of structure, function, and morphology studies Interplexiform cells Feedback, IPL to OPL ?
is immunocytochemical methods, which can be used to stain
  Total ~65
structural components, enzymes, neurotransmitters, synaptic
proteins, and postsynaptic receptors (Fig. 15.5). Specific primary OPL, outer plexiform layer; IPL, inner plexiform layer.

antibodies are currently available for many of these components


or they can be generated. Secondary antibodies conjugated to an
ever-increasing variety of fluorochromes are readily available as
standard reagents and allow for multiple staining. Single neurons synapses.42,43 As we learn more about neuronal circuits and the
can also be filled with fluorescent dyes via glass microelectrodes. functional anatomy we see repeated patterns across the retina,
Diffusible tracers such as Neurobiotin can be used to label a and we are reminded that it is a two-dimensional array of recur-
network of coupled cells.5 The optic nerve can be back-filled with ring circuits.
fluorescent dyes, which in one useful variant are concentrated The classification of retinal neurons, which dates back to the
in vacuoles that explode when excited to release dye, which dif- work of Cajal more than a century ago, is not a simple matter.44
fuses throughout the dendritic structure of individual ganglion However, it is important that we are at least aware of all the
cells.35 Certain ganglion cell types can be selectively labeled by major cell types in the retina. Even a basic understanding of a
stereotactically injecting a central target with retrograde tracers.36 circuit cannot be achieved if critical parts are missing. Classifica-
Another method to label single cells involves labeling the whole tion is complicated by the large number of different cell types,
retina with dihydrorhodamine, which can then be photoconver- the low probability of obtaining rare types, variation in size with
ted to a fluorescent product in selected cells by illumination.37,38 eccentricity, and the normal morphological variation within a
Individual neurons of all types can be randomly labeled with cell type. While the staining and sampling issues have largely
ballistic particles coated with fluorescent dyes or DNA to syn- been resolved by modern methods, the question of what consti-
thesize a specific marker.39 Finally, there is increasing use of tutes a separate cell type, as opposed to subtle differences
transgenic animals such as mice engineered to express green between examples from the same type, is still a problem. The
fluorescent protein (GFP) variants under the control of a specific resolution lies partly in understanding the significance of some
promoter.40 morphological features and partly in considering more vari-
ables, i.e., independent criteria to classify a specific cell type.
Six major neuronal cell classes Finally, as a kind of parity check, it is necessary to consider the
The retina contains six major neuronal classes. Photoreceptors properties of a whole population for a given cell type to see if it
are located in the ONL and can be subdivided into rods and is consistent.
cones. Bipolar cells take the signals from photoreceptors and Neurons may be classified into unique cell types on the basis
transmit them to the inner retina. Horizontal cells and amacrine of several properties:
cells are laterally extensive interneurons in the outer and inner
retina, respectively. Ganglion cells receive input from bipolar 1. their morphology, meaning the size, shape, and structure of
and amacrine cells and form the output from the retina. In addi- a neuron, especially its dendritic field. This may include
tion, interplexiform cells share many properties with amacrine measures of dendritic density and branching patterns
cells but project back to the outer retina. Finally, the radially 2. the depth within the retina, particularly the level in the IPL
oriented Müller cells are the predominant glial cells (Table 15.2). where its dendrites or axons stratify. We have come to
realize that depth in the IPL functions as a simple
addressing system to determine which target neurons are
CLASSIFICATION OF RETINAL CELLS in synaptic contact
Although the retina contains six major cell classes, these may be 3. their electrophysiology, particularly their excitatory and
further divided into many distinct subtypes for a total of about inhibitory inputs
65 different neurons. Thus, the retina is certainly complex, but 4. their biochemistry, particularly with regard to the different
it is not chaotic. Cells of a given type form nonrandom mosaics neurotransmitters used to communicate between neurons
across the retina.41 A single type shares essential morphological and structural proteins or receptors
features and uses the same neurotransmitter(s). Furthermore, 5. the pattern of connections with other neurons, which should
they ramify at a characteristic depth in the retina and make be consistent for a unique cell type
stereotyped synaptic connections, including the same type and 6. finally, the properties of the whole population for a specific
number of postsynaptic cells, and even the same number of cell type.
A group of cells that are all the same type have certain special Cones support color vision and, in old-world primates and
properties. The retina is a two-dimensional array and most cell humans, there are three classes: blue, green and red. They are
types tile the retina in a consistent manner so that an even maximally sensitive to 430, 530, and 561 nm light, respectively.53–55 365
coverage is obtained. Cells of the same type are usually not Other mammals, including cats, rabbits, and rodents, have an
adjacent. The position of the cells relative to others of the same evolutionary ancient form of color vision based on green and

Chapter 15
class is also an indicator of cell type because unique popula- blue cones only. The presence of red and green cone opsins in a
tions form nonrandom mosaics.41 Consider a handful of marbles tandem array on the X chromosome is thought to be due to a
dropped on the floor. Some will stop close by but others may recent gene duplication and underlies the preponderance of
roll into a corner. If a measure is taken between the nearest color blindness among males.56 Using adaptive optics to correct
neighbors, it will be found to have a high variation. In contrast, for blurring in the lens and cornea, the distribution of red, green,
for a nonrandom mosaic of neurons, the spacing is controlled and blue cones can be mapped in the living human eye.57 Sur-
such that the distance to the nearest cell of the same type is prisingly, the distribution of cones was random and clumpy

Function and Anatomy of the Mammalian Retina


relatively constant and the variance of this measure is low. (Fig. 15.6). In addition, there appears to be enormous variation
The ratio of mean nearest neighbor distance to the variance in the red/green cone ratio among individuals with normal color
gives an index of regularity for a cell population. We would vision.
expect the ratio to be low for the randomly scattered marbles, Blue cones are present as a minority: they make up approxi-
but indices of three or higher are common for the typical non- mately 10% of the cone population (Fig. 15.6). This is not enough
random, orderly mosaics of retinal neurons. The most satisfying to support high acuity but calculations show that the blue cone
results for classification come from the convergence of these density is sufficient to support the reduced resolution caused by
methods to indicate the function of a retinal cell in the process visual aberration at the blue end of the spectrum.30 At the very
of vision. center of the fovea, blue cones are absent.29,58 The fact that this is
not readily apparent is due to the relatively poor spatial acuity
Photoreceptors of color vision compared to the luminance-driven pathways.
Cones Red and green cones cannot be differentiated morphologically
The mammalian retina contains two types of photoreceptor, rods and the red and green cone opsins are so closely related that they
and cones.45 Rods account for 95% of all photoreceptors. They are also indistinguishable. However, blue cones can be mapped
are numerous, with slender outer segments, densely packed and
specialized for high sensitivity under dark or starlight condi-
tions. Cones are larger, with tapering outer segments, and they
are found in the top row of the ONL (see Fig. 15.3). Cones make
up only ~5% of photoreceptors28,45 but they provide high-acuity
color vision in daylight conditions when photons are abundant.
This versatile combination of rods and cones and their associated
circuits covers an intensity range of around 10 log units from the
darkest night to bright sunlight.46 While the average visual scene
has a range of intensities covering 2–3 log units, the continual
adaptation of retinal sensitivity slides this operating range
through the entire range of light intensities. This is a critical
function of the retina because outside the normal operating
range we are functionally blind. Common examples include the
inability to see momentarily when entering a dark cinema or
driving into the setting sun. Much of the adaptation takes place
in photoreceptors but, as we shall see below, this is accompanied
by major changes in the neural pathways through the retina.
This is arguably the most important function of the retina, after
light detection itself.
There are approximately 5 million cones in the human retina
and 190 000 in the mouse retina.47,48 Cones make up approxi-
mately 5% of the total photoreceptors in humans, compared to
2.8% in the mouse,49 so we are all rod-dominated in terms of
absolute numbers. One exception is the ground squirrel, which
is truly cone-dominated. Importantly, cones are not evenly dis-
tributed. In human retina, there is a massive peak at the fovea
(Fig. 15.3) where the density reaches around 200 000/mm2,
approximately 100 times the density in the periphery.28,47 This Fig. 15.6 The mosaic of red, green, and blue cones in the human
retina. This image, taken from a human subject using adaptive optics,
is the region of maximum acuity, although the peak density shows the distribution of the three cone classes. Blue cones make up
slightly exceeds experimentally measured visual acuity due to a small fraction, <10%, but make a regular mosaic. Red and green
blurring by the optics of the eye.50–52 Where the ganglion cell cones have a clumpy, random distribution. In this subject, the red
cones outnumber the green cones but this ratio is highly variable, even
axons gather to form the optic nerve there are no photoreceptors
in subjects who have normal color vision. (Courtesy of Austin Roorda;
and their absence from this location is the cause of the blind after Roorda A, Williams DR. The arrangement of the three cone
spot (Fig. 15.3). classes in the living human eye. Nature 1999;397:520–2.)
with a selective antibody against blue cone opsin29,59 or by their single large horseshoe-shaped ribbon with two bipolar cell den-
selective accumulation of certain dyes.58 drites nestled within. The cones have numerous and slightly
366 smaller ribbons with each also nestled against an mGluR6-
Rods labeled dendrite. What is not visible are the horizontal cell den-
The ONL contains photoreceptor cell bodies, both rods and drites that also invaginate, but are slightly lower than the bipolar
cones.45 Even in the cone-dominated human retina, rods far out- cells.
Section 1

number cones, by a factor of 20 : 1, so they account for most of Around 10–12 cone bipolar cells plus multiple horizontal cell
the ONL except at the fovea. The human retina contains approxi- dendrites contact each cone, so under the pedicle there may be
mately 100 million rods, and they pool signals to provide high as many as 200 postsynaptic processes.64 In fact, the cone pedicle
sensitivity for dark-adapted vision, say starlight, which appears may be the most complex synaptic structure in the brain. The
monochromatic. A lack of color vision is the hallmark of rod- distribution of postsynaptic processes under the cone pedicle is
mediated vision. Rods are absent within 350 µm of the fovea but laminated in a stereotyped fashion (Fig. 15.8).63,64
Anatomy and Physiology
Basic Science and Translation to Therapy

reach a peak density in an annular region at about 20° eccentric- The fundamental problem for rods is to detect a small brief
ity (Fig. 15.3). This does not match the area of maximum scotopic hyperpolarization due to a single photon against a noisy back-
acuity, which occurs around 5°,60 so it has been suggested that ground of thermal noise and the stochastic nature of transmitter
another component of the rod pathway, such as the AII amacrine release.65,66 One strategy is to minimize background variation
cells, present at a much lower density, forms a bottleneck to limit by maintaining a high sustained-release rate in the dark. The
acuity61,62 (see below). synaptic terminal of a rod, or rod spherule, is about 2  µm in
diameter and contains a very large synaptic ribbon (Fig. 15.9),
Cone pedicles and rod spherules a specialization thought to be associated with a high rate of
Cones and rods make contact with bipolar and horizontal cells transmitter release. The spherule is packed with synaptic ves-
in the OPL. Cone axons descend through the massed ranks of icles, containing the neurotransmitter glutamate, to support
rod somas in the ONL to terminate in a two-dimensional array sustained transmitter release. There is a single invagination
of cone pedicles at the OPL (Fig. 15.7). Near the fovea in primate (imagine a closed fist and insert a finger between thumb and
retinas, the cone axons are splayed out radially so that the ped- forefinger to make a pocket) containing a tetrad of processes,
icles form an annular array around the center (Fig. 15.7B). Cones two from horizontal cells and two rod bipolar dendrites.67 This
form two specialized contacts, ribbon synapses and flat contacts, structure brings the postsynaptic processes close to the release
with postsynaptic neurons. Ribbon synapses are so named site and may prevent spillover to adjacent rods. These anatomi-
because of electron-dense structures at invaginations where they cal specializations appear to reduce variation in rod signaling
contact depolarizing (or ON) bipolar and horizontal cells (Fig. so that small single-photon responses can be detected with high
15.8). Cone pedicles also make flat contacts along the base of the reliability.
pedicle with hyperpolarizing (or OFF) bipolar cells. Rod axons
also descend to form synapses with a single type of depolarizing Photoreceptor coupling
bipolar cell as well as horizontal cells. The terminals of rods are While the synapses between photoreceptors and second-order
called spherules, and, similar to cones, they use a ribbon synapse. neurons are extremely complex, there are still additional connec-
In contrast to cones, there is only a single invagination in each, tions between photoreceptors. These take the form of electrical
containing two rod bipolar and two horizontal cell dendrites. coupling, mediated by gap junctions. Close to the fovea, the cone
Each cone pedicle is 6–8 µm in diameter and, near the fovea, pedicles are densely packed, almost touching, and connected by
contains 20 synaptic ribbons and, in the periphery, around 40.63 very fine processes known as telodendria (Fig. 15.7C).45 Rod
Cones release glutamate constantly in the dark and the synaptic terminals are either absent or displaced slightly higher, towards
ribbons are thought to support this high rate of release. As we the outer segments, in this region. More peripherally, the cone
will describe further below, the multiple ribbon and flat syn- pedicles are widely spaced and the telodendria are much more
apses on each cone pedicle form connections with many differ- prominent (Fig. 15.7D). The contact points between telodendria
ent types of bipolar cell. of neighboring cones are the sites of connexin (Cx)36 gap junc-
A sense of the complexity of these synapses can be obtained tions, which mediate electrical coupling between cones.68,69
by labeling some of the individual components. Antibodies reac- Recordings between neighboring cones also show that they are
tive against a vesicle protein such as synaptophysin provide a coupled.68,70 This may seem puzzling at first because it should
way to label photoreceptor terminals in the OPL, because pho- lead to a loss of acuity and blurring. However, the cone array
toreceptor axon terminals are filled with vesicles. To visualize actually oversamples the signal so this leads to little or no loss.
the synaptic ribbons, antibodies to kinesin II can be used. Finally, Instead, the coupling is thought to reduce noise, which is
antibodies to the mGluR6 receptor, which is located on the den- random, while the light-driven signals, which are correlated
dritic tip of the depolarizing bipolar cells, mark one of the post- between close neighbors, will be reinforcing. It has been calcu-
synaptic processes. When visualizing just the cone pedicle, there lated that this may improve the signal-to-noise ratio by 77% – a
are a number of indentations or invaginations (Fig. 15.8). Within large gain for little loss.71 Coupling between red/green cones is
each invagination, horizontal cell dendrites are lateral and high, indiscriminate, which may reflect the close absorption curves of
approaching the synaptic ribbons. ON cone bipolar cell pro- red and green cones.70 Morphologically, blue cone pedicles are
cesses are central but slightly lower at the synaptic ribbon. In slightly smaller than those of red/green cones and they have
contrast, OFF bipolar dendrites form basal synapses with the only a few withered dendrites, which rarely touch neighboring
cone pedicle at a distinctly lower level. Staining a piece of retina cones.72 Hence, blue cones are not coupled into the red/green
for synaptophysin, kinesin II, and mGluR6, begins to show the network.68,70 This may serve to preserve spectral information in
complexity of the structures (Fig. 15.9). Rod spherules contain a the color pathways.
367

Chapter 15
Function and Anatomy of the Mammalian Retina
A Primate Cones 10 µm B Fovea 50.00 µm

C Cone Pedicles – Central 5 µm D Cone Pedicles – Peripheral 10 µm

Fig. 15.7 Primate cones. (A) Vertical section of macaque retina shows cones stained with an antibody against cone arrestin. The outer segments
of a few cones, <10%, have been double-labeled with an antibody against blue cone opsin. From each cone, an axon descends to the cone
terminal or pedicle. (B) Whole-mount view shows how the axons run obliquely away from the fovea to terminate in an annulus of cone pedicles.
(C) Close to the fovea, the cone pedicles form a tightly packed array connected by very fine telodendria. The dark regions within each pedicle
are the invaginations penetrating from beneath. (D) More peripherally, the cone pedicles are widely spaced and connected by longer telodendria
which are the sites of cone-to-cone coupling. (Antibody donated by Peter MacLeish, images courtesy of J O’Brien and SC Massey.)

There are also gap junctions between cone pedicles and rods.73 coupling forms an alternative rod-driven pathway that may be
These allow rod signals to enter cones, and rod-mediated signals important at intermediate light intensities, in the mesopic
can be detected in second-order neurons that are thought to be range.76,77 This influence may relate to the enhancement of cone
connected exclusively to cones.74 Responses with the signature electroretinogram (ERG) amplitudes in humans and mice by
of rod origin have also been recorded in primate cones.75 Because light adaptation, where the cone ERG gradually increases in
rods far outnumber cones, the influence of many rods on a single amplitude following the onset of a steady adapting field.78–81
cone may be substantial. It is now thought that rod–cone Blocking gap junctions inhibits this effect in the mouse retina. In
A
368

H
Section 1


Anatomy and Physiology
Basic Science and Translation to Therapy

C 5 µm

Fig. 15.8 Laminated postsynaptic structure of the cone pedicle. (A) Cross-section of a single cone pedicle contains synaptic ribbons and synaptic
vesicles. Horizontal cell dendrites (red) invaginate the cone pedicle deeply and laterally. ON cone bipolar dendrites (green) assume a central
position beneath the ribbon. (B) Whole-mount view, in the plane just below the multiple synaptic ribbons, shows the relative positions of
horizontal and ON bipolar processes. The OFF bipolar contacts are beneath this plane of focus. (C) Electron microscopic view of a cone pedicle.
Arrowheads mark synaptic ribbons. Horizontal cell processes (H) are lateral, ON bipolar dendrites (star) central. OFF bipolar cells (asterisk)
form basal synapses away from the synaptic ribbon. Beneath each cone pedicle lies a snake’s nest of postsynaptic processes. There are also
specialized desmosome-like contacts of unknown function between horizontal cell processes (small arrows). (Reproduced with permission from
Haverkamp S, Grunert V, Wassle H. The cone pedicle, a complex synapse in the retina. Neuron 2000;27:85–95.)

the human, however, the adaptation dependence of these neurotransmission because the clearance of glutamate must be
changes has a photopic (cone) signature. Rod–cone coupling also rapid to provide a fast postsynaptic response to light.85 The
is very dynamic and influenced by circadian rhythms, increasing hyperpolarizing light response of photoreceptors is now well
at night and decreasing during the day.82 established and it is supported by studies of vesicle turnover,
which is much higher in the dark.86 Furthermore, synaptic block-
Photoreceptors release glutamate in the dark ing studies produce the appropriate responses in the different
The first intracellular recordings from cones were surprising second-order neurons and there is a stereotyped array of distinct
because they showed that photoreceptors hyperpolarize in glutamate receptors associated with specific cell types. Gluta-
response to light.4 This means they are relatively depolarized mate, with its library of postsynaptic receptors, seems particu-
and release their neurotransmitter, glutamate, in the dark. From larly suitable to orchestrate the large variety of postsynaptic
the viewpoint of signal information, the sign of the photorecep- responses at the cone pedicle.
tor response makes no logical difference; photoreceptors produce Rods operate in a manner essentially similar to cones: they
graded responses modulated around the mean light level. When hyperpolarize in response to light increases, albeit there are
a photon is absorbed, the visual pigment is activated and then a many molecular differences. However, everything about the rod
cascade of other biochemical events is triggered.83 This is known pathway in the retina is designed for maximum sensitivity. This
as phototransduction and it leads to the closure of a cGMP-gated is reflected in the anatomical details and synaptic connections of
cation channel in the cell membrane to produce hyperpolariza- rods.66 Rods can respond to single photons, obviously the design
tion and a reduction in the release of glutamate.84 The postsyn- limit, with a binary (all or nothing) signal, but visual threshold
aptic responses to light are in fact due to a decrease of glutamate requires a signal in 5–10 rods. Depending on the species, about
release. Thus, glutamate uptake also plays a key role in retinal 20–100 rods converge on to a single rod bipolar cell87,88 and this
369

Chapter 15
Function and Anatomy of the Mammalian Retina
A B

C D

Fig. 15.9 Synaptic structure of rod spherules and cone pedicles. (A) The terminals of rods and cones are packed with synaptic vesicles and
contain synaptic ribbons. An antibody against synaptophysin (green), a synaptic vesicle protein, labels rod spherules which appear round (some
are outlined to the lower right) and cone pedicles which are polygonal (larger outlines). Each rod spherule contains a single large synaptic ribbon
which is curved like a horseshoe, stained with an antibody against kinesin II (red). The cone pedicles contain a complex of smaller synaptic
ribbons (also red). (B) The dendritic tips of rod bipolar and ON cone bipolar cells can be marked with an antibody against the glutamate receptor,
mGluR6 (blue). The central invagination of each rod spherule contains a pair of mGluR6-positive rod bipolar dendrites. At each cone pedicle,
there are a number of finer ON bipolar dendrites. (C) The rod bipolar dendrites (blue) are nestled within the horseshoe formed by the synaptic
ribbon (red) at each rod spherule. At the cone pedicles, the ON bipolar dendrites also approach closely to the cone synaptic ribbons. (D) A
triple-label image showing rod spherules and cone pedicles (green, some outlined), synaptic ribbons (red) and the postsynaptic bipolar dendrites
tagged for the glutamate receptor mGluR6 (blue). Scale bars = 10 μm. (Courtesy of W Li and SC Massey.)

high convergence also contributes to the high sensitivity of the


rod pathway. If 100 rods converge to a single rod bipolar cell
Second-order neurons: horizontal
and 100 rod bipolar cells converge to a ganglion cell, then the and bipolar cells
absolute threshold for vision is determined by the product, Rods and cones make synaptic connections with bipolar
approximately 1 photoisomerization per 10 000 rods. and horizontal cells. Horizontal cells are laterally extensive
interneurons located in the outer row of the INL, adjacent to the the axon terminal whose branches contact rods instead of cones.98
OPL. They respond to diffuse light with a large hyperpolariza- B-type horizontal cells are also coupled by gap junctions and so
370 tion. This is the same as cones so we say that the input is sign- are the axon terminals (Fig. 15.11B).95,99–101 For both types of hori-
conserving. It appears to be mediated by excitatory glutamate zontal cell, as the cell density falls with eccentricity, the dendritic
receptors of the AMPA subtype, which have been located on field size increases so that an even coverage of 6–8 is maintained
horizontal cell dendrites.89,90 In most mammals there are across the retina. B-type horizontal cells are always smaller and
Section 1

two morphological types of horizontal cell, but only one in more numerous – two to three times the A-type density.95
rodents.91–94 In all species, horizontal cells are extensively In primates, there are also two kinds of horizontal cell, both
coupled, which dramatically increases the size of the receptive axon-bearing, but the H2 only contacts cones and the axon is not
field. In Figure 15.10 the entire A-type horizontal cell network well developed.102 H1 is a large, well-coupled cell that contacts
in the rabbit retina has been labeled by injecting several cells all red/green cones but not blue cones.103 H2 has a smaller soma
with a diffusible tracer, Neurobiotin, which readily passes and finer dendrites that make sparse contacts with red/green
Anatomy and Physiology
Basic Science and Translation to Therapy

through gap junctions.95 A-type horizontal cells are axonless and cones but densely innervate blue cone pedicles.103 Recording
have a large irregular shape, giving rise to many fine terminals from both horizontal cell types shows that they receive sign-
which contact every cone pedicle within the dendritic field. conserving inputs from both red and green cones (plus blue for
A high-resolution image shows how fine horizontal cell den- H2).103 The wiring of the H2 horizontal cell suggests it plays a
drites converge at cone terminals while ignoring the numerous role in blue/yellow (red +green) processing.
rod spherules (Fig. 15.10). The cone terminals were marked by In central retina, there are four times as many H1s as H2s,
the labeling of two different glutamate receptors, GluR5, which decreasing to twice as many in peripheral retina.104 Increasing
marks the basal contacts of certain OFF bipolar cells,96 and size with eccentricity compensates for decreasing density so cov-
mGluR6, which is expressed by ON cone bipolar cells17,97 (see erage for both types is 3–5 evenly across most of the retina. The
below). It should be noted that the three labels are nonoverlap- peak density for H1 horizontal cells reaches about 18 000/mm2
ping at the cone terminal, consistent with the presence of three near the fovea. This is an order of magnitude higher than rabbit
separate neurons, horizontal cells, ON cone bipolar cells, and retina. Packer and Dacey105 have recorded from many primate
OFF cone bipolar cells, which all converge independently at the H1 cells and they make the interesting observation that central
cone pedicles (Fig. 15.10).64 cells are not only smaller but less coupled, perhaps because they
B-type horizontal cells in the rabbit retina are smaller and overlap less. Thus, the H1 receptive field in the fovea may be
radially symmetrical (Fig. 15.11). While the somatic dendrites of small enough, 20–30 µm, to match the receptive field surround
the B-type horizontal cell also contact cones, each cell gives rise of midget ganglion cells.
to a long axon that meanders randomly before branching into Horizontal cells are extensively coupled by gap junctions and
an elaborate terminal structure (Fig. 15.11C).95 The electrotonic their molecular identity has been determined for many species.
length of the axon apparently isolates the somatic region from In the rabbit retina, the gap junctions between A-type horizontal

5 µm

Fig. 15.10 A-type horizontal cells in the rabbit retina. The coupled matrix of A-type horizontal cells following the intracellular injection of
Neurobiotin (green) in the rabbit retina and the rod/cone mosaic is shown by labeling postsynaptic glutamate receptors, GluR5 (blue) and
mGluR6 (red). The A-type horizontal cells contact every cone in the frame. The high-resolution images (right) show that the fine horizontal cell
dendrites (green) converge at individual cone pedicles. The green, red, and blue labels are not colocalized. They label independent neuronal
structures: horizontal cells, ON bipolar cells, and OFF bipolar cells, respectively. (Courtesy of F Pan and SC Massey.)
371

Chapter 15
Function and Anatomy of the Mammalian Retina
A B

Fig. 15.11 B-type horizontal cells in the rabbit retina. (A) A single
B-type horizontal cell filled with Lucifer Yellow contacts all the cone
pedicles (white outlines) within its dendritic field. Arrow shows the axon
leaving the frame. (B) When B-type horizontal cells are filled with
Neurobiotin, the coupled network is revealed. (C) Fine details of the
elaborate axon terminal of a B-type horizontal cell. Each terminal
varicosity contacts a rod spherule. (Panel A, courtesy of W Li and SC
Massey; panels B and C, reproduced with permission from Mills SL,
C Massey SC. A series of biotinylated tracers distinguishes three types
of gap junction in retina. J Neurosci 2000;20:8629–36.)

cells are labeled with an antibody against Cx50 at many contact make it possible to test theories concerning horizontal cell
points in the matrix (Fig. 15.12).106 Some of the gap junction coupling.
plaques are very large and the unitary conductance of Cx50
channels is also high. This explains why A-type horizontal cells Horizontal cell function
form an electrical syncytium. B-type horizontal cells are not As pointed out by Sterling, reading high-contrast images in
labeled for Cx50. These gap junctions have different properties bright artificial light is one thing,108 but step into the outside
so they are likely to be assembled from different connexins. In world of the naturalist and visual objects often have very low
rodents, there is only one type of axon-bearing horizontal cell.94 contrast from the background. What we need is a way to subtract
In a Cx57 knockout mouse, these cells are no longer coupled.107 the common background so we can focus on the small, low-
This suggests that multiple neuronal connexins are present in contrast details in the image. At least in part, this role is accom-
the retina and that Cx57 gap junctions may be responsible for plished by horizontal cells in the outer retina. The horizontal cell
coupling in axon-bearing horizontal cells. The situation in the network holds a big slow picture of the visual scene that is sub-
primate retina is still unknown but future progress should tracted by feedback to cones. Because horizontal cells have a
vesicular GABA transporter,115 and there is mounting evidence
that many components required for GABA release also are
372 present.116–118 In addition, GABA receptors are located on bipolar
cell dendrites119 or perhaps cone pedicles themselves,120 provid-
ing another mechanism for feedback. However, GABA antagon­
ists do not block surround antagonism either in photoreceptors121
Section 1

or at the level of the ganglion cell receptive field.122 An ephaptic


mechanism is proposed in which a change in the transmembrane
potential of the cone photoreceptor occurs via hemichannels and
glutamate receptors on the horizontal cell dendrites and creates
a voltage drop in the synapse; a subsequent increase in photo-
receptor calcium channel activity modifies the gain in the cone
Anatomy and Physiology
Basic Science and Translation to Therapy

synapse.123,124 Finally, a model is proposed in which horizontal


cell depolarization produces an efflux of protons, which acidifies
the extracellular space and inhibits cone voltage-gated calcium
channels.125,126 While these discrepancies in the mechanism of
negative feedback remain to be resolved, results of a recent pub-
lication suggest that a positive-feedback mechanism may also
contribute to signaling at this synapse.127
Studies of this important mechanism have been undertaken
primarily in species other than mouse, which has meant the
10 µm
Inj. AHC Cx50-CT GluR5 analyses have required pharmacological manipulations, rather
than genetic approaches. However, recently a zebrafish model,
Fig. 15.12 Gap junctions between A-type horizontal cells. A-type in which a specific connexin was thought to make up the hori-
horizontal cells are extensively coupled, as observed following
Neurobiotin injection (red). An antibody against connexin 50 (green) zontal cell hemichannels, was identified. In these fish feedback
labels gap junctions in the matrix of A-type horizontal cells. Some was reduced, providing support for the ephaptic hypothesis.128
large plaques are located at the crossing of two major dendrites Studies in mice also have shown that feedback is present. This
(yellow). The background mosaic of cone pedicles is marked by GluR5
model organism provides the possibility that the underlying
labeling (blue). (Reproduced with permission from O’Brien JJ, Li W,
Pan F, et al. Coupling between A-type horizontal cells is mediated by mechanism can be dissected genetically. No doubt in the coming
connexin 50 gap junctions in the rabbit retina. J Neurosci years the details of this feedback mechanism, which is critical to
2006;26:11624–36.) vision, will be fully characterized.

Bipolar cell function


large receptive field, due to their size and coupling, they subtract Bipolar cells receive input from photoreceptors and then conduct
the mean illumination, allowing the photoreceptors to respond the visual signal to the inner retina. They are excitatory interneu-
to local changes in visual illumination. Because they are slow, rons, using glutamate as a neurotransmitter, specialized for sus-
horizontal cells always lag behind and damp the visual signal tained transmitter release, with terminals containing synaptic
without blocking the rapidly changing peaks that carry the most ribbons, similar to those of photoreceptors, only smaller. There
important visual information. In one in silico model of the retina, are approximately 9–12 kinds of cone bipolar cell in the mam-
the bipolar cells amplify the difference between the cone signal malian species that have been studied thoroughly, i.e., primates,
and the horizontal cell network.109 Horizontal cells also may rabbits, cats, rats, and mice (Fig. 15.13).12,129–132 In contrast to the
have other specific roles in vision. Recent studies have led to the cone bipolar cells, there is only one type of rod bipolar cell,
hypothesis that they are responsible for red-green opponency in which is morphologically and physiologically distinct from the
the primate midget ganglion cells.110 cone bipolar cells. Rod bipolar cells are numerous – three times
There is general consensus that horizontal cells provide a the density of any diffuse cone bipolar type. However, there are
negative-feedback signal to cones and rods. When a light flash around 10 types of cone bipolar cell so, in total, they outnumber
hyperpolarizes the photoreceptors and the horizontal cells, the the rod bipolar cells by a factor of three to four.
horizontal cell input results in sign-inverted voltage change in Cone bipolar cells are difficult to identify in a retinal slice
rods.111 This negative feedback results in an antagonistic center preparation. Once filled with a tracer they can be distinguished
surround receptive field in the photoreceptors. The mechanism by the locations of their somas in the INL, by the morphology of
is thought to consist of a shift in the voltage sensitivity of the their dendritic and axon terminals, and by the depth of termina-
calcium channels in the photoreceptors, which results in a left- tion in the IPL. An increasing number of selective antibodies are
ward shift of the calcium channels I/V relationship and increased now available to aid in the identification of specific cone bipolar
currents. Increased currents in photoreceptors inhibit the light- cells.133–136 In addition, several transgenic reporter mouse lines
mediated hyperpolarization and alter glutamate release. that mark specific types of bipolar cells have been created.137,138
The mechanism that mediates feedback remains under consid- There is a major functional division in the cone pathways that
erable debate. Three models have been proposed as the basis of arises at the first synapse and is preserved in all higher visual
negative feedback at this synapse. The first proposed that hori- centers. The retina is divided into ON and OFF channels served
zontal cells release GABA to hyperpolarize the cone photorecep- by ON and OFF cone bipolar cells (Fig. 15.13).15 The ON pathway
tor.112 There are data to support this idea. GABA has been found is optimized to detect increases in intensity and the OFF pathway
in some mammalian horizontal cells,113,114 as has VGAT, the decreases in intensity. Reflecting the importance of depth in the
Rat bipolar cells

373
(OPL)

INL

Chapter 15
IPL

Function and Anatomy of the Mammalian Retina


1 2 3 4 5 6 7 8 9 RB GCL

Primate bipolar cells


DB1 DB2 DB3 DB4 DB5 DB6

FMB IMB

30µm

Fig. 15.13 Bipolar cells. Top: Lucifer Yellow-filled examples of bipolar cell types in the rat retina. (Reproduced with permission from Harveit E.
Functional organization of cone bipolar cells in the rat retina. J Neurophysiol 1997;77:1716–30, with permission from the authors and the
American Physiological Society.) Bottom: drawings of Golgi-impregnated bipolar cells from the primate retina. Note the presence of midget
bipolar cells in the primate retina. (Reproduced with permission from Boycott BB, Wassle H. Morphological classification of bipolar cells of the
primate retina. Eur J Neurosci 1991;3:1069–88.) In both species, it may be observed that the main difference between cell types is the depth of
stratification in the inner plexiform layer. OFF bipolar cells terminate in sublamina a and ON bipolar cells stratify at a deeper level, in sublamina
b. OPL, outer plexiform layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; RB, rod bipolar cells; FMB, flat midget
bipolar; IMB, invaginating midget bipolar.

retina, these two types of bipolar cells terminate at different shown by a + in Figure 15.14, served by ionotropic glutamate
levels in the IPL. OFF bipolar cells ramify in the top half, sub- receptors, i.e., conventional glutamate receptors of the AMPA/
lamina a, where they synapse with OFF ganglion cells. ON kainate type. Thus, the dark release of glutamate from cones
bipolar cells descend further in the IPL to sublamina b, where holds OFF bipolar cells at a relatively depolarized potential
they synapse with ON ganglion cells. Now, ON and OFF bipolar and the reduction of glutamate release in response to light pro-
cells produce opposite signals in response to changes in light duces a hyperpolarization in OFF bipolar cells. In this regard,
intensity. But how is this achieved if both bipolar types contact the photoreceptor/OFF bipolar cell synapse is similar to the
the same cones? The short answer is via different postsynaptic photoreceptor/horizontal cell synapse.
receptors and, indeed, glutamate produces opposing responses Recordings from cone and OFF bipolar cell pairs in ground
in ON and OFF bipolar cells15,139,140 This simple trick, dividing squirrel, which has very large cones particularly suitable for
the cone signal into ON and OFF components, is thought to recordings, has shown that three different OFF bipolar cells use
double the dynamic range of the retina. Half the bipolar cells three different glutamate receptors – one AMPA receptor and
carry signals greater than the local mean and the other half two kainate receptors (Fig. 15.15).141 This dovetails very well
dimmer than the average. with the differential distribution of glutamate receptors at the
cone pedicle complex.90,96 Furthermore, the characteristic rate at
OFF cone bipolar cells which each receptor recovers from desensitization effectively
The dendrites of OFF bipolar cells branch in the OPL and contact divides the cone signal into different bandwidths. The fast
every cone within the dendritic field at basal synapses (Fig. AMPA receptors are well suited to convey transient signals,
15.14). OFF bipolar cells, like cones themselves, are hyperpolar- while the slower kainate receptors may transmit sustained
ized by light. So these are sign-conserving excitatory synapses, responses.141 All three OFF bipolar types terminate in sublamina
374
Section 1

- +

ON OFF
Anatomy and Physiology
Basic Science and Translation to Therapy

B B

+
A
IPL
B +

OFF
ON GC
A GC B
5 µm

Fig. 15.14 ON and OFF pathways. (A) The opposite responses of ON and OFF cone bipolar cells are generated by the expression of different
glutamate receptors. OFF bipolar cells are depolarized by glutamate, the photoreceptor transmitter, hence the + for a sign-conserving synapse.
ON bipolar cells are hyperpolarized by glutamate, hence the – for a sign-inverting synapse. This unusual receptor is called the mGluR6
receptor. It is selectively activated by the glutamate analog 2-amino-4-phosphonobutyrate and it is responsible for the separation of ON and
OFF signals through the visual system. Furthermore, the inner retina is functionally stratified. OFF bipolar cells ramify in sublamina a and ON
bipolar cells descend to sublamina b. Both bipolar cell types use glutamate and they make excitatory synapses with amacrine and ganglion
cells. IPL, inner plexiform layer; GC, ganglion cell. (B) The image shows an OFF bipolar cell that was filled with Lucifer Yellow (green). In
whole mount, with the focus at the outer plexiform layer, fine dendrites extend to contact every cone within the dendritic field. The positions of
rod and cone terminals are marked with a combination of glutamate receptor antibodies mGluR6 (red) and GluR5 (blue). (Panel B reproduced
with permission from Li W, Keung JW, Massey SC. Direct synaptic connections between rods and OFF cone bipolar cells in the rabbit retina.
J Comp Neurol 2004;474:1–12, with permission of the authors and Wiley-Liss, a subsidiary of John Wiley.)

a, as expected, but each one branches at a different depth within to be the transient receptor melastatin 1 (TRPM1) channel.144–146
the OFF sublamina (Fig. 15.15). Thus, the division of the visual The exact mechanism by which mGluR6 activation closes TRPM1
signal into different channels, for delivery to different addresses is not known, but it does not seem to use a cyclic nucleotide-
in the IPL, begins at the first synapse in the retina. mediated mechanism.147,148 While the details of the mechanisms
remain uncertain, the signal inversion that occurs at the ON
ON cone bipolar cells bipolar mGluR6 receptor underlies the separation between ON
and OFF channels throughout the visual system.
The dendrites of ON bipolar cells invaginate into the cone
Labeling the retina with an antibody against the mGluR6
pedicle and approach the synaptic ribbons in a central position.
receptor stains a narrow band at the level of bipolar dendrites in
ON bipolar cells are depolarized by light. This is opposite to the
the OPL.17,97 In whole mount, the distribution of mGluR6 labeling
cone signal so we refer to this as a sign-inverting synapse, hence
shows a distinct pattern with two types of terminals (Fig. 15.15C).
the minus sign at the cone/ON bipolar cell synapse in Figure
There are lightly labeled mGluR6-positive clusters of fine ON
15.14.15 The dark release of glutamate from the photoreceptors
cone bipolar terminals at each cone pedicle and bright mGluR6-
holds ON bipolar cells relatively hyperpolarized. Light turns off
positive terminals, which insert into each rod spherule. These are
the cone transmitter, and the decrease of glutamate release pro-
the terminal dendrites of rod bipolar cells. This mGluR6 pattern
duces a depolarization in ON bipolar cells. Bipolar cells obey the
also provides a simple way to map the location of rod and cone
division of the IPL and so ON bipolar cells are stratified in sub-
terminals in the outer retina. All ON bipolar cells, which include
lamina b (Fig. 15.13).
ON cone bipolar cells, blue cone bipolar cells, and rod bipolar
A hyperpolarizing response to glutamate is unusual and this
cells, express mGluR6 at the tips of their dendrites.17 This is quite
is an unusual receptor, which is only expressed in the retina. It
different from the multiple glutamate receptors used by OFF
has now been identified as mGluR6, one in a series of eight
cone bipolar cells. However, a variety of responses may still be
metabotropic glutamate receptors, so called because activation
produced by the different types of ON cone bipolar cell due to
of these receptors turns on an intracellular signaling cascade.142,143
modulation of the mGluR6 cascade or to the action of calcium
The mGluR6 receptor is selectively activated by glutamate or the
channels or amacrine cells at the bipolar cell terminal.149
glutamate analog 2-amino-4-phosphonobutyrate (APB).15 Acti-
vation of the mGluR6 receptor leads to the closure of a cation Midget bipolar cells
channel, producing a hyperpolarization in ON bipolar cells. The primate retina is unusual in that the central retina is domi-
Light decreases glutamate release from photoreceptors and pro- nated by midget bipolar cells. Within the central 10 mm, there
duces the opposite response. The cation channel is now known is one OFF midget bipolar cell and one ON midget bipolar cell
MASSEY & MILLS 96

375
RB

Chapter 15
RB CB HC
RB HC
RB RB

Function and Anatomy of the Mammalian Retina


B CABP
PKC - rod bipolar cells

b7
b2

b6

A C GluR5
5 µm
mGluR6

Fig. 15.15 Different OFF bipolar cells and rod/cone mosaic. (A) Three bipolar cells from the ground squirrel filled with different-colored dyes. The
two OFF bipolar cells have responses mediated by different classes of glutamate receptor and they stratify at different levels, or addresses, in
the inner plexiform layer. (Reproduced with permission from DeVries Sh. Bipolar cells use kainite and AMPA receptors to filter visual information
into separate channels. Neuron 2000;28:847–56.) (B) A single ON bipolar cell type in the rabbit retina stained for the calcium-binding protein
calbindin (red). For comparison, the more numerous rod bipolar cells are stained for protein kinase C (blue). The calbindin bipolar cell has an
axon that descends to sublamina b where it is narrowly stratified just above the rod bipolar terminals. This is a different address in the inner
plexiform layer. In the outer plexiform layer, horizontal cells are also stained for calbindin. (Reproduced with permission from Massey SC, Mills
SL. A calbindin-immunoreactions cone bipolar cell type in the rabbit retina. J Comp Neurol 1996;366:15–33, with permission of the authors and
Wiley-Liss, a subsidiary of John Wiley.) (C) Staining for mGluR6 receptors (red) mark the tips of rod bipolar cells where they enter the rod
spherule. A small cluster of ON cone bipolar dendrites is also lightly marked at each cone pedicle. GluR5 receptors (blue) mark OFF bipolar cell
basal contacts at each cone pedicle. (Courtesy of F Pan and SC Massey.)

for each cone. In this area, they account for more than 80% of all OFF midget bipolar cells make flat or basal contacts with cones
cone bipolar cells.130,150 Midget bipolar cells have very small den- and terminate in sublamina a where they contact OFF midget
dritic fields and receive input from single cones and make output ganglion cells. Most investigators think this specialization of the
to single midget ganglion cells. This is the so-called private line, primate retina was designed to achieve maximum acuity at high
one cone to one midget bipolar cell to one midget ganglion cell. cone density. It may also, by virtue of the single cone connec-
The ON midget bipolar cells invaginate the cone pedicle, ramify tions, which are automatically color-coded, serve red/green
in sublamina b of the IPL, and contact ON midget ganglion cells. color vision.
Blue cone bipolar cells Rod bipolar cells
In general, diffuse cone bipolar cells contact every cone within In contrast to the cone bipolar cells, there is only one morpho-
376 the dendritic field and this gives them a characteristic appear- logical type of rod bipolar cell. They are numerous, with a
ance. However, in the primate retina, one bipolar cell type is mop-head of fine dendrites that may receive input from as
distinctly different in that it has long dendrites that bypass many many as 80–120 rods in the rabbit retina (Fig. 15.16A).88 This
Section 1

cones to seek out only blue cones.151,152 The dendrites are labeled very high convergence contributes to the sensitivity of the
for mGluR6 and the axon descends deep into sublamina b so the primary rod pathway. A single dye-injected rod bipolar cell is
blue cone bipolar cells are ON cells.17,153 shown in Figure 15.16B. The dendrites branch profusely but

A B
Anatomy and Physiology
Basic Science and Translation to Therapy

10 µm
PKC
5 µm mGluR6
RBP

Fig. 15.16 Rod bipolar cells and contacts in the rabbit retina. (A) There
is a single morphological type of rod bipolar cell that can be stained
with antibodies against protein kinase C (PKC). (Courtesy of W Li and
SC Massey.) (B) A single rod bipolar cell from the rabbit retina, filled
with Lucifer Yellow (green), focus at the outer plexiform layer, has a
spray of fine dendrites that contact many rod spherules (red puncta).
(C) At higher resolution, mGluR6 receptors, which stain the dendritic
tips of the rod bipolar cells (red), are doublets where they enter into
the rod spherule invagination. The general rule is that each pair of rod
bipolar dendrites at a rod spherule originates from different rod bipolar
cells. A cone pedicle, marked by a cluster of dimmer, finer ON cone
bipolar dendrites, is also outlined (dashed line). (B, C, Reproduced
from Li W, Keung JW, Massey SC. Direct synaptic connections
2 µm between rods and OFF cone bipolar cells in the rabbit retina. J Comp
Neurol 2004;474:1–12, with permission from the authors and
Wiley-Liss, a subsidiary of John Wiley.)
Primate Retina

377

Chapter 15
R

ROD

Function and Anatomy of the Mammalian Retina


B

S1/S2 AII


B
Rod
bipolar
+ + B Rod Bipolar Cells
A 10 µm
All amacrine cells

Fig. 15.17 The primary rod pathway. (A) There is only one type of rod bipolar cell (rod B), which produces ON responses to light and terminates
at the bottom of the inner plexiform layer (IPL) in sublamina 5. Rod bipolar cells express mGluR6 receptors and are hyperpolarized by glutamate
released from rods at a sign-inverting synapse, marked with a minus sign. Rod bipolar cells release glutamate and are connected to AII
amacrine cells and S1/S2 amacrine cells via excitatory receptors, marked by a plus sign. In turn, S1/S2 amacrine cells make inhibitory reciprocal
synapses mediated by γ-aminobutyric acid, back on to the rod bipolar terminal. (B) Vertical section of macaque retina, the AIIs amacrine cells,
stained for calretinin (green) descend around the rod bipolar cell axons (protein kinase C, red) as they descend through the IPL. Arrow marks
fainter staining of cone bipolar cell DB4 at a different level in the IPL. (Panel B reproduced with permission from Massey SC, Mills SL. Antibody
to calretinin stains AII amacrine cells in the rabbit retina: double-label and confocal analyses. J Comp Neurol 1999;411:3–18.)

avoid the cone pedicles within the dendritic field. Instead, all pathways, referred to as primary, secondary, and tertiary, by
the terminal dendrites invaginate a rod spherule and they are which rod signals reach ganglion cells (Fig. 15.18). They are
double-labeled for mGluR6. Only one terminal at each rod thought to process signals under slightly different conditions,
spherule comes from this rod bipolar cell. The other half of the primary being the most sensitive and the tertiary being the
each mGluR6 doublet is claimed by another unlabeled rod least sensitive.
bipolar dendrite. Thus, each rod contacts two different rod The primary rod pathway utilizes rod bipolar cells, which
bipolar cells.97 Each rod bipolar cell gives rise to a long slender make ribbon synapses with two postsynaptic amacrine cells in
axon that descends to sublamina 5 of the IPL (Fig. 15.16). Physi- sublamina 5 of the IPL (Fig. 15.17). One of these postsynaptic
ologically, rod bipolar cells give ON responses to light stimula- neurons is the AII amacrine cell, and the other is either an S1
tion and this is consistent with the labeling for mGluR6 and (A17 in the cat) or S2 amacrine cell (Fig. 15.17).42,154 These two
the depth of stratification.13,154 Rod bipolar cells do not usually wide-field GABA amacrine cells both make reciprocal synapses
contact ganglion cells directly. Instead, the primary output of with rod bipolar terminals and thus provide another stage of
rod bipolar cells is to AII amacrine cells, which pass on the negative feedback. The conspicuous terminals of rod bipolar
rod signal, or to S1 and S2 amacrine cells42,154 that provide a cells literally plug into holes in the meshwork of dendrites pro-
powerful negative-feedback signal to rod bipolar terminals (Fig. vided by these amacrine cell types. Rod bipolar cells, like other
15.17; see below). More than 90% of the rod bipolar output is bipolar cells, release glutamate and the contact points with the
on to these amacrine cells. AII matrix are covered with glutamate receptors of the AMPA
The high convergence allows the rod bipolar cell to collect subtype (Fig. 15.19).157,158 The glutamate receptors of S1/S2 ama-
signals from many rods but is also potentially noisy. However, crine cells have not been completely clarified, but the A17 (prob-
the rod-to-rod bipolar synapse has a nonlinearity by which small ably the S1 equivalent in the rodent) uses a Ca2+-permeable
signals are thresholded.155 The price for this is that many small AMPA glutamate receptor via L-type Ca2+ and Ca2+-activated
signals are rejected but the reduction in noise is worth it. Some potassium (BK) channels159 to modulate feedback inhibition on
near-threshold signals may be lost but when a photon signal is to the rod bipolar cell and control glutamate release at this
captured it has a high signal-to-noise ratio and is transmitted synapse. On the rod bipolar cell the postsynaptic targets are the
very reliably.156 GABAA and GABAC receptors.160–162
So, there are two obvious questions: how do rod signals reach
Multiple rod pathways ganglion cells and, if there is only one type of rod bipolar cell,
Rods are utilized under low light conditions and provide infor- how are both ON and OFF signals generated in dark-adapted
mation over 5 log units. It is now clear that there are at least three conditions? The answer lies in the way the rod pathway is
Primary rod pathway Secondary rod pathway Tertiary rod pathway
378
Section 1

Cone Cone Cone


Anatomy and Physiology
Basic Science and Translation to Therapy

OFF- OFF- OFF-


Rod BC Cone BC Cone BC
ON- ON- ON-
BC AII- BC BC
AC

OFF- OFF- OFF-


GC GC GC
ON- ON-
GC GC

Gap-junction Excitation Inhibition

Fig. 15.18 Rod and cone pathways. The primary rod pathway (rod → ON bipolar cell → AII amacrine cell) provides an excitatory input to AII
amacrine cells, which are electrically coupled to cone ON bipolar cells and inhibit cone OFF bipolar cells. The secondary rod pathway (rod →
cone → bipolar cell) arises from electrical coupling between rods and cones and allows the flow of rod signals into the cones and glutamate
release into the cone ON and OFF bipolar cells. The tertiary rod pathway (rod → OFF bipolar cell) is a direct glutamatergic input from rods to
cone OFF bipolar cells. Note that the secondary and tertiary pathways are in principle independent of the primary pathway. Red arrows indicate
sign-inverting synapses, and green arrows indicate sign-preserving synapses. (Modified with permission from van Genderen MM, Bijveld MM,
Claassen YB, et al. Mutations in TRPM1 are a common cause of complete congenital stationary night blindness. Am J Hum Genet 2009;85:
730–6.)

integrated into the cone pathways via the AII amacrine cell heterocellular network is modulated by dopamine and, perhaps
(Fig. 15.18). AII amacrine cells are glycinergic neurons and they more importantly, by light.169 The underlying mechanism is by
also make conventional inhibitory glycinergic synapses with the phosphorylation of Cx36, and this regulation can be achieved on
axon terminals of OFF cone bipolar cells in sublamina a via alpha a cell-by-cell basis.170
1 glycine receptors.163 In turn, the AII itself is modulated by a These gap junctions are bidirectional. This means that electri-
glycinergic input at synapses expressing alpha 3 glycine recep- cal signals and tracers can pass through the gap junction in both
tors.164 Their distal processes make electrical synapses or gap directions.171,172 One consequence is that glycine from the AII
junctions with ON cone bipolar cells in sublamina b and provide amacrine cell can enter ON cone bipolar cells by this route and
a direct, presumably sign-conserving, input signal to the ON indeed most ON bipolar cells contain glycine, even though they
cone bipolar cells.43,165,166 The signal transfer via gap junctions is use glutamate as a neurotransmitter. The source of the bipolar
presumed to be sign-conserving. Thus, while the cone pathways cell glycine was definitively established by blocking gap junc-
split via different postsynaptic glutamate receptors in the outer tions with carbenoxolone. This changed the labeling pattern for
retina, the rod pathway bifurcates at the level of the AII amacrine glycine, which was subsequently diminished in bipolar cells.167
cell. It is often said that the rod signals piggyback on the cone Another more relevant consequence of bidirectional coupling is
pathways. that not only do rod signals pass into the cone pathways but, in
AII amacrine cells themselves are well coupled by gap junc- fact, cone signals can pass from ON bipolar cells into the AII
tions, as can be demonstrated by injecting a single AII amacrine network. The implication is that the AII network can also influ-
cell with the diffusible tracer Neurobiotin, which passes through ence cone signals.
gap junctions to label all the coupled cells.166,167 Figure 15.20 The importance of these gap junction pathways has been ele-
shows that many AII amacrine cells are coupled as well as an gantly demonstrated by the development of a Cx36 knockout
overlying group, consisting of four or five different types, of ON mouse.168 In these animals, filling an AII amacrine cell with Neu-
cone bipolar cells. The AII-to-AII gap junctions occur preferen- robiotin yielded only one cell: there was no coupling without the
tially at dendritic crossings (Fig. 15.21) and AII coupling is expression of Cx36. In contrast to the wild type, in the knockout
absent in a Cx36 knockout mouse.168 Coupling in this complex animals, no rod signals are detectable in recordings from ON
379

Chapter 15
Function and Anatomy of the Mammalian Retina
A 5 µm B
GluR4 All(CR) RB(PKC)

Fig. 15.19 Rod bipolar input to AII is mediated by glutamate receptors. (A) The matrix of AII amacrine cells in the rabbit retina, stained for the
calcium-binding protein calretinin (green), is decorated with punctate glutamate receptors (red) of the α-amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid (AMPA) subtype. (B) The AII dendrites carefully wrap around the rod bipolar terminals (blue) at this level. Critically, the
glutamate receptors only occur at the contact sites between rod bipolar cells and AII amacrine cells (red + green + blue = white). Intervening sections
of AII dendrite, between rod bipolar terminals, have no glutamate receptors. This indicates that AMPA receptors are located at synaptic contacts
between rod bipolar cells and AII amacrine cells. (Reproduced from Li W, Trexler EB, Massey SC. Glutamate receptors at rod bipolar ribbon synapses
in the rabbit retina. J Comp Neurol 2002;448:230–48, with permission from the authors and Wiley-Liss, a subsidiary of John Wiley.)

ON Cone
Bipolar Cells

All amacrine
Cells

Fig. 15.20 Coupling between AII amacrine cells and ON cone bipolar
cells. This three-dimensional reconstruction from a series of confocal
images shows the result of injecting a diffusible tracer, such as
Neurobiotin, into an AII amacrine cell in the rabbit retina. A matrix
of coupled AII amacrine cells (khaki), all with the characteristic
morphology, is labeled. In addition, the tracer also spreads into a
complex of ON cone bipolar cells (blue) which consists of 3–5 types.
(Courtesy of EB Trexler and SC Massey.)

Connexin 36 5 µm
ganglion cells. In the absence of Cx36 gap junctions, rod level All Amacrine Cells
signals do not pass into the ON cone pathways. Of course, the
Fig. 15.21 AII/AII gap junctions contain connexin (Cx)36. A confocal
OFF pathways are not dependent on transmission via gap junc- image of the AII dendrites (red) in sublamina b of the inner plexiform
tions and so rod-driven OFF signals are maintained. One obvious layer. Staining for Cx36 (green) shows that Cx36 plaques are abundant
explanation is the absence of AII/ON bipolar gap junctions, in the AII matrix. Image analysis indicates that there is a high probability
described above. However, the Cx36 knockout may also inter- of a Cx36 plaque when two AII dendrites cross. (Reproduced from Mills
SL, O’Brien JJ, Li W, et al. Rod pathways in the mammalian retina use
fere with rod/cone coupling in the outer retina. In fact, both connexin 36. J Comp Neurol 2001;436:336–50, with permission from the
these pathways must be missing to eliminate the transmission authors and Wiley-Liss, a subsidiary of John Wiley.)
of rod signals to ON pathways. In either case, this is the first time pathways may be designed to cover different intensity ranges
that a gap junction connection has been shown to be essential and perhaps they are selectively connected to specific ganglion
380 for the function of a neuronal pathway in the mammalian CNS. cell types but, as yet, there are no data on this point. There is
In the rabbit retina, the gap junctions between one type of ON physiological evidence from the mouse retina that different gan-
cone bipolar cell that is selectively labeled by antibodies against glion cell types have different intensity response functions but
the calcium-binding protein calbindin also involve Cx36.173 the ganglion cell types have not been identified and the contrib-
Section 1

However, the properties of AII/bipolar gap junctions are differ- uting pathways are unknown.168 The function of these novel
ent from AII/AII gap junctions,166 suggesting that the bipolar retinal pathways under different light intensities is an important
side of the gap junction is different in some way, perhaps due and active area of current research.
to phosphorylation or even the expression of a different con-
nexin by bipolar cells. There is some evidence that bipolar cells Amacrine cells
express Cx36.168 However, there is convincing evidence that in
Anatomy and Physiology
Basic Science and Translation to Therapy

Amacrine cells form a morphologically and physiologically


some ON bipolar cells the coupling is mediated by Cx36 (AII)/
diverse group of mostly inhibitory interneurons. Approximately
Cx45 (BC) heterotypic gap junctions.174
30 types of amacrine cell have been identified morphologically
Secondary and tertiary rod pathways in mammalian retina (Fig. 15.22).37,38,181 The cell bodies of the
The primary rod pathway is used under threshold conditions amacrine cells are primarily located in the innermost portion of
such as starlight108,154 and it is absolutely dependent on synaptic the INL, unofficially known as the amacrine cell layer. Amacrine
transmission between rods and rod bipolar cells via mGluR6 cells are also found in the GCL, where they are known as dis-
receptors. However, when these synapses are blocked with APB placed amacrine cells. Most are GABAergic medium- to wide-
it was still possible to record rod level signals in OFF ganglion field cells49,182 and many recognized classes of amacrine cells are
cells.175 This alternative pathway was attributed to rod/cone found in both layers. In fact, in the mouse retina, more than half
coupling, for which there is both anatomical and physiological the cells in the GCL are amacrine cells. In general, the three major
evidence. This second rod pathway runs from rods via gap junc- connections made by amacrine cells are feedback inhibition to
tions to cones and then into cone bipolar cells and ganglion cells bipolar cell terminals, feedforward inhibition to ganglion cells
in the usual way (Fig. 15.18). It has been suggested that this and serial inhibitory connections where amacrine cells are the
pathway operates in mesopic conditions of intermediate light postsynaptic targets of other amacrine cells.183
intensity.76 Amacrine cells can be divided by their dendritic arbor size into
This pathway has also been reported for the mouse retina. narrow- (<200 µM) and wide-field. The narrow-field amacrine
However, the rod-driven responses persisted even in a trans- cells are generally glycinergic and frequently provide inputs
genic mouse line in which cones were eliminated.176 The absence across the ON/OFF strata in the IPL.184 Wide-field amacrine cells
of cones would seem to rule out rod/cone coupling as a pathway. are mostly GABAergic.181,182,185–187 Some provide lateral inhibition
Therefore, a third rod pathway was proposed involving direct within a substratum184 and others provide reciprocal feedback.188
connections between rods and OFF cone bipolar cells and sub- They are involved in surround inhibition189 or contrast adapta-
sequently these connections were found.177,178 This pathway was tion.190 Several of the wide-field cells also release catecholamines
first thought to be a specialization of the rodent retina but it has or other neuropeptides.114 The best characterized in this group
now been identified in cats and rabbits.97,179 It also is present in are the dopaminergic wide-field amacrine cells that are involved
ground squirrel, where a specific type of OFF bipolar cell, the in light adaptation, and those releasing ACh.
b2, was shown to contact rods and provide a means for rods to In a large population of rabbit amacrine cells, acquired ran-
provide fast photoreceptor signaling.180 Thus, this third rod domly using modern imaging methods, 24 different morpho-
pathway may be a general feature of the mammalian retina. logical types were identified (Fig. 15.22).37,38 Similar studies
It has been suggested that the rod to OFF bipolar pathway have been extended to the mouse, where several groups have
operates at high mesopic/low photopic conditions, around the identified large numbers of narrow-, medium-, and wide-field
transition from rod to cone vision. In general, the different rod amacrine cells.181,182,185–187

Amacrine cells

Fig. 15.22 Amacrine cell morphology. This figure shows the comparative morphology of 24 distinct amacrine cell types from the rabbit retina.
(Reproduced with permission from Masland D. Neuronal diversity in the retina. Curr Opin Neurobiol 2001;11:431–6.)
The most numerous amacrine cell type is the AII amacrine AII amacrine cells
cell, approximately 11%, while other relatively common types, The AII amacrine cell, also known as the rod amacrine cell, is the
such as the ChACs, make up 3–5% of the total population. Half most numerous of the amacrine cells, accounting for 11% 381
of the amacrine cells are narrow-field, with coverage factors of the total. It is correctly written with a Roman numeral, which
around 1. Many of these are broadly stratified, which suggests is retained from an early classification scheme, while other num-

Chapter 15
they may interconnect the ON and OFF substrata. Approxi- bered amacrine cells use Arabic numerals. AII amacrine cells
mately a quarter of the amacrine cells are wide-field and nar- have a distinctive bistratified morphology, which makes them
rowly stratified with very high coverage – as high as 100–500. easy to identify, even in retinal slices.199 The soma protrudes into
A cell with an overlap this high forms an extremely dense plexus the IPL and turns into a thick axon that descends to sublamina b
of fine dendrites that blankets the entire retina. Examples are of the IPL and branches into an overlapping matrix.200 This is the
S1 amacrine cells, DACs, and starburst amacrine cells (see site of glutamate input from rod bipolar cells (Fig. 15.20) and
below). Thus, in the rabbit retina, the identification of all the output, via gap junctions to ON cone bipolar cells. There are also

Function and Anatomy of the Mammalian Retina


amacrine cell types, started so long ago by Cajal and his con- fine dendrites, which terminate in lobules in sublamina a. At this
temporaries, is now almost complete. The mouse is probably level, AII amacrine cells make glycinergic inhibitory contacts
the next best characterized because of the array of genetic tools with OFF cone bipolar cells. They also receive input from OFF
that can be used for their analyses. cone bipolar cells at this level, the function of which is unclear.154
The reasons for such a variety of amacrine cells are unknown. Primate AII amacrine cells are similar in morphology to those
However, two functions seem to be commonly repeated. First, of other mammalian species.61,62 The density of rods reaches a
all bipolar cell terminals seem to receive GABA-mediated nega- maximum at about 20° from the fovea but psychophysical mea-
tive feedback. In the case of the rod bipolar cell, feedback is surements in humans indicate that, in dark-adapted conditions,
mediated by two amacrine cell types, S1 and S2.191 Bipolar cells the maximum acuity occurs at approximately 5° from the fovea,
(approximately 10 types) are narrowly stratified and if different a substantial mismatch. The rod pathway for maximum acuity
amacrine cells mediate feedback to each one, this could account is thought to be rods → rod bipolar cells → AII → midget bipolar
for 10–20 different cell types. Secondly, about half the ganglion cells → midget ganglion cells (Fig. 15.18). There is good evidence
cells (approximately eight types) are coupled to one or two ama- that AII amacrine cells contact midget bipolar cells and other
crine cell types by gap junctions.192–194 Again, if different ama- ganglion cell types are too sparse to support the maximum reso-
crine cells are coupled to each ganglion cell type, this could lution. Now, rods and rod bipolar cells far outnumber AII ama-
account for 10–15 types. These two groups need not be mutually crine cells so they will not present a limit to scotopic acuity. In
exclusive and doubtless amacrine cells have many functions. In addition, both ON and OFF midget bipolar cells and ON and
another calculation, we estimate there are 15 ganglion cell types OFF midget ganglion cells have a 1 : 1 : 1 correspondence with the
forming independent circuits or channels. If each channel tightly packed cones of the central retina. That leaves the AII
required two amacrine cells, that would produce a total of 30 amacrine cell as the lowest density, the bottleneck, in the
amacrine cell types. While necessarily vague, these simple esti- pathway. In macaque retina, the peak density of AII amacrine
mates indicate the variety of amacrine cell types may not be cells is about 5000 cells/mm2 around 5°, which matches the peak
extravagant. of dark-adapted acuity. If we consider the mosaic of AII ama-
General functions attributed to amacrine cells include feed- crine cells and, from sampling theory, calculate the maximum
back inhibition, surround inhibition, some forms of adaptation, resolution for such an array, we find that it closely matches the
signal averaging, and noise reduction.195,196 In a rapid eye move- peak acuity from psychophysical experiments. Together, these
ment or saccade, the visual image does not blur. This is because results indicate that the density of AII amacrine cells sets the
a wave of inhibition sweeps the inner retina, like a vertical blank limit of dark-adapted resolution in central retina.
on a television screen. In one study, the inhibitory wave associ- The AII network seems to be very important since it is found
ated with saccades was attributed to wide-field amacrine cells.197 in all mammalian retinas. In general terms, the AII network is
However, in terms of specific connections and neuronal circuits, thought to reduce noise by canceling out random events which
the functional anatomy of amacrine cells, it has only been pos- are not correlated and by reinforcing light-driven events that are
sible to study a few types, four of which are selected below. synchronized.201,202 Coupling is also strongly modulated by
We know almost nothing except the shape of 75% of the ama- dopamine169 and processes from the DACs make a ring around
crine cells, not their connections, physiology, or circuit functions the neck of each AII where it protrudes into the IPL (Fig. 15.23).
– in other words, little more than Cajal knew 100 years ago. As Of course, we think that dopamine is the general signal for light
the means of targeting, imaging, and recording from visually adaptation and, in fact, light itself seems to modulate the strength
identified cell types improves, we can expect further progress of coupling in the AII network.203 Disrupting the AII network
in this area. interrupts the rod-driven ON pathways, as described above.
An example of these new approaches was demonstrated by However, even in the rod-driven OFF pathways, which are not
Knop and colleagues,198 who used a GFP transgenic mouse line routed via AII gap junctions, the sensitivity is reduced by
that marked a type 2 wide-field amacrine cell. They character- approximately 1 log unit in the Cx36 knockout mouse.203 This
ized the inputs, stratification, and physiology of the cell, showing indicates that coupling in the AII network adapts to ambient
it was ON-OFF and GABAergic and that its function was main- lighting conditions to optimize rod signaling over a large
tained over a wide range of light adaptation conditions. A major dynamic range.
goal of visual neuroscientists is to characterize fully all the ama- The AII amacrine cells also appear to have a day job.204–206
crine cells and integrate their function into the retinal circuitry. During the day, dark objects that approach the viewer activate
Several amacrine cells have been studied extensively, and their a particular class of ganglion cells. An important component of
characteristics are described below. the circuit is the AII. However, unlike in the dark, the
information flow is effectively reversed. Depolarization of cone and the varicosities are smaller but more numerous (Fig.
ON bipolar cells results in depolarization of the AII through gap 15.24B).208,209 Both cell types contribute to a dense overlapping
382 junctions, which increases inhibition by the AII on to OFF bipolar meshwork at the level of the rod bipolar terminals (Fig. 15.25).
cells. This is a crossover inhibition pathway by which ON chan- These large cells are relatively numerous so the dendritic overlap
nels inhibit OFF pathways.207 The dual use of the AII amacrine is huge with coverage factors as high as 500. Although the rabbit
depending on lighting condition maximizes circuit efficiency. retina contains little endogenous serotonin, for some unknown
Section 1

reason these cells take up serotonin. Therefore, they are some-


S1 and S2 amacrine cells
times known as indoleamine-accumulating amacrine cells and
The S1 amacrine cell of the rabbit retina (A17 in the cat) is a
this provides a simple way to label the entire population. Elec-
wide-field GABA amacrine cell with straight radiating dendrites
tron microscopy shows that S1/S2 amacrine cells make recipro-
decorated with prominent varicosities (Fig. 15.24A). The S2
cal synapses with rod bipolar terminals, i.e., they receive input
GABA amacrine cell is smaller, the dendrites are more tangled,
at a rod bipolar ribbon synapse and nearby they synapse back
Anatomy and Physiology
Basic Science and Translation to Therapy

on to the rod bipolar terminal.42


The varicosities contain presynaptic markers; they are wrapped
around rod bipolar terminals (alternating with AII dendrites)
and opposed by synaptic ribbons and GABA receptors (Fig.
15.26). In fact, the varicosities are synaptic sites. Confocal analy-
sis of double-label material shows that every varicosity is in
synaptic contact with a rod bipolar terminal.209 This means that
the functional role of the S1/S2 amacrine cells is to provide a
level of GABA-mediated negative feedback to rod bipolar termi-
nals. This is their job and they do nothing else; they have no
output to any other cell besides the rod bipolar cell.
These cells provide a massive inhibitory input to rod bipolar
terminals. S1/S2 amacrine cells carry about 300 and 500 varicosi-
ties, respectively, and the density of varicosities has been calcu-
lated as 330 000/mm2. Each rod bipolar terminal receives input
from about 25 S1 and 50 S2 varicosities. They are both apposed
by GABAA and GABAC receptors.210 Thus, the inhibitory input
to bipolar terminals will have two components: a large sustained
component from GABAC receptors, which are only expressed by
bipolar terminals, and rapid initial transient input from GABAA
receptors, which are more widespread.138,160,189,211–213 The S1 and
All amacrime cell S2 amacrine cells clearly have different spatial and coupling
Dopamine amacrine cell
properties.191 Hence, the S2 input dominates within 200 µm and
provides an inhibitory signal that matches the size of the sur-
Fig. 15.23 Dopaminergic amacrine cells form a dense matrix. The round recorded from AII amacrine cells. The larger, well-coupled
matrix of dopaminergic dendrites, stained for tyrosine hydroxylase
S1 amacrine cell may provide a more distant network or global
(green), blankets the retina. There are many small varicosities
surrounding AII amacrine somas, stained for calretinin (red). signal. This analysis suggests that the presence of both S1 and
(Courtesy of SC Massey.) S2 amacrine cells is not redundant: each cell contributes different

A B

100 µm
S1 amacrine cell S2 amacrine cell

Fig. 15.24 (A) S1 and (B) S2 amacrine cells filled with Lucifer Yellow, from the rabbit retina. These amacrine cells have prominent varicosities,
which are synaptic structures, along slender dendrites. (Reproduced with permission from Zhang J, Li W, Trexler EB, et al. Confocal analysis of
reciprocal feedback at rod bipolar terminals in the rabbit retina. J Neurosci 2002;22:10871–82.)
383

Chapter 15
Function and Anatomy of the Mammalian Retina
A B

C 10 µm D

Fig. 15.25 S1/S2 matrix. (A) The S1/S2 matrix (green), stained by serotonin uptake, from whole-mount rabbit retina. (B) Rod bipolar terminals
(blue) fill holes in the S1/S2 matrix. (C) AII dendrites (red) also contact the same rod bipolar terminals. (D) Merged triple-label image shows S1/
S2 processes and AII dendrites surrounding every rod bipolar terminal. (Reproduced with permission from Zhang J, Li W, Massey SC. Confocal
analysis of reciprocal feedback at rod bipolar terminals in the rabbit retina. J Neurosci 2002;22:10871–82.)

components of lateral inhibition in the rod pathway. Together, Ca2+ channels. In conjunction with the cable properties of the A17
these components will summate to modulate the spatial and neurites the outputs are compartmentalized, keeping individual
temporal properties of rod bipolar output. Further, each of the reciprocal synaptic dyads independent.159,188,214 Such local pro-
synaptic sites of S1 (A17) amacrine cells may operate indepen- cessing has an important consequence: it allows a single A17 to
dently of the whole. This is possible because each varicosity is process upwards of 500 signals independently of each other.
electrically isolated from its neighbor. This was tested recently This represents a large increase in processing power based on
by Diamond and colleagues, who showed that at this reciprocal what amounts to parallel circuits in a single cell.188
synapse of the rod ON bipolar cell and the A17 amacrine cell, It should be emphasized that the reciprocal feedback described
BK channels control calcium influx via L-type voltage-dependent at the rod bipolar terminal is, in fact, a general case. It is likely
plexus is in sublamina 1, adjacent to the INL, but there are minor
bands in sublaminae 3 and 5 of the IPL. In some species, particu-
384 larly fish, the dopamine neurons project to the OPL. In other
words, they are interplexiform cells. In the rabbit retina, a few
stunted processes run towards the OPL but they do not form a
plexus. In the macaque retina, some dopaminergic dendrites run
Section 1

into the INL where they surround the somas of AII amacrine
cells (Fig. 15.23).62
Electron microscopy shows that the dendrites in layer 1 receive
direct bipolar input at ribbon synapses, many of which appear
to be monads, as opposed to the usual dyadic arrangement with
two postsynaptic processes.218 Yet, recordings from GFP-labeled
Anatomy and Physiology
Basic Science and Translation to Therapy

cells in the mouse retina have shown that DACs produce a


variety of light responses: ON transient, ON sustained and light
independent.23,24 But how do these ON responses arise if most
dopaminergic processes are stratified in the OFF sublamina? If
DACs are ON cells, then they break the rules of stratification for
the IPL.
Several different mechanisms have been proposed to over-
come this difficulty. For example, it has been shown that ON
bipolar inputs to the DACs occur on the minor bands in layer
3.219 However, it is also clear that there are bipolar ribbon inputs
to the DAC in sublamina 1.218 Most recently, it has been sug-
Fig. 15.26 S1 synapse on a rod bipolar terminal. This three- gested that the melanopsin-containing ipRGCs may provide an
dimensional reconstruction shows a single varicosity from an S1
amacrine cell (green) wrapped around a rod bipolar terminal (blue). input to the DACs, because the sustained features of the DAC
The transparency has been adjusted so we can see through these response and the spectral signature match those of the melan­
structures. GABAC (red) receptors are found on the surface of the rod opsin ganglion cells.24 Furthermore, the light-evoked DAC
bipolar terminal, including at the interface with the S1 (red + green +
response is maintained when photoreceptors degenerate,
blue = white). This is an example of locating a synaptic structure by
confocal microscopy. (Courtesy of W Li and SC Massey.) although transient light responses are blocked by APB, suggest-
ing they arise from the ON bipolar pathway.23 In addition, mela-
nopsin ganglion cell dendrites that ramify in sublamina a also
have ON responses, thus breaking the stratification rules them-
that the terminals of all bipolar cells make reciprocal synapses
selves.25,27 Most recent evidence suggests that many ON cone
with GABA amacrine cells and bipolar terminals are literally
bipolar cells make axonal ribbon or ectopic synapses as their
surrounded by GABA-positive profiles. In the mouse retina, as
axons descend through stratum 1 (Fig. 15.27). These axonal
well as other species, GABAC receptors, a postsynaptic target of
ribbons provide ON excitatory input via AMPA-type glutamate
feedback inhibition, are expressed on cone ON and OFF bipolar
receptors to DACs (and to ipRGCs) in the OFF layer of the IPL.
cells. The current mediated by this receptor is slow and sus-
Thus, axonal ribbons break the stratification rules of the IPL and
tained, making it a likely target of feedback control on the cone
provide an additional accessory ON sublayer in the outer IPL.26,27
bipolar cells, as it does in rod ON bipolar cell. The absence (in
In the mouse retina, DACs also contain GABA220 but DACs
GABAC receptors knockout mice) or pharmacological block of
from the rabbit retina do not display a GABA signature.34 Dis-
this receptor increases both the spontaneous activity and gain of
sociated DACs are spontaneously active221 and, in contrast to
ON-center ganglion cells relative to wild-type.213,215,216
GABA, dopamine seems to be steadily released in a paracrine
There seem to be GABAC receptors on all bipolar cell termi-
fashion to diffuse throughout the retina.222 Dopamine levels are
nals, although the density for each cell type varies.211–213 If each
higher in the daytime and seem to be under circadian control.
bipolar cell type, with its unique address, had one GABA ama-
The most recent work indicates that the dopaminergic cells
crine cell type, that would account for 10–12 of the amacrine cell
themselves express clock genes.223–226 This is appropriate because
types. The “sharing” of some broadly stratified amacrine cells
dopamine is intimately involved in the diurnal rhythms associ-
would tend to reduce this number while those cases, like the rod
ated with the processes of light and dark adaptation. Dopamine
bipolar cell, which receive feedback inhibition from two ama-
acts at a variety of sites in the retina, many of which seem obvi-
crine cell types, will inflate it. In either case, negative feedback
ously linked to light or dark adaptation.227 For example, coupling
provides greater stability, increased frequency response, and a
in the AII network is dramatically reduced by dopamine169,170
wider bandwidth at the level of bipolar cell terminals. A major
and, in the outer retina, dopamine acts to promote cone input to
role for at least some of the multiple types of GABA amacrine
second-order neurons. Conversely, in the dark and absence of
cells is to provide negative feedback for all bipolar cells.
dopamine, rod inputs predominate. Specifically, by controlling
Dopaminergic amacrine cells rod/cone coupling, it seems like dopamine in the light, or its
A small number of amacrine cells, less than 0.1%, contain dopa- absence in the night, presets rod and cone pathways as appropri-
mine. To make up for their low density, these cells have very ate for the lighting conditions.82,228
long axon-like processes that can run for millimeters across the A role for D1 receptors has recently been shown to be critical
retina.217 The net result is a dense high-coverage plexus of dopa- in the GABAergic control of the rod ON bipolar cell circuit that
minergic dendrites, which covers the entire retina. Most of the enhances light sensitivity and increases the dynamic range of
TOH
RIBEYE 385
Calbindin

Chapter 15
Function and Anatomy of the Mammalian Retina
A 20 µm B 5 µm

Fig. 15.27 Dopaminergic amacrine cells, stained with an antibody against tyrosine hydroxylase (TOH: blue), receive ectopic, axonal ribbon
synaptic input. (A) The dopaminergic dendrites run in sublamina a. At this level, ON cone bipolar cells, labeled with an antibody against calbindin
(red), are passing through so the descending axons appear as dots. However, they are nearly all immediately adjacent to the dopaminergic
dendrites (arrows). (B) At higher magnification, it can be seen that synaptic ribbons (green) also occur at these contact points. Thus, the arrows
show ON synaptic input, via axonal ribbons, in sublamina a, the OFF layer of the inner plexiform layer. (Reproduced with permission from Hoshi
H, Liu WL, Massey SC, et al. ON inputs to the OFF layer: bipolar cells that break the stratification rules of the retina. J Neurosci 2009;29:
8875–83.)

their responses at dim-light levels.229 The location of this control vesicles but whether cotransmission occurs at the same sites is
remains to be discovered and could occur via a novel dopamin­ unknown.233
ergic to GABAergic amacrine serial synapse in the IPL or a hori- The starburst amacrine cells have been a focus of attention
zontal cell-mediated input modulated by dopamine in the outer because they seem to be directly involved in neuronal circuits
retina. The retina is like a self-optimizing network controlled, at that produce DS responses in certain ganglion cells. DS ganglion
least in part, by the circadian release of dopamine. cells were discovered nearly 50 years ago and, although the
mechanism has been under intense investigation, the details
Starburst amacrine cells have still not been resolved. However, starburst amacrine cells
The ChACs, also known as starburst amacrine cells on account are in the middle of the puzzle. Ablating starburst amacrine
of their unique morphology, form two mirror-symmetric popu- cells, in a mouse line engineered to respond to an immunotoxin,
lations on either side of the IPL in all mammals.20 They are the blocked DS responses in ganglion cells.234 Furthermore, opto­
only source of ACh in the retina. In whole-mount view, they are kinetic nystagmus, a type of reflex eye movement elicited by a
radially symmetric with dendrites radiating from the soma in all moving stimulus, was also blocked. This combination of physi-
directions (Fig. 15.28). The bipolar cell inputs, via AMPA-type ological and behavioral data provides convincing evidence that
glutamate receptors, occur all along the branches but the outputs starburst amacrine cells are required for directional selectivity
seem to be restricted to the varicosities in the terminal third of and optokinetic eye movements.
each dendrite. The cells in the INL produce OFF responses and Starburst amacrine cells are found at the same depth as the
stratify in sublamina a while the displaced starburst amacrine bistratified ON/OFF DS ganglion cells, which are the most sensi-
cells are ON cells branching in sublamina b. They are relatively tive of all ganglion cells to ACh. But cholinergic input alone is
numerous wide-field amacrine cells. Consequently, they have a not sufficient to produce DS responses.235 Rather, the mechanism
very high coverage factor and in cross-section their dendrites seems to depend on asymmetrical GABA inhibition that comes
form two continuous bands in the IPL that look like train tracks from the starburst amacrine cells. In dual recordings, stimulating
in cross-section (Fig. 15.28). This is a very dense, narrowly strati- a starburst amacrine cell on the null side produced an inhibitory
fied matrix of overlapping dendrites that are cofasciculated with GABA input to DS ganglion cells.233,236
the direction selective (DS) ganglion cell types.230,231 Furthermore, calcium imaging by two-photon microscopy
Starburst amacrine cells also contain GABA.232 In fact, we showed that individual dendrites of starburst amacrine cells can
should probably think of these cells as wide-field GABA ama- themselves produce directional responses, even though record-
crine cells, which also contain ACh. The presence of two classical ings from the soma do not.33 The dendrites respond better to a
neurotransmitters in the same cell is unusual and it has aroused centrifugal stimulus, moving away from the soma, than a cen-
a great deal of interest. The release of both transmitters seems to tripetal stimulus, towards the soma. The directional signals are
be dependent on calcium, which indicates a conventional mech- due to the intrinsic morphological properties of starburst ama-
anism using synaptic vesicles. But GABA and ACh release have crine cells and a voltage gradient generated by the asymmetric
a differential sensitivity to calcium. This implies that the two distribution of voltage-dependent channels.237,238 It has also been
transmitters may be released from different populations of suggested that a chloride gradient generated by the regional
synaptic connections, identified by varicosities containing pre-
synaptic machinery, with ON/OFF DS ganglion cells with an
386 antiparallel null axis. Thus, preferred excitation in starburst den-
drites can provide the GABA input for null inhibition to ON/
OFF DS ganglion cells. Often, the starburst dendrites and the
A
ON/OFF DS ganglion cell dendrites are oriented nearly parallel
Section 1

DAPI
and it is well known that these two cell types are cofasciculated
Starburst at the level of the two cholinergic bands. Once a local DS response
amacrine cell is generated in the ganglion cell, dendritic spikes propagate to
the soma with a high probability of generating somatic spikes.241
After 50 years, the neuronal circuitry underlying directional
selectivity has finally been revealed. How this arises develop-
Anatomy and Physiology
Basic Science and Translation to Therapy

mentally and what the role is for cholinergic input are further
important questions that remain.
Finally, the network of starburst amacrine cells appears to play
a key role in retinal development.242 Imaging of neonatal retina
shows the presence of calcium waves, which propagate across
the retina and elicit bursts of firing in all ganglion cells. These
waves apparently are not required to establish directional selec-
tivity, which arises independently of both waves and visual
experience.243 However, in one model, wave activity is thought
to promote the development of ganglion cell connections in a
B retinotopic manner. The cholinergic plexus is present surpris-
Massey and Mills ingly early in development and, in the early stages, the calcium
ChAC
waves are blocked by cholinergic antagonists.244–246 This suggests
that spontaneous activity in the ChACs may initiate the calcium
INL waves. This was thought to be light-independent; however, it
now appears that light input via ipRGCs (light-sensitive gan-
IPL
glion cells: see below) is required for aspects of the spiking that
are important for retinogeniculate segregation.247
C GCL Ganglion cells
Fig. 15.28 Starburst amacrine cells. (A) The somas of displaced Ganglion cells are the output neurons of the retina and they
starburst amacrine cells stained with 4’,6-diamidino-2-phenylindole receive input from bipolar cells, at ribbon synapses, via AMPA-
(DAPI). This method is used to target this cell type. (B) A single type glutamate receptors.248 In addition, there are even more
starburst amacrine cell, filled with Lucifer Yelllow. (C) A section of amacrine cell inputs, through conventional synapses and via gap
retina stained for choline acetyltransferase (red) showing conventional
and displaced somas and two bands in the inner plexiform layer. INL, junctions, which are thought to tune ganglion cell properties in
inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer. a complex way that is not well understood. Ganglion cells obey
(Courtesy of SC Massey.) the stratification rules governing the inner retina. Thus OFF gan-
glion cells receive input from OFF bipolar cells in sublamina a
and ON ganglion cells receive input from ON bipolar cells in
expression of chloride transporters may contribute to the direc- sublamina b. There are a few ON/OFF types of ganglion cell,
tional asymmetry.239 In addition, there is GABA-mediated such as the famous DS type, and they are bistratified.
starburst-to-starburst inhibition, which may enhance or fine- Ganglion cell axons run across the vitreal surface of the retina
tune the DS responses.233 These important results confirm a long- and unite to form the optic nerve. Unlike many other retinal
held suspicion that there is a great deal of local processing neurons, ganglion cells produce conventional action potentials
among amacrine cell dendrites and it suggests that starburst suitable for transmission over the relatively long distance to the
amacrine cells are the source of directional signals in the retina. brain. The vast majority of ganglion cells use the excitatory neu-
Given that starburst amacrine cells generate directional signals, rotransmitter glutamate to communicate with higher visual
they release GABA, and they synapse directly with ON/OFF DS centers. Everything you see about the visual world comes to you
ganglion cells, then the final requirement for specific wiring is through the responses of ganglion cells. If they don’t signal it,
that individual starburst dendrites with the appropriate direc- you don’t see it! If there are about 20 ganglion cell types (see
tional asymmetry are connected to ON/OFF DS ganglion cells below) and the coverage factors range from one to three, then
with the same orientation of the null/preferred axis. (Remem- the total coverage of the retina by the whole ganglion cell popu-
ber, there are four subsets of ON/OFF DS ganglion cells with lation is about 50. In other words, each point on the retina is
different axes, roughly aligned with the four main compass covered by about 50 ganglion cells. The number of ganglion cells
points.) By a combination of calcium imaging to identify the in each human retina is approximately 1.5 million, equal to the
directional preferences combined with serial block-face electron number of axons in each optic nerve. In macaque, there are
microscopic imaging to reconstruct the underlying retinal cir- 1.8 million,108 in rabbit, 380 000,249 and in mouse retina approxi-
cuits, the specific synaptic connections have now been identi- mately 60 000 ganglion cells.49 The actual number of cells has
fied.240 Starburst dendrites with a specific orientation make a large genetic component, which produces up to threefold
variation in numbers, as demonstrated by quantitative analyses imaging has produced a surprising consensus on the number of
of a large number of mouse strains.250 ganglion cell types. The Masland lab used four different staining
Ganglion cells are physiologically and morphologically strategies (intracellular dye injection, photofilling, and gene gun- 387
diverse. Best estimates are in the range of 15–20 different gan- mediated entry of DiI particles or DNA-coated particle coding
glion cell types. We think the number of distinct types is for GFP expression) to label over 700 ganglion cells in the rabbit

Chapter 15
extremely important because each ganglion cell type is thought retina.256 Based on dendritic field size and structure and, most
to represent an independent visual channel. They can be classi- importantly, the depth of stratification in the IPL, this set of
fied by many physiological and anatomical criteria, including ganglion cells was divided into 13 different types (Fig. 15.29). A
size, response, receptive field, color, bandwidth, ON or OFF, few cells were unclassified, which could allow additional gan-
conduction velocity, morphology, branch pattern, stratification, glion cell types of low density. A calculation based on the
coupling, coverage, and central projections. The rabbit retina has expected coverage and density for each cell type showed that
long been favored for physiological recording and the classic this number of classes would fit within the total number of rabbit

Function and Anatomy of the Mammalian Retina


studies of Levick251 and, later, Caldwell and Daw252 identified ganglion cells. In the macaque retina, 13 different ganglion cell
about 12 different physiological types. An array of microelec- types were labeled by back-filling from the lateral geniculate
trodes makes it possible to record from a large number of gan- nucleus with rhodamine-linked dextrans followed by photo-
glion cells simultaneously. Using this technique, DeVries and staining.35 In summary, large-scale surveys have now been
Baylor253 identified 11 different physiological types in the rabbit conducted for human,257–259 macaque,260 rabbit,256 cat, rat, and
retina, many of which were arranged in independent mosaics mouse,261,262 and some morphological varieties of ganglion cell
with a characteristic spacing, such that a gaussian fit of the recep- are common to all these species. An important goal now is to
tive fields touched at the border. This corresponds to a coverage characterize fully each RGC type with respect to physiology and
of 1.8 and suggests a general principle for the optimal spacing synaptic connections. An understanding of the central projec-
of ganglion cells. OFF α cells, in particular, showed synchro- tions of each ganglion cell type will be necessary before we have
nized firing patterns consistent with the presence of gap junction a true understanding of the visual process.
coupling in this cell type.193,254,255 New tools by way of transgenic mouse lines that express a
The classification of ganglion cell types, especially when based marker protein such as GFP in a single type of cell are becoming
on morphological details, has been difficult and contentious. available.40,263–268 A particularly striking example of the power of
However, a quiet revolution in staining techniques and digital this approach was the ability of Michal Rivlin-Etzion and

INL
0%
20%
40%
60%
80%
100%
G1 G2 G3 G4 G4 G5 GCL

INL
0%
20%
40%
60%
80%
100%
G6 G7 G8 G9 GCL

INL
0%
20%
40%
60%
80%
100%
G10 G11 G11 GCL

Fig. 15.29 A summary of the main types of ganglion cell from the rabbit retina. G11 are the alpha ganglion cells, G7 is the ON/OFF directionally
selective (DS) cell, and G10 is the ON DS cell. INL, inner nuclear layer; GCL, ganglion cell layer. (Reproduced with permission from Rockhill RL,
Daly FJ, MacNeil MA, et al. The diversity of ganglion cells in a mammalian retina. J Neurosci 2002;22:3831–43, with permission from the authors
and the Society for Neuroscience.)
colleagues to show that two ON/OFF DS RGCs with similar Importantly, cluster analysis in multivariate space showed that
direction selectivity had different projection patterns to the the different ganglion cell types are statistically separate.
388 brain. These data suggest there are at least eight ON/OFF DS
RGCs with indistinguishable dendritic arbors. If a similar situa- Does each ganglion cell type represent
tion arises with other classes of RGCs then the number of inde- a visual channel?
pendent cell types may increase dramatically, also increasing the The simple answer to this question is yes, we think so. However,
Section 1

number of visual channels leaving the retina. In fact, the total we still do not have a definitive value for the number of channels
number of ganglion cell types has not yet been determined for and, for those ganglion cell types that have been classified mor-
any species. Seemingly rare cell types which account for a small phologically, it is not always clear which physiological class they
fraction of the total, e.g., ipRGCs, make up less than 1%, but may belong to. The correspondence between functional anatomy and
be split into five subtypes with different morphology and differ- physiological type is only partially complete.273 A problem that
ent central projections, further adding to the types of RGCs. has yet to be adequately resolved is what features of the func-
Anatomy and Physiology
Basic Science and Translation to Therapy

There seems to be general agreement that the number of gan- tional responses of the ganglion cells are the ones that set indi-
glion cell types will be more than 20.269 vidual classes apart. In fact, function alone is probably not
The importance of depth in the IPL was emphasized when sufficient and projection pattern, axon diameter, and terminal
Roska and Werblin showed that at least 10 ganglion cell types, morphology may also be important. Regardless, it is widely
most narrowly stratified at different depths, received distinct believed that the retina, with the ganglion cells as the output,
physiological inputs in the rabbit retina.270,271 Apparently, the IPL produces a large set of neural codes, most of which have not
contains a stack of parallel versions of the visual scene coded by been deciphered, that are transmitted to the brain in parallel
depth. Each layer receives a different combination of excitatory both on a temporal scale and to a variety of central structures.
and inhibitory inputs to produce at least 10 spatiotemporal chan- These are the visual channels, some of which carry different
nels. Ganglion cells with transient or sustained responses were versions of Mona Lisa (Fig. 15.1), where we began. Below are
located at different levels of the IPL. Some ganglion cells received some examples of different ganglion cells/visual channels that
inhibitory inputs that originated in another layer. This type of we are beginning to understand.
vertical inhibition could be carried by narrow-field amacrine In a few cases, a particular ganglion cell type seems to have a
cells, many of which are broadly stratified.38 The functional specific role. Midget ganglion cells in the primate retina are a
anatomy of the IPL is very precise. In the rabbit retina, OFF and good example. There are two types, ON and OFF, which are
ON alpha ganglion cells are stratified just below the two cholin- broadly stratified towards the middle of the IPL. Midget gan-
ergic bands (Fig. 15.30).10,272 They are separated by as little as glion cells are numerically dominant in central retina, where
1–3 µm but they are clearly different addresses serving different they may account for as many as 95% of all ganglion cells.150
ganglion cells with distinct morphological and physiological Importantly, we know that in central retina there is a 1 : 1 : 1 cor-
properties. If a single stratum can be as narrow as this, there is respondence from cones → midget bipolar cells → midget gan-
certainly room for a dozen or more different layers within the glion cells. Beyond 2 mm or 10°, the midget ganglion cells form
IPL, each serving one or more different ganglion cell types. separate dendritic clusters that receive input from multiple
A parallel attempt to classify rabbit ganglion cells relied on an midget bipolar cells. Sampling theory, based on the density of
entirely different strategy. The molecular signature for six dif- midget ganglion cells, closely predicts the curve describing a
ferent amino acids and a glutamate excitation signal (1-amino- psychophysical measure of human acuity, except close to the
4-guanidobutane: AGB) allowed a set of 465 cells in the GCL to fovea where the cone pedicles are laterally displaced.274 Thus, it
be sorted into 14 different ganglion cell types and three small seems clear that midget ganglion cells transmit a high-acuity
amacrine cell types.193 All ganglion cells contained glutamate, version of the visual scene from the center of your gaze, the
but the levels of GABA and glycine, which are thought to reflect central retina. The 1 : 1 : 1 ratio also means that individual midget
gap junction coupling to amacrine cells, were more variable. ganglion cells will be color-coded according to the type of cone

B
A
ON/OFF DS
OFF

Fig. 15.30 Stratification of the inner plexiform layer. (A) The dendrites
of an OFF alpha ganglion cell (green) lie above the cholinergic a band
(red). (B) The dendrites of an ON/OFF directionally selective ganglion
cell are bistratified and lie within the two cholinergic bands (green +
red + yellow). (C) The dendrites of an ON alpha ganglion cell lie
immediately below the lower cholinergic b band. This is at the same
depth as the terminals of calbindin bipolar cells (blue), so green + blue
= cyan. Thus, the inner plexiform layer can be very finely divided into
C different layers which are actually different addresses for synaptic
ON α interactions. (Courtesy of W Li and SC Massey.)
at the beginning of the chain. Midget ganglion cells project to exactly to alpha cells is an open question. There appear to be
the parvocellular layers of the lateral geniculate nucleus which wide-field ganglion cells in the primate retina that are mor­
also carry color signals. Midget ganglion cells are a specializa- phologically closer to alpha ganglion cells in other species.260 389
tion of the primate retina, perhaps the final point in the relentless The ON/OFF DS ganglion cells of the rabbit retina, reported
survival-driven goal of maximum acuity. There is no exact by Barlow and Levick nearly 50 years ago,276are one of the best

Chapter 15
match in other mammalian species but an equivalent role could characterized of the retinal ganglion cells. In these ganglion cells,
be played by beta ganglion cells in the cat retina. In the rabbit a bar moving in the preferred direction through their receptive
retina, there are neither midget nor beta ganglion cells but an field produces a strong burst of spikes, but the same stimulus
ON and OFF pair of relatively small ganglion cells, G4, which moving in the opposite direction produces little or no response.
are rather broadly stratified toward the middle of the IPL.256 As first shown by Amthor and colleagues,277 bistratified cells
These are at least morphologically similar. Consistent with the fasciculate extensively within the two bands of starburst ama-
idea that the midget/beta cells are a specialization of animals crine cell processes and they have a distinct retroflexive branch-

Function and Anatomy of the Mammalian Retina


with a fovea or foveal-like specialization, the rodent retina lacks ing pattern (Fig. 15.32). DS ganglion cells are exquisitely sensitive
these types of ganglion cells. to nicotinic cholinergic agonists but, surprisingly, directional
Some of the most convincing evidence that ganglion cells carry selectivity is not dependent on this cholinergic input.235
specific channels of visual information comes from comparative Paired recordings showed that starburst amacrine cells on the
anatomy, where it can be seen that certain ganglion cell types null side of the DS ganglion cells provide asymmetric GABA
recur frequently across species. Perhaps the best example is the inhibition to DS ganglion cells.236,278 The source of the GABAergic
alpha ganglion cell, which has been reported in many mamma- input is from starburst amacrine cells, which contain GABA as
lian retinas.275 Alpha ganglion cells have large somas, come in well as ACh.232 Individual dendrites of the starburst amacrine
paramorphic ON and OFF pairs, make up less than 5% of all cells generate directional responses due to their intrinsic proper-
ganglion cells, and have a large dendritic field with a character- ties and this is the origin of directional signals in several other
istic radiate branching pattern (Fig. 15.31). In the rabbit retina, types of DS ganglion cells.33 This difference is consistent with
alpha ganglion cells are narrowly stratified just outside the cho- recent morphological results using an extensive three-
linergic bands. Physiologically, they have transient nonlinear dimensional reconstruction of functionally identified DS gan-
(Y-like) properties, they respond to stimuli of high temporal glion cells by serial block-face electron microscopy that showed
frequency, and project to the magnocellular layers of the lateral specific inhibitory inputs from starburst dendrites with the
geniculate nucleus. Their responses are not color-coded. Alpha appropriate orientation.240
ganglion cells seem well suited for detection of low luminance At present, four ganglion cell types with different null/
contrast over large areas of the visual field, such as a blurred preferred axes have been described, one of which is dye-coupled
copy of the Mona Lisa. Their occurrence over such a wide range and shows a precise dendrite-to-dendrite tiling pattern with a
of species suggests they play a fundamental role common to the coverage close to 1.279 DS cells with the other preferred axes
visual needs of many animals, e.g., detection of motion. Parasol are thought to tile the retina independently so that if the den-
ganglion cells in the primate retina and A-type cells of rodents drites of two DS ganglion cells overlap, they must have different
have similar properties, although whether they correspond preferred directions. They can also be differentiated by molecu-
lar markers and have slightly different central projections.280
The four null/preferred axes are aligned with the extraocular
muscles281 so it is often suggested that the ON/OFF DS cells
play a role in the control of eye movements. Cells with a similar
morphology have been noted in mouse,261,262 rat, cat (the iota
cell),282 and even primate retina.260 In the mouse two posteriorly
tuned, essentially morphologically identical DS cells have been
defined that have subtle receptive field differences, and differ-
ent central projection patterns.268 If each compass point is dupli-
cated in the same manner, this would predict there should be
a total of eight ON/OFF DS ganglion cells. It is not unreason-
able to assume a similar situation will exist in most other
mammals.
There is another DS cell in the rabbit retina that has interesting
properties. Its dendrites are monostratified in the ON starburst
band and have a similar retroflexive branch pattern. They also
receive specific null-side inhibition from starburst amacrine
200 µm
cells.283 However, these ON DS cells respond to much slower
movements and its three variants have different null/preferred
Fig. 15.31 Dye-injected ganglion cells in the rabbit retina. The white axes, which are aligned with the semicircular canals.284 Consis-
cell is an ON/OFF directionally selective ganglion cell surrounded by
four much larger transient ON DS ganglion cells. These two ganglion tent with the hypothesis that DS ganglion cells are critical to eye
cell types have obviously different dendritic morphology. In addition, movement control, the ON DS ganglion cells project to the
the transient ON DS ganglion cells form a nonrandom mosaic. medial terminal nucleus of the accessory optic system.284,285 Gan-
There are also some dye-coupled amacrine cells in the background. glion cells with a strikingly similar morphology have been
(Reproduced with permission from Hoshi H, Tian LM, Massey SC,
et al. Two distinct types of ON directionally selective ganglion cells in reported in mouse, rat, and primate retina.260 The value of this
the rabbit retina. J Comp Neurol 2011;519:2509–21.) comparative anatomy becomes clear when we consider an
390
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

A B
OFF ON

Fig. 15.32 ON/OFF directionally selective ganglion cell. A single


ON/OFF directionally selective ganglion cell was dye-injected with
Neurobiotin and then, using the confocal microscope, the two dendritic
fields were imaged and color-coded. Note the retroflexive branching
pattern which fills most of the available space, in contrast to the
branch pattern of the alpha ganglion cell above. (Reproduced with
ON/OFF DIRECTIONALLY permission from Kittila CA, Massey SC. Pharmacology of directionally
C selective ganglion cells in the rabbit retina. J Neurophysiol 1997;77:
SELECTIVE GANGLION CELL
675–89.)

experiment in the mouse retina. When starburst amacrine cells amacrine cells are the source of directional signals in the retina,
are ablated, DS responses in ganglion cells are abolished and the it is not clear how DS signals are generated in this cell type.
optokinetic nystagmus is also blocked.234 This is a type of reflex Finally, in a stunning example of a cell type-specific molecular
eye movement involved with tracking moving objects with the marker, a whole population of ganglion cells from the mouse
same velocity range as the ON DS ganglion cell. It suggests that retina was stained using a junctional adhesion molecule B (JAM-
ON DS cells provide the input to a system for tracking or stabi- B)-driven GFP mouse line.265 This procedure identified a uniform
lizing the retinal image. At least one gene associated with an eye population of ganglion cells with asymmetrical dendritic trees
movement disorder is exclusively expressed in starburst ama- aligned dorsal to ventral. A similar OFF cell type, G3 in the
crine cells, which provide the directional input to the ON DS Masland catalog, has been reported for the rabbit retina.256,289 In
ganglion cell.267 the mouse, this cell type has been characterized as OFF DS,
A second type of ON DS ganglion cell has recently been identi- although in the rabbit retina it was identified as orientation-
fied in the rabbit retina.285–287 This cell type, originally reported selective.290 If it is truly DS, then the mechanism may be unusual
by Ackert and colleagues,288 is distinct from the cell described because it does not stratify with the starburst amacrine cells.
above in terms of morphology and dendritic stratification, just Together, these observations suggest that there may be multiple
above the lower band of ChAC processes. If the starburst circuits to generate directional signals.291
A ganglion cell for the control of pupil diameter Based on a combination of morphology and physiology, five
and circadian rhythm subtypes of melanopsin-containing ganglion cells, M1–M5, have
It has long been known that certain visually driven activities been identified in the mouse retina.298,299 The M1 subtype is the 391
persist in mice where rods and cones have completely degener- only one stratified in sublamina a; it contains the most melanop-
ated. These apparently blind mice retain pupillary light reflexes sin and has strong intrinsic photoresponses and relatively weak

Chapter 15
as well as circadian rhythms and a similar profile has been rod/cone input. M2 cells have dendrites in sublamina b but less
reported in totally blind humans.292 It was concluded that these melanopsin and weaker intrinsic responses. M3 cells are bistrati-
reflexes are driven by connections separate from the usual fied and do not form a uniform population.300 M4 and M5 cells
image-forming pathways. This issue was resolved when a set of have very low levels of melanopsin and correspondingly weak
ganglion cells were found to express an additional visual intrinsic responses.298 Using combinations of markers, the
pigment called melanopsin. They form a relatively sparse mosaic M1-class ipRGCs can be further divided into two types, one
(only 2000 per mouse retina) with loopy dendrites (Fig. 15.33) in projecting to the shell of the OPN, which is required for the

Function and Anatomy of the Mammalian Retina


sublamina a and b and they respond with a slow sustained dis- pupillary light reflex, and one targeting the SCN, required for
charge dependent on the light intensity.36 Even when these cells circadian photo entrainment.301 This Brn-3b-negative group of
are isolated, they still produce light responses.293 In other words, M1 ipRGCs comprises approximately only 10% of all melan­
the melanopsin-containing ganglion cells are intrinsically pho- opsin ganglion cells. Thus, the specific nonimage-forming visual
tosensitive and they are referred to as ipRGCs. pathway that resets the circadian clock every morning consists
The melanopsin-containing ganglion cells project to the supra- of only 200 ganglion cells.301 It is very surprising that a distinct
chiasmatic nucleus (SCN), the brain’s circadian pacemaker.36,294 visual pathway can be formed by such a small number of cells.
Labeled axons were also found in the olivary pretectal nucleus The roles of non-M1 types are still unclear but they may con-
(OPN), which is involved with the circuit controlling pupil tribute to light avoidance in mouse pups, sleep regulation and,
diameter. These are referred to as nonimage-forming pathways. in humans, the light sensitivity of migraine and seasonal affec-
In mice lacking melanopsin, photoentrainment of the circadian tive disorder.298 The activity of ipRGCs is also important for reti-
rhythm and the control of the pupillary light reflex were attenu- nogeniculate segregation, which is now known to involve light
ated at high light intensities.295 sensitivity prior to function of the photoreceptors.247
However, melanopsin ganglion cells also receive the usual rod
and cone inputs, which can drive these circuits at low light Color vision and ganglion cells
intensities. The M1 subtype is stratified in sublamina a but it still Humans are trichromatic, with red, green, and blue cones. Color
has ON-driven light responses due to the presence of ectopic or vision provides an additional variable to improve visual dis-
axonal ribbon synapses from ON cone bipolar cells as they tra- crimination. It is organized into two opponent systems, red-
verse sublamina a (Fig. 15.33).27,28,296 In crossbred mice with pho- green and blue-yellow (where yellow = red + green).302 Hence,
toreceptor degeneration as well as no melanopsin, the pupillary there are no red/green hues and no mixtures of blue and yellow;
light reflex and the photoentrainment of the circadian clock were they are said to be color-opponent. The blue-yellow system is
completely eliminated.297 The conclusion is that, at low irradi- found widely in mammals and is thought to be older on an
ance, the normal rod/cone inputs drive these circuits but at evolutionary timescale.56 In primates, once the midget system
higher light levels the intrinsic melanopsin system drives these had reached maximum resolution in the 1 : 1 : 1 pathway, a com-
accessory visual functions. These experiments provide anatomi- paratively recent gene duplication produced red and green pig-
cal, physiological, and behavioral evidence that the ganglion cell ments. By reason of the connection with a single cone, midget
type defined by the expression of melanopsin forms a specific ganglion cells are automatically color-coded.
channel in the mammalian visual system that is connected to the Midget ganglion cells are concentrically organized as red-
SCN and the OPN among other targets. This is a prime example green or green-red. The center input is spectrally pure because
that different ganglion cell types provide parallel channels in the of the 1 : 1 : 1 connection with single cones. There has been some
visual system that serves specific functions. controversy concerning the origin of the color-opponent

Fig. 15.33 Melanopsin ganglion cell ectopic


A B inputs. (A) Low-resolution picture shows
melanopsin ganglion cells in the rabbit retina
have sparse randomly arranged dendrites.
(B) Staining certain ON cone bipolar cells
with an antibody against calbindin showed
that the descending axons were frequently
adjacent to the melanopsin-positive ganglion
cells. These are the sites of axonal ribbon
inputs which provide ON input to melanopsin
ganglion cells that stratify in sublamina a of
the inner plexiform layer. (Reproduced with
permission from Hoshi H, Liu WL, Massey
SC, et al. ON inputs to the OFF layer: bipolar
cells that break the stratification rules of the
retina. J Neurosci 2009;29:8875–83.)

50 µm 20 µm
surround, whether it is spectrally pure, and whether it arises search of the blue ones.152,306,307 Blue ON cone bipolar cells have
from the outer or inner retina. Recent recordings from midget direct input to a small bistratified ganglion cell that gives blue
392 ganglion cells in the primate retina indicate that the surround is ON/yellow OFF responses.308 Blue-yellow small bistratified gan-
chromatically mixed. In other words, it is the average of the sur- glion cells have also been identified in multielectrode array
rounding cones. On average, the ratio of red to green in the recordings from primate retina. These experiments suggested
surround was approximately 50%.110 Furthermore, both center that most of the color opponency arose in the outer retina.309,310
Section 1

and surround inputs modulated an excitatory conductance with Further physiological evidence suggests that blue-yellow color
a reversal potential around 0 mV. This means that the surround opponency originates in the outer retina.311 Indeed, blue cones
signal does not arise from amacrine cell inhibition in the inner themselves are blue-yellow color-opponent, indicating that
retina. Rather, the color opponency must be generated in the the yellow-opponent signal is generated by horizontal cell
outer retina by horizontal cell feedback.110 Horizontal cells are feedback.312
suitably wired to provide the spectrally mixed surround signal In nonprimate mammalian retina, blue-driven ganglion cells
Anatomy and Physiology
Basic Science and Translation to Therapy

because they contact both red and green cones indiscrimi- were reported as monostratified ON cells.313 This is quite differ-
nately.303 Enhanced buffering to block pH-dependent horizontal ent from the small bistratified blue-yellow ganglion cell of the
cell feedback eliminated both the spatial and chromatic sur- primate retina. The color opponency of this monostratified gan-
round of midget ganglion cells. Although the exact mechanism glion cell could arise in the outer retina, as suggested for primate
of horizontal cell feedback is controversial, this strongly indi- retina.309,312
cates that color opponency is produced by horizontal cell feed-
back in the outer retina.110,304 Finally, neither GABA nor glycine GENE THERAPY TO CURE
antagonists blocked the surround inputs. Thus inhibition in COLOR BLINDNESS
either the outer or inner retina is not required. Nor does hori-
Mice are dichromats, expressing a short-wavelength photo­
zontal cell feedback in the primate retina depend on GABA or
pigment and a single medium-wavelength pigment on the X
glycine.110
chromosome. Jacobs and colleagues314 showed that adding a
Large-scale multielectrode array recording has also been used
human long-wavelength photopigment allowed mice to have a
to analyze the color inputs to primate ganglion cells.305 Hun-
form of color vision. This idea has been extended to primates
dreds of ganglion cells may be recorded simultaneously and
and could potentially be used in humans. About 5–8% of the
then sorted by spike size and shape to identify individual gan-
population has red-green color blindness, because they lack
glion cells. Types of ganglion cell may be identified by their
either the long- or middle-wavelength visual pigments. Some
receptive field sizes, center/surround organization, and the
squirrel monkeys also are dichromats and recently Mancuso and
mosaic properties by which they tile the retina. Using a fine-
colleagues315 showed that adding a third photopigment, using
grained textual stimulus, local hot spots were found within each
gene therapy approaches could restore color discrimination,
receptive field that matched the distribution of the underlying
even though the monkeys had been dichromats since birth.
cone array. The spectral characteristics of each hot spot, recorded
in several nearby ganglion cells, were used to identify each cone NEW TOOLS TO IDENTIFY GANGLION
locus as red, green, or blue and then the contributions of each
individual cone were mapped to the overlying array of ganglion
CELL TYPES
cells. Parasol ganglion cells sampled uniformly from red and The lack of genetic markers for individual ganglion cell types is
green cones but not blue cones. In contrast, many midget gan- a major handicap. As shown above, classification schemes that
glion cells showed color-opponent responses. As above, the sur- rely on morphology are very difficult to implement and there is
rounds were spectrally mixed but the center inputs to both ON a less-than-perfect mesh with physiology and biochemistry. This
and OFF midget ganglion cells had a small nonrandom tendency is not a minor issue: the comparatively recent flood of informa-
to sample from more red or green cones.305Obviously, in central tion about ipRGCs depends in large part on the identification of
retina, where midget bipolar cells may contact a single cone, this this cell type(s) due to the expression of melanopsin. Mouse lines
would provide a spectrally pure center with a mixed color- with GFP-labeled melanopsin ganglion cells, knockout mouse
opponent surround. Intellectually, we understand that tracing strains, and melanopsin antibodies have all been developed.
back from ganglion cells through the retinal circuitry must lead This one simple attribute allows the identification of melanopsin-
to cones. However, the direct demonstration of ganglion cell containing ganglion cells for recording, filling, manipulating,
sampling from the complete array of identified cones is an exper- or deleting. Thus, the connections, subtypes, physiology, light
imental triumph. These experiments indicate that parasol gan- responses, and central projections of the ipRGCs have been
glion cells are not color-coded while color signals are carried by uncovered, obtaining essentially a full description of this
the midget ganglion cells.305 nonimage-forming pathway and its functions in the pupillary
In contrast to the red-green system, the circuits underlying light reflex and circadian entrainment.
blue-yellow opponency have been relatively well described. However, new molecular techniques are starting to provide a
Some of this success is due to the morphological details which way to mark certain ganglion cell types as a complete popula-
make cells in the blue pathway more recognizable. This begins tion. Thus, it is becoming possible to study a specific ganglion
with blue cones themselves, which can be labeled with anti­ cell type as opposed to a spectrum of mixed cell types with vari-
bodies against blue cone opsin.59 Red and green cone opsins are able properties. One research group has screened a library of
so close they cannot be discriminated by current antibodies. In bacterial artificial chromosome (BAC) transgenic mice with GFP
addition, blue cone pedicles are noticeably smaller, with fewer expressed under the control of different promoters. In particular,
telodendria.72 The blue cone bipolar cells can also be recognized they looked for GFP expression in a nonrandom mosaic. This is
because of their long dendrites, which bypass many cones in one of the known properties of specific ganglion cell types,
which tile the retina in a uniform manner.41 In calretinin- The sum of these inputs to a particular type of ganglion cell
enhanced GFP (EGFP) mice, a mosaic of large ganglion cells, forms a single channel in the visual system. The overall complex-
stratified in sublamina a, with OFF transient responses to light ity appears daunting but it may be broken down into a repeated 393
was found. Collectively, these properties identify the GFP- set of stereotyped local circuits, which are strictly governed by
labeled cells as OFF α ganglion cells.263 Centrally, this specific a set of rules concerning position and appropriate synaptic part-

Chapter 15
ganglion cell type projects to both the superior colliculus, an area ners. Below we discuss briefly two obvious examples where the
integrating sensory input and guiding visual attention, and the basic layering and organization of the retina are disrupted, with
dorsal lateral geniculate nucleus, a relay station on the way to important clinical consequences.
the visual cortex. Furthermore, the axonal projections in both In glaucoma, the basic defect is in the GCL. Increased intraocu-
areas are precisely organized in columns at a specific laminar lar pressure apparently induces programmed cell death in retinal
depth. Importantly, disrupting cholinergic retinal waves during ganglion cells. Factors involved may include anoxia, reduced
development prevented the columnar organization of OFF α axonal transport, and excess glutamate, leading to excitatory

Function and Anatomy of the Mammalian Retina


ganglion cell axons.263 This suggests that spontaneous activity damage known as excitotoxicity. Ganglion cells appear to be
during retinal development controls the development of a reti- particularly sensitive while other retinal neurons such as ama-
notopic map. It is worth repeating that these experiments were crine cells are unaffected. Figure 15.34 shows a retinal section
made possible because a genetic marker for a single ganglion cell from the same eccentricity of a control macaque eye compared
type, the OFF α ganglion cell, was identified. to the operated eye with experimentally induced high intraocu-
In a cadherin-3 GFP mouse line, a small percentage of ganglion lar pressure. All retinal layers appear normal, with the exception
cells was labeled, some of which also expressed melanopsin. of the GCL, which is severely depleted compared to the control
They also contained cadherin-6 and projected to nonimage- eye. The absence of retinal ganglion cells may be correlated with
forming visual nuclei such as the ventral lateral geniculate dramatically reduced visual acuity, especially in the peripheral
nucleus, the intergeniculate leaflet, and the OPN. These nuclei, retina.318
known targets of ipRGCs. Importantly, in cadherin-6-deficient A large number of photoreceptor mutations, often in the
mice (cadherin-3/GFP crossed with a cadherin-6 knockout), the phototransduction cascade, lead to photoreceptor degeneration
cadherin-3-labeled ganglion cells failed to innervate the appro- in conditions such as retinitis pigmentosa (RP). The defect often
priate visual nuclei. Instead, the GFP-labeled axons projected arises in rods but eventually both rods and cones degenerate.
through and past their usual targets. These experiments, utiliz- Whether this occurs because rods secrete a maintenance factor
ing genetically labeled mouse lines, indicate that cadherin-6 is for cones, or via rod/cone coupling, known as the bystander
necessary for certain ganglion cell types to recognize their syn- effect, or due to the accumulation of cellular debris is a matter
aptic targets and form functional circuits.316 of debate beyond the scope of this chapter. However, photo­
As mouse lines in which different cell types are labeled by receptor degeneration has severe consequences for the organiza-
molecular methods become widely available, the methods to tion of the remaining retina. The neural retina apparently
differentiate among retinal cell types will continue to expand. requires input from the sensory retina to maintain its well-
One group has published a “genetic address book” of retinal organized structure. When the neural retina has been deaffer-
cell types.40 Moreover, several groups have used GFP-labeled ented, the second-order neurons, such as rod bipolar cells,
cell types to develop single-cell expression libraries for specific produce new dendrites and sprout profusely, making many
cell types.280,317 This advance in technique is likely to be a inappropriate contacts.3 Many neurons develop novel processes
game-changer. It will enable a molecular fingerprint to be that invade other layers where they proliferate to produce
developed for each cell type, as well as identifying the tran- tangled microneuromas. Glial cells are extensively remodeled;
scription factors and signaling molecules which govern devel- there is mass neuronal migration and almost total disruption of
opment and circuit connections.317 In particular, a transcription the layered structure of the retina. Figure 15.35 shows examples
factor-based code was used to sort the cells into relevant groups. from two rodent lines and a human RP case. In each case, the
Perhaps this methodology will provide the definitive answer resulting tangle of misplaced cells and inappropriate contacts is
to how many ganglion cell types are present in the mammalian essentially unrecognizable as a section of retina. The basic
retina. Finally, by correlating these single-cell libraries against layered organization of the retina has been destroyed. One con-
a database of gene mutations associated with visual system sequence of this neural reorganization is that the remaining
disorders, it may be possible to identify defects arising in retina is unlikely to retain sufficient organization to operate in
specific cell types or circuits. For example, an eye movement any kind of coherent manner. Thus, in the absence of photo­
disorder was correlated with starburst amacrine cells, which receptors, strategies aimed at stimulating the remaining neural
may be the source of DS signals required to control reflex retina by means of a retinal implant seem unlikely to succeed,
eye movements.317 unless the process of retinal remodeling can be controlled.
Of course, one major advance in the last few years is our ability
CLINICAL RELEVANCE OF to identify mutations that cause RP by whole-genome sequenc-
ing. In the near future we will no doubt be able to sequence each
FUNCTIONAL ANATOMY person’s entire genome, and this could be done at birth. Identify-
As we have seen, the retina is fundamentally organized as a ing individuals who will get RP, well before the retina under-
layered structure. This applies not only to the major cellular and goes extensive degeneration, might allow curative strategies to
synaptic layers but particularly within the IPL where the level be implemented at times that avoid some of the problems associ-
of stratification is an address. It appears that stratification at the ated with remodeling.
appropriate depth in the IPL allows a ganglion cell to receive In addition to the above diseases that disrupt the gross mor-
input from a certain set of bipolar and amacrine cell processes. phology of the retina, there also are diseases with less severe
394
ONL
Section 1

OPL

INL
Anatomy and Physiology
Basic Science and Translation to Therapy

IPL

GCL

A Normal B Glaucoma

Fig. 15.34 Depletion of ganglion cells in glaucoma. (A) Control retina, near central, as shown by the depth of ganglion cell somas. (B) While the
rest of the retina is normal, the ganglion cell layer has been severely depleted by experimental glaucoma. (Courtesy of Louvenia
Carter-Dawson.)

effects. One group results in night blindness, without any degen- obvious of which are a lack of a fovea and red-green color
eration of retinal cell types. These are called congenital station- vision.
ary night blindness (CSNB). The result is a greatly diminished Similar data, though less complete, are available for other
amplitude of the electroretinogram b-wave, indicating lack of or mammals, particularly rodent and primate. In fact, experimental
poor signal transmission between photoreceptors and bipolar reasons often govern the choice of species for investigation and,
cells. Careful analyses of the electroretinogram indicate that in this regard, fish and salamander still have much to contribute
there are two forms, incomplete and complete.319 The genetic to the understanding of visual processing.
defects that give rise to these defects are now known to be Rods are specialized for high sensitivity at night, while cones
caused by mutations in genes that are involved in release of provide high acuity and color vision in daylight. Adaptation
glutamate from photoreceptors (incomplete, iCSNB) or absence in the phototransduction cascade plays a crucial role in adjust-
of signaling in depolarizing bipolar cells (complete, cCSNB). ing sensitivity. Now, we know there are dedicated pathways
Mouse models for iCSNB reveal that the dendritic arbors of ON for rod and cone vision throughout the retina. Horizontal cells
bipolar and horizontal cells are abnormal. They extend across provide feedback to photoreceptors and probably subtract a
the OPL into the ONL and often reach the outer limiting mem- large version of the average background. Visual pathways
brane.320 By contrast, models of cCSNB have no morphological diverge dramatically into multiple bipolar cell types with
abnormalities, although in some there appears to be some abnor- outputs at different levels or addresses in the IPL. One role
mal expression of synaptic proteins, resulting in ribbon synapses of amacrine cells is to provide another level of negative feed-
being present in dendrites of depolarizing bipolar cells.321 back at bipolar cell terminals but amacrine cells are extremely
diverse and probably serve many other functions, including
feature extraction, lateral inhibition, and various types of adap-
CONCLUSIONS tation to the image parameters. Ganglion cells are the output
In one sense, our understanding of the mammalian retina is neurons of the retina. In central primate retina, the midget
very advanced in that most of the cell types have been described system dominates with one cone to one midget bipolar cell
and numerically accounted for, more so than in any other part to one midget ganglion cell. This system may also carry color
of the CNS. While the rabbit was the preferred model for a long signals. There are at least 15–20 different types of ganglion
time, our ability to manipulate the mouse genetically means it cell, each of which is thought to carry a different channel of
is increasingly becoming the main model organism. Of course visual information. Some of these parallel channels represent
it has major drawbacks with respect to human retina, the most spatial vision at several bandwidths but others are involved
RPE Theme Class Key Fig. 15.35 Disruption of retinal organization
following photoreceptor degeneration.
A OSL GABA+ACs (A) Control retina: a theme map shows the
Gly+ACs well-organized, layered structure of the 395
Adult ISL GABA/Gly+ACs normal retina. (B, C) Rodent retina following
SD Rat ON cone BCs photoreceptor degeneration shows severe
ONL disruption of the normal layered structure.

Chapter 15
rod/OFF cone BCs
Many cells have migrated to inappropriate
OPL HCs locations. (D) A human retinitis pigmentosa
Anomalies GCs case where the organization of the neural
INL MCs retina has been severely disrupted. SD,
C Columns Sprague–Dawley; OSL, outer segment layer;
VCs
S Strictures ISL, inner segment layer; ONL, outer nuclear
RPE layer; OPL, outer plexiform layer; INL, inner
M MC seals
IPL nuclear layer; IPL, inner plexiform layer;
E Everted INL cells

Function and Anatomy of the Mammalian Retina


GCL, ganglion cell layer; GABA, gamma-
I Inverted INL cells
aminobutyric acid; Gly, glycine; ACs,
GCL amacrine cells; BCs, bipolar cells; HCs,
horizontal cells; GCs, ganglion cells; MCs,
Müller cells; VCs, vascular cells ; RPE,
M retinal pigment epithelium. (Reproduced with
B permission from Jones BW, Watt CB,
INL Frederick JM, et al. Retinal remodeling
triggered by photoreceptor degenerations.
S E J Comp Neurol 2003;464:1–16.)
Adult
RCS Rat C IPL
I

GCL

C M
INL
Adult C IPL
P23H Rat GCL
E

D
Human RP
M INL
E

IPL
S
I GCL

with eye movements, image tracking, pupil control, and reset- electrophysiology will provide a major contribution to many
ting the circadian clock. of these problems and rapid progress may be anticipated in
While an outline and the gross cell count may have been the next few years. The requirement of functional anatomy
completed, much work remains to describe specific circuits is to describe the exact neuronal connections that underlie
and cellular connections, which we refer to as the functional specific pathways and circuits in the retina. These are the
anatomy of the retina. Several obvious problems challenge building blocks of the visual system.
our current understanding: the role of horizontal cells and For online acknowledgments visit http://www.
the mechanism of horizontal cell feedback, the reason for the expertconsult.com
presence of two horizontal cell types, the circuitry underlying
color vision, the connections of the diffuse cone bipolar cells, REFERENCES
a full description of the mGluR6 cascade, the switch from 1. Masland RH. The functional architecture of the retina. Sci Am 1986;255:
102–11.
rod to cone pathways, the mechanism of directional selectivity, 2. Rodieck R. The first steps in seeing. Sunderland, MA: Sinauer Associates;
the reasons for the extensive electrical coupling of neurons 1998.
3. Marc RE, Jones BW, Watt CB, et al. Neural remodeling in retinal degeneration.
in the retina, the circuits that control sensitivity and adapta- Prog Retin Eye Res 2003;22:607–55.
tion, the specific visual function of the many ganglion cell 4. Dowling JE. The retina: an approachable part of the brain. Cambridge, MA:
Belknap Press of Harvard University Press; 1987.
types – in other words, the role of each channel. We could 5. Vaney DI. Patterns of neuronal coupling in the retina. Prog Retin Eye Res
go on. Our ability to combine new imaging modes with 1994;13:301–55.
ACKNOWLEDGMENTS Thanks to past and present members of our laboratories who
contributed figures: Chris Kittila, Brady Trexler, Steve Mills, Wei
This work was supported by National Eye Institute grants EY Li, Jennifer O’Brien, Feng Pan, and John Koomen. Special thanks 395.e1
12354 (RGG), EY 014701 (MMC), EY 06515 (SCM), National Eye to Jennifer O’Brien, who prepared many of the figures, and
Institute core grant EY 10608 (SCM), and unrestricted grants David Marshak, Steve Mills, and John O’Brien for many discus-
from Research to Prevent Blindness to the Departments of Oph-

Chapter 15
sions. And finally, thanks to all those investigators who contrib-
thalmology and Visual Science (University of Texas and Univer- uted figures. The authors regret that due to space limitations the
sity of Louisville). SCM is the Elizabeth Morford Professor of outstanding work of many investigators could not be included
Ophthalmology and the grateful recipient of fellowships from or referenced in this brief review.
the Hanse-Wissenschaftskolleg, Delmenhorst, Germany and
Research to Prevent Blindness.

Function and Anatomy of the Mammalian Retina


6. Vaney DI, Weiler R. Gap junctions in the eye: evidence for heteromeric, het- 43. Strettoi E, Raviola E, Dacheux RF. Synaptic connections of the narrow-field,
erotypic and mixed-homotypic interactions. Brain Res Brain Res Rev 2000;32: bistratified rod amacrine cell (AII) in the rabbit retina. J Comp Neurol 1992;
115–20. 325:152–68.
396 7. Galarreta M, Hestrin S. Electrical synapses between GABA-releasing interneu- 44. Masland RH. Neuronal cell types. Curr Biol 2004;14:R497–500.
rons. Nat Rev Neurosci 2001;2:425–33. 45. Ahnelt PK, Kolb H. The mammalian photoreceptor mosaic-adaptive design.
8. Harris AL. Emerging issues of connexin channels: biophysics fills the gap. Prog Retin Eye Res 2000;19:711–77.
Q Rev Biophys 2001;34:325–472. 46. Sterling P, Demb JB. Retina. In: Shepard GM, editor. The synaptic organiza-
9. Goodenough DA, Paul DL. Beyond the gap: functions of unpaired connexon tion of the brain. New York: Oxford University Press; 2004. p. 217–93.
Section 1

channels. Nat Rev Mol Cell Biol 2003;4:285–94. 47. Curcio CA, Sloan Jr KR, Packer O, et al. Distribution of cones in human and
10. Zhang J, Yang Z, Wu SM. Development of cholinergic amacrine cells is visual monkey retina: individual variability and radial asymmetry. Science
activity-dependent in the postnatal mouse retina. J Comp Neurol 1987;236:579–82.
2005;484:331–43. 48. Whitney IE, Raven MA, Lu L, et al. A QTL on chromosome 10 modulates cone
11. Wassle H. Parallel processing in the mammalian retina. Nat Rev Neurosci photoreceptor number in the mouse retina. Invest Ophthalmol Vis Sci
2004;5:747–57. 2011;52:3228–36.
12. Hartveit E. Functional organization of cone bipolar cells in the rat retina. 49. Jeon CJ, Strettoi E, Masland RH. The major cell populations of the mouse
J Neurophysiol 1997;77:1716–30. retina. J Neurosci 1998;18:8936–46.
Anatomy and Physiology

13. Dacheux RF, Raviola E. The rod pathway in the rabbit retina: a depolarizing 50. Williams DR. Visibility of interference fringes near the resolution limit. J Opt
Basic Science and Translation to Therapy

bipolar and amacrine cell. J Neurosci 1986;6:331–45. Soc Am A 1985;2:1087–93.


14. DeVries SH, Schwartz EA. Kainate receptors mediate synaptic transmission 51. Williams DR. Aliasing in human foveal vision. Vis Res 1985;25:195–205.
between cones and ‘Off’ bipolar cells in a mammalian retina. Nature 52. Hirsch J, Curcio CA. The spatial resolution capacity of human foveal retina.
1999;397:157–60. Vis Res 1989;29:1095–101.
15. Slaughter MM, Miller RF. 2-amino-4-phosphonobutyric acid: a new pharma- 53. Baylor DA, Nunn BJ, Schnapf JL. Spectral sensitivity of cones of the monkey
cological tool for retina research. Science 1981;211:182–5. Macaca fascicularis. J Physiol 1987;390:145–60.
16. Nakanishi S, Masu M, Bessho Y, et al. Molecular diversity of glutamate recep- 54. Schnapf JL, Kraft TW, Baylor DA. Spectral sensitivity of human cone photo-
tors and their physiological functions. EXS 1994;71:71–80. receptors. Nature 1987;325:439–41.
17. Vardi N, Duvoisin R, Wu G, et al. Localization of mGluR6 to dendrites of ON 55. Schnapf JL, Kraft TW, Nunn BJ, et al. Spectral sensitivity of primate photore-
bipolar cells in primate retina. J Comp Neurol 2000;423:402–12. ceptors. Vis Neurosci 1988;1:255–61.
18. Nelson R, Famiglietti Jr EV, Kolb H. Intracellular staining reveals different 56. Nathans J. The evolution and physiology of human color vision: insights from
levels of stratification for on- and off-center ganglion cells in cat retina. molecular genetic studies of visual pigments. Neuron 1999;24:299–312.
J Neurophysiol 1978;41:472–83. 57. Roorda A, Williams DR. The arrangement of the three cone classes in the
19. Schiller PH, Sandell JH, Maunsell JH. Functions of the ON and OFF channels living human eye. Nature 1999;397:520–2.
of the visual system. Nature 1986;322:824–5. 58. DeMonasterio FM, Schein SJ, McCrane EP. Staining of blue-sensitive cones of
20. Tauchi M, Masland RH. The shape and arrangement of the cholinergic the macaque retina by a fluorescent dye. Science 1981;213:1278–81.
neurons in the rabbit retina. Proc R Soc Lond B Biol Sci 1984;223:101–19. 59. Szel A, Diamantstein T, Rohlich P. Identification of the blue-sensitive cones
21. Peichl L, Buhl EH, Boycott BB. Alpha ganglion cells in the rabbit retina. in the mammalian retina by anti-visual pigment antibody. J Comp Neurol
J Comp Neurol 1987;263:25–41. 1988;273:593–602.
22. Peichl L, Ott H, Boycott BB. Alpha ganglion cells in mammalian retinae. 60. Lennie P, Fairchild MD. Ganglion cell pathways for rod vision. Vis Res
Proc R Soc Lond B Biol Sci 1987;231:169–97. 1994;34:477–82.
23. Zhang DQ, Zhou TR, McMahon DG. Functional heterogeneity of retinal dopa- 61. Wassle H, Grunert U, Chun MH, et al. The rod pathway of the macaque
minergic neurons underlying their multiple roles in vision. J Neurosci monkey retina: identification of AII-amacrine cells with antibodies against
2007;27:692–9. calretinin. J Comp Neurol 1995;361:537–51.
24. Zhang DQ, Wong KY, Sollars PJ, et al. Intraretinal signaling by ganglion cell 62. Mills SL, Massey SC. AII amacrine cells limit scotopic acuity in central
photoreceptors to dopaminergic amacrine neurons. Proc Natl Acad Sci USA macaque retina: A confocal analysis of calretinin labeling. J Comp Neurol
2008;105:14181–6. 1999;411:19–34.
25. Dacey DM, Liao HW, Peterson BB, et al. Melanopsin-expressing ganglion 63. Chun MH, Grunert U, Martin PR, et al. The synaptic complex of cones in the
cells in primate retina signal colour and irradiance and project to the LGN. fovea and in the periphery of the macaque monkey retina. Vis Res
Nature 2005;433:749–54. 1996;36:3383–95.
26. Hoshi H, Liu WL, Massey SC, et al. ON inputs to the OFF layer: bipolar cells 64. Haverkamp S, Grunert U, Wassle H. The cone pedicle, a complex synapse in
that break the stratification rules of the retina. J Neurosci 2009;29:8875–83. the retina. Neuron 2000;27:85–95.
27. Dumitrescu ON, Pucci FG, Wong KY, et al. Ectopic retinal ON bipolar cell 65. Sterling P. Needle from a haystack. Optimal signaling by a nonlinear synapse.
synapses in the OFF inner plexiform layer: contacts with dopaminergic ama- Neuron 2002;34:670–2.
crine cells and melanopsin ganglion cells. J Comp Neurol 2009;517:226–44. 66. Sterling P, Smith RG. Design for a binary synapse. Neuron 2004;41:313–5.
28. Curcio CA, Sloan KR, Kalina RE, et al. Human photoreceptor topography. 67. Rao-Mirotznik R, Harkins AB, Buchsbaum G, et al. Mammalian rod terminal:
J Comp Neurol 1990;292:497–523. architecture of a binary synapse. Neuron 1995;14:561–9.
29. Curcio CA, Allen KA, Sloan KR, et al. Distribution and morphology of human 68. Li W, DeVries SH. Separate blue and green cone networks in the mammalian
cone photoreceptors stained with anti-blue opsin. J Comp Neurol 1991; retina. Nat Neurosci 2004;7:751–6.
312:610–24. 69. O’Brien J, Nguyen HB, Mills SL. Cone photoreceptors in bass retina use two
30. Wandell B. Foundations of vision. Sunderland, MA: Sinauer Associates; 1995. connexins to mediate electrical coupling. J Neurosci 2004;24:5632–42.
31. Masland RH, Raviola E. Confronting complexity: strategies for understanding 70. Hornstein EP, Verweij J, Schnapf JL. Electrical coupling between red and
the microcircuitry of the retina. Annu Rev Neurosci 2000;23:249–84. green cones in primate retina. Nat Neurosci 2004;7:745–50.
32. Denk W, Detwiler PB. Optical recording of light-evoked calcium signals in 71. DeVries SH, Qi X, Smith R, et al. Electrical coupling between mammalian
the functionally intact retina. Proc Natl Acad Sci USA 1999;96:7035–40. cones. Curr Biol 2002;12:1900–7.
33. Euler T, Detwiler PB, Denk W. Directionally selective calcium signals in 72. Ahnelt P, Keri C, Kolb H. Identification of pedicles of putative blue-sensitive
dendrites of starburst amacrine cells. Nature 2002;418:845–52. cones in the human retina. J Comp Neurol 1990;293:39–53.
34. Anderson JR, Jones BW, Watt CB, et al. Exploring the retinal connectome. 73. Raviola E, Gilula NB. Gap junctions between photoreceptor cells in the ver-
Mol Vis 2011;17:355–79. tebrate retina. Proc Natl Acad Sci U S A 1973;70:1677–81.
35. Dacey DM, Peterson BB, Robinson FR, et al. Fireworks in the primate retina: 74. Nelson R. Cat cones have rod input: a comparison of the response properties
in vitro photodynamics reveals diverse LGN-projecting ganglion cell types. of cones and horizontal cell bodies in the retina of the cat. J Comp Neurol
Neuron 2003;37:15–27. 1977;172:109–35.
36. Hattar S, Liao HW, Takao M, et al. Melanopsin-containing retinal ganglion 75. Schneeweis DM, Schnapf JL. The photovoltage of macaque cone photorecep-
cells: architecture, projections, and intrinsic photosensitivity. Science 2002; tors: adaptation, noise, and kinetics. J Neurosci 1999;19:1203–16.
295:1065–70. 76. Smith RG, Freed MA, Sterling P. Microcircuitry of the dark-adapted cat
37. MacNeil MA, Masland RH. Extreme diversity among amacrine cells: implica- retina: functional architecture of the rod-cone network. J Neurosci 1986;6:
tions for function. Neuron 1998;20:971–82. 3505–17.
38. MacNeil MA, Heussy JK, Dacheux RF, et al. The shapes and numbers of 77. Hornstein EP, Verweij J, Li PH, et al. Gap-junctional coupling and absolute
amacrine cells: matching of photofilled with Golgi-stained cells in the rabbit sensitivity of photoreceptors in macaque retina. J Neurosci 2005;25:11201–9.
retina and comparison with other mammalian species. J Comp Neurol 78. Peachey NS, Alexander KR, Fishman GA. Visual adaptation and the cone
1999;413:305–26. flicker electroretinogram. Invest Ophthalmol Vis Sci 1991;32:1517–22.
39. Gan WB, Grutzendler J, Wong WT, et al. Multicolor “DiOlistic” labeling of 79. Peachey NS, Arakawa K, Alexander KR, et al. Rapid and slow changes in the
the nervous system using lipophilic dye combinations. Neuron 2000;27: human cone electroretinogram during light and dark adaptation. Vis Res
219–25. 1992;32:2049–53.
40. Siegert S, Scherf BG, Del Punta K, et al. Genetic address book for retinal cell 80. Peachey NS, Ball SL. Electrophysiological analysis of visual function in
types. Nat Neurosci 2009;12:1197–204. mutant mice. Adv Ophthalmol 2003;107:13–36.
41. Wassle H, Riemann HJ. The mosaic of nerve cells in the mammalian retina. 81. Heikkinen H, Vinberg F, Nymark S, et al. Mesopic background lights enhance
Proc R Soc Lond B Biol Sci 1978;200:441–61. dark-adapted cone ERG flash responses in the intact mouse retina: a possible
42. Strettoi E, Dacheux RF, Raviola E. Synaptic connections of rod bipolar cells in role for gap junctional decoupling. J Neurophysiol 2011;105:2309–18.
the inner plexiform layer of the rabbit retina. J Comp Neurol 1990;295: 82. Ribelayga C, Cao Y, Mangel SC. The circadian clock in the retina controls
449–66. rod–cone coupling. Neuron 2008;59:790–801.
83. Arshavsky VY, Lamb TD, Pugh Jr EN. G proteins and phototransduction. 121. Verweij J, Hornstein EP, Schnapf JL. Surround antagonism in macaque cone
Annu Rev Physiol 2002;64:153–87. photoreceptors. J Neurosci 2003;23:10249–57.
84. Copenhagen DR, Jahr CE. Release of endogenous excitatory amino acids from 122. McMahon MJ, Packer OS, Dacey DM. The classical receptive field surround
turtle photoreceptors. Nature 1989;341:536–9. of primate parasol ganglion cells is mediated primarily by a non-GABAergic 397
85. Sarantis M, Mobbs P. The spatial relationship between Müller cell processes pathway. J Neurosci 2004;24:3736–45.
and the photoreceptor output synapse. Brain Res 1992;584:299–304. 123. Kamermans M, Fahrenfort I, Schultz K, et al. Hemichannel-mediated inhibi-
86. Schaeffer SF, Raviola E. Membrane recycling in the cone cell endings of the tion in the outer retina. Science 2001;292:1178–80.

Chapter 15
turtle retina. J Cell Biol 1978;79:802–25. 124. VanLeeuwen M, Fahrenfort I, Sjoerdsma T, et al. Lateral gain control in the
87. Grunert U, Martin PR. Rod bipolar cells in the macaque monkey retina: outer retina leads to potentiation of center responses of retinal neurons.
immunoreactivity and connectivity. J Neurosci 1991;11:2742–58. J Neurosci 2009;29:6358–66.
88. Young HM, Vaney DI. Rod-signal interneurons in the rabbit retina: 1. Rod 125. Hirasawa H, Kaneko A. pH changes in the invaginating synaptic cleft mediate
bipolar cells. J Comp Neurol 1991;310:139–53. feedback from horizontal cells to cone photoreceptors by modulating Ca2+
89. Vardi N, Morigiwa K, Wang TL, et al. Neurochemistry of the mammalian cone channels. J Gen Physiol 2003;122:657–71.
‘synaptic complex’. Vis Res 1998;38:1359–69. 126. Vessey JP, Stratis AK, Daniels BA, et al. Proton-mediated feedback inhibition
90. Haverkamp S, Grunert U, Wassle H. The synaptic architecture of AMPA of presynaptic calcium channels at the cone photoreceptor synapse. J Neurosci
receptors at the cone pedicle of the primate retina. J Neurosci 2001;21: 2005;25:4108–17.
2488–500. 127. Jackman SL, Babai N, Chambers JJ, et al. A positive feedback synapse from

Function and Anatomy of the Mammalian Retina


91. Kolb H. The connections between horizontal cells and photoreceptors in the retinal horizontal cells to cone photoreceptors. PLoS Biol 2011;9:e1001057.
retina of the cat: electron microscopy of Golgi preparations. J Comp Neurol 128. Klaassen LJ, Sun Z, Steijaert MN, et al. Synaptic transmission from horizontal
1974;155:1–14. cells to cones is impaired by loss of connexin hemichannels. PLoS Biol
92. Boycott BB, Peichl L, Wassle H. Morphological types of horizontal cell in the 2011;9:e1001107.
retina of the domestic cat. Proc R Soc Lond B Biol Sci 1978;203:229–45. 129. Boycott BB, Wassle H. Morphological classification of bipolar cells of the
93. Dacheux RF, Raviola E. Horizontal cells in the retina of the rabbit. J Neurosci primate retina. Eur J Neurosci 1991;3:1069–88.
1982;2:1486–93. 130. Grunert U, Martin PR, Wassle H. Immunocytochemical analysis of bipolar
94. Peichl L, Gonzalez-Soriano J. Morphological types of horizontal cell in rodent cells in the macaque monkey retina. J Comp Neurol 1994;348:607–27.
retinae: a comparison of rat, mouse, gerbil, and guinea pig. Vis Neurosci 131. Ghosh KK, Bujan S, Haverkamp S, et al. Types of bipolar cells in the mouse
1994;11:501–17. retina. J Comp Neurol 2004;469:70–82.
95. Mills SL, Massey SC. Distribution and coverage of A- and B-type horizontal 132. MacNeil MA, Heussy JK, Dacheux RF, et al. The population of bipolar cells
cells stained with Neurobiotin in the rabbit retina. Vis Neurosci 1994;11: in the rabbit retina. J Comp Neurol 2004;472:73–86.
549–60. 133. Haverkamp S, Ghosh KK, Hirano AA, et al. Immunocytochemical description
96. Haverkamp S, Grunert U, Wassle H. Localization of kainate receptors at the of five bipolar cell types of the mouse retina. J Comp Neurol 2003;455:
cone pedicles of the primate retina. J Comp Neurol 2001;436:471–86. 463–76.
97. Li W, Keung JW, Massey SC. Direct synaptic connections between rods and 134. Haverkamp S, Specht D, Majumdar S, et al. Type 4 OFF cone bipolar cells of
OFF cone bipolar cells in the rabbit retina. J Comp Neurol 2004;474:1–12. the mouse retina express calsenilin and contact cones as well as rods. J Comp
98. Nelson R, von Litzow A, Kolb H, et al. Horizontal cells in cat retina with Neurol 2008;507:1087–101.
independent dendritic systems. Science 1975;189:137–9. 135. Puller C, Haverkamp S. Cell-type-specific localization of protocadherin beta16
99. Vaney DI. Many diverse types of retinal neurons show tracer coupling when at AMPA and AMPA/Kainate receptor-containing synapses in the primate
injected with biocytin or Neurobiotin. Neurosci Lett 1991;125:187–90. retina. J Comp Neurol 2011;519:467–79.
100. Vaney DI. The coupling pattern of axon-bearing horizontal cells in the mam- 136. Puthussery T, Gayet-Primo J, Taylor WR. Localization of the calcium-binding
malian retina. Proc Biol Sci 1993;252:93–101. protein secretagogin in cone bipolar cells of the mammalian retina. J Comp
101. Mills SL, Massey SC. The kinetics of tracer movement through homologous Neurol 2010;518:513–25.
gap junctions in the rabbit retina. Vis Neurosci 1998;15:765–77. 137. Dhingra A, Sulaiman P, Xu Y, et al. Probing neurochemical structure and
102. Boycott BB, Hopkins JM, Sperling HG. Cone connections of the horizontal function of retinal ON bipolar cells with a transgenic mouse. J Comp Neurol
cells of the rhesus monkey’s retina. Proc R Soc Lond B Biol Sci 1987;229: 2008;510:484–96.
345–79. 138. Schubert T, Kerschensteiner D, Eggers ED, et al. Development of presynaptic
103. Dacey DM, Lee BB, Stafford DK, et al. Horizontal cells of the primate retina: inhibition onto retinal bipolar cell axon terminals is subclass-specific. J Neu-
cone specificity without spectral opponency. Science 1996;271:656–9. rophysiol 2008;100:304–16.
104. Wassle H, Dacey DM, Haun T, et al. The mosaic of horizontal cells in the 139. Murakami M, Otsuka T, Shimazaki H. Effects of aspartate and glutamate on
macaque monkey retina: with a comment on biplexiform ganglion cells. the bipolar cells in the carp retina. Vis Res 1975;15:456–8.
Vis Neurosci 2000;17:591–608. 140. Slaughter MM, Miller RF. The role of excitatory amino acid transmitters in
105. Packer OS, Dacey DM. Receptive field structure of H1 horizontal cells in the mudpuppy retina: an analysis with kainic acid and N-methyl aspartate.
macaque monkey retina. J Vis 2002;2:272–92. J Neurosci 1983;3:1701–11.
106. Massey SC, O’Brien JJ, Trexler EB, et al. Multiple neuronal connexins in the 141. DeVries SH. Bipolar cells use kainate and AMPA receptors to filter visual
mammalian retina. Cell Commun Adhes 2003;10:425–30. information into separate channels. Neuron 2000;28:847–56.
107. Hombach S, Janssen-Bienhold U, Sohl G, et al. Functional expression of con- 142. Nomura A, Shigemoto R, Nakamura Y, et al. Developmentally regulated
nexin57 in horizontal cells of the mouse retina. Eur J Neurosci 2004;19: postsynaptic localization of a metabotropic glutamate receptor in rat rod
2633–40. bipolar cells. Cell 1994;77:361–9.
108. Sterling P. How retinal circuits optimize the transfer of visual information. In: 143. Dhingra A, Lyubarsky A, Jiang M, et al. The light response of ON bipolar
Chalupa LM, editor. The visual neurosciences. Cambridge, MA: MIT Press; neurons requires Gαo. J Neurosci 2000;20:9053–8.
2004. p. 234–59. 144. Shen Y, Heimel JA, Kamermans M, et al. A transient receptor potential-like
109. Mahowald MA, Mead C. The silicon retina. Sci Am 1991;264:76–82. channel mediates synaptic transmission in rod bipolar cells. J Neurosci
110. Crook JD, Manookin MB, Packer OS, et al. Horizontal cell feedback without 2009;29:6088–93.
cone type-selective inhibition mediates “red–green” color opponency in 145. Morgans CW, Zhang J, Jeffrey BG, et al. TRPM1 is required for the depolar-
midget ganglion cells of the primate retina. J Neurosci 2011;31:1762–72. izing light response in retinal ON-bipolar cells. Proc Natl Acad Sci U S A
111. Baylor DA, O’Bryan PM. Electrical signaling in vertebrate photoreceptors. 2009;106:19174–8.
Fed Proc 1971;30:79–83. 146. Koike C, Numata T, Ueda H, et al. TRPM1: a vertebrate TRP channel respon-
112. Wu SM. Feedback connections and operation of the outer plexiform layer of sible for retinal ON bipolar function. Cell Calcium 2010;48:95–101.
the retina. Curr Opin Neurobiol 1992;2:462–8. 147. Nawy S. The metabotropic receptor mGluR6 may signal through G(o), but not
113. Vardi N, Sterling P. Subcellular localization of GABAA receptor on bipolar phosphodiesterase, in retinal bipolar cells. J Neurosci 1999;19:2938–44.
cells in macaque and human retina. Vis Res 1994;34:1235–46. 148. Dhingra A, Faurobert E, Dascal N, et al. A retinal-specific regulator
114. Marc R. Retinal neurotransmitters. In: Claw JS, editor. The visual sciences. of G-protein signaling interacts with Galpha(o) and accelerates an
Cambridge, MA: MIT Press; 2004. p. 301–19. expressed metabotropic glutamate receptor 6 cascade. J Neurosci 2004;24:
115. Cueva JG, Haverkamp S, Reimer RJ, et al. Vesicular gamma-aminobutyric 5684–93.
acid transporter expression in amacrine and horizontal cells. J Comp Neurol 149. Awatramani GB, Slaughter MM. Intensity-dependent, rapid activation of pre-
2002;445:227–37. synaptic metabotropic glutamate receptors at a central synapse. J Neurosci
116. Hirano AA, Brandstatter JH, Morgans CW, et al. SNAP25 expression in mam- 2001;21:741–9.
malian retinal horizontal cells. J Comp Neurol 2011;519:972–88. 150. Dacey DM. The mosaic of midget ganglion cells in the human retina.
117. Mercer AJ, Rabl K, Riccardi GE, et al. Location of release sites and calcium- J Neurosci 1993;13:5334–55.
activated chloride channels relative to calcium channels at the photoreceptor 151. Mariani AP. Bipolar cells in monkey retina selective for the cones likely to be
ribbon synapse. J Neurophysiol 2011;105:321–35. blue-sensitive. Nature 1984;308:184–6.
118. Zampighi GA, Schietroma C, Zampighi LM, et al. Conical tomography of a 152. Kouyama N, Marshak DW. Bipolar cells specific for blue cones in the macaque
ribbon synapse: structural evidence for vesicle fusion. PloS ONE 2011;6:e16944. retina. J Neurosci 1992;12:1233–52.
119. Shields CR, Tran MN, Wong RO, et al. Distinct ionotropic GABA receptors 153. Breuninger T, Puller C, Haverkamp S, et al. Chromatic bipolar cell pathways
mediate presynaptic and postsynaptic inhibition in retinal bipolar cells. in the mouse retina. J Neurosci 2011;31:6504–17.
J Neurosci 2000;20:2673–82. 154. Bloomfield SA, Dacheux RF. Rod vision: pathways and processing in the
120. Picaud S, Pattnaik B, Hicks D, et al. GABAA and GABAC receptors in adult mammalian retina. Prog Retin Eye Res 2001;20:351–84.
porcine cones: evidence from a photoreceptor-glia co-culture model. J Physiol 155. Field GD, Rieke F. Nonlinear signal transfer from mouse rods to bipolar cells
1998;513:33–42. and implications for visual sensitivity. Neuron 2002;34:773–85.
156. Sampath AP, Rieke F. Selective transmission of single photon responses by 192. Xin D, Bloomfield SA. Tracer coupling pattern of amacrine and ganglion cells
saturation at the rod-to-rod bipolar synapse. Neuron 2004;41:431–43. in the rabbit retina. J Comp Neurol 1997;383:512–28.
157. Ghosh KK, Haverkamp S, Wassle H. Glutamate receptors in the rod pathway 193. Marc RE, Jones BW. Molecular phenotyping of retinal ganglion cells.
398 of the mammalian retina. J Neurosci 2001;21:8636–47. J Neurosci 2002;22:413–27.
158. Li W, Trexler EB, Massey SC. Glutamate receptors at rod bipolar ribbon syn- 194. Hu EH, Pan F, Volgyi B, et al. Light increases the gap junctional coupling of
apses in the rabbit retina. J Comp Neurol 2002;448:230–48. retinal ganglion cells. J Physiol 2010;588:4145–63.
159. Chavez AE, Singer JH, Diamond JS. Fast neurotransmitter release triggered 195. Berry 2nd MJ, Brivanlou IH, Jordan TA, et al. Anticipation of moving stimuli
by Ca influx through AMPA-type glutamate receptors. Nature 2006;443: by the retina. Nature 1999;398:334–8.
Section 1

705–8. 196. Olveczky BP, Baccus SA, Meister M. Segregation of object and background
160. Eggers ED, McCall MA, Lukasiewicz PD. Presynaptic inhibition differentially motion in the retina. Nature 2003;423:401–8.
shapes transmission in distinct circuits in the mouse retina. J Physiol 197. Roska B, Werblin F. Rapid global shifts in natural scenes block spiking in
2007;582:569–82. specific ganglion cell types. Nat Neurosci 2003;6:600–8.
161. Eggers ED, Lukasiewicz PD. Interneuron circuits tune inhibition in retinal 198. Knop GC, Feigenspan A, Weiler R, et al. Inputs underlying the ON-OFF light
bipolar cells. J Neurophysiol 2010;103:25–37. responses of type 2 wide-field amacrine cells in TH::GFP mice. J Neurosci
162. Chavez AE, Grimes WN, Diamond JS. Mechanisms underlying lateral GABA­ 2011;31:4780–91.
ergic feedback onto rod bipolar cells in rat retina. J Neurosci 2010;30: 199. Famiglietti Jr EV, Kolb H. A bistratified amacrine cell and synaptic circuitry
Anatomy and Physiology

2330–9. in the inner plexiform layer of the retina. Brain Res 1975;84:293–300.
Basic Science and Translation to Therapy

163. Ivanova E, Müller U, Wassle H. Characterization of the glycinergic input to 200. Massey SC, Mills SL. Antibody to calretinin stains AII amacrine cells in the
bipolar cells of the mouse retina. Eur J Neurosci 2006;23:350–64. rabbit retina: double-label and confocal analyses. J Comp Neurol 1999;411:
164. Weiss J, O’Sullivan GA, Heinze L, et al. Glycinergic input of small-field ama- 3–18.
crine cells in the retinas of wildtype and glycine receptor deficient mice. 201. Smith RG, Vardi N. Simulation of the AII amacrine cell of mammalian retina:
Mol Cell Neurosci 2008;37:40–55. functional consequences of electrical coupling and regenerative membrane
165. Strettoi E, Dacheux RF, Raviola E. Cone bipolar cells as interneurons in the properties. Vis Neurosci 1995;12:851–60.
rod pathway of the rabbit retina. J Comp Neurol 1994;347:139–49. 202. Vardi N, Smith RG. The AII amacrine network: coupling can increase
166. Mills SL, Massey SC. Differential properties of two gap junctional pathways correlated activity. Vis Res 1996;36:3743–57.
made by AII amacrine cells. Nature 1995;377:734–7. 203. Bloomfield SA, Volgyi B. Function and plasticity of homologous coupling
167. Vaney DI, Nelson JC, Pow DV. Neurotransmitter coupling through gap between AII amacrine cells. Vis Res 2004;44:3297–306.
junctions in the retina. J Neurosci 1998;18:10594–602. 204. Manookin MB, Beaudoin DL, Ernst ZR, et al. Disinhibition combines with
168. Deans MR, Volgyi B, Goodenough DA, et al. Connexin36 is essential for excitation to extend the operating range of the OFF visual pathway in day-
transmission of rod-mediated visual signals in the mammalian retina. Neuron light. J Neurosci 2008;28:4136–50.
2002;36:703–12. 205. Munch TA, da Silveira RA, Siegert S, et al. Approach sensitivity in the retina
169. Hampson EC, Vaney DI, Weiler R. Dopaminergic modulation of gap junction processed by a multifunctional neural circuit. Nat Neurosci 2009;12:1308–16.
permeability between amacrine cells in mammalian retina. J Neurosci 206. Oesch NW, Kothmann WW, Diamond JS. Illuminating synapses and circuitry
1992;12:4911–22. in the retina. Curr Opin Neurobiol 2011;21:238–44.
170. Kothmann WW, Massey SC, O’Brien J. Dopamine-stimulated dephosphoryla- 207. Werblin FS. Six different roles for crossover inhibition in the retina: correcting
tion of connexin 36 mediates AII amacrine cell uncoupling. J Neurosci the nonlinearities of synaptic transmission. Vis Neurosci 2010;27:1–8.
2009;29:14903–11. 208. Vaney DI. Morphological identification of serotonin-accumulating neurons in
171. Trexler EB, Li W, Mills SL, et al. Coupling from AII amacrine cells to ON cone the living retina. Science 1986;233:444–6.
bipolar cells is bidirectional. J Comp Neurol 2001;437:408–22. 209. Zhang J, Li W, Trexler EB, et al. Confocal analysis of reciprocal feedback at
172. Veruki ML, Hartveit E. Electrical synapses mediate signal transmission in the rod bipolar terminals in the rabbit retina. J Neurosci 2002;22:10871–82.
rod pathway of the mammalian retina. J Neurosci 2002;22:10558–66. 210. Fletcher EL, Wassle H. Indoleamine-accumulating amacrine cells are presyn-
173. Mills SL, O’Brien JJ, Li W, et al. Rod pathways in the mammalian retina use aptic to rod bipolar cells through GABA(C) receptors. J Comp Neurol
connexin 36. J Comp Neurol 2001;436:336–50. 1999;413:155–67.
174. Maxeiner S, Dedek K, Janssen-Bienhold U, et al. Deletion of connexin45 in 211. Eggers ED, Lukasiewicz PD. Receptor and transmitter release properties set
mouse retinal neurons disrupts the rod/cone signaling pathway between AII the time course of retinal inhibition. J Neurosci 2006;26:9413–25.
amacrine and ON cone bipolar cells and leads to impaired visual transmis- 212. Eggers ED, Lukasiewicz PD. GABA(A), GABA(C) and glycine receptor-
sion. J Neurosci 2005;25:566–76. mediated inhibition differentially affects light-evoked signalling from mouse
175. DeVries SH, Baylor DA. An alternative pathway for signal flow from rod retinal rod bipolar cells. J Physiol 2006;572:215–25.
photoreceptors to ganglion cells in mammalian retina. Proc Natl Acad Sci 213. Lukasiewicz PD, Eggers ED, Sagdullaev BT, et al. GABAC receptor-mediated
U S A 1995;92:10658–62. inhibition in the retina. Vis Res 2004;44:3289–96.
176. Soucy E, Wang Y, Nirenberg S, et al. A novel signaling pathway from rod 214. Grimes WN, Li W, Chavez AE, et al. BK channels modulate pre- and postsyn-
photoreceptors to ganglion cells in mammalian retina. Neuron 1998;21: aptic signaling at reciprocal synapses in retina. Nat Neurosci 2009;12:
481–93. 585–92.
177. Hack I, Frech M, Dick O, et al. Heterogeneous distribution of AMPA 215. Freed MA, Smith RG, Sterling P. Timing of quantal release from the
glutamate receptor subunits at the photoreceptor synapses of rodent retina. retinal bipolar terminal is regulated by a feedback circuit. Neuron 2003;38:
Eur J Neurosci 2001;13:15–24. 89–101.
178. Tsukamoto Y, Morigiwa K, Ueda M, et al. Microcircuits for night vision in 216. Sagdullaev BT, McCall MA, Lukasiewicz PD. Presynaptic inhibition modu-
mouse retina. J Neurosci 2001;21:8616–23. lates spillover, creating distinct dynamic response ranges of sensory output.
179. Fyk-Kolodziej B, Qin P, Pourcho RG. Identification of a cone bipolar cell in Neuron 2006;50:923–35.
cat retina which has input from both rod and cone photoreceptors. J Comp 217. Dacey DM. The dopaminergic amacrine cell. J Comp Neurol 1990;301:
Neurol 2003;464:104–13. 461–89.
180. Li W, Chen S, DeVries SH. A fast rod photoreceptor signaling pathway in the 218. Hokoc JN, Mariani AP. Synapses from bipolar cells onto dopaminergic ama-
mammalian retina. Nat Neurosci 2010;13:414–6. crine cells in cat and rabbit retinas. Brain Res 1988;461:17–26.
181. Lin B, Masland RH. Populations of wide-field amacrine cells in the mouse 219. Contini M, Lin B, Kobayashi K, et al. Synaptic input of ON-bipolar cells onto
retina. J Comp Neurol 2006;499:797–809. the dopaminergic neurons of the mouse retina. J Comp Neurol 2010;518:
182. Perez De Sevilla Müller L, Shelley J, Weiler R. Displaced amacrine cells of the 2035–50.
mouse retina. J Comp Neurol 2007;505:177–89. 220. Contini M, Raviola E. GABAergic synapses made by a retinal dopaminergic
183. Zhang C, Mccall MA. Receptor targets of amacrine cells. Vis Neurosci 2012 neuron. Proc Natl Acad Sci U S A 2003;100:1358–63.
(in press). 221. Feigenspan A, Gustincich S, Bean BP, et al. Spontaneous activity of solitary
184. Hsueh HA, Molnar A, Werblin FS. Amacrine-to-amacrine cell inhibition in dopaminergic cells of the retina. J Neurosci 1998;18:6776–89.
the rabbit retina. J Neurophysiol 2008;100:2077–88. 222. Puopolo M, Hochstetler SE, Gustincich S, et al. Extrasynaptic release of dopa-
185. Badea TC, Nathans J. Quantitative analysis of neuronal morphologies in the mine in a retinal neuron: activity dependence and transmitter modulation.
mouse retina visualized by using a genetically directed reporter. J Comp Neuron 2001;30:211–25.
Neurol 2004;480:331–51. 223. Witkovsky P, Veisenberger E, LeSauter J, et al. Cellular location and circadian
186. Dedek K, Breuninger T, de Sevilla Müller LP, et al. A novel type of rhythm of expression of the biological clock gene Period 1 in the mouse retina.
interplexiform amacrine cell in the mouse retina. Eur J Neurosci 2009;30: J Neurosci 2003;23:7670–6.
217–28. 224. Gustincich S, Contini M, Gariboldi M, et al. Gene discovery in genetically
187. Majumdar S, Weiss J, Wassle H. Glycinergic input of widefield, displaced labeled single dopaminergic neurons of the retina. Proc Natl Acad Sci U S A
amacrine cells of the mouse retina. J Physiol 2009;587:3831–49. 2004;101:5069–74.
188. Grimes WN, Zhang J, Graydon CW, et al. Retinal parallel processors: more 225. Dorenbos R, Contini M, Hirasawa H, et al. Expression of circadian clock genes
than 100 independent microcircuits operate within a single interneuron. in retinal dopaminergic cells. Vis Neurosci 2007;24:573–80.
Neuron 2010;65:873–85. 226. Pozdeyev N, Tosini G, Li L, et al. Dopamine modulates diurnal and circadian
189. Lukasiewicz PD. Synaptic mechanisms that shape visual signaling at the inner rhythms of protein phosphorylation in photoreceptor cells of mouse retina.
retina. Prog Brain Res 2005;147:205–18. Eur J Neurosci 2008;27:2691–700.
190 Demb JB. Functional circuitry of visual adaptation in the retina. J Physiol 227. Witkovsky P. Dopamine and retinal function. Adv Ophthalmol 2004;108:
2008;586:4377–84. 17–40.
191. Li W, Zhang J, Massey SC. Coupling pattern of S1 and S2 amacrine cells in 228. Ribelayga C, Mangel SC. Identification of a circadian clock-controlled neural
the rabbit retina. Vis Neurosci 2002;19:119–31. pathway in the rabbit retina. PloS ONE 2010;5:e11020.
229. Herrmann R, Heflin SJ, Hammond T, et al. Rod vision is controlled by 265. Kim IJ, Zhang Y, Yamagata M, et al. Molecular identification of a retinal cell
dopamine-dependent sensitization of rod bipolar cells by GABA. Neuron type that responds to upward motion. Nature 2008;452:478–82.
2011;72:101–10. 266. Kim IJ, Zhang Y, Meister M, et al. Laminar restriction of retinal ganglion cell
230. Vaney DI, Pow DV. The dendritic architecture of the cholinergic plexus in the dendrites and axons: subtype-specific developmental patterns revealed with 399
rabbit retina: selective labeling by glycine accumulation in the presence of transgenic markers. J Neurosci 2010;30:1452–62.
sarcosine. J Comp Neurol 2000;421:1–13. 267. Yonehara K, Ishikane H, Sakuta H, et al. Identification of retinal ganglion cells
231. Dacheux RF, Chimento MF, Amthor FR. Synaptic input to the on-off direction- and their projections involved in central transmission of information about

Chapter 15
ally selective ganglion cell in the rabbit retina. J Comp Neurol 2003;456: upward and downward image motion. PloS ONE 2009;4:e4320.
267–78. 268. Rivlin-Etzion M, Zhou K, Wei W, et al. Transgenic mice reveal unexpected
232. Brecha N, Johnson D, Peichl L, et al. Cholinergic amacrine cells of the rabbit diversity of ON-OFF direction-selective retinal ganglion cell subtypes and
retina contain glutamate decarboxylase and gamma-aminobutyrate immuno- brain structures involved in motion processing. J Neurosci 2011;31:8760–9.
reactivity. Proc Natl Acad Sci U S A 1988;85:6187–91. 269. Volgyi B, Chheda S, Bloomfield SA. Tracer coupling patterns of the ganglion
233. Lee S, Kim K, Zhou ZJ. Role of ACh-GABA cotransmission in detecting image cell subtypes in the mouse retina. J Comp Neurol 2009;512:664–87.
motion and motion direction. Neuron 2010;68:1159–72. 270. Roska B, Werblin F. Vertical interactions across ten parallel, stacked represen-
234. Yoshida K, Watanabe D, Ishikane H, et al. A key role of starburst amacrine tations in the mammalian retina. Nature 2001;410:583–7.
cells in originating retinal directional selectivity and optokinetic eye move- 271. Roska B, Molnar A, Werblin FS. Parallel processing in retinal ganglion cells:
ment. Neuron 2001;30:771–80. how integration of space-time patterns of excitation and inhibition form the

Function and Anatomy of the Mammalian Retina


235. Kittila CA, Massey SC. Pharmacology of directionally selective ganglion cells spiking output. J Neurophysiol 2006;95:3810–22.
in the rabbit retina. J Neurophysiol 1997;77:675–89. 272. van Wyk M, Wassle H, Taylor WR. Receptive field properties of ON- and
236. Fried SI, Munch TA, Werblin FS. Mechanisms and circuitry underlying OFF-ganglion cells in the mouse retina. Vis Neurosci 2009;26:297–308.
directional selectivity in the retina. Nature 2002;420:411–4. 273. Farrow K, Masland RH. Physiological clustering of visual channels in the
237. Borg-Graham LJ. The computation of directional selectivity in the retina mouse retina. J Neurophysiol 2011;105:1516–30.
occurs presynaptic to the ganglion cell. Nat Neurosci 2001;4:176–83. 274. Dacey DM. Morphology of a small-field bistratified ganglion cell type in the
238. Hausselt SE, Euler T, Detwiler PB, et al. A dendrite-autonomous mechanism macaque and human retina. Vis Neurosci 1993;10:1081–98.
for direction selectivity in retinal starburst amacrine cells. PLoS Biol 275. Peichl L. Alpha ganglion cells in mammalian retinae: common properties,
2007;5:e185. species differences, and some comments on other ganglion cells. Vis Neurosci
239. Gavrikov KE, Nilson JE, Dmitriev AV, et al. Dendritic compartmentalization 1991;7:155–69.
of chloride cotransporters underlies directional responses of starburst ama- 276. Barlow HB, Levick WR. The mechanism of directionally selective units in
crine cells in retina. Proc Natl Acad Sci U S A 2006;103:18793–8. rabbit’s retina. J Physiol 1965;178:477–504.
240. Briggman KL, Helmstaedter M, Denk W. Wiring specificity in the direction- 277. Amthor FR, Oyster CW, Takahashi ES. Morphology of ON-OFF direction-
selectivity circuit of the retina. Nature 2011;471:183–8. selective ganglion cells in the rabbit retina. Brain Res 1984;298:187–90.
241. Oesch N, Euler T, Taylor WR. Direction-selective dendritic action potentials 278. Wei W, Elstrott J, Feller MB. Two-photon targeted recording of GFP-expressing
in rabbit retina. Neuron 2005;47:739–50. neurons for light responses and live-cell imaging in the mouse retina. Nature
242. Wei W, Feller MB. Organization and development of direction-selective protocols 2010;5:1347–52.
circuits in the retina. Trends Neurosci 2011;34:638–45. 279. Vaney DI. Territorial organization of direction-selective ganglion cells in
243. Elstrott J, Anishchenko A, Greschner M, et al. Direction selectivity in the retina rabbit retina. J Neurosci 1994;14:6301–16.
is established independent of visual experience and cholinergic retinal waves. 280. Kay JN, De la Huerta I, Kim IJ, et al. Retinal ganglion cells with distinct
Neuron 2008;58:499–506. directional preferences differ in molecular identity, structure, and central
244. Feller MB. The role of nAChR-mediated spontaneous retinal activity in visual projections. J Neurosci 2011;31:7753–62.
system development. J Neurobiol 2002;53:556–67. 281. Oyster CW, Barlow HB. Direction-selective units in rabbit retina: distribution
245. Syed MM, Lee S, Zheng J, et al. Stage-dependent dynamics and modulation of preferred directions. Science 1967;155:841–2.
of spontaneous waves in the developing rabbit retina. J Physiol 2004;560: 282. Isayama T, Berson DM, Pu M. Theta ganglion cell type of cat retina. J Comp
533–49. Neurol 2000;417:32–48.
246. Syed MM, Lee S, He S, et al. Spontaneous waves in the ventricular zone of 283. Yonehara K, Balint K, Noda M, et al. Spatially asymmetric reorganization of
developing mammalian retina. J Neurophysiol 2004;91:1999–2009. inhibition establishes a motion-sensitive circuit. Nature 2011;469:407–10.
247. Renna JM, Weng S, Berson DM. Light acts through melanopsin to alter retinal 284. Oyster CW, Simpson JI, Takahashi ES, et al. Retinal ganglion cells projecting
waves and segregation of retinogeniculate afferents. Nat Neurosci to the rabbit accessory optic system. J Comp Neurol 1980;190:49–61.
2011;14:827–9. 285. Buhl EH, Peichl L. Morphology of rabbit retinal ganglion cells projecting to
248. Lukasiewicz PD, Wilson JA, Lawrence JE. AMPA-preferring receptors the medial terminal nucleus of the accessory optic system. J Comp Neurol
mediate excitatory synaptic inputs to retinal ganglion cells. J Neurophysiol 1986;253:163–74.
1997;77:57–64. 286. Hoshi H, Tian LM, Massey SC, et al. Two distinct types of ON directionally
249. Vaney DI. A quantitative comparison between the ganglion cell populations selective ganglion cells in the rabbit retina. J Comp Neurol 2011;519:
and axonal outflows of the visual streak and periphery of the rabbit retina. 2509–21.
J Comp Neurol 1980;189:215–33. 287. Kanjhan R, Sivyer B. Two types of ON direction-selective ganglion cells in
250. Williams RW, Strom RC, Rice DS, et al. Genetic and environmental control of rabbit retina. Neurosci Lett 2010;483:105–9.
variation in retinal ganglion cell number in mice. J Neurosci 1996;16: 288. Ackert JM, Farajian R, Volgyi B, et al. GABA blockade unmasks an OFF
7193–205. response in ON direction selective ganglion cells in the mammalian retina.
251. Levick WR. Receptive fields of rabbit retinal ganglion cells. Am J Optom J Physiol 2009;587:4481–95.
1965;42:337–43. 289. Hoshi H, Mills SL. Components and properties of the G3 ganglion cell circuit
252. Caldwell JH, Daw NW. New properties of rabbit retinal ganglion cells. in the rabbit retina. J Comp Neurol 2009;513:69–82.
J Physiol 1978;276:257–76. 290. Venkataramani S, Taylor WR. Orientation selectivity in rabbit retinal ganglion
253. DeVries SH, Baylor DA. Mosaic arrangement of ganglion cell receptive fields cells is mediated by presynaptic inhibition. J Neurosci 2010;30:15664–76.
in rabbit retina. J Neurophysiol 1997;78:2048–60. 291. Trenholm S, Johnson K, Li X, et al. Parallel mechanisms encode direction in
254. DeVries SH. Correlated firing in rabbit retinal ganglion cells. J Neurophysiol the retina. Neuron 2011;71:683–94.
1999;81:908–20. 292. Klerman EB, Shanahan TL, Brotman DJ, et al. Photic resetting of the human
255. Hu EH, Bloomfield SA. Gap junctional coupling underlies the short-latency circadian pacemaker in the absence of conscious vision. J Biol Rhythms
spike synchrony of retinal alpha ganglion cells. J Neurosci 2003;23:6768–77. 2002;17:548–55.
256. Rockhill RL, Daly FJ, MacNeil MA, et al. The diversity of ganglion cells in a 293. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells
mammalian retina. J Neurosci 2002;22:3831–43. that set the circadian clock. Science 2002;295:1070–3.
257. Dacey DM. Physiology, morphology and spatial densities of identified gan- 294. Berson DM. Strange vision: ganglion cells as circadian photoreceptors. Trends
glion cell types in primate retina. Ciba Found Symposium 1994;184:12–28; Neurosci 2003;26:314–20.
discussion -34, 63–70. 295. Lucas RJ, Hattar S, Takao M, et al. Diminished pupillary light reflex at high
258. Peterson BB, Dacey DM. Morphology of human retinal ganglion cells with irradiances in melanopsin-knockout mice. Science 2003;299:245–7.
intraretinal axon collaterals. Vis Neurosci 1998;15:377–87. 296. Grunert U, Jusuf PR, Lee SC, et al. Bipolar input to melanopsin containing
259. Dacey DM. Primate retina: cell types, circuits and color opponency. Prog Retin ganglion cells in primate retina. Vis Neurosci 2011;28:39–50.
Eye Res 1999;18:737–63. 297. Hattar S, Lucas RJ, Mrosovsky N, et al. Melanopsin and rod-cone photorecep-
260. Yamada ES, Bordt AS, Marshak DW. Wide-field ganglion cells in macaque tive systems account for all major accessory visual functions in mice. Nature
retinas. Vis Neurosci 2005;22:383–93. 2003;424:76–81.
261. Sun W, Li N, He S. Large-scale morophological survey of rat retinal ganglion 298. Schmidt TM, Chen SK, Hattar S. Intrinsically photosensitive retinal ganglion
cells. Vis Neurosci 2002;19:483–93. cells: many subtypes, diverse functions. Trends Neurosci 2011;34:572–80.
262. Sun W, Li N, He S. Large-scale morphological survey of mouse retinal 299. Schmidt TM, Do MT, Dacey D, et al. Melanopsin-positive intrinsically photo-
ganglion cells. J Comp Neurol 2002;451:115–26. sensitive retinal ganglion cells: from form to function. J Neurosci 2011;31:
263. Huberman AD, Manu M, Koch SM, et al. Architecture and activity-mediated 16094–101.
refinement of axonal projections from a mosaic of genetically identified retinal 300. Berson DM, Castrucci AM, Provencio I. Morphology and mosaics of
ganglion cells. Neuron 2008;59:425–38. melanopsin-expressing retinal ganglion cell types in mice. J Comp Neurol
264. Huberman AD, Wei W, Elstrott J, et al. Genetic identification of an ON-OFF 2010;518:2405–22.
direction-selective retinal ganglion cell subtype reveals a layer-specific sub- 301. Chen SK, Badea TC, Hattar S. Photoentrainment and pupillary light reflex are
cortical map of posterior motion. Neuron 2009;62:327–34. mediated by distinct populations of ipRGCs. Nature 2011;476:92–5.
302. Lennie P. Color vision: putting it together. Curr Biol 2000;10:R589–91. 313. Yin L, Smith RG, Sterling P, et al. Physiology and morphology of color-
303. Dacey DM. Circuitry for color coding in the primate retina. Proc Natl Acad opponent ganglion cells in a retina expressing a dual gradient of S and M
Sci U S A 1996;93:582–8. opsins. J Neurosci 2009;29:2706–24.
400 304. Fahrenfort I, Steijaert M, Sjoerdsma T, et al. Hemichannel-mediated and 314. Jacobs GH, Williams GA, Cahill H, et al. Emergence of novel color vision in
pH-based feedback from horizontal cells to cones in the vertebrate retina. mice engineered to express a human cone photopigment. Science 2007;315:
PloS ONE 2009;4:e6090. 1723–5.
305. Field GD, Gauthier JL, Sher A, et al. Functional connectivity in the retina at 315. Mancuso K, Hauswirth WW, Li Q, et al. Gene therapy for red-green colour
the resolution of photoreceptors. Nature 2010;467:673–7. blindness in adult primates. Nature 2009;461:784–7.
Section 1

306. Mariani AP. The neuronal organization of the outer plexiform layer of the 316. Osterhout JA, Josten N, Yamada J, et al. Cadherin-6 mediates axon-target
primate retina. Int Rev Cytol 1984;86:285–320. matching in a non-image-forming visual circuit. Neuron 2011;71:632–9.
307. Haverkamp S, Wassle H, Duebel J, et al. The primordial, blue-cone color 317. Siegert S, Cabuy E, Gross Scherf B, et al. Transciptional code and disease map
system of the mouse retina. J Neurosci 2005;25:5438–45. for adult retinal cell types. Nat Neurosci 2012;15:487–95.
308. Dacey DM, Lee BB. The ‘blue-on’ opponent pathway in primate retina origi- 318. Harwerth RS, Carter-Dawson L, Shen F, et al. Ganglion cell losses underlying
nates from a distinct bistratified ganglion cell type. Nature 1994;367:731–5. visual field defects from experimental glaucoma. Invest Ophthalmol Vis Sci
309. Field GD, Sher A, Gauthier JL, et al. Spatial properties and functional organi- 1999;40:2242–50.
zation of small bistratified ganglion cells in primate retina. J Neurosci 319. Miyake Y, Yagasaki K, Horiguchi M, et al. Congenital stationary night blind-
Anatomy and Physiology

2007;27:13261–72. ness with negative electroretinogram. A new classification. Arch Ophthalmol


Basic Science and Translation to Therapy

310. Field GD, Greschner M, Gauthier JL, et al. High-sensitivity rod photoreceptor 1986;104:1013–20.
input to the blue-yellow color opponent pathway in macaque retina. 320. Chang B, Heckenlively JR, Bayley PR, et al. The nob2 mouse, a null mutation
Nat Neurosci 2009;12:1159–64. in Cacna1f: anatomical and functional abnormalities in the outer retina and
311. Crook JD, Davenport CM, Peterson BB, et al. Parallel ON and OFF cone their consequences on ganglion cell visual responses. Vis Neurosci 2006;23:
bipolar inputs establish spatially coextensive receptive field structure of blue- 11–24.
yellow ganglion cells in primate retina. J Neurosci 2009;29:8372–87. 321. Ishii M, Morigiwa K, Takao M, et al. Ectopic synaptic ribbons in dendrites
312. Packer OS, Verweij J, Li PH, et al. Blue-yellow opponency in primate S cone of mouse retinal ON- and OFF-bipolar cells. Cell Tissue Res 2009;338:
photoreceptors. J Neurosci 2010;30:568–72. 355–75.
Anatomy and Physiology Section 1
For additional online content visit http://www.expertconsult.com

Cell Biology of the Retinal Pigment Epithelium Chapter

Gabriele Thumann, Guorui Dou, Yusheng Wang, David R. Hinton


16 
The retinal pigment epithelium (RPE) forms a monolayer of of the photoreceptors and choroid. A role for RPE in the induc-
highly specialized neuroectodermally derived pigmented cells tion and modulation of scleral development has also been
located between the neurosensory retina and the vascular described.12 Readers are referred to Chapter 13, Development of
choroid. The RPE mediates functions essential for normal outer the retina, for a more comprehensive review of the development
retinal physiology, including participation in the visual cycle, of the RPE and retina.
phagocytosis of shed photoreceptor outer segments (OS), main-
tenance of the outer blood–retinal barrier, secretion of neuro- ANATOMY AND HISTOLOGY
trophic, inflammatory, and vasculotrophic growth factors, water
There are approximately 3.5 × 106 RPE in the adult human eye,
transport out of the subretinal space, and regulation of bidirec-
and this population remains relatively stable during young adult
tional ion and metabolic transport between the retina and
life in the absence of diseases in which RPE are induced to
choroid. The RPE is a primary site of pathology in major causes
undergo proliferation or cell death.13 Studies evaluating cell
of blindness, including age-related macular degeneration (AMD)
cycle and cell proliferation markers show that a very low level
and proliferative vitreoretinopathy (PVR).
of mature RPE are retained in the cell cycle and proliferate in
vivo in both rats and an aged human sample.14 The RPE layer
EMBRYOLOGY
extends from the optic nerve to the ora serrata and continues as
At day 27 postconception, the optic vesicles invaginate to form the pigment epithelium of the ciliary body. The foveal area con-
the optic cup and the neuroepithelium undergoes early differ- tains the highest density of RPE cells, while the cell density
entiation and thickening. By day 30, the basic cellular relation- exhibits in-gradient decrease to the periphery. These cobblestone-
ship of the outer retina is established with the prospective RPE like cells with highly polarized structure form a monolayer
layer found closely apposed to the prospective neural retina. between the neurosensory retina and the choroid. The apical
Several transcription factors have been found to play a critical microvilli of the RPE cells interdigitate with the OS of the pho-
role in the determination and specification of RPE, among which toreceptors, while the RPE basal side is attached firmly to the
are microphthalmia-associated transcription factor (Mitf), ortho- underlying Bruch’s membrane; the basement membrane of the
denticle homeobox (Otx)1/2 and paired box (Pax) 6.1 The Sonic RPE represents the innermost layer of Bruch’s membrane.
hedgehog (Shh) signaling pathway is also critical; the distinct Readers are referred to Chapter 20, Structure and function of
expression pattern of its members in central and peripheral RPE Bruch’s membrane, for a detailed description of Bruch’s mem-
suggests multiple regulatory roles in the process of RPE differ- brane physiology and interaction with the RPE.
entiation.2 By day 35, melanin pigment granules are identified The brown color of the RPE is imparted by its melanin gran-
in the presumptive RPE; this is the earliest site of pigmentation ules and the typical patterned appearance of the fundus results
in the body. By the 6th week of gestation, RPE cells elaborate from variations in the pigmentation of the RPE layer. In a healthy
basement membrane material that participates in the formation retina, the highest concentration of pigment is found in the
of the first recognizable Bruch’s membrane. Between 4 weeks peripheral retina, the lowest in the macular area.15 With age, RPE
and 6 months of gestation, RPE cells exhibit a high rate of pro- gradually lose melanin granules, possibly related to effects of
liferation that peaks at 4 months of gestation.3 Meanwhile, most photooxidation.16 The retina and the RPE are separated by a
of the RPE cells have developed polarized characteristics with potential space known as the subretinal space. Although the
apical microvillous projections that extend into the subretinal retina conforms to the shape of the adjacent pigment epithelium
space.4 The RPE’s consequent interaction with neurosensory and underlying sclera, it is not firmly attached to the pigment
retina coordinates the differentiation of primordial photorecep- epithelium except at the optic disc and ora serrata; attachments
tors. While RPE fate may be reversible for several days following elsewhere are weak and can be disrupted by relatively weak
the initial activation of differentiation,5 recent studies suggest forces. Readers are referred to Chapter 19, Retinal adhesion and
that, the Shh pathway,6 retinoic acid,7 bone morphogenetic fluid physiology of the subretinal space, for a complete discus-
protein (BMP) signaling,8 Notch9 and Wnt/β-catenin10 pathways sion of retinal adhesion.
all play a role in the maintenance of RPE differentiation. In addi-
tion, ezrin, a member of the ezrin/radixin/moesin (ERM) family, Heterogeneity and polarity of the RPE
is important for morphogenesis of apical microvilli and basal RPE cells exhibit regional heterogeneity in morphology and
infoldings in RPE cells.11 Polarization of the differentiated RPE function.17 The cuboidal RPE cells that form the RPE monolayer
monolayer is required for the normal development and function appear polygonal in shape when viewed en face, a pattern that
is maintained during early passage in culture when grown to requirements for RPE function, RPE cells vary regionally in
confluence (Fig. 16.1). In situ, cell shape varies throughout the growth potential,18 level of expression of vimentin and phospho-
402 fundus; in the macular area the cells are tall and narrow, whereas tyrosine,17 distribution of Na/K adenosine triphosphatase
in the periphery they are flatter, more spread out, and may (ATPase) pumps,19 and kinetics of rod outer-segment binding
be binucleated.13 Perhaps due to regional differences in and ingestion.20 Regionally heterogeneous age-related altera-
tions in expression of lysosomal enzymes, Mn-superoxide
Section 1

dismutase, and accumulation of lipofuscin have been identified


in RPE,21–23 suggesting that this heterogeneity is probably
involved in pathological changes in RPE. Quantification of RPE
phenotypic variation can be achieved using laser scanning
cytometry.22
The polarity of RPE in the monolayer is characterized by dis-
Anatomy and Physiology
Basic Science and Translation to Therapy

tinct ultrastructural features and specialized functions in the


apical and basolateral domains (Fig. 16.2). The apical cell mem-
brane elaborates numerous microvilli (3–7 µm in length) that
interdigitate with and ensheath the OS of the retinal photorecep-
tors.24 (Figs 16.3 and 16.4). These interdigitations, in conjunction
with the extracellular matrix (ECM), and neural cell adhesion
molecule (N-CAM) expressed on the RPE apical surface, allow
some degree of adhesion between the retina and the RPE.
Between 30 and 45 photoreceptors are in contact with each RPE
cell.25 RPE cells engulf and degrade shed rod OS each day such
Fig. 16.1 Cultured human retinal pigment epithelium (RPE) cells.
A monolayer of early-passage human RPE cells demonstrates the that one human RPE cell will ingest and degrade hundreds of
polygonal appearance of the cells when grown to confluence (phase millions of discs during its lifetime.26
microscopy, ×25). The functional polarity of RPE cells is expressed in the dif-
ferential distribution of membrane proteins along the apical–
basal axis (Table 16.1). Sharing many common characteristics
with other transporting epithelia, RPE exhibit “reversed polar-
ity” of certain proteins, such as the Na/K-ATPase pump, ECM
metalloproteinase inducer (EMMPRIN), and N-CAM. In RPE
these proteins are found at the apical surface, rather than
OS at the basolateral surface where they are found in other epi-
thelia.27 In a proteomic study of RPE apical microvilla,27 283
proteins were identified, which could be divided into different
functional categories, including retinoid-metabolizing, cytoskel-
mv etal, enzymes, ECM components, membrane proteins, and

OS

N
OS

mit

bm 0.3
m
Fig. 16.2 Electron micrograph of a retinal pigment epithelium (RPE)
cell and adjacent outer segments (OS) of the rod photoreceptor cells. Fig. 16.3 Scanning electron micrograph of rod photoreceptor outer
Note the apical microvilli (mv) of the RPE cell. Darkly stained segments (OS) fractured across their long axis, surrounded by apical
melanin granules are seen apically, while mitochondria (mit) are folds extending from the surface of the retinal pigment epithelium. The
predominantly basal. The cell rests on the basal Bruch’s membrane membrane forms a sheath around each of the rod OS (scale bar =
(bm). The nucleus is present in the basal third of the cytoplasm. 0.3 µm). (Reproduced with permission from Hollenberg MJ, Lea PJ.
A phagolysosome (arrowhead) containing degrading rod OS material High resolution scanning electron microscopy of the retinal pigment
is seen in the cytoplasm at low (×21 500) and high (inset, ×50 000) epithelium and Bruch’s layer. Invest Ophthalmol Vis Sci
magnification. 1988;29:1380–9.)
transporters. αvβ5 integrin (Fig. 16.5), mannose receptors, and
CD36 are localized to the apical membrane domain, specifically
in the microvilli, where they play critical roles in rod outer- 403
segment phagocytosis.28–30 Na/K-ATPase is mainly expressed
at the apical RPE cell membrane where it is critical for tran-

Chapter 16
sepithelial ion transport in the RPE.31 A newly identified mem-
brane protein chloride intracellular channel 4 (CLIC4) has been
shown to be enriched in apical RPE microvilli, possibly for
modulating the activity of cell surface channels/transporters.32
The RPE basal membrane domain is characterized by infoldings
that are approximately 1 µm in length (Fig. 16.6). The basal
membrane expresses α3β1, α6β1, and αvβ3 integrins and medi-

Cell Biology of the Retinal Pigment Epithelium


ates attachment to Bruch’s membrane (Fig. 16.5). The basal
infolding also express Bestrophin-1, which is a chloride anion
channel that regulates voltage-dependent L-type Ca2+ channels
5 by interacting with the beta subunits of the Ca2+ channels.33
m
Ezrin is found in both apical and basolateral membranes where
Fig. 16.4 Scanning electron micrograph of the apical surface of a it promotes morphogenesis of apical microvilli and basal infold-
bovine retinal pigment epithelium (RPE) explant. Note the abundant ings.34 A number of transporters work together in apical and
microvilli on the apical surface of the RPE cells. Approximately a basal domains. For example, glucose transport from the choroid
dozen RPE are shown. Bar = 5 µm.
to the photoreceptors is mediated by glucose transporter (GLUT)
1 located both basally and apically.35 In contrast, elimination of
lactic acid from the subretinal space is mediated by different
Table 16.1 Polarized cell surface molecule expression in retinal monocarboxylate transporters in the apical (MCT1) and baso-
pigment epithelium lateral (MCT3) membranes.36,37 The lateral membrane domain
of the RPE cell demonstrates specialized junctions important
Location Protein Function
for cell–cell attachment and communication, which will be
Apical Na/K-ATPase Na flux discussed below.
membrane The intracellular distribution of cell organelles in RPE also
N-CAM Adhesion to retina, exhibits heterogeneity and polarization (Figs 16.2 and 16.6).38
phagocytosis Melanin granules, which are ovoid or spherical in shape, 2–3 µm
αvβ5 Integrin Phagocytosis
in length, and 1 µm in diameter, are localized at the apical region
of the cell, as is the endoplasmic reticulum, the site of protein
CD36 Phagocytosis synthesis. At the ora serrata, the RPE contain many dense round
melanin granules throughout the cell; at the equator and macula,
Ezrin Apical microvilli melanin granules are more apical, less frequent, and often elon-
CLIC4 Channels/
gated. The nucleus, with a diameter of 8–12 µm, is located in the
transporters basal part of the cell. The localization of mitochondria in the
basal half of the RPE results from the oxygen pressure which is
GLUT1 Glucose transport highest in this cell region and is most apparent at the macula.39
Other cytoplasmic elements, such as microperoxisomes, lyso-
MCT1 Monocarboxylate
transporter somes, and phagosomes, do not appear to have a distinct distri-
bution. However, inhibition of lysosomal degradation in RPE
Lateral Occludin Tight junction cells could induce exocytosis of phagocytic residual material at
membrane the basolateral plasma membrane.40
Cadherin Adherens junction

Connexin Gap junction


Cellular junctions
The outer blood–retinal barrier is formed by the RPE monolayer,
Basolateral α3β1, α6β1, ανβ3 integrin Attachment to in which the lateral domains of adjacent RPE cells connected
membrane Bruch’s membrane by apical zonulae occludens (tight junctions) and adjacent
Ezrin Basal infoldings zonulae adherentes (adherens junctions) enable the epithelium
to form a barrier by joining neighboring cells together and
GLUT1 Glucose transport regulating transepithelial diffusion through the paracellular
spaces (Fig. 16.6).41 These junctions seal off the subretinal space
MCT3 Monocarboxylate where the exchange of macromolecules with the choriocapillaris
transporter
takes place, and form the so-called Verhoeff’s membrane. Two
Bestrophin-1 Chloride anion major groups of proteins of zonulae occludens are the claudins
channel and the occludins. The interaction between the extracellular
domains of adjacent occludin molecules leads to high transepi-
Na/K-ATPase, Na/K adenosine triphosphatase; N-CAM, neural cell
adhesion molecule; CLIC4, chloride intracellular channel 4; GLUT1, thelial resistance and an intact blood–retinal barrier. They are
glucose transporter 1; MCT1, monocarboxylate transporter 1. responsible for maintaining the polarity of cells by preventing
Outer segments
404 RPE

Bruch's membrane
Section 1

Choroid

x 1 v x
Anatomy and Physiology
Basic Science and Translation to Therapy

V Control

5 3

Fig. 16.5 Subcellular distribution of integrin receptors in adult rat retinal pigment epithelium by indirect immunofluorescence. For reference,
a methylene blue-stained section of intact retina is shown at the top. β1 Integrins are exclusively basal. αv Integrin receptors are apical
and basolateral and stain cytoplasmic vesicles. β5 Integrins are located apically, and β3 integrins are restricted to a basal location.
Bars = 5 µm. (Reproduced with permission from Finnemann SC, Bonilha VL, Mamorstein AD, et al. Phagocytosis of rod outer segments by
retinal pigment epithelial cells requires αvβ5 integrin for binding but not for internalization. Proc Natl Acad Sci U S A 1997;94:12932–7.)

mv the lateral diffusion of integral membrane proteins between the


mv
apical and lateral/basal surfaces, allowing the specialized func-
tions (such as molecule transport ) of each to be preserved.42
af The cytoplasmic domain of occludin interacts with several other
proteins, including zonula occludens (ZO)-1 and ZO-2, to form
jc
a complex that interacts with the actin cytoskeleton and com-
ponents of various signal transduction pathways (Fig. 16.7).
ZO-1 regulates cell proliferation and gene expression by inhibit-
ing the activity of the Y-box transcription factor ZONAB in
cultured epithelial cells, indicating they are critical for the dif-
ferentiation and homeostasis of the RPE monolayer.43 Early
pg expression of the assembly proteins, including junctional adhe-
sion molecule-A (JAM-A), AF-6, PAR-3, and PAR-6, corresponds
to the initial establishment of the adherens and tight junctions,44
whereas diffusible factors secreted by the neural retina act syn-
ergistically with basolateral stimulation to regulate the structure
and function of RPE tight junctions.45 Claudin 19 has been shown
to be dominantly expressed in human fetal RPE cells.46 JAM-C
localizes specifically in the tight junctions of human fetal RPE
bi
and adult native RPE, and functions to regulate the recruitment
bm
of N-cadherin and ZO-1 to the cell–cell contacts.47 In pathologi-
cal conditions, oxidative stress,48 inhibition of Na/K-ATPase,49
matrix metalloproteinase (MMP)-9, interferon (IFN)-γ, tumor
Fig 16.6 Transmission electron micrograph of intact bovine retinal necrosis factor,50,51 and amyloid-beta(1–42)52 could decrease tight
pigment epithelium (RPE) cells. Note the apical microvilli (mv), the junction protein expression, resulting in the disruption of the
pigment granules (pg) in the apical region of the RPE cytoplasm, and
the basal infoldings (bi) in contact with Bruch’s membrane (bm). In the integrity of the outer blood–retina barrier; in contrast, nitric
upper left portion of the figure, the apically located junctional complex oxide is involved in the maintenance of blood–retina barrier
is seen between two retinal pigment epithelia, including the more integrity.51
apical zonula occludens (tight junction) and adjacent zonula adherens. The zonulae adherentes (adherens junction) form a junction
The junctional complex (jc) is shown at higher magnification in the
inset. The jc is associated with a cytoplasmic band of actin filaments with a separation of 200 Å and are associated with circumferen-
(af). Bar = 1.8 µm (bar in inset = 1 µm). tial microfilament bundles.53–55 The transmembrane cadherin
Microfilaments and microtubules are dynamic structures that
undergo polymerization and depolymerization and are critical
for intracellular transport. Microtubules play a role in mitosis, 405
and the movement of subcellular organelles and pigment gran-
ules. Actin microfilaments are located in the microvilli and

Chapter 16
throughout the cytoplasm where they are arranged in loose
arrays or bundles. They play an important role in the generation
and maintenance of cellular shape and cell migration.65 In fish
RPE, actin is involved in melanosome transport within apical
projections.66 Myosins are cytoskeletal motors critical for gener-
ating the forces necessary for establishing cell structure and
mediating actin-dependent cell motility. The expression of mul-

Cell Biology of the Retinal Pigment Epithelium


tiple myosin family members has been shown in fish RPE.67 In
addition, myosins are proposed to play a role in the movement
and biogenesis of the melanosomes in RPE.68 Intermediate fila-
ments provide a structural framework for the cell linking the
nucleus with the cell membrane and to microfilaments and
microtubules. In human RPE cells, intermediate filaments of
type I (acidic keratins), type II (basic/neutral keratins), and type
V (lamins) have been identified. In RPE, the pattern of cytokera-
tin filament expression varies with cellular differentiation,
Fig 16.7 Immunofluorescent stain of a bovine retinal pigment
epithelium (RPE) monolayer explant stained with an antibody against changes in cellular polarity, intercellular association, and condi-
the ZO-1 protein, a protein associated with the zonula occludens, or tions of cell culture.69,70 Human RPE cells in vivo express keratins
tight junction, demonstrating the polygonal shape and tight adherence 8 and 18.71 Under conditions such as cell culture, keratins 7 and
of the RPE (×440).
19 are also coexpressed. The presence of keratin 18/19 has been
correlated with migration in cultured RPE cells.71
molecules of the adherens junction require calcium for ensuring
that cells within tissues are bound together. Their cytoplasmic
domains interact with catenins, which in turn form a complex
Role of RPE in Bruch’s membrane
with proteins such as α-actinin and vinculin. The adherens junc- synthesis and remodeling
tions play a role in maintenance of the polygonal shape of the The basal surface of the RPE monolayer has elaborate basal
RPE cell and in the organization of the actin cytoskeleton.56 Gap infoldings that attach to Bruch’s basement membrane, an acel-
junctions, which are also present in the lateral cell membranes, lular layer separating the RPE from the choriocapillaris.72 Bruch’s
are associated with the expression of connexin and are important membrane is a complex structure that not only serves as the
for the exchange of ions and metabolites between cells. The gap attachment site for the RPE cells, but also serves as a selective
junction protein connexin 43-mediated communication between conduit for nutrients transported to the retina from the choroidal
the retina and RPE is essential for the correct pacing of retinal vasculature and for metabolic wastes transported from the retina
organogenesis.57 In addition, ATP released from RPE hemichan- to the circulation. Bruch’s membrane consists of five distinct
nels via connexin 43 speeds both cell division and proliferation layers.73 From the RPE to the choroid the following layers can be
in the neural retina.58 The basal cell membrane expresses various distinguished: the basement membrane of the RPE, the inner
integrins and shows focal adhesion points with the ECM.59 The collagenous layer, the elastic layer, the outer collagenous layer,
presence of desmosomes between RPE cells varies among and the choriocapillaris endothelium basement membrane. In
species; they are often difficult to detect, and they do not appear normal mice the RPE basement membrane and the choroid base-
to be necessary for establishment of a polar, functioning RPE ment membrane are synthesized first, followed by the deposi-
layer.60 Occasional desmosomal–mitochondrial complexes have tion of a collagenous layer between the two basement membranes.
been described between RPE cells in human specimens, similar Later the elastic layer is synthesized and deposited within the
in appearance to those observed in nonpigmented ciliary epithe- collagenous layer, eventually separating it into an inner and
lia.61 While the desmosome-associated protein desmoplakin is outer collagenous layer.74
found in cultured bovine RPE, it has not been identified in The basement membrane of the RPE and choroid (1.4–1.5  µm
human RPE cultures. thick) are similar to other basement membranes in composi-
tion; they contain collagens, laminin, fibronectin, and sulfated
Cytoskeleton polysaccharides75–77 and serve the same functions, including
Apart from the general role of the cytoskeleton in all nucleated anchoring subjacent cells, acting as a barrier and a filter, and
cells, the RPE cytoskeleton is highly associated with its distinct stabilizing the structure of the tissue.78 The choriocapillaris
functions such as melanosome transport and phagocytosis.62,63 basement membrane also contains collagen type VI, which is
These functions are disrupted in some diseases like PVR, caused not present in the RPE basement membrane, and may be
by RPE cell proliferation and myofibroblastic transdifferentia- involved in capillary endothelial cell stabilization. The inner
tion, where there are prominent rearrangements of the cytoskel- collagenous (1.4  µm thick) and outer collagenous (0.7  µm thick)
eton.64 The cytoskeleton is composed of three major elements: layers are composed of collagens type I, II, and V organized
the actin microfilaments (diameter 7 nm), microtubules (diam- in a lattice-like network embedded in an amorphous collection
eter 25 nm), and intermediate filaments (diameter 10 nm). of glycosaminoglycans.79
Distributed throughout all the layers of Bruch’s membrane is potential to researchers working in the field of tissue engineering
collagen XVIII,80 which gives rise to the endostatin, an inhibitor and cell transplantation. Culture of well-differentiated and char-
406 of choroidal neovascularization.81 Extensive gene expression acterized RPE cells from various sources may be a future cellular
studies by Booij et al.82,83 have shown that both the RPE and therapy for several retinal diseases.98 Methods to generate RPE
choroid express most of the genes necessary for the formation with varying degrees of polarization have been reported for
and maintenance of Bruch’s membrane. Since both the RPE and immortalized human RPE cell lines, such as ARPE19 and D407,
Section 1

choroid are able to synthesize the major components of Bruch’s and fetal human RPE99,100 (reviewed by Sonoda et al.101 and
membrane, it appears that the basal lamina of each RPE would Maminishkis and Miller102). Polarized RPE monolayers derived
be maintained by RPE cells, the choroid basement membrane by from human fetal RPE develop high transepithelial resistance,
choroidal cells, whereas the inner, outer collagenous layer and and show polarized membrane specialization, including promi-
the elastic layer would be maintained cooperatively by both nent apical microvilli and apical expression of Na/Ka-ATPase.101
tissues. Age-related biochemical alterations in the composition More recently, cells from the ciliary margin progenitor zone,103
Anatomy and Physiology
Basic Science and Translation to Therapy

and three-dimensional organization of ECM molecules in Bruch’s retinal stem cells,104 human embryonic stem cells,105,106 and
membrane may affect this function and are described in detail human induced pluripotent stem cells107 have also been shown
in Chapter 20, Structure and function of Bruch’s membrane. to have the potential to be directed into RPE cells. The reader is
The apical domain of the RPE cells is embedded in the inter- referred to Chapters 35, Stem cells and cellular therapy, and 125,
photoreceptor matrix (IPM), which is produced by the RPE and Transplantation frontiers, for an indepth discussion of RPE
the inner segments of the photoreceptors. Major protein compo- cellular therapies.
nents of IPM that are involved in retinoid transport between the
photoreceptors and the RPE include the interphotoreceptor- SPECIALIZED FUNCTIONS OF THE RPE
binding protein (IRBP), retinol-binding protein (RBP), and trans-
thyretin (TTR).84–87 Retinal adhesion to the RPE is mediated in Absorption of light
part by RPE transport of water and ions from the IPM toward The first step in the visual process is absorption of light by the
the choriocapillaris. It has been identified that RPE predomi- opsins of rod and cone photoreceptors and by the melanopsin
nantly secrete the neurotrophic pigment epithelial-derived factor of retina ganglion cells. Melanopsin is involved in the regulation
(PEDF) from the apical side into the IPM, which is about 1000- of circadian rhythms, pupillary light reflex, and other nonvisual
fold greater than that for vascular endothelial growth factor responses to light,108 although recently it has been shown that
(VEGF), which is mainly from the basolateral side.88 In addition, melanopsin in ganglion cells may contribute directly to pattern
proteomic analysis of the porcine IPM indicated that a set of vision.109 The light-absorbing chromophore of opsins is 11-cis
potentially neuroprotective proteins could be extracted, includ- retinaldehyde (11-cis-RAL), which is delivered to the rod photo-
ing phosphoprotein enriched in astrocytes (PEA)-15, peroxi­ receptors by the RPE cells. The RPE cells possess the enzymatic
redoxin 5, αB crystallin, macrophage migration inhibitory factor, mechanism to convert vitamin A to 11-cis-RAL as well as the
78-kDa glucose-regulated protein (GRP78), protein disulfide mechanism to deliver it to the photoreceptors. An important
isomerase (PDI), and PEP-19.89 Notably, αB crystallin has been function of the RPE is the absorption of light that passes beyond
shown to be secreted by RPE and plays an important role in the OS of the photoreceptors. The melanin pigment within the
maintaining and facilitating a neuroprotective outer retinal pigment granules (Fig. 16.6) of the RPE absorbs stray photons of
microenvironment.90 light which minimizes light scatter within the retina and protects
Degradation of ECM is regulated, in part, by both MMPs and from excess light.15
the urokinase-type plasminogen activator (uPA) cascade,91
which are activated by multiple types of cells including leuko- Phagocytosis of rod outer segments
cytes, endothelial cells, and RPE. MMPs are a family of zinc- The RPE is a monolayer of cells that apically extends microvilli
binding, Ca+-dependent endopeptidases that can degrade both that interdigitate with the photoreceptor OS. This arrangement
collagen and proteoglycans in ECM during physiological and is critical to vision since the renewal of the phototransduction
pathological ECM remodeling. The MMPs are tightly regulated machinery and maintenance of a constant OS length require the
by its tissue inhibitors of metalloproteinases (TIMPs), whose shedding of apical stacks of discs, which are ingested and
impairment may lead to progressive alterations in Bruch’s mem- degraded by RPE cells.
brane.92 Normal RPE express the membrane-bound type 1 (MT1 In all vertebrates examined, nocturnal and diurnal, cold-
MMP) as well as type 2 (MMP-2) metalloproteinase,93 as well as blooded and warm-blooded, rod OS shedding occurs maximally
the metalloproteinase inhibitors TIMP-1 and TIMP-3.94 Degrada- at or shortly after light onset, a daily rhythm which is established
tion of the ECM is also promoted by uPA, a serine protease that early after birth, irrespective of the lighting conditions during
catalyzes conversion of inactive plasminogen to active plasmin.95 development. In fact, in rats the daily rod OS shedding rhythm
Plasmin is a broad-spectrum protease that degrades fibrin, fibro- is established by 2 weeks postnatally and it is maintained even
nectin, laminin, and other ECM components. uPA activity can after optic nerve section, indicating that this rhythmic process
be blocked by two endogenous inhibitors, plasminogen activator of OS shedding is controlled by intrinsic signals within the
inhibitor-1 and -2 (PAI-1 and PAI-2),96 which belong to the serpin eye.110,111The temporal pattern of cone OS shedding is much more
protease superfamily. Cultured human RPE cells produce im- variable since in some species it occurs at night,112,113 whereas in
munoreactive uPA and a protein similar to PAI-1.97 others cones and rod OS are shed just after light onset.114,115
The disposal of the continuously shed OS is accomplished by
Cell culture models of RPE phagocytosis of the OS at the expense of elevated metabolic
Cell culture models of RPE play a critical role in gaining basic activity and energy expenditure.116,117 It has been estimated that,
knowledge about native tissue, evaluating polarized RPE func- during an 80-year span, one RPE cell has internalized and
tion, facilitating drug discovery and toxicity, and offering degraded about 200 million discs.118 The phagocytosis of
on their phagocytic targets. A number of soluble molecules have
been shown to be possible bridging elements, such as Gas6,
RPE protein S, and milk fat globule epidermal growth factor (MFG)- 407
E8.121 Recently Tubby and tubby-like protein (Tulp) 1, which are
secreted by photoreceptors, have been identified as playing a

Chapter 16
OS role in RPE phagocytosis. Tubby and Tulp1 are bridging mole-
cules that bind to MerTk (a member of the TAM receptor tyro-
sine kinase subfamily) at their N-terminal region and likely to
the rod OS at their C-terminal region.122
Binding of the rod OS is followed by invagination of the
IS plasma membrane around the OS fragment, leading to its inges-
tion into a phagosome. A reorganization of cytoskeletal ele-

Cell Biology of the Retinal Pigment Epithelium


ments, particularly of microfilaments in the microvilli, appears
to be involved in the initial stages of ingestion. An actin feltwork
forms at the sites of attachment that extend into the pseudopods
which surround and engulf the fragments to form the phago-
some.123 This process includes an undefined, presumably
G-protein-coupled transmembrane signal that stimulates the
assembly of an appropriate contractile apparatus that, in turn,
provides the motive force for internalization of the attached
particle.124 Internalization appears to be mediated by the recep-
tor tyrosine kinase, c-mer, and its ligand Gas6.125 Once inside the
ONL cytoplasm, phagosomes are transported basally. In the presence
of colchicine, the phagosomes remain in the apical domain of the
RPE, suggesting that microtubules are involved.126,127 Transport
may also be partially mediated by myosin VIIa, an unconven-
tional myosin that is mutated in patients with Usher syndrome.128
Cytokines, which play an important role in development, dif-
ferentiation, and survival of retinal cells, are also involved in the
phagocytic process. In vitro studies of RPE cells from normal rats
and rats with defective phagocytosis have shown that transform-
ing growth factor (TGF)-β1 and basic fibroblast growth factor
10 mm (FGF) regulate rod OS phagocytosis.129
Basally transported phagosomes then fuse with lysosomes
Fig. 16.8 Phagocytosis of rod outer segments (OS). A section of
and are degraded. The lysosome–phagosome interaction occurs
outer retina was obtained from an eye enucleated 2 hours after light in two steps. First, smaller lysosomes fuse with phagosomes.
exposure in a pigmented rat. The section was evaluated using Subsequently, larger lysosomes appear to interact with phago-
confocal microscopy after fluorescent labeling for rhodopsin (red), somes via pore-like structures. Following fusion, lysosomal
cyoplasm (green), and nuclei (blue). The figure shows the rhodopsin+
OS and punctate labeling of rhodopsin in the phagolysosomes within enzymes hydrolyze the sequestered OS into small molecules that
the retinal pigment epithelium (RPE: arrows); shown at higher diffuse out of the RPE cell or are reused within the cell. Of the
magnification in insert. ONL, outer nuclear layer; IS, inner segments wide array of lysosomal enzymes capable to hydrolyze photo­
of photoreceptors. Scale bar = 10 µm.
receptor OS, cathepsin D is the most important and plays a
dominant role in the degradation of rhodopsin, the major glyco-
sylated protein present in the OS.130–132 Another enzyme involved
ROS disc material can be visualized in normal rat eyes using in degradation of rod OS is the cysteine protease, cathepsin S.133
confocal microscopy to visualize rhodopsin+ phagolysosomes With age and pathologic changes degradation of OS materials
by enucleating the eye 2 hours after onset of light (Fig. 16.8). The within the phagolysosomes leads to the formation of lipofuscin
mechanisms involved in rod OS phagocytosis have recently granules composed of rod OS residue.134,135
been reviewed.119 Rod OS phagocytosis is a highly specialized
receptor-mediated, multistep process that comprises recogni- Role in visual cycle
tion, attachment (receptor–ligand interactions), internalization Light perception in the retina is initiated by the reaction of
(transmembrane signaling and contractile proteins), and degra- photons with light-sensitive pigments that are part of the mem-
dation of the ingested OS.119 The first step in the phagocytic branes of photoreceptor OS. Cooperation between the photo-
pathway is recognition. It appears the recognition is a receptor- receptors and the RPE cells allows these pigments to be recycled
mediated event that requires soluble molecules to bridge the through a complex series of oxidation–reduction reactions and
receptor on the RPE cell and some functional molecules on the transport mechanisms that are referred as the “visual cycle”
rod OS. Studies have shown that the receptor on the RPE cell is (Fig. 16.9).
αvβ5 and the molecule on the rod OS is phosphatidylserine, The visual cycle is initiated by a photon reacting with rhodop-
which has been described on the OS plasma membranes.120 sin, which comprises a G-coupled receptor protein, opsin (rods)
Although αvβ3 and αvβ5 have been demonstrated to be instru- or cone-opsin (cones), and the chromophore 11-cis-RAL.
mental for substrate binding, they do not bind substrate directly. Reaction with light changes 11-cis-RAL into all-trans-RAL, which
Rather they bind an opsonin that recognizes the “eat-me” signal is released from the opsin and reduced to all-trans-retinol
Fig. 16.9 The visual cycle takes place in
Photoreceptor RPE cell the photoreceptors (left), the retinal pigment
Rho–Lys epithelium (right), and the intervening
408 11-cis-RAL 11-cis-RAL-CRALBP
interphotoreceptor matrix. See text (visual
Photon 11-cis-RAL cycle) for description of components of
IRBP the cycle. RAL, retinal; RE, retinyl
NADH
11-cis-RAL 11-cis-RDH esters; ROL, retinol; CRALBP, cellular
Meta-Rho II
Section 1

+ 11-cis-RAL NAD retinaldehyde-binding protein; LRAT, lecithin


Apo-Rho + 11-cis-ROL-CRALBP retinol acyltransferase; RDH, retinol
IRBP dehydrogenase; Rho, rhodopsin; IRBP,
All-trans-RAL + Apo-Rho interphotoreceptor-binding protein.
NADPH RPE65
All-trans-ROL
NADP All-trans-RDH
All-trans-RE-CRALBP
IRBP
Anatomy and Physiology
Basic Science and Translation to Therapy

All-trans-ROL (Vit A) LRAT

Interphotoreceptor Matrix

(all-trans-ROL). ABCA4 (a member of the A subfamily of ATP- FGF1 and FGF2, which act as survival factors by preventing
binding cassette proteins) supports the processing of highly reac- apoptosis and retinal degeneration.148,149
tive all-trans-RAL and prevents the buildup of A2E, an insoluble A variety of antioxidants and their enzymes play a critical role
toxic compound that is shed from the photoreceptors, phagocy- in cell protection and are abundantly expressed in RPE. The
tosed by RPE, and accumulates in lipofuscin.136 All-trans-ROL major ones among them are glutathione (GSH),150 thioredoxins
passes into the IPM, where it binds to the IRBP and is transported (Trx), and glutaredoxins (Grx),151,152 methionine sulfoxide reduc-
into the adjacent RPE cells. IRBP is a 140-kDa glycoprotein tases (Msrs),153 catalase,154 superoxide dismutases,155 and gluta-
secreted by the photoreceptors, which enhances the translocation thione peroxidase.156 The antioxidants as well as the enzymes
of 11-cis-RAL from the RPE to the photoreceptors137 and the trans- have been shown to be either upregulated or downregulated
location of all-trans-ROL from the photoreceptors to the RPE.138 depending on the severity of oxidative stress stimuli,154 which
IRBP is essential to the normal functioning of the visual cycle. In can include H2O2, 4-hydroxy-2-nonenal, hypoxia/hyperoxia,
fact, in knockout mice (irbp−/−) that lack IRBP there is significant inflammation, and toxic drugs, as well as lipid signaling agents
photoreceptor degeneration.139 such as ceramide.157
In the RPE cells all-trans-ROL is esterified to fatty acids through Depletion of GSH causes apoptosis in RPE cells with an
the action of lecithin retinol acyltransferase (LRAT) to yield all- accompanying compensatory upregulation of glutathione
trans-retinyl esters (all-trans-RE), which are the substrates for S-transferase, gamma glutamylcysteine synthetase, and glutathi-
RPE65 isomerase. RPE65 isomerase isomerizes the all-trans-RE one peroxidase-1 through a signaling pathway that involves the
to 11-cis-ROL, which is bound by cellular retinaldehyde-binding transcription factor nuclear factor-erythroid 2 p45-related factor
protein, reduced to 11-cis-RAL, and transferred to the IPM where 2 (NRF2) and phase 2 enzyme induction.158,159 Since mitochon-
it binds to IRBP and is translocated to the photoreceptors, where dria are directly involved in the generation of reactive oxygen
it binds to opsin to start the cycle anew. species , the maintenance of the mitochondrial GSH pool is criti-
cal during oxidative injury. Regulation of superoxide dismutase
Protection from oxidative stress has been shown to be effective in protection from retinal
The RPE layer resides in a perilous environment and is subjected injury.156,160 Among the Trx and Grx family, overexpression of
to oxidative stress (in part due to the continuous flux of polyun- Trx1 protected RPE cells from oxidative injury.152 The intravitre-
saturated fatty acids), to high-energy radicals (as a result of the ous injection of Trx effectively attenuated N-methyl-d-aspartate
high oxygen consumption of the retina), and to exposure to (NMDA)-induced retinal cell damage, and suppression of oxida-
radiant radiation.140 Confronted with such a risky environment, tive stress and inhibition of apoptotic signaling pathways were
RPE cells produce neurotrophin 1,141 a potent neuroprotective involved in this neuroprotection.161 Similarly, the protective
docosahexaenoic acid-derived lipid that inhibits the expression function of Grx in oxidant-induced apoptosis is also known.162,163
of proinflammatory genes and enhances the expression of In addition to the enzymes of GSH metabolism, the key reduc-
antiapoptotic molecules of the Bcl-2 family, thus promoting tases of methionine oxidation, namely Msrs, play an important
survival.142 role in RPE protection.164 Suppression of MsrA augments H2O2-
The photoreceptors and neural retina depend on the RPE to induced apoptosis while overexpression prevented cell death.153
provide metabolic balance for the maintenance of normal func- Recently, the antiapoptotic action of small heat shock proteins
tions. RPE cell degeneration leads to secondary photoreceptor in the retina has received attention.90,165–167 The antiapototic func-
degeneration and neural dysfunction. PEDF is a critical factor tion of alpha crystallins may be related to their chaperone activ-
produced by RPE that provides cytoprotection. PEDF is an ity as well as to the metabolism of GSH metabolism in RPE.168,169
endogenous regulator of proliferation, an inhibitor of neovascu-
larization, and acts to protect neural cells from various environ- Role in maintaining avascular outer retina
mental insults, such as glutamate stress, oxidative stress, and The avascularity of the subretinal space is dependent on the
ischemic conditions.143–147 RPE cells also synthesize and secrete antiangiogenic activity of PEDF, a factor that is expressed by
many cells, including RPE cells, and the antiangiogenic activity waste products from the retina and photoreceptors to the circu-
of endostatin, the 20-kDa C-terminal fragment derived from type lation. Since the RPE cells form a tight barrier to diffusion, nutri-
XVIII collagen by proteolysis. PEDF is synthesized and secreted ents are taken up via active transport mechanisms that are 409
by RPE cells into the IPM,143,144 where it acts as a neuroprotective specific to each nutrient and ion.
factor for photoreceptors and neural retina ganglion cells. PEDF Of the nutrients that RPE cells must deliver, glucose is one of

Chapter 16
is also a most potent and selective antiangiogenic factor that the most important. RPE cells express very high levels of the
inhibits the growth and mediates regression of newly formed glucose transporters, GLUT1 and GLUT3.180,181 GLUT1 is respon-
blood vessels without affecting pre-existing vessels and in the sible for transport of glucose that is dependent on metabolic
retina prevents the growth of blood vessels into the subretinal demand and its expression is regulated by the level of glucose:
space.170–172 The importance of PEDF in maintaining the avascu- reduced glucose levels increase expression whereas increased
larity of the outer retina is apparent from the distribution of glucose levels decrease expression.182 GLUT3 is a high-affinity
PEDF in the retina. The expression of PEDF is highest in the glucose transporter that is responsible for the basal transport

Cell Biology of the Retinal Pigment Epithelium


developing fovea of midgestation human retinas and in the of glucose to maintain resting-level activity. It is interesting that
fovea of monkeys aged between fetal day 55 and 11 years of GLUT1 also mediates the uptake of vitamin C. In RPE cells this
age.173 In addition, in normal adult eyes PEDF concentration is may be important since the high energy requirements of the
10-fold higher than VEGF in the macular region but not in the retina will produce high-energy radicals whose harmful effects
peripheral region of the retina, strongly suggesting that PEDF is may be moderated by the high levels of vitamin C.183
responsible for macular avascularity.174 Of the many substances that are essential to vision, vitamin A
In addition to PEDF, avascularity of the subretinal space is occupies a prominent place. Vitamin A is transported from the
dependent on endostatin. Endostatin is cleaved from collagen blood to the RPE bound to a complex of RBP and TTR. Upon
XVIII in Bruch’s membrane by the action of RPE-derived prot­ reaching the basal membrane of RPE cells, retinol is released to
eases, such as MMP-9 and cathepsin L. Laser-induced choroidal the membrane retinol receptor STRA6 (stimulated by retinoic
neovascular lesions in Col18a1–/– mice, which lack endostatin, acid 6), which transfers vitamin A into the cells, where it is con-
formed large confluent areas of neovascularization and led to verted to the active chromophore 11-cis-retinal.184
increased vascular permeability; in contrast, choroidal neovas- Ions are transported in and out of RPE cells via selective chan-
cular lesions in control mice remained small and clearly nels. Intracellular calcium, for example, which is essential to
circumscribed. Administration of recombinant endostatin to many functions of RPE cells, including growth factor secretion,
Col18a1–/– mice decreased the lesion size to the sizes observed in phagocytosis, ion exchange, and water transport, is mediated by
control mice.175 a number of channels; these include L-type and T-type voltage-
gated calcium channels,31,185,186 calcium channels of the transient
Immune privilege receptor potential canonical (TRPC), specifically TRPC1 and
Immune privilege refers to the fact that foreign-tissue grafts TRPC4.187,188 Recently, two calcium transport channels, TRPV5
placed in the immune-privileged site are tolerated and survive and TRPV6, with a calcium–sodium selectivity of more than
for prolonged, often indefinite, intervals, while placement of 100 : 1 for calcium versus sodium, have also been identified in
such grafts at conventional body sites leads to acute irreversible RPE cells.189
immune rejection. Ocular immune privilege was demonstrated RPE cells are responsible for removing water, ions, and catabo-
by Sir Peter Medawar in 1948 when skin allografts survived lites from the retina to the choriocapillaries; however, RPE cell
when implanted into the anterior chamber of the eye.176 Only in tight junctions establish a barrier between the subretinal space
the past 20 years has evidence accumulated that the subretinal and the choriocapillaris such that water, ions or other molecules
space was also an immune-privileged space.177,178 Subsequently, cannot leave via the paracellular route, but must be actively
the RPE has been shown to be a major source of immune privi- transported transcellularly. Na+,K+-ATPase, which is localized at
lege in the subretinal space. Immune privilege has evolved from the apical surface of RPE cells, provides the energy for transepi-
a concept of passive physical barriers (tight junctions between thelial transport, and controls the flux of sodium and potassium
RPE; lack of lymphatic drainage from subretinal space; low ions across the plasma membrane, thereby maintaining the
levels of major histocompatibility antigen expression) to more of proper balance of these ions in the IPM and establishing mem-
an active process. The RPE express both soluble (TGF-β, PEDF) brane potentials. Water is eliminated from the subretinal space
and cell surface molecules (TGF-β, CD95 (FAS) ligand, CD59, by active transport by the RPE and is mediated by the transepi-
CD46) that promote immune privilege. TGF-β has been identi- thelial transport of chloride ion from the subretinal space, across
fied as a major factor in the inhibition of T-cell proliferation and the RPE to the choroid.190 Water movement across the RPE is also
IFN-γ production, and the generation of T-regulatory cells facilitated by Aquaporin-1 channels.191,192 It is critical that water
(Tregs) in ocular immune-privileged sites.178 Recently, immune formed in the retina as a result of high metabolic activity is
privilege in the aqueous humor was shown to require a role for removed, since excess water in the retina will result in macular
both TGF-β and retinoic acid in the generation of Tregs; however, edema, RPE and photoreceptor degeneration, and eventually
whether this is also true for the subretinal space is currently blindness. Elimination of lactic acid from the subretinal space is
unknown.179 A full discussion of the blood–retinal barrier and mediated by different monocarboxylate transporters in the
immune privilege is found in Chapter 27, Blood–retinal barrier, apical (MCT1) and basolateral (MCT3) membranes.36,37
immune privilege, and autoimmunity.
Secretion of cytokines and growth factors
Transport of nutrients, ions, and water RPE cells secrete many cytokines and growth factors that regu-
RPE cells are responsible for delivering nutrients from the circu- late many of the cellular pathways necessary for function, sur-
lation to the photoreceptors and retina and water and metabolic vival, and response to injury. The best known of these factors
Table 16.2 Growth factors produced by retinal pigment epithelium cells

410 Factor Function References

Pigment epithelial-derived factor (PEDF) Neuroprotection, neurogenesis, antiangiogenesis 88,143–147


Section 1

Vascular endothelial growth factor (VEGF) Angiogenesis, endothelial cell survival factor, maintenance of 88,174,193
choriocapillaris fenestrations

Nerve growth factor (NGF) Survival and maintenance of sympathetic and sensory neurons. Ocular 194,195
immune response

Brain-derived neurotrophic factor (BDNF) Supports the survival of existing neurons, and fosters the growth and 195,196
differentiation of new neurons and synapses
Anatomy and Physiology
Basic Science and Translation to Therapy

Neurotrophin-3 (NT-3) Stimulation and control of neurogenesis 196

Insulin-like growth factor (IGF-1) Regulation of neurogenesis, myelination, synaptogenesis, and dendritic 197–199
branching and neuroprotection after neuronal damage

Neuroprotectin 1 (NPD1) Protects neural tissues from injury caused by free radicals and other 200–202
oxidative stress

Transforming growth factor-β (TGFβ) Regulation of many cellular processes in the adult and developing 203–206
embryo, including cell growth and differentiation and cellular homeostasis

Granulocyte–macrophage colony- Stimulation of stem cells to differentiate into granulocytes and monocytes 207–209
stimulating factor (GM-CSF)

Monocyte chemotactic protein-1 (MCP-1) Monocyte chemoattractant 210,211

Hepatocyte growth factor (HGF) Hepatocyte growth factor regulates cell growth, cell adhesion, cell 212–214
motility, and morphogenesis

Erythropoietin (EPO) Regulates red blood cell production. EPO protects retina from physiologic 215–217
and pathologic light-induced oxidative injury

Platelet-activating factor (PAF) Phospholipid activator and mediator of platelet aggregation, inflammation, 218
and anaphylaxis

Melanoma growth-stimulatory activity/ Enhances chemotaxis of neutrophils and enhances the secretion of 219,220
growth-regulated protein (MGSA/GRO) elastase and other matrix-degrading enzymes

Endothelin 1 Vasoconstriction 221,222

Fibroblast growth factor Mitogenic and cell survival activities. Involved in embryonic development, 223–226
cell growth, morphogenesis, tissue repair, tumor growth, and invasion

Bone morphogenetic protein Regulation of differentiation, senescence, and apoptosis 227,228

Interleukin-6 Pleiotropic cytokine with a role in inflammation, hematopoiesis, 229,230


angiogenesis, cell differentiation, and neuronal survival

Interleukin-8 Interleukin-8 attracts and activates neutrophils 231,232

Connective tissue growth factor Intraocular fibrosis 233

are VEGF and PEDF. VEGF, which is constitutively secreted For online acknowledgments visit http://www.
from the basal surface of the RPE towards the choriocapillaris, expertconsult.com
has two main functions: (1) to provide prosurvival signals to the
choroidal vascular endothelial cells; and (2) to maintain the fen- REFERENCES
1. Martinez-Morales JR, Rodrigo I, Bovolenta P. Eye development: a view from
estrations of the choriocapillaris endothelium.193 PEDF is pre- the retina pigmented epithelium. Bioessays 2004;26:766–77.
dominantly secreted from the apical surface where it promotes 2. Perron M, Boy S, Amato MA, et al. A novel function for Hedgehog signalling
an antiangiogenic and neuroprotective environment for the pho- in retinal pigment epithelium differentiation. Development 2003;130:
1565–77.
toreceptors.88 Many of these factors show dysregulated expres- 3. Stroeva OG, Mitashov VI. Retinal pigment epithelium: proliferation and dif-
sion in retinal diseases such as AMD, diabetic retinopathy, ferentiation during development and regeneration. Int Rev Cytol 1983;83:
221–93.
and PVR. The most common of these factors are listed in 4. Marmorstein AD. The polarity of the retinal pigment epithelium. Traffic
Table 16.2. 2001;2:867–72.
ACKNOWLEDGMENTS
The authors would like to thank Ernesto Barron for preparation 410.e1
of figures and Ram Kannan, PhD and Sreekumar P.G.
Parameswaran, PhD for their insightful additions and
comments.

Chapter 16
Cell Biology of the Retinal Pigment Epithelium
5. Fuhrmann S. Eye morphogenesis and patterning of the optic vesicle. Curr Top 37. Gallagher-Colombo S, Maminishkis A, Tate S, et al. Modulation of MCT3
Dev Biol 2010;93:61–84. expression during wound healing of the retinal pigment epithelium. Invest
6. Zhang XM, Yang XJ. Temporal and spatial effects of Sonic hedgehog signaling Ophthalmol Vis Sci 2010;51:5343–50.
in chick eye morphogenesis. Dev Biol 2001;233:271–90. 38. Korte GE, Perlman JI, Pollack A. Regeneration of mammalian retinal pigment 411
7. Matt N, Ghyselinck NB, Pellerin I, et al. Impairing retinoic acid signalling in epithelium. Int Rev Cytol 1994;152:223–63.
the neural crest cells is sufficient to alter entire eye morphogenesis. Dev Biol 39. Gouras P, Ivert L, Neuringer M, et al. Topographic and age-related changes
2008;320:140–8. of the retinal epithelium and Bruch’s membrane of rhesus monkeys. Graefes

Chapter 16
8. Adler R, Belecky-Adams TL. The role of bone morphogenetic proteins Arch Clin Exp Ophthalmol 2010;248:973–84.
in the differentiation of the ventral optic cup. Development 2002;129: 40. Peters S, Reinthal E, Blitgen-Heinecke P, et al. Inhibition of lysosomal degra-
3161–71. dation in retinal pigment epithelium cells induces exocytosis of phagocytic
9. Schouwey K, Aydin IT, Radtke F, et al. RBP-Jkappa-dependent Notch signal- residual material at the basolateral plasma membrane. Ophthalmic Res
ing enhances retinal pigment epithelial cell proliferation in transgenic mice. 2006;38:83–8.
Oncogene 2011;30:313–22. 41. Fine BS. Limiting membranes of the sensory retina and pigment epithelium.
10. Burke JM. Epithelial phenotype and the RPE: is the answer blowing in the An electron microscopic study. Arch Ophthalmol 1961;66:847–60.
Wnt? Prog Retin Eye Res 2008;27:579–95. 42. Rodriguez-Boulan E. Genesis of polarity in renal tubular cells. Miner
11. Bonilha VL, Rayborn ME, Saotome I, et al. Microvilli defects in retinas of ezrin Electrolyte Metab 1986;12:20–4.
knockout mice. Exp Eye Res 2006;82:720–9. 43. Georgiadis A, Tschernutter M, Bainbridge JW, et al. The tight junction associ-

Cell Biology of the Retinal Pigment Epithelium


12. Lawrence MS, Azar DT. Myopia and models and mechanisms of refractive ated signalling proteins ZO-1 and ZONAB regulate retinal pigment epithe-
error control. Ophthalmol Clin North Am 2002;15:127–33. lium homeostasis in mice. PLoS ONE 2010;5:e15730.
13. Panda-Jonas S, Jonas JB, Jakobczyk-Zmija M. Retinal pigment epithelial cell 44. Luo Y, Fukuhara M, Weitzman M, et al. Expression of JAM-A, AF-6, PAR-3
count, distribution, and correlations in normal human eyes. Am J Ophthalmol and PAR-6 during the assembly and remodeling of RPE tight junctions.
1996;121:181–9. Brain Res 2006;1110:55–63.
14. Al-Hussaini H, Kam JH, Vugler A, et al. Mature retinal pigment epithelium 45. Peng S, Rahner C, Rizzolo LJ. Apical and basal regulation of the permeability
cells are retained in the cell cycle and proliferate in vivo. Mol Vision 2008; of the retinal pigment epithelium. Invest Ophthalmol Vis Sci 2003;44:
14:1784–91. 808–17.
15. Schmidt SY, Peisch RD. Melanin concentration in normal human retinal 46. Peng S, Rao VS, Adelman RA, et al. Claudin-19 and the barrier properties of
pigment epithelium. Regional variation and age-related reduction. Invest the human retinal pigment epithelium. Invest Ophthalmol Vis Sci 2011;52:
Ophthalmol Vis Sci 1986;27:1063–7. 1392–403.
16. Sarna T, Burke JM, Korytowski W, et al. Loss of melanin from human RPE 47. Economopoulou M, Hammer J, Wang F, et al. Expression, localization, and
with aging: possible role of melanin photooxidation. Exp Eye Res 2003;76: function of junctional adhesion molecule-C (JAM-C) in human retinal pigment
89–98. epithelium. Invest Ophthalmol Vis Sci 2009;50:1454–63.
17. Burke JM, Skumatz CM, Irving PE, et al. Phenotypic heterogeneity of retinal 48. Bailey TA, Kanuga N, Romero IA, et al. Oxidative stress affects the junctional
pigment epithelial cells in vitro and in situ. Exp Eye Res 1996;62:63–73. integrity of retinal pigment epithelial cells. Invest Ophthalmol Vis Sci 2004;
18. Burke JM, Soref C. Topographical variation in growth in cultured bovine 45:675–84.
retinal pigment epithelium. Invest Ophthalmol Vis Sci 1988;29:1784–8. 49. Rajasekaran SA, Hu J, Gopal J, et al. Na,K-ATPase inhibition alters tight junc-
19. Burke JM, McKay BS, Jaffe GJ. Retinal pigment epithelial cells of the posterior tion structure and permeability in human retinal pigment epithelial cells.
pole have fewer Na/K adenosine triphosphatase pumps than peripheral cells. Am J Physiol Cell Physiol 2003;284:C1497–507.
Invest Ophthalmol Vis Sci 1991;32:2042–6. 50. Zhou J, He S, Zhang N, et al. Neutrophils compromise retinal pigment
20. McLaren MJ. Kinetics of rod outer segment phagocytosis by cultured retinal epithelial barrier integrity. J Biomed Biotechnol 2010:289–360.
pigment epithelial cells. Relationship to cell morphology. Invest Ophthalmol 51. Zech JC, Pouvreau I, Cotinet A, et al. Effect of cytokines and nitric oxide on
Vis Sci 1996;37:1213–24. tight junctions in cultured rat retinal pigment epithelium. Invest Ophthalmol
21. Cabral L, Unger W, Boulton M, et al. Regional distribution of lysosomal Vis Sci 1998;39:1600–8.
enzymes in the canine retinal pigment epithelium. Invest Ophthalmol Vis Sci 52. Bruban J, Glotin AL, Dinet V, et al. Amyloid-beta(1–42) alters structure and
1990;31:670–6. function of retinal pigmented epithelial cells. Aging Cell 2009;8:162–77.
22. Hjelmeland LM, Fujikawa A, Oltjen SL, et al. Quantification of retinal pigment 53. Crawford BJ. Development of the junctional complex during differentiation
epithelial phenotypic variation using laser scanning cytometry. Mol Vision of chick pigmented epithelial cells in clonal culture. Invest Ophthalmol Vis
2010;16:1108–21. Sci 1980;19:223–37.
23. Gulcan HG, Alvarez RA, Maude MB, et al. Lipids of human retina, retinal 54. Hudspeth AJ, Yee AG. The intercellular junctional complexes of retinal
pigment epithelium, and Bruch’s membrane/choroid: comparison of macular pigment epithelia. Invest Ophthalmol 1973;12:354–65.
and peripheral regions. Invest Ophthalmol Vis Sci 1993;34:3187–93. 55. Kalnins VI, Sandig M, Hergott GJ, et al. Microfilament organization and
24. Bonilha VL, Rayborn ME, Bhattacharya SK, et al. The retinal pigment epithe- wound repair in retinal pigment epithelium. Biochem Cell Biol 1995;73:
lium apical microvilli and retinal function. Adv Exp Med Biol 2006;572: 709–22.
519–24. 56. Sandig M, Kalnins VI. Subunits in zonulae adhaerentes and striations in the
25. Young RW. The renewal of rod and cone outer segments in the rhesus associated circumferential microfilament bundles in chicken retinal pigment
monkey. J Cell Biol 1971;49:303–18. epithelial cells in situ. Exp Cell Res 1988;175:1–14.
26. Bonnel S, Mohand-Said S, Sahel JA. The aging of the retina. Exp Gerontol 57. Tibber MS, Becker D, Jeffery G. Levels of transient gap junctions between the
2003;38:825–31. retinal pigment epithelium and the neuroblastic retina are influenced by cat-
27. Bonilha VL, Bhattacharya SK, West KA, et al. Proteomic characterization of echolamines and correlate with patterns of cell production. J Comp Neurol
isolated retinal pigment epithelium microvilli. Mol Cell Proteomics 2004;3: 2007;503:128–34.
1119–27. 58. Pearson RA, Dale N, Llaudet E, et al. ATP released via gap junction hemichan-
28. Finnemann SC, Bonilha VL, Marmorstein AD, et al. Phagocytosis of rod outer nels from the pigment epithelium regulates neural retinal progenitor prolif-
segments by retinal pigment epithelial cells requires alpha(v)beta5 integrin eration. Neuron 2005;46:731–44.
for binding but not for internalization. Proc Natl Acad Sci U S A 1997;94: 59. Opas M, Kalnins VI. Light-microscopical analysis of focal adhesions of retinal
12932–7. pigmented epithelial cells. Invest Ophthalmol Vis Sci 1986;27:1622–33.
29. Tarnowski BI, Shepherd VL, McLaughlin BJ. Mannose 6-phosphate receptors 60. Owaribe K, Kartenbeck J, Rungger-Brandle E, et al. Cytoskeletons of retinal
on the plasma membrane on rat retinal pigment epithelial cells. Invest Oph- pigment epithelial cells: interspecies differences of expression patterns indi-
thalmol Vis Sci 1988;29:291–7. cate independence of cell function from the specific complement of cytoskel-
30. Ryeom SW, Sparrow JR, Silverstein RL. CD36 participates in the phagocytosis etal proteins. Cell Tissue Res 1988;254:301–15.
of rod outer segments by retinal pigment epithelium. J Cell Sci 1996;109: 61. Schlotzer-Schrehardt U, Muller HG, Wirtz PM, et al. Desmosomal–
387–95. mitochondrial complexes in human nonpigmented ciliary and retinal pigment
31. Wimmers S, Karl MO, Strauss O. Ion channels in the RPE. Prog Retin Eye Res epithelia. Invest Ophthalmol Vis Sci 1990;31:664–9.
2007;26:263–301. 62. Hunt RC, Davis AA. Altered expression of keratin and vimentin in human
32. Chuang JZ, Chou SY, Sung CH. Chloride intracellular channel 4 is critical for retinal pigment epithelial cells in vivo and in vitro. J Cell Physiol 1990;
the epithelial morphogenesis of RPE cells and retinal attachment. Mol Biol 145:187–99.
Cell 2010;21:3017–28. 63. Grisanti S, Guidry C. Transdifferentiation of retinal pigment epithelial cells
33. Milenkovic VM, Krejcova S, Reichhart N, et al. Interaction of bestrophin-1 and from epithelial to mesenchymal phenotype. Invest Ophthalmol Vis Sci
Ca2+ channels beta-subunits: identification of new binding domains on the 1995;36:391–405.
bestrophic-1 C-terminus. PLoS ONE 2011;6:e19364. 64. Lee J, Ko M, Joo CK. Rho plays a key role in TGF-beta1-induced cytoskeletal
34. Bonilha VL, Finnemann SC, Rodriguez-Bouan E. Ezrin promotes morphogen- rearrangement in human retinal pigment epithelium. J Cell Physiol 2008;216:
esis of apical microvilli and basal infoldings in retinal pigment epithelium. 520–6.
J Cell Biol 1999;147:1533–47. 65. Burnside B, Adler R, O’Connor P. Retinomotor pigment migration in the
35. Senanayake P, Calabro A, Hu JG, et al. Glucose utilization by the retinal teleost retinal pigment epithelium. I. Roles for actin and microtubules in
pigment epithelium: evidence for rapid uptake and storage in glycogen, pigment granule transport and cone movement. Invest Ophthalmol Vis Sci
followed by glycogen utilization. Exp Eye Res 2006;83:235–46. 1983;24:1–15.
36. Philp NJ, Wang D, Yoon H, et al. Polarized expression of monocarboxylate 66. McNeil EL, Tacelosky D, Basciano P, et al. Actin-dependent motility of mela-
transporters in human retinal pigment epithelium. Invest Ophthalmol Vis Sci nosomes from fish retinal pigment epithelial (RPE) cells investigated using in
2003;44:1716–21. vitro motility assays. Cell Motil Cytoskeleton 2004;58:71–82.
67. Lin-Jones J, Sohlberg L, Dose A, et al. Identification and localization of myosin 100. Maminishkis A, Chen S, Jalickee S, et al. Confluent monolayers of cultured
superfamily members in fish retina and retinal pigmented epithelium. J Comp human fetal retinal pigment epithelium exhibit morphology and physiology
Neurol 2009;513:209–23. of native tissue. Invest Ophthalmol Vis Sci 2006;47:3612–24.
412 68. Coudrier E. Myosins in melanocytes: to move or not to move? Pigment Cell 101. Sonoda S, Spee C, Barron E, et al. A protocol for the culture and differentiation
Res 2007;20:153–60. of highly polarized human retinal pigment epithelial cells. Nat Protoc
69. Turksen K, Opas M, Aubin JE, et al. Microtubules, microfilaments and adhe- 2009;4:662–73.
sion patterns in differentiating chick retinal pigment epithelial (RPE) cells in 102. Maminishkis A, Miller SS. Experimental models for study of retinal pigment
vitro. Exp Cell Res 1983;147:379–91. epithelial physiology and pathophysiology. J Vis Exp 2010 doi: 10.3791/2032.
Section 1

70. Korte GE. New ultrastructure of rat RPE cells: basal intracytoplasmic tubules. 103. Vossmerbaeumer U, Kuehl S, Kern S, et al. Induction of retinal pigment epi-
Exp Eye Res 1984;38:399–409. thelium properties in ciliary margin progenitor cells. Clin Exp Ophthalmol
71. Robey HL, Hiscott PS, Grierson I. Cytokeratins and retinal epithelial cell 2008;36:358–66.
behaviour. J Cell Sci 1992;102:329–40. 104. Aruta C, Giordano F, De Marzo A, et al. In vitro differentiation of retinal
72. Bok D. The retinal pigment epithelium: a versatile partner in vision. J Cell Sci pigment epithelium from adult retinal stem cells. Pigment Cell Melanoma Res
Suppl 1993;17:189–95. 2011;24:233–40.
73. Hogan MJ. Ultrastructure of the choroid. Its role in the pathogenesis of cho- 105. Idelson M, Alper R, Obolensky A, et al. Directed differentiation of human
rioretinal disease. Transactions of the Pacific Coast Oto-Ophthalmological embryonic stem cells into functional retinal pigment epithelium cells.
Anatomy and Physiology

Society Annual Meeting 1961;42:61–87. Cell Stem Cell 2009;5:396–408.


Basic Science and Translation to Therapy

74. Hirabayashi Y, Fujimori O, Shimizu S. Bruch’s membrane of the brachymor- 106. Zhu D, Deng X, Spee C, et al. Polarized secretion of PEDF from human
phic mouse. Med Electron Microsc 2003;36:139–46. embryonic stem cell-derived RPE promotes retinal progenitor cell survival.
75. Timpl R. Structure and biological activity of basement membrane proteins. Invest Ophthalmol Vis Sci 2011;52:1573–85.
Eur J Biochem 1989;180:487–502. 107. Buchholz DE, Hikita ST, Rowland TJ, et al. Derivation of functional retinal
76. Marshall GE, Konstas AG, Lee WR. Collagens in ocular tissues. Br J Ophthal- pigmented epithelium from induced pluripotent stem cells. Stem Cells
mol 1993;77:515–24. 2009;27:2427–34.
77. Aisenbrey S, Zhang M, Bacher D, et al. Retinal pigment epithelial cells 108. Hattar S, Liao HW, Takao M, et al. Melanopsin-containing retinal ganglion
synthesize laminins, including laminin 5, and adhere to them through cells: architecture, projections, and intrinsic photosensitivity. Science 2002;295:
alpha3- and alpha6-containing integrins. Invest Ophthalmol Vis Sci 2006; 1065–70.
47:5537–44. 109. Allen AE, Cameron MA, Brown TM, et al. Visual responses in mice lacking
78. Libby RT, Hunter DD, Brunken, WJ. The roles of the extracellular matrix in critical components of all known retinal phototransduction cascades.
retinal development and maintenance. In: Fini E, editor. Vertebrate eye devel- PLoS ONE 2010;5:e15063.
opment. Berlin: Springer-Verlag; 2000. p. 115–40. 110. Tamai M, Chader GJ. The early appearance of disc shedding in the rat retina.
79. Hewitt AT, Nakazawa K, Newsome DA. Analysis of newly synthesized Invest Ophthalmol Vis Sci 1979;18:913–7.
Bruch’s membrane proteoglycans. Invest Ophthalmol Vis Sci 1989;30: 111. Tamai M, Teirstein P, Goldman A, et al. The pineal gland does not control rod
478–86. outer segment shedding and phagocytosis in the rat retina and pigment epi-
80. Bhutto IA, Kim SY, McLeod DS, et al. Localization of collagen XVIII and the thelium. Invest Ophthalmol Vis Sci 1978;17:558–62.
endostatin portion of collagen XVIII in aged human control eyes and eyes 112. Young RW. The daily rhythm of shedding and degradation of cone outer
with age-related macular degeneration. Invest Ophthalmol Vis Sci 2004;45: segment membranes in the lizard retina. J Ultrastruct Res 1977;61:172–85.
1544–52. 113. Anderson DH, Fisher SK. Disc shedding in rod like and cone like photorecep-
81. Ma DH, Yao JY, Kuo MT, et al. Generation of endostatin by matrix metallo- tors of tree squirrels. Science 1975;187:953–5.
proteinase and cathepsin from human limbocorneal epithelial cells cultivated 114. Anderson DH, Fisher SK, Erickson PA, et al. Rod and cone disc shedding in
on amniotic membrane. Invest Ophthalmol Vis Sci 2007;48:644–51. the rhesus monkey retina: a quantitative study. Exp Eye Res 1980;30:559–74.
82. Booij JC, Baas DC, Beisekeeva J, et al. The dynamic nature of Bruch’s mem- 115. Fisher SK, Pfeffer BA, Anderson DH. Both rod and cone disc shedding are
brane. Prog Retin Eye Res 2010;29:1–18. related to light onset in the cat. Invest Ophthalmol Vis Sci 1983;24:844–56.
83. Booij JC, Jacoline B, ten Brink JB, et al. A new strategy to identify and annotate 116. Young RW. The renewal of photoreceptor cell outer segments. J Cell Biol
human RPE-specific gene expression. PLoS ONE 2010;5:e9341. 1967;33:61–72.
84. Chader GJ. Interphotoreceptor retinoid-binding protein (IRBP): a model 117. Young RW, Bok D. Participation of the retinal pigment epithelium in the rod
protein for molecular biological and clinically relevant studies. Friedenwald outer segment renewal process. J Cell Biol 1969;42:392–403.
lecture. Invest Ophthalmol Vis Sci 1989;30:7–22. 118. Young RW. Biological renewal; applications to the eye. Trans Ophthalmol Soc
85. Adler AJ, Evans CD. Proteins of the bovine interphotoreceptor matrix: reti- UK 1982;102:42–75.
noid binding and other functions. Prog Clin Biol Res 1985;190:65–88. 119. Kevany BM, Palczewski K. Phagocytosis of retinal rod and cone photorecep-
86. Semenova EM, Converse CA. Comparison between oleic acid and docosa- tors. Physiology 2010;25:8–15.
hexaenoic acid binding to interphotoreceptor retinoid-binding protein. Vision 120. Boesze-Battaglia K, Albert AD. Phospholipid distribution among bovine rod
Res 2003;43:3063–7. outer segment plasma membrane and disk membranes. Exp Eye Res
87. Cunningham LL, Gonzalez-Fernandez F. Internalization of interphotorecep- 1992;54:821–3.
tor retinoid-binding protein by the Xenopus retinal pigment epithelium. 121. Nandrot EF, Anand M, Dena Almeida D, et al. Essential role for MFG-E8 as
J Comp Neurol 2003;466:331–42. ligand for αvβ5 integrin in diurnal retinal phagocytosis Proc Natl Acad Sci
88. Sonoda S, Sreekumar PG, Kase S, et al. Attainment of polarity promotes U S A 2007;104:12005–10
growth factor secretion by retinal pigment epithelial cells: relevance to age- 122. Caberoy NB, Maiguel D, Kim Y, et al. Identification of tubby and tubby-like
related macular degeneration. Aging 2009;2:28–42. protein 1 as eat-me signals by phage display. Exp Cell Res 2010;316:245–57
89. Hauck SM, Schoeffmann S, Deeg CA, et al. Proteomic analysis of the porcine 123. Chaitin MH, Hall MO. The distribution of actin in cultured normal and dys-
interphotoreceptor matrix. Proteomics 2005;5:3623–36. trophic rat pigment epithelial cells during the phagocytosis of rod outer seg-
90. Sreekumar PG, Kannan R, Kitamura M, et al. alphaB crystallin is apically ments. Invest Ophthalmol Vis Sci 1983;24:821–31.
secreted within exosomes by polarized human retinal pigment epithelium 124. Hall MO, Abrams T, Mittag TW. ROS ingestion by RPE cells is turned off by
and provides neuroprotection to adjacent cells. PLoS ONE 2010;5:e12578. increased protein kinase C activity and by increased calcium. Exp Eye Res
91. Elner SG, Elner VM, Kindzelskii AL, et al. Human RPE cell lysis of extracel- 1991;52:591–8.
lular matrix: functional urokinase plasminogen activator receptor (uPAR), 125. Hall MO, Obin MS, Prieto AL et al. Gas6 binding to photoreceptor outer
collagenase and elastase. Exp Eye Res 2003;76:585–95. segments requires gamma-carboxyglutamic acid (Gla) and Ca2+ and is
92. Alexander JP, Bradley JM, Gabourel JD, et al. Expression of matrix metallo- required for OS phagocytosis by RPE cells in vitro. Exp Eye Res 2002;75:
proteinases and inhibitor by human retinal pigment epithelium. Invest 391–400.
Ophthalmol Vis Sci 1990;31:2520–8. 126. Besharse JC, Dunis DA. Rod photoreceptor disc shedding in vitro: inhibition
93. Smine A, Plantner JJ. Membrane type-1 matrix metalloproteinase in human by cytochalasins and activation by colchicine. In: Hollyfield JG, editor. The
ocular tissues. Curr Eye Res 1997;16:925–9. structure of the eye. New York: Elsevier Biomedical; 1982.
94. Jomary C, Neal MJ, Iwata K, et al. Localization of tissue inhibitor of 127. Herman KG, Steinberg RH. Phagosome movement and the diurnal pattern of
metalloproteinases-3 in neurodegenerative retinal disease. Neuroreport phagocytosis in the tapetal retinal pigment epithelium of the opossum. Invest
1997;8:2169–72. Ophthalmol Vis Sci 1982;23:277–90.
95. Siren V, Immonen I. uPA, tPA and PAI-1 mRNA expression in periretinal 128. Gibbs D, Kitamoto J, Williams, DS. Abnormal phagocytosis by retinal pig-
membranes. Curr Eye Res 2003;27:261–7. mented epithelium that lack myosin VIIa, the Usher syndrome 1B protein.
96. Andreasen PA, Kjoller L, Christensen L, et al. The urokinase-type plasmin­ Proc Natl Acad Sci U S A 2003;100:6481–64860.
ogen activator system in cancer metastasis: a review. Int J Cancer 1997;72: 129. Hayashi A, Nakae K, Naka H, et al. Cytokine effects on phagocytosis of rod
1–22. outer segments by retinal pigment epithelial cells of normal and dystrophic
97. Siren V, Stephens RW, Salonen EM, et al. Retinal pigment epithelial cells rats. Curr Eye Res 1996;15:487–99.
secrete urokinase-type plasminogen activator and its inhibitor PAI-1. 130. Rakoczy PE, Baines M, Kennedy CJ, et al. Correlation between autofluorescent
Ophthalmic Res 1992;24:203–12. debris accumulation and the presence of partially processed forms of cathep-
98. Singh MS, MacLaren RE. Stem cells as a therapeutic tool for the blind: biology sin D in cultured retinal pigment epithelial cells challenged with rod outer
and future prospects. Proc R Soc B 2011;278:3009–16. segments. Exp Eye Res 1996;63:159–67.
99. Hu J, Bok D. A cell culture medium that supports the differentiation of human 131. Regan CM, De Grip WJ, Daemen FJM, et al. Degradation of rhodopsin by a
retinal pigment epithelium into functionally polarized monolayers. Mol Vis lysosomal fraction of retinal pigment epithelium: biochemical aspects of the
2001;7:14–9. visual process, XLI. Exp Eye Res 1980;30:183–91.
132. Zimmerman WF, Godchaux III W, Belkin M. The relative proportions of 163. Wu H, Xing K, Lou MF. Glutaredoxin 2 prevents H(2)O(2)-induced cell apop-
lysosomal enzyme activities in bovine retinal pigment epithelium. Exp Eye tosis by protecting complex I activity in the mitochondria. Biochim Biophys
Res 1983;36:151–8. Acta 2010;1797:1705–15.
133. Rakoczy PE, Mann K, Cavaney DM, et al. Detection and possible functions of 164. Sreekumar PG, Kannan R, Yaung J, et al. Protection from oxidative stress by 413
a cysteine protease involved in digestion of rod outer segments by retinal methionine sulfoxide reductases in RPE cells. Biochem Biophys Res Commun
pigment epithelial cells. Invest Ophthalmol Vis Sci 1994;35:4100–8. 2005;334:245–53.
134. Feeney L, Mixon RL. An in vitro model of phagocytosis in bovine and human 165. Mao YW, Liu JP, Xiang H, et al. Human alphaA- and alphaB-crystallins bind

Chapter 16
retinal pigment epithelium. Exp Eye Res 1976;22:533–48. to Bax and Bcl-X(S) to sequester their translocation during staurosporine-
135. Feeney-Burns L, Hilderbrand ES, Eldridge S. Aging human RPE: morphomet- induced apoptosis. Cell Death Differ 2004;11:512–26.
ric analysis of macular, equatorial, and peripheral cells. Invest Ophthalmol 166. Yaung J, Jin M, Barron E, et al. alpha-Crystallin distribution in retinal pigment
Vis Sci 1984;25:195–200. epithelium and effect of gene knockouts on sensitivity to oxidative stress.
136. Pollock NL, Callaghan R. The lipid translocase, ABCA4: seeing is believing. Mol Vis 2007;13:566–77.
FEBS J 2011;278:3204–14. 167. Yaung J, Kannan R, Wawrousek EF, et al. Exacerbation of retinal degeneration
137. Edwards RB, Adler AJ. IRBP enhances removal of 11-cis-retinaldehyde from in the absence of alpha crystallins in an in vivo model of chemically induced
isolated RPE membranes. Exp Eye Res 2000;70:235–45. hypoxia. Exp Eye Res 2008;86:355–65.
138. Wu Q, Blakeley LR, Cornwall MC, et al. Interphotoreceptor retinoid binding 168. Pasupuleti N, Matsuyama S, Voss O, et al. The anti-apoptotic function of
protein is the physiologically relevant carrier that removes retinol from rod human αA-crystallin is directly related to its chaperone activity. Cell Death

Cell Biology of the Retinal Pigment Epithelium


photoreceptor outer segments. Biochemistry 2007;46:8669–79. Dis 2010;1:e31.
139. Jin M, Li S, Nusinowitz S, et al. The role of interphotoreceptor retinoid- 169. Kannan R, Sreekumar PG, Hinton DR. Glutathione metabolism and its con-
binding protein on the translocation of visual retinoids and function of cone tribution to antiapoptotic properties of alpha crystallins. In: Stratton RD,
photoreceptors. J Neurosci 2009;29:1486–95. Hauswirth WW, Gardner TW, editors. Studies on retinal and choroidal
140. Cai J, Nelson KC, Wu M, et al. Oxidative damage and protection of the RPE. disorders. New York, NY: Springer; 2012, Chapter IX.
Prog Retin Eye Res 2000;19:205–2. 170. Filleur S, Volz K, Nelius T, et al. Two functional epitopes of pigment epithelial-
141. Mukherjee PK, Marcheselli VL, Serhan CN, et al. Neuroprotectin D1: a doco- derived factor block angiogenesis and induce differentiation in prostate
sahexaenoic acid-derived docosatriene protects human retinal pigment cancer. Cancer Res 2005;65:5144–52.
epithelial cells from oxidative stress. Proc Natl Acad Sci U S A 2004;101: 171. Bouck N. PEDF: anti-angiogenic guardian of ocular function. Trends Mol Med
8491–6. 2002;8:330–4.
142. Lukiw WJ, Cui JG, Marcheselli VL, et al. A role for docosahexaenoic acid- 172. Bhutto IA, Mcleod DS, Hasegawa T, et al. Pigment epithelium-derived factor
derived neuroprotectin D1 in neural cell survival and Alzheimer disease. (PEDF) and vascular endothelial growth factor (VEGF) in aged human
J Clin Invest 2005;115:2774–83. choroid and eyes with age-related macular degeneration. Exp Eye Res 2006;
143. Wu YQ, Notario V, Chader GJ, et al. Identification of pigment epithelium- 82:99–110.
derived factor in the interphotoreceptor matrix of bovine eyes. Protein Expr 173. Kozulin P, Natoli R, Bumsted O’Brien KM, et al. The cellular expression of
Purif 1995;6:447–56. antiangiogenic factors in fetal primate macula. Invest Ophthalmol Vis Sci
144. Perez-Mediavilla LA, Chew C, Campochiaro PA, et al. Sequence and expres- 2010;51:4298–306.
sion analysis of bovine pigment epithelium-derived factor. Biochim Biophys 174. Kociok N, Joussen AM. Varied expression of functionally important genes of
Acta 1998;1398:203–14. RPE and choroid in the macula and in the periphery of normal human eyes.
145. Bilak MM, Corse AM, Bilak SR, et al. Pigment epithelium-derived factor Graefes Arch Clin Exp Ophthalmol 2007;245:101–13.
(PEDF) protects motor neurons from chronic glutamate-mediated neurode- 175. Marneros AG, She H, Zambarakji H, et al. Endogenous endostatin inhibits
generation. J Neuropathol Exp Neurol 1999;58:719–28. choroidal neovascularization. FASEB J 2007;21:3809–18.
146. Duh EJ, Yang HS, Suzuma I, et al. Pigment epithelium-derived factor sup- 176. Medawar PB. Immunity to homologous grafted skin; the fate of skin homo-
presses ischemia-induced retinal neovascularization and VEGF-induced grafts transplanted to the brain, to subcutaneous tissue, and to the anterior
migration and growth. Invest Ophthalmol Vis Sci 2002;43:821–9. chamber of the eye. Br J Exp Pathol 1948;29:58–69.
147. Ogata N, Wang L, Jo N, et al. Pigment epithelium derived factor as a neuro- 177. Streilein JW, Ma N, Wenkel H, et al. Immunobiology and privilege of neuronal
protective agent against ischemic retinal injury. Curr Eye Res 2001;22: retina and pigment epithelium transplants. Vis Res 2002;42:487–95.
245–52. 178. Masli S, Vega JL, Ocular immune privileged sites. Methods Mol Biol
148. Faktorovich E, Steinberg R, Yasumara D, et al. Photoreceptor degeneration in 2011;677:449–58.
inherited retinal dystrophy delayed by basic fibroblast growth factor. Nature 179. Zhou R, Horai R, Mattapallil MJ, et al. A new look at immune privilege of the
1990;347:83–6. eye: dual role for the vision-related molecule retinoic acid. J Immunol 2011;
149. Campochiaro PA, Chang M, Ohsato M, et al. Retinal degeneration in trans- 187;4170–7.
genic mice with photoreceptor-specific expression of a dominant-negative 180. Ban Y, Rizzolo LJ. Regulation of glucose transporters during development of
fibroblast growth factor receptor. J Neurosci 1996;16:1679–88. the retinal pigment epithelium. Dev Brain Res 2000;121:89–95.
150. Marí M, Morales A, Colell A, et al. Mitochondrial glutathione, a key survival 181. Bergersen L, Jóhannsson E, Veruki ML, et al. Cellular and subcellular expres-
antioxidant. Antioxid Redox Signal 2009;11:2685–700. sion of monocarboxylate transporters in the pigment epithelium and retina of
151. Holmgren A, Lu J. Thioredoxin and thioredoxin reductase: current research the rat. Neuroscience 1999;90:319–31.
with special reference to human disease. Biochem Biophys Res Commun 182. Kim D-I, Lim S-K, Park M-J, et al. The involvement of phosphatidylinositol
2010;396:120–4. 3-kinase/Akt signaling in high glucose-induced downregulation of GLUT-1
152. Sreekumar PG, Ding Y, Ryan SJ, et al. Regulation of thioredoxin by ceramide expression in ARPE cells. Life Sci 2007;80:626–32.
in retinal pigment epithelial cells. Exp Eye Res 2009;88:410–7. 183. Montel-Hagen A, Kinet S, Manel N, et al. Erythrocyte Glut1 triggers dehydro-
153. Sreekumar PG, Hinton DR, Kannan R. Methionine sulfoxide reductase A: ascorbic acid uptake in mammals unable to synthesize vitamin C.
Structure, function and role in ocular pathology. World J Biol Chem Cell 2008;132:1039–48.
2011;2:184–92. 184. Bridges CD, Alvarez RA, Fong SL, et al. Visual cycle in the mammalian eye.
154. Kannan R, Jin M, Gamulescu, M-A, et al. Ceramide-induced apoptosis: role Retinoid-binding proteins and the distribution of 11-cis retinoids. Vision Res
of catalase and hepatocyte growth factor. Free Radic Biol Med 2004;37: 1984;24:1581–94.
166–75. 185. Wimmers S, Coeppicus L, Rosenthal R, et al. Expression profile of voltage-
155. Dong A, Shen J, Krause M, et al. Superoxide dismutase 1 protects retinal cells dependent Ca2+ channel subunits in the human retinal pigment epithelium.
from oxidative damage. J Cell Physiol 2006;208:516–26. Graefes Arch Clin Exp Ophthalmol 2008;246:685–92.
156. Usui S, Oveson BC, Iwase T, et al. Overexpression of SOD in retina: need 186. Wimmers S, Halsband C, Seyler S, et al. Voltage-dependent Ca2+
for increase in H2O2-detoxifying enzyme in same cellular compartment. channels, not ryanodine receptors, activate Ca2+-dependent BK potassium
Free Radic Biol Med 2011;51:1347–54. channels in human retinal pigment epithelial cells. Mol Vis 2008;14:
157. Zhu D, Sreekumar PG, Hinton DR, et al. Expression and regulation of 2340–8.
enzymes in the ceramide metabolic pathway in human retinal pigment epi- 187. Bollimuntha S, Cornatzer E, Singh BB. Plasma membrane localization and
thelial cells and their relevance to retinal degeneration. Vision Res 2010;50: function of TRPC1 is dependent on its interaction with beta-tubulin in retinal
643–51. epithelium cells. Vis Neurosci 2005;22:163–70.
158. Jin M, Yaung J, Kannan R, et al. Hepatocyte growth factor protects RPE cells 188. Wimmers S, Strauss O. Basal calcium entry in retinal pigment epithelial
from apoptosis induced by glutathione depletion. Invest Ophthalmol Vis Sci cells is mediated by TRPC channels. Invest Ophthalmol Vis Sci 2007;48:
2005;46:4311–9. 5767–72.
159. Johnson J, Maher P, Hanneken A. The flavonoid, eriodictyol, induces long- 189. Kennedy BG, Torabi AJ, Kurzawa R, et al. Expression of transient receptor
term protection in ARPE-19 cells through its effects on Nrf2 activation and potential vanilloid channels in retinal pigment epithelium, Mol Vis 2010;16:
phase 2 gene expression. Invest Ophthalmol Vis Sci 2009;50:2398–406. 665–75.
160. Kowluru RA, Kowluru V, Xiong Y, et al. Overexpression of mitochondrial 190. Moseley H, Foulds WS, Allan D, et al. Routes of clearance of radioactive water
superoxide dismutase in mice protects the retina from diabetes-induced oxi- from the rabbit vitreous. Br J Ophthalmol 1984;68:145–5.
dative stress. Free Radic Biol Med 2006;41:1191–6. 191. Stamer WD, Bok D, Hu J, et al. Aquaporin-1 channels in human retinal
161. Inomata Y, Nakamura H, Tanito M, et al. Thioredoxin inhibits NMDA- pigment epithelium: role in transepithelial water movement. Invest Ophthal-
induced neurotoxicity in the rat retina. J Neurochem 2006;98:372–85. mol Vis Sci 2003;44:2803–8.
162. Wu H, Lin L, Giblin F, et al. Glutaredoxin 2 knockout increases sensitivity to 192. Motulsky E, Philippe Koch P, Sarah Janssens S, et al. Aquaporin expression
oxidative stress in mouse lens epithelial cells. Free Radic Biol Med 2011;51: in blood–retinal barrier cells during experimental autoimmune uveitis.
2108–17. Mol Vision 2010;16:602–10.
193. Blaauwgeers HG, Holtkamp GM, Rutten H, et al. Polarized vascular endothe- 213. Ohtaka K, Machida S, Ohzeki T, et al. Protective effect of hepatocyte growth
lial growth factor secretion by human retinal pigment epithelium and localiza- factor against degeneration of the retinal pigment epithelium and photorecep-
tion of vascular endothelial growth factor receptors on the inner tor in sodium iodate-injected rats. Curr Eye Res 2006;31:347–55.
414 choriocapillaris. Evidence for a trophic paracrine relation. Am J Pathol 1999; 214. Jin M, Chen Y, He S, et al. Hepatocyte growth factor and its role in the patho-
155:421–8. genesis of retinal detachment. Invest Ophthalmol Vis Sci 2004;45:323–9.
194. Dicou E, Nerriere V, Naud MC, et al. NGF involvement in ocular inflamma- 215. Gawad AE, Schlichting L, Strauss O, et al. Antiapoptotic properties of eryth-
tion: secretion by rat resident retinal cells. Neuroreport 1994;6:26–8. ropoietin: novel strategies for protection of retinal pigment epithelial cells.
195. Ishida K, Yoshimura N, Yoshida M, et al. Expression of neurotrophic factors Eye 2009;23:2245–50.
Section 1

in cultured human retinal pigment epithelial cells. Curr Eye Res 1997;16: 216. Wang ZY, Shen LJ, Tu L, et al. Erythropoietin protects retinal pigment epithe-
96–101. lial cells from oxidative damage. Free Radic Biol Med 2009;46:1032–41.
196. Hackett SF, Friedman Z, Freund J, et al. A splice variant of trkB and brain- 217. Xie Z, Wu X, Qiu Q, et al. Expression pattern of erythropoietin and erythro-
derived neurotrophic factor are co-expressed in retinal pigmented epithelial poietin receptor in experimental model of retinal detachment. Curr Eye Res
cells and promote differentiated characteristics. Brain Res 1998;789: 2007;32:757–64.
201–12. 218. He YG, Wang H, Zhao B, et al. Functional analysis of platelet-activating factor
197. Rosenthal R, Wohlleben H, Malek G, et al. Insulin-like growth factor-1 con- in the retinal pigment epithelial cells and choroidal endothelial cells. Curr Eye
tributes to neovascularization in age-related macular degeneration. Biochem Res 2009;34:957–65.
Anatomy and Physiology

Biophys Res Commun 2004;323:1203–8. 219. Shattuck RL, Wood LD, Jaffe GJ, et al. MGSA/GRO transcription is differen-
Basic Science and Translation to Therapy

198. Lambooij AC, van Wely KH, Lindenbergh-Kortleve DJ, et al. Insulin-like tially regulated in normal retinal pigment epithelial and melanoma cells.
growth factor-I and its receptor in neovascular age-related macular degenera- Mol Cell Biol 1994;14:791–802.
tion. Invest Ophthalmol Vis Sci 2003;44:2192–8. 220. Jaffe GJ, Richmond A, Van Le L, et al. Expression of three forms of melanoma
199. Weng CY, Kothary PC, Verkade AJ, et al. MAP kinase pathway is involved growth stimulating activity (MGSA)/gro in human retinal pigment epithelial
in IGF-1-stimulated proliferation of human retinal pigment epithelial cells cells. Invest Ophthalmol Vis Sci 1993;34:2776–85.
(hRPE). Curr Eye Res 2009;34:867–76. 221. Narayan S, Brun AM, Yorio T. Endothelin-1 distribution and basolateral secre-
200. Halapin NA, Bazan NG. PD1 induction of retinal pigment epithelial cell tion in the retinal pigment epithelium. Exp Eye Res 2004;79:11–9.
survival involves PI3K/Akt phosphorylation signaling. Neurochem Res 222. Narayan S, Prasanna G, Tchedre K, et al. Thrombin-induced endothelin-1
2010;35:1944–7. synthesis and secretion in retinal pigment epithelial cells is rho kinase depen-
201. Calandria JM, Bazan NG. Neuroprotectin d1 modulates the induction of pro- dent. J Ocul Pharmacol Ther 2010;26:389–97.
inflammatory signaling and promotes retinal pigment epithelial cell survival 223. Mascarelli F, Hecquet C, Guillonneau X, et al. Control of the intracellular
during oxidative stress. Adv Exp Med Biol 2010;664:663–70. signaling induced by fibroblast growth factors (FGF) over the proliferation
202. Bazan NG. Cell survival matters: docosahexaenoic acid signaling, neuropro- and survival of retinal pigment epithelium cells: example of the signaling
tection and photoreceptors. Trends Neurosci 2006;29:263–71. regulation of growth factors endogenous to the retina. J Soc Biol 2001;
203. Tan J, Deng ZH, Liu SZ, et al. TGF-beta2 in human retinal pigment epithelial 195:101–6.
cells: expression and secretion regulated by cholinergic signals in vitro. 224. Bryckaert M, Guillonneau X, Hecquet C, et al. Regulation of proliferation-
Curr Eye Res 2010;35:37–44. survival decisions is controlled by FGF1 secretion in retinal pigmented epi-
204. Yu AL, Fuchshofer R, Kook D, et al. Subtoxic oxidative stress induces senes- thelial cells. Oncogene 2000;19:4917–29.
cence in retinal pigment epithelial cells via TGF-beta release. Invest Ophthal- 225. Bryckaert M, Guillonneau X, Hecquet C, et al. Both FGF1 and bcl-x synthesis
mol Vis Sci 2009;50:926–35. are necessary for the reduction of apoptosis in retinal pigmented epithelial
205. Uchida H, Kuroki M, Shitama T, et al. Activation of TGF-beta1 through cells by FGF2: role of the extracellular signal-regulated kinase 2. Oncogene
up-regulation of TSP-1 by retinoic acid in retinal pigment epithelial cells. 1999;18:7584–93.
Curr Eye Res 2008;33:199–203. 226. Seidler DG, Schwegler JS, Liesenhoff H. Expression of fibroblast growth
206. Nagineni CN, Cherukuri KS, Kutty V, et al. Interferon-gamma differentially factors 1 and 2 in a human retinal pigment epithelium cell line (K1034).
regulates TGF-beta1 and TGF-beta2 expression in human retinal pigment Ophthalmic Res 1999;31:280–6.
epithelial cells through JAK-STAT pathway. J Cell Physiol 2007;210: 227. Zhu D, Wu J, Spee C, et al. BMP4 mediates oxidative stress-induced retinal
192–200. pigment epithelial cell senescence and is overexpressed in age-related macular
207. Crane IJ, Kuppner MC, McKillop-Smith S, et al. Cytokine regulation of degeneration. J Biol Chem 2009;284:9529–39.
granulocyte–macrophage colony-stimulating factor (GM-CSF) production by 228. Zhu D, Deng X, Xu J, et al. What determines the switch between atrophic and
human retinal pigment epithelial cells. Clin Exp Immunol 1999;115:288–9. neovascular forms of age related macular degeneration? The role of BMP4
208. Crane IJ, Wallace CA, Forrester JV. Regulation of granulocyte–macrophage induced senescence. Aging 2009;1:740–5.
colony-stimulating factor in human retinal pigment epithelial cells by IL-1beta 229. Benson MT, Shepherd L, Rees RC, et al. Production of interleukin-6 by human
and IFN-gamma. Cell Immunol 2001;209:132–9. retinal pigment epithelium in vitro and its regulation by other cytokines.
209. Fukuoka Y, Strainic M, Medof ME. Differential cytokine expression of human Curr Eye Res 1992;11:173–9.
retinal pigment epithelial cells in response to stimulation by C5a. Clin Exp 230. Chong DY, Boehlke CS, Zheng QD, et al. Interleukin-6 as a photoreceptor
Immunol 2003;131:248–53. neuroprotectant in an experimental model of retinal detachment. Invest
210. Yang D, Elner SG, Chen X, et al. MCP-1-activated monocytes induce apoptosis Ophthalmol Vis Sci 2008;49:3193–200.
in human retinal pigment epithelium. Invest Ophthalmol Vis Sci 2011;52: 231. Bian ZM, Elner SG, Yoshida A, et al. Differential involvement of phos-
6026–34. phoinositide 3-kinase/Akt in human RPE MCP-1 and IL-8 expression.
211. Pons M, Marin-Castaño ME. Cigarette smoke-related hydroquinone dysregu- Invest Ophthalmol Vis Sci 2004;45:1887–96.
lates MCP-1, VEGF and PEDF expression in retinal pigment epithelium in 232. Relvas LJ, Bouffioux C, Marcet B, et al. Extracellular nucleotides and
vitro and in vivo PLoS ONE 2011;6:e16722. interleukin-8 production by ARPE cells: potential role of danger signals in
212. Chu R, Zheng X, Chen D, et al. Blue light irradiation inhibits the production blood–retinal barrier activation. Invest Ophthalmol Vis Sci 2009;50:1241–6.
of HGF by human retinal pigment epithelium cells in vitro. Photochem 233. He S, Chen Y, Khankan R, et al. Connective tissue growth factor as a mediator
Photobiol 2006;82:1247–50. of intraocular fibrosis. Invest Ophthalmol Vis Sci 2008;49:4078–88.
Anatomy and Physiology Section 1

Cell Biology of the Müller Cell Chapter

Andreas Reichenbach, Andreas Bringmann


17 
astrocytes play a crucial role in retinal vascularization; the local-
INTRODUCTION ization of these cells in the mature retina is restricted to the nerve
In addition to microglial cells, which are the resident immune fiber and ganglion cell layers.
cells of the retina, the retina contains two types of macroglial
cell: astrocytes and Müller cells (Fig. 17.1A). Macroglial cells
MORPHOLOGY OF MÜLLER CELLS
support the functioning and metabolism of retinal neurons. For Heinrich Müller (1820−1864) in 1851 described the “radial fibers”
this reason, they constitute an anatomical and functional link of the retina which later became known as Müller cells. Müller
between these neurons and the compartments with which they cells are specialized radial glial cells which span the entire thick-
need to exchange molecules (blood vessels, vitreous chamber, ness of the neural retina, from the subretinal space to the vitreal
and subretinal space). Most nutrients, waste products, ions, surface (Fig. 17.1A, B). The somata of the cells are located in the
water, and other molecules are transported through macroglial inner nuclear layer, and two stem (trunk) processes radiate from
cells between the retinal vessels and neurons. As a peculiarity, the soma in opposite directions. The outer stem process draws

A B C
GCL

AG P
G BV
A G
IPL

MG
M
INL

A B A B
B H G
M
OPL

D
ONL

C R
PRS
RPE

Fig. 17.1 Müller cells (M) span the entire thickness of the neuroretina, and are arranged in a regular pattern. (A) Schematic drawing of the
cellular constituents of a human retina. The perikarya of Müller cells are localized in the inner nuclear layer (INL). The funnel-shaped endfeet of
Müller cells form the inner surface of the retina. In the outer (OPL) and inner plexiform layers (IPL), side branches which form perisynaptic
membrane sheaths originate at the stem processes. Both astrocytes (AG) and Müller cells contact the superficial blood vessels (BV) and the
inner surface of the retina. In the outer nuclear layer (ONL), the stem process of Müller cells forms membrane sheaths which envelop the
perikarya of rods (R) and cones (C). Microvilli of Müller cells extend into the subretinal space which surrounds the photoreceptor segments
(PRS). Microglial (MG) cells are located in both plexiform layers and the ganglion cell layer (GCL). A, amacrine cell; B, bipolar cell; G, ganglion
cell; H, horizontal cell; P, pericyte; RPE, retinal pigment epithelium. (B–D) Confocal images of living guinea pig retina preparations. (B) Radial
section, illustrating the tight package of the cellular elements shown in (A). The Müller cells are labeled with the vital dye, Mitotracker Orange
(green); synapses and the outer segments of photoreceptor cells are counterlabeled with another vital dye, FM-43. (C, D) Optical horizontal
sections through a flat-mounted retina, illustrating the regular pattern of Müller cell stem processes in the inner plexiform layer (D) and the almost
total occupation of the ganglion cell layer by the Müller cell endfeet (green; M); only the somata of the ganglion cells (G) appear “empty” (C).
towards the subretinal space; here, microvilli of Müller cells sur- form elaborate sheaths around the neuronal synapses, as well as
round the photoreceptor inner segments. Zonulae adherentes into the nuclear layers where they embed neuronal perikarya.
416 between Müller and photoreceptor cells form the outer limiting Astrocytes and Müller cells ensheath the vessels of the superfi-
membrane of the retina. The inner stem process projects towards cial vascular plexus; the deep capillaries are ensheathed by en
the vitread surface of the retina; the end of this process is passant endfeet of Müller cells. Within the macula, Müller cells
expanded into a funnel-shaped endfoot. Both the basement display a Z-shaped morphology because the outer processes
Section 1

membrane at the inner retinal surface and the Müller cell endfoot accompany the centrifugally running cone axons in the Henle
membranes together constitute the inner limiting membrane. fiber layer (Figs 17.2 and 17.3C). The central fovea contains 20–30
Lateral processes extend into the plexiform layers where they atypical Müller cells. These cells are not associated with cone

Fig. 17.2 Müller cells of the primate macula


Anatomy and Physiology
Basic Science and Translation to Therapy

are elongated and run in parallel with


A IPL the Henle fibers. (A) Scanning electron
microphotograph of the fovea of a macaque.
The extremely high packing density of
INL cone inner and outer segments in the
OPL photoreceptor segment layer (PSL), the
complex centrifugal course of their axons
crowded in the Henle fiber layer (HFL), and
HFL the absence of the ganglion cell (GCL), inner
plexiform (IPL), inner nuclear (INL), and outer
ONL plexiform layers (OPL) are illustrated. The
outer processes of Müller cells run among
the Henle fibers but cannot be reliably
identified. (B) When the Müller cell
PSL processes are visualized by vimentin
immunohistochemistry (in brown;
counterstaining of cell nuclei by hematoxylin
10 µm
and eosin; siamang retina), they can be
RPE traced along their path from the outer margin
of the outer nuclear layer (ONL) up to their
endfeet; it becomes obvious that they run in
B parallel with the Henle fibers.
GCL

IPL

INL
OPL
HFL
ONL

25 µm PSL
RPE

Fig. 17.3 Müller cells form the cores of radial


units of retinal neurons. Every Müller cell
A B C is surrounded by a distinct group of retinal
neurons (A) with which it interacts specifically
during development, in the mature
functioning retina, and after injury of the
retina. These repetitive radial units are
almost identical throughout most regions of
the retina (B) but differ in the (peri-)fovea
(C) by the lack of rods, an increased number
of neurons of the inner nuclear and ganglion
cell layers, and by an elongation and
Z-shaped course of the outer processes of
Müller cells (cf. also Fig. 17.2). Blue, Müller
cells; yellow, retinal neurons; orange, cones;
brown, rods.
axons; instead, their outer process runs straight towards the photoreceptors and neurons, are responsible for the structural
inner retinal surface where the soma is located and where their stabilization of the retina, and are modulators of immune
large, flat endfeet contribute to the inner limiting membrane of and inflammatory responses. They guide the light to the pho- 417
the fovea proper.1 toreceptors and buffer mechanical tissue deformations. Müller
cells become activated upon virtually all pathogenic stimuli.
Müller cells constitute the cores of

Chapter 17
Reactive Müller cells are neuroprotective but may also stop
functional retinal columns supporting the neurons, and rather contribute to neuronal
The human retina contains 8–10 million regularly arranged degeneration. Currently, the many roles of Müller cells in the
Müller cells (Figs 17.1D and 17.4C). Each Müller cell constitutes regulation of retinal function are still not fully resolved, and
the core of a column of retinal neurons which represents the are the subject of ongoing intensive research. Noteworthy,
smallest functional unit for “forward information processing.”2 much of the current knowledge about Müller cell (dys-)func-
Such a column contains one cone per Müller cell (Figs 17.3B, C tion was obtained in animal models and thus awaits confirma-

Cell Biology of the Müller Cell


and 17.4B, C) and up to 10 rods, as well as 6 (fovea) and 4 tion on human cells.
(periphery) inner nuclear layer neurons, and 2.5 (fovea) and 0.3
(periphery) ganglion cell layer neurons, respectively.1 Light guidance
Müller cells interact with the neurons of their columns in Because the retina is inverted, light has to pass the entire depth
a kind of symbiotic relationship. They are responsible for all of the neuroretina before it arrives at the photoreceptors. In the
functional and metabolic support of their associated neurons. retina as in any other organ, all cells and their processes and
Müller cells provide trophic substances to neurons, and remove organelles are phase objects which scatter the incoming light. In
metabolic waste. They play a critical role in regulation of the particular, the synapses in the plexiform layers have dimensions
extracellular space volume, ion and water homeostasis, and close to 500 nm, i.e., within the wavelength range of visible light;
in the maintenance of the inner blood–retinal barrier. They this makes them light-scattering structures.3 The backscattering
release gliotransmitters and other neuroactive substances, and of light from the retinal cell structures is clinically used by optical
impact synaptic activity by neurotransmitter recycling which coherence tomography. However, light scattering should reduce
involves the supply of neurons with precursors of neurotrans- visual sensitivity and acuity, particularly outside the fovea
mitters. All of these functions directly or indirectly modify (where the inner layers are shifted aside, and the light directly
the neuronal activity. Müller cells support the survival of hits the cones).

Fig. 17.4 Müller cells are “optical fibers”


guiding the incoming light to the
A B photoreceptor cells. (A) Artist’s view of the
optical path through the retina. Light arrives
at the inner retinal surface where it hits
the Müller cell endfeet. Then it propagates
through the Müller cell stem processes
(thus bypassing the light-scattering retinal
structures, particularly in the two plexiform
layers) until it arrives at the photoreceptor
cells. Every Müller cell illuminates a distinct
group of photoreceptors, about 10 rods and
one cone. (B, C) Vertical (B) and horizontal
(C) sections through the guinea pig retina.
Müller cells were immunolabeled for
vimentin (green), and cones were labeled by
peanut agglutinin (red). (B) The cone inner
segments in the photoreceptor segment
ONL layer (RSL) appear continuous with the
outer processes of Müller cells which
envelop the cone perikarya in the outer
nuclear layer (ONL). (C) The focus of the
image is on the outer plexiform layer (OPL).
The number of cone pedicles (red) roughly
RSL
equals that of Müller cell processes (green);
often the two elements appear as pairs.
Thus, each cone receives its part of the
image of the outside world by its “individual”
light-guiding Müller cell.

C
To reduce light scattering, nonfoveal Müller cells act as living generates an inward current (the transporter is “electrogenic”;
optical fibers that guide the light through the inner retinal layers Figs 17.6A and 17.7A, B) and cellular depolarization. The ampli-
418 towards the photoreceptors (Fig. 17.4A).3 The funnel-shaped tude of the glutamate transporter currents is voltage-dependent
endfeet of Müller cells act as light collectors at the vitread surface (Fig. 17.6A); a very negative membrane potential is essential for
of the retina. The refractory index of Müller cells is slightly lower an efficient uptake of glutamate.17 Sodium-dependent carriers
than that of photoreceptor segments (which are also light- allow uphill transport of substrates into the cells against a con-
Section 1

guiding fibers) but higher than that of the surrounding retinal centration gradient. The driving force for the transport is the
tissue.3 On the other hand, the refractory index of Müller cell electrochemical sodium gradient across the plasma membrane,
endfeet is rather low, about halfway between that of Müller cell generated by the Na+,K+-ATPase. Other transporters, e.g., for
stem processes and that of the vitreous.3 This allows a “soft GABA, are also electrogenic (Figs 17.6B and 17.7F). The subcel-
coupling” of the light path between the vitreous and the retina, lular distribution of the GABA transporter currents (Fig. 17.6B)
and reduces light reflection at the inner retinal surface. By light corresponds with the observation that GABA is taken up by
Anatomy and Physiology
Basic Science and Translation to Therapy

guidance, Müller cells transport an image (like a fiberoptic plate) amacrine and Müller cells in the inner retina but exclusively by
with minimal loss of light intensity from the inner retinal surface Müller cells in the outer retina.18
to the photoreceptors; this image is resolved in pixels corre- In the outer plexiform layer, the photoreceptor-derived gluta-
sponding to individual Müller cells (Fig. 17.4A). Because the mate is mainly removed by photoreceptor, horizontal, and
local densities of cones and Müller cells are roughly equal (Fig. bipolar cells.12,19 whereas Müller cells prevent the lateral spread
17.4C), every cone has its “private” Müller cell which delivers of neurotransmitters beyond the synapse(s) where they are
its part of the image, while several (up to 10) rods are illuminated released, thus ensuring visual resolution (Fig. 17.5). In the inner
by one Müller cell (Fig. 17.4A). plexiform layer, Müller cell-provided glutamate uptake takes a
more active part in shaping the synaptic responses. The rapid
Recycling of cone photopigments termination of synaptic glutamate action in nonspiking inner
retinal neurons and in retinal ganglion cells is predominantly
Müller cells support the function of photoreceptors by other
mediated by the uptake of glutamate into Müller cells.20,21 Gener-
mechanisms, as well. There exist two cycles in the retina that
ally, Müller cells remove the bulk of extracellular glutamate in
regenerate the photopigments, the rod and the cone visual cycle.4
the inner retina.13,22,23
In the photoreceptor outer segments, all-trans retinal is reduced
to all-trans retinol. Rod-derived all-trans retinol is regenerated to
11-cis retinal in the pigment epithelium while cone-derived all- Malfunction of glial glutamate uptake
trans retinol is processed by Müller cells. Müller cells convert contributes to glutamate toxicity
all-trans retinol to 11-cis retinol which is subsequently oxidized
Glutamate toxicity is a major cause of neuronal loss in many
to 11-cis retinal by a retinol dehydrogenase, and released into
retinal disorders, including glaucoma, ischemia, diabetes, and
the extracellular space for uptake by the cone photoreceptors.
inherited photoreceptor degeneration. A malfunction and/or
For the transport of the retinoids, Müller cells express cellular
downregulation of GLAST may cause, or contribute to, a rise in
retinol-binding protein and cellular retinal-binding protein
extracellular glutamate towards excitotoxic levels.23–25 Hypoxia
(CRALBP).5 Furthermore, Müller cells contribute to the assembly
facilitates the formation of free radicals in mitochondria; the
of the photoreceptor outer segments,6 in part by the release of
lactose and pigment epithelium-derived factor (PEDF),7,8 and resulting lipid peroxidation disrupts glutamate transport into
phagocytose outer-segment discs shed from cones.9 Docosa- Müller cells.26 This mechanism is probably implicated in the
neuronal dysfunction in diabetic retinopathy27 and in ganglion
hexaenoic acid is required for outer-segment renewal and for the
cell death in Leber hereditary optic neuropathy.28 Inflammatory
preservation of mitochondria.10 Müller cells incorporate docosa-
lipids such as arachidonic acid, a reduction in extracellular pH
hexaenoic acid into phospholipids, and channel it to the
as occurs in ischemia, and zinc ions released from photorecep-
photoreceptors.10
tors29 all inhibit glial glutamate uptake.30,31
Because glutamate transport is voltage-dependent (Fig. 17.6A),
Regulation of the synaptic activity by depolarization decreases the efficiency of glial glutamate
neurotransmitter uptake uptake.17,32 Depolarization of Müller cells occurs in response to
Precise shaping (i.e., control in time and space) of synaptic activ- pathological rises in extracellular potassium (as occurs in isch-
ity depends upon the kinetics of the presynaptic neurotransmit- emia and glaucoma), to the opening of cation channels in Müller
ter release and of the (re-)uptake of the transmitter molecules cells (Fig. 17.7C), and to a decrease in the potassium permeability
into the cells. In the neuroretina, photoreceptor cells, neurons, of glial membranes due to inactivation and/or downregulation
and macroglial cells express high-affinity transporters for neu- of inwardly rectifying potassium (Kir) channels (Fig. 17.8A, B).
rotransmitters. Müller cells possess uptake and exchange systems An inactivation of Kir channels was observed in animal models
for various neurotransmitters, particularly for glutamate and of various retinopathies (see below). Depolarization of Müller
γ-aminobutyric acid (GABA) (Fig. 17.5). cells can also be evoked by inflammatory lipids such as arachi-
The major glutamate uptake carrier of Müller cells is the glu- donic acid and prostaglandins which potently inhibit Na+,K+-
tamate aspartate transporter (GLAST, or excitatory amino acid ATPase. Depolarization of Müller cells may even cause a reversal
transporter-1, EAAT1).11–13 In addition, EAAT2 (GLT1) and of the glutamate transport.33 A nonvesicular release of glutamate
EAAT3 were found in human Müller cells.14,15 EAATs mediate and aspartate via reversed operation of glial glutamate trans-
the cotransport of three sodium ions and one proton, and the porters was implicated in the excitotoxic damage of retinal gan-
countertransport of one potassium ion, with each glutamate glion cells.34,35 A release of glial glutamate might also be mediated
anion.16 The transport of an excess of sodium into the cell by the cystine glutamate antiporter (which is activated under
Müller cell
Cystine Cysteine
CyT
Glutamate γ-glutamyl-cysteine Glycine
CGA γ-glutamyl-cysteine 419
Glutamine synthetase GSH synthetase
H 2O GSH
GLAST

Chapter 17
PAG
NH4+ R˙ GP GR NADPH
2 Na+
Glutamate 1 Cl– GSSG
GAT
GAD GABA
EAAC1
α-KG
GLT1
GABA-T
GABA TCA

Cell Biology of the Müller Cell


1 K+
BC BC HC NH4+
EAAT5 GLAST NADPH SSA
GLT1
3 Na+ GDH
EAAT5 1 H+ NADPH
GLT1 Glutamate
α-KG
NH4+ Glucose OAA
PC AAT
TCA
ATP GS Pyruvate
Aspartate
LDH
Glutamate Lactate ALAT
Glutamine AGC
+
NH4 Alanine
PAG Glutamate MAS
H2O Malate
α-KG
Glutamine OAA

Fig. 17.5 Recycling of amino acid neurotransmitters in the outer plexiform layer. The ribbon synapse of a photoreceptor cell (PC) synthesizes
glutamate which is released in the dark. The postsynaptic elements are dendrites of bipolar (BC) and horizontal cells (HC). Horizontal cells
release γ-aminobutyric acid (GABA) which is formed from glutamate. The synaptic complexes are surrounded by Müller cell sheets. The right
side shows neurotransmitter uptake systems and some metabolic pathways within Müller cells. Glutamate, GABA, and ammonia (NH4+) are
transported into Müller cells and transformed to glutamine, alanine, and α-ketoglutarate (α-KG). These products are released from Müller
cells, and taken up by neurons. Glutamine serves as a precursor for the transmitter synthesis in neurons (glutamate–glutamine cycle). Lactate,
alanine, α-ketoglutarate, and glutamine are utilized by neurons as substrates for their energy metabolism. Glutamate is also used for
the production of glutathione (GSH), which is an antioxidant, released from Müller cells and taken up by neurons under oxidative stress
conditions. ALAT, alanine aminotransferase; AAT, aspartate aminotransferase; CGA, cystine-glutamate antiporter; CyT, cystine transporter;
EAAC1, excitatory amino acid carrier 1; EAAT5, excitatory amino acid transporter 5; GABA-T, GABA transaminase; GAD, glutamic acid
decarboxylase; AGC, aspartate-glutamate carrier; GAT, GABA transporter; GDH, glutamate dehydrogenase; GLAST, glutamate-aspartate
transporter; GLT-1, glutamate transporter-1; GlyT, glycine transporter; GP, glutathione peroxidase; GR, glutathione reductase; GS, glutamine
synthetase; GSH, glutathione; GSSG, glutathione disulfide; LDH, lactate dehydrogenase; MAS, malate-aspartate shuttle; OAA, oxaloacetate;
PAG, phosphate-activated glutaminase; R•, free radicals; SSA, succinate semialdehyde; TCA, tricarboxylic acid cycle.

A B
Glutamate Membrane potential (mV) GABA
–80 –40 0 +40
+40
Membrane potential (mV)

0
Relative current amplitude

–0.5
–40
5 pA
–80
1s
–1

1 min 20 pA
0 0.5 1
Relative current

Fig. 17.6 The main glutamate and γ-aminobutyric acid (GABA) uptake carriers of Müller cells are electrogenic transporters. The membrane
currents were recorded in whole Müller cells. (A) Administration of glutamate (1 mM) to a rabbit Müller cell evokes inward currents at negative
membrane potentials (left). The current-voltage relation of the glutamate transporter currents in guinea pig Müller cells (right) shows that the
efficiency of the glutamate transport increases with increasing (i.e., more negative) membrane potentials. (B) Subcellular distribution of the GABA
transporter currents in a guinea pig Müller cell. The distribution of the currents was determined by focal ejections of GABA (1 mM) on to the
following membrane domains of the cell: endfoot, inner stem process, soma, and inner and outer parts of the outer stem process.
A Glutamate Kainate NMDA C ATP BzATP
420

0.1
Section 1

nA

10 s 30 pA 30 s

Current at
BK current

+120 mV
Anatomy and Physiology
Basic Science and Translation to Therapy

–60 mV
B –100 mV cation
Glutamate ATP
current

1 P2Y receptor P2X7 receptor


nA activation activation

1 min D

Current at

+120 mV

–60 mV
–100 mV

E F
1 min Glutamate GABA
0.3
Open probability

0.2

0.1

0
10 pA
–C 10 pA 30 s
–1 250 ms

Fig. 17.7 Neurotransmitters modify the membrane conductance of Müller cells. (A) Administration of glutamate (100 µM) to a rat Müller cell evoked
inward currents through electrogenic glutamate transporters, while the glutamate receptor agonists, kainate (500 µM) and N-methyl-D-aspartate
(NMDA) (100 µM, in the presence of 10 µM glycine and absence of magnesium) did not evoke membrane currents. This suggests that rat Müller
cells express metabotropic but no functional ionotropic glutamate receptors. The current traces were recorded in whole-cell records at
a membrane potential of –80 mV. (B) Time-dependent record of the whole-cell currents in a human Müller cell. The currents at +120 mV were
mainly mediated by big-conductance potassium (BK) channels. The BK currents were transiently increased in response to extracellular adenosine
triphosphate (ATP) (500 µM). Extracellular glutamate (500 µM) evoked a delayed transient increase in BK currents. The increase in the currents
at –60 and –100 mV during the exposure of glutamate (arrow) reflects the activation of the electrogenic glutamate transporters. The arrowheads
indicate transient activation of calcium-evoked cation channels. (C) Activation of metabotropic P2Y and ionotropic P2X7 receptors in a human
Müller cell. Activation of P2Y receptors by ATP (100 µM) evoked repetitive transient calcium-evoked activation of BK currents (that were recorded
at the potential of +120 mV; left). Activation of P2X7 receptors by 2’-/3’-O-(4-benzoylbenzoyl)-ATP (BzATP; 50 µM) evoked a sustained calcium-
evoked activation of BK currents, as well as cation currents through P2X7 receptor channels (right). (D) Immunostaining of two isolated human
Müller cells against the P2X7 receptor protein using two different antibodies. Arrows, perikarya; arrowheads, cell endfeet. Bars, 20 µm.
(E) Extracellular glutamate (200 µM) increased the activity of a single BK channel that was recorded in a membrane patch localized at the
perikaryum of a human Müller cell. Above, Time dependency of the opening probability of the channel. Below, Original records of the channel
activity. The pipette potential was 0 mV (i.e., the recording was made near the resting membrane potential of the cell). C, closed-state current level;
1, open-state current level. (F) γ-aminobutyric acid (GABA) (100 µM) evoked two currents in a human Müller cell, a transient, rapidly inactivating
current mediated by GABAA receptor channels (arrow), and a sustained current mediated by electrogenic GABA transporters (asterisk).
A
Control 3 days after 7 days after 421
ischemia ischemia
+20

Chapter 17
–80 mV
–160

1
nA
50 ms
Kir

Cell Biology of the Müller Cell


B C
–90 120

Hypo-osmolarity

–80
RMP
Relative current

Soma area (%)


Kir 110
RMP (mV)
–70
Ischemia
Relative
capacitance
–60 100

Healthy control
–50
Control 3h 1d 2d 3d 0 2 4 min

Time after reperfusion

Fig. 17.8 In the course of retinal ischemia–reperfusion, Müller cells of the rat change their membrane conductance(s) and their osmotic swelling
properties. Transient retinal ischemia was induced by elevation of intraocular pressure above systolic blood pressure for 1 hour. (A) Examples of
the whole-cell potassium currents of Müller cells isolated from a control retina, and from retinas 3 and 7 days after transient retinal ischemia.
(B) The amplitude of the Kir currents decreases time-dependently after transient retinal ischemia. Simultaneously, the resting membrane potential
(RMP) of the cells decreases and the whole-cell capacitance (that is proportional to the cell membrane area) increases, indicating cellular
hypertrophy. The parameters were measured at different periods (3 hours to 3 days) after transient ischemia. (C) Under hypo-osmotic stress,
Müller cells in slices of retinas 3 days postischemia display cellular swelling which is not observed in Müller cells in slices of control retinas. The
time-dependent alteration in the cross-sectional area of Müller cell somata is shown. The images display Müller cell somata in a postischemic
retina, before (above) and after (below) hypotonic challenge. Bar, 5 µm.

oxidative stress conditions when an elevated production of glu- Glutamine is released from Müller cells, and taken up by neurons
tathione requires an increased uptake of cystine; Fig. 17.5) and as a precursor for the resynthesis of glutamate and GABA
by exocytosis of glutamate-containing secretory vesicles.1,36 (glutamate–glutamine cycle; Fig. 17.5).39 In the photoreceptor
Under various pathological conditions including ischemia, glau- cells, this cycle accounts only for a part of glutamate supply; they
coma, retinal detachment, and proliferative retinopathies, an directly take up glutamate from the synaptic cleft, and, in addi-
enhanced expression or activity of GLAST was observed.11,37 This tion, synthesize glutamate from α-ketoglutarate as well as from
might counterbalance the depolarization-induced malfunction Müller cell-derived glutamine.19,40 By contrast, the production of
of glial glutamate uptake. glutamate in bipolar and ganglion cells almost entirely depends
on Müller cell-derived glutamine.40 Pharmacological blockade of
Production of neurotransmitter precursors the glutamine synthetase in Müller cells causes a rapid loss of
To sustain the mechanism of transmitter recycling at the syn- the glutamate content of bipolar and ganglion cells, and (within
apses, the Müller cells are endowed with specific enzymes. After 2 minutes) the animals become functionally blind.40 Likewise, a
GABA has been taken up by Müller cells, it is readily converted significant amount of GABA in amacrine cells is synthesized
to glutamate by the GABA transaminase, and glutamate is con- from Müller cell-derived glutamine.40
verted to glutamine by the glutamine synthetase (Fig. 17.5). Glu- Downregulation of the glial glutamine synthetase contributes
tamine synthetase is exclusively localized to glial cells.38 to neuronal dysfunction and glutamate toxicity in inherited
photoreceptor degeneration and retinal detachment.41,42 The Removal of carbon dioxide
downregulation is mediated (at least in part) by the basic fibro-
Photoreceptor cells display the highest rate of oxidative metab­
422 blast growth factor (bFGF).43 bFGF is rapidly released within the
olism of all cells in the body. This high metabolic activity is
retina after detachment, and its expression level increases under
associated with high oxygen and glucose demands. The active
ischemic conditions, after light injury, and in response to inher-
glucose metabolism, in turn, generates much carbon dioxide and
ited photoreceptor degeneration and mechanical injury.1 Though
Section 1

water. Via transcellular transport, Müller cells redistribute meta-


bFGF is a neurotrophic factor which supports the survival of
bolic waste into the blood and vitreous. Carbon dioxide is cleared
photoreceptors and neurons,44–46 the bFGF-induced downregula-
by a process called carbon dioxide siphoning57; it is rapidly
tion of glutamine synthetase might rather aggravate neuronal
hydrated to bicarbonate and protons by carbonic anhydrases.58
degeneration. Under ischemic conditions, the GABA transami-
Carbonic anhydrase II is localized within Müller and amacrine
nase activity is decreased, and GABA accumulates in Müller
cells, whereas the membrane-bound carbonic anhydrase XIV is
cells.47,48 This impairs the efficiency of glial GABA uptake due to
Anatomy and Physiology
Basic Science and Translation to Therapy

present in glial cells and vascular endothelia.57,59,60 Bicarbonate is


the decrease in the extra- to intracellular gradient as a driving
transported to the blood and vitreous by sodium bicarbonate
force.
cotransporters and anion exchangers, both enriched in glial
endfoot membranes.57
Trophic support of photoreceptors
and neurons Regulation of the extracellular pH
The uptake of glutamate and GABA by Müller cells links neu- By the removal of carbon dioxide, Müller cells also regulate the
ronal excitation with the release of metabolic substrates, as extracellular pH. Light-evoked neuronal activity is associated
well as with the defense against oxidative stress. Müller cells with extracellular alkalinization.61 The pH shift is balanced by
produce various substrates of the oxidative neuronal metab­ an efflux of protons from photoreceptors62 and by the consecu-
olism such as glutamine, lactate, alanine, and α-ketoglutarate tive action of carbonic anhydrases and glial sodium bicarbonate
(Fig. 17.5)49; these substrates are used by photoreceptors and cotransporters.63 Because sodium bicarbonate transporters are
neurons in periods of metabolic stress in the dark. The meta- electrogenic, depolarization of Müller cells by potassium released
bolic support is regulated by neuron-derived glutamate (which from active neurons causes an influx of bicarbonate into the
stimulates the uptake of glucose and the production of lactate) cells.63 The carbonic anhydrase-generated protons remain
and potassium (which induces hydrolyzation of glycogen in outside the cells and cause an extracellular acidification.63
Müller cells).1,50 Because extracellular acidification inhibits synaptic transmis-
sion,64 Müller cell-mediated pH regulation may protect neurons
Antioxidative support of photoreceptors against overexcitation.
and neurons
Glutamate in Müller cells is also utilized for the production of Spatial potassium buffering
the antioxidant, glutathione (Fig. 17.5).39 The retina has a high Neuronal activity is associated with rapid ion shifts between the
need of antioxidant protection; this results from light exposure intra- and extracellular spaces. Sodium, chloride, and calcium
together with high oxygen consumption and the presence of ions flow into active neurons, and potassium ions are released
high polyunsaturated fatty acid levels in photoreceptors. In from neurons. Light onset causes increases in extracellular potas-
addition, the outer retina underlies circadian periods of hypoxia sium in the plexiform layers, and a decrease in the subretinal
(dark) and hyperoxia (light)51 which both increase oxidative space.65,66 If not corrected, increased potassium will cause neu-
stress. Usually, retinal glutathione is confined to glial and hori- ronal depolarization and hyperexcitation. Müller cells buffer
zontal cells.39,52 In Müller cells, glutathione constitutes about 2% imbalances in the extracellular potassium concentration via per-
of the total protein.1 In response to oxidative stress such as mission of transcellular potassium currents, a process termed
during ischemia, glutathione is rapidly released from Müller “spatial potassium buffering” or “potassium siphoning.”67,68
cells and provided to the neurons,52 where it acts as cofactor of They take up excess potassium from the extracellular fluid, and
the enzyme glutathione peroxidase. The glial release of antioxi- release equal amounts of potassium into fluid-filled spaces
dants like glutathione, and of neuroprotective factors like bFGF outside the neuroretina where the potassium concentration is
and adenosine, is implicated in the protection of photoreceptors constant or decreased, i.e., into blood vessels, the vitreous, and
from the harmful effects of circadian light exposure. the subretinal space (Fig. 17.9C).
Müller cells from aged animals contain reduced levels of Because Müller cell membranes are highly permeable to potas-
glutathione; this is associated with mitochondrial damage, mem- sium, Müller cells have a very negative resting membrane po-
brane depolarization, and reduced cell viability.53 An age- tential, close to the potassium equilibrium potential, around
dependent decrease in retinal glutathione may accelerate the –80 mV.69,70 The potassium permeability is mainly mediated by
pathogenesis of retinopathies in the elderly. In diabetic retin­ special potassium channels belonging to the Kir subfamily of K+
opathy, the decreased glutamate uptake of Müller cells results channels.71 In particular, Kir4.1 channels (which mediate bidirec-
in reduced glutathione synthesis54,55 and in an upregulation of tional potassium currents; Fig. 17.9A) and Kir2.1 channels (which
glutaredoxin which catalyzes the deglutathionylation of pro- mediate inward potassium currents) are implicated in potassium
teins; the latter mechanism induces nuclear translocation of siphoning.72,73 The channel proteins are localized in a polarized
nuclear factor-κB and upregulation of proinflammatory factors.56 fashion in the Müller cell membrane (Fig. 17.9B). Homomeric
Other antioxidants provided by Müller cells include pyruvate, Kir4.1 channels are mainly localized in membrane domains
α-ketoglutarate, metallothionein, lysozyme, ceruloplasmin, across which the cells extrude potassium into spaces outside the
heme oxygenase, and reduced ascorbate.1 neuroretina, i.e., in perivascular and vitread endfoot membranes,
A B C
Kir4.1 Vitreous body
Kir4.1 I 423

GCL GCL

Chapter 17
IPL
IPL
–150 –50 V
INL
INL
K+
ONL
ONL IPL K+

Cell Biology of the Müller Cell


Kir2.1 I
K+

Kir2.1
–150 –50 V
GCL
K+
IPL GCL OPL
IPL
INL Kir4.1
TASK I INL
Kir2.1 +
Kir4.1/5.1
ONL
ONL

–150 –50 V
Control Ischemia Subretinal space

Fig. 17.9 The subcellular localization of Kir channel subtypes determines the direction of spatial buffering potassium currents through Müller
cells. (A) Current-voltage (I-V) relations of Kir4.1 and Kir2.1 channels. Kir4.1 channels mediate inward and outward potassium currents with
similar amplitudes, whereas Kir2.1 channels mediate inward currents but almost no outward currents. (B) Immunolocalization of glial Kir channel
proteins in the normal and postischemic rat retina. The Kir4.1 protein is predominantly localized at the limiting membranes of the neuroretina
(arrowheads) and around the blood vessels (arrows). The Kir2.1 protein is localized in the inner retina in membrane domains of Müller cells that
abut neuronal compartments such as processes that traverse the inner plexiform layer (IPL) (arrowheads). Seven days after a 1-hour transient
retinal ischemia, the expression of Kir4.1 protein is largely downregulated whereas the localization of the Kir2.1 protein is not altered. Note the
decrease in the thickness of the inner retina which is a characteristic feature of retinal ischemia–reperfusion injury. (C) Schematic drawing of the
potassium buffering currents flowing through Müller cells during neuronal activation. Activated neurons release potassium which is absorbed by
Müller cells through Kir2.1 and Kir5.1/4.1 channels, and distributed into the blood vessels, the vitreous, and the subretinal space through Kir4.1
channels. Kir4.1 channels mediate in- and outward currents and, thus, contribute to the osmohomeostasis between the neuroretina and
extraretinal fluid-filled spaces. GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer; OPL, outer plexiform layer.
Bars, 20 µm.

and in the microvilli that extend into the subretinal space.72,74 channels.72 Kir4.1 channels are also involved in cell volume regu-
Membranes which abut neuronal cell structures contain Kir lation which compensates for changes in the osmotic condi-
channels which mediate inward potassium currents, i.e., Kir2.1 tions.1,77 Under various pathological conditions, Kir4.1 channels
(Fig. 17.9B) and heterotetrameric Kir4.1/Kir5.1 channels.73,75 of Müller cells are downregulated and/or dislocated (Fig. 17.9B);
Thus, the direction of the transcellular potassium currents is this is associated with functional inactivation and results in a
determined by the subcellular localization of different Kir decrease of the potassium currents through Müller cells (Fig.
channel subtypes. Local increases in extracellular potassium 17.8A, B). Such alterations have been observed in animal
cause the driving force for passive currents through Kir models of various ischemic-hypoxic and inflammatory diseases,
channels. Müller cells also express other types of potassium including ischemia-reperfusion, ocular inflammation, diabetic
channels which may contribute to potassium buffering, includ- retinopathy, blue-light injury, retinal detachment, retinal vein
ing voltage-gated potassium (KA, KDR), calcium-activated big- occlusion, and proliferative vitreoretinopathy, as well as in
conductance potassium (BK) channels (Figs 17.7E and 17.10A), Müller cells from patients with proliferative retinopathies (Fig.
and tandem-pore domain potassium channels.76 In addition, the 17.10A).1,77 The impaired transcellular potassium transport and
Na+,K+-ATPase and transporter molecules, e.g., the K/Na/2Cl the depolarization of the cells (which impairs the glutamate
cotransporter, contribute to Müller cell-mediated potassium uptake) contribute to neuronal hyperexcitation and glutamate
homeostasis.1 toxicity.32 Human Müller cells display an age-dependent decrease
Many of the homeostatic functions of Müller cells, including in Kir currents, on average by 50% between the ages of 40 and
potassium siphoning and neurotransmitter uptake, depend on 80 years (Fig. 17.10C). This may enhance the susceptibility for
the very negative membrane potential constituted by the Kir4.1 age-dependent retinopathies because dysfunctional Müller cells
A Postmortem donor Proliferative vitreoretinopathy
424
BK
Section 1

Inward/Outward currents

+160

KA
KDR –80 mV
–160

50 ms 0.5
Anatomy and Physiology
Basic Science and Translation to Therapy

Kir nA

Postmortem donor Proliferative diabetic retinopathy

+80

–80 mV
–160

Kir 0.5
50 ms
nA

B C

0
Membrane potential (mV)

Relative current density

Kir
–50

VGCC
–100

150 ms 30 40 50 60 70 80
Age (years)

Fig. 17.10 Age- and disease-related alterations in the membrane conductance of human Müller cells. (A) Four different types of potassium
currents can be recorded in Müller cells from postmortem donors without apparent eye diseases (left): BK, currents mediated by big-conductance
potassium channels; KA, fast transient (A-type) potassium currents; KDR, delayed rectifying potassium currents; Kir, inwardly rectifying potassium
currents. Müller cells obtained from patients with proliferative vitreo- and diabetic retinopathies (right) display an almost complete absence of Kir
currents. The inward potassium currents (evoked by membrane hyperpolarization from a holding potential of –80 mV) are depicted downwardly.
The outward potassium currents (evoked by membrane depolarization) are depicted upwardly. The arrow indicates transient inward currents
through voltage-gated sodium channels. (B) Single-action potential-like discharges (arrow) can be evoked in the current-clamp mode
by large depolarizing current steps in a Müller cell of a patient with proliferative vitreoretinopathy. Depolarizing currents from 40 to 200 pA
(increment, 20 pA) were applied after administration of a hyperpolarizing current of 250 pA (resulting in a membrane potential between –100 and
–125 mV). (C) Age-dependent alterations in the densities of Kir and L-type voltage-gated calcium channel (VGCC) currents in human Müller
cells. While the Kir currents display an age-dependent decrease, the currents through L-type calcium channels increase in the course of aging.
cannot efficiently buffer pathological complications such as
Vitreous
neuronal hyperexcitation due to hypoxia caused by diabetic AQP4 Kir4.1
alterations of the vessels. 425

Water clearance

Chapter 17
Usually, water accumulates in the retinal tissue due to various
mechanisms, including a water influx from the blood coupled to
the glucose uptake, and metabolic water production by the oxi-
dative degradation of glucose. The accumulation of metabolic Inner
water is especially abundant in macular tissue which displays a retina
high metabolic activity and a high density of cells. This generates AQP4

Cell Biology of the Müller Cell


the necessity for a substantial constitutive efflux of water out of
the retina into the blood. In addition, the transmembrane ion
shifts associated with neuronal activity (in particular, with iono-
Kir4.1
tropic glutamatergic signaling) needs, for osmotic reasons, to be
buffered by rapid water transport across glial membranes.
Retinal water clearance is mediated by an osmotically driven
transcellular water transport coupled to transport of osmolytes,
and in particular to potassium transport.1,74 The Müller cell-
mediated clearance of the retinal tissue from excess potassium
will simultaneously redistribute metabolic water out of the neu-
roretina. The transmembrane water transport is facilitated by
water-selective channels, the aquaporins. Aquaporin-4 is mainly
localized in glial membranes within the inner retina and the
Subretinal
outer plexiform layer,74 while aquaporin-1 is present in photore- space
ceptor and pigment epithelial cells (Fig. 17.11).1,78 In Müller cell
membranes that contact fluid-filled spaces outside the neuro-
RPE AQP1
retina, aquaporin-4 (Fig. 17.11) and Kir4.1 channels (Fig. 17.9B)
are colocalized.74 This suggests that osmotic gradients between
the neuroretina on the one hand and the blood, vitreal fluid, and Choroidea
subretinal fluid on the other hand are compensated by bidirec-
tional potassium and water flux across Müller cell membranes.
Fig. 17.11 Müller cells mediate the osmohomeostasis predominantly
In addition, Müller cells express aquaporin-4 in their perisynap- of the inner retina. Water fluxes through the retina are shown. Under
tic membranes within the plexiform layers. Because the plexi- normal conditions, water accumulates in the neural retina and
form layers are high-resistance barriers for paracellular fluid subretinal space (blue arrows) due to an influx from the blood
(coupled to the uptake of nutrients such as glucose) and vitreous
movement,79 any water flowing into the neurons (associated chamber (due to the intraocular pressure), and due to the oxidative
with ion flux through ionotropic receptors) will be delivered synthesis of adenosine 5’-triphosphate (ATP) in the mitochondria that
from Müller cells through aquaporin-4.1 Aquaporin-4 knockout generates carbon dioxide and water. The excess water is redistributed
mice display reduced electroretinogram b-waves,80 suggesting into the blood by a transcellular water transport through Müller cells
and the retinal pigment epithelium (RPE). The water transport across
that Müller cell-mediated water transport is implicated in the cell membranes is facilitated by aquaporin (AQP) water channels.
maintenance of regular neuronal activity. Ischemia-reperfusion RPE cells express aquaporin-1, while Müller cells express
results in a decreased thickness of the inner retina (Fig. 17.9B) aquaporin-4. The transcellular water transport is osmotically coupled
to the transport of osmolytes, especially potassium (orange arrows)
due to neuronal degeneration caused by reactive oxygen radicals
and chloride ions (yellow arrow). The ion fluxes across the cell
and glutamate toxicity.81 Deletion of aquaporin-4 in mice pro- membranes are facilitated by transporter molecules and ion channels.
tects against ischemic injury of the retina,82 suggesting that In Müller cells, the Kir4.1 potassium channel is colocalized with AQP4
Müller cell-mediated water transport is implicated in the in membranes that surround the vessels, and at both limiting
membranes of the retina.
pathomechanisms of neuronal cell death after ischemia.

Contribution to edema development


and resolution contribute to edema development. Edema is characterized by a
Müller cells in animal models of retinopathies, which are known thickening of the retinal tissue, caused by water accumulation
to be associated with the development of edema in the human due to vascular leakage and/or by a swelling of neuronal and
retina, undergo an altered expression of aquaporins.1 In response glial cells. Whereas neuronal swelling is observed early after
to outer retinal and/or subretinal edema caused by blue-light ischemia-hypoxia, and is mainly caused by an overexcitation of
illumination, Müller cells strongly increase their expression of ionotropic glutamate receptors,81 Müller cell swelling occurs
aquaporin-4 in the outer nuclear layer (Fig. 17.12B). In ischemia within days of hypoxia.83 Because the water transport through
and diabetes, Müller cells and astrocytes express aquaporin-1 in Müller cells is coupled to the potassium currents,74 downregula-
membranes which contact the vitreous and the innermost blood tion or functional inactivation of Kir4.1 channels observed under
vessels.1 These alterations might be helpful for edema resolution. pathological conditions (Fig. 17.8A, B) should also disturb the
On the other hand, dysfunctional Müller cells might also transcellular water transport. Indeed, Müller cell bodies in slices
A AQP4 GFAP Merge
426
Control Diabetes
Section 1

GCL

GCL
IPL
IPL
Anatomy and Physiology
Basic Science and Translation to Therapy

INL INL
OPL
OPL

ONL
ONL

B
Control Blue light injury

GCL GCL

IPL IPL

INL
INL

OPL OPL

ONL
ONL

Fig. 17.12 Upregulation of glial fibrillary acidic protein (GFAP) is a characteristic marker of Müller cell gliosis under pathological conditions.
(A) Retinal slices from 6-month diabetic (right) and age-matched control rats (left) were immunostained against GFAP (green) and the glial water
channel, aquaporin-4 (AQP4; red). Cell nuclei were labeled with Hoechst 33258 (blue). In the control retina, only astrocytes at the inner margin
of the retina contain GFAP. In the retina of the diabetic animal, hypertrophied Müller cell fibers, traversing the whole neuroretina, display strong
labeling for GFAP. (B) Retinal slices of a rat were stained 3 days after illumination of a circumscribed area of the retina with blue light which
resulted in apoptotic death of photoreceptor cells. The light-injured retinal area is shown on the right (asterisk); the uninjured area is shown on
the left. The strong upregulation of AQP4 in Müller cell processes within the outer nuclear layer (ONL) is likely a response to the outer retinal
edema which developed due to the light-induced injury of the retinal pigment epithelium (resulting in leakage of the outer blood–retinal barrier)
and the volume decrease of apoptotic cells which is mediated by an efflux of osmolytes (especially of potassium and chloride) and water. GCL,
ganglion cell layer; INL, inner nuclear layer; IPL, inner plexiform layer; OPL, outer plexiform layer. Bars, 20 µm.

of diseased retinas swell upon hypo-osmotic challenge which is between the Müller cell interior and the hypo-osmotic environ-
not observed in control tissues (Fig. 17.8C). The swelling of ment. This results in water influx and cellular swelling. Because
Müller cells reflects an alteration in the osmotically driven water Müller cells are still capable of taking up excess potassium from
transport across Müller cell membranes. In the absence of func- the extracellular space through Kir2.1 channels which are not
tional Kir4.1 channels, the cells are not capable of rapidly releas- altered in their expression after ischemia (Fig. 17.9B), the impair-
ing potassium and, thus, to compensate the osmotic gradient ment in the release of potassium from Müller cells will result in
a potassium accumulation within the cells and, thus, in increased which cause dilation (epoxyeicosatrienoic acids) or constriction
intracellular osmotic pressure.77 The increased osmotic pressure (20-hydroxyeicosatetraenoic acid) of arterioles.97 Whether vaso-
draws water from fluid-filled spaces outside the neuroretina dilating or vasoconstricting responses are produced depends 427
(blood, vitreous) into the perivascular and endfeet regions of on nitric oxide.97 ATP and adenosine, which are released from
Müller cells, resulting in Müller cell swelling. The water influx both neurons and glial cells,98 evoke constriction and relaxation,

Chapter 17
from blood and vitreous is supported by aquaporin-4 water respectively, of pericytes.99,100
channels which are not altered, or even increased, in their
expression (Fig. 17.12).83 Thus, dysfunctional water clearance, Regulation of the extracellular
and possibly Müller cell swelling, contribute to the development space volume
of edema. Neuronal activity is also associated with alterations in the
Further factors which induce osmotic Müller cell swelling are volume of neuronal cell compartments (in particular, of syn-
reactive oxygen radicals and inflammatory lipids (arachidonic apses).1 The ion flux through ionotropic receptors is (for osmotic

Cell Biology of the Müller Cell


acid, prostaglandins).1 Increased vascular permeability is associ- reasons) coupled to a water flux. The ion flux into neurons (and
ated with the extravasation of albumin; in the presence of a of excess potassium into Müller cells) decreases the osmolarity
hypo-osmotic environment, albumin induces Müller cell swell- of the extracellular fluid,101 and the influx of water into neurons
ing.84 The effect of albumin is mediated by activation of trans- results in a swelling of neuronal cell processes and, thus, in a
forming growth factor-ß receptor type II, oxidative stress, decrease in the extracellular space volume.1 To avoid a decrease
production of inflammatory lipids, and intracellular sodium in the extracellular space volume (which would cause neuronal
overload.84 Thus, extravasated albumin may represent one factor hyperexcitation), Müller cells inhibit their swelling even if the
which links vascular leakage, Müller cell swelling, and dysregu- extracellular fluid is hypo-osmotic, which rather favors a water
lation of retinal fluid clearance. Other blood-derived factors such influx. In addition, Müller cells elongate and reduce the thick-
as thrombin and glutamate (as a constituent of the plasma), ness of their inner stem processes, at least in vitro.1 The rapid,
which close Kir channels and depolarize Müller cells, might be activity-dependent alterations in cell shape are supported by the
involved.85,86 A reduced efficiency of glial glutamate uptake, due viscoelastic properties of Müller cells (see below).
to cellular depolarization, may explain in part the clinical obser- To regulate cellular volume, Müller cells possess a purinergic
vation that the presence of hemorrhages is associated with an signaling mechanism which is triggered by glutamate released
aggravated reduction in retinal function.87 from neurons or Müller cells.1 The final step of this autocrine
cascade is the release of adenosine from Müller cells, which
Regulation of the blood–retinal barrier activates their adenosine A1 receptors, resulting in the opening
Vascular leakage caused by opening of the tight junctions of potassium and chloride channels. The ion efflux compensates
between vascular endothelial cells and/or by increased vesicular the osmotic gradient across the Müller cell membrane and thus
transport of serum proteins across vascular endothelia is an prevents cellular swelling. This mechanism might also accelerate
important pathogenic mechanism of retinal edema.88 Retinal the clearance of excess fluid from the retina through Müller
capillaries are closely ensheathed by glial processes. Usually, cells.1 The anti-inflammatory corticosteroid triamcinolone ace-
Müller cells enhance the barrier function of vascular endothelia89 tonide, which is clinically used to induce a rapid clearance of
by the secretion of factors such as PEDF, thrombospondin-1, and retinal edema, was shown to inhibit the osmotic swelling of
glial cell line-derived neurotrophic factor (GDNF).90,91 However, Müller cells, via inducing a release of endogenous adenosine.102
in response to hypoxia, inflammation, or glucose deprivation, The adenosine-induced opening of ion channels may re-establish
Müller cells produce factors such as vascular endothelial growth the ion and water transport through Müller cells under condi-
factor (VEGF) and tumor necrosis factor that increase vascular tions when the Kir4.1 channels are inactivated.
permeability.90,92,93 Müller cells are also a source of matrix metal-
loproteinases93,94 which degrade the tight junction protein, Responses to mechanical stress
occludin.95 Neuronal activity is associated with rapid alterations in the size
of neuronal synapses and cell bodies, in particular, in the plexi-
Mediation of neurovascular coupling form layers which contain many ionotropic receptors. Müller
Changes in neuronal activity induce different responses in cells respond by reshaping their processes; this is supported by
Müller cells, including alterations in their membrane potential the viscoelastic properties of the cells. Müller cells are softer than
and intracellular calcium responses triggered by activation of neurons; in particular, the processes within the plexiform layers
transmitter receptors.1 Müller cells in the rat retina generate are very soft.103 Because growing neurites prefer soft substrates,104
spontaneous calcium transients that start within the inner soft glial processes may support the growth of neuronal pro-
plexiform layer and propagate to the endfeet of the cells.96 cesses implicated in synaptic plasticity.103 Gliotic Müller cells
Flickering light increases the frequency of the calcium tran- display an increase in their stiffness due to upregulation of inter-
sients; this is likely mediated by adenosine 5’-triphosphate mediate filaments, particularly, of glial fibrillary acidic protein
(ATP) released from neurons (amacrine and ganglion cells) (GFAP) (Fig. 17.12); this will inhibit neurite growth and might
and subsequent activation of metabotropic purinergic (P2Y) be one reason for the aberrant tissue repair after retinal injury.105
receptors on Müller cells.96 Such purinergic neuron-to-glia sig- The increased stiffness of gliotic Müller cells might be also
naling is involved in the activity-dependent regulation of the responsible for the fact that, in retinal neovascularization, new
local retinal blood flow.97 Calcium responses in Müller cells blood vessels grow towards the vitreous rather than within the
evoked by neuron-derived ATP (Fig. 17.13B, C) activate the retinal tissue.106
calcium-dependent phospholipase A2; arachidonic acid metab­ Müller cells sense mechanical deformations of the retina by
olites are then released from perivascular glial membranes calcium-dependent mechanisms. Stretching of the retinal tissue
A C
428 Control Ischemia
ATP
Section 1

F
1

2 min
Anatomy and Physiology
Basic Science and Translation to Therapy

B Control ATP Washout

Fig. 17.13 Purinergic signaling in the inner retina involves Müller cells and blood vessels. (A-C) P2Y receptor-evoked calcium responses in Müller
cell endfeet. (A) Images taken from a whole mount of a control porcine retina (left) and a porcine retina that was isolated 3 days after a transient
retinal ischemia of 1 hour (right). The tissues were exposed to adenosine triphosphate (ATP) (200 µM) for 1 minute, and the peak calcium
responses in Müller cell endfeet within the ganglion cell layer are shown. In the image from the control retinal tissue, only one Müller cell endfoot
displayed a calcium response, whereas numerous endfeet displayed responses in the image from the postischemic retina. (B) Intracellular peak
calcium responses in Müller cell endfeet at the vitreal surface of a rat retina evoked by ATP (500 µM). Note the constriction of the arteriole in
response to ATP. (C) Time dependence of the calcium responses in three Müller cell endfeet marked by the circles in (B).

induces calcium responses, activation of extracellular signal- detachment of the vitreous from the retina, exert tractional forces
regulated kinases (ERK1/2), and upregulation of the transcrip- on to the cells; this activates Müller cells and results in cell
tion factor c-Fos and of bFGF in Müller cells.107 This may have hypertrophy and proliferation, as well as in vascular leakage.110
implications also for postnatal eye growth. Because bFGF coun- Mechanically stressed Müller cells release growth factors (e.g.,
teracts the development of form deprivation-induced axial bFGF) and ATP.107,111,112 Calcium influx (through stretch-activated
myopia,108 Müller cell-derived bFGF might prevent retinal over- and/or voltage-gated channels) and activation of BK channels
stretching and the development of myopia during ocular are required for the growth factor- and ATP-induced prolifera-
growth.107 tion of Müller cells.1,113,114
Müller cells have stretch-activated calcium-permeable cation
channels.109 Activation of the channels results in an increased
Regulation of neuronal activity by release
activity of calcium-activated BK channels.109 The potassium of gliotransmitters
efflux through BK channels is associated with an efflux of cell In addition to the uptake of neurotransmitters, Müller cells regu-
water and results in decreased Müller cell volume.109 Stretch- late the synaptic transmission by the release of “gliotransmit-
induced Müller cell responses may be implicated in epiretinal ters,” in particular of glutamate and of the purines, ATP and
membrane formation. Vitreous fibers, which adhere to Müller adenosine. Gliotransmitters provide excitatory (glutamate, ATP)
cell endfeet at sites of vitreoretinal attachment after partial and inhibitory effects (adenosine) on neighboring neurons.98,115
Retinal glial cells release glutamate in response to electrical bicarbonate, they may be implicated in the regulation of the
and mechanical stimuli, and after activation of receptors which extracellular pH. The chloride efflux through the receptor chan-
are coupled to intracellular calcium responses, including VEGF nels may also stimulate GABA uptake by the cells (which is 429
receptor-2 and P2Y receptors.1,116 Glia-derived glutamate acti- driven by a cotransport of sodium and chloride) and may com-
vates inhibitory amacrine cells which results in a depression of pensate the decrease in the extracellular chloride level caused by

Chapter 17
the light-evoked activity in a population of ganglion cell layer chloride influx into activated neurons.
neurons.116,117 Glutamate released from Müller cells by calcium- P2X7 receptor channels are calcium-permeable cation chan-
dependent exocytosis directly activates ganglion cell layer nels. Opening of the channels by extracellular ATP results in a
neurons.1 Glia-derived glutamate (acting at ionotropic glutamate calcium influx into the cells, calcium release from intracellular
receptors) and ATP (acting at ionotropic purinergic P2X7 recep- stores, and activation of BK channels (Fig. 17.7B, C).1,130 More-
tors) might contribute to the swelling and degeneration of retinal over, the opening of P2X7 channels causes cellular depolarization
ganglion cells, e.g., in glaucoma.1,118,119 In addition, Müller cell- which decreases the efficiency of the electrogenic neurotransmit-

Cell Biology of the Müller Cell


derived glutathione and polyamines, which increase the cur- ter uptake.130 However, the depolarization is counterregulated
rents of ionotropic glutamate receptors, might contribute to by hyperpolarization due to a potassium flux through BK chan-
glutamate-induced neuronal degeneration.120,121 nels. The BK channel-mediated hyperpolarization increases the
Receptor agonists such as glutamate as well as electrical and driving force for the calcium influx. P2X7 receptor currents are
mechanical stimuli trigger calcium waves in the glial network of increased when the extracellular fluid contains low concentra-
the rat retina.111,116 The propagation of the waves among Müller tions of divalent cations,130 suggesting that the light-induced
cells depends upon the release of ATP and autocrine/paracrine decrease in extracellular calcium will facilitate the gating of P2X7
activation of P2Y receptors.111,116 The glial calcium waves are receptors in Müller cells.
associated with both enhancement and depression of light-
evoked activity of distinct ganglion cells.117 Glial ATP released Müller cell gliosis
into the inner plexiform layer is converted extracellularly In response to virtually every pathological stimulus, Müller cells
to adenosine that activates A1 receptors in a population of gan- become activated.1 Reactive gliosis is thought to represent a cel-
glion cells, resulting in reduced spontaneous activity.112 Müller lular attempt to protect the tissue from further damage, to
cell-derived adenosine, which is also released via nucleoside promote tissue repair, and to limit tissue remodeling. It includes
transporters,1 may act as a negative-feedback regulator of neu- morphological, biochemical, and physiological changes; these
rotransmission; this adapts the level of neuronal activity to the responses vary with the type and severity of the insult. An
capacity of Müller cells to maintain retinal homeostasis. upregulation of the intermediate filaments, vimentin and GFAP
Müller cells are involved in setting the sensitivity of ganglion in glial cells (Fig. 17.12) is a very sensitive and early retinal stress
and amacrine cells by the release of D-serine, which potentiates indicator.1,131
N-methyl-D-aspartate receptor currents.122 They are implicated In fact, Müller cell gliosis has both cytoprotective and cyto-
in providing the direction selectivity of direction-selective gan- toxic effects on retinal neurons.1 Particularly under conditions of
glion cells by the secretion of acyl coenzyme A-binding protein.123 massive (proliferative) gliosis, the regular glial–neuronal inter-
Müller cells also produce nitric oxide; the production of nitric actions are disrupted and the retina degenerates (Fig. 17.14). An
oxide is strongly enhanced during dark adaptation.124 Nitric inverse relation between Müller cell proliferation and the
oxide diffuses out of the cells and has multiple effects on neigh- expression of neuron glia symbiosis-mediating proteins has
boring neurons, including a potentiation of phototransduction, been observed. However, also in the case of “conservative”
closure of gap junctions and N-methyl-D-aspartate receptor gliosis (associated with short-term, or low-level, Müller cell
channels, and an increase of calcium channel currents.1 Under proliferation), a dysfunction of Müller cells may contribute to
ischemic-hypoxic conditions, glial cells are a major source of neurodegeneration. After retinal detachment, for example, the
retinal nitric oxide. Nitric oxide dilates blood vessels, prevents downregulation of CRALBP and of glutamine synthetase132,133
platelet aggregation, and protects neurons from apoptosis by disrupts glia–neuronal interactions involved in photopigment
closure of N-methyl-D-aspartate receptor channels.125 However, and neurotransmitter recycling. The downregulation of carbonic
production of excess nitric oxide and subsequnt formation of anhydrase132,133 and Kir channels (Fig. 17.8A, B)1 results in dis-
free nitrogen radicals and protein nitrosylation have toxic effects turbances of the retinal acid–base, ion, and water homeostasis.
on surrounding neurons; these effects are implicated in the Because the electrogenic uptake carriers are voltage-dependent
pathogenesis of diabetic retinopathy, for example.126–128 (Fig. 17.6A), membrane depolarization reduces the efficiency of
glial neurotransmitter recycling which, together with an increase
Ionotropic receptors of Müller cells in extracellular potassium, will aggravate neurotoxicity. Because
Müller cells can sense neuronal activity,129 primarily by mem- the potassium currents of Müller cells are also a major driving
brane depolarization upon activation of receptor channels. force for the water transport through the cells,1 downregulation
Human Müller cells express GABAA and P2X7 receptors (Fig. of functional Kir channels also impairs retinal water homeosta-
17.7D),37,130 which are ligand-gated chloride and cation channels, sis, contributing to the development of edema.134
respectively. GABA evokes two types of inward currents in The involvement of reactive Müller cells in immune and
Müller cells, a fast transient GABAA receptor current and a sus- inflammatory responses may also exert detrimental effects. After
tained current mediated by electrogenic GABA transporters (Fig. retinal injuries such as retinal detachment, Müller cells upregu-
17.7F).37 Both currents depolarize the Müller cells. Zinc ions late inflammatory factors, including monocyte chemoattractant
released from photoreceptors29 increase the GABAA receptor protein (MCP)-1, which recruit phagocytotic monocytes/
currents.37 GABAA receptors may have different functional macrophages and microglial cells to the injured area.135,136 Blood-
roles; because the receptor channels are also permeable for derived monocytes/macrophages and neutrophils release
Vitreous body Migration
A B Proliferation C
430 Control
Section 1

MC

N
Anatomy and Physiology
Basic Science and Translation to Therapy

Subretinal space Migration

Fig. 17.14 Müller cell reactivity in proliferative vitreoretinopathy (PVR). (A) Ophthalmoscopic image of an experimentally induced PVR in the
rabbit eye. Note the large, folded cellular masses on the vitreal surface of the retina (arrows). (B) Schematic drawing of increasing degrees of
Müller cell reactivity (from left to right). Müller cells re-enter the proliferation cycle, migrate out of the neural retina, and participate in the
formation of periretinal fibrocellular membranes. (C) Transmission electron micrograph of a reactive Müller cell (MC) of the rabbit retina which is
migrating through a hole in the inner limiting membrane (arrowheads) into the vitreous body (asterisk). The nucleus (N) of the Müller cell is
translocated to the innermost retinal layer.

oxygen free radicals and cytotoxic cytokines which play critical that a better understanding of gliotic mechanisms is essential for
roles in the photoreceptor apoptosis after retinal detachment137 the development of efficient therapeutic strategies which increase
and in neuronal degeneration after ischemia-reperfusion.138 Mice the protective and regenerative, and decrease the destructive,
deficient in vimentin and GFAP display an attenuation of the roles of reactive Müller cells.
detachment-induced glial cell responses (activation of ERK1/2 REFERENCES
and c-Fos, induction of MCP-1) and, as a consequence, a decrease 1. Reichenbach A, Bringmann A. Müller cells in the healthy and diseased retina.
in monocyte infiltration and photoreceptor apoptosis was New York: Springer, 2010.
2. Reichenbach A, Robinson SR. Phylogenetic constraints on retinal organization
observed.139 and development: an Haeckelian perspective. Prog Retin Eye Res 1995;15:
In contrast, reactive Müller cells might protect photoreceptors 139–71.
3. Franze K, Grosche J, Skatchkov SN, et al. Müller cells are living optical fibers
and neurons from cell death by various mechanisms, including in the vertebrate retina. Proc Natl Acad Sci U S A 2007;104:8287–92.
the secretion of neurotrophic and growth factors, in particular 4. Muniz A, Villazana-Espinoza ET, Hatch AL, et al. A novel cone visual cycle
bFGF.1,46,140 Various cytokines might reduce (e.g., nerve growth in the cone-dominated retina. Exp Eye Res 2007;85:175–84.
5. Bunt-Milam AH, Saari JC. Immunocytochemical localization of two retinoid-
factor) or increase (e.g., GDNF) the production of bFGF by binding proteins in vertebrate retina. J Cell Biol 1983;97:703–12.
Müller cells.46,141,142 GDNF induces phosphorylation of ERK1/2 6. Jablonski MM, Iannaccone A. Targeted disruption of Müller cell metabolism
induces photoreceptor dysmorphogenesis. Glia 2000;32:192–204.
in Müller cells, resulting in a transcriptional upregulation of 7. Jablonski MM, Wohabrebbi A, Ervin CS. Lactose promotes organized photo-
bFGF that in turn supports photoreceptor survival.141,142 GDNF, receptor outer segment assembly and preserves expression of photoreceptor
proteins in retinal degeneration. Mol Vis 1999;5:16.
which is produced by photoreceptor and glial cells,141,143 also 8. Wang X, Iannaccone A, Jablonski MM. Contribution of Müller cells toward
protects photoreceptors and ganglion cells from apoptosis the regulation of photoreceptor outer segment assembly. Neuron Glia Biol
through upregulation of the glial glutamate transporter 2005;1:1–6.
9. Long KO, Fisher SK, Fariss RN, et al. Disc shedding and autophagy in the
GLAST.144,145 Retinal preconditioning with bright light or cone-dominant ground squirrel retina. Exp Eye Res 1986;43:193–205.
mechanical stress protects photoreceptors from apoptosis 10. Polit L, Rotstein N, Carri N. Effects of docosahexaenoic acid on retinal devel-
opment: cellular and molecular aspects. Lipids 2001;36:927–35.
because these stimuli induce an upregulation of bFGF and ciliary 11. Otori Y, Shimada S, Tanaka K, et al. Marked increase in glutamate-aspartate
neurotrophic factor in Müller cells.45,140 Likewise, argon laser transporter (GLAST/GluT-1) mRNA following transient retinal ischemia.
Mol Brain Res 1994;27:310–4.
photocoagulation slows photoreceptor degeneration by induc- 12. Rauen T, Rothstein JD, Wässle H. Differential expression of three glutamate
tion of bFGF in Müller cells.146 transporter subtypes in the rat retina. Cell Tissue Res 1996;286:325–36.
13. Rauen T, Taylor WR, Kuhlbrodt K, et al. High-affinity glutamate transporters
Müller stem cells in the rat retina: a major role of the glial glutamate transporter GLAST-1 in
transmitter clearance. Cell Tissue Res 1998;291:19–31.
As a precondition for proliferation and migration, Müller cells 14. Kugler P, Beyer A. Expression of glutamate transporters in human and rat
retina and rat optic nerve. Histochem Cell Biol 2003;120:199–212.
reactivate distinct cellular programs normally used in ontoge- 15. Ward MM, Jobling AI, Kalloniatis M, et al. Glutamate uptake in retinal glial
netic development. In the injured retina, a population of Müller cells during diabetes. Diabetologia 2005;48:351–60.
16. Barbour B, Brew H, Attwell D. Electrogenic glutamate uptake in glial cells is
cells dedifferentiates to cells with properties similar to pluripo- activated by intracellular potassium. Nature 1988;335:433–5.
tent retinal progenitor/stem cells, and may even express distinct 17. Brew H, Attwell D. Electrogenic glutamate uptake is a major current carrier
in the membrane of axolotl retinal glial cells. Nature 1987;327:707–9.
neuronal and photoreceptor markers.147,148 The decrease in Kir 18. Marc RE. Structural organization of GABAergic circuitry in ectotherm retinas.
currents (Fig. 17.10A) and the increase in voltage-gated sodium Prog Brain Res 1992;90:61–92.
currents (Fig. 17.10B) observed in Müller cells from patients with 19. Hasegawa J, Obara T, Tanaka K, et al. High-density presynaptic transporters
are required for glutamate removal from the first visual synapse. Neuron
proliferative retinopathies37 may reflect such a transdifferentia- 2006;50:63–74.
tion of the cells. However, the neuron-regenerating capability of 20. Matsui K, Hosoi N, Tachibana M. Active role of glutamate uptake in the
synaptic transmission from retinal nonspiking neurons. J Neurosci 1999;19:
mammalian Müller cells in situ is very restricted. Attempts to 6755–66.
facilitate the neurogenic program of Müller cells, e.g., by trans- 21. Higgs MH, Lukasiewicz PD. Glutamate uptake limits synaptic excitation of
retinal ganglion cells. J Neurosci 2002;19:3691–700.
differentiation of cultured Müller cells, are ongoing.148,149 The 22. Pow DV, Barnett NL, Penfold P. Are neuronal transporters relevant in retinal
tedious progress of this approach underlines the general issue glutamate homeostasis? Neurochem Int 2000;37:191–8.
23. Holcombe DJ, Lengefeld N, Gole GA, et al. The effects of acute intraocular 57. Newman EA. A physiological measure of carbonic anhydrase in Müller cells.
pressure elevation on rat retinal glutamate transport. Acta Ophthalmol Scand Glia 1994;11:291–9.
2008;86:408–14. 58. Oakley B, Wen R. Extracellular pH in the isolated retina of the toad in
24. Naskar R, Vorwerk CK, Dreyer EB. Concurrent downregulation of a gluta- darkness and during illumination. J Physiol 1989;419:353–78. 431
mate transporter and receptor in glaucoma. Invest Ophthalmol Vis Sci 59. Linser PJ, Moscona AA. Carbonic anhydrase-C in the neural retina: transition
2000;41:1940–4. from generalized to glia-specific cell localization during embryonic develop-
25. Barnett NL, Pow DV, Bull ND. Differential perturbation of neuronal and glial ment. Proc Natl Acad Sci U S A 1981;78:7190–4.

Chapter 17
glutamate transport systems in retinal ischaemia. Neurochem Int 2001;39: 60. Nagelhus EA, Mathiisen TM, Bateman AC, et al. Carbonic anhydrase XIV is
291–9. enriched in specific membrane domains of retinal pigment epithelium, Müller
26. Müller A, Maurin L, Bonne C. Free radicals and glutamate uptake in the cells, and astrocytes. Proc Natl Acad Sci U S A 2005;102:8030–5.
retina. Gen Pharmacol 1998;30:315–8. 61. Borgula GA, Karwoski CJ, Steinberg RH. Light-evoked changes in extracel-
27. Li Q, Puro DG. Diabetes-induced dysfunction of the glutamate transporter in lular pH in frog retina. Vision Res 1989;29:1069–77.
retinal Müller cells. Invest Ophthalmol Vis Sci 2002;43:3109–16. 62. DeVries SH. Exocytosed protons feedback to suppress the Ca2+ current in
28. Beretta S, Wood JP, Derham B, et al. Partial mitochondrial complex I inhibition mammalian cone photoreceptors. Neuron 2001;32:1107–17.
induces oxidative damage and perturbs glutamate transport in primary 63. Newman EA. Acid efflux from retinal glial cells generated by sodium
retinal cultures. Relevance to Leber hereditary optic neuropathy (LHON). bicarbonate cotransport. J Neurosci 1996;16:159–68.
Neurobiol Dis 2006;24:308–17. 64. Barnes S, Merchant V, Mahmud F. Modulation of transmission gain by

Cell Biology of the Müller Cell


29. Wu SM, Qiao X, Noebels JL, et al. Localization and modulatory actions of zinc protons at the photoreceptor output synapse. Proc Natl Acad Sci U S A
in vertebrate retina. Vision Res 1993;33:2611–6. 1993;90:10081–5.
30. Barbour B, Szatkowski M, Ingledew N, et al. Arachidonic acid induces a 65. Steinberg RH, Oakley II B, Niemeyer G. Light-evoked changes in [K+]0 in
prolonged inhibition of glutamate uptake into glial cells. Nature 1989;342: retina of intact cat eye. J Neurophysiol 1980;44:897–921.
918–20. 66. Karwoski CJ, Newman EA, Shimazaki H, et al. Light-evoked increases in
31. Spiridon M, Kamm D, Billups B, et al. Modulation by zinc of the glutamate extracellular K+ in the plexiform layers of amphibian retinas. J Gen Physiol
transporters in glial cells and cones isolated from the tiger salamander retina. 1985;86:189–213.
J Physiol 1998;506:363–76. 67. Newman EA, Frambach DA, Odette LL. Control of extracellular potassium
32. Napper GA, Pianta MJ, Kalloniatis M. Reduced glutamate uptake by retinal levels by retinal glial cell K+ siphoning. Science 1984;225:1174–5.
glial cells under ischemic/hypoxic conditions. Vis Neurosci 1999;16:149–58. 68. Karwoski CJ, Lu HK, Newman EA. 1989. Spatial buffering of light-evoked
33. Szatkowski M, Barbour B, Attwell D. Non-vesicular release of glutamate from potassium increases by retinal Müller (glial) cells. Science 1989;244:
glial cells by reversed electrogenic glutamate uptake. Nature 1990;348:443–6. 578–80.
34. Maguire G, Simko H, Weinreb RN, et al. Transport-mediated release of end­ 69. Newman EA. Regional specialization of retinal glial cell membrane. Nature
ogenous glutamate in the vertebrate retina. Pflügers Arch 1998;436:481–4. 1984;309:155–7.
35. Marcaggi P, Hirji N, Attwell D. Release of L-aspartate by reversal of glutamate 70. Brew H, Gray PT, Mobbs P, et al. Endfeet of retinal glial cells have higher
transporters. Neuropharmacology 2005;49:843–9. densities of ion channels that mediate K+ buffering. Nature 1986;324:466–8.
36. Kato S, Ishita S, Sugawara K, et al. Cystine/glutamate antiporter expression 71. Newman EA. Inward-rectifying potassium channels in retinal glial (Müller)
in retinal Müller glial cells: implications for DL-α-aminoadipate toxicity. cells. J Neurosci 1993;13:3333–45.
Neuroscience 1993;57:473–82. 72. Kofuji P, Ceelen P, Zahs KR, et al. Genetic inactivation of an inwardly rectify-
37. Reichelt W, Pannicke T, Biedermann B, et al. Comparison between functional ing potassium channel (Kir4.1 subunit) in mice: phenotypic impact in retina.
characteristics of healthy and pathological human retinal Müller glial cells. J Neurosci 2000;20:5733–40.
Surv Ophthalmol 1997;42:S105–17. 73. Kofuji P, Biedermann B, Siddharthan V, et al. Kir potassium channel subunit
38. Riepe RE, Norenburg MD. Müller cell localisation of glutamine synthetase in expression in retinal glial cells: implications for spatial potassium buffering.
rat retina. Nature 1977;268:654–5. Glia 2002;39:292–303.
39. Pow DV, Crook DK. Direct immunocytochemical evidence for the transfer of 74. Nagelhus EA, Horio Y, Inanobe A, et al. Immunogold evidence suggests that
glutamine from glial cells to neurons: use of specific antibodies directed coupling of K+ siphoning and water transport in rat retinal Müller cells is
against the D-stereoisomers of glutamate and glutamine. Neuroscience mediated by a coenrichment of Kir4.1 and AQP4 in specific membrane
1996;70:295–302. domains. Glia 1999;26:47–54.
40. Pow DV, Robinson SR. Glutamate in some retinal neurons is derived solely 75. Ishii M, Fujita A, Iwai K, et al. Differential expression and distribution of
from glia. Neuroscience 1994;60:355–66. Kir5.1 and Kir4.1 inwardly rectifying K+ channels in retina. Am J Physiol
41. Fletcher EL, Kalloniatis M. Neurochemical architecture of the normal and 2003;285:C260–C267.
degenerating rat retina. J Comp Neurol 1996;376:343–60. 76. Skatchkov SN, Eaton MJ, Shuba YM, et al. Tandem-pore domain potassium
42. Marc RE, Murry RF, Fisher SK, et al. Amino acid signatures in the detached channels are functionally expressed in retinal (Müller) glial cells. Glia 2006;53:
cat retina. Invest Ophthalmol Vis Sci 1998;39:1694–702. 266–76.
43. Kruchkova Y, Ben-Dror I, Herschkovitz A, et al. Basic fibroblast growth factor: 77. Pannicke T, Iandiev I, Uckermann O, et al. A potassium channel-linked mech-
a potential inhibitor of glutamine synthetase expression in injured neural anism of glial cell swelling in the postischemic retina. Mol Cell Neurosci
tissue. J Neurochem 2001;77:1641–9. 2004;26:493–502.
44. Faktorovich EG, Steinberg RH, Yasumura D, et al. Photoreceptor degenera- 78. Stamer WD, Bok D, Hu J, et al. Aquaporin-1 channels in human retinal pigment
tion in inherited retinal dystrophy delayed by basic fibroblast growth factor. epithelium: role in transepithelial water movement. Invest Ophthalmol Vis
Nature 1990;347:83–6. Sci 2003;44:2803–8.
45. Liu C, Peng M, Laties AM, et al. Preconditioning with bright light evokes a 79. Antcliff RJ, Hussain AA, Marshall J. Hydraulic conductivity of fixed retinal
protective response against light damage in the rat retina. J Neurosci 1998;18: tissue after sequential excimer laser ablation: barriers limiting fluid distribu-
1337–44. tion and implications for cystoid macular edema. Arch Ophthalmol 2001;
46. Harada T, Harada C, Nakayama N, et al. Modification of glial–neuronal cell 119:539–44.
interactions prevents photoreceptor apoptosis during light-induced retinal 80. Li J, Patil RV, Verkman AS. Mildly abnormal retinal function in transgenic
degeneration. Neuron 2000;26:533–41. mice without Müller cell aquaporin-4 water channels. Invest Ophthalmol Vis
47. Barnett NL, Osborne NN. Redistribution of GABA immunoreactivity follow- Sci 2002;43:573–9.
ing central retinal artery occlusion. Brain Res 1995;677:337–40. 81. Osborne NN, Casson RJ, Wood JP, et al. Retinal ischemia: mechanisms of
48. Ishikawa A, Ishiguro S, Tamai M. Changes in GABA metabolism in damage and potential therapeutic strategies. Prog Retin Eye Res 2004;23:
streptozotocin-induced diabetic rat retinas. Curr Eye Res 1996;15:63–71. 91–147.
49. Poitry S, Poitry-Yamate C, Ueberfeld J, et al. Mechanisms of glutamate meta- 82. Da T, Verkman AS. Aquaporin-4 gene disruption in mice protects against
bolic signaling in retinal glial (Müller) cells. J Neurosci 2000;20:1809–21. impaired retinal function and cell death after ischemia. Invest Ophthalmol Vis
50. Poitry-Yamate CL, Poitry S, Tsacopoulos M. Lactate released by Müller glial Sci 2004;45:4477–83.
cells is metabolized by photoreceptors from mammalian retina. J Neurosci 83. Kaur C, Sivakumar V, Yong Z, et al. Blood–retinal barrier disruption and
1995;15:5179–91. ultrastructural changes in the hypoxic retina in adult rats: the beneficial effect
51. Linsenmeier RA. Effects of light and darkness on oxygen distribution and of melatonin administration. J Pathol 2007;212:429–39.
consumption in the cat retina. J Gen Physiol 1986;88:521–42. 84. Löffler S, Wurm A, Kutzera F, et al. Serum albumin induces osmotic swelling
52. Schütte M, Werner P. Redistribution of glutathione in the ischemic rat retina. of rat retinal glial cells. Brain Res 2010;1317:268–76.
Neurosci Lett 1998;246:53–6. 85. Puro DG, Stuenkel EL. Thrombin-induced inhibition of potassium currents in
53. Paasche G, Gärtner U, Germer A, et al. Mitochondria of retinal Müller (glial) human retinal glial (Müller) cells. J Physiol 1995;485:337–48.
cells: the effects of aging and of application of free radical scavengers. 86. Kusaka S, Kapousta-Bruneau NV, Puro DG. Plasma-induced changes in the
Ophthalmic Res 2000;32:229–36. physiology of mammalian retinal glial cells: role of glutamate. Glia 1999;
54. Kern TS, Kowluru RA, Engerman RL. Abnormalities of retinal metabolism in 25:205–15.
diabetes or galactosemia: ATPases and glutathione. Invest Ophthalmol Vis Sci 87. Gass JD. Stereoscopic atlas of macular diseases: diagnosis and treatment.
1994;35:2962–7. St Louis, MO: Mosby, 1997. p.1061.
55. Reichelt W, Stabel-Burow J, Pannicke T, et al. The glutathione level of retinal 88. Cunha-Vaz JG, Travassos A. Breakdown of the blood–retinal barriers and
Müller glial cells is dependent on the high-affinity sodium-dependent uptake cystoid macular edema. Surv Ophthalmol 1984;28:S485–92.
of glutamate. Neuroscience 1997;77:1213–24. 89. Tout S, Chan-Ling T, Hollander H, et al. The role of Müller cells in the
56. Shelton MD, Kern TS, Mieyal JJ. Glutaredoxin regulates nuclear factor κ-B and formation of the blood–retinal barrier. Neuroscience 1993;55:291–301.
intercellular adhesion molecule in Müller cells: model of diabetic retinopathy. 90. Eichler W, Yafai Y, Wiedemann P, et al. Angiogenesis-related factors derived
J Biol Chem 2007;282:12467–74. from retinal glial (Müller) cells in hypoxia. Neuroreport 2004;15:1633–7.
91. Nishikiori N, Osanai M, Chiba H, et al. Glial cell-derived cytokines attenuate 123. Barmack NH, Bilderback TR, Liu H, et al. Activity-dependent expression of
the breakdown of vascular integrity in diabetic retinopathy. Diabetes 2007; acyl-coenzyme A-binding protein in retinal Müller glial cells evoked by
56:1333–40. optokinetic stimulation. J Neurosci 2004;24:1023–33.
432 92. Aiello LP, Northrup JM, Keyt BA, et al. Hypoxic regulation of vascular endo- 124. Zemel E, Eyal O, Lei B, et al. NADPH diaphorase activity in mammalian
thelial growth factor in retinal cells. Arch Ophthalmol 1995;113:1538–44. retinas is modulated by the state of visual adaptation. Vis Neurosci 1996;13:
93. Noda K, Ishida S, Shinoda H, et al. Hypoxia induces the expression of 863–71.
membrane-type 1 matrix metalloproteinase in retinal glial cells. Invest Oph- 125. Goldstein IM, Ostwald P, Roth S. Nitric oxide: a review of its role in retinal
thalmol Vis Sci 2005;46:3817–24. function and disease. Vision Res 1996;36:2979–94.
Section 1

94. Behzadian MA, Wang XL, Windsor LJ, et al. TGF-ß increases retinal endothe- 126. Roth S. Role of nitric oxide in retinal cell death. Clin Neurosci 1997;4:
lial cell permeability by increasing MMP-9: possible role of glial cells in endo- 216–23.
thelial barrier function. Invest Ophthalmol Vis Sci 2001;42:853–9. 127. Goureau O, Regnier-Ricard F, Courtois Y. Requirement for nitric oxide in
95. Giebel SJ, Menicucci G, McGuire PG, et al. Matrix metalloproteinases in early retinal neuronal cell death induced by activated Müller glial cells. J Neuro-
diabetic retinopathy and their role in alteration of the blood–retinal barrier. chem 1999;72:2506–15.
Lab Invest 2005;85:597–607. 128. Koeberle PD, Ball AK. Nitric oxide synthase inhibition delays axonal
96. Newman EA. Calcium increases in retinal glial cells evoked by light-induced degeneration and promotes the survival of axotomized retinal ganglion cells.
neuronal activity. J Neurosci 2005;25:5502–10. Exp Neurol 1999;158:366–81.
Anatomy and Physiology

97. Metea MR, Newman EA. Glial cells dilate and constrict blood vessels: a 129. Rillich K, Gentsch J, Reichenbach A, et al. Light stimulation evokes two
Basic Science and Translation to Therapy

mechanism of neurovascular coupling. J Neurosci 2006;26:2862–70. different calcium responses in Müller glial cells of the guinea pig retina.
98. Housley GD, Bringmann A, Reichenbach A. Purinergic signaling in special Eur J Neurosci 2009;29:1165–76.
senses. Trends Neurosci 2009;32:128–41. 130. Pannicke T, Fischer W, Biedermann B, et al. P2X7 receptors in Müller glial cells
99. Li Q, Puro DG. Adenosine activates ATP-sensitive K+ currents in pericytes of from the human retina. J Neurosci 2000;20:5965–72.
rat retinal microvessels: role of A1 and A2A receptors. Brain Res 2001;907: 131. Bignami A, Dahl D. The radial glia of Müller in the rat retina and their
93–9. response to injury: an immunofluorescence study with antibodies to the glial
100. Peppiatt CM, Howarth C, Mobbs P, et al. Bidirectional control of CNS fibrillary acidic (GFA) protein. Exp Eye Res 1979;28:63–9.
capillary diameter by pericytes. Nature 2006;443:700–4. 132. Lewis GP, Erickson PA, Guerin CJ, et al. Changes in the expression of specific
101. Dmitriev AV, Govardovskii VI, Schwahn HN, et al. Light-induced changes of Müller cell proteins during long-term retinal detachment. Exp Eye Res 1989;
extracellular ions and volume in the isolated chick retina-pigment epithelium 49:93–111.
preparation. Vis Neurosci 1999;16:1157–67. 133. Lewis GP, Mervin K, Valter K, et al. Limiting the proliferation and reactivity
102. Uckermann O, Kutzera F, Wolf A, et al. The glucocorticoid triamcinolone of retinal Müller cells during experimental retinal detachment: the value of
acetonide inhibits osmotic swelling of retinal glial cells via stimulation of oxygen supplementation. Am J Ophthalmol 1999;128:165–72.
endogenous adenosine signaling. J Pharmacol Exp Ther 2005;315:1036–45. 134. Jackson TL, Hillenkamp J, Williamson TH, et al. An experimental model of
103. Lu Y-B, Franze K, Seifert G, et al. Viscoelastic properties of individual glial rhegmatogenous retinal detachment: surgical results and glial cell response.
cells and neurons in the CNS. Proc Natl Acad Sci U S A 2006;103: Invest Ophthalmol Vis Sci 2003;44:4026–34.
17759–64. 135. Nakazawa T, Matsubara A, Noda K, et al. Characterization of cytokine
104. Flanagan LA, Ju YE, Marg B, et al. Neurite branching on deformable sub- responses to retinal detachment in rats. Mol Vis 2006;12:867–78.
strates. Neuroreport 2002;13:2411–5. 136. Hollborn M, Francke M, Iandiev I, et al. Early activation of inflammation- and
105. Lu YB, Iandiev I, Hollborn M, et al. Reactive glial cells: increased stiffness immune response-related genes after experimental detachment of the porcine
correlates with increased intermediate filament expression. FASEB J 2011;25: retina. Invest Ophthalmol Vis Sci 2008;49:1262–73.
624–31. 137. Nakazawa T, Hisatomi T, Nakazawa C, et al. Monocyte chemoattractant
106. Lundkvist A, Reichenbach A, Betsholtz C, et al. Under stress, the absence of protein 1 mediates retinal detachment-induced photoreceptor apoptosis. Proc
intermediate filaments from Müller cells in the retina has structural and func- Natl Acad Sci U S A 2007;104:2425–30.
tional consequences. J Cell Sci 2004;117:3481–8. 138. Neufeld AH, Kawai S, Das S, et al. Loss of retinal ganglion cells following
107. Lindqvist N, Liu Q, Zajadacz J, et al. Retinal glial (Müller) cells: sensing and retinal ischemia: the role of inducible nitric oxide synthase. Exp Eye Res
responding to tissue stretch. Invest Ophthalmol Vis Sci 2010;51:1683–90. 2002;75:521–8.
108. Rohrer B, Stell WK. Basic fibroblast growth factor (bFGF) and transforming 139. Nakazawa T, Takeda M, Lewis GP, et al. Attenuated glial reactions and pho-
growth factor ß (TGF-ß) act as stop and go signals to modulate postnatal toreceptor degeneration after retinal detachment in mice deficient in glial
ocular growth in the chick. Exp Eye Res 1994;58:553–61. fibrillary acidic protein and vimentin. Invest Ophthalmol Vis Sci 2007;48:
109. Puro DG. Stretch-activated channels in human retinal Müller cells. 2760–8.
Glia 1991;4:456–60. 140. Wen R, Song Y, Cheng T, et al. Injury-induced upregulation of bFGF and
110. Schubert HD. Cystoid macular edema: the apparent role of mechanical CNTF mRNAs in the rat retina. J Neurosci 1995;15:7377–85.
factors. Prog Clin Biol Res 1989;312:277–91. 141. Harada C, Harada T, Quah HMA, et al. Potential role of glial cell line-derived
111. Newman EA. Propagation of intercellular calcium waves in retinal astrocytes neurotrophic factor receptors in Müller glial cells during light-induced retinal
and Müller cells. J Neurosci 2001;21:2215–23. degeneration. Neuroscience 2003;122:229–35.
112. Newman EA. Glial cell inhibition of neurons by release of ATP. J Neurosci 142. Hauck SM, Kinkl N, Deeg CA, et al. GDNF family ligands trigger indirect
2003;23:1659–66. neuroprotective signaling in retinal glial cells. Mol Cell Biol 2006;26:
113. Puro DG, Mano T. Modulation of calcium channels in human retinal glial cells 2746–57.
by basic fibroblast growth factor: a possible role in retinal pathobiology. 143. Igarashi Y, Chiba H, Utsumi H, et al. Expression of receptors for glial cell
J Neurosci 1991;11:1873–80. line-derived neurotrophic factor (GDNF) and neurturin in the inner blood–
114. Puro DG, Roberge F, Chan C-C. Retinal glial cell proliferation and ion chan- retinal barrier of rats. Cell Struct Funct 2000;25:237–41.
nels: a possible link. Invest Ophthalmol Vis Sci 1989;30:521–9. 144. Delyfer MN, Simonutti M, Neveux N, et al Does GDNF exert its neuroprotec-
115. Newman EA. Glial modulation of synaptic transmission in the retina. tive effects on photoreceptors in the rd1 retina through the glial glutamate
Glia 2004;47:268–74. transporter GLAST? Mol Vis 2005;11:677–87.
116. Newman EA, Zahs KR. Calcium waves in retinal glial cells. Science 145. Koeberle PD, Bähr M. The upregulation of GLAST-1 is an indirect antiapop-
1997;275:844–7. totic mechanism of GDNF and neurturin in the adult CNS. Cell Death Differ
117. Newman EA, Zahs KR. Modulation of neuronal activity by glial cells in the 2008;15:471–83.
retina. J Neurosci 1998;18:4022–8. 146. Chu Y, Humphrey MF, Alder VV, et al. Immunocytochemical localization of
118. Zhang X, Zhang M, Laties AM, et al. Stimulation of P2X7 receptors elevates basic fibroblast growth factor and glial fibrillary acidic protein after laser
Ca2+ and kills retinal ganglion cells. Invest Ophthalmol Vis Sci 2005;46: photocoagulation in the Royal College of Surgeons rat. Aust N Z J Ophthalmol
2183–91. 1998;26:87–96.
119. Resta V, Novelli E, Vozzi G, et al. Acute retinal ganglion cell injury caused by 147. Fischer AJ, Reh TA. Müller glia are a potential source of neural regeneration
intraocular pressure spikes is mediated by endogenous extracellular ATP. in the postnatal chicken retina. Nat Neurosci 2001;4:247–52.
Eur J Neurosci 2007;25:2741–54. 148. Ooto S, Akagi T, Kageyama R, et al. Potential for neural regeneration after
120. Janaky R, Ogita K, Pasqualotto BA, et al. Glutathione and signal transduction neurotoxic injury in the adult mammalian retina. Proc Natl Acad Sci U S A
in the mammalian CNS. J Neurochem 1999;73:889–902. 2004;101:13654–9.
121. Pernet V, Bourgeois P, Di Polo A. A role for polyamines in retinal ganglion 149. Lawrence JM, Singhal S, Bhatia B, et al. MOI-M1 cells and similar Müller glial
cell excitotoxic death. J Neurochem 2007;103:1481–90. cell lines derived from adult human retina exhibit neural stem cell character-
122. Miller RF. D-Serine as a glial modulator of nerve cells. Glia 2004;47:275–83. istics. Stem Cells 2007;25:2033–43.
Anatomy and Physiology Section 1

Retinal and Choroidal Vasculature: Chapter

Retinal Oxygenation
Maria B. Grant, Gerard A. Lutty 18 
INTRODUCTION vasculature is supplied with blood directly by the central retinal
artery in humans. The retinal vasculature has a traditional end-
Oxygen is necessary for the existence of mammals because it is arterial hierarchy: arteries branch to arterioles, which supply a
required to generate adenosine triphosphate (ATP) oxidatively. capillary network that is drained by venules, and then veins
Although the partial pressure of oxygen (pO2) is 149 mmHg (21% remove the blood from the retina (Fig. 18.1). The retinal vascu-
of atmospheric oxygen) at sea level, arterial oxygen content is as lature forms the inner blood–retinal barrier (BRB), restricting
low as 75–100 mmHg (10–14%) and tissue pO2 is much lower. The passage of molecules that do not have receptors or transporters
oxygen level in inner segments of photoreceptors (mitochondria- on the luminal surface of the endothelial cells. Capillaries have
rich) after dark adaptation is between 0 and 5 mmHg (0.7%) but a lumen diameter of 3.5–6 µm, permitting passage of red blood
up to 20 mmHg in the light. Inner retinal oxygen is normally cells only after deformation of their disc shape. The retinal capil-
20 mmHg, so normoxia depends on the area of retina and dark/ laries and venules have perivascular pericytes and the retina has
light state.1,2 Hypoxia is an oxygen level below normoxia while the highest endothelial cell-to-pericyte ratio in the body, 1 : 1.8
hyperoxia is achieved by inhaling high levels of oxygen as in the The choroidal vasculature forms well before the retinal vessels
isolette of the neonatal intensive care unit. (6–9 WG), although its maturation is only completed after
The retina is one of the most metabolically active tissues in the 20 WG. It develops by hemovasculogenesis, formation of blood
body. It has two unique zones of oxygenation.1 The inner retina cells and blood vessel cells from a common progenitor, the
is supplied with oxygen by the retinal vasculature. The retinal hemangioblast.9 The choroidal vasculature provides oxygen and
vasculature is autoregulated because it is responsive to changes nutrients to the photoreceptors. The capillary system, the chorio-
in systemic oxygen levels, keeping the inner retina at a relatively capillaris, lies directly under the Bruch’s membrane, while inter-
constant level. If the retinal vasculature is compromised, as in mediate and large blood vessels of the system lie posterior to the
ischemic retinopathies, the retina becomes hypoxic in that area. capillaries. The short and long ciliary arteries supply blood to
The outer retina is supplied solely by the choroidal vasculature. the choroidal vasculature while 4–6 vortex veins remove blood
Unlike retina, choroidal vessels are not autoregulated, so sys- from this vast system. Unlike the retina, the hierarchy in choroid
temic levels of oxygen control the level of oxygen in choroid. is lobular, similar to kidney glomeruli (Fig. 18.1). The lobules
Supply of oxygen to choroid is diminished by stenosis of the change in shape, vascular density, and size depending upon area
ophthalmic artery, which is the branch off the internal carotid and the location of feeding arterioles and draining venules also
that is most likely to be stenosed because it is a right-angle varies by geographic location of the lobule.10 The capillaries are
branch. broad and flat, having luminal diameters ranging from 10 to
38 µm in diameter. Another major difference from retina is that
Comparison of retinal and the capillaries are fenestrated, allowing the passage of small
choroidal vasculatures molecules and solutes through these 60–70-nm pores. The cho-
riocapillaris is sided in that the majority of the fenestrations are
Although less than 300 µm apart in distance, the retinal and
on the retinal side as well as all three types of vascular endothe-
choroidal vasculatures are vastly different in many attributes in
lial growth factor (VEGF) receptors.11 Pericytes, however, are
addition to autoregulation. The initial retinal vasculature in
mostly on the scleral side of these capillaries. Control of vascular
human starts forming around 14 weeks’ gestation (WG) by vas-
tone in choroid may be accomplished by mast cells, which lie
culogenesis, development by differentiation and assembly of
abluminal to arteries and arterioles, or choroidal ganglion cells.12
vascular precursors, angioblasts.3–5 The deep capillary network
forms after 20 WG by angiogenesis, development by migration,
and proliferation of endothelial cells from existing blood vessels.
HISTORY OF RETINAL ISCHEMIA
The driving force for vascular development is physiological Ischemia is the restriction in oxygen supply without considering
hypoxia; metabolic requirements of developing neurons are only actual levels of oxygen. Michaelson13,14 and Wise15 hypothesized
met by stimulating the development of a retinal vasculature.6,7 that areas of vascular loss in retina must be hypoxic because the
It is mostly a bilayered system, a superficial network, and a deep high metabolic rate requires a continuous supply of oxygen.
capillary plexus; however, there are multiple layers of capillaries They observed that neovascularization always formed adjacent
in the peripapillary region, the radial peripapillary capillaries. to these nonperfused areas and, therefore, an angiogenic factor
There is only one layer of blood vessels in the periphery at must be produced by the hypoxic retina. They hypothesized
the ora serrata where the retina thins to 100 µm. The retinal that this factor X must be hypoxia-inducible and diffusible.
Fig. 18.1 Comparison of retinal (A-C) and
choroidal (D-F) vasculatures. (A) The retinal
vasculature, stained here with adenosine
434 diphosphatase (ADPase) enzyme
histochemistry, has an end-arterial hierarchy:
arteries (arrow) have a capillary-free zone,
and arterioles and then capillaries, which
Section 1

are drained by venules, and then veins


(arrowhead). (B) A capillary in the deep
capillary network has a pericyte and
endothelial cell. (C) Transmission electron
A D microscopy (TEM) of a retinal capillary shows
a pericyte (p) within the shared basement
membrane (arrow) with an endothelial cell
(e). (Courtesy of DB Archer, TA Gardiner,
Anatomy and Physiology
Basic Science and Translation to Therapy

and AW Stitt.) (D) The choriocapillaris,


p stained by alkaline phosphatase (APase)
enzyme histochemistry, has a lobular
e hierarchy. (E) When sectioned, the
endothelial cell APase activity in
c
c choriocapillaris (c) shows the broad flat
B E lumens of the choriocapillaris which are
positioned under retinal pigment epithelial
cells (top) and Bruch’s membrane between
them. (F) With TEM the thin processes of the
choriocapillaris endothelial cells (arrow) are
p apparent and, at higher magnification (inset),
the fenestrations in these endothelial cells
are visible.

e
C F

Subsequently, oxygen was measured directly in retinas of several children are placed in 40% oxygen, making their tissue further
species and it demonstrated that nonperfused areas were indeed hyperoxic, which yields vaso-obliteration (endothelial cells die
hypoxic.1,2 It was not until 1989 that factor X was discovered, and pericytes and progenitors survive).19 The only direct mea-
purified, and characterized as vascular endothelial growth factor surements of oxygen in a model of retinopathy of prematurity
(VEGF).16 This factor was first shown to be responsible for the (ROP) were performed by Ernest and Goldstick.20 They found in
increased vascular permeability seen in some retinopathies.17 kitten after 80–90% O2 that preretinal pO2 over avascular retina
was close to zero but was normal over vascularized retina. Vaso-
NORMOXIA obliteration from hyperoxia does not occur in the choroid of
The studies of Wangsa-Wirawan and Linsenmeier1 and Yu and humans and dogs19 but does occur when rats are exposed to
Cringle2 using oxygen electrodes directly assessed oxygen levels hyperoxia.21 Loss of vasculature in vaso-obliteration makes the
from choroid to vitreous in various species. The oxygen tension retina hypoxic when the child is returned to room air. Exposure
is around 70 mmHg in choroid and plummets to zero at the of the adult vasculature to hyperoxia causes constriction but not
inner segments in the dark (Fig. 18.2). Inner retina is around vaso-obliteration. During hyperoxia breathing (100% oxygen),
10–20 mmHg. There are regional variations in oxygen concentra- the inner retinal pO2 remains unchanged due to autoregulation
tion within the retina. Yu et al.18 showed that oxygen consump- while the choroidal pO2 rises to 250 mmHg in cat22 and 220 mmHg
tion in outer retina is highest in the parafoveal region while inner in the minipig, due to a lack of metabolic control of the choroidal
retinal oxygen in the fovea (approximately 5 mmHg) reflected vasculature.23
the lack of a retinal vasculature and the predominantly choroidal
source of oxygen. HYPOXIA
Complex homeostatic mechanisms are designed to maintain
HYPEROXIA O2 concentration in each cell within a narrow range. While
Life in utero is hypoxic, so when a child is born prematurely, the O2 consumption increases with the metabolic activity of the
normoxic environment is actually hyperoxic. Prematurely born organism, exposure to O2 must be limited due to the potentially
Fig. 18.2 Oxygen profile measured with
microelectrodes in cat retina during light
and dark adaptations. The schematic
at the top shows where, anatomically, 435
80 the measurements were taken from
choriocapillaris (left, retinal depth = 100)
to the internal limiting membrane (right,

Chapter 18
retinal depth = 0). (Reproduced with
Light permission from Wangsa-Wirawan ND,
60 Linsenmeier RA. Retinal oxygen. Arch
Dark
Ophthalmol 2003;121:547–55.)
Oxygen tension, mmHg

40

Retinal and Choroidal Vasculature:


20

110 100 90 80 70 60 50 40 30 20 10 0
Retinal depth, %

damaging effects of reactive oxygen species (ROS). Hypoxia, which facilitates heterodimerization, and a C-terminus, which
the state of low oxygen concentration, promotes the formation recruits transcriptional coregulatory proteins.
of blood vessels and is important for the formation of a vascular The activity of HIF depends on the intracellular levels of its
system in embryos.24 Disease occurs when the retina and choroid inducible alpha subunit. In the presence of oxygen, HIF-1α is
are deprived of adequate oxygen supply; this can also be hydroxylated on two critical proline residues (Pro402 and Pro564)
described as a mismatch of oxygen supply versus demand at in the so-called oxygen-dependent degradation domain. Three
the cellular level within ocular tissues. prolyl hydroxylases have been identified in mammalian cells
The blood O2-carrying capacity is maintained by the O2- and use O2 as a substrate to generate 4-hydroxyproline at resi-
regulated production of erythropoietin (EPO), which stimulates dues 402 and/or 564 of HIF-1α. The hydroxylation reaction
the proliferation and survival of red blood cell progenitors. also requires 2-oxoglutarate (α-ketoglutarate) as a substrate and
Semenza and coworkers25,26 performed seminal studies to iden- generates succinate as a side product. These prolyl hydroxylases
tify hypoxia-inducible factor-1 (HIF-1). HIF-1 orchestrates a have a high Km for O2 that is slightly above atmospheric con-
pleiotropic adaptive response to hypoxia by inducing the expres- centration; thus O2 is rate-limiting for enzymatic activity under
sion of more than 100 genes encoding glycolytic enzymes and physiological conditions and any change in cellular O2 concen-
glucose transporters (thereby facilitating the glycolytic switch in tration is directly transduced into changes in the rate of HIF-1α
energy metabolism typically observed under hypoxic condi- hydroxylation.29
tions), matrix metalloproteinases, and angiogenic, mitogenic, Factor-inhibiting HIF-1 (FIH-1), which was identified in a
and survival factors, including EPO.27,28 Other molecules upreg- yeast two-hybrid screen as a protein that interacts with, and
ulated by HIF-1 that have profound effects on vasculature inhibits the activity of the HIF-1α transactivation domain,30
include 5’ nucleotidase, an enzyme that is the major source of functions as asparaginyl hydroxylase.31 As in the case of the
the potent vasodilator adenosine in the body, and VEGF. HIFs prolyl hydroxylases, FIH-1 appears to use O2 and 2-oxoglutarate,32
are vital to development and, in mammals, deletion of the HIF-1 although it has a Km for O2 that is three times lower than
genes results in perinatal death. HIF-1 is expressed in all cell the prolyl hydroxylases.33 Hydroxylation provides a mechanism
types and functions as a master regulator of oxygen homeostasis for regulating protein–protein interactions, similar to the effect
by playing critical roles in embryonic development and post­ of phosphorylation and other posttranslational modifications.
natal physiology. However, this hydroxylation occurs in an O2-dependent manner,
thus establishing a direct link between cellular oxygenation and
Hypoxia-inducible factor HIF-1 activity. Following HIF-1α hydroxylation, the protein
HIF is a highly conserved transcriptional complex which is a becomes targeted for ubiquitination by an E3 ligase complex
heterodimer composed of an alpha and a beta subunit. HIF-1 (including the von Hippel–Lindau (VHL) tumor suppressor
belongs to the PER-ARNT-SIM (PAS) subfamily of the basic protein) and subsequent proteasomal degradation.
helix–loop–helix (bHLH) family of transcription factors. The Under hypoxic conditions, the HIF prolyl-hydroxylases are
alpha and beta subunit both contain an N-terminus bHLH inhibited, because these HIF prolyl-hydroxylases utilize oxygen
domain for DNA binding, a central region with PAS domain, as a cosubstrate. Hypoxia results in an increase in succinate,
Fig. 18.3 Hypoxia-inducible factor 1 (Hif-1α)
in hypoxia and hyperoxia.
Hyperoxia Ub
436 OH OH
Prolyl E3 Ligase
hydroxylase Degradation
complex
Hif-1α Hif-1α Hif-1α
Section 1

Hypoxia

Angiogenic growth factors


Hif-1β
Angiogenic survival factors
(ARNT)
Anatomy and Physiology
Basic Science and Translation to Therapy

Cytoskeletal proteins
Proapoptotic proteins
Glucose transporters
Glycolytic enzymes
Nucleus Hif-1β
(ARNT)
Transcription

Hif-1α
Hif-1β
(ARNT)

due to inhibition of the electron transport chain in the mito- system is required for embryonic survival by embryonic day 9
chondria, which serves to inhibit further HIF prolyl-hydroxylase (E9) in the mouse. In wild-type mouse embryos, HIF-1α expres-
activity. When stabilized by hypoxic conditions, HIF increases sion increases dramatically between E8.5 and E9.5, whereas
the expression of critical genes that promote survival in low- embryos that lack HIF-1α expression die between E9.5 and E10.5
oxygen conditions, including glycolytic enzymes, which allow and show cardiac malformations, vascular regression, and
ATP synthesis in an oxygen-independent manner. HIF activates massive cell death.40 Complete HIF-2α deficiency is also associ-
the transcription of genes encoding secreted signaling mole- ated with embryonic lethality41 and because the embryos survive
cules, including angiogenic growth factors and survival factors, longer than HIF-1a–/– mice, effects on multiple organ systems can
cell surface receptors, extracellular matrix proteins and modify- be demonstrated.42
ing enzymes, transcription factors, cytoskeletal proteins, pro- Complete HIF-1α deficiency results in developmental defects;
apoptotic proteins, and glucose transporters and glycolytic however, partial HIF-1α deficiency is sufficient to result in
enzymes (Fig. 18.3).29 impaired responses to physiological stimuli. A particularly dra-
HIF-induced VEGF, stromal-derived factor-1 (SDF-1), and matic example is the loss of O2 sensing in the carotid body of
EPO promote neovascularization. HIF-1 acts by binding to HIF- HIF-1a+/– mice.43 Although the carotid bodies are anatomically
responsive elements in promoters that contain the sequence and histologically normal and depolarize normally in response
NCGTG, which is present in the promoters for VEGF, SDF-1, to cyanide application, they show essentially no response to
EPO, and many other genes. In addition to hypoxia, other factors hypoxia. Thus partial HIF-1α deficiency in the carotid body
such as nuclear factor κB (NF-κB) modulate HIF-1α expression results in a complete loss of the ability to sense and/or respond
in the presence of normal oxygen pressure. Thus, conditions to changes in the arterial PO2 by stimulation of the central
such as tissue inflammation can lead to local HIF-1α expres- nervous system cardiorespiratory centers. The HIF-1 target
sion.34 HIF-1 DNA-binding activity and target gene expression genes that are critical for O2 sensing and/or efferent responses
are induced in cells exposed not only to hypoxia but also to the by the carotid body have not been identified.
iron chelator desferrioxamine or to cobalt chloride.35 Mice with HIF-1α conditionally knocked out using PAX6-Cre
A structurally and functionally related protein to HIF-1α, des- have delayed development of the outer retinal plexus but not
ignated HIF-2α, is the product of the EPAS1 gene. HIF-2α can the superficial or deep plexus.44 However, when HIF-1α was
also heterodimerize with HIF-1ß.36 HIF-1α:HIF-1ß and HIF- knocked down only in Müller cells using a Cre-LOX system, and
2α:HIF-1ß heterodimers have overlapping yet distinct target the animals were made diabetic with streptozotocin, vascular
gene specificities.37 HIF-2α, unlike HIF-1α, is not expressed in all permeability in retina was reduced and leukostasis and overpro-
cell types and HIF-2α can be inactivated by cytoplasmic seques- duction of VEGF and intercellular adhesion molecule (ICAM)-1
tration. This “compartmentalization” of oxygen-sensitive signal- were attenuated in adult mice.45
ing components also influences the hypoxic response.38,39 Another dramatic phenotype is the complete inability of HIF-
1α–/– myeloid cells (granulocytes and macrophages) to respond
HIF deficiency and its resultant pathology to inflammatory stimuli.46 Myeloid cells are dependent on gly-
O2 delivery to cells of the developing embryo becomes limited colysis for ATP generation, perhaps reflecting the hypoxic
by diffusion such that establishment of a functioning circulatory microenvironment that is often associated with inflammation
and infection. HIF-1α deficiency results in ATP deficiency, which While VEGF is critical to maintaining normal ocular func-
impairs critical myeloid cell functions such as aggregation, tion, overproduction of VEGF is deleterious. Elevated levels
motility, invasion, and bacterial killing. HIF-1 also plays of VEGF have been strongly implicated in the pathogenesis 437
critical roles in B-lymphocyte development47 and T-lymphocyte of ocular neovascular diseases such as neovascular age-related
activation.48 macular degeneration (NV in AMD)66 and proliferative diabetic

Chapter 18
retinopathy67 as well as diabetic macular edema.68 Elevated
HIF-activated genes relevant to VEGF levels are observed in central and branch retinal vein
physiological and pathological occlusion (CRVO and BRVO),69 neovascular glaucoma,70 and
ROP.71 Blocking VEGF action is now an established strategy
ocular angiogenesis for the treatment of NV in AMD, with two agents (the RNA
The paragraphs above provide a brief summary of the critical aptamer pegaptanib sodium72 and the humanized murine
role of HIF-1α in oxygen sensing, development, and physiology. monoclonal antibody antigen-binding fragment ranibizumab73)

Retinal and Choroidal Vasculature:


HIF-1α plays an equally important role in disease pathophysiol- having received regulatory approval for the intravitreal treat-
ogy, including retinal diseases. As a result, there is considerable ment of NV in AMD.
interest in HIF-1 α as a therapeutic target.49 In cardiovascular While current standard of care for NV in AMD uses intravitreal
diseases, increased HIF-1α activity induced as a result of HIF-1α delivery of an anti-VEGF agent on a repeated basis, this routine
gene therapy,50 small-molecule inhibitors of prolyl hydroxylase clinical practice does not address all the nuances of VEGF biology.
activity,51 or inhibitors of HIF-1α–VHL interaction52 may provide The biology of the VEGF proteins is extremely complex. The
a means to stimulate neovascularization in ischemic tissue. In VEGF family members are part of a superfamily of cysteine
contrast, small-molecule inhibitors of HIF-1 activity may be knot proteins and include VEGF-B, -C, -D, and placental growth
useful as antiangiogenic agents. However, because HIF-1α func- factor (PlGF). Alternative splicing events for VEGFA give rise to
tions as a global regulator of oxygen homeostasis, it may not be at least 14 subtypes of VEGF, namely, VEGF111, VEGF121, VEGF121b,
a useful therapeutic target if the treatment results in unintended VEGF145, VEGF145b, VEGF148, VEGF162, VEGF165, VEGF165b, VEGF183,
and undesirable side-effects. VEGF183b, VEGF189, VEGF189b,74 and VEGF206.75,76 Following the
An alternative therapeutic approach that may be particularly discovery of the antiangiogenic isoform of VEGF, VEGF165b, and
relevant to the treatment of ocular pathology is to focus on its associated family of isoforms, a further layer of complexity
modulation of HIF-1α target genes. However, the protein prod- was added to understanding the regulation of VEGF.
ucts of these target genes must also be delivered in a precise and All VEGF isoforms are essential regulators of angiogenesis
perfectly timed manner. EPO is an oxygen-regulated hormone and vascular permeability. VEGFs elicit their intracellular activi-
stimulating erythrocyte production and is critical for retinal ties via the activation of two receptor tyrosine kinases (RTKs):
angiogenesis. Increasing EPO expression in phase 1 of the VEGFR-1 and -2. VEGFR-1, a high-affinity fms-like tyrosine
murine ROP model (postnatal days 7–12) is protective and kinase-1,77 and VEGFR-2, a kinase insert domain-containing
results in less neovascularization during phase 2 (postnatal days receptor,78 are transmembrane glycoproteins consisting of a
12–17).53 In contrast, EPO mRNA expression levels in retina are seven-tandem immunoglobulin-like domains, which serves as
highly elevated during the hypoxia-induced proliferation phase the extracellular ligand-binding region, a single-transmembrane
of retinopathy (phase 2) and inhibition of retinal EPO mRNA domain, and a cytoplasmic domain consisting of two tyrosine
expression with RNA interference results in suppressed retinal kinase catalytic domains. Moreover, it has also been reported
neovascularization.53 that a family of cell surface glycoproteins, particularly
The best-known gene activated by HIF-1α is VEGF, first iden- neuropilin-1, act as isoform-specific coreceptors for VEGF-A.79
tified as a potent promoter of vascular permeability17 and endo- The VEGF family and their respective receptors are outlined in
thelial cell proliferation.15 VEGF has become known as a master Fig. 18.4.
regulator of angiogenesis.54 Tight control of physiologic VEGF Ligand binding to the extracellular domain of VEGFR-2 results
levels is required for proper embryological development.55 in a maximal increase of kinase activity following the induction
Although it was initially thought that the postembryonic role of of receptor dimerization and subsequent phosphorylation of
VEGF was restricted to a few processes, it is now quite clear that tyrosine residues on the intracellular domain of the receptor.80
VEGF acts as a pluripotent growth factor essential for a wide This event is crucial for the recruitment of additional signaling
variety of physiological processes,56 including maintenance of molecules that contain Src homology 2 or phosphotyrosine
the adult microvasculature,57 neuronal survival,58 and trophic binding domains, which mediate further downstream signaling
maintenance of ocular tissues. cascades.81 The association of RTKs with coreceptors, such as
NP-1, in the case of VEGFR-2:VEGF165 signaling/interaction,
VEGF in health and in ocular disease can enhance the functional signal transduction and facilitate
VEGF is produced by many cell types in the retina, including diverse cellular responses.80 VEGFR1/R2 signaling activates
retinal pigment epithelium (RPE),59 vascular endothelial cells,60 RAS, raf1, MEK1, and ERK1/ERK2 and stimulates PI3K/AKT/
pericytes,60 retinal neurons,61 Müller cells,61 and astrocytes,62 sug- PKCz/MAPK pathways to mediate proliferation, migration, and
gesting that VEGF has important functions in ocular homeosta- cell survival (Fig. 18.4).
sis. RPE-secreted VEGF plays an important role in maintaining VEGFR-2 is the major mediator of angiogenic signaling in
the choriocapillaris.11,63,64 VEGF secretion by retinal cells and the endothelial cells and is required for de novo vessel formation,
RPE is stimulated in response to hypoxia.60 VEGF administration vasculogenesis, and for angiogenesis, the formation of vessels
protects retinal neurons from apoptosis.65 Moreover, chronic from pre-existing vasculature.82 The pathways leading to VEGFR
VEGF inhibition can lead to a significant loss of retinal ganglion internalization and the role of receptor degradation in VEGF
cells in normal adult animals.65 signaling remain controversial and differ for VEGFR-1 and -2.
VEGFRs generate signal output at the plasma membrane and on including the nucleus,88 where receptors encounter distinct sig-
their way to degradation through endocytic vesicles,83 whereas, naling molecules.89
438 in unstimulated cells, VEGFR-2 is predominantly located in recy- In addition to EPO and VEGF, SDF-1 is hypoxia-regulated and
cling endosomes identified by Rab4 and/or Rab5.84 VEGFR inter- ischemic tissues express high levels of SDF-1 to recruit repara-
nalization is clathrin-mediated and transport is further directed tive cells to the injured region. SDF-1 activation of either CXCR-4
by the endosomal sorting complex required for transport pro- or CXCR-7 results in stimulation of the Ras/Raf/Mek/ERK
Section 1

teins.85 VEGFR signaling is also regulated by ubiquitination, not pathway and the PI3K/Akt pathway to promote endothelial cell
only of the receptor itself, but also of receptor-associated signal- proliferation and neovascularization. Thus ligand–receptor
ing molecules.86 Specific VEGFR trafficking regulates biological interaction of VEGF to VEGF-R1 and VEGF-R2 and SDF-1 to
output, as shown for arterial morphogenesis, for example.87 CXCR4/CXCR-7 and their subsequent internalization sets in
The molecular basis for ligand specificity of VEGFR signaling motion the cascade of cellular effects of VEGF and SDF-1
is poorly understood. It is well accepted however that VEGF (Fig. 18.4). The VEGF and SDF-1 signaling pathways appear
Anatomy and Physiology
Basic Science and Translation to Therapy

receptors can associate with distinct coreceptors such as neuro- to be intimately connected with HIF-1 activation (Fig. 18.3), as
pilins, integrins, semaphorins, or heparan sulfate glycosamino- the promoters of each of these factors contain a HIF response
glycans, and engage distinct signaling molecules giving rise to element. Because hypoxic tissue releases SDF-1 and VEGF,
specific signal output. Ligand-specific signaling may also result varying O2 concentrations would have an effect on expression of
from receptor trafficking to specific cellular compartments, the receptors for these factors.

VEGF-A
VEGF-B VEGF-E
Insulin
PIGF
IGF-1

IGF-2 SDF-1

VEGF-R1 VEGF-R2 IGF-1R CXCR4/CXCR7

Signaling by
intracellular RAS PI3K/Akt PLCg
translocation

RAF1 PKCz PKC

MEK1 MAPK

ERK1

ERK2

Fig. 18.4 Vascular endothelial growth factor (VEGF) family and its receptors. PlGF, placental growth factor; IGF, insulin-like growth factor;
SDF, stromal-derived factor.
Bone marrow-derived progenitor cells endothelial-like cells109; however, the blood vessels formed by
the endothelial cells of CD14+ origin eventually generate patho-
(BMPC) and vascular repair
logical blood vessels with increased permeability and contribute 439
In conditions like diabetic retinopathy and ROP, areas of retinal to the pathology of diabetic retinopathy.
vasodegeneration occur and lead to retinal ischemia which in We and others have been particularly interested in a novel
turn induces the expression of hypoxia-regulated angiogenic

Chapter 18
factor expressed in increased concentrations in hypoxic tissue,
factors. Typically, BMPCs robustly respond to these factors, insulin-like growth factor-binding protein 3 (IGFBP-3). While
including VEGF and SDF-1.90 the standard IGF-dependent actions of the family of IGF-binding
Importantly, all hypoxia-regulated angiogenic factors are proteins have been well described, recently several IGF-
modulators of bone marrow-derived stem cells. Specifically, independent actions have been discovered for IGFBPs, including
hematopoietic stem cells and other BMPCs reside in the bone IGFBP-3. These IGF-1 independent actions have been charac­
marrow and are mobilized into the peripheral circulation by terized as regulating cell fate and apoptosis.110–113 The role of

Retinal and Choroidal Vasculature:


increased levels of EPO, SDF-1, and VEGF released by the isch- IGFBP-3 in the control of cell growth remains an area under
emic tissue. Hematopoietic stem cell/BMPC mobilization occurs intense study, since IGFBP-3 may enhance or suppress cell
in response to vascular injury throughout the body and, when growth, depending on specific conditions.112,114 In the retina,
functioning properly, leads to revascularization of injured multiple forms of IGFBP (2–5) are secreted by retinal endothelial
areas.91 In healthy individuals, bone marrow-derived CD34+ cells.113 IGFBP-3 has been shown to enhance cell proliferation
endothelial progenitor cells as well as other BMPC successfully in retinal endothelial cells and to decrease the formation of
orchestrate the reparative process. These BMPC demonstrate a neovascular tufts in a murine model of oxygen-induced reti-
limited ability to differentiate directly into endothelial cells and nopathy.115,116 These studies suggest that IGFBP-3 may be
form components of new blood vessels by vasculogenesis, but vascular-protective in the retina. Recently, we also have dem-
show marked ability to provide paracrine support for the resi- onstrated that IGFBP-3 is neuroprotective in the retina and
dent vasculature. This paracrine support facilitates resident reduces injury-induced retinal inflammation.117
endothelial cell recovery.

Disease-associated BMPC dysfunction Key factors that modulate VEGF function


In diabetes, for example, BMPC are dramatically altered and in the retina
cannot facilitate the repair process. Diabetic individuals have The retina is one of the last organs to become vascularized in the
fewer circulating CD34+ cells and an increased number of fetus. The vasculogenic part of the process of human retinal
inflammatory BMPC such as CD14+ cells than nondiabetics.92–94 vascularization begins in about gestational week 14 and appears
This diabetes-related bone marrow dysfunction is closely linked to depend on a physiological hypoxia7,118 brought about by an
to the impaired healing response experienced by many diabetic increase in metabolic demand within the developing retina.3–5
patients and to the vasodegenerative aspect of diabetic macro- This physiological hypoxia induces the local release of VEGF
and microvascular complications.94–96 Diabetes-induced BMPC which, together with IGF-1, regulates angiogenesis and therefore
defects occur in part due to uncoupling of nitric oxide synthases, normal vascularization of the retina.118,119 Retinal vascularization
enhanced NADPH oxidase activity, and increased generation of is complete by 36–40 WG.
ROS such as superoxide and peroxynitrite (ONOO-)97 within ROP is a two-phase disease in which the phases are mirror
BMPC. While stem and progenitor cells are deemed more resis- images; the controlling growth factors are deficient in phase 1
tant to oxidative stress,98 the highly oxidative diabetic milieu has and in excess in phase 2. Phase 2 involves uncontrolled prolifera-
a clearly detrimental effect on the function of these cells.99 Pro- tive growth of retinal blood vessels in response to hypoxia. The
longed oxidant exposure reduces reparative function100 by therapeutic intention would be to prevent the first phase of ces-
impairing antioxidant defense enzymes. Previously, we and sation of vessel and neural retinal development and then the
others have shown that diabetic CD34+ cells exhibit decreased second destructive phase would be prevented. This has been
migration and adhesion activities in vitro, and consequently successfully performed using exogenous EPO, VEGF, and
reduce recruitment to areas of injury.96 In addition to oxidative IGFBP-3 during phase 1 to prevent phase 2. These two phases,
stress, other key mechanisms implicated in diabetes-induced seen in premature infants, can be duplicated in animal models
BMPC dysfunction include a reduction of cathepsin L activity101 of ROP. Hyperoxia causes vaso-obliteration and cessation of
and an upregulation of thrombospondin-1.100,102 While these normal retinal blood vessel development, which mimics phase
functional defects are profound, strategies that successfully 1 of ROP. IGF-1 levels rise in the third trimester of pregnancy
reverse BMPC defects in diabetics have included: (1) enhance- but not in the preterm infant. The low IGF-1 in the preterm infant
ment of angiogenic stimulus by increasing BMPC mobilization is due to the infant no longer being exposed to the support of
using granulocyte colony-stimulating factor and targeting the maternal environment, including maternal sources of IGF-1.
SDF-1103; (2) use of nitric oxide donors to correct migration and IGF-1 levels rise slowly after preterm birth, as these babies are
promote cell deformability104; (3) enhancing cell interactions unable to produce adequate IGF-1 compared to term infants.120
with substrate proteins to increase attachment to basement In these premature infants, IGF-1 is further reduced by poor
membranes105; (4) reducing high levels of endogenous trans- nutrition,121 acidosis, hypothyroxinemia, and sepsis.122 IGF-1
forming growth factor-β to normal levels106; and (5) treatment appears important for retinal and brain growth.119 Low levels of
with rosiglitazone107 or atorvastatin.108 IGF-1 appear to play an important role in the early cessation of
In addition to CD34+ cells, other populations of bone retinal growth that precipitates ROP.
marrow cells may attempt vascular repair, such as CD14+ cells, IGF-1 mediates its effects through activation of the IGF-1 recep-
discussed above, which can, under select circumstances, form tor (IGF-1R) (Fig. 18.3). IGF-1R is a receptor tyrosine kinase and
is well established as a key regulator of cell growth and survival independent predictors.128 These observations are supported by
with activation of Ras-ERK pathway, the PI3K/Akt pathway and several other prospective studies, in which tissue plasminogen
440 PKC (Fig. 18.4). Insulin and IGF-2 can also signal using the IGF-R. activator, another marker of reduced fibrinolysis,129 and von
There is also a growing body of data to support a role for the Willebrand factor, a marker of endothelial injury, were predic-
structurally and functionally related insulin receptor (IR) in cell tive.130 In one clinical study of type 2 diabetics, adhesion mol-
survival, even though its major function has been to modulate ecules were higher in subjects with retinopathy than those
Section 1

metabolism. Bidirectional cross-talk between IGF-1R and IR is without,131 and in another population-based cohort, composite
observed, where specific inhibition of either receptor confers a scores of both inflammatory and endothelial function markers
compensatory increase in activity for the reciprocal receptor. were strongly associated with the presence of diabetic retinopa-
Although fluctuating oxygen has long been associated with thy.132 Similarly, E-selectin values were found to be increased
the development of ROP, oxygen-regulated factors like VEGF in a group with type 1 diabetes and retinopathy.133 Adiponectin
appear to be directly modulated by IGF-1. IGF-1 is a key growth was increased in the advanced stages of retinopathy.134 These
Anatomy and Physiology
Basic Science and Translation to Therapy

factor in early retinal development. IGF-1 controls maximum results should be interpreted cautiously, however, as serum
VEGF activation of the Akt endothelial cell survival pathway markers are not necessarily indicators of tissue events. However,
(Fig. 18.3). Thus loss of IGF-1 leads to loss of VEGF signaling P-selectin, ICAM-1, and polymorphonuclear leukocyte numbers
and retinal vaso-obliteration. This vaso-obliteration leads to are all elevated in human diabetic retina.135 Experimental work
phase 2 of ROP which is the proliferative phase. At this point, in diabetic rat retinas later demonstrated that inflammatory
suppression of IGF-1 and VEGF can reduce neovascularization. cytokine-mediated leukostasis occurs early in diabetic retina
Thus IGF-1 is critical to normal retinal vascular development and neutralizing ICAM-1 and CD-18 prevents it.136–138 From these
and a lack of IGF-1 in the early neonatal period is associated with studies, it appears that inflammation-mediated EC injury and
lack of vascular growth and with subsequent proliferative ROP. vaso-occlusion may cause nonperfusion and subsequrent
In IGF-1-null mice, the retinal blood vessels grow more slowly hypoxia in diabetic retina.139,140
than in those of normal mice, a pattern very similar to that seen The hypothesis that inflammation is critical to the develop-
in premature babies with ROP. ment of diabetic retinopathy arose from initial reports that dia-
betic patients taking salicylates to treat rheumatoid arthritis had
ADULT RETINAL HYPOXIA AND ETIOLOGY a lower-than-expected incidence of diabetic retinopathy.141 The
subsequent decades demonstrated an increase in inflammatory
Diabetic retinopathy markers and growth factors in the diabetic vitreous and retina.
Much like ROP, diabetic retinopathy has a vaso-obliteration Recently microarray analyses substantiated a marked inflamma-
phase that leads to a proliferative phase. The vaso-obliteration tory response in the retinas of diabetic rodents.142 Confirmation
is not due to hyperoxia but rather to vaso-occlusion and vaso­ of the importance of inflammatory factors and growth factors is
degeneration of the microvasculature, setting up the ischemic supported by additional rodent studies that show that blocking
environment that leads to the vasoproliferative end-stage condi- these factors prevents the development of lesions characteristic
tion. Clinically the early pathology has been classified as non- of the retinopathy in animals. Specific inflammatory molecules
proliferative (microaneurysms, exudates, leakage, capillary that have been shown to contribute to structural or functional
nonperfusion) resulting in hypoxia and the end-stage pathology alterations that are characteristic of the retinopathy include
as proliferative (preretinal neovascularization). The purely vaso- NF-κβ143; inducible nitric oxide synthase143; cytochrome c
degenerative, nonproliferative form of the disease is by far the oxidase143; ICAM140; 5-lipoxygenase144; interleukin-1β145; tumor
most common and represents a disease of the neurovascular necrosis factor (TNF)-α146; and VEGF.67,147 Inflammation can
unit, resulting in dysfunction and eventual death of several of intensify the generation of AGEs that are produced in response
the key cells that maintain the BRB: pericytes, vascular endothe- to hyperglycemia and increased oxidative stress.
lial cells, Müller glia and neurons. Kohner and Henkind ele-
gantly demonstrated that, in diabetic individuals, areas of Retinal vein occlusion (RVO)
nonperfusion on fluorescein angiography are associated with Occlusion of large retinal blood vessels is a common occurrence,
acellular capillaries in trypsin digests.123 Direct measurement of which results in retinal hypoxia. RVO is the second most
oxygen in diabetic cat retinas demonstrated that even small common sight-threatening retinal vascular disorder after dia-
aneurysms can result in a decrease in retinal interstitial oxygen.124 betic retinopathy.148 RVO represents an obstruction of the retinal
There are many mechanisms implicated in the pathogenesis venous system that involves either the central retinal vein or
of diabetic retinopathy but one that has gained considerable a branch retinal vein. RVO is typically due to external compres-
attention in the last decade is inflammation. The environment sion or disease of the vein wall, such as is seen in vasculitis.149
of hyperglycemia, abnormal lipids, increased oxidative stress, Central retinal artery occlusion (CRAO) results in sudden, cata-
elevated serum and tissue advanced glycation endproducts strophic visual loss and branch retinal arteriolar occlusion
(AGE)/receptor for AGE, increased serum/tissue cytokines, (BRAO) causes sudden segmental visual loss and may recur to
elevated blood pressure, and endoplasmic reticulum stress are involve other branch retinal arterioles. CRAO studies have
the likely initiators of inflammation.125 Pathways of inflamma- shown that the ischemic retinal whitish opacity and swelling
tion converge with pathways of endothelial dysfunction and of CRAO are essentially located in the perifoveolar region of
coagulation to accelerate the pathogenesis of this disease.126 the macula. Oxygen supply and nutrition from the choroidal
Initially nonspecific indicators of inflammation such as white- vascular bed to the thinner peripheral retina help in its much
cell count and fibrinogen were found to be predictive of incident longer survival and the maintenance of peripheral visual fields.
diabetes.127 Subsequently plasminogen activator inhibitor-1 The diagnosis is clinical and based on the observation of the
(PAI-1), C-reactive protein, and fibrinogen were shown to be ocular fundus: venous dilatation and tortuosity, flame-shaped
retinal hemorrhages, retinal edema and cotton-wool exudates It is also important to consider that fibrinolytic agents can dis-
affecting all the retinal sectors (in CRVO) or the sector of the solve only platelet fibrin emboli.157 Retinal emboli are made of
retina drained by the affected vein in BRVO. Open-angle glau- 74% cholesterol, 10.5% calcific material, and only 15.5% of plate- 441
coma is the most frequent local alteration predisposing to RVO let fibrin. Fibrinolytic agents cannot dissolve cholesterol or calci-
as it compromises venous outflow by increasing intraocular fied material. Therefore, there is no scientific rationale for the use

Chapter 18
pressure. Raised intraocular pressure causes external compres- of fibrinolytic agents in at least 85% of CRAO cases. For the
sion of the central retinal vein as it passes through the lamina remaining 15%, if the diagnosis is made within 15 days from
cribrosa, resulting in turbulent blood flow distal to the compres- onset of clinical manifestations, low-molecular-weight heparins
sion leading to thrombus formation. at anticoagulant doses are typically used 10–15 days followed by
The natural history of RVO is highly variable; in some cases half dose for a total of 90 days.158 No data are available on the
the retinal findings progressively disappear and there is a possible role of antithrombotic/antiplatelet strategies in the
good visual outcome, while in other cases severe complications long-term prevention of recurrent RVO159; however, they are

Retinal and Choroidal Vasculature:


like ocular neovascularization (proliferative retinopathy), vitre- routinely prescribed to most patients with diabetes, hyperten-
ous hemorrhage, neovascular glaucoma, and macular edema sion, or arterial disease. Of Virchow’s three classical factors that
develop. Using retinal oximetry in BRVO, venular saturation play a role in thrombogenesis – stasis, vessel wall damage, and
was found to be highly variable between patients (12–93%)150; hypercoagulability – the first two have long been reported in
this was attributed to variable severity of the disease, recana- patients with RVO, whereas the third has not been sufficiently
lization, degree of occlusion, collateral vasculature, or tissue investigated until recently.
atrophy. The majority of patients with CRVO have signs of
macular edema at presentation whereas only 5–15% of eyes Sickle-cell disease (SCD)
with BRVO develop macular edema over the first year. Venous The pathological processes involved in SCD can affect virtually
collateral channels represent tortuous vessels that develop every vascular bed including the retina and in its advanced
locally, mainly around the optic disc, and are usually associ- stages has the potential to cause blindness.160 Classification of
ated with a long-standing vascular obstruction. Vitreous hem- ophthalmic manifestations of SCD in the retina is based on the
orrhage develops in 10% of eyes with CRVO within 9 months presence or absence of vascular proliferation, which is the most
of presentation and in about 40% of eyes with BRVO.148 If important precursor of blinding complications and precedes
there is restoration of circulation in the central retinal artery, development of a vitreous hemorrhage or retinal detachment.
the retinal capillaries in the central, thickest part of the macular Ischemia occurs due to obstruction of capillaries with sickle red
region do not refill because of compression by the surround- blood cells and subsequent thrombus formation. The subsequent
ing swollen superficial retinal tissue, resulting in the “no-reflow scenario for disease pathogenesis is similar to that for ROP,
phenomenon,”151and consequently in permanent ganglion cell diabetic retinopathy, and RVO, described above, except that
death in the nonperfused retina; the area of central retinal there is no retinal leakage of retinal blood vessels only from
capillary nonfilling may vary from eye to eye depending upon preretinal neovascularization, even though there is loss of
the severity of retinal swelling in the macular region. This peripheral vasculature and elevation of VEGF.161 Neovascular-
results in the variable size of the permanent central scotoma. ization that occurs can lead to fibrosis that can then result in
In considering the outcome of CRAO, it is important to con- retinal detachment.
sider retinal tolerance time to acute retinal ischemia. The chance
of recovery of vision only exists as long as the retina has Ocular ischemic syndrome (OIS)
reversible ischemic damage. The retina suffers no detectable OIS occurs at a mean age of 65 years and is rare before the
damage with CRAO of up to 97 minutes, but after that, the age of 50. Men are affected twice as often as women,162 reflect-
longer the CRAO, the more extensive the irreversible ischemic ing their higher incidence of atherosclerotic disease; however,
retinal damage. no racial predilection exists. Bilateral involvement may occur
It is generally believed that the CRAO is always either embolic in up to 22% of cases.163,164 Sturrock and Mueller estimated 7.5
or thrombotic in origin. Embolism is far more common than cases per million persons every year,165 but this is likely an
thrombosis,152 as was pointed out almost a century ago by underestimation as OIS can be easily misdiagnosed. Kearns166
Coats.153,154 An inflammatory etiology is also postulated in RVO reported that, of patients with occlusion of the internal carotid
as RVO is associated with immunological diseases in young artery undergoing surgical anastomosis between the superficial
patients. To support this contention, patients can experience temporal artery and the middle cerebral artery, 18% presented
prompt resolution of symptoms with the use of periocular ste- with OIS. Up to 29% of patients with a symptomatic carotid
roids.155 Giant cell arteritis is an important and well-known cause artery occlusion manifest retinal vascular changes that are
of CRAO, and is an ophthalmic emergency because of the high usually asymptomatic; however, 1.5% of them progress per year
risk of bilateral visual loss, which is preventable. to symptomatic OIS.167 OIS develops especially in patients with
Elevated levels of PAI-1, lipoprotein(a), and hyperhomocyste- poor collateral circulation between the internal and external
inemia, and low circulating levels of folic acid, vitamin B12 carotid arterial systems. Insufficient collateral vascular flow in
and vitamin B6 have been implicated in the pathogenesis of OIS patients explains the frequent association with cerebral
this disease.156 While the pathogenesis is complex and largely infarctions and the poor neurologic outcomes.168 Degree of inter-
unknown, medical treatment includes identification and correc- nal carotid artery stenosis, presence of collateral vessels, and
tion of vascular risk factors. The use of fluorescein fundus angi- compensation by collaterals are important in assessing OIS
ography before (showing occlusion of the central retinal artery) disease severity. Also if the OIS is bilateral or there are associ-
and immediately after thrombolysis may show improvement in ated systemic vascular diseases, this tends to worsen the
and/or restoration of retinal circulation and retinal function. prognosis.
GA: Nonatrophic GA: Atrophic Wet AMD

442
Section 1

A D G
Anatomy and Physiology
Basic Science and Translation to Therapy

B E H

c c c
C F I

Fig. 18.5 The choroid and retinal pigment epithelium (RPE) in geographic atrophy (A-F) and wet (neovascular) age-related macular degeneration
(AMD) (G-I). (A) Alkaline phosphatase (APase)-stained choroidal blood vessels in a nonatrophic region of geographic atrophy (GA) choroid.
(B) When sectioned, this area has normal-appearing RPE cells (arrowhead) and viable choriocapillaris lumens filled with serum APase.
(C) Higher magnification shows the intimate relationship between RPE, Bruch’s membrane, and choriocapillaris (c). (D) The atrophic region of
this GA choroid has no RPE and an attenuated choriocapillaris with narrow lumens. (E) Cross-sections of this area demonstrate the APase
reaction product in a few remaining choriocapillaris lumens and the endothelial cells of artery (bottom). (F) At higher magnification it is apparent
that the APase– capillaries (c) are only collagenous tubes between intercapillary septa. (G) In a flat choroid preparation of a wet AMD subject,
a large fan-shaped choroidal neovascular formation is present and the RPE monolayer to the left appears normal. (H) A section of this
subject immediately in advance of the choroidal neovascularization demonstrates viable hypertrophic RPE (arrowhead) over an attenuated
choriocapillaris. (I) At higher magnification, the remnants of atrophic choriocapillaris lumens (c) are present between intercapillary septa and
a single APase+ lumen remains, yet RPE are still present.

Retinal detachment center of our understanding of ocular angiogenesis. Retinal


neovascularization is defined as a state where new pathologic
Retinal detachment causes the sensory retina to be distant from
vessels originate from the existing retinal veins and extend
choriocapillaris, thus reducing its oxygen supply and resulting
along the inner surface of the retina.
in photoreceptor degeneration. Linsenmeier and Padnick-
Silver169 demonstrated in cat that retinal detachment resulted in Vascular permeability
a significant decrease in outer retinal oxygen, having a serious
Growth factors such as VEGF have been implicated in both
metabolic effect on photoreceptors. In subsequent work, Linsen-
retinal neovascularization and vascular hyperpermeability.173
meier demonstrated that hyperoxia may have clinical benefit
Antibodies to VEGF improve visual function in patients with
after retinal detachment because it normalized photoreceptor
diabetic macular edema.174 In experimental diabetes and in
oxygen consumption and prevented photoreceptor dysfunc-
VEGF-induced permeability, alterations of the tight junction (TJ)
tion.170 Furthermore, hyperoxia prevents proliferation and reac-
complex of microvascular endothelial cells alters the BRB.175
tivity of retinal Müller cells in the detached feline retina, limiting
A role of classical PKC isoforms (cPKCs) but especially PKCβ
retinal injury.171
in regulating VEGF-induced vascular permeability is well
accepted.176 VEGF activation of PKCβ leads to phosphorylation
Consequences of retinal ischemia and reorganization of the TJ complex, increasing vessel wall
In 1971, Judah Folkman reported in the New England Journal permeability.177 VEGF increases the phosphorylation of the
of Medicine that all cancer tumors are angiogenesis-dependent.172 TJ protein, occludin, at multiple sites,178 including Ser490.179
If a tumor could be stopped from growing its own blood Phosphorylation at Ser490 allows subsequent ubiquitination
supply, he surmised, it would wither and die. Though his and endocytosis of occludin and fosters breaks in the TJ.180
hypothesis was initially disregarded by most experts in the Despite this well-supported mechanism, the PKCβ inhibitor
field, Folkman persisted with his research. After more than a ruboxistaurin failed to achieve Food and Drug Administration
decade, his theory became widely accepted and is now at the approval for diabetic retinopathy. cPKC inhibition also failed to
prevent TNF-α-induced permeability,181 a proinflammatory REFERENCES
cytokine also implicated in diabetic retinopathy. Thus targeting 1. Wangsa-Wirawan ND, Linsenmeier RA, Retinal oxygen. Fundamental and
clinical aspects. Arch Ophthalmol 2003;121:547–57.
permeability in the retina continues to be a difficult clinical 2. Yu DY, Cringle SJ. Oxygen distribution and consumption within the retina in 443
problem. vascularised and avascular retinas and in animal models of retinal disease.
Prog Retin Eye Res 2001;20:175–208.
3. Chan-Ling T, McLeod DS, Hughes S, et al. Astrocyte–endothelial cell relation-
ADULT CHOROIDAL ISCHEMIA

Chapter 18
ships during human retinal vascular development. Invest Ophthalmol Vis Sci
2004;45:2020–32.
The choroidal vasculature is an expansive vascular plexus that 4. Hasegawa T, McLeod DS, Prow T, et al. Vascular precursors in developing
human retina. Invest Ophthalmol Vis Sci 2008;49:2178–92.
handles 90% of the blood from the ophthalmic artery. It was 5. McLeod DS, Hasegawa T, Prow T, et al. The initial fetal human retinal vas-
often assumed that the choriocapillaris was so vast that genera- culature develops by vasculogenesis. Dev Dyn 2006;235:3336–47.
tion of choroidal ischemia rarely occurred. However, there are 6. Stone J, Itin A, Alon T, et al. Development of retinal vasculature is mediated
by hypoxia-induced vascular endothelial growth factor (VEGF) expression by
several diseases in which the loss of choroidal vasculature is neuroglia. J Neurosci 1995;15:4738–47.

Retinal and Choroidal Vasculature:


extensive, resulting in apparent choroidal ischemia. 7. Chan-Ling T, Gock B, Stone J. The effect of oxygen on vasoformative cell
division: Evidence that ‘physiological hypoxia’ is the stimulus for normal
Diabetic choroidopathy was first described by Hidiyat and retinal vasculogenesis. Invest Ophthalmol Vis Sci 1995;36:1201–14.
Fine.182 The use of alkaline phosphatase enzyme histochemistry 8. Cogan DG, Kuwabara T. The mural cell in perspective. Arch Ophthalmol
1967;78:133–9.
on human choroid permitted quantification of vascular loss in 9. Hasegawa T, McLeod DS, Bhutto IA, et al. The embryonic human choriocapil-
choroid. Viable vessels were positive for alkaline phosphatase laris develops by hemo-vasculogenesis. Dev Dyn 2007;236:2089–100.
10. McLeod DS, Lutty GA. High resolution histologic analysis of the human
while acellular, dysfunctional capillaries lacked it, and choroidal choroidal vasculature. Invest Ophthalmol Vis Sci 1994;35:3799–811.
neovascularization had the greatest activity.10 This technique 11. Blaauwgeers HG, Holtkamp GM, Rutten H, et al. Polarized vascular
demonstrated that there was four times greater loss in chorio- endothelial growth factor secretion by human retinal pigment epithelium and
localization of vascular endothelial growth factor receptors on the inner cho-
capillaris in diabetic subjects than in aged control subjects.183 riocapillaris. Evidence for a trophic paracrine relation. Am J Pathol 1999;
Loss of alkaline phosphatase activity was associated with the 155:421–8.
12. Lutjen-Drecoll E. Choroidal innervation in primate eyes. Exp Eye Res 2006;82:
presence of polymorphonuclear leukocytes.184 Areas of chorio- 357–61.
capillaris loss were also associated with choroidal neovascular- 13. Michaelson IC. The mode of development of the vascular system of the retina,
with some observations on its significance for certain retinal diseases.
ization and Bruch’s membrane deposits. A consequence of Trans Ophthalmol Soc UK 1948;68:137–80.
choriocapillaris dropout is loss of outer retinal oxygenation. This 14. Michaelson IC. Retinal circulation in man and animals. Springfield: CC
may be the reason for loss of blue cones and other cones in Thomas; 1954.
15. Wise GN. Retinal neovascularization. Trans Am Ophthalmol Soc 1956;96:
diabetic retina when there is no retinopathy present.185,186 729–826.
Decreased choroidal blood flow has been measured in 16. Leung DW, Cachianes G, Kuang WJ, et al. Vascular endothelial growth factor
is a secreted angiogenic mitogen. Science 1989;246:1306–9.
AMD.187–190 Using alkaline phosphatase activity and quantifica- 17. Senger DR, Galli SJ, Dvorak AM, et al. Tumor cells secrete a vascular perme-
tion of RPE in AMD, the loss of choriocapillaris has been dem- ability factor that promotes accumulation of ascites fluid. Science 1983;219:
983–5.
onstrated in exudative or wet AMD as well as geographic 18. Yu DY, Cringle SJ, Su EN. Intraretinal oxygen distribution in the monkey
atrophy (Fig. 18.5). RPE loss occurs first in geographic atrophy retina and the response to systemic hyperoxia. Invest Ophthalmol Vis Sci
2005;46:4728–33.
and then choriocapillaris degeneration occurs. Although some 19. McLeod DS, Brownstein R, Lutty GA. Vaso-obliteration in the canine model
capillaries survive in the area of RPE atrophy, a 50% loss of of oxygen-induced retinopathy. Invest Ophthalmol Vis Sci 1996;37:300–11.
vasculature occurs and the surviving capillaries are highly con- 20. Ernest JT, Goldstick TK. Retinal oxygen tension and oxygen reactivity in reti-
nopathy of prematurity in kittens. Invest Ophthalmol Vis Sci 1984;25:
stricted.191,192 This undoubtedly contributes to photoreceptor loss 1129–34.
in the area of degeneration. Capillary loss occurs in exudative 21. Hardy P, Peri KG, Lahaie I, et al. Increased nitric oxide synthesis and action
preclude choroidal vasoconstriction to hyperoxia in newborn pigs. Circ Res
AMD as well but in the presence of a complete RPE monolayer 1996;79:504–11.
(Fig. 18.5). This suggests that the RPE in this area are hypoxic 22. Linsenmeier RA, Yancy CM. Effects of hyperoxia on the oxygen distribution
in the intact cat retina. Invest Ophthalmol Vis Sci 1989;30:612–8.
and VEGF production may be increased, causing choroidal neo- 23. Pournaras CJ, Riva CE, Tsacopoulos M, et al. Diffusion of O2 in the retina of
vascularization. Large-vessel stenosis is also often observed in anesthetized miniature pigs in normoxia and hyperoxia. Exp Eye Res 1989;
AMD choroid, suggesting that the choriocapillaris blood supply 49:347–60.
24. Benizri E, Ginouves A, Berra E. The magic of the hypoxia-signaling cascade.
may be limited as well. Unfortunately, we have no way of mea- Cell Mol Life Sci 2008;65:1133–49.
suring oxygen directly in the choroid so we can only assume 25. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo
protein synthesis binds to the human erythropoietin gene enhancer at a site
ischemia occurs when there is loss of viable choriocapillaris and required for transcriptional activation. Mol Cell Biol 1992;12:5447–54.
stenosis of large and intermediate choroidal blood vessel. 26. Wang GL, Jiang BH, Rue EA, et al. Hypoxia-inducible factor 1 is a basic-helix-
loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad
Sci U S A 1995;92:5510–4.
CONCLUSIONS 27. Gariboldi MB, Ravizza R, Monti E. The IGFR1 inhibitor NVP-AEW541 dis-
rupts a pro-survival and pro-angiogenic IGF-STAT3-HIF1 pathway in human
Both too little and too much oxygen can be damaging to the glioblastoma cells. Biochem Pharmacol 2010;80:455–62.
28. Jiang BH, Zheng JZ, Leung SW, et al. Transactivation and inhibitory domains
retina. Retina cannot function properly nor survive without the of hypoxia-inducible factor 1alpha. Modulation of transcriptional activity by
support of two vasculatures: retinal and choroidal. If either vas- oxygen tension. J Biol Chem 1997;272:19253–60.
29. Semenza GL. Hydroxylation of HIF-1: oxygen sensing at the molecular level.
culature is dysfunctional, retinal hypoxia occurs and HIF-1α Physiol (Bethesda) 2004;19:176–82.
becomes stable and induces expression of key factors to assist 30. Mahon PC, Hirota K, Semenza GL. FIH-1: a novel protein that interacts with
in retinal cell survival. Reduced retinal oxygenation causes HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity.
Genes Dev 2001;15:2675–86.
impaired neuroretinal activity in young healthy persons.193 One 31. Lando D, Peet DJ, Whelan DA, et al. Asparagine hydroxylation of the HIF
key factor upregulated by HIF-1α is VEGF, which stimulates transactivation domain a hypoxic switch. Science 2002;295:858–61.
32. Lee C, Kim SJ, Jeong DG, et al. Structure of human FIH-1 reveals a unique
new angiogenesis and increased vascular permeability. These active site pocket and interaction sites for HIF-1 and von Hippel–Lindau.
pathological events can be controlled by destroying ischemic J Biol Chem 2003;278:7558–63.
33. Kukkola L, Koivunen P, Pakkanen O, et al. Collagen prolyl 4-hydroxylase
retina (photocoagulation), inhibiting VEGF, or reducing levels of tetramers and dimers show identical decreases in Km values for peptide sub-
HIF-1α. strates with increasing chain length: mutation of one of the two catalytic sites
in the tetramer inactivates the enzyme by more than half. J Biol Chem 64. Saint-Geniez M, Kurihara T, Sekiyama E, et al. An essential role for RPE-
2004;279:18656–61. derived soluble VEGF in the maintenance of the choriocapillaris. Proc Natl
34. van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor- Acad Sci U S A 2009;106:18751–6.
444 1alpha by NF-kappaB. Biochem J 2008;412:477–84. 65. Nishijima K, Ng YS, Zhong L, et al. Vascular endothelial growth factor-A is
35. Wang GL, Semenza GL. Desferrioxamine induces erythropoietin gene expres- a survival factor for retinal neurons and a critical neuroprotectant during the
sion and hypoxia-inducible factor 1 DNA-binding activity: implications for adaptive response to ischemic injury. Am J Pathol 2007;171:53–67.
models of hypoxia signal transduction. Blood 1993;82:3610–5. 66. Ng EW, Adamis AP. Targeting angiogenesis, the underlying disorder in neo-
36. Tian H, McKnight SL, Russell DW. Endothelial PAS domain protein 1 vascular age-related macular degeneration. Can J Ophthalmol 2005;40:
Section 1

(EPAS1), a transcription factor selectively expressed in endothelial cells. 352–68.


Genes Dev 1997;11:72–82. 67. Kunz Mathews M, Merges C, McLeod DS, et al. Vascular endothelial growth
37. Sowter HM, Raval RR, Moore JW, et al. Predominant role of hypoxia-inducible factor (VEGF) and vascular permeability changes in human diabetic retinopa-
transcription factor (Hif)-1alpha versus Hif-2alpha in regulation of the tran- thy. Invest Ophthalmol Vis Sci 1997;38:2729–41.
scriptional response to hypoxia. Cancer Res 2003;63:6130–4. 68. Starita C, Patel M, Katz B, et al. Vascular endothelial growth factor and the
38. Park SK, Dadak AM, Haase VH, et al. Hypoxia-induced gene expression potential therapeutic use of pegaptanib (Macugen) in diabetic retinopathy.
occurs solely through the action of hypoxia-inducible factor 1alpha (HIF- Dev Ophthalmol 2007;39:122–48.
1alpha): role of cytoplasmic trapping of HIF-2alpha. Mol Cell Biol 2003;23: 69. Aiello LP, Cahill MT, Cavallerano JD. Growth factors and protein kinase C
Anatomy and Physiology

4959–71. inhibitors as novel therapies for the medical management diabetic retinopa-
Basic Science and Translation to Therapy

39. Makino Y, Cao R, Svensson K, et al. Inhibitory PAS domain protein is a nega- thy. Eye (Lond) 2004;18:117–25.
tive regulator of hypoxia-inducible gene expression. Nature 2001;414:550–4. 70. Tripathi RC, Li J, Tripathi BJ, et al. Increased level of vascular endothelial
40. Compernolle V, Brusselmans K, Franco D, et al. Cardia bifida, defective heart growth factor in aqueous humor of patients with neovascular glaucoma.
development and abnormal neural crest migration in embryos lacking Ophthalmology 1998;105:232–7.
hypoxia-inducible factor-1alpha. Cardiovasc Res 2003;60:569–79. 71. Lashkari K, Hirose T, Yazdany J, et al. Vascular endothelial growth factor
41. Peng J, Zhang L, Drysdale L, et al. The transcription factor EPAS-1/hypoxia- and hepatocyte growth factor levels are differentially elevated in patients
inducible factor 2alpha plays an important role in vascular remodeling. with advanced retinopathy of prematurity. Am J Pathol 2000;156:
Proc Natl Acad Sci U S A 2000;97:8386–91. 1337–44.
42. Scortegagna M, Morris MA, Oktay Y, et al. The HIF family member EPAS1/ 72. Chakravarthy U. Age related macular degeneration. Br Med J 2006;333:
HIF-2alpha is required for normal hematopoiesis in mice. Blood 2003;102: 869–70.
1634–40. 73. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizumab for neovascular age-
43. Kline DD, Peng YJ, Manalo DJ, et al. Defective carotid body function and related macular degeneration. N Engl J Med 2006;355:1419–31.
impaired ventilatory responses to chronic hypoxia in mice partially deficient 74. Miller-Kasprzak E, Jagodzinski PP. 5-Aza-2’-deoxycytidine increases the
for hypoxia-inducible factor 1 alpha. Proc Natl Acad Sci U S A 2002;99: expression of anti-angiogenic vascular endothelial growth factor 189b variant
821–6. in human lung microvascular endothelial cells. Biomed Pharmacother 2008;
44. Caprara C, Thiersch M, Lange C, et al. HIF1A is essential for the development 62:158–63.
of the intermediate plexus of the retinal vasculature. Invest Ophthalmol Vis 75. Anthony FW, Wheeler T, Elcock CL, et al. Short report: identification of
Sci 2011; 52:2109–17. a specific pattern of vascular endothelial growth factor mRNA expression
45. Lin M, Chen Y, Jin J, et al. Ischaemia-induced retinal neovascularisation and in human placenta and cultured placental fibroblasts. Placenta 1994;15:
diabetic retinopathy in mice with conditional knockout of hypoxia-inducible 557–61.
factor-1 in retinal Muller cells. Diabetologia 2011;54:1554–66. 76. Woolard J, Bevan HS, Harper SJ, et al. Molecular diversity of VEGF-A as a
46. Cramer T, Yamanishi Y, Clausen BE, et al. HIF-1alpha is essential for myeloid regulator of its biological activity. Microcirculation 2009;16:572–92.
cell-mediated inflammation. Cell 2003;112:645–57. 77. de Vries C, Escobedo JA, Ueno H, et al. The fms-like tyrosine kinase, a receptor
47. Kojima H, Gu H, Nomura S, et al. Abnormal B lymphocyte development and for vascular endothelial growth factor. Science 1992;255:989–91.
autoimmunity in hypoxia-inducible factor 1alpha -deficient chimeric mice. 78. Quinn TP, Peters KG, De Vries C, et al. Fetal liver kinase 1 is a receptor for
Proc Natl Acad Sci U S A 2002;99:2170–4. vascular endothelial growth factor and is selectively expressed in vascular
48. Makino Y, Nakamura H, Ikeda E, et al. Hypoxia-inducible factor regulates endothelium. Proc Natl Acad Sci U S A 1993;90:7533–7.
survival of antigen receptor-driven T cells. J Immunol 2003;171:6534–40. 79. Soker S, Takashima S, Miao HQ, et al. Neuropilin-1 is expressed by endothe-
49. Giaccia A, Siim BG, Johnson RS. HIF-1 as a target for drug development. lial and tumor cells as an isoform-specific receptor for vascular endothelial
Nat Rev Drug Discov 2003;2:803–11. growth factor. Cell 1998;92:735–45.
50. Kelly BD, Hackett SF, Hirota K, et al. Cell type-specific regulation of angio- 80. Schlessinger J. New roles for Src kinases in control of cell survival and angio-
genic growth factor gene expression and induction of angiogenesis in non­ genesis. Cell 2000;100:293–6.
ischemic tissue by a constitutively active form of hypoxia-inducible factor 1. 81. Hubbard SR. Structural analysis of receptor tyrosine kinases. Prog Biophys
Circ Res 2003;93:1074–81. Mol Biol 1999;71:343–58.
51. Hirsila M, Koivunen P, Gunzler V, et al. Characterization of the human prolyl 82. Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-island formation
4-hydroxylases that modify the hypoxia-inducible factor. J Biol Chem 2003; and vasculogenesis in Flk-1-deficient mice. Nature 1995;376:62–6.
278:30772–80. 83. Scott A, Mellor H. VEGF receptor trafficking in angiogenesis. Biochem Soc
52. Willam C, Masson N, Tian YM, et al. Peptide blockade of HIFalpha degrada- Trans 2009;37:1184–8.
tion modulates cellular metabolism and angiogenesis. Proc Natl Acad Sci 84. Jopling HM, Odell AF, Hooper NM, et al. Rab GTPase regulation of VEGFR2
U S A 2002;99:10423–8. trafficking and signaling in endothelial cells. Arterioscler Thromb Vasc Biol
53. Chen J, Smith LE. A double-edged sword: erythropoietin eyed in retinopathy 2009;29:1119–24.
of prematurity. J AAPOS 2008;12:221–2. 85. Salikhova A, Wang L, Lanahan AA, et al. Vascular endothelial growth factor
54. Ferrara N. Vascular endothelial growth factor as a target for anticancer and semaphorin induce neuropilin-1 endocytosis via separate pathways.
therapy. Oncologist 2004;9(Suppl 1):2–10. Circ Res 2008;103:e71–79.
55. Carmeliet P, Ferreira V, Breier G, et al. Abnormal blood vessel development 86. Ewan LC, Jopling HM, Jia H, et al. Intrinsic tyrosine kinase activity is required
and lethality in embryos lacking a single VEGF allele. Nature 1996;380: for vascular endothelial growth factor receptor 2 ubiquitination, sorting and
435–9. degradation in endothelial cells. Traffic 2006;7:1270–82.
56. Alon T, Hemo I, Itin A, Pe’er J, et al. Vascular endothelial growth factor acts 87. Lanahan AA, Hermans K, Claes F, et al. VEGF receptor 2 endocytic trafficking
as a survival factor for newly formed retinal vessels and has implications for regulates arterial morphogenesis. Dev Cell 2010;18:713–24.
retinopathy of prematurity. Nat Med 1995;1:1024–8. 88. Cai J, Jiang WG, Ahmed A, et al. Vascular endothelial growth factor-induced
57. Baffert F, Le T, Thurston G, et al. Angiopoietin-1 decreases plasma leakage by endothelial cell proliferation is regulated by interaction between VEGFR-2,
reducing number and size of endothelial gaps in venules. Am J Physiol Heart SH-PTP1 and eNOS. Microvasc Res 2006;71:20–31.
Circ Physiol 2006;290:H107–118. 89. Chen J, Braet F, Brodsky S, et al. VEGF-induced mobilization of caveolae and
58. Saint-Geniez M, Maharaj AS, Walshe TE, et al. Endogenous VEGF is required increase in permeability of endothelial cells. Am J Physiol Cell Physiol
for visual function: evidence for a survival role on muller cells and photore- 2002;282:C1053–1063.
ceptors. PLoS ONE 2008;3:e3554. 90. Grant MB, Boulton ME, Ljubimov AV. Erythropoietin: when liability becomes
59. Adamis AP, Shima DT, Yeo KT, et al. Synthesis and secretion of vascular asset in neurovascular repair. J Clin Invest 2008;118:467–70.
permeability factor/vascular endothelial growth factor by human retinal 91. Takahashi T, Ueno H, Shibuya M. VEGF activates protein kinase C-dependent,
pigment epithelial cells. Biochem Biophys Res Commun 1993;193:631–8. but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in
60. Aiello LP, Northrup JM, Keyt BA, et al. Hypoxic regulation of vascular endo- primary endothelial cells. Oncogene 1999;18:2221–30.
thelial growth factor in retinal cells. Arch Ophthalmol 1995;113:1538–44. 92. Fadini GP, Miorin M, Facco M, et al. Circulating endothelial progenitor cells
61. Famiglietti EV, Stopa EG, McGookin ED, et al. Immunocytochemical localiza- are reduced in peripheral vascular complications of type 2 diabetes mellitus.
tion of vascular endothelial growth factor in neurons and glial cells of human J Am Coll Cardiol 2005;45:1449–57.
retina. Brain Res 2003;969:195–204. 93. Tepper OM, Galiano RD, Capla JM, et al. Human endothelial progenitor cells
62. Sandercoe TM, Geller SF, Hendrickson AE, et al. VEGF expression by gan- from type II diabetics exhibit impaired proliferation, adhesion, and incorpora-
glion cells in central retina before formation of the foveal depression in tion into vascular structures. Circulation 2002;106:2781–6.
monkey retina: evidence of developmental hypoxia. J Comp Neurol 2003;462: 94. Loomans CJ, de Koning EJ, Staal FJ, et al. Endothelial progenitor cell dysfunc-
42–54. tion: a novel concept in the pathogenesis of vascular complications of type 1
63. Marneros AG, Fan J, Yokoyama Y, et al. Vascular endothelial growth factor diabetes. Diabetes 2004;53:195–9.
expression in the retinal pigment epithelium is essential for choriocapillaris 95. Schatteman GC, Hanlon HD, Jiao C, et al. Blood-derived angioblasts acceler-
development and visual function. Am J Pathol 2005;167:1451–9. ate blood-flow restoration in diabetic mice. J Clin Invest 2000;106:571–8.
96. Caballero S, Sengupta N, Afzal A, et al. Ischemic vascular damage can be 125. Tang J, Kern TS. Inflammation in diabetic retinopathy. Prog Retin Eye Res
repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes 2011; 30:343–58.
2007;56:960–7. 126. Kern TS. Contributions of inflammatory processes to the development of the
97. Jarajapu YP, Caballero S, Verma A, et al. Blockade of NADPH oxidase restores early stages of diabetic retinopathy. Exp Diabetes Res 2007;2007:95103. 445
vasoreparative function in diabetic CD34+ cells. Invest Ophthalmol Vis Sci 127. Schmidt MI, Duncan BB, Sharrett AR, et al. Markers of inflammation and
2011;52:5093–104. prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities
98. Case J, Ingram DA, Haneline LS. Oxidative stress impairs endothelial progeni- study): a cohort study. Lancet 1999;353:1649–52.

Chapter 18
tor cell function. Antioxid Redox Signal 2008;10:1895–907. 128. Festa A, D’Agostino Jr R, Tracy RP, et al. Elevated levels of acute-phase pro-
99. Togliatto G, Trombetta A, Dentelli P, et al. Unacylated ghrelin rescues endo- teins and plasminogen activator inhibitor-1 predict the development of type
thelial progenitor cell function in individuals with type 2 diabetes. Diabetes 2 diabetes: the insulin resistance atherosclerosis study. Diabetes 2002;51:
2010;59:1016–25. 1131–7.
100. Ingram DA, Lien IZ, Mead LE, et al. In vitro hyperglycemia or a diabetic 129. Eliasson MC, Jansson JH, Lindahl B, et al. High levels of tissue plasminogen
intrauterine environment reduces neonatal endothelial colony-forming cell activator (tPA) antigen precede the development of type 2 diabetes in a
numbers and function. Diabetes 2008;57:724–31. longitudinal population study. The Northern Sweden MONICA study.
101. Urbich C, Dernbach E, Rossig L, et al. High glucose reduces cathepsin L activ- Cardiovasc Diabetol 2003;2:19.
ity and impairs invasion of circulating progenitor cells. J Mol Cell Cardiol 130. Meigs JB, Dupuis J, Liu C, et al. PAI-1 gene 4G/5G polymorphism and risk
2008;45:429–36. of type 2 diabetes in a population-based sample. Obesity (Silver Spring)

Retinal and Choroidal Vasculature:


102. Li SS, Liu Z, Uzunel M, et al. Endogenous thrombospondin-1 is a cell-surface 2006;14:753–8.
ligand for regulation of integrin-dependent T-lymphocyte adhesion. Blood 131. Orasanu G, Plutzky J. The pathologic continuum of diabetic vascular disease.
2006;108:3112–20. J Am Coll Cardiol 2009;53:S35–42.
103. Butler JM, Guthrie SM, Koc M, et al. SDF-1 is both necessary and sufficient to 132. van Hecke MV, Dekker JM, Nijpels G, et al. Inflammation and endothelial
promote proliferative retinopathy. J Clin Invest 2005;115:86–93. dysfunction are associated with retinopathy: the Hoorn study. Diabetologia
104. Segal MS, Shah R, Afzal A, et al. Nitric oxide cytoskeletal-induced alterations 2005;48:1300–6.
reverse the endothelial progenitor cell migratory defect associated with 133. Spijkerman AM, Gall MA, Tarnow L, et al. Endothelial dysfunction and low-
diabetes. Diabetes 2006;55:102–9. grade inflammation and the progression of retinopathy in type 2 diabetes.
105. Bhatwadekar AD, Glenn JV, Li G, et al. Advanced glycation of fibronectin Diabet Med 2007;24:969–76.
impairs vascular repair by endothelial progenitor cells: implications for vaso- 134. Hadjadj S, Aubert R, Fumeron F, et al. Increased plasma adiponectin concen-
degeneration in diabetic retinopathy. Invest Ophthalmol Vis Sci 2008;49: trations are associated with microangiopathy in type 1 diabetic subjects.
1232–41. Diabetologia 2005;48:1088–92.
106. Bhatwadekar AD, Guerin EP, Jarajapu YP, et al. Transient inhibition of trans- 135. McLeod DS, Lefer DJ, Merges C, et al. Enhanced expression of intracellular
forming growth factor-beta1 in human diabetic CD34+ cells enhances vascular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid.
reparative functions. Diabetes 2010; 59:2010–9. Am J Pathol 1995;147:642–53.
107. Sorrentino SA, Bahlmann FH, Besler C, et al. Oxidant stress impairs in vivo 136. Joussen AM, Murata T, Tsujikawa A, et al. Leukocyte-mediated endothelial
reendothelialization capacity of endothelial progenitor cells from patients cell injury and death in the diabetic retina. Am J Pathol 2001;158:147–52.
with type 2 diabetes mellitus: restoration by the peroxisome proliferator- 137. Joussen AM, Poulaki V, Qin W, et al. Retinal vascular endothelial growth
activated receptor-gamma agonist rosiglitazone. Circulation 2007;116: factor induces intracellular adhesion molecule-1 and endothelial nitric oxude
163–73. synthase expression and initiates early diabetic retinal leukocyte adhesion in
108. Mohler 3rd ER, Shi Y, Moore J, et al. Diabetes reduces bone marrow and vivo. Am J Pathol 2002;160:501–9.
circulating porcine endothelial progenitor cells, an effect ameliorated by ator- 138. Joussen AM, Poulaki V, Mitsiades N, et al. Nonsteroidal anti-inflammatory
vastatin and independent of cholesterol. Cytometry A 2009;75:75–82. drugs prevent early diabetic retinopathy via TNF-α suppression. FASEB J
109. Anghelina M, Krishnan P, Moldovan L, et al. Monocytes/macrophages coop- 2002;16:438–40.
erate with progenitor cells during neovascularization and tissue repair: con- 139. Adamis AP. Is diabetic retinopathy an inflammatory disease? Br J Ophthalmol
version of cell columns into fibrovascular bundles. Am J Pathol 2006;168: 2002;86:363–5.
529–41. 140. Joussen AM, Poulaki V, Le ML, et al. A central role for inflammation in the
110. Chang KH, Chan-Ling T, McFarland EL, et al. IGF binding protein-3 regulates pathogenesis of diabetic retinopathy. Faseb J 2004;18:1450–2.
hematopoietic stem cell and endothelial precursor cell function during 141. Powell ED, Field RA. Diabetic retinopathy and rheumatoid arthritis. Lancet
vascular development. Proc Natl Acad Sci U S A 2007;104:10595–600. 1964;2:17–8.
111. Granata R, Trovato L, Garbarino G, et al. Dual effects of IGFBP-3 on endothe- 142. Brucklacher RM, Patel KM, VanGuilder HD, et al. Whole genome assessment
lial cell apoptosis and survival: involvement of the sphingolipid signaling of the retinal response to diabetes reveals a progressive neurovascular inflam-
pathways. FASEB J 2004;18:1456–8. matory response. BMC Med Genomics 2008;1:26.
112. Franklin SL, Ferry Jr RJ, Cohen P. Rapid insulin-like growth factor (IGF)- 143. Kern TS, Miller CM, Du Y, et al. Topical administration of nepafenac inhibits
independent effects of IGF binding protein-3 on endothelial cell survival. diabetes-induced retinal microvascular disease and underlying abnormalities
J Clin Endocrinol Metab 2003;88:900–7. of retinal metabolism and physiology. Diabetes 2007;56:373–9.
113. Liu LQ, Sposato M, Liu HY, et al. Functional cloning of IGFBP-3 from human 144. Gubitosi-Klug RA, Talahalli R, Du Y, et al. 5-Lipoxygenase, but not 12/15-
microvascular endothelial cells reveals its novel role in promoting prolifera- lipoxygenase, contributes to degeneration of retinal capillaries in a mouse
tion of primitive CD34+CD38– hematopoietic cells in vitro. Oncol Res model of diabetic retinopathy. Diabetes 2008;57:1387–93.
2003;13:359–71. 145. Vincent JA, Mohr S. Inhibition of caspase-1/interleukin-1beta signaling
114. Vasylyeva TL, Chen X, Ferry Jr RJ. Insulin-like growth factor binding protein-3 prevents degeneration of retinal capillaries in diabetes and galactosemia.
mediates cytokine-induced mesangial cell apoptosis. Growth Horm IGF Res Diabetes 2007;56:224–30.
2005;15:207–14. 146. Behl Y, Krothapalli P, Desta T, et al. Diabetes-enhanced tumor necrosis factor-
115. Lofqvist C, Chen J, Connor KM, et al. IGFBP3 suppresses retinopathy through alpha production promotes apoptosis and the loss of retinal microvascular
suppression of oxygen-induced vessel loss and promotion of vascular cells in type 1 and type 2 models of diabetic retinopathy. Am J Pathol
regrowth. Proc Natl Acad Sci U S A 2007;104:10589–94. 2008;172:1411–8.
116. Giannini S, Cresci B, Pala L, et al. IGFBPs modulate IGF-I- and high glucose- 147. Lutty GA, McLeod DS, Merges C, et al. Localization of VEGF in human retina
controlled growth of human retinal endothelial cells. J Endocrinol 2001;171: and choroid. Arch Ophthalmol 1996;114:971–7.
273–84. 148. Rogers SL, McIntosh RL, Lim L, et al. Natural history of branch retinal vein
117. Kielczewski JL, Hu P, Shaw LC, et al. Novel protective properties of IGFBP-3 occlusion: an evidence-based systematic review. Ophthalmology 2010;
result in enhanced pericyte ensheathment, reduced microglial activation, 117:1094–101 e1095.
increased microglial apoptosis, and neuronal protection after ischemic retinal 149. Laouri M, Chen E, Looman M, et al. The burden of disease of retinal vein
injury. Am J Pathol 2011; 178:1517–28. occlusion: review of the literature. Eye (Lond) 2011; 25(8):981–8.
118. Chan-Ling TL, Halasz P, Stone J. Development of retinal vasculature in the 150. Hardarson HS, Stefansson E. Oxygen saturation in branch vein occlusion.
cat: processes and mechanisms. Curr Eye Res 1990;9:459–78. Acta Ophthalmol 2011;Epub ahead of printing.
119. Hellstrom A, Engstrom E, Hard AL, et al. Postnatal serum insulin-like growth 151. Hayreh SS, Weingeist TA. Experimental occlusion of the central artery of the
factor I deficiency is associated with retinopathy of prematurity and other retina. IV: Retinal tolerance time to acute ischaemia. Br J Ophthalmol
complications of premature birth. Pediatrics 2003;112:1016–20. 1980;64:818–25.
120. Giudice LC, de Zegher F, Gargosky SE, et al. Insulin-like growth factors and 152. Hayreh SS, Podhajsky P. Ocular neovascularization with retinal vascular
their binding proteins in the term and preterm human fetus and neonate with occlusion. II. Occurrence in central and branch retinal artery occlusion.
normal and extremes of intrauterine growth. J Clin Endocrinol Metab Arch Ophthalmol 1982;100:1585–96.
1995;80:1548–55. 153. Coats G. Obstruction of the central artery of the retina. R. Lond Ophthalmic
121. Smith WJ, Underwood LE, Keyes L, et al. Use of insulin-like growth factor I Hosp Rep 1905;16:262–306.
(IGF-I) and IGF-binding protein measurements to monitor feeding of prema- 154. Coats G. Pathology of obstruction of the centeral artery of the retina. R. Lond
ture infants. J Clin Endocrinol Metab 1997;82:3982–8. Ophthalmic Hosp Rep 1913;19:45–70.
122. Smith LE. IGF-1 and retinopathy of prematurity in the preterm infant. 155. Fong AC, Schatz H, McDonald HR, et al. Central retinal vein occlusion in
Biol Neonate 2005;88:237–44. young adults (papillophlebitis). Retina 1992;12:3–11.
123. Kohner EM, Henkind P. Correlation of fluorescein angiogram and retinal 156. Sofi F, Marcucci R, Fedi S, et al. High lipoprotein (a) levels are associated with
digest in diabetic retinopathy. Am J Ophthalmol 1970;69:403–14. an increased risk of retinal vein occlusion. Atherosclerosis 2010; 210:278–81.
124. Linsenmeier RA, Braun RD, McRipley MA, et al. Retinal hypoxia in long term 157. Arruga J, Sanders MD. Ophthalmologic findings in 70 patients with evidence
diabetic cats. Invest Ophthalmol Vis Sci 1998;39:1647–57. of retinal embolism. Ophthalmology 1982;89:1336–47.
158. Ageno W, Cattaneo R, Manfredi E, et al. Parnaparin versus aspirin in the 177. Barber AJ, Antonetti DA, Gardner TW. Altered expression of retinal
treatment of retinal vein occlusion. A randomized, double blind, controlled occludin and glial fibrillary acidic protein in experimental diabetes. The
study. Thromb Res 2010; 125:137–41. Penn State Retina Research Group. Invest Ophthalmol Vis Sci 2000;41:
446 159. Marcucci R, Sofi F, Grifoni E, et al. Retinal vein occlusions: a review for the 3561–8.
internist. Intern Emerg Med 2011; 6:307–14. 178. Harhaj NS, Felinski EA, Wolpert EB, et al. VEGF activation of protein kinase
160. Nagpal KC, Goldberg MF, Rabb MF. Ocular manifestations of sickle hemo- C stimulates occludin phosphorylation and contributes to endothelial perme-
globinopathies. Surv Ophthalmol 1977;21:391–411. ability. Invest Ophthalmol Vis Sci 2006;47:5106–15.
161. Cao J, Kunz Mathews M, McLeod DS, et al. Angiogenic factors in human 179. Sundstrom JM, Sundstrom CJ, Sundstrom SA, et al. Phosphorylation site
Section 1

proliferative sickle cell retinopathy. Br J Ophthalmol 1999;83:838–46. mapping of endogenous proteins: a combined MS and bioinformatics
162. Brown GC, Magargal LE. The ocular ischemic syndrome. Clinical, fluorescein approach. J Proteome Res 2009;8:798–807.
angiographic and carotid angiographic features. Int Ophthalmol 1988;11: 180. Murakami T, Felinski EA, Antonetti DA. Occludin phosphorylation
239–51. and ubiquitination regulate tight junction trafficking and vascular
163. Boto de los Bueis A, Fernandez-Prieto A, Ruiz-Martin MM, et al. [Bilateral endothelial growth factor-induced permeability. J Biol Chem 2009;284:
carotid occlusion in young woman. Clinical and hemodynamic ocular results.] 21036–46.
Arch Soc Esp Oftalmol 2003;78:227–30. 181. Aveleira CA, Lin CM, Abcouwer SF, et al. TNF-alpha signals through
164. Baatz H, Lange S, Buchner H, et al. [Pseudoangiitis in bilateral ocular isch- PKCzeta/NF-kappaB to alter the tight junction complex and increase retinal
Anatomy and Physiology

emia.] Ophthalmologe 2007;104: 243–245. endothelial cell permeability. Diabetes 2010; 59:2872–82.
Basic Science and Translation to Therapy

165. Sturrock GD, Mueller HR. Chronic ocular ischaemia. Br J Ophthalmol 182. Hidayat A, Fine B. Diabetic choroidopathy: light and electron microscopic
1984;68:716–23. observations of seven cases. Ophthalmology 1985;67:512–22.
166. Kearns TP. Differential diagnosis of central retinal vein obstruction. 183. Cao J, McLeod S, Merges CA, et al. Choriocapillaris degeneration and related
Ophthalmology 1983;90:475–80. pathologic changes in human diabetic eyes. Arch Ophthalmol 1998;116:
167. Klijn CJ, Kappelle LJ, van Schooneveld MJ, et al. Venous stasis retinopathy in 589–97.
symptomatic carotid artery occlusion: prevalence, cause, and outcome. Stroke 184. Lutty GA, Cao J, McLeod DS. Relationship of polymorphonuclear leukocytes
2002;33:695–701. (PMNs) to capillary dropout in the human diabetic choroid. Am J Pathol
168. Costa VP, Kuzniec S, Molnar LJ, et al. Clinical findings and hemo­ 1997;151:707–14.
dynamic changes associated with severe occlusive carotid artery disease. 185. Lovasik J, Kergoat H. Electroretinographic results and ocular vascular perfu-
Ophthalmology 1997;104:1994–2002. sion in type I diabetes. Invest Ophthalmol Vis Sci 1993;34:1731–43.
169. Linsenmeier RA, Padnick-Silver L. Mtabolic dependence of photoreceptors on 186. Holopigian K, Greenstein VC, Seiple W, et al. Evidence for photoreceptor
the choroid in the normal and detached retina. Invest Ophthalmol Vis Sci changes in patients with diabetic retinopathy. Invest Ophthalmol Vis Sci
2000;41:3117–23. 1997;38:2355–65.
170. Wang S, Linsenmeier RA. Hyperoxia improves oxygen consumption in the 187. Grunwald J, Hariprasad S, DuPont J, et al. Foveolar choroidal blood flow in
detached feline retina. Invest Ophthalmol Vis Sci 2007;48:1335–41. age-related macular degeneration. Invest Ophthalmol Vis Sci 1998;39:
171. Lewis G, Mervin K, Valter K, et al. Limiting the proliferation and reactivity 385–90.
of retinal Muller cells during experimental retinal detachment: the value of 188. Grunwald JE, Metelitsina TI, Dupont JC, et al. Reduced foveolar choroidal
oxygen supplementation. Am J Ophthalmol 1999;128:165–72. blood flow in eyes with increasing AMD severity. Invest Ophthalmol Vis Sci
172. Folkman, J. Tumor angiogenesis: therapeutic implications. N. Engl J Med 285: 2005;46:1033–8.
1182–6, 1971. 189. Metelitsina TI, Grunwald JE, DuPont JC, et al. Foveolar choroidal circulation
173. Penn JS, Madan A, Caldwell RB, et al. Vascular endothelial growth factor in and choroidal neovascularization in age-related macular degeneration. Invest
eye disease. Prog Retin Eye Res 2008;27:331–71. Ophthalmol Vis Sci 2008;49:358–63.
174. Salam A, Mathew R, Sivaprasad S. Treatment of proliferative diabetic reti- 190. Pournaras CJ, Rungger-Brandle E, Riva CE, et al. Regulation of retinal blood
nopathy with anti-VEGF agents. Acta Ophthalmol 2011; 89:405–11. flow in health and disease. Prog Retin Eye Res 2008;27:284–330.
175. Antonetti DA, Barber AJ, Khin S, et al. Vascular permeability in experimental 191. McLeod DS, Taomoto M, Otsuji T, et al. Quantifying changes in RPE
diabetes is associated with reduced endothelial occludin content: vascular and choriocapillaris in eyes with age-related macular degeneration. Invest
endothelial growth factor decreases occludin in retinal endothelial cells. Penn Ophthalmol Vis Sci 2002;43:1986–93.
State Retina Research Group. Diabetes 1998;47:1953–9. 192. McLeod DS, Grebe R, Bhutto I, et al. Relationship between RPE and chorio-
176. Aiello L, Bursell S, Clermont A, et al. Vascular endothelial growth factor- capillaris in age-related macular degeneration. Invest Ophthalmol Vis Sci
induced retinal permeability is mediated by protein kinase C in vivo and 2009;50:4982–91.
suppressed by an orally effective beta-isoform-selective inhibitor. Diabetes 193. Feigl B. Age-related maculopathy-linking aetiology and pathophysiological
1997;46:1473–80. changes to the ischaemia hypothesis. Prog Retin Eye Res 2009;28:63–86.
Anatomy and Physiology Section 1

Mechanisms of Normal Retinal Adhesion Chapter

Michael F. Marmor
19 
In some cases of retinal detachment, the reasons for retinal pressure that is required to expand the detachment can be con-
separation are obvious, such as penetrating trauma that snags verted mathematically, by Laplace’s law, into a value for adhe-
the retina or gross vitreous traction bands that pull the retina sive force at the margin of the detachment (where the separation
from the retinal pigment epithelium (RPE). Any physiologic is taking place). This technique allows the evaluation in living
mechanism of adhesion would be overwhelmed by such forces. animals of the effects of drugs and other agents that modify
However, detachment is also caused by less dramatic forces retinal adhesion.9,10
and under conditions in which stronger adhesion might prevent,
or at least delay, the separation and its spread. The physiol- ADHESIVE FORCE AND
ogy of attachment is especially important with respect to ENVIRONMENTAL FACTORS
nonrhegmatogenous detachments that cannot develop unless
A substantial force maintains attachment of the sensory retina
local mechanisms for keeping the subretinal space dry are
to the underlying RPE. This force can be altered by environmen-
overwhelmed.
tal factors.
Independent of its clinical relevance, the study of retinal adhe-
sion is also of considerable physiologic interest. No anatomic Magnitude of adhesive force
junctions bridge the mammalian subretinal space,1 yet this pri- In vitro measurements, 5–20 minutes after enucleation, have
mordial dural cavity remains collapsed and, indeed, tightly shown that about 25 mg of force is required to peel a 5-mm strip
closed throughout a lifetime of ocular movement and tugging of rabbit retina from the RPE.3,11,12 Pressure measurements in
from the vitreous. Our current knowledge of this system is still living eyes have shown an adhesive force in the rabbit of
incomplete, but evidence suggests that adhesion depends on a 100–180 dyn/cm.8,9 Adhesion is stronger in cats and monkeys,
mixture of anatomic, physical, and metabolic factors.2 This which show mean values for adhesive force that are 180% (for
chapter reviews these factors and considers clinical and thera- cats) and 140% (for monkeys) of that in the rabbits.9
peutic implications.
Sensitivity to temperature and
MODELS FOR MEASURING ionic environment
RETINAL ADHESION Retinal adhesiveness drops rapidly postmortem at 37 °C, but
The strength of the adhesion between the sensory retina and the remains near control levels for hours at 4 °C7,13–15 (Fig. 19.1A).
RPE can be measured with in vitro and in vivo methods. These effects of temperature are reversible. Figure 19.1B shows
that changing the temperature from 37 °C to 4 °C or vice versa
In vitro methods causes retinal adherence in the rabbit16 to rise or fall, respec-
Adhesive force falls rapidly after enucleation or death,3,4 as is tively, and repeatedly. This reversibility has also been docu-
discussed later, and this puts a constraint of time on techniques mented in primate and human tissue.14,15,17 The mechanism of
for measuring adhesiveness in vitro.5,6 One approach is to peel these reversible temperature effects is still obscure. Temperature
the retina within a fluid bath while recording the required force could act directly on physicochemical components of adhesion
with a transducer.3 A faster method is to measure the amount of or modulate metabolic systems. Some of the adhesion-enhancing
RPE pigment that remains attached to the retina after separation effect of cold temperature may result from sodium pump inhibi-
as the index of adhesiveness.7 An eye can be enucleated and tion and secondary tissue swelling, which would make the inter-
small strips of the eyecup prepared within 30 seconds, after digitated outer segments and RPE microvilli harder to separate.3,13
which the retina is gently peeled by hand from the RPE. The Because cellular swelling is a pathologic event, the actions of
stronger the adhesive force, the more pigment adheres to the cold temperature may not be entirely relevant to normal adhe-
peeled retina. sive processes.
The ionic environment appears to be critical to adhesive
In vivo methods strength. In in vitro experiments using tissue from rabbits,
Kita et al.8 developed the most direct technique for quantifying humans, and other primates, lowering the pH from 7.4 to 5.5, or
adhesiveness within the living eye. A small detachment is made removing calcium and magnesium ions from the bathing solu-
in the eye by injecting fluid into the subretinal space through a tion, weakened adhesive force and accelerated the rate at which
micropipette, and a second micropipette is inserted simultane- adhesion falls postmortem11,13,15,17 (Fig. 19.2A). These changes can
ously into the detachment cavity to measure fluid pressure. The be rapidly reversible16 (Fig. 19.2B), although cold temperature
100
448 Pigment adherence to retina (%) 30
4°C Control

Peeling force (mg)


80
0
Section 1

60
30
25°C ∅pH
40
0
Anatomy and Physiology
Basic Science and Translation to Therapy

20 37°C 30
∅Ca
0
0
A 0 5 10 15 20 A 0 30 60 90
Incubation time (min) Time (min)

Pigment adherence to retina (%)


100 100
Pigment adherence (%)

80 4°C 80

60 60
Hanks'
40 4°C 4°C 40

20 37°C 20
37°C pH6 Ca/Mg-free
37°C
0 0
B 2 4 6 8 10 12 14 16 0 1 2 3 4 5 6
Incubation time (min) B Incubation time (min)

Fig. 19.1 Relationship between retinal adhesiveness and temperature Fig. 19.2 Effects of pH and calcium/magnesium ion concentration on
in the rabbit. (A) Cold temperatures slow down the postmortem failure in retinal adhesiveness in the rabbit. (A) The force required to peel retina
adhesiveness to the point that firm adhesion is maintained at 4°C for from retinal pigment epithelium (RPE) drops within seconds after
many hours. (Modified with permission from Endo EG, Yao XY, Marmor lowering pH (by injecting 1 mL of 1 mol/L HCl near the tissue) or
MF. Pigment adherence as a measure of retinal adhesion: dependence lowering external Ca/Mg-free solution. The tracings represent a
on temperature. Invest Ophthalmol Vis Sci 1988;29:1390–6.) (B) The continuous measurement of the peeling force; chemical changes
effects of temperature on retinal adhesion are reversible within minutes, were made at time 0. (Modified with permission from Marmor MF,
and adhesiveness can be repeatedly strengthened or weakened by Maack T. Local environmental factors and retinal adhesion in the
cooling or warming the same piece of tissue. (Modified with permission rabbit. Exp Eye Res 1982;34:727–33.) (B) The effects of changing pH
from Yao XY, Endo EG, Marmor MF. Reversibility of retinal adhesion in or Ca/Mg concentration are rapidly reversible. Pigment adherence was
the rabbit. Invest Ophthalmol Vis Sci 1989;30:220–4.) used as an index of adhesiveness. Dotted line, tissue maintained in
Hanks balanced salt solution. Dashed line, pH changed from 7.4 to 6.0
and back again. Solid line, Hanks solution changed to Ca/Mg-free
will block or mask the effects of pH and calcium ions. In vivo, solution and back again. (Modified with permission from Yao XY, Endo
EG, Marmor MF. Reversibility of retinal adhesion in the rabbit. Invest
the removal of calcium ions from the subretinal space weakens Ophthalmol Vis Sci 1989;30:220–4.)
retinal adhesion in rabbits to about 30% of normal.18 In other
words, calcium appears to be a necessary element for the main-
tenance of normal adhesiveness in the living eye. the eye through anterior drainage channels.19,21 The posterior
route is limited because the retina and RPE22,23 provide a sub-
Mechanical forces outside the stantial resistance to water movement, and little fluid can cross
subretinal space these layers under the available heads of pressure. A side-effect
A number of forces play upon the retina from outside the sub- of this retinal flow resistance is that the outward movement of
retinal space, strengthening or weakening retinal adhesion. Most fluid acts to push the retina against the RPE (Fig. 19.3). This is
important are fluid and vitreous pressure, not only because they one mechanism contributing to normal retinal attachment. The
contribute to adhesion, but also because they may cause detach- actual pressure difference across retina or RPE is probably small
ment when altered by pathologic conditions. because intraocular pressure is ultimately contained by the
sclera, and tissue pressures will therefore equalize to a large
Fluid pressure: hydrostatic and osmotic degree between the vitreous and choroid. However, Fatt and
Fluid is driven passively from vitreous to choroid by both intra- Shantinath22 have calculated that even a very small pressure dif-
ocular pressure and the osmotic pressure of the extracellular ference (only 0.52 × 10−3 mmHg) across the retina would gener-
fluid in the choroid (estimated to be about 12 mmHg in the ate a force sufficient to keep the retina firmly fixed against the
rabbit,19 but possibly lower in humans20). However, under wall of the eye. This low value may explain the fact that no
normal conditions there appears to be relatively little posterior one14,19 has been able to measure a pressure difference between
flow, and most of the fluid that enters the eye as aqueous leaves the vitreous cavity and subretinal space.
Fig. 19.3 Intraocular pressure and retinal flow
resistance as a factor in retinal adhesion.
(A) Intraocular pressure is evenly distributed
within the vitreous cavity so that fluid is 449
continually being pushed through the coats
of the eye. (B) Magnified view of the tissue
layers in the posterior of the eye, including

Chapter 19
cortical vitreous. The flow resistance of each
layer is indicated on the right; note that the
retinal pigment epithelium (RPE) and choroid
can also modify fluid movement by active
transport and osmotic pressure, respectively.
Intraocular
pressure

Mechanisms of Normal Retinal Adhesion


A

Intraocular pressure
Flow resistance
Cortical
vitreous Uncertain
Neural
retina High
RPE High (active transport)
Choroid Low (high osmotic
pressure)

Sclera High

These physical forces can also work in the other direction and transport at a maximal rate. Measurements in humans for the
cause retinal detachment. For example, if hyperosmotic fluid is resorption of detachments showed a rate of 0.11 µL/h/mm2 of
introduced into the vitreous cavity, fluid moving from the RPE, and the rate may well be higher in eyes without pathol-
choroid into the vitreous will elevate the retina24,25 (Fig. 19.4). ogy.31 This translates into roughly 3.5 mL of fluid per day, which
This is an important concern in the evaluation or use of intra­ explains why a rhegmatogenous detachment can settle within 24
vitreal drugs that may damage the retina osmotically indepen- hours, and emphasizes the power of RPE fluid transport.
dently of pharmacologic effects. The clinical importance of fluid pressure as a factor in prevent-
It may seem puzzling that, although most intraocular fluid ing retinal detachment is uncertain. It undoubtedly helps to keep
leaves the eye anteriorly rather than posteriorly, ample animal the retina in place when the retina is intact, but raising intraocu-
data26–30 indicated that the RPE can pump fluid from the subreti- lar pressure in rabbits to 38 mmHg or lowering it to 0 mmHg
nal space to choroid at a very high rate (about 0.3 µL/h/mm2 of had only a modest effect on the rate of subretinal fluid absorp-
RPE) comparable with that of aqueous secretion. How can the tion.32 Furthermore, clinical retinal (as opposed to choroidal)
RPE have such enormous power for fluid removal, yet very little detachment is uncommon in hypotonus eyes, and even a small
fluid leaves the normal eye by a posterior route? The answer hole in the retina would theoretically allow fluid to reach the
probably lies in the flow resistance of the retina. Under normal subretinal space and circumvent fluid pressure mechanisms of
conditions, given the flow resistance of the tissue and the tiny adhesion. Retinal holes are, in fact, found in about 10% of
pressure differential across it,22,23 water will cross the retina only autopsy eyes without detachment,33 but many of these holes may
at a very slow rate. In other words, the tissue resistance is rate- not be functionally open because of surrounding pigmentation
limiting, and most aqueous fluid must leave by the anterior or because of “tamponade” by the cortical vitreous gel.34
route. Under pathologic conditions of retinal detachment Once the retina has detached and adhesion mechanisms that
(whether rhegmatogenous or nonrhegmatogenous), fluid is depend on retina–RPE contact can no longer work, fluid dynam-
present within the subretinal space and is thus available for ics become much more important, even in the presence of a hole.
Degree of retinal detachment
Large *
450
Moderate
Section 1

Low **

Incipient *

None

0 1000 2000 3000


Anatomy and Physiology
Basic Science and Translation to Therapy

Osmolarity of midvitreous injection (mOsm)

NaCl
Na aspartate
Penicillin G
Penicillin G with systemic probenecid (Benemid)
Mannitol
Sucrose
Mixture of penicillin, NaCl and sucrose
A B * Ethylenediaminetetraacetic acid (EDTA)

Fig. 19.4 Effects of injecting hyperosmolar solution into the vitreous. (A) Monkey eye enucleated after a mid-vitreal hyperosmolar injection.
Bullous detachment was present in the posterior pole and peripapillary region. (B) Degree of serous retinal detachment in rabbit eyes 15 minutes
after injecting 0.05 mL of different solutions and osmolarities into the vitreous. Incipient detachment, the vitreoretinal interface glistened but did
not visibly separate; low detachment, separation occurred without bullous elevation; moderate detachment, the bullae were confined to the
posterior pole; large detachment, there was extension near or beyond the equator. (Reproduced with permission from Marmor MF. Retinal
detachment from hyperosmotic intravitreal injection. Invest Ophthalmol Vis Sci 1979;18:1237–44.)

with blood osmolality. Mannitol is known to enhance the absorp-


200 tion of fluid out of the subretinal space.38 Some of the adhesive
Mannitol
Monkey
(2 g/kg) effect may come from fluid absorption “pulling” the retina
against the RPE; however, most of the effect probably comes
from dehydration of the subretinal space (which enhances the
Retinal adhesive force (%)

150
binding properties of the interphotoreceptor matrix (IPM)),
because the mannitol effect is still evident in excised tissue
where there is no flow toward the choriocapillaris.
100
Vitreous support and other physical
aspects of adhesion
50 The role of the vitreous in creating retinal detachments through
Previous rabbit data
traction and contraction is well known. The role of the vitreous
in maintaining retinal attachment is less clear. Vitreous gel has
0 a physical structure that may help to keep the retina in place,34,39
0 1 2 3 4 5 although retinas do not just come off when vitreous detachment
Time after injection (hr) or syneresis occurs. A thin cortical layer of vitreous might remain
after vitreous detachment or syneresis and serve as a seal or
Fig. 19.5 Time course of change in retinal adhesive force after tamponade for retinal holes,34 thus aiding the action of fluid
intravenous injection of mannitol in rabbits (broken line, 2.5 g/kg) and pressure in keeping the retina apposed (Fig. 19.3).
monkeys (solid lines, 2.0 g/kg). (Reproduced with permission from
Kita M, Marmor MF. Retinal adhesive force in living rabbit, cat, and There is other, indirect, evidence for a vitreous role in adhe-
monkey eyes: normative data and enhancement by mannitol and sion. It is very difficult to produce and maintain an experimental
acetazolamide. Invest Ophthalmol Vis Sci 1992;33:1879–82.) rhegmatogenous detachment if the vitreous body remains
intact.24,40 Vitreous removal (either mechanical or with hyaluron-
Hammer35 has calculated that fluid entering a retinal hole over idase) presumably weakens the tamponade and the structural
a scleral buckle will, by its flow pattern, create a suction force qualities of the gel, but it also allows liquid vitreous to reach the
that pulls the retina backward against the buckle. subretinal space. The status of the gel may help account for the
Osmotic pressure can be manipulated more easily and may vastly different incidence of retinal detachment among young
turn out to have therapeutic applications. Both in vitro and in individuals, in whom the gel is largely intact, and among older
vivo experiments, using rabbits and primates,9,36,37 have shown individuals, in whom syneresis and vitreous detachment have
that an intravenous injection of mannitol will increase retinal occurred. A retinal hole in a young eye is blocked by gel pressure
adhesiveness by roughly 50% within 1–2 hours of injection and can seal uneventfully; a hole in an old eye is more likely to
(Fig. 19.5). The magnitude of the effect correlates rather closely allow fluid to enter the subretinal space and cause detachment.
On the other hand, the integrity of the vitreous gel seems of little develop within 3 days of reattachment, but retinal adhesiveness
consequence to the formation or persistence of experimental does not return to normal until 5–6 weeks after reapposition of
nonrhegmatogenous detachments32 or to the retinal adhesive experimentally detached retina in the rabbit45 (Fig. 19.7). 451
force measured by peeling,11 so the role of the vitreous gel in The mechanisms by which interdigitation could produce
attachment may be more one of preventing pathologic fluid adhesion are not known. During outer-segment phagocytosis,

Chapter 19
access to the subretinal space than one of providing a direct the microvilli indent the outer segments and must physically
adhesive support or force. impede attempts to withdraw them. Close ensheathment might
The weight of retina is potentially a factor in attachment and also provide a frictional resistance to withdrawal, just as a finger
detachment, as influenced by gravity and ocular movement. is hard to pull from a narrow tube. There may be electrostatic
Once a detachment has occurred, the influence of gravity is well forces that oppose separation of the membranes.46 The magni-
known, because appropriate positioning of the patient can be tude of all of these effects will also depend on other factors, such
enormously effective in inducing retina to settle. However, it is as the composition of the intervening matrix and capability of

Mechanisms of Normal Retinal Adhesion


hard to envision retinal weight as a major factor in retinal attach- the RPE microvilli for motility and remodeling.
ment under normal conditions,27,32 because our upright posture
and eye movements would dictate that benefits and adverse Interphotoreceptor matrix properties
effects be simultaneously present in different parts of the eye. The IPM is a viscous material composed largely of proteins,
glycoproteins, and proteoglycans47,48 but containing a substantial
MECHANICAL FORCES INSIDE concentration of glycosaminoglycans.49 The idea that IPM might
THE SUBRETINAL SPACE serve as a viscous glue was proposed many years ago by
Berman,50 but current evidence indicates the IPM is not just a
Anatomic bridges do not exist between retina and RPE,1 but it is
layer of glue but has component structures that play a role in the
reasonable to ask to what extent matrix material between the
adhesive process. For example, the cones are surrounded by a
layers serves as “glue” and to what extent the physical interdigi-
specialized sheath of matrix51,52 that can be recognized histo-
tation of photoreceptor outer segments and RPE microvilli
chemically by the binding of peanut agglutinin (PNA) (Fig.
serves to hold the tissues together.
19.8A). These cone matrix sheaths remain attached to both RPE
Mechanical interdigitation and photoreceptor cells when the neural retina is peeled from
the RPE.48,51,53 If fresh primate or human retina is peeled from the
RPE microvilli wrap closely around the tips of the outer seg-
RPE, the matrix material will stretch markedly before breaking
ments (Fig. 19.6), and this connection is strong enough to allow
(Fig. 19.8B), suggesting a rather strong bonding at both the
for daily phagocytosis of outer-segment fragments as the photo-
outer-segment and RPE interfaces.17,54 The evidence for a bonding
receptors renew their disc material.1,41,42 However, as a factor in
function of the IPM is strongest with respect to the cone matrix
adhesion, interdigitation seems of variable importance. The RPE
sheaths, but the structural material that surrounds the rods may
microvilli are subject to continuous cellular remodeling during
play a similar role. It can be recognized histochemically by stain-
the outer-segment renewal cycle. After surgical repair of a retinal
ing with wheatgerm agglutinin (WGA) or other lectins,55,56 and
detachment, reattachment takes place rather promptly, before
the outer segments have regenerated and microvillous connec-
tions have been re-established.43,44 Interdigitation begins to
100
Peeling force relative to normal retina (%)

80

Cone
60
Rod Rod
Rod

40

20

Microvilli 0
0 20 40 60 80 100
Days after reattachment

RPE
cytoplasm Fig. 19.7 Time course of the recovery of adhesive strength after
separated retina becomes reattached. Experimental detachments were
made in rabbit eyes by injecting a balanced salt solution into the
subretinal space; the fluid absorbed within several hours, and at
intervals after settling of the detachment, a comparison was made
between the force required to peel the retina from normal and
Fig. 19.6 Interdigitation between photoreceptor outer segments and reattached areas of retinal pigment epithelium (RPE). (Reproduced
the microvilli of the retinal pigment epithelium (RPE). Note that the with permission from Yoon YH, Marmor MF. Rapid enhancement
microvilli actually indent the outer segments during phagocytosis of of retinal adhesion by laser photocoagulation. Ophthalmology
“outdated” discs. The cone outer segments are shorter, and the 1988;95:1385–8. Copyright 1988, with permission from the American
microvilli form long pedicles in order to reach them. Academy of Ophthalmology.)
452
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

RPE
RPE

A B

Fig. 19.8 Fluorescence micrographs of monkey retina showing cone sheaths of the interphotoreceptor matrix (IPM) stained with peanut
agglutinin. Photoreceptors are above and retinal pigment epithelium (RPE) below. (A) Normal retina. The cone sheaths are short and thick.
(B) Partially peeled retina. The cone sheaths have been stretched markedly between the photoreceptors and the RPE surface, showing the
strength of IPM bonding to both sides (original magnification ×130). (Reproduced with permission from Hageman GS, Marmor MF, Yao XY, et al.
The interphotoreceptor matrix mediates primate retinal adhesion. Arch Ophthalmol 1995;113:655–60. Copyright © (1995) American Medical
Association. All rights reserved.)

stretching of WGA-staining material may be observed on peel­ with cytochemical evidence of damage to the IPM (Figs 19.9
ing freshly enucleated eyecup material.17,54,57 The role of newly and 19.10). It is unknown whether the effects of these enzymes
recognized matrix components such as galectins and IPM on adhesiveness are a result of changes in viscosity or damage
proteoglycans remains to be determined.58,59 to the structural elements of the IPM. However, adhesiveness
The ability of cone or rod matrix sheaths to serve as a struc- returns to normal roughly 3 weeks after exposure of an eye to
tural bond between retina and RPE may well depend on the neuraminidase or chondroitinase ABC, and the recovery of
presence of specific receptors that bind IPM components to the adhesiveness correlates closely with the recovery of normal
cell membranes. (“Cell adhesion molecules” represent a group PNA-binding properties in the IPM.72
of substances that mediate adhesion between individual cells or Experiments have shown that the chemical nature of the
between cells and substrates.60) Experiments have shown that IPM may change with light and dark adaptation.73 To the
extracted IPM material does not automatically stick cells to one extent that photic effects might modify the viscosity or bonding
another,61 but adhesion is unlikely unless specific receptors are properties of the IPM, light could be a modulating factor of
present to bind the macromolecular substances. Several receptor retinal adhesion. However, experimental data on adhesion in
systems, including sites for fibronectin, integrins, and mannose, the light and dark have been contradictory and of uncertain
have been proposed and disputed as being involved with retinal significance.11,74,75
adhesion.48,62–65 There is functional evidence that inhibition of
chondroitin sulfate proteoglycan synthesis with xylosides causes Subcellular components and mobility
spontaneous detachments in primates.48,66 Subcellular components of the RPE, such as microtubules and
Other types of evidence support the concept that IPM con- microfilaments, will be relevant to retinal adhesion to the extent
tributes significantly to retinal adhesion. Physical factors that that they influence remodeling and movement of the RPE micro-
affect adhesiveness, such as temperature, pH, and calcium con- villi.76 Actin filaments control melanin movement in the amphib-
centration, are known to be modulators of the physicochemical ian RPE11 and are present in mammalian RPE,77,78 but a movement
properties of IPM macromolecules and may also affect the of melanin granules has never been observed in mammals. A
bonding of large-molecular species at receptoral sites.67,68 There few attempts have been made to alter adhesion by inhibiting
is also a marked loss of retinal adhesiveness when the subretinal subcellular components of the RPE. Cytochalasin B, which
space is exposed to enzymes that degrade matrix compo- blocks microfilaments, was found to have no effect on adhesion
nents,15,69 such as chondroitinase ABC (which degrades chon- when the drug was presented to excised tissue briefly after enu-
droitin sulfate70) or neuraminidase (which breaks sialic acid cleation.12 However, injecting cytochalasin into the vitreous 4–72
bonds71). These enzymes, given intravitreally or into the sub- hours before enucleation caused a 70–90% reduction in pigment
retinal space itself, weaken adhesiveness markedly in correlation adherence as a measure of adhesiveness.79
RPE
453

Chapter 19
OPL
A B

Mechanisms of Normal Retinal Adhesion


Fig. 19.9 Fluorescence light micrographs depicting binding of peanut
agglutinin (PNA) to rabbit retinas in the region between the retinal
pigment epithelium (RPE) and outer plexiform layer (OPL). (A) In
a normal eye, PNA bound intensely and specifically to cone matrix
sheaths (arrows). (B) After injection of neuraminidase, PNA binds
throughout the interphotoreceptor matrix, with binding in the region
of cones being diminished (note gaps indicated by arrows). (C) After
intravitreal injection of testicular hyaluronidase, cone matrix sheaths
were disrupted, especially at their apices (asterisks), and shallow
separations between the photoreceptors and RPE were commonly
observed. (Reproduced with permission from Yao XY, Hageman GS,
Marmor MF. Retinal adhesiveness is weakened by enzymatic
C modification of the interphotoreceptor matrix in vivo. Invest Ophthalmol
Vis Sci 1990;31:2051–8.)

A B

C D

Fig. 19.10 Peeled whole-mount retinas after subretinal injection of enzymes. The areas of strong adhesion show retinal pigment epithelium
(RPE) pigment adherent to the retina after peeling; the areas of weak adhesion are clear of pigment. (A) Three days after injection of control
(Hanks) solution, adhesion is normal (strong) except at the injection site. One (B), 2 (C), and 3 (D) days after testicular hyaluronidase, there was
a progressive increase in the area of weakened adhesion beyond the injection site. A similar progression of adhesive loss was observed after
neuraminidase. (Reproduced with permission from Yao XY, Hageman GS, Marmor MF. Retinal adhesiveness is weakened by enzymatic
modification of the interphotoreceptor matrix in vivo. Invest Ophthalmol Vis Sci 1990;31:2051–8.)
When retina detaches, the surface morphology of the RPE One possible explanation for the postenucleation loss of
changes literally within minutes.23 The evolution of these changes adhesion is that the rapid release of lysosomal enzymes by
454 can be altered by both colchicine (which dissembles micro­ injured RPE alters the IPM.84 Enzyme release can occur quickly
tubules) and cytochalasin,80 suggesting that microtubules and after injury,84,85 and lysosomal enzyme activity is generally
microfilaments are important to apical RPE morphology. Actin enhanced by warm temperature, low pH, and low calcium
filament contraction and subcellular motility are known to levels, all of which reduce retinal adhesiveness. However, the
Section 1

require calcium,81 the absence of which is severely detrimental loss of adhesion under warm temperature, low pH, and low
to retinal adhesion. However, we do not know whether the calcium conditions is rapidly reversible by restoring normal
action of calcium in weakening adhesiveness relates to this environmental conditions,16 which would seem unlikely if irre-
mechanism or to effects that calcium may have on transport versible degradation of the IPM had taken place. We have
systems and matrix-binding properties. also been unable to find any cytochemical or morphologic
changes in the IPM that correlate with the postmortem period
Anatomy and Physiology
Basic Science and Translation to Therapy

Metabolic factors loss of adhesion.


The question of whether or why metabolic activity is necessary The postmortem failure can be retarded or even reversed
or even contributory to retinal adhesion is extremely important. simply by improving or restoring oxygenation.82 With enhanced
From a physiologic standpoint, we would like to know the inter- oxygenation, retinal adhesion in excised rabbit tissue can be
action between RPE transport, IPM properties, and the passive maintained for 15–30 minutes (instead of 1–2 minutes) without
physical forces that impinge on the subretinal space. From a postmortem failure (Fig. 19.11B); adhesive failure that occurred
clinical standpoint, if adhesion is purely passive (i.e., dependent after a few minutes without oxygen was rapidly reversed by
on pressure, or “glue”), therapy for clinical detachments would oxygenating the bath. In monkeys, adhesiveness may persist in
be primarily mechanical, whereas if active metabolism is neces- oxygenated Ames solution for 1 hour or more.83
sary for adhesion, physicians may have a very different range of These results are extremely important with respect to the
therapeutic options. mechanisms of adhesion because they show that, although
IPM may account for much of the mechanical bond between
Critical dependence on oxygen retina and RPE, continuous oxygenation and active metabolic
It was noted previously that adhesiveness in living rabbit eyes activity by RPE and photoreceptors are vital to the maintenance
falls severely within a few minutes of death5 (Fig. 19.1A). Fur- of adhesive strength. The critical activity might be subretinal
thermore, just 1 minute of ocular ischemia in living rabbits fluid transport, which dehydrates the subretinal space, or it
weakens adhesion dramatically, whereas restoration of circula- might be the metabolic control of membrane systems that
tion restores adhesiveness.82,83 The postmortem adhesive loss modulate local pH, calcium concentration, or other factors that
appears to be slower in primates and humans, and under similar influence the bonding properties of the IPM. Metabolic failure
in vitro conditions, at least a moderate adhesive force was mea- could also affect the synthesis of IPM components and the
surable for 30 minutes or more after enucleation of primate and function of subcellular organelles that influence microvillous
human tissue15,17,83 (Fig. 19.11A). motility.

100
Retinal coverage with pigment (%)

90 Circulating
80 O2
70 Static
Circulating O2
60 (PO2 500)
50
40
30 Static
20 (PO2 205)
10
0
0 5 10 15 20 25 0 5 10 15 20
A B Incubation time (min)

Fig. 19.11 Dependence of retinal adhesion on metabolic activity. (A) Scanning electron micrograph of the photoreceptor surface of human retina
peeled from the retinal pigment epithelium (RPE) within 4 minutes of enucleation. The outer segments are largely covered by apical fragments of
RPE cells, indicating that the strength of retinal adhesion in living or fresh tissue is greater than that of the RPE cell membrane. Retinas peeled
1 hour after enucleation showed little or no adherence of RPE fragments. Bar = 10 µm. (Reproduced with permission from Marmor MF, Yao XY.
The metabolic dependency of retinal adhesion in rabbit and primate. Arch Ophthalmol 1995;113:232–8.) (B) Effect of oxygenation on the
adhesiveness of rabbit retina in vitro. Left, Postmortem adhesive failure was delayed markedly by the incubation of tissue in an oxygenated
rather than static bath. Right, Switching tissue from a static bath to an oxygenated one caused a prompt recovery of adhesive strength. (Modified
with permission from Marmor MF, Yao XY. The metabolic dependency of retinal adhesion in rabbit and primate. Arch Ophthalmol 1995;113:
232–8.)
Metabolic inhibitors and other agents of fluid across the RPE,3,89 and its effects on adhesion may well
be a result of fluid movement, since the enhancement of outward
A number of experiments have shown that general metabolic
fluid movement by other means such as mannitol36 also increases 455
inhibitors have a deleterious effect on the process of retinal ad­
adhesiveness.36,37
hesion. For example, the introduction of cyanide into the experi-
Cyclic adenosine monophosphate (cAMP), which has been
mental bath reduces the measured adhesive force and hastens

Chapter 19
shown by several groups to inhibit the outward transport of
the postmortem failure of adhesion.3 In vivo experiments with
fluid from the subretinal space,92–94 reversibly weakens retinal
dinitrophenol placed in the subretinal space showed that retinal
adhesive force in vitro12 (Fig. 19.14). cAMP is present in the RPE,
adhesiveness was rapidly reduced to an unmeasurable level10
but it is not known whether it actively modulates adhesive force
(Fig. 19.12). Sodium iodate and hemicholinium-3 also cause
in the living eye. In rabbits with experimental detachments,
retinal adhesiveness to fall rapidly to a low level,86–89 but these
cyclic guanosine monophosphate (cGMP) has the opposite effect
effects may involve a variety of mechanisms because these
of cAMP on RPE fluid transport (i.e., it facilitates outward

Mechanisms of Normal Retinal Adhesion


agents affect not only metabolic functions of the RPE (and, more
slowly, the photoreceptors) but also the physical integrity of the
RPE cells and blood–retinal barrier.87,90
Specific transport inhibitors, in contrast, may act very selec-
tively on different aspects of the adhesive process. For example, 30
ouabain, which blocks the electrogenic sodium pump, will ulti- Control
mately kill photoreceptor and RPE cells. However, in the short Mannitol
25
term its effect is to strengthen adhesiveness3,10 (Fig. 19.12)
because sodium entry causes cellular swelling that tightens

Peeling force (mg)


20
(presumably) the interdigitation between RPE microvilli and
outer segments.13 This is a pathologic effect, of course, and is
probably of little relevance to normal mechanisms of adhesion. 15
Acetazolamide, which inhibits carbonic anhydrase, causes an
increase in retinal adhesiveness, measured in vitro in rabbits3,88 10
and in vivo in rabbit and monkey7 (Fig. 19.13). The drug is
effective only when given systemically and does not enhance 5
adhesion if placed in the subretinal space8 or in an experimental
bath.3,91 Acetazolamide enhances the apical-to-basal transport 0
A 4 6 8 10 12
Time after enucleation (min)

200
15 Monkey Acetazolamide
(18) * 175
(15 mg/kg)
(24)
Retinal adhesive force (%)

150
Retinal adhesive force (dyn/cm)

(36) (21)
125
10 (24) *
(19) *
100
(41) *
75

5 50

25

0
(3) * B 0 1 2 3 4 5
0 Time after injection (h)
Hanks'

DNP

cAMP

cGMP

Ouabain

Acetazolamide

Furosemide

Amiloride

Fig. 19.13 Effects of acetazolamide on retinal adhesiveness.


(A) Retinal adhesion in the rabbit is increased by systemically
administered acetazolamide. The graph shows the force required to
peel retina from the retinal pigment epithelium (RPE) at four time
intervals after enucleation, with and without acetazolamide. (Modified
Fig. 19.12 The retinal adhesive force after injection of active agents with permission from Marmor MF, Maack T. Local environmental
into the subretinal space. Columns show mean value; bars show factors and retinal adhesion in the rabbit. Exp Eye Res 1982;34:727–
standard error; the numbers of experiments are in parentheses; 33.) (B) The time course of change in retinal adhesive force in three
asterisks indicate a significant difference from control (P ≤ 0.05). monkeys after intravenous injection of a clinical dose (15 mg/kg) of
DNP, dinitrophenol; cAMP, cyclic adenosine monophosphate; cGMP, acetazolamide. A control value (100%) was derived from each eye
cyclic guanosine monophosphate. (Reproduced with permission from before administration of the drug. (Reproduced with permission from
Kita M, Marmor MF. Retinal adhesive force in living rabbit, cat, and Kita M, Marmor MF. Retinal adhesive force in living rabbit, cat, and
monkey eyes: normative data and enhancement by mannitol and monkey eyes: normative data and enhancement by mannitol and
acetazolamide. Invest Ophthalmol Vis Sci 1992;33:1879–82.) acetazolamide. Invest Ophthalmol Vis Sci 1992;33:1879–82.)
choroidal pressure is relatively elevated (e.g., through venous
obstruction, ocular hypotony, systemic hypertension, or other
20 Control
456 causes), there will be a risk of fluid following the pressure
+ cAMP
gradient to enter the subretinal space and cause a nonrheg-
15
matogenous (serous) detachment. Passive adhesive systems are
Peeling force (mg)

important to minimize the spread of fluid laterally, but the criti-


Section 1

cal determinant of whether a detachment forms and how large


10 it grows will be the ability of the RPE surrounding the “leak” to
remove fluid as quickly as it enters the subretinal space. This
concept of serous detachment2 is discussed later.
5 Since the subretinal fluid of both serous and rhegmatogenous
detachments has a high concentration of protein, the question
Anatomy and Physiology
Basic Science and Translation to Therapy

may be asked whether oncotic pressure from this protein con-


0 tributes to the formation or persistence of detachments by
4 6 8 10 12 drawing in fluid. However, in experiments that introduced
Minutes after enucleation albumin (serum) or larger molecules into the subretinal space of
rabbits (Fig. 19.15), fluid was absorbed within a few hours
Fig. 19.14 Retinal adhesive force is weakened by exposure to cyclic regardless of whether protein or other larger molecules remained
adenosine monophosphate (cAMP). The graph shows the force in the subretinal space.100–102 Furthermore, retina is sufficiently
required to peel rabbit retina from the retinal pigment epithelium (RPE)
at various times after enucleation. For each strip of eyecup, three permeable to ions and water that the osmotic pressure of the
measurements of peeling force were made: one in control solution, subretinal space is continuously equilibrating with that of the
one after cAMP was added to the bath, and one after the tissue was vitreous to neutralize gradients102,103 (Fig. 19.15B). In fact, albumin
returned to control solution. The addition of cAMP always weakened can also diffuse slowly across the retina. When serum is injected
adhesion, but the effect was reversible on removing it from the bath.
(Modified with permission from Yoon YH, Marmor MF. Effects on into the subretinal space, the subretinal fluid loses roughly
retinal adhesion of temperature, cyclic AMP, cytochalasin B, and 5% of its protein concentration per hour into the vitreous
enzymes. Invest Ophthalmol Vis Sci 1988;29:910–4.) (Fig. 19.15A and C). Thus, the high protein content of subretinal
fluid is not self-maintaining and is not the cause of detachment;
rather it is the result of continued protein influx from the RPE
or vitreous. Faulty RPE transport may also contribute to the
movement),93,95 but it has not been shown to have an effect on retention of fluid within the retina, i.e., macular edema.104
retinal adhesiveness.
Retinal adhesiveness in vivo in the rabbit was also decreased,
relative to normal, to 86% with furosemide and 81% with
PHARMACOLOGIC MODIFICATION
amiloride10 (Fig. 19.12). Both these agents reduce apical–basal OF ADHESION
fluid transport across epithelial cells.96,97 Pharmacologic agents that modify retinal adhesive force could
be useful clinically in several ways. Agents that enhance adhe-
Relationship of adhesion to subretinal fluid sion could help reduce the risk of retinal detachment in high-risk
transport and subretinal protein eyes. They might minimize the spread of an existing detachment
The RPE actively transports water from the subretinal space to while awaiting surgical repair. They could improve or hasten
the choroid23,92,98 and experiments on the control of subretinal the healing process after surgical repair of a detachment. To the
fluid transport3,27,28,32,91,93,95,99 show that many of the factors affect- extent that they enhance the absorption of subretinal fluid, they
ing adhesion also influence transport in a similar direction. This would speed the recovery of nonrhegmatogenous, as well as
concurrence may, in part, be fortuitous, since both physiologic rhegmatogenous, detachments and minimize the risk of rede-
processes involve the RPE; thus, conditions that nonspecifically tachment. If drugs can compensate in part for metabolic factors
stimulate or inhibit the RPE will have a similar effect on adhe- that fail in older eyes (which are more susceptible to detachment
sion and fluid absorption. However, there may also be a more than younger ones) or in specific diseases that are predisposed
direct connection insofar as conditions that dehydrate the sub- to detachment, the risk of detachment might be decreased in
retinal space should serve to keep the retina closely apposed, the these selected populations. Agents that reduce adhesion could
matrix viscous, and the matrix molecules tightly interactive at facilitate an atraumatic separation of the retina in surgical pro-
ionic or receptor-binding sites. Conversely, conditions that block cedures such as the removal of subretinal lesions, macular trans-
subretinal fluid transport and allow the subretinal space to location, RPE transplantation, and injections of therapeutic
become hydrated would seem intuitively to weaken adhesive- agents such as a viral vector for gene transfer or stem cells for
ness. Thus, part of the metabolic requirement for retinal adhe- retinal regeneration.105,106
sion is almost certainly a direct result of the fact that subretinal
fluid transport under normal conditions takes place primarily by Mannitol
active metabolic transport across the RPE. Adhesiveness is also Mannitol is an accepted therapeutic agent in humans for glau-
increased by raising systemic osmolality with mannitol,9,36,37 coma and cerebral edema. In systemic doses that are used clini-
which passively enhances the absorption of subretinal fluid.38 cally, it causes a 50% increase in retinal adhesiveness, measured
In certain conditions the efficacy of subretinal fluid transport both in excised tissue in vitro37 and in the living eyes of primates
is clearly a critical factor in maintaining adhesion. For example, and rabbits9,36 (Fig. 19.5). The magnitude and duration (2–3
under conditions where defects occur in the RPE barrier and hours) of the mannitol effect are directly related to the increase
35
1200
457
Subretinal protein Vitreal protein

Chapter 19
1000

30
800
mg/mL

Mechanisms of Normal Retinal Adhesion


µg/mL
600

25
400

200

A 20 C 0
0 1 2 3 4 0 1 2 3 4
Hours
300
Subretinal osmolality

295
mOsm/kg

Fig. 19.15 Changes in protein concentration and osmolality after


injecting highly proteinaceous fluid into the subretinal space of rabbit
290 eyes. (A) Protein concentration of subretinal fluid. There is a steady
decrease despite simultaneous water absorption. (B) Osmolality of the
subretinal fluid. The osmolality of the injected fluid was initially low, but
it equilibrated to vitreous levels (293 mosmol/kg) within 2 minutes and
then remained steady despite the changing subretinal protein
285 concentration. (C) Protein concentration of vitreous. There was
0 1 2 1 2 3 4 a steady increase as protein diffused from the subretinal space.
B (min) Hours (Modified with permission from Takeuchi A, Kricorian G, Marmor MF.
Albumin movement out of the subretinal space after experimental
retinal detachment. Invest Ophthalmol Vis Sci 1995;36:1298–305.)

in blood osmolality. However, the effect is not simply a result of drugs, but could in theory be used to produce or facilitate retinal
fluid movement “pulling” the retina against the RPE, because it separation in conjunction with procedures that require surgical
is measurable in vitro (where there is no transretinal flow). More elevation of the macula. Limitations are the minutes required for
likely, it depends on dehydration of the IPM, which strengthens detachment to develop and the associated cellular damage if the
bonding characteristics. osmotic load is too great.
Intravenous mannitol might be useful preoperatively for
short-term stabilization of retinal attachment (in a case with Acetazolamide
partial detachment) or postoperatively to enhance the absorp- The acetazolamide effect is slightly weaker than mannitol in
tion of fluid and increase the initial strength of bonding of retina the clinical doses we have used, causing a 30–45% increase in
to RPE. The drug might even be given intraoperatively to help retinal adhesiveness in primates and rabbits9,17 (Fig. 19.13). The
minimize retinal separation during vitrectomy. The benefits are acetazolamide effect lasts for 3–4 hours after a single injection,
limited, however, because the drug cannot be given repeatedly but acetazolamide can be given safely over long periods
over long periods. This would seem to reduce its value in the (although currently it is unknown whether the positive effects
management of serous nonrhegmatogenous detachments, on adhesion would persist). Acetazolamide increases subretinal
although it might help to clear the accumulated fluid in cases fluid transport91 and thus could theoretically help to absorb
where the source of leakage is no longer active. fluid postoperatively or in cases of serous detachment. However,
When hyperosmotic solutions are injected into the vitreous judging by anecdotal reports, researchers have not found acet-
cavity,25 the gradient is in the opposite direction and retinal azolamide to be dramatically effective in the management of
detachment can be induced (Fig. 19.4). This is a risk with certain central serous retinopathy. The problem may be that the effect
of acetazolamide on subretinal fluid transport is not very strong adhesiveness. Conversely, when detachment is created surgi-
and can only occur when the RPE is basically healthy and cally such as during macular translocation, it may be useful to
458 receptive to metabolic stimulation.107 In a disorder such as create conditions that weaken adhesiveness to make the proce-
central serous retinopathy, where RPE transport is probably dure as atraumatic as possible.
compromised as a part of the disease, there may not be a Two agents have already been documented to enhance retinal
normal substrate of RPE on which the acetazolamide can work. adhesiveness to a significant degree – mannitol and acetazol-
Section 1

Although acetazolamide and related carbonic anhydrase amide. cGMP might theoretically be of use, and a study of
inhibitors have not been clinically useful for detachment disor- related agents may reveal some that are clinically beneficial.
ders, they have proved quite useful for the removal of foveal Other options for the future include agents that modify local pH
cystic fluid in a number of disorders. They have not been of great or calcium concentration, strengthen the binding properties of
use in postcataract cystoid edema and diabetic macular edema, adhesion molecules within the IPM, or stimulate microvillous
where fluid continuously enters from vascular sources, but they growth and ensheathment of outer segments. Even a “patho-
Anatomy and Physiology
Basic Science and Translation to Therapy

can clear intraretinal cysts dramatically where the fluid is rela- logic” effect of cold temperature in strengthening adhesion
tively static in disorders such as retinitis pigmentosa, X-linked through cellular edema might prove useful in temporary clinical
juvenile retinoschisis, enhanced S-cone syndrome and some- situations such as vitreous surgery.
times macular epiretinal membrane formation.104,107–111 The effec- Conversely, conditions that reduce adhesiveness, such as
tiveness probably depends on reasonable preservation of RPE intravitreal hyperosmolarity, metabolic inhibition, low Ca2+, and
metabolic function under the fovea, so that the drug can augment low pH, might prove useful in subretinal surgery where retinal
transport. One limitation to its use for retinal edema is the sys- separation needs to be produced transiently and with little cel-
temic side-effects, which may relate in part to its action on intra- lular damage. Low-Ca2+, low-Mg2+ solutions have already been
cellular carbonic anhydrase throughout the body. Agents like shown to weaken adhesion and facilitate experimental detach-
benzolamide, which affect only the membrane-bound enzymes ment in rabbits.75,105,117 Metabolic inhibitors, such as dinitrophe-
that are relevant to RPE transport, may have fewer side-effects.112 nol, should in theory also be effective if their effects could be
Recent work has shown that enough drug may enter from topical made transient and nontoxic. Another agent that might reduce
carbonic anhydrase drops to produce a clinical effect, although adhesiveness is cAMP. Some of these approaches are likely to
the onset may take months instead of days.113,114 The action of reach clinical practice within the next few years.
both oral and topical carbonic anhydrase inhibitors seems
subject to tachyphylaxis (rebound of cystic edema), and cysts Recovery after rhegmatogenous
may return after roughly a year of usage; however, the effects retinal detachment
will usually return after several months off treatment.115,116 Retinal reattachment forces return following reapproximation of
the sensory retina and RPE. This is observed both without
Cold temperature and ouabain retinopexy and following such therapy, which augments the
Both cold temperature and ouabain increase retinal adhesive- adhesion.
ness,3,7 but their action depends on pathologic changes (cellular
swelling) rather than a true enhancement of the mechanisms of Recovery of adhesiveness
adhesion.13 Neither cold nor ouabain therefore can be used for without retinopexy
any long-term clinical applications, but they may be valuable in Recovery after retinal detachment depends on a variety of
special situations. For example, cooling the irrigating solution factors, including the length of detachment (which may affect
during vitrectomy might reduce the risk of retinal separation the survival of cells) and effectiveness with which adhesive
during the manipulation of surgery. mechanisms can be re-established. We have measured in rabbits
the retinal peeling force in vitro45 after making small experimen-
Ionic changes
tal detachments that spontaneously settled within hours to a few
The removal of local calcium and magnesium ions, or lowering days. The results show that restoration of normal adhesive
the pH, causes a dramatic fall in retinal adhesive force. Thus strength requires 4–6 weeks (Fig. 19.7). Keep in mind that these
transient irrigation of the retina with calcium-free solution, or a experiments involved the injection of a small amount of bal-
solution of low pH, might facilitate the production of atraumatic anced salt solution into the subretinal space; in clinical detach-
surgical separation in association with submacular surgery. ments, where the fluid is serous and the retina is separated for
Alternatively a small amount of such solution might be injected longer periods, there could well be more trauma to the RPE or
into the subretinal space to weaken adhesion and make enlarge- outer segments and thus an even more prolonged recovery
ment of the surgical detachment easier. Preliminary studies on period. It remains to be shown which adhesive factors are rate-
this approach have been reported,75,105 but RPE or retinal toxicity limiting for the recovery process, considering that full recovery
is a risk, and guidelines for safe administration still need to be involves the restoration of anatomic interdigitation, RPE syn-
established. thetic capabilities, IPM structure and binding, and subretinal
fluid transport. These data show how clinical recovery after
IMPLICATIONS FOR retinal detachment is possible without retinopexy,118–120 although
VITREORETINAL SURGERY it is slow and would only be advisable when mechanical (vitre-
ous) traction forces have been fully relieved.
Understanding mechanisms of retinal adhesion, both positive
and negative, is relevant to vitreoretinal surgery. During proce- Effects of retinopexy
dures of repair, or whenever retinal detachment would be a Retinal surgeons frequently use retinopexy (i.e., scarification of
complication, it may be useful to create conditions that maximize the retina and RPE with laser photocoagulation, diathermy, or
cryotherapy) as a means of creating an extra-strong adhesion Thus, all forms of retinopexy appear to be effective in the
between retina and RPE that will strengthen attachment and long run, presumably by inducing scar formation; however, if a
prevent the lateral movement of subretinal fluid. Experiments rapid bond is required, laser photocoagulation or diathermy is 459
on the in vitro peeling force45 and the in vivo adhesive force120 preferable.
show that laser photocoagulation produces a bond that
Effects of vitreous in the subretinal space

Chapter 19
approaches normal adhesive strength within 24 hours, possibly
from local effects such as fibrin formation (Fig. 19.16). The ad­ One of the factors contributing to many rhegmatogenous detach-
hesive strength continues to increase gradually and reaches ments is vitreous syneresis and liquefaction, which allow fluid
levels roughly twice normal by 2–3 weeks after the photocoagu- to percolate through the retinal tear and expand or maintain the
lation.45,121,122 Diathermy effects, measured in vivo, have a similar elevation of retina. Although there is some evidence that hyal-
time course. Cryotherapy, however, weakens adhesion for the uronic acid may be toxic in high concentrations to RPE,123 clinical
first week, after which the adhesive force rises to the same levels data indicate that liquid vitreous is tolerated insofar as a reason-

Mechanisms of Normal Retinal Adhesion


as with other forms of retinopexy. The initial weakening after able recovery of retinal function generally occurs if detachments
cryotherapy probably relates to local inflammation or edema. are repaired within a few days or weeks. The presence of lique-
fied vitreous in the subretinal space has no immediate effect on
the rate of fluid transport across the RPE.124 The concentration of
protein in normal vitreous is only about 1% of that in serum, but
when vitreous enters the subretinal space the concentration of
subretinal protein rises slowly.124 This occurs because water is
Peeling force relative to normal retina (%)

320
pumped out faster than the protein can be removed, whereas the
280 Immediate laser
continued entry of liquid vitreous brings in more protein. This
photocoagulation
240
mechanism may contribute to the high protein concentration of
subretinal fluid in chronic rhegmatogenous detachments. Serum
200 and protein may also leak in from the choroid to the extent
that the RPE has been damaged under the conditions of
160 detachment.
120 PATHOPHYSIOLOGY OF
80 SEROUS DETACHMENT
No treatment For detachment to occur there must be forces that cause the
40
retina to separate and also conditions that allow subretinal fluid
0 4 8 12 16 20 24 28 to persist. The clinical condition of serous detachment provides
Days after reattachment a case in point. In central serous chorioretinopathy (CSC), fluo-
rescein angiography demonstrates apparent “leaks” through the
Fig. 19.16 Laser photocoagulation of freshly reattached retina can RPE, defects through which fluid enters the subretinal space
rapidly enhance the strength of retinal adhesion. Two experimental from the choroid. However, experimental work has clearly
detachments were made in each eye (Fig. 19.7), and after the
shown that subretinal fluid overlying an intact RPE is removed
subretinal fluid had spontaneously absorbed, the base of one
detachment was covered with laser photocoagulation. The force very actively by active metabolic transport and that damage to
required to peel retina from the reattached areas, with or without the RPE barrier in general makes the absorption of fluid faster
photocoagulation, was compared to the force required in the rather than slower (Fig. 19.17). For example, damaging the RPE
intervening normal areas. The adhesive force in the photocoagulated
regions exceeded normal within hours and reached an apparent focally by mechanical micropipette injury or laser burns or dif-
maximum after about 2 weeks (although at these levels of force the fusely by the systemic administration of sodium iodate hastens
retina frequently tore rather than separated, and the “true” adhesive the absorption of subretinal fluid.125,126 This occurs because intra-
strength may be even greater). (Reproduced from Yoon YH, Marmor ocular pressure and the osmotic absorption of the choroid both
MF. Rapid enhancement of retinal adhesion by laser photocoagulation.
Ophthalmology 1988;95:1385–8. Copyright 1988, with permission from move fluid in an outward direction, and their effectiveness is
the American Academy of Ophthalmology.) increased when the RPE flow barrier is removed. Even serous

Fig. 19.17 Conditions that control the


Subretinal fluid movement of fluid across the retinal pigment
epithelium (RPE). Left, When the tight
junctions are intact between normal RPE
cells, fluid is transported across by active
transport. Middle, If the tight junctions are
damaged, fluid can cross passively under the
ATP ATP ATP ATP influence of hydrostatic and osmotic
ATP ATP
pressure. Right, If the tight junctions are
intact and the metabolic transport capability
of the RPE has been compromised, then fluid
is unable to cross. ATP, adenosine
Normal RPE Breakdown of Intact barrier but triphosphate.
blood–retinal barrier compromised transport
fluid is absorbed at least as fast over damaged as over intact example, the absorption of subretinal fluid in rabbits is slowed
RPE.101,103 Furthermore, to cause detachment the entering fluid by the ligation of vortex veins.130 Furthermore the choroid is
460 must overcome the retinal adhesive forces at the margin of the typically thickened in CSC, which may signify increased hydro-
leak. Thus the mere presence of a focal defect in the RPE does static pressure that would foster serous detachment both directly
not “cause” serous detachment.127 and through effects upon RPE transport.106 Under conditions
How then do serous detachments form?2,128,129 The question is that compromise fluid absorption and possibly adhesiveness but
Section 1

not whether the leaks are the source of fluid – they must be – but do not destroy the blood–retinal barrier, fluid that enters from a
why the fluid accumulates when entry is slow and one would focal leak may persist and accumulate within the subretinal
expect subretinal fluid to be cleared rapidly by active transport space (Fig. 19.19). One may ask whether serous detachment can
(across a normal RPE) or by passive mechanisms (across be accounted for simply by oncotic pressure when a protein-rich
damaged RPE). On reflection, the one condition that would fluid leaks into the subretinal space. However, as noted earlier,
allow fluid to accumulate is when the RPE barrier function protein in the subretinal space does not prevent fluid absorption;
Anatomy and Physiology
Basic Science and Translation to Therapy

remains intact around the leak, whereas active fluid transport is it is osmotically neutralized by the diffusion of water and ions
impaired (Fig. 19.18). In this situation fluid cannot leave pas- from the vitreous, and it will not persist unless there is a con-
sively or actively. I suspect that many serous detachments form tinual influx of new proteinaceous fluid.
because there is widespread functional damage to the process of Three conditions appear to be necessary for serous fluid to
transport across the RPE (either from RPE disease directly or accumulate129,130: (1) a defect in the RPE barrier that allows access
from disease of the underlying choroid and choriocapillaris). For to the subretinal space; (2) a source of fluid pressure, to move

Fig. 19.18 Transport capabilities of the


Serous detachment retinal pigment epithelium (RPE) can
determine whether or not a serous
detachment will form in the presence of fluid
leakage through an RPE defect. Left, When
No detachment the active systems are working at normal
efficiency, fluid can be pumped out as fast
as it enters the subretinal space, and little
Retina or no detachment will develop. Right, When
RPE transport has been compromised, then
a serous detachment will form until a large
enough area of RPE is exposed to subretinal
RPE fluid to allow outward transport to balance
the inward leakage of fluid.

Healthy metabolic Leak Compromised


transport transport

A B

Fig. 19.19 Fluorescein angiograms from different fundus regions of the same cat, showing the effects of diffuse retinal pigment epithelium (RPE)
damage on the accumulation of serous fluid. (A) Control region. Weak laser burns were applied after the animal was given intravenous rose
Bengal to photosensitize the choriocapillaris. Fluorescein leaks from the burn sites but spreads very little. (B) RPE-damaged region. This area
was pretreated with intense white light to injure the RPE diffusely before giving the rose Bengal and laser burns. The dye leakage extends much
farther beyond the burn sites and slowly fills an overlying detachment. (Unpublished data reproduced with permission from Marmor MF, Yao XY.
Conditions necessary for the formation of serous detachment: experimental evidence from the cat. Arch Ophthalmol 1994;112:830–8.)
fluid in; and (3) an impairment of outward fluid transport (or a inhibitors that enhance RPE transport has not shown great value,
broad area of leakage), so fluid spreads and persists in the sub- presumably because the RPE transport mechanisms are compro-
retinal space. Weakness of retinal adhesion may be an additional mised by the disease. However, as noted earlier, these agents can 461
facilitating factor. Unless all three basic conditions are present, be very effective in clearing cystic intraretinal fluid, especially
detachment will not occur. For example, attempts to induce in retinal dystrophies including retinitis pigmentosa, X-linked

Chapter 19
serous detachment in rabbits by damaging the RPE focally while retinoschisis, and enhanced S-cone syndrome.
altering intraocular pressure (conditions 1 and 2 only) have been
largely unsuccessful.118 However, diffuse injury to the RPE and CONCLUSIONS AND
underlying choriocapillaris (by photosensitization with rose
Bengal dye131 or by toxic effects of N-ethylmaleimide132) leads to
GENERAL IMPLICATIONS
the formation of bullae (Fig. 19.20), since the damaged capillaries One important conclusion to be derived from the evidence
not only serve as a source of fluid, but also compromise RPE reviewed in this chapter is that retina “wants” to adhere. A

Mechanisms of Normal Retinal Adhesion


transport and barrier functions. In clinical practice, choroidal variety of systems, including passive hydrostatic forces, the
vascular damage is probably the most frequent primary cause of interdigitation of outer segments and RPE microvilli, the active
serous detachments and may underlie even the focal RPE defects transport of subretinal fluid, and the complex structure and
of CSC.106,128 binding properties of the IPM, all work to keep the retina prop-
If CSC is more a disease of diffuse transport dysfunction, erly apposed to the wall of the eye. Thus retinas do not detach
involving RPE or choriocapillaris, than a result of the focal simply because there is a hole or a tear or even a leak in the RPE;
“leak” seen on angiography,2 why does laser photocoagulation there must be positive traction pulling it off or a positive force
of the leak help to clear the fluid? Because laser therapy may pushing fluid into the subretinal space (often in conjunction with
effectively seal the visible leak, it removes the immediate source underlying pathologic conditions of the RPE or choriocapillaris
of fluid and allows the existing fluid to absorb slowly. However, that block the normal transport processes that remove subretinal
recurrences will be common as long as the underlying transport fluid). Retinal detachment is not simply a disease of physical
dysfunction persists, and this is indeed the clinical course of the separation but is the result of a vital battle between a destructive
disease. In further support of the concept of diffuse dysfunction, influence and the collection of forces routinely at work con­
serous retinopathy is often associated with systemic disease of structively to maintain attachment.
the vascular system (especially choroidal ischemic disorders) or This multifactorial nature of retinal adhesiveness is impor­
with emotional stress,133–136 and the serous fluid accumulations tant clinically, as well as physiologically. To function optimally,
are often bilateral and multifocal. Serous detachments may occur adhesive systems require proper anatomic relations, healthy
in patients taking high levels of corticosteroids,137,138 and the metabolic activity, and an appropriate local environment (pH,
chronic administration of epinephrine can produce serous temperature, Ca2+ concentration) in the subretinal space. This
detachments in experimental animals.139 The most effective treat- means that adhesion may be weakened by diseases with various
ment for CSC may ultimately be pharmacologic intervention sites and modes of action; at the same time, the complementary
that blocks the adverse adrenergic and corticosteroid effects.128 mechanisms for attachment undoubtedly provide safety cover-
Treatment with acetazolamide or other carbonic anyhdrase age for one another, so injury or illness of one adhesive system
does not necessarily doom the eye to total detachment. The
multiplicity of factors also allows us to consider a variety of
therapeutic options that might otherwise not be available. For
example, the presence of holes in the retina obviates the use
of fluid pressure to aid retinal apposition but leaves open
options for encouraging fluid transport pharmacologically or
for creating scars as a means of restoring local adhesion. As
the different mechanisms of adhesion become better understood,
more specific medical and surgical approaches to the preven-
tion, as well as the management, of detachments should become
available.
The importance of metabolic activity in maintaining retinal
adhesion cannot be emphasized too strongly. If retinal attach-
ment were solely a passive process, then detachment would be
solely at the mercy of passive forces and physical means of
repair. To the extent that the metabolic health of the RPE appears
to be critical to adhesion, we can better understand the failure
Fig. 19.20 Histology from the margin of an experimental serous of adhesion in certain disease states or ocular locations, and we
detachment in the rabbit, induced by illumination of the fundus after may ultimately realize more effective means of treatment. The
injection of the photosensitizing dye, rose Bengal. This treatment propensity of aged eyes to suffer detachment much more than
causes vascular injury, and the animal was cooled to prevent choroidal
thrombosis. There was diffuse fluorescein leakage across the base of younger ones, for example, may in part lie in the gradual meta-
the detachment, and the detached area (right) shows retinal pigment bolic failure of the RPE. The retinal periphery has a less effective
epithelium damage over patent (but presumably injured) vascular supply than the posterior pole, and this, too, is where
choriocapillaris. (Reproduced with permission from Yao XY, Marmor
most detachments begin. This discussion is not intended to mini-
MF. Induction of serous retinal detachment in rabbit eyes by pigment
epithelial and choriocapillary injury. Arch Ophthalmol 1992;110:541–6. mize the well-documented role of vitreous syneresis and vitre-
Copyright © 1982 American Medical Association. All rights reserved.) ous traction in creating detachments. However, by improving
the metabolic status of peripheral retina and RPE, we might be 32. Negi A, Kawano S, Marmor MF. Effects of intraocular pressure and other
factors on subretinal fluid resorption. Invest Ophthalmol Vis Sci 1987;28:
able to reduce the incidence of detachment and improve the 2099–102.
462 success rate of surgical repair. Cutting vitreous bands and 33. Rutnin U, Schepens CL. Fundus appearance in normal eyes. IV. Retinal breaks
and other findings. Am J Ophthalmol 1978;64:1063–78.
sealing holes manage only one aspect of the disease; we will treat 34. Foulds WS. The vitreous in retinal detachment. Trans Ophthalmol Soc UK
the whole disease only when we can also treat the pathologic 1975;95:412–6.
conditions within the subretinal space. 35. Hammer ME. Retinal re-attachment forces created by absorption of subretinal
Section 1

fluid. Doc Ophthalmol 1981;25:61–75.


36. Kita M, Marmor MF. Systemic mannitol increases the retinal adhesive force
REFERENCES in vivo. Arch Ophthalmol 1991;109:1449–50.
1. Steinberg RH, Wood I. Pigment epithelial cell ensheathment of cone outer 37. Yao XY, Moore KT, Marmor MF. Systemic mannitol increases retinal adhe-
segments in the retina of the domestic cat. Proc R Soc Lond B 1974;187: siveness measured in vitro. Arch Ophthalmol 1991;109:275–7.
461–78. 38. Negi A, Marmor MF. Effects of subretinal and systemic osmolality on the rate
2. Marmor MF. New hypothesis on the pathogenesis and treatment of serous of subretinal fluid resorption. Invest Ophthalmol Vis Sci 1984;25:616–20.
retinal detachment. Graefes Arch Clin Exp Ophthalmol 1988;226:548–52. 39. Osterlin S. On the molecular biology of the vitreous in the aphakic eye.
Anatomy and Physiology

3. Marmor MF, Abdul-Rahim AS, Cohen DS. The effect of metabolic inhibitors Acta Ophthalmol 1977;55:353–61.
Basic Science and Translation to Therapy

on retinal adhesion and subretinal fluid resorption. Invest Ophthalmol Vis Sci 40. Foulds WS. Experimental retinal detachment. Trans Ophthalmol Soc UK
1980;19:893–903. 1963;83:153–70.
4. Zauberman H, DeGuillebon H. Retinal traction in vivo and postmortem. 41. Anderson DH, Fisher SK. The relationship of primate foveal cones to the
Arch Ophthalmol 1972;87:549–54. pigment epithelium. J Ultrastruct Res 1986;67:23–32.
5. De Guillebon H, De la Tribonniere MM, Pomerantzeff O. Adhesion between 42. Young RW. Visual cells and the concept of renewal. Invest Ophthalmol
retina and pigment epithelium. Arch Ophthalmol 1971;86:679–84. 1976;15:700–25.
6. De Guillebon H, Zauberman H. Experimental retinal detachment: biophysi- 43. Anderson DH, Guerin CJ, Erickson PA, et al. Morphological recovery in the
cal aspects of retinal peeling and stretching. Arch Ophthalmol 1972;87: reattached retina. Invest Ophthalmol Vis Sci 1986;227:168–83.
545–8. 44. Kroll AJ, Machemer R. Experimental retinal detachment in the owl monkey.
7. Endo EG, Yao XY, Marmor MF. Pigment adherence as a measure of retinal V. Electron microscopy of the reattached retina. Am J Ophthalmol 1969;67:
adhesion: dependence on temperature. Invest Ophthalmol Vis Sci 1988;29: 117–30.
1390–6. 45. Yoon YH, Marmor MF. Rapid enhancement of retinal adhesion by laser pho-
8. Kita M, Negi A, Kawano SI, et al. Measurement of retinal adhesive force in tocoagulation. Ophthalmology 1988;95:1385–8.
the in vivo rabbit eye. Invest Ophthalmol Vis Sci 1990;31:624–8. 46. Gingell D, Fornes JA. Demonstration of intermolecular forces in cell adhesion
9. Kita M, Marmor MF. Retinal adhesive force in living rabbit, cat, and monkey using a new electrochemical technique. Nature 1975;256:210–1.
eyes: normative data and enhancement by mannitol and acetazolamide. 47. Adler AJ, Klucznik KM. Proteins and glycoproteins of the bovine interpho-
Invest Ophthalmol Vis Sci 1992;33:1879–82. toreceptor matrix: composition and fractionation. Exp Eye Res 1982;34:
10. Kita M, Marmor MF. Effects on retinal adhesive force in vivo of metabolically 423–34.
active agents in the subretinal space. Invest Ophthalmol Vis Sci 1992;33: 48. Hageman GS, Kirchoff MA, Anderson DH. Biochemical characterization and
1883–7. distribution of retinal interphotoreceptor matrix glycoconjugates. Glycoconj J
11. Marmor MF, Maack T. Local environmental factors and retinal adhesion in 1990;7:512.
the rabbit. Exp Eye Res 1982;34:727–33. 49. Porello K, LaVail MM. Histochemical demonstration of spatial heterogeneity
12. Yoon YH, Marmor MF. Effects on retinal adhesion of temperature, cyclic in the interphotoreceptor matrix of the rat retina. Invest Ophthalmol Vis Sci
AMP, cytochalasin B, and enzymes. Invest Ophthalmol Vis Sci 1988;29: 1986;27:1577–86.
910–4. 50. Berman ER. Mucopolysaccharides (glycosaminoglycans) of the retina: identi-
13. Marmor MF, Yao XY. The enhancement of retinal adhesiveness by ouabain fication, distribution, and possible biological role. Mod Probl Ophthalmol
appears to involve cellular edema. Invest Ophthalmol Vis Sci 1989;30: 1969;8:5–31.
1511–4. 51. Hollyfield JG, Varner H, Rayborn ME, et al. Retinal attachment to the pigment
14. Maurice DM, Salmon J, Zauberman H. Subretinal pressure and retinal epithelium. Retina 1989;9:59–68.
adhesion. Exp Eye Res 1971;12:212–7. 52. Johnson LV, Hageman GS, Blanks MC. Interphotoreceptor matrix domains
15. Yao XY, Hageman GS, Marmor MF. Retinal adhesiveness in the monkey. ensheath vertebrate cone photoreceptor cells. Invest Ophthalmol Vis Sci 1986;
Invest Ophthalmol Vis Sci 1994;35:744–8. 27:129–35.
16. Yao XY, Endo EG, Marmor MF. Reversibility of retinal adhesion in the rabbit. 53. Hageman GS, Marmor MF, Yao XY, et al. The interphotoreceptor matrix medi-
Invest Ophthalmol Vis Sci 1989;30:220–4. ates primate retinal adhesion. Arch Ophthalmol 1995;113:655–60.
17. Marmor MF, Yao XY, Hageman GS. Retinal adhesiveness in surgically enucle- 54. Hageman GS, Johnson LV. Structure, composition, and function of the retinal
ated human eyes. Retina 1994;14:181–6. interphotoreceptor matrix. In: Osborne N, Chader J, editors. Progress in
18. Kita M, Negi A, Marmor MF. Lowering the calcium concentration in the retinal research. Oxford: Pergamon Press; 1991.
subretinal space in vivo loosens retinal adhesion. Invest Ophthalmol Vis Sci 55. Hageman GS, Johnson LV. The “cone matrix sheath”: structural, compo-
1992;33:23–9. sitional, and functional analyses. Invest Ophthalmol Vis Sci 1988;29(suppl):
19. Bill A. Some aspects of tissue fluid dynamics in the eye. In: Cant JS, editor. 108.
Vision and circulation. St Louis: Mosby; 1976. 56. Kirchoff MA, Anderson K, Johnson LF, et al. Composition and distribution of
20. Toris CB, Pederson JE, Tsuboi S, et al. Extravascular albumin concentration insoluble interphotoreceptor matrix components. Invest Ophthalmol Vis Sci
of the uvea. Invest Ophthalmol Vis Sci 1990;31:43–53. 1990;31(suppl):153.
21. Maurice DM. Flow of water between aqueous and vitreous compartments in 57. Marmor MF. Mechanisms of retinal adhesion. In: Osborne NN, Chader GJ,
the rabbit eye. Am J Physiol 1987;252:104–8. editors. Progress in retinal research. Oxford: Pergamon Press; 1993.
22. Fatt I, Shantinath K. Flow conductivity of retina and its role in retinal 58. Kuehn MH, Wietecki DT, Hageman GS. Molecular characterization of the
adhesion. Exp Eye Res 1971;12:218–26. murine orthologue of the human retinal proteoglycan IPM 150. Mol Vis
23. Tsuboi S. Measurement of the volume flow and hydraulic conductivity across 2000;6:148–56.
the isolated dog retinal pigment epithelium. Invest Ophthalmol Vis Sci 59. Uebara F, Ohba N, Ozawa M. Isolation and characterization of galectins in
1987;28:1776–82. the mammalian retina. Invest Ophthalmol Vis Sci 2001;42:2164–72.
24. Marmor MF. Retinal detachment from hyperosmotic intravitreal injection. 60. Edelman GM. Cell adhesion molecules. Science 1983;219:450–7.
Invest Ophthalmol Vis Sci 1979;18:1237–44. 61. Adler AJ, Klucznik KM. Interaction of bovine pigment epithelium cells, pho-
25. Marmor MF, Martin LJ, Tharpe S. Osmotically induced retinal detachment in toreceptor outer segments, and interphotoreceptor matrix: a model for retinal
the rabbit and primate: electron microscopy of the retinal pigment epithelium. adhesion. Curr Eye Res 1982;1:579–89.
Invest Ophthalmol Vis Sci 1980;19:1016–29. 62. Kohno T, Sorgente N, Ishibashi T, et al. Immunofluorescent studies of fibro-
26. Cantrill HL, Pederson JE. Experimental retinal detachment. VI. The permea- nectin and laminin in the human eye. Invest Ophthalmol Vis Sci 1987;
bility of the blood–retinal barrier. Arch Ophthalmol 1984;102:747–51. 28:506–14.
27. Frambach DA, Marmor MF. The rate and route of fluid resorption from 63. Philp NJ, Nachmias VT. Polarized distribution of integrins and fibronectin in
the subretinal space of the rabbit. Invest Ophthalmol Vis Sci 1982;22: retinal pigment epithelium. Invest Ophthalmol Vis Sci 1987;28: 1275–80.
292–302. 64. Shirakawa H, Ishiguro SI, Itoh Y, et al. Are sugars involved in the binding of
28. Negi A, Marmor MF. Quantitative estimation of metabolic transport of sub- rhodopsin-membranes by the retinal pigment epithelium? Invest Ophthalmol
retinal fluid. Invest Ophthalmol Vis Sci 1986;27:1564–8. Vis Sci 1987;28:628–32.
29. Pederson JE, Cantrill HL. Experimental retinal detachment. V. Fluid move- 65. Opas M, Kalnins VI. Distribution of spectrin and lectin-binding materials in
ment through the retinal hole. Arch Ophthalmol 1984;102:136–9. surface lamina of RPE cells. Invest Ophthalmol Vis Sci 1985;26: 621–7.
30. Hamann S, Kiilgaard JF, la Cour M, et al. Cotransport of H+, lactate, and 66. Lazarus HS, Hageman GS. Xyloside-induced disruption of interphotoreceptor
H2O in porcine retinal pigment epithelial cells. Exp Eye Res 2003;76: matrix proteoglycans results in retinal detachment. Invest Ophthalmol Vis Sci
493–504. 1992;33:364–76.
31. Chihara E, Nao-IN. Resorption of subretinal fluid by transepithelial flow 67. Ishikawa M, Johnson LV, DeWing MD, et al. pH-Dependent changes in inter-
of the retinal pigment epithelium. Graefes Arch Ophthalmol 1985;223: photoreceptor matrix domains surrounding cone photoreceptors. Ophthalmol
202–4. Res 1996;28:117–24.
68. Ishikawa M, Fujiwara T, Yoshitomi T. Temperature-dependent ultrastructural 102. Takeuchi A, Kricorian G, Marmor MF. Albumin movement out of the subreti-
changes in the cone interphotoreceptor matrix. Jpn J Ophthalmol 2009;53: nal space after experimental retinal detachment. Invest Ophthalmol Vis Sci
536–40. 1995;36:1298–305.
69. Yao XY, Hageman GS, Marmor MF. Retinal adhesiveness is weakened 103. Takeuchi A, Kricorian G, Yao XY, et al. The rate and source of albumin entry 463
by enzymatic modification of the interphotoreceptor matrix in vivo. Invest into saline-filled experimental retinal detachments. Invest Ophthalmol Vis Sci
Ophthalmol Vis Sci 1990;31:2051–8. 1994;35:3792–8.
70. Yamada K. The effect of digestion with chondroitinases upon certain 104. Wolfensberger TJ, Gregor ZJ. Macular edema – rationale for therapy. In:

Chapter 19
histochemical reactions of mucosaccharide-containing tissues. J Histochem Coscas G editor. Macular edema, vol 47. Basel: Karger; 2010. p. 49–58.
Cytochem 1974;22:266. 105. Szurman P, Roters S, Grisanti S, et al. Ultrastructural changes after artificial
71. Uehara F, Muramatsu T, Sameshima M, et al. Effects of neuraminidase on retinal detachment with modified retinal adhesion. Invest Ophthalmol Vis Sci
lectin binding sites in photoreceptor cells of monkey retina. Jpn J Ophthalmol 2006;47:4983–9.
1985;29:54. 106. Imamura Y, Fujiwara T, Margolis R, et al. Enhanced depth imaging optical
72. Yao XY, Hageman GS, Marmor MF. Recovery of retinal adhesion after enzy- coherence tomography of the choroid in central serous chorioretinopathy.
matic perturbation of the interphotoreceptor matrix. Invest Ophthalmol Vis Retina 2009;29:1469–73.
Sci 1992;33:498–503. 107. Marmor MF. Hypothesis concerning carbonic anhydrase treatment of
73. Uehara F, Yasumura D, LaVail MM. Rod- and cone-associated interphoto- CME: example with epiretinal membrane. Arch Ophthalmol 1990;108:
receptor matrix in the rat retina. Invest Ophthalmol Vis Sci 1991;32: 1524–5.

Mechanisms of Normal Retinal Adhesion


285–92. 108. Cox SN, Hay E, Bird AC. Treatment of chronic macular edema with acetazol-
74. Owczarek FR, Marak GE, Pilkerton AR. Retinal adhesion in light- and dark- amide. Arch Ophthalmol 1988;106:1190–5.
adapted rabbits. Invest Ophthalmol 1975;14:353–8. 109. Fishman GA, Gilbert COT, Fiscella RG, et al. Acetazolamide for treatment
75. Faude F, Wendt S, Biedermann B, et al. Facilitation of artificial retinal detach- of chronic macular edema in retinitis pigmentosa. Arch Ophthalmol
ment for macular translocation surgery tested in rabbits. Invest Ophthalmol 1989;107:1445–52.
Vis Sci 2001;42:1328–37. 110. Apushkin MA, Fishman GA. Use of dorzolamide for patients with X-linked
76. Burnside MB. Possible roles of microtubules and actin filaments in retinal retinochisis. Retina 2006;26:741–5.
pigmented epithelium. Exp Eye Res 1976;23:257–75. 111. Genead MA, Fishman GA, McAnany JJ. Efficacy of topical dorzolamide for
77. Burnside B, Laties AM. Actin filaments in apical projections of the primate treatment of cystic macular lesions in a patient with enhanced S-cone syn-
pigmented epithelial cell. Invest Ophthalmol 1976;15:570–5. drome. Doc Ophthalmol 2010;121:231–40.
78. Burnside B, Adler R, O’Connor P. Retinomotor pigment migration in the 112. Wolfensberger TJ, Chiang R, Takeuchi A, et al. Inhibition of membrane-bound
teleost retinal pigment epithelium. I. Roles for actin and microtubules in carbonic anhydrase enhances subretinal fluid absorption and retinal
pigment granule transport and cone movement. Invest Ophthalmol Vis Sci adhesiveness. Graefes Arch Clin Exp Ophthalmol 2000;238:76–80.
1983;24:1–15. 113. Fishman GA, Apushkin MA. Continued use of dorzolamide for the treatment
79. Moore KT, Yao XY, Marmor MF. Intravitreal cytochalasin D decreases retinal of cystoid macular oedema in patients with retinitis pigmentosa. Br J
adhesion. Invest Ophthalmol Vis Sci 1991;32(suppl):772. Ophthalmol 2007;91:743–5.
80. Immel J, Negi A, Marmor MF. Acute changes in RPE apical morphology after 114. Genead MA, Fishman GA. Efficacy of sustained topical dorzolamide therapy
retinal detachment in rabbit: a SEM study. Invest Ophthalmol Vis Sci for cystic macular lesions in patients with retinitis pigmentosa and usher
1986;27:1770–6. syndrome. Arch Ophthalmol 2010;128:1146–50.
81. Dearry A, Burnside B. Effects of extracellular Ca++, K+, and Na+ on cone and 115. Apushkin MA, Fishman GA, Grover S, et al. Rebound of cystoid macular
retinal pigment epithelium retinomotor movements in isolated teleost retinas. edema with continued use of acetazolamide in patients with retinitis pigmen-
J Gen Physiol 1984;83:589–611. tosa. Retina 2007;27:1112–8.
82. Kim RY, Yao XY, Marmor MF. Oxygen dependency of retinal adhesion. 116. Thobani A, Fishman GA. The use of carbonic anhydrase inhibitors in the
Invest Ophthalmol Vis Sci 1993;34:2074–8. retreatment of cystic macular lesions in retinitis pigmentosa and X-linked
83. Marmor MF, Yao XY. The metabolic dependency of retinal adhesion in rabbit retinoschisis. Retina 2011;31:312–5.
and primate. Arch Ophthalmol 1995;113:232–8. 117. Fang XY, Hayashi A, Cekic O, et al. Effect of Ca2+-free and Mg2+-free
84. Kain HL, Libondi T. Experimentelle Netzhautabldaung-Untersuchungen zum BSS plus solution on the retinal pigment epithelium and retina in rabbits.
Pathomechanismus. Fortschr Ophthalmol 1986;83:590–6. Am J Ophthalmol 2001;131:481–8.
85. Adler AJ, Martin KJ. Lysosomal enzymes in the interphotoreceptor matrix: 118. Chignell AH, Markham RHC. Retinal detachment surgery without cryother-
acid protease. Curr Eye Res 1983;2:359–66. apy. Br J Ophthalmol 1981;65:371–3.
86. Ashburn FS Jr, Pilkerton A, Rao NA, et al. The effects of iodate and iodoacetate 119. Machemer R. The importance of fluid absorption, traction, intraocular
on the retinal adhesion. Invest Ophthalmol Vis Sci 1980;19:1427–32. currents, and chorioretinal scars in the therapy of rhegmatogenous retinal
87. Negi A, White MP, Marmor MF. Effects of hemicholinium-3, a photoreceptor detachments. Am J Ophthalmol 1984;98:681–93.
and pigment epithelial toxin, on retinal adhesiveness and subretinal fluid 120. Zauberman H, Rosell FG. Treatment of retinal detachment without inducing
absorption. Doc Ophthalmol 1993;83:331–6. chorioretinal lesions. Trans Am Acad Ophthalmol Otolaryngol 1975;79:
88. Takeuchi A, Negi A, Yamamoto F, et al. Effects of sodium iodate on retinal 835–44.
adhesive force in vivo. Invest Ophthalmol Vis Sci 1991;32(suppl):667. 121. Kita M, Negi A, Kawano SI, et al. Photothermal, cryogenic, and diathermic
89. Yoon YH, Marmor MF. Effects of hemicholinium-3 and sodium iodate effects on retinal adhesive force in vivo. Retina 1991;11:441–4.
on RPE and retinal adhesiveness. Invest Ophthalmol Vis Sci 1991;32(suppl): 122. Kwon OW, Kim SY. Changes in adhesive force between the retina and the
667. retinal pigment epithelium by laser photocoagulation in rabbits. Yonsei Med
90. Nilsson SEG, Knave B, Persson HE. Changes in ultrastructure and function J 1995;36:243–50.
of the sheep pigment epithelium and retina induced by sodium iodate. II. 123. Zhu ZR, Goodnight R, Sorgente N, et al. Cellular proliferation induced by
Early effects. Acta Ophthalmol 1977;55:1007–26. subretinal injection of vitreous in the rabbit. Arch Ophthalmol 1988;106:
91. Marmor MF, Maack T. Enhancement of retinal adhesion and subretinal 406–11.
fluid resorption by acetazolamide. Invest Ophthalmol Vis Sci 1982;23: 124. Takeuchi A, Kricorian G, Marmor MF. When vitreous enters the sub­
121–4. retinal space: implications for subretinal fluid protein. Retina 1996;16:
92. Hughes BA, Miller SS, Machen TE. Effects of cyclic AMP on fluid absorption 426–30.
and ion transport across frog retinal pigment epithelium: measurements in 125. Negi A, Marmor MF. The resorption of subretinal fluid after diffuse damage
the open-circuit state. J Gen Physiol 1984;83:875–99. to the retinal pigment epithelium. Invest Ophthalmol Vis Sci 1983;24:
93. Marmor MF, Negi A. Pharmacologic modification of subretinal fluid 1475–9.
absorption in the rabbit eye. Arch Ophthalmol 1986;104:1674–7. 126. Negi A, Marmor MF. Experimental serous retinal detachment and focal
94. Miller S, Farber D. Cyclic AMP modulation of ion transport across frog retinal pigment epithelial damage. Arch Ophthalmol 1984;102:445–9.
pigment epithelium: measurements in the short-circuit state. J Gen Physiol 127. Tsukahara Y, Marmor MF. Experimental studies on the accumulation of
1984;83:853–74. subretinal fluid. Ophthalmologica 1991;202:202–7.
95. Kawano SI, Marmor MF. Metabolic influences on the absorption of serous 128. Marmor MF. On the cause of serous detachments and acute central serous
subretinal fluid. Invest Ophthalmol Vis Sci 1988;29:1255–7. chorioretinopathy. Br J Ophthalmol 1997;81:812–3.
96. Frambach DA, Misfeldt DS. Furosemide-sensitive Cl transport in embryonic 129. Marmor MF, Yao XY. Conditions necessary for the formation of serous detach-
chicken retinal pigment epithelium. Am J Physiol 1983;244:679–85. ment: Experimental evidence from the cat. Arch Ophthalmol 1994;112:
97. Tsuboi S, Pederson JE. Experimental retinal detachment. XI. Furosemide- 830–8.
inhibitable fluid absorption across retinal pigment epithelium in vivo. 130. Kita M, Negi A, Kawano S, et al. Lamellar scleral resection enhances subretinal
Arch Ophthalmol 1986;104:602–3. fluid absorption in eyes with choroidal congestion. Jpn J Ophthalmol 1991;
98. Frambach DA, Weiter JJ, Adler AJ. A photogrammetric method to measure 35:394–401.
fluid movement across isolated frog retinal pigment epithelium. Biophys J 131. Yao XY, Marmor MF. Induction of serous retinal detachment in rabbit eyes
1985;47:547–52. by pigment epithelial and choriocapillary injury. Arch Ophthalmol 1992;
99. Negi A, Marmor MF. Mechanisms of subretinal fluid resorption in the cat eye. 110:541–6.
Invest Ophthalmol 132. Chon CH, Yao XY, Dalal R, et al. An experimental model of retinal pigment
100. Marmor MF, Negi A, Maurice DM. Kinetics of macromolecules injected into epithelial and serous detachment. Retina 1996;16:139–44.
the subretinal space. Exp Eye Res 1985;40:687–96. 133. De Venecia G, Jampol LM. The eye in accelerated hypertension. II. Localized
101. Negi A, Marmor MF. Healing of photocoagulation lesions affects the rate of serous detachments of the retina in patients. Arch Ophthalmol 1984;102:
subretinal fluid resorption. Ophthalmology 1984;91:1678–83. 68–73.
134. Gelber GS, Schatz H. Loss of vision due to central serous chorioretinopathy 138. Wakakura M, Ishikawa S. Central serous chorioretinopathy complicating
following psychological stress. Am J Psychiatry 1987;144:46–50. systemic corticosteroid treatment. Br J Ophthalmol 1984;68:329–31.
135. Yannuzzi LA. Type A behavior and central serous chorioretinopathy. 139. Yoshioka H, Katsume Y, Akune H. Experimental central serous chorioreti-
464 Retina 1987;7:111–30. nopathy in monkey eyes: fluorescein angiographic findings. Ophthalmologica
136. Fastenberg DM, Ober RR. Central serous choroidopathy in pregnancy. 1982;185:168–78.
Arch Ophthalmol 1983;101:1055–8.
137. Gass JDM, Little H. Bilateral bullous exudative retinal detachment complicat-
ing idiopathic central serous chorioretinopathy during systemic corticosteroid
Section 1

therapy. Ophthalmology 1995;102:737–47.


Anatomy and Physiology
Basic Science and Translation to Therapy
Anatomy and Physiology Section 1
For additional online content visit http://www.expertconsult.com

Structure, Function, and Pathology of Chapter

Bruch’s Membrane
Christine A. Curcio, Mark Johnson 20 
INTRODUCTION, HISTORY, EMBRYOLOGY the choroidal vasculature, and Bruch’s as part of it, depends on
differentiated RPE and its production of inductive signals,
Bruch’s membrane is a thin (2–4  µm), acellular, five-layered including basic fibroblast growth factor and vascular endothelial
extracellular matrix located between the retina and choroid.1,2 growth factor (VEGF).13
It extends anteriorly to the ora serrata, interrupted only by the
optic nerve. Tissue resembling Bruch’s membrane is visible STRUCTURE OF BRUCH’S MEMBRANE IN
anterior to the ora serrata extending forward to the pigmented
epithelium of the ciliary body. Bruch’s membrane lies between
THE YOUNG ADULT EYE
the metabolically active retinal pigment epithelium (RPE) and Hogan’s five-layer nomenclature for Bruch’s membrane14 is com-
a capillary bed (choriocapillaris) and thus serves two major func- monly used. Gass proposed a three-layer system that did not
tions as the substratum of the RPE and a vessel wall. It has include the cellular basal laminas as part of Bruch’s proper.15
major clinical significance because of its involvement in age- These layers are shown in Fig. 20.1 and their constituents are
related macular degeneration (AMD) and other chorioretinal given in Table 20.1.
diseases.
RPE basal lamina (RPE-BL)
Early history This ∼0.15-µm-thick layer is a meshwork of fine fibers like other
basal laminas in the body.16,17 The RPE-BL resembles that of the
Carl Ludwig Wilhelm Bruch first isolated the “lamina vitrea”
choriocapillaris endothelium but does not contain collagen VI.
that we now know as Bruch’s membrane, and described it in his
The RPE-BL contains collagen IV α3–5,18 like that of kidney
1844 doctoral thesis,3,4 where he also first described the tapetum
glomerulus, another organ with specialized filtration and trans-
found in many mammals. By light microscopy, Bruch’s mem-
port functions. The RPE synthesizes specific laminins that pre­
brane appeared transparent with little internal structure. Later
ferentially adhere Bruch’s membrane to the RPE through
studies by Smirnow5 divided this membrane into an outer elastic
interaction with integrins.19
layer (first described by Sattler in 1877) and an inner cuticular
layer, separated by a dense plexus of very fine elastic fibers.6,7 Inner collagenous layer (ICL)
The ICL is ∼1.4  µm thick and contains 70-nm-diameter fibers
Development of Bruch’s membrane of collagens I, III, and V in a multilayered criss-cross, parallel
The bipartite character of Bruch’s membrane arises from the to the plane of Bruch’s membrane.1 The collagen grid is asso-
embryology of its tissue. When the optic cup invaginates and ciated with interacting molecules, particularly the negatively
folds, its inner layer forms the neural retina, and its outer layer charged proteoglycans chondroitin sulfate and dermatan
forms the RPE. The RPE lies in contact with mesenchyme. At this sulfate.20,21
apposition, Bruch’s membrane forms by 6–7 weeks’ gestation.
Thus, its inner layer is composed of ectodermal tissue and its Elastic layer (EL)
outer layer is composed of mesodermal tissue. At the border of The EL consists of stacked layers of linear elastin fibers, criss-
two layers, the elastic layer forms last, becoming histologically crossing to form a 0.8-µm-thick sheet with interfibrillary spaces
visible by 11–12 weeks.8–10 of ∼1 µm. This sheet extends from the edge of the optic nerve to
The collagen that fills the extracellular space and the later- the ciliary body pars plana.1 In addition to elastin fibers, the EL
appearing elastin appear to be made by invading fibroblasts and contains collagen VI, fibronectin, and other proteins, and colla-
the filopodia of endothelial cells lining the adjacent choriocapil- gen fibers from the ICL and outer collagenous layer (OCL) can
laris. The two basal laminas are produced by their associated cell cross the EL. Some EL elastin fibers are said to cross the tissue
layers.11 In addition to collagen IV subunits specific to special- space between the choriocapillaris and join bundles of choroidal
ized basal lamina, RPE expresses genes for structural collagen elastic tissue.22 The EL confers biomechanical properties, vascu-
III and angiostatic collagen XVIII in a developmentally regulated lar compliance, and antiangiogenic barrier functions. It is more
manner linked to photoreceptor maturation.12 discontinuous in the macula, perhaps explaining why choroidal
By week 13, fenestrations are apparent in the endothelium neovascularization (CNV) is more prominent there.23 This
facing Bruch’s membrane,10 indicating that, at this stage, trans- concept is supported by the extensive laser-induced neovascu-
port across this tissue may be functional. Choroidal endothelial larization in mice deficient in lysyl oxidase-like 1, an enzyme
cells originate from paraocular mesenchyme. Development of required for elastin polymerization.24
466
L
Section 1

ICL
Anatomy and Physiology
Basic Science and Translation to Therapy

OCL

A 17 y
B 46 y
C 65 y

Fig. 20.1 Macular Bruch’s membrane throughout the lifespan. Retinal pigment epithelium (RPE) is at the top of all panels. RPE basal lamina
(arrowheads) and elastic layer (EL, yellow arrows, discontinuous in macula) are shown. (A) 17 years: electron-dense amorphous debris and
lipoproteins are absent. ICL, inner collagenous layer; OCL, outer collagenous layer. Bar = 1 µm. (B) 46 years: electron-dense amorphous debris
and lipoproteins are present. Coated membrane-bound bodies (green arrow) contain lipoproteins. L, lipofuscin. (C) 65 years: electron-dense
amorphous debris and lipoproteins are abundant. Membranous debris, also called lipoprotein-derived debris (red arrow), has electron-dense
exteriors within basal laminar deposit (*).Within OCL, banded material is type VI collagen, often found in basal laminar deposit.

Table 20.1 Structural and molecular components of Bruch’s membrane

Layer (common abbreviation) Component; age change References

Basal laminar deposit (BlamD) + Fibronectin, laminin, IV α4–5, VI, endostatin, EFEMP1 164, 167, 206–209

RPE basal lamina (RPE-BL) IV α1–5, V, laminins 1, 5, 10, and 11, nidogen-1, heparan sulfate, 18, 19, 21, 66, 210,
chondroitin sulfate 211

Lipid wall/basal linear deposit + Lipoproteins 38, 39, 212


(BlinD)

Inner collagenous layer (ICL) I, III, V, fibronectin, chondroitin sulfate, dermatan sulfate, lipoproteins ↑, 34, 35, 38, 39, 50, 66,
apoE, heme, clusterin, vitronectin 146, 152, 210, 213–215

Elastic layer (EL) Elastin ↑, calcium phosphate ↑ 14, 66–68, 210, 216

Outer collagenous layer I, III, V, fibulin-5, fibronectin, chondroitin sulfate, dermatan sulfate, 21, 39, 50, 152, 210,
(OCL) lipoproteins ↑, apoE, clusterin 215, 217

ChC-basal lamina IV α1, 2, V, VI, laminin, heparan sulfate, chondroitin sulfate, endostatin 18, 208, 210, 211, 218

Bruch’s, throughout or layer I ↑, collagen solubility ↓, perlecan, MMP-2 ↑, MMP-9 ↑, TIMP-2; TIMP-3 62, 66, 138, 139, 147,
not specified ↑, pentosidine ↑, CML ↑, GA-AGE ↑, RGR-d, apoB, oxidized apoB-100, 218–230
7-KCh, LHP, HHE ↑, DHP-lys ↑, C3d ↑, C5b-9 ↑, pentraxin-3 ↑,
thrombospondin-1, zinc
Table shows definitely localized components. Most determinations were made in macula. Studies showing histochemical/immunohistochemical verification of
biochemistry and ultrastructural validation of structures identified by light microscopy techniques were given greater weight. Localizations were assigned to specific
layers if immunogold-electron microscopy or high-magnification confocal microscopy images were available. Roman numerals denote collagens. Components are
ordered within each layer: structural components, lipoproteins, extracellular matrix and its regulation, modified lipids and proteins, complement/immunity, cellular
response/activity, metals. Known changes with advancing age are bold with an arrow indicating direction of change. New additions with age are shown with a plus
(+). Plain text means no change or not tested.
CML, carboxymethyl-lysine226; 7-KCh, 7 keto-cholesterol229; GA-AGE, glycolaldehyde-derived advanced glycation end products221; HHE, 4-hydroxyhexenal66,218;
DHP-lys, dihydropyridine lysine.66
Fig. 20.2 Surface of the endothelium of the
choriocapillaris showing fenestrations with a
bicycle-spoke pattern (yellow arrow) and
presumed artifactual openings arising from 467
tissue preparation (cyan arrow); quick-freeze/
deep-etch, 64-year-old eye, macula. Bar =
100 nm. (Reproduced with permission from

Chapter 20
Johnson M, Huang J-D, Presley JB, et al.
Comparison of morphology of human
macular and peripheral Bruch’s membrane in
older eyes. Curr Eye Res 2007;32:791–9.)

Structure, Function, and Pathology of Bruch’s Membrane


Outer collagenous layer Lipid accumulation: Bruch’s
The OCL contains many of the same molecular components membrane lipoproteins
as the ICL, and the collagen fibrils running parallel to the Early electron microscopists described aged Bruch’s membrane
choriocapillaris additionally form prominent bundles. This as being filled with debris, including amorphous electron-dense
layer, unlike the ICL, has periodic outward extensions between material, membrane fragments, vesicles, and calcification.1,25
individual choriocapillary lumens called intercapillary pillars, Debris deposition in ICL and OCL begins in the second decade
where thickness cannot be determined due to the lack of a in the macula and is delayed in equatorial regions, a regional lag
boundary. Between pillars, OCL thickness can range from 1 also reported for individual components.30 Identifying this mat­
to 5  µm.25 erial has been a fruitful approach to understanding antecedents
of disease.
Most prominent among the changes in Bruch’s membrane is
Choriocapillaris basal lamina (ChC-BL) a profound accumulation of lipids. Clinical observations on
This 0.07-µm-thick layer is discontinuous with respect to fluid-filled RPE detachments in older adults led to Bird and
Bruch’s membrane due to the interruptions of the intercapil- Marshall’s hypothesis that a lipophilic barrier in Bruch’s blocked
lary pillars of the choroid. It is continuous with respect to a normal, outwardly directed fluid efflux from the RPE31 (as
the complex network of spaces defined by the choriocapillary opposed to leakage from CNV). This hypothesis motivated a
lumens because the basal lamina envelops the complete cir- seminal histochemical study by Pauleikhoff et al.32 that demon-
cumference of the endothelium. A remarkable structural strated oil red O-binding material (esterified cholesterol (EC),
feature of the adjacent choriocapillary endothelium is fenestra- triglyceride (TG), fatty acid) localized exclusively to Bruch’s
tions that are permeable to macromolecules (Fig. 20.2).26 This membrane, unlike other stains. This lipid was absent <30 years,
basal lamina may inhibit endothelial cell migration into variably present at 31–60 years, and abundant at ≥61 years.33,34
Bruch’s membrane, as do basal laminas associated with retinal A specific fluorescent marker, filipin, which binds the 3β-hydroxy
capillaries.27 group of sterols to reveal unesterified (free) cholesterol (UC) or
EC depending on tissue pretreatment,35 indicated that EC is a
prominent component of the oil red O-binding deposition.35
BRUCH’S MEMBRANE IN AN AGED EYE Macular EC rose linearly from near zero at age 22 years to reach
Aging is the largest risk factor for developing AMD,28 and high and variable levels in aged donors. EC was detectable in
Bruch’s membrane undergoes significant age-related changes. periphery at ∼1/7 macular levels and increased significantly
Identification of factors predisposing to disease progression is a with age. Hot-stage polarizing microscopy34 similarly demon-
priority. This task has been challenged by difficulty imposed by strated prominent age-related increases in EC in Bruch’s mem-
the thinness of the tissue, and the closely integrated functions of brane, manifest as liquid crystals (“Maltese crosses”) when
RPE, Bruch’s, and choriocapillaris. Current opinion holds that examined through a polarizing filter. Few birefringent crystals
RPE and Bruch’s membrane age in concert, and normal Bruch’s signifying the neutral lipid TG were found.
membrane aging transforms insidiously into AMD pathol- Histochemical, ultrastructural, biochemical, gene expression,
ogy.1,16,17,29 This section covers aging, to inform the following and cell biological evidence now indicate that the EC-rich
section on function. material accumulating with age in Bruch’s membrane is a
lipoprotein-containing apolipoprotein B, assembled by the inherited disorder that includes a pigmentary retinopathy.43,44
RPE.36 This process, ongoing throughout life yet first revealed by The combination of apoB and MTP within native RPE marks
468 aging, has implications for the formation of AMD-specific these cells as constitutive lipoprotein secretors.45 Secretion of
lesions, intraocular transport, RPE physiology, nutrition of outer full-length apoB has been demonstrated in rat-derived and
retina, and maintenance of photoreceptor health. In 1926, Ver- human-derived RPE cell lines.46,47 Consistent with an RPE
hoeff and Sisson speculated that lipid deposition might precede origin, particles first appear in the elastic layer of Bruch’s mem-
Section 1

Bruch’s membrane basophilia and fragmentation, common in brane and fill in towards the RPE.39
older eyes due to “lime salts [calcification] in the elastic layer.”37 Indirect evidence that Bruch’s membrane lipoproteins are of
Ultrastructural studies described in Bruch’s membrane of intraocular origin also emerges from the epidemiologic litera-
older eyes36 numerous small (<100 nm), round, electron-lucent ture. If the EC deposition in Bruch’s membrane and AMD-
vesicular profiles, implying aqueous interiors. Lipid-preserving associated lesions were a manifestation of systemic perifibrous
preparation techniques together with extraction studies show lipid and atherosclerosis, then a strong positive correlation
Anatomy and Physiology
Basic Science and Translation to Therapy

that these so-called vesicles are actually solid, lipid-containing between disease status and plasma lipoprotein levels, like that
particles, now considered lipoproteins (Fig. 20.3B). These documented for coronary artery disease,48 might be expected but
methods include postfixation in osmium paraphenylenediamine has not emerged.49
(OTAP)35 and, most strikingly, quick-freeze/deep-etch (QFDE), Identifying the upstream sources of Bruch’s membrane lipo-
a freeze fracture method with an etching step to remove frozen protein constituents is essential for understanding the biological
water.38–40 Particles vary in size from 60 to 100 nm but could be purpose of this pathway and the prospects for eventual clinical
as large as 300 nm, occasionally appearing to coalesce (Fig. 20.3). exploitation. Studies using isolated lipoproteins from Bruch’s
Lipoprotein particles are first seen among fibrils of the elastic membrane50 and Bruch’s membrane choroid EC51 report a high
layer in early adulthood, extending inward ultimately to fill mole percentage of linoleate (>40%) and low docosahexaenoate
most of the open space of the ICL by the seventh decade of life.40 (<1%) for all lipid classes.52 This composition strongly points
Most fatefully, a new layer, the lipid wall,38 then forms with solid away from photoreceptor outer segments (35% docosahexaeno-
particles stacked 3–4 deep occupying nearly 100% of a space ate in membrane phospholipids) as an upstream source, as long
between RPE basal lamina and OCL of many older eyes. The postulated,53,54 and towards plasma lipoproteins (45–55% linole-
lipid wall displaces ICL collagen fibrils that anchor the RPE basal ate in all lipid classes). These data have been interpreted to
lamina (Fig. 20.3). It is considered a precursor to basal linear signify that plasma lipoproteins are major contributors upstream
deposits, a specific lesion of AMD (see below). to an apoB lipoprotein of RPE origin. In contrast, the sources of
Lipoprotein composition can provide clues to sources of its UC in Bruch’s lipoproteins are not yet known and could be
components.41 When isolated (Fig. 20.4A), Bruch’s membrane outer segments, plasma lipoproteins, endogenous synthesis,
lipoproteins are found to be EC-enriched (EC/total cholesterol or a combination.
= 0.56; EC/TG = 4–11; Fig. 20.4B). For comparison, hepatic very- Lipoproteins may thus be assembled from several sources,
low-density lipoprotein (VLDL), of similar diameter, is TG-rich. including outer segments, remnant components from the photo-
An early report of TG-enriched Bruch’s membrane neutral receptor nutrient supply system, and endogenous synthesis.
lipid42 was not replicated. Abundant EC points to the only According to this model,52 plasma lipoproteins serve as vehicles
mechanism by which neutral lipids are released directly from for delivery of lipophilic nutrients (carotenoids,55 vitamin E, and
cells, an apoB-containing lipoprotein, like hepatic VLDL or cholesterol56) to photoreceptors by RPE, which has functional
intestinal chylomicrons. Significantly, RPE expresses the apoB receptors for low-density lipoprotein (LDL) and high-density
gene and protein, along with microsomal triglyceride transfer lipoprotein.57,58 Nutrients are stripped from these lipoproteins by
protein (MTP), required for apoB lipidation and secretion. Lack the RPE for delivery to the photoreceptors, and the remnants are
of functional MTP is the basis of abetalipoproteinemia, a rare repackaged for secretion into Bruch’s membrane as part of

L
RPE

BrM

A B

Fig. 20.3 Lipid wall, a layer of lipoproteins on the inner surface of Bruch’s membrane. (A) Lipoproteins (spherical vesicles of uniform diameter)
accumulate 3–4 deep between the retinal pigment epithelium (RPE), basal lamina (black arrowheads), and Bruch’s membrane (BrM), inner
collagenous layer (white arrowheads). Thin-section transmission electron micrograph following osmium postfixation. L, lipofuscin. Sectioning plane
is vertical; bar = 1 µm. (B) Quick-freeze/deep-etch shows tightly packed Bruch’s membrane lipoproteins in the lipid wall, and that lipoproteins have
classic core and surface morphology. Fracture plane is oblique; bar = 200 nm. (Reproduced with permission from Huang J-D, Presley JB, Chimento
MF, et al. Age-related changes in human macular Bruch’s membrane as seen by quick-freeze/deep-etch. Exp Eye Res 2007;85:202–18.)
469

Chapter 20
TG
A-I B-100

Structure, Function, and Pathology of Bruch’s Membrane


EC

UC
C-I
?

PL

C-II
E
A Apo
B

Fig. 20.4 Bruch’s membrane lipoprotein composition. (A) Lipoprotein particles isolated from Bruch’s membrane are large and spherical; negative
stain.153 (Source: Li C-M, Chung BH, Presley JB, et al. Lipoprotein-like particles and cholesteryl esters in human Bruch’s membrane: initial
characterization. Invest Ophthalmol Vis Sci 2005;46:2576–86). Bar = 50 nm. (B) Bruch’s membrane lipoprotein composition inferred from direct
assay,50,153 (Sources: Wang L, Li C-M, Rudolf M, et al. Lipoprotein particles of intra-ocular origin in human Bruch membrane: an unusual lipid
profile. Invest Ophthalmol Vis Sci 2009;50:870–7) (Li C-M, Chung BH, Presley JB, et al. Lipoprotein-like particles and cholesteryl esters in human
Bruch’s membrane: initial characterization. Invest Ophthalmol Vis Sci 2005;46:2576–86), druse composition, and retinal pigment epithelium gene
expression.139,154 (Sources: Malek G, Li C-M, Guidry C, et al. Apolipoprotein B in cholesterol-containing drusen and basal deposits in eyes with
age-related maculopathy. Am J Pathoi 2003;162:413–25) and (TG, Li C-M, Clark ME, Chimento MF, et al. Apolipoprotein localization in isolated
drusen and retinal apolipoprotein gene expression. Invest Ophthalmol Vis Sci 2006;47:3119–28) TG, triglyceride; EC, esterified cholesterol; UC,
unesterified cholesterol; PL, phospholipid; Apo, apolipoproteins. The question mark signifies that not all apolipoproteins are known.

apoB-containing lipoproteins, where they begin to accumulate The EL thickens with age but decreases relative to overall
during age and become toxically modified to instigate inflam- thickening of Bruch’s membrane.23 Thus elastin referenced to
mation in AMD. other Bruch’s constituents, as detected by Raman spectroscopy,
decreases with age.66 Similar arguments can be made for colla-
Other aging changes gen III and IV. A prominent age change,67 noted early,37 is calci-
Bruch’s membrane thickens throughout adulthood (20–100 fication and ensuing brittleness. This process involves fine
years) two- to threefold under the macula and becoming more deposition of electron-dense particulate matter,14 confirmed as
variable between individuals at older ages.25,59,60 Equatorial calcium phosphate68 on individual elastin fibrils.
Bruch’s membrane changes little while Bruch’s membrane near Long-lived proteins like collagens are modified in vivo by
the ora serrata increases twofold during this time.25 In the nonenzymatic Maillard and free radical reactions to yield
macula, the OCL thickens more prominently than the ICL.61 A advanced glycation end products (AGEs) and the formation of
large ultrastructural study of 121 human donor eyes demon- lipid-derived reactive carbonyl species like 4-hydroxyhexenal
strated that the macular EL is 3–6 times thinner than peripheral and linoleate hydroperoxide, collectively called age-related lipo-
EL23 at all ages. peroxidation end products (ALEs). Accumulation of AGEs and
Unbalanced regulation of extracellular matrix molecules and ALEs, characteristic of diabetes and atherosclerosis, also occurs
their modulator matrix are thought to result in Bruch’s mem- in aging Bruch’s membrane (Table 20.1). Finally, other compo-
brane thickening. Increased histochemical reactivity for glyco- nents more prominent in aged eyes include complement compo-
conjugates, glycosaminoglycans (GAGs), collagen, and elastin is nents C3d, C5b-9, and pentraxin-3, a homolog of the acute-phase
seen in the macula relative to equator and near the ora serrata.25 respondent C-reactive protein. Thus, at the molecular level,
Collagen solubility declines with age.62 Matrix metalloprotein- aging Bruch’s membrane contains evidence of many biological
ases MMP-2 and MMP-3 increase with age, as does a potent activities, including remodeling, oxidative damage, and inflam-
tissue inhibitor of metalloproteinases, TIMP-3. TIMP-3 immuno- mation, in addition to lipoprotein accumulation.
reactivity reaches adult levels at 30 years of age near vasculature
in lung, kidney, and in Bruch’s membrane, signifying the end of
developmental organogenesis.63 The reduction or absence of
FUNCTION OF BRUCH’S MEMBRANE
TIMP-3 is proangiogenic, as this protein not only regulates As a vessel wall of the choroid, Bruch’s membrane’s primary
metalloproteinases during the normal turnover of Bruch’s mem- function is structural, like other vessel walls. Its architecture is
brane matrix components, but it also binds to VEGF.64,65 similar to vascular intima, with a subendothelial extracellular
matrix and elastic layer corresponding to the internal elastic pressure (the osmotic pressure generated by plasma proteins).
lamina. The abluminal surface of Bruch’s differs from other This balance is embodied by Starling’s law that characterizes the
470 vessel walls in that it abuts a basal lamina, that of the RPE. The relationship between fluid flux (q = flow per unit area; positive
luminal surface faces a fenestrated vascular endothelium and when flow is out of the blood vessel) across a capillary vessel
basal lamina, making Bruch’s membrane structurally analogous wall and the forces driving this flow:
to the renal glomerulus and providing a basis for commonality
Section 1

between retinal and kidney disease.69–71 The importance of fluid q = Lp ( ∆P − σ∆Π)  (equation 1)
and macromolecular transport across the renal glomerulus is
Lp is hydraulic conductivity, which characterizes the ease with
well known.72 Transport is a second important function of
which fluids flow cross the vessel wall. If the surface area of the
Bruch’s membrane.
blood vessel is A, then 1/(Lp A) is the flow resistance of the vessel
wall. ΔP is the difference between the fluid pressure within the
Structural role of Bruch’s membrane
Anatomy and Physiology
Basic Science and Translation to Therapy

blood vessel (Pcc) and the pressure at the basal surface of the RPE
Bruch’s membrane encircles more than half the eye and stretches (PRPE). ΔΠ is the difference between the oncotic pressure within
with the corneoscleral envelope as intraocular pressure (IOP) the blood vessel (Πcc) and that at the basal surface of the RPE
increases. It therefore withstands this stretch and returns to its (ΠRPE). σ is the reflection coefficient that characterizes the extent
original shape when IOP decreases. This tissue also stretches to to which the vessel wall rejects the plasma protein species gen-
accommodate changes in choroidal blood volume. Finally, the erating ΔΠ. σ ranges from 0 for a freely permeable species to 1
choroid (and Bruch’s membrane with it) may act as a spring that when a species is completely rejected by the membrane.
pulls the lens during accommodation.73,74 For these reasons, then, We can estimate the magnitude of ΔP – σΔΠ using measured
Bruch’s membrane requires elasticity. Marshall and Hussain’s value of q and Lp. The fluid pumping rate by human RPE has
group estimated the modulus of elasticity in Bruch’s membrane been measured as q = 11 µL/h/cm2, similar to that in other
choroid preparations to be 7–19 MPa.75 These values are similar animals (Table 20.2). The hydraulic conductivity of macular
to those of sclera (although sclera is much thicker and thus can Bruch’s membrane/choroid of healthy young humans ranges
support more load), consistent with the notion that Bruch’s from 20 to 100 × 10−10 m/s/Pa.84 Then, using q = 11 µL/h/cm2
membrane contributes to load bearing. After early adulthood, and Lp = 50 × 10−10 m/s/Pa, we can calculate that the magnitude
the modulus of elasticity of human Bruch’s membrane–choroid of (ΔP − σΔΠ) necessary to drive this flow through Bruch’s mem-
complex increases (P < 0.001) at a rate of ∼1% per year. Bruch’s brane is roughly 0.05 mmHg. (This does not include the flow
membrane stiffness in AMD eyes does not differ from age- resistance of choriocapillaris endothelium, which is not mea-
matched normals.76 sured when Lp of a Bruch’s membrane/choroidal preparation is
determined. For this highly fenestrated endothelium, Lp can be
Transport role of Bruch’s membrane estimated as roughly 25 × 10−10 m/s/Pa,85 which does not affect
The choroid services the metabolic needs of the outer retina, our conclusions below.)
facilitated in part by fenestrated endothelium. Oxygen, electro- σ can be roughly estimated by assuming that the fluid in the
lytes, nutrients, and cytokines destined for the RPE and photo- suprachoroidal space is in equilibrium with blood in the choroid.
receptors pass from the choriocapillaris and through Bruch’s Using measurements of fluid pressure and of the plasma protein
membrane, and waste products travel back in the opposite direc- concentration (to estimate oncotic pressure) inside and outside
tion for elimination. Vitamins, signaling molecules, and other the choriocapillaris,86–88 equation (1) can be used to find σ ≈ 0.5.
factors needed for photoreceptor function are carried to the RPE Allowing that Πcc = 27 mmHg,86 Pcc = IOP + 8 mmHg,87 and
by lipoprotein particles passing through Bruch’s membrane, as assuming that ΠRPE = 0 mmHg (fluid pumped by the RPE is
do the RPE-produced lipoproteins that are eliminated in the assumed protein-free) and PRPE = IOP (assuming no pressure is
opposite direction. The RPE pumps water from the subretinal generated by the RPE above that necessary for crossing Bruch’s),
space to counter the swelling of the interphotoreceptor matrix we find that ΔP – σΔΠ is approximately –5.5 mmHg pulling fluid
GAGs. This fluid also flows across Bruch’s membrane to reach into the choroid. Thus, in normal young adults, oncotic pressure
the circulation. Thus, many transport processes involve Bruch’s
membrane, as reviewed here.
Table 20.2 Retinal pigment epithelium (RPE) fluid pumping rates
Hydraulic conductivity of Bruch’s membrane
GAGs are concentrated in the interphotoreceptor matrix77,78 and Fluid transport rate across RPE
Species (µL/h/cm2) References
corneal stroma.79 In both locations, these highly charged macro-
molecules maintain geometric fidelity essential for vision Frog 4.8–7.6 231, 232
(periodic collagen spacing for corneal transparency, orderly
photoreceptor spacing for visual sampling78,80,81). GAGs generate Rabbit 12 ± 4 233, 234
significant swelling pressure (up to 50 mmHg in cornea).82,83
Without a mechanism to maintain tissue deturgescence, GAGs Canine 6.4 235
would imbibe fluid, swell, destroy tissue geometry, and interfere Primate* 14 ± 3 236, 237
with visual function. Corneal endothelium forestalls swelling by
continuously pumping fluid out. This function is accomplished Human 11 238
for retina by the RPE, and its failure can lead to retinal detach- RPE pumping rates were measured by readsorption of subretinal fluid or by
ment. A driving force adequate to overcome the collective flow direct measurement in culture.
*Cantrill and Pederson236 measured a much higher transport rate than that
resistance of RPE, Bruch’s membrane, and choriocapillaris endo- reported here, but used fluorescein as a tracer which likely does not track fluid
thelium is provided by a gradient in fluid pressure and oncotic flow due to its high diffusion coefficient.
within the choroid is more than sufficient to adsorb all the fluid They reported that Lp of macular Bruch’s membrane exhibited
pumped by the RPE. We can also use equation (1) to calculate a dramatic, exponential decline throughout life (Fig. 20.5), drop-
that the lowest value of Lp that still adsorbs fluid pumped by the ping from 130 × 10−10 m/s/Pa in young children to 0.52 × 471
RPE without generating an elevated pressure at the RPE basal 10−10 m/s/Pa in old age. Lp of macular Bruch’s membrane
surface is Lp > 0.4 × 10−10 m/s/Pa. dropped more rapidly with age than did that of the periphery,

Chapter 20
Experiments using laser ablation of Bruch’s membrane/ consistent with an accelerated process occurring in the
choroid explants allowed Starita et al.89 to conclude that the ICL macula.1,84,94,95 Note that the lowest value measured for Lp of
was responsible for most of the flow resistance in Bruch’s mem- Bruch’s membrane in normal eyes is similar to the calculated
brane. Attempts to localize further the flow resistance using minimum value of Lp that allows complete fluid resorption
morphometric methods are complicated by first, stereological (0.4 × 10−10 m/s/Pa; see above). Marshall and Hussain’s group
issues90 and second, the loss of ultrastructural fidelity from con- reached similar conclusions regarding this process.94
nective tissue conventionally processed for electron micros- Determining Lp of Bruch’s membrane in isolated macular

Structure, Function, and Pathology of Bruch’s Membrane


copy.38 Failure to appreciate the former difficulty can lead to samples of AMD eyes is difficult due to scar formation and
unphysiologically low estimates for tissue porosity and thereby other changes.94 However, Marshall and Hussein’s group
hydraulic conductivity.1 showed that, in the periphery, Lp of Bruch’s membrane is
decreased in AMD eyes as compared to age-matched normal
Age-related changes in hydraulic conductivity eyes (Fig. 20.5).94 Assuming that similar processes occur in
and disease macular Bruch’s membrane due to the profound lipid accumu-
Fisher was the first to measure Lp of human Bruch’s membrane,91 lation in this region, then in diseased eyes, the RPE must gen-
finding that Lp decreased significantly with age. However, his erate higher pressures at its basal surface to drive fluid into
values for Lp of Bruch’s membrane and other tissues are much the choriocapillaris, with further pathological consequences.31
lower than those found by later investigators.85,92,93 Marshall and Above an unknown threshold level, higher pressure will cause
Hussain’s group carefully revisited these measurements using the RPE-BL to separate from the ICL, leading to RPE detach-
Bruch’s membrane/choroid with RPE removed, a preparation ment and fluid accumulation, as seen in 12–20% of AMD
that was simpler to create. They showed using laser ablation that patients.94
the flow resistance of these preparations was entirely due to What causes the dramatic age-related decrease in Lp of Bruch’s
Bruch’s membrane.89 They also found that flow rate increased membrane? It is natural to suspect the age-related lipid accu-
linearly with driving pressure, indicating that Lp of Bruch’s mulation. In fact, McCarty et  al.96 showed that lipid particles
membrane is relatively insensitive to pressure up to 25 mmHg. trapped in an extracellular matrix can generate very significant

Fig. 20.5 Hydraulic conductivity (Lp) of


Bruch’s membrane as a function of age.
1000
Dotted lines are exponential fits to data from
macular and peripheral regions, respectively.
Macular Note that all of the data from eyes with
age-related macular degeneration (AMD)
Peripheral
(taken only in peripheral region) have lower
Peripheral (AMD) values of Lp than the best fit to data taken
from peripheral Bruch’s membrane of
100 nondiseased eyes. (Reproduced with
permission from Hussain AA, Starita C,
Marshall J. Transport characteristics of
ageing human Bruch’s membrane:
implications for age-related macular
degeneration (AMD). In: Ioseliani OR, editor.
LP (m/s/Pa x 10-10)

Focus on macular degeneration research.


Hauppauge, New York: Nova Biomedical
10 Books; 2004.)

0.1
0 20 40 60 80 100
Age (years)
flow resistance, more than would be expected based simply on membrane.97 The agreement between the trends and the fits to
their size and number. However, Marshall and Hussain’s group the data is striking. This is strong evidence that the increasing
472 observed that most of the marked change in Lp occurred before lipid content and progressively hydrophobic character of Bruch’s
age 40 (Fig. 20.6A) while the increase in Bruch’s membrane membrane are responsible for impairing fluid transfer with age,
lipid content occurred largely after this age. They thus con- as postulated.31 The strong correlation between flow resistivity
cluded that other age-related changes must be responsible for of Bruch’s membrane and lipid content was likewise found by
Section 1

changes in Lp.1,84 Marshall and Hussain’s group.1,84,95 Laser ablation studies local-
A different conclusion can be reached from examining age- izing flow resistance to the ICL89 further support this conclusion,
effects on flow resistivity the inverse of Lp. Resistivity increases because lipids accumulate prominently in the ICL with aging.39
from a low of roughly R = 108 Pa/m/s for young individuals to Further, more laser pulses were required to abolish flow resis-
R = 1010 Pa/m/s for aged persons. Thus, when hydraulic con- tance in the oldest eyes, consistent with presence of a lipid wall,
ductivity Lp drops from roughly 100 × 10−10 m/s/Pa to 25 × requiring prior removal.
Anatomy and Physiology
Basic Science and Translation to Therapy

10−10 m/s/Pa between birth and 40 years of age, 75% of its total Thus, it appears that decreased Lp and increased resistivity of
possible decrease, resistivity R increases from 1 × 108 Pa/m/s to Bruch’s membrane with aging are closely related to the age-
4 × 108 Pa/m/s, only 4% of the ultimate increase. Simply put, related accumulation of lipids, primarily EC. Lipids accumulate
hydraulic conductivity drops more rapidly with age at young more rapidly in the macular Bruch’s membrane than in the
ages because its value is high to start with. Fig. 20.6B plots resis- periphery.35,98 Thus, Lp of the macula decreases more rapidly
tivity and histochemically detected EC against age for Bruch’s with age than it does in the periphery.

Fig. 20.6 (A) Hydraulic conductivity of


human macular Bruch’s membrane/choroidal
150 Hydraulic conductivity 250
preparations as a function of age, as
Hydraullic conductivity (m/sec Pa) x 10-10

compared to lipid accumulation in human


Lipid content
macular Bruch’s membrane; lines as
125 exponential fits to the data. (B) Hydraulic
200
resistivity of human macular Bruch’s
membrane/choroidal preparations as a
100
Total lipid (mg/m2)
function of age,1 as compared to esterified
150 cholesterol accumulation in human macular
Bruch’s membrane35; lines are exponential
75 fits to the data (the fits nearly overlie one
another). (Modified with permission from
100 Marshall J, Hussain AA, Starita C, et al,
50 editors. The retinal pigment epithelium:
function and disease. New York: Oxford
University Press; 1998. pp. 669–92.)
50
25

0 0
0 20 40 60 80 100
Age (years)

1 25
Hydraulic resistivity
Hydraullic resistivity (Pa sec/m x 10-10)

Esterified cholesterol
0.8
Fluorescence (arbitrary units)

20

0.6 15

0.4 10

0.2 5

0 0
0 20 40 60 80 100
Age (years)
Permeability of Bruch’s membrane to opposite direction of transport and thus complicating the results.
solute transport Nonetheless, useful comparative results can be generated.
Along with bulk fluid flow, there is significant transport of indi- The transport rate across human Bruch’s membrane declines 473
vidual molecular species across Bruch’s membrane, including linearly with age for all molecules measured. Amino acids
dissolved gases, nutrients, cytokines, and waste products driven exhibited permeabilities of 0.6 × 10−4  cm/s (phenylalanine) to

Chapter 20
by passive diffusion. Flow crossing Bruch’s membrane is too 1.2 × 10−4  cm/s (glycine) for young Bruch’s membrane and
slow to influence this process. This can be seen through calcula- exhibited a modest decline (twofold or less) with aging.101
tion of the Peclet number, the relative magnitude of convection Serum proteins decrease more markedly, dropping from
of a species due to bulk flow to that of diffusion99: 3.5 × 10−6  cm/s in the first decade to 0.2 × 10−6  cm/s in the
ninth decade, a >10-fold decrease.102 In particular, proteins
VL larger than 100  kDa have significantly decreased flux through
 (equation 2)
D0 Bruch’s membrane of older individuals. Macular Bruch’s

Structure, Function, and Pathology of Bruch’s Membrane


membrane showed a steeper decrease with age than did the
where V is the velocity of the flow, L is the transport path length, periphery.105 Permeability was reduced in eyes with AMD
and D0 the free diffusion coefficient of the species being trans- relative to age-matched normal eyes.105
ported. (The free diffusion coefficient in saline is used rather Decreased permeability of Bruch’s membrane to transport is
than its value in tissue, since the species carried by flow is con- likely due to a decrease in diffusion coefficients, especially for
strained to the same extent by the tissue as is its diffusion). Using the larger species affected by interaction with extracellular
the RPE pumping rate (Table 20.2) for V, Bruch’s membrane matrix and lipoproteins. As indicated in equation (3), increased
thickness (average of 3 µm59) for L, and a range of diffusion coef- path length due to age-related thickening of Bruch’s membrane59
ficients of species crossing Bruch’s membrane (2 × 10−7 cm2/s for could also have a significant effect.
LDL to 2 × 10−5 cm2/s for oxygen99,100), we find that the Peclet An original proposal of a molecular weight exclusion limit to
number ranges in value from 5 × 10−5 to 5 × 10−3. Thus, convection Bruch’s membrane macromolecule transport of 66–200 kDa101,102
is negligible in transporting species across Bruch’s membrane has been questioned by more recent work suggesting that, if
under physiological conditions. such a limits exists, it is much higher.105 Because of the impor-
Diffusion follows Fick’s law whereby the diffusive flux per tance of lipoproteins in transporting lipophilic nutrients to the
unit area (j) is proportional to the diffusion coefficient (D) of that RPE for ultimate use by the photoreceptors, and also because
species in the medium through which it passes and to the con- lipoproteins accumulate with age in Bruch’s membrane, Cankova
centration difference across the medium (ΔC), and inversely pro- et al.104 specifically examined the reflection coefficient of bovine
portional to the diffusion length: Bruch’s membrane to plasma LDL. They measured a reflection
coefficient of 0.58 (compared to a reflection coefficient of arterial
j = D∆C/L  (equation 3) endothelium to LDL of 0.998 and arterial intima to LDL of
0.827106). Thus, while LDL did not pass freely through Bruch’s
The permeability of a tissue to a given species is defined as P = membrane, it could nonetheless pass. Hussain et al.105 also con-
j/ΔC. We see then that P = D/L. For example, the permeability of cluded that particles as large as LDL could cross Bruch’s mem-
Bruch’s membrane to oxygen is ∼ 0.067 cm/s. Note that since brane. Accordingly, RPE cells have been shown to internalize
diffusion moves down a concentration gradient, one species plasma LDL from the choroid.56,107,108
might be diffusing across Bruch’s membrane toward the RPE These considerations are relevant not only to understanding
(e.g., oxygen) while another species (e.g., carbon dioxide) dif- mass transfer between the choriocapillaris and the RPE, but also
fuses simultaneously in the other direction. for transscleral drug delivery strategies, including antiangio-
With high diffusion coefficient and little interaction with genic agents for treating AMD and steroids for treating diabetic
extracellular matrix, small molecules (e.g., oxygen) diffuse retinopathy.109,110 Lipophilic solutes are significantly hindered in
quickly across Bruch’s membrane. However, macromolecules their transport by Bruch’s membrane/choroid,103 while hydro-
have much smaller free solution diffusion coefficients due to philic moieties are blocked by the RPE.111
their size. Coefficients are further reduced by interactions with
extracellular matrix or lipoproteins that accumulate with age, Summary and implications
so macromolecule transport across Bruch’s membrane is Bruch’s membrane’s physiological roles are structural and facili-
impeded. tating transport. Transport across Bruch’s membrane is increas-
The transport of amino acids,101 serum proteins,102 drugs,103 ingly hindered with age, due at least partly to the marked
and LDL104 across Bruch’s membrane has been examined. There age-related accumulation of EC-rich lipoproteins in this tissue,
are technical challenges to these experiments. First, as indicated impeding pumping of fluid from RPE.94 A ≥90% decrease in
in equation (3), diffusional flux depends on the length of the transport of some species from the choroid102,105 may include
tissue. Since the diffusion coefficient of the transported species lipophilic essentials delivered by lipoproteins. This decline in
is likely different in Bruch’s membrane than in the choroid in a transport capability is thought to have functional consequences
combined preparation, but the path lengths of both tissue com- for photoreceptors.112 A well-characterized change occurring
ponents are usually not determined, it is difficult to use the through the lifespan of individuals with healthy maculas is
measured values to determine absolute values of permeability. slowed dark adaptation,113 attributed to impaired translocation
Instead the more easily measured flux rate (j: see equation 3) is of retinoids across the RPE–Bruch’s interface. This slowing,
usually presented. Second, since larger macromolecules used in worse in AMD patients,114,115 can be partly ameliorated by short-
diffusion studies are hindered in their passage into the tissue, an term administration of high-dose vitamin A,116 presumably over-
oncotic pressure could develop, generating a fluid flow in the coming the translocation deficit via mass action.
PATHOLOGY OF BRUCH’S MEMBRANE sub-RPE tissue compartment as the lipid wall and BlinD. Found
in most older adults,67,119 drusen are more numerous in periph-
474 AMD lesions eral retina than in macula.120–122 Drusen are typically classified as
In aging and AMD, characteristic extracellular lesions accumu- “hard” and “soft” by the appearance of their borders. Soft drusen
late in tissue compartments anterior to the ICL. Known as drusen confer high risk of advanced disease123–126 and, importantly, are
found only in the macula.122 Other rare druse types exist and are
Section 1

and basal deposits,29,117 these lipid-containing aggregations ulti-


mately impact RPE and photoreceptor health by impairing trans- less well characterized.127
port, causing inflammation, and predisposing to CNV (Fig. 20.7). In separate 1854 publications, Donders (a Dutch ophthalmolo-
Basal linear deposit (BlinD) forms consequent to lipoprotein gist) and Wedl (an Austrian pathologist) described “colloid
accumulation in Bruch’s membrane and formation of the lipid bodies” (Colloidkugeln) or “hyaline deposits” on the inner surface
wall, likely involving oxidation of individual lipid classes and of the choroid in older or diseased human eyes3,128,129 (translated
local inflammation. Drusen could form by similar mechanisms, by Busk). Both authors interpreted the droplets that filled these
Anatomy and Physiology
Basic Science and Translation to Therapy

plus lipoprotein aggregation and other undefined processes that deposits as “fat globules.” The term “drusen” originated with
cause the distinctive dome shape of these lesions. Basal laminar Müller in 1856, from the German word for the geological term
deposit (BlamD) forms in parallel with lipid deposition in geode (not to be confused with Drüse, meaning gland).130 The
Bruch’s and may indicate RPE stressed by it. We begin by dis- name drusen was adopted by English writers early in the 20th
cussing drusen, due to their importance in AMD. century,131 yet “colloid body” was used by Holloway and Ver-
hoeff into the 1920s.132 The basis of the fatty content emerged
Drusen slowly. Lauber133,134 noted that deposits between the lamina
In a fundus view, drusen are yellow-white deposits 30–300 µm vitrea and the RPE were sudanophilic in 1924. Wolter and Falls135
in diameter posterior to the RPE. By optical coherence tomogra- stated that “hyaline bodies [drusen] … stain reddish with … oil
phy, they appear as variably hyporeflective spaces in the same red O” in 1962.
location.118 Histologically, drusen are focal, domed lesions Extant theories for druse formation, extending back to their
between the RPE basal lamina and the ICL, i.e., in the same discovery,130 fit into two general categories: transformation of the

Fig. 20.7 Bruch’s membrane and


characteristic age-related macular
degeneration lesions. (A) Bruch’s membrane
has five layers in a normal eye: 1, basal
lamina of the retinal pigment epithelium
(RPE); 2, inner collagenous layer; 3, elastic
layer; 4, outer collagenous layer; and 5,
basal lamina of the choriocapillary
endothelium (fenestrated cells).
L L, lipofuscin. (B) Older eyes have basal
laminar deposit (BlamD) and basal linear
deposit (BlinD) and its precursor, the lipid
wall. Drusen, BlinD, and the lipid wall occupy
RPE
the same tissue compartment. Basal mounds
1 are soft druse material within BlamD.
2 (Adapted with permission from Curcio CA,
3 Johnson M, Huang J-D, et al. Apolipoprotein
4
5 B-containing lipoproteins in retinal aging and
age-related maculopathy. J Lipid Res
A 2010;51:451–67.)

Basal mound

L
L

RPE

B Basal laminar Lipid wall Basal linear Druse


overlying RPE and deposition of materials on to Bruch’s mem- These lesions are richer in histochemically detectable UC than
brane. The latter is now accepted.129,135 The RPE has been impli- surrounding cellular membranes.141,142 By transmission electron
cated as a source of many druse components, via budding of microscopy following osmium tetroxide postfixation, membra- 475
membrane-bound packets of cytoplasm or secretion. The contri- nous debris appears as variably sized, contiguous coils of
bution of plasma-derived components, in contrast, has not been uncoated membranes consisting of uni- or multilamellar electron-

Chapter 20
well characterized. The existence of druse subregions addition- dense lines, denser than cellular membranes, surrounding an
ally suggests remodeling in the extracellular compartment, such electron-lucent center (Fig. 20.1). Since conventional ultrastruc-
as cellular invasion and enzymatic activity23,136–138 and uplifting tural preparation methods can remove lipids, the building blocks
of the lipid wall.35,139 of membranous debris are more plausibly the UC-rich exteriors
Most prominent among druse constituents are lipids, an of lipoproteins (native and fused) whose neutral lipid interiors
observation made by their earliest discoverers. All drusen are not well preserved in postmortem tissue.39,153,157 Rather than
contain EC and UC, in addition to phosphatidylcholine, other vesicles, then, membranous debris is likely aggregated or fused

Structure, Function, and Pathology of Bruch’s Membrane


phospholipids, and ceramides.34,35,137,139–142 Extractable lipids particles that could collectively account for the abundant EC in
account for ≥40% of hard druse volume143 and likely more for sub-RPE deposits. EC abundance and ultrastructural evidence
macular soft drusen.139 This includes large EC-rich lakes in soft for solid, nonvesicular particles suggest that the major lipid-
drusen (Fig. 20.8), reminiscent of atherosclerotic plaques.144 Only containing component of AMD-specific lesions can be called
half of macular drusen take up hydrophilic fluorescein in angi- “lipoprotein-derived debris” rather than membranous debris.
ography,145 possibly reflecting differing proportions of polar and Basal linear deposit
neutral lipids in individual lesions.141 Discrete nonlipid compo-
BlinD is a thin (0.4–2 µm) layer located in the same sub-RPE
nents in some drusen include amyloid assemblies and granules
compartment as soft drusen. BlinD is not visible clinically except
of lipofuscin or melanin, indicating cellular origin (Table 20.3,
as associated with other pathology. By OTAP and QFDE, BlinD
online). Other constituents present in all drusen include vitro-
is rich in solid lipoprotein particles and lipid pools (Fig. 20.9A,
nectin, TIMP-3, complement factor H, complement components
C). BlinD and soft drusen are considered alternate forms (layer
C3 and C8, crystallins, and zinc.23,136,138,146–150
and lump) of the same entity158 and may interchange over time.
Apolipoprotein immunoreactivity appears in drusen with
Soft drusen are oily, difficult to isolate individually, and are
high frequency (100%, apoE; >80% apoB; 60%, A–I).139,151–154
biomechanically more fragile than hard drusen,122 properties
Plasma-abundant apoC-III is present in fewer drusen than
applicable to BlinD by inference. Both lesions could thus be
plasma-sparse apoC-I, indicating a specific retention mechanism
permissive to invading capillaries of type I CNV.159,160 ApoE and
for plasma-derived apolipoproteins or an intraocular source.
apoB are present in BlinD and its precursor, the lipid wall.139,151,152
Importantly, hard drusen contain many solid, Folch-extractable
Transitional morphologies between lipid wall and BlinD have
electron-dense particles of the same diameter as the lipoproteins
been reported.161
that accumulate with age in Bruch’s membrane. These observa-
tions together with the appearance of membranous debris in soft Basal laminar deposit
drusen (below) make an RPE-secreted apoB-containing lipopro- BlamD forms small pockets between the RPE and the RPE-BL
tein particle an efficient mechanism to place multiple lipids and in many older normal eyes or a continuous layer as thick as
apolipoproteins within lesion compartments. 15  µm in AMD eyes142,156,162 (Fig. 20.7). Some authors consider
The principal lipid-containing component of soft drusen and a continuous layer of BlamD a histological definition of AMD.163
BlinD was called “membranous debris” by the Sarks.123,155,156 Ultrastructurally, BlamD resembles basement membrane

A B

Fig. 20.8 Esterified cholesterol (EC) forms lakes in macular soft drusen. (A) EC lakes in a macular soft druse revealed by filipin fluorescence
(arrow). Bar = 25 µm. (Adapted with permission from Malek G, Li C-M, Guidry C, et al. Apolipoprotein B in cholesterol-containing drusen and basal
deposits in eyes with age-related maculopathy. Am J Pathol 2003;162:413–25.) (B) Macular soft druse from an eye with age-related macular
degeneration has lakes of homogeneous electron-dense lipid (arrow) among partially preserved lipoprotein-like material. Basal laminar deposit
(asterisk) overlying the druse has similar material, called membranous- or lipoprotein-derived debris (to the right of the asterisk). Bar = 1 µm.
Table 20.3 (online) Localized components of drusen

Component Phase Direct assay Abundance References 475.e1


Lipoproteins (EC, UC, phospholipid) P ✓ All drusen; >40% of hard druse volume; 122, 239, 240
EC pools in soft drusen

Chapter 20
Apolipoproteins (apoB, A-I, C-I, E) P apoE ✓ 60–100% of hard drusen; higher rates 152, 154, 240
in periphery than macula

Melanin/lipofuscin granules P 6% of hard and soft drusen 122

Cells (dendritic, others) P 3–6% of hard drusen only 122, 241

Structure, Function, and Pathology of Bruch’s Membrane


Amyloid vesicles (0.25-10 µm) P 2% of hard drusen, 40% of compound 122, 136, 239, 242
drusen, frequent in eyes with many
drusen, some AMD eyes

Calcification P ✓ 43% of macular hard drusen, 1.6% of 122


soft drusen, 2% of peripheral hard
drusen

Clusterin, TIMP3, vitronectin, D ✓ Reliably detected; abundance inferred 143, 191


apolipoprotein E, complement factor H,
complement components 8, 9

Components of classic, lectin, alternative, D Some ✓ Many pathway components evidence 243
terminal complement pathways; C3 key role of complement
fragments indicating activation

RGR-d D All drusen 244

αA- and αB-crystallin D ✓ NA; higher in BrM, more in AMD drusen 150, 191

Ubiquitin D Most drusen in most eyes 245

Exosome markers CD63, CD81, and P NA 246


LAMP2

Bestrophin, membrane-bound P NA 247

Carbohydrates D All drusen 191

Zinc D ✓ Many drusen 138


EC, esterified cholesterol; UC, unesterified cholesterol; P, particulate; D, dispersed; NA, not available; AMD, age-related macular degeneration; BrM, Bruch’s membrane.
Localization methods: immunohistochemistry, histochemistry, immunogold transmission electron microscopy. Direct assays: proteomics, Western blot, microprobe
synchrotron X-ray fluorescence for zinc. Varying estimates of particulate druse components are due to differences in location of samples and druse types examined.
Fig. 20.9 Lipoprotein-derived debris and lipid
pools in age-related macular degeneration
lesions are solid rather than vesicular
476 material. (A) Above retinal pigment epithelium
(RPE) basal lamina (arrowheads) is basal
laminar deposit with individual particles
indicated (arrow). Below RPE basal lamina is
Section 1

numerous solid particles in basal linear


deposit. Transmission electron microscopy,
osmium paraphenylenediamine fixation.
Bar = 500 nm. (Reproduced with permission
from Curcio CA, Presley JB, Millican CL, et al.
Basal deposits and drusen in eyes with
age-related maculopathy: evidence for solid
lipid particles. Exp Eye Res 2005;80:761–75.)
Anatomy and Physiology
Basic Science and Translation to Therapy

(B) Basal laminar deposit appears as a solid


column of basal lamina-like material, with
solid particles embedded within (arrow).
Bar = 500 nm. (Courtesy of J-D Huang.)
(C) Basal linear deposit has lipoproteins of
heterogeneous sizes and shapes as well as
B pooled lipid, consistent with a model of
surface degradation and particle fusion.
Bar = 200 nm. (Reproduced from Curcio CA,
Johnson M, Rudolf M, et al. The oil spill in
ageing Bruch’s membrane. Br J Ophthalmol
2011;95:1638–45.)

A C

mat­erial (Fig. 20.9B), containing laminin, fibronectin, type IV macula.174 This coherent morphology suggests a specific forma-
and type VI collagen.164–167 The latter is a distinctive banded tive process, possibly involving microglia resident in that
material with 120  nm periodicity, called wide- or long-spacing compartment.175,176
collagen, which also appears in other ocular locations like
epiretinal membranes. Thick BlamD, associated with advanced Summary
AMD risk,156 contains histochemically detectable lipid, including Levels of significance ascribed to molecules sequestered
UC and EC141,142 and is a classically described site for membra- in drusen (Table 20.3, online), and by inference, BlinD,
nous debris (Fig. 20.1C). By lipid-preserving methods, solid include toxicity to the overlying RPE, stigmata of formative pro-
particles are seen in BlamD (Fig. 20.9A, B). Especially enriched cesses (extrusion of cellular materials, secretion, extracellular
in basal mounds156 (Fig. 20.7), lipoprotein-derived debris in enzymatic processing, cellular activity), and markers of a dif-
BlamD may be considered as retained in transit from the RPE fusely distributed disease process affecting RPE and Bruch’s
to BlinD and/or drusen.139,141,142 Morphologically heterogeneous membrane. Additional significance can be ascribed to these
BlamD also contains vitronectin, MMP-7, TIMP-3, C3, and C5b- lesions as physical objects that increase path length between
9,162 EC, and UC.142 Evoked in numerous mouse models of aging, choriocapillaries and retina and provide a biomechanically
stress, and genetic manipulation, BlamD is a reliable marker of unstable cleavage plane between RPE BL and ICL.
RPE stress.168
Subretinal drusenoid debris Response-to-retention hypothesis of AMD
Hypotheses of druse formation must eventually also account for The parallels between the pathology of arterial intima of large
subretinal drusenoid debris (SDD). Located adjacent to RPE in arteries and that of Bruch’s membrane are striking. Both diseases
the subretinal space, SDD was first described in AMD eyes by feature cholesterol-rich lesions in subendothelial compartments
the Sarks.155 Ultrastructurally similar to soft drusen, these depos- within the systemic circulation, involving many of the same
its are enriched in UC, apoE, vitronectin, and complement factor molecules and biological processes at multiple steps, as long
H, and, like drusen, they lack markers for photoreceptors, Müller anticipated.177,178 According to the response-to-retention theory
cells, and RPE apical processes.142,169 Clinically this material is of atherosclerosis, plasma lipoproteins cross the vascular endo-
called reticular drusen in a fundus view170 and subretinal thelium of large arteries, and bind to extracellular matrix. By
drusenoid debris in a cross-sectional view.171 Conferring lower itself, this process is not pathological. However, lipoprotein
risk for advanced AMD than conventional drusen,172 SDD appear components become modified via oxidative and nonoxidative
in up to 60% of geographic atrophy eyes,172,173 appearing as focal processes, and launch numerous downstream deleterious
deposits near the fovea and part of large sheets elsewhere in the events, including inflammation, macrophage recruitment, and
neovascularization, leading to disease.179,180 Parallel with apoB Angioid streaks are associated with abetalipoproteinemia,192–195
lipoprotein-instigated disease in arterial intima, an intraocular an extremely rare disorder with low plasma apoB-containing
response to retention involving the RPE and Bruch’s membrane lipoproteins, acanthocytosis of erythrocytes, neuropathy, and 477
in aging and AMD would begin with age-related accumulation pigmentary retinopathy. It is historically attributed to lack of
of lipoproteins of local origin. Oxidation, perhaps driven by lipophilic vitamins delivered by plasma LDL.44 The RPE is

Chapter 20
reactive oxygen species from adjacent RPE mitochondria, would now known to express the abetalipoproteinemia gene (MTP),46
then initiate a pathological process resembling that in the vas- which cotranslationally lipidates apoB (see above). How MTP
cular system with inflammation-driven downstream events deficiency leads to angioid streaks is unknown. The finding,
including complement activation and structurally unstable however, highlights that lack of apoB lipoproteins has negative
lesions.36 consequences for Bruch’s membrane health, just as an excess of
retained apoB lipoproteins has negative consequences via lesion
Neovascular AMD formation and impaired transport (see above). Good chorioreti-

Structure, Function, and Pathology of Bruch’s Membrane


CNV, the major sight-threatening complication of AMD, involves nal function thus requires an optimal balance between these
angiogenesis along vertical and horizontal vectors: vertically extremes.
across Bruch’s membrane, and laterally external to the RPE (type
1 CNV181), laterally within the subretinal space (type 2 CNV), or Thick basal laminar deposits
further anteriorly into the retina (type 3 CNV).181–183 Of 40+ con- (TIMP-3, CTRP5, EFEMP1 genes)
ditions involving CNV, AMD is the most prevalent, followed by Three autosomal dominant inherited disorders with adult
ocular histoplasmosis,181 and including angioid streaks (see onset – Sorsby fundus dystrophy, late-onset retinal degeneration
below). CNV is a multifactorial nonspecific wound-healing (LORD) and malattia leventinese-Doyne honeycomb retinal dys-
response to various specific stimuli, involving VEGF stimulation trophy (ML-DH) – share phenotypic similarities with AMD and
of choriocapillaris endothelium, compromise to Bruch’s mem- provide mechanistic support for many aspects of Bruch’s mem-
brane, and participation of macrophages.181 Impaired transport brane physiology and pathophysiology, discussed above. All
across Bruch’s membrane in AMD increasingly isolates the RPE three conditions result from mutations in genes encoding extra-
from its metabolic source in the choriocapillaries and enhances cellular matrix proteins or their regulators (Sorsby, TIMP3196;
the challenge in waste product disposal. VEGF released by RPE LORD - CTRP5197; and ML-DH, EFEMP1198). All three can prog-
as a stress signal initiates an angiogenic response by the endo- ress to CNV to varying degrees (Sorsby > LORD > ML-DH). All
thelium. However, Bruch’s membrane compromise is essential three have visual dysfunction, especially rods, attributed to a
for CNV to proceed, as evidenced by intrachoroidal neovascu- nutritional night blindness that is responsive to short-term
larization without CNV in a mouse overexpressing VEGF in the administration of high-dose vitamin A in Sorsby and LORD.199–201
setting of an intact Bruch’s membrane.184 Sorsby and LORD are notable for thick BlamD and areas of RPE
Bruch’s membrane in a state of compromise can be breached atrophy202 and may involve macula and periphery, while ML-DH
easily by new vessels in AMD. It is notable that the EL is is notable for radially distributed drusen and peripapillary
thinner and more interrupted in eyes with neovascular AMD.23 deposits. In Sorsby eyes mutant TIMP-3 localizes to BlamD. In
The length of gaps in the EL is greater in eyes with early AMD ML-DH, EFEMP1 localizes to BlamD and not to the pathogno-
and any CNV.23 In paired donor eyes with and without CNV monic drusen themselves, suggesting an important role of
secondary to AMD, progressed eyes are distinguished by cal- BlamD in druse formation.
cification and breaks in Bruch’s membrane.185 In contrast, cal- BlamD in Sorsby and LORD, like that in AMD, is notably rich
cification in a small number of geographic atrophy eyes is in oil red O-binding lipid.203–205 The significance of these findings
unremarkable.186 was unclear until a model of a Bruch’s membrane lipoprotein
BlinD furthers this process by presenting a horizontal cleavage was articulated. In LORD eyes,205 deposits contain EC, UC, and
plane for vessel formation to exploit. The lipid-rich composition, apoB, and lipid-preserving ultrastructural methods revealed
relative lack of structural elements like collagen fibrils, lesion solid electron-dense particles tracking in intersecting networks
biomechanical instability,122 and proinflammatory, proangio- across the BlamD. In hindsight, these may represent native lipo-
genic compounds like 7-ketocholesterol and linoleate hydroper- proteins in transit from RPE to the choriocapillaris rather than
oxide160,181,187 likely promote vessel growth in this plane.160 depositions/aggregations of plasma LDL, as originally specu-
lated. Lipid particle disposition within these thick deposits has
Angioid streaks (ABCC6, MTP genes) been replicated in a mouse model expressing the R345W EFEMP1
Angioid streaks are ruptures in Bruch’s membrane associated mutation.168
with multiple disorders, caused by excess calcification of the
elastic layer188 and often accompanied by CNV. They are
prominent ocular manifestation of pseudoxanthoma elasticum
CONCLUSION
(PXE), a systemic connective tissue disorder. PXE patients Bruch’s membrane serves essential functions as substrate to
harbor mutations of a hepatically expressed lipid transporter the RPE and vessel wall of the outer retina. Its layers and
ABCC6.189 Clinical presentation includes, in addition to streaks constituent proteins collectively represent a barrier that keeps
and CNV, peau d’orange (flat, yellow, drusen-like lesions), choroidal vessels at bay, provides a route for water, solutes,
optic nerve head drusen, outer retinal tubulations, subretinal and macromolecules that transfer between RPE and choroid,
fluid, and pigmentary changes.190 PXE clinical manifestations while supporting the structural integrity of both. It is unusual
are believed to be related to ectopic mineralization of nonhe- among human tissues in accumulating a high content of EC-rich
patic tissues, suggesting a defect in the transport of antimin- neutral lipid over the lifespan. A natural history and biochemi-
eralization agents.191 cal model now suggest this lipid is due to apoB lipoprotein
secretion by RPE, which may be part of an outer retinal nutri- 32. Pauleikhoff D, Harper CA, Marshall J, et al. Aging changes in Bruch’s mem-
tion system. This deposition can account for the impaired brane: a histochemical and morphological study. Ophthalmology
1990;97:171–8.
478 outward movement of fluid from RPE, increasing risk for RPE 33. Pauleikhoff D, Wojteki S, Müller D, et al. [Adhesive properties of basal mem-
detachments, more common in older persons, and impaired branes of Bruch’s membrane. Immunohistochemical studies of age-dependent
changes in adhesive molecules and lipid deposits.] Ophthalmologe 2000;97:
macromolecular transport also leading to RPE stress. Oxidation 243–50.
of these lipid deposits in Bruch’s membrane likely initiates 34. Haimovici R, Gantz DL, Rumelt S, et al. The lipid composition of drusen,
Section 1

an inflammatory process that leads to lesion formation and Bruch’s membrane, and sclera by hot stage polarizing microscopy. Invest
Ophthalmol Vis Sci 2001;42:1592–9.
CNV in AMD. 35. Curcio CA, Millican CL, Bailey T, et al. Accumulation of cholesterol with
age in human Bruch’s membrane. Invest Ophthalmol Vis Sci 2001;42:
265–74.
REFERENCES 36. Curcio CA, Johnson M, Huang J-D, et al. Aging, age-related macular degen-
1. Marshall J, Hussain AA, Starita C, et al., editors. The retinal pigment epithe- eration, and the response-to-retention of apolipoprotein B-containing lipopro-
lium: function and disease. New York: Oxford University Press; 1998. teins. Prog Ret Eye Res 2009;28:393–422.
p. 669–92.
Anatomy and Physiology

37. Verhoeff FH, Sisson RJ. Basophilic staining of Bruch’s membrane. Arch
Basic Science and Translation to Therapy

2. Harris A, Moss A, Erhlich R. The choroid. In: Tasman W, Jaeger EA, editors. Ophthalmol 1926;55:125–7.
Duane’s ophthalmology. 18th ed. Philadelphia, PA: Lippincott Williams & 38. Ruberti JW, Curcio CA, Millican CL, et al. Quick-freeze/deep-etch visualiza-
Wilkins; 2011. tion of age-related lipid accumulation in Bruch’s membrane. Invest Ophthal-
3. Wedl C. Grundzüge der pathologischen Histologie. Vienna: Carl Gerold & mol Vis Sci 2003;44:1753–9.
Sohn; 1854. 39. Huang J-D, Presley JB, Chimento MF, et al. Age-related changes in human
4. Bruch CLW. Untersuchungen zur Kenntniss des Kornigen Pigments der macular Bruch’s membrane as seen by quick-freeze/deep-etch. Exp Eye Res
Wirbelthiere. Zurich: University of Zurich; 1844. 2007;85:202–18.
5. Smirnow AE. Zum Baue der Chorioidea propia des erwachsenen Menschen 40. Huang J-D, Curcio CA, Johnson M. Morphometric analysis of lipoprotein-like
(Stratum elasticum supracapillare). Graefes Arch 1899;57:451. particle accumulation in aging human macular Bruch’s membrane.
6. Salzmann M. The anatomy and histology of the human eye in the normal Invest Ophthalmol Vis Sci 2008;49:2721–7.
state. Its development and senescence. Chicago: University of Chicago; 1912. 41. Smith E. The influence of age and atherosclerosis on the chemistry of aortic
7. Wolfensberger TJ. The historical discovery of the retinal pigment epithelium. intima. I. The lipids. J Atheroscler Res 1965;5:224–40.
In: Marmor MF, Wolfensberger TJ, editors. The retinal pigment epithelium: 42. Holz FG, Sheraidah G, Pauleikhoff D, et al. Analysis of lipid deposits extracted
function and disease. New York: Oxford University Press; 1998. p. 13–22. from human macular and peripheral Bruch’s membrane. Arch Ophthalmol
8. Duke-Elder S, Cook C. Part 1, Embryology. St Louis: Mosby; 1967. 1994;112:402–6.
9. Mann I. The development of the human eye. New York: Grune and Stratton; 43. Shoulders CC, Shelness GS. Current biology of MTP: implications for selective
1964. inhibition. Curr Top Med Chem 2005;5:283–300.
10. Scales D, Fryczkowski A, Opremcak E. The choroid. In: Jakoviec F, editor. 44. Chowers I, Banin E, Merin S, et al. Long-term assessment of combined vitamin
Principle and practices of ophthalmology. Philadelphia, PA: WB Saunders; A and E treatment for the prevention of retinal degeneration in abetalipopro-
1994. p. 252–61. teinaemia and hypobetalipoproteinaemia patients. Eye 2001;15:525–30.
11. Takei Y, Ozanics V. Origin and development of Bruch’s membrane in monkey 45. Shelness GS, Ledford AS. Evolution and mechanism of apolipoprotein
fetuses: an electron microscopic study. Invest Ophthalmol 1975;14:903–16. B-containing lipoprotein assembly. Curr Opin Lipidol 2005;16:325–32.
12. Rizzolo LJ, Chen X, Weitzman M, et al. Analysis of the RPE transcriptome 46. Li C-M, Presley JB, Zhang X, et al. Retina expresses microsomal triglyceride
reveals dynamic changes during the development of the outer blood–retinal transfer protein: implications for age-related maculopathy. J Lipid Res
barrier. Mol Vis 2007;13:1259–73. 2005;46:628–40.
13. Saint-Geniez M, D’Amore PA. Development and pathology of the hyaloid, 47. Wu T, Fujihara M, Tian J, et al. Apolipoprotein B100 secretion by cultured
choroidal and retinal vasculature. Int J Dev Biol 2004;48:1045–58. ARPE-19 cells is modulated by alteration of cholesterol levels. J Neurochem
14. Hogan MJ, Alvarado JA, Weddell JE. Histology of the human eye. An atlas 2010; 114:1734–44.
and textbook. Philadelphia, PA: WB Saunders; 1971. 48. Contois JH, McConnell JP, Sethi AA, et al. Apolipoprotein B and cardiovas-
15. Gass JDM. Stereoscopic atlas of macular diseases: diagnosis and treatment. cular disease risk: position statement from the AACC Lipoproteins and
4th ed. St Louis: Mosby; 1997. Vascular Diseases Division Working Group on Best Practices. Clin Chem
16. Guymer R, Bird A. Age changes in Bruch’s membrane and related structures. 2009;55:407–19.
In: Ryan SJ, editor. Retina. St Louis: Mosby; 2006. p. 1030–9. 49. Dashti N, McGwin Jr G, Owsley C, et al. Plasma apolipoproteins and risk for
17. Booij JC, Baas DC, Beisekeeva J, et al. The dynamic nature of Bruch’s mem- age-related maculopathy. Br J Ophthalmol 2006;90:1028–33 (Erratum Br J
brane. Prog Retin Eye Res 2009;9:1–18. Ophthalmol. 2007;91:403.).
18. Chen L, Miyamura N, Ninomiya Y, et al. Distribution of the collagen IV iso- 50. Wang L, Li C-M, Rudolf M, et al. Lipoprotein particles of intra-ocular origin
forms in human Bruch’s membrane. Br J Ophthalmol 2003;87:212–5. in human Bruch membrane: an unusual lipid profile. Invest Ophthalmol Vis
19. Aisenbrey S, Zhang M, Bacher D, et al. Retinal pigment epithelial cells syn- Sci 2009;50:870 – 7.
thesize laminins, including laminin 5, and adhere to them through alpha3- 51. Bretillon L, Thuret G, Gregoire S, et al. Lipid and fatty acid profile of the
and alpha6-containing integrins. Invest Ophthalmol Vis Sci 2006;47:5537–44. retina, retinal pigment epithelium/choroid, and the lacrimal gland, and asso-
20. Hewitt TA, Nakasawa K, Newsome DA. Analysis of newly synthesized ciations with adipose tissue fatty acids in human subjects. Exp Eye Res
Bruch’s membrane proteoglycans. Invest Ophthalmol Vis Sci 1989;30: 2008;87:521–8.
478–86. 52. Curcio CA, Johnson M, Rudolf M, et al. The oil spill in aging Bruch’s mem-
21. Call TW, Hollyfield JG. Sulfated proteoglycans in Bruch’s membrane of brane. Br J Ophthalmol 2011; 95:1638–45.
the human eye: localization and characterization using cupromeronic blue. 53. Hogan MJ. Role of the retinal pigment epithelium in macular disease.
Exp Eye Res 1990;51:451–62. Trans Am Acad Ophthalmol Otolaryngol 1972;76:64–80.
22. Korte GE, D’Aversa G. The elastic tissue of Bruch’s membrane. Arch 54. Grindle CFJ, Marshall J. Ageing changes in Bruch’s membrane and their
Ophthalmol 1989;107:1654–8. functional implications. Trans Ophthalmol Soc U K 1978;98:172–5.
23. Chong NH, Keonin J, Luthert PJ, et al. Decreased thickness and integrity of 55. Loane E, Nolan JM, O’Donovan O, et al. Transport and retinal capture of
the macular elastic layer of Bruch’s membrane correspond to the distribution lutein and zeaxanthin with reference to age-related macular degeneration.
of lesions associated with age-related macular degeneration. Am J Pathol Surv Ophthalmol 2008;53:68–81.
2005;166:241–51. 56. Tserentsoodol N, Sztein J, Campos M, et al. Uptake of cholesterol by the retina
24. Yu HG, Liu X, Kiss S, et al. Increased choroidal neovascularization following occurs primarily via a low density lipoprotein receptor-mediated process.
laser induction in mice lacking lysyl oxidase-like 1. Invest Ophthalmol Vis Sci Mol Vis 2006;12:1306–18.
2008;49:2599–605. 57. Tserentsoodol N, Gordiyenko NV, Pascual I, et al. Intraretinal lipid transport
25. Newsome DA, Huh W, Green WR. Bruch’s membrane age-related changes is dependent on high density lipoprotein-like particles and class B scavenger
vary by region. Curr Eye Res 1987;6:1211–21. receptors. Mol Vis 2006;12:1319–33.
26. Lutty GA, Hasegawa T, Baba T, et al. Development of the human choriocapil- 58. Provost A, Vede L, Bigot K, et al. Morphological and electroretinographic
laris. Eye 2010;24:408–15. phenotype of SRBI knock-out mice after a long term atherogenic diet. Invest
27. Roberts JM, Forrester JV. Factors affecting the migration and growth of endo- Ophthalmol Vis Sci 2009;50:3931–42.
thelial cells from microvessels of bovine retina. Exp Eye Res 1990;50:165–72. 59. Ramrattan RS, van der Schaft TL, Mooy CM, et al. Morphometric analysis of
28. Smith W, Assink J, Klein R, et al. Risk factors for age-related macular degen- Bruch’s membrane, the choriocapillaris, and the choroid in aging. Invest
eration. Pooled findings from three continents. Ophthalmology 2001;108: Ophthalmol Vis Sci 1994;35:2857–64.
697–704. 60. Okubo A, Rosa Jr RH, Bunce CV, et al. The relationships of age changes in
29. Sarks SH. Ageing and degeneration in the macular region: a clinico- retinal pigment epithelium and Bruch’s membrane. Invest Ophthalmol Vis Sci
pathological study. Br J Ophthalmol 1976;60:324–41. 1999;40:443–9.
30. Johnson M, Huang J-D, Presley JB, et al. Comparison of morphology of human 61. Killingsworth MC. Age-related components of Bruch’s membrane. Graefes
macular and peripheral Bruch’s membrane in older eyes. Curr Eye Res Arch Clin Exp Ophthalmol 1987;225:406–12.
2007;32:791–9. 62. Karwatowski WSS, Jeffried TE, Duance VC, et al. Preparation of Bruch’s
31. Bird AC, Marshall J. Retinal pigment epithelial detachments in the elderly. membrane and analysis of the age-related changes in the structural collagens.
Trans Ophthalmol Soc UK 1986;105:674–82. Br J Ophthalmol 1995;79:944–52.
63. Macgregor AM, Eberhart CG, Fraig M, et al. Tissue inhibitor of matrix 98. Holz FG, Piguet B, Minassian DC, et al. Decreasing stromal iris pigmentation
metalloproteinase-3 levels in the extracellular matrix of lung, kidney, and eye as a risk factor for age-related macular degeneration. Am J Ophthalmol
increase with age. J Histochem Cytochem 2009;57:207–13. 1994;117:19–23.
64. Qi JH, Ebrahem Q, Moore N, et al. A novel function for tissue inhibitor of 99. Truskey GA, Yuan F, Katz DF. Transport phenomena in biological systems. 479
metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of Upper Saddle River, NJ: Pearson Prentice Hall; 2004.
VEGF binding to VEGF receptor-2. Nat Med 2003;9:407–15. 100. Fisher WR, Granade ME, Mauldin JL. Hydrodynamic studies of human low
65. Ebrahem Q, Qi JH, Sugimoto M, et al. Increased neovascularization in mice density lipoproteins. Evaluation of the diffusion coefficient and the preferen-

Chapter 20
lacking tissue inhibitor of metalloproteinases-3. Invest Ophthalmol Vis Sci tial hydration. Biochemistry 1971;10:1622–9.
2011;52:6117–23. 101. Hussain AA, Rowe L, Marshall J. Age-related alterations in the diffusional
66. Beattie JR, Pawlak AM, Boulton ME, et al. Multiplex analysis of age-related transport of amino acids across the human Bruch’s–choroid complex. J Opt
protein and lipid modifications in human Bruch’s membrane. FASEB J Soc Am A 2002;19:166–72.
2010;24:4816–24. 102. Moore DJ, Clover GM. The effect of age on the macromolecular permeability
67. van der Schaft TL, Mooy CM, de Bruijn WC, et al. Histologic features of the of human Bruch’s membrane. Invest Ophthalmol Vis Sci 2001;42:2970–5.
early stages of age-related macular degeneration. Ophthalmol 1992;99: 103. Cheruvu NP, Kompella UB. Bovine and porcine transscleral solute transport:
278–86. influence of lipophilicity and the choroid-Bruch’s layer. Invest Ophthalmol
68. Davis WL, Jones RG, Hagler HK. An electron microscopic histochemical and Vis Sci 2006;47:4513–22.
analytical X-ray microprobe study of calcification in Bruch’s membrane from 104. Cankova Z, Huang, J-D, Kruth, H, et al. 2011. Passage of low-density lipo­

Structure, Function, and Pathology of Bruch’s Membrane


human eyes. J Histochem Cytochem 1981;29:601–8. proteins through Bruch’s membrane and choroid. Exp Eye Res 2011;93:
69. Colville D, Savige J. Alport syndrome. Review of the ocular manifestations. 947–55.
Ophthalm Genet 1997;18:161–73. 105. Hussain AA, Starita C, Hodgetts A, et al. Macromolecular diffusion charac-
70. Kim RY, Faktorovich EG, Kuo CY, et al. Retinal function abnormalities in teristics of ageing human Bruch’s membrane: implications for age-related
membranoproliferative glomerulonephritis type II. Am J Ophthalmol macular degeneration (AMD). Exp Eye Res 2010;90:703–10.
1997;123:619–28. 106. Yang N, Vafai K. Modeling of low-density lipoprotein (LDL) transport in
71. Weiner DE, Tighiouart H, Reynolds R, et al. Kidney function, albuminuria the artery – effects of hypertension. Int J Heat Mass Transfer 2006;49:
and age-related macular degeneration in NHANES III. Nephrol Dialysis 850–67.
Transplant. 2011; 26:3159–65. 107. Elner VM. Retinal pigment epithelial acid lipase activity and lipoprotein
72. Maddox DA, Brenner BM. Glomerular filtration of fluid and macromolecules: receptors: effects of dietary omega-3 fatty acids. Trans Am Ophthalmol Soc
the renal response to injury. Annu Rev Med 1977;28:91–102. 2002;100:301–38.
73. Beers APA, Van Der Heijde GL. In vivo determination of the biomechanical 108. Gordiyenko N, Campos M, Lee JW, et al. RPE cells internalize low-density
properties of the component elements of the accommodation mechanism. lipoprotein (LDL) and oxidized LDL (oxLDL) in large quantities in vitro and
Vision Res 1994;34:2897–905. in vivo. Invest Ophthalmol Vis Sci 2004;45:2822–9.
74. Charman WN. The eye in focus: accommodation and presbyopia. Clin Exp 109. Martidis A, Duker JS, Greenberg PB, et al. Intravitreal triamcinolone for
Optom 2008;91:207–25. refractory diabetic macular edema. Ophthalmology 2002;109:920–7.
75. Chan WH, Hussain AA, Marshall J. Young’s modulus of Bruch’s membrane: 110. Anderson OA, Bainbridge JW, Shima DT. Delivery of anti-angiogenic
implications for AMD. Invest Ophthalmol Vis Sci 2007;48:2187. molecular therapies for retinal disease. Drug Discov Today 2010;15:
76. Ugarte M, Hussain AA, Marshall J. An experimental study of the elastic 272–82.
properties of the human Bruch’s membrane–choroid complex: relevance to 111. Pitkanen L, Ranta VP, Moilanen H, et al. Permeability of retinal pigment
ageing. Br J Ophthalmol 2006;90:621–6. epithelium: effects of permeant molecular weight and lipophilicity.
77. Tawara A, Varner HH, Hollyfield JG. Proteoglycans in the mouse interphoto- Invest Ophthalmol Vis Sci 2005;46:641–6.
receptor matrix. I. Histochemical studies using cuprolinic blue. Exp Eye Res 112. Jackson GR, Curcio CA, Sloan KR, et al. Photoreceptor degeneration in aging
1988;46:689–704. and age-related maculopathy. In: Penfold PL, Provis JM, editors. Macular
78. Hollyfield JG. Hyaluronan and the functional organization of the interphoto- degeneration. Berlin: Springer-Verlag; 2005. p. 45–62.
receptor matrix. Invest Ophthalmol Vis Sci 1999;40:2767–9. 113. Jackson GR, Owsley C, McGwin G. Aging and dark adaptation. Vision Res
79. Klintworth GK. The cornea – structure and macromolecules in health and 1999;39:3975–82.
disease. A review. Am J Pathol 1977;89:718–808. 114. Owsley C, McGwin Jr G, Jackson GR, et al. Cone- and rod-mediated dark
80. Maurice DM. The structure and transparency of the cornea. J Physiol adaptation impairment in age-related maculopathy. Ophthalmology 2007;
1957;136:263–86. 114:1728–35.
81. Hassell JR, Birk DE. The molecular basis of corneal transparency. Exp Eye Res 115. Steinmetz RL, Haimovici R, Jubb C, et al. Symptomatic abnormalities of
2010;91:326–35. dark adaptation in patients with age-related Bruch’s membrane change.
82. Dohlman CH, Hedbys BO, Mishima S. The swelling pressure of the corneal Br J Ophthalmol 1993;77:549–54.
stroma. Invest Ophthalmol 1962;1:158–62. 116. Owsley C, McGwin Jr G, Jackson GR, et al. Effect of short-term, high-dose
83. Wiig H. Cornea fluid dynamics. I: measurement of hydrostatic and colloid retinol on dark adaptation in aging and early age-related maculopathy.
osmotic pressure in rabbits. Exp Eye Res 1989;49:1015–30. Invest Ophthalmol Vis Sci 2006;47:1310–8.
84. Moore DJ, Hussain AA, Marshall J. Age-related variation in the hydraulic 117. Green WR, Enger C. Age-related macular degeneration histopathologic
conductivity of Bruch’s membrane. Invest Ophthalmol Vis Sci 1995;36: studies: the 1992 Lorenz E. Zimmerman Lecture. Ophthalmology 1993;100:
1290–7. 1519–35.
85. Levick JR, Smaje LH. An analysis of the permeability of a fenestra. Microvasc 118. Khanifar AA, Koreishi AF, Izatt JA, et al. Drusen ultrastructure imaging with
Res 1987;33:233–56. spectral domain optical coherence tomography in age-related macular degen-
86. Navar PD, Navar LG. Relationship between colloid osmotic pressure and eration. Ophthalmology 2008;115:1883–90.
plasma protein concentration in the dog. Am J Physiol 1977;233:H295–8. 119. Klein R, Klein BEK, Linton KLP. Prevalence of age-related maculopathy.
87. Maepea O. Pressures in the anterior ciliary arteries, choroidal veins and cho- Ophthalmol 1992;99:933–43.
riocapillaris. Exp Eye Res 1992;54:731–6. 120. Friedman E, van Buskirk EM, Fineberg E, et al. Pathogenesis of senile disci-
88. Emi K, Pederson JE, Toris CB. Hydrostatic pressure of the suprachoroidal form degeneration of the macula. XXI Concilium Ophthalmologicum. Mexico:
space. Invest Ophthalmol Vis Sci 1989;30:233. Elsevier; 1971. p. 454–8.
89. Starita C, Hussain AA, Patmore A, et al. Localization of the site of major 121. Lengyel I, Tufail A, Hosaini HA, et al. Association of drusen deposition with
resistance to fluid transport in Bruch’s membrane. Invest Ophthalmol Vis Sci choroidal intercapillary pillars in the aging human eye. Invest Ophthalmol
1997;38:762–7. Vis Sci 2004;45:2886–92.
90. Overby D, Johnson M. Studies of depth-of-field effects in microscopy 122. Rudolf M, Clark ME, Chimento M, et al. Prevalence and morphology of druse
supported by numerical simulations. J Microscopy 2005;220:176–89. types in the macula and periphery of eyes with age-related maculopathy.
91. Fisher RF. The influence of age on some ocular basement membranes. Invest Ophthalmol Vis Sci 2008;49:1200–9.
Eye 1987;1:184–9. 123. Sarks SH. Council lecture: Drusen and their relationship to senile macular
92. Daniels BS, Hauser EB, Deen WN, et al. Glomerular basement membrane: in degeneration. Aust J Ophthalmol 1980;8:117–30.
vitro studies of water and protein permeability. Am J Physiol 1992;262: 124. Klein R, Davis MD, Magli YL, et al. The Wisconsin Age-Related Maculopathy
F919–F26. Grading System. Ophthalmol 1991;98:1128–34.
93. Renkin EM. Multiple pathways of capillary permeability. Circ Res 1977;41: 125. Davis MD, Gangnon RE, Lee LY, et al. The Age-Related Eye Disease Study
735–43. severity scale for age-related macular degeneration: AREDS report no. 17.
94. Hussain AA, Starita C, Marshall J. Transport characteristics of ageing human Arch Ophthalmol 2005;123:1484–98.
Bruch’s membrane: implications for age-related macular degeneration (AMD). 126. Klein R, Klein BE, Knudtson MD, et al. Fifteen-year cumulative incidence of
In: Ioseliani OR, editor. Focus on macular degeneration research. Hauppauge, age-related macular degeneration: the Beaver Dam Eye Study. Ophthalmology
New York: Nova Biomedical Books; 2004. 2007;114:253–62.
95. Starita C, Hussain AA, Pagliarini S, et al. Hydrodynamics of ageing Bruch’s 127. Querques G, Guigui B, Leveziel N, et al. Insights into pathology of cuticular
membrane: implications for macular disease. Exp Eye Res 1996;62: drusen from integrated confocal scanning laser ophthalmoscopy imaging and
565–72. corresponding spectral domain optical coherence tomography. Graefes Arch
96. McCarty WJ, Chimento MF, Curcio CA, et al. Effects of particulates and Clin Exp Ophthalmol 2011;249:1617–25.
lipids on the hydraulic conductivity of Matrigel. J Appl Physiol 2008;105: 128. Donders F. Beitrage zur pathologischen Anatomie des Auges. Arch Ophthal-
621–8. mol 1854;1:106–18.
97. Ethier CR, Johnson M, Ruberti J. Ocular biomechanics and biotransport. 129. Hageman GS, Mullins RF. Molecular composition of drusen as related to
Annu Rev Biomed Eng 2004;6:249–73. substructural phenotype. Mol Vis 1999;5:28.
130. Müller H. Anatomische Beiträge zur Ophthalmologie. Graefes Arch Clin Exp immunhistochemical analysis of AMD associated with basal deposits. Graefes
Ophthalmol 1856;2:1–69. Arch Clin Exp Ophthalmol 2008;246:803–10.
131. Oatman EL. Diagnostics of the fundus oculi. Troy, NY: Southworth; 1913. 163. Yamada Y, Ishibashi K, Ishibashi K, et al. The expression of advanced glyca-
480 132. Holloway TB, Verhoeff FH. Disc-like degeneration of the macula with micro- tion endproduct receptors in RPE cells associated with basal deposits in
scopic report concerning a tumor-like mass in the macular region. Trans Am human maculas. Exp Eye Res 2006;82:840–8.
Ophthalmol Soc 1928;26:206–28. 164. Löffler KU, Lee WR. Basal linear deposit in the human macula. Graefes Arch
133. Lauber H. The origin of hyaline formations within the eye. Ber ü d deutsch Clin Exp Ophthalmol 1986;224:493–501.
ophth Gesellsch 1924;44:216. 165. Marshall GE, Konstas AGP, Reid GG, et al. Type IV collagen and laminin
Section 1

134. Rones B. Formation of drusen of the lamina vitrea. Arch Ophthalmol 1937;18: in Bruch’s membrane and basal linear deposit in the human macula. Br J
288–402. Ophthalmol 1992;76:607–14.
135. Wolter JR, Falls HF. Bilateral confluent drusen. Arch Ophthalmol 1962;68: 166. Knupp C, Munro PM, Luther PK, et al. Structure of abnormal molecular
219–26. assemblies (collagen VI) associated with human full thickness macular holes.
136. Luibl V, Isas JM, Kayed R, et al. Drusen deposits associated with aging and J Struct Biol 2000;129:38–47.
age-related macular degeneration contain nonfibrillar amyloid oligomers. 167. Reale E, Groos S, Eckardt U, et al. New components of “basal laminar depos-
J Clin Invest 2006;116:378–85. its” in age-related macular degeneration. Cells, Tissues, Organs 2008;190:
137. Li C-M, Clark M, Rudolf M, et al. Distribution and composition of esterified 170–81.
Anatomy and Physiology

and unesterified cholesterol in extra-macular drusen. Exp Eye Res 2007;85: 168. Marmorstein LY, McLaughlin PJ, Peachey NS, et al. Formation and progres-
Basic Science and Translation to Therapy

192–201. sion of sub-retinal pigment epithelium deposits in Efemp1 mutation knock-in


138. Lengyel I, Flinn JM, Peto T, et al. High concentration of zinc in sub-retinal mice: a model for the early pathogenic course of macular degeneration. Hum
pigment epithelial deposits. Exp Eye Res 2007;84:772–80. Mol Genet 2007;16:2423–32.
139. Malek G, Li C-M, Guidry C, et al. Apolipoprotein B in cholesterol-containing 169. Rudolf M, Malek G, Messinger JD, et al. Sub-retinal drusenoid deposits in
drusen and basal deposits in eyes with age-related maculopathy. Am J Pathol human retina: organization and composition. Exp Eye Res 2008;87:402–8.
2003;162:413–25. 170. Smith RT, Sohrab MA, Busuioc M, et al. Reticular macular disease. Am J
140. Farkas TG, Sylvester V, Archer D, et al. The histochemistry of drusen. Ophthalmol 2009;148:733–43 e2.
Am J Ophthalmol 1971;71:1206–15. 171. Zweifel SA, Spaide RF, Curcio CA, et al. Reticular pseudodrusen are subreti-
141. Pauleikhoff D, Zuels S, Sheraidah GS, et al. Correlation between biochemical nal drusenoid deposits. Ophthalmology 2010;117:303–12.e.1.
composition and fluorescein binding of deposits in Bruch’s membrane. 172. Zweifel SA, Imamura Y, Spaide TC, et al. Prevalence and significance of
Ophthalmol 1992;99:1548–53. subretinal drusenoid deposits (reticular pseudodrusen) in age-related macular
142. Curcio CA, Presley JB, Medeiros NE, et al. Esterified and unesterified choles- degeneration. Ophthalmology 2010;117:1775–81.
terol in drusen and basal deposits of eyes with age-related maculopathy. 173. Schmitz-Valckenberg S, Alten F, Steinberg JS, et al. Reticular drusen
Exp Eye Res 2005;81:731–41. associated with geographic atrophy in age-related macular degeneration.
143. Wang L, Clark ME, Crossman DK, et al. Abundant lipid and protein compo- Invest Ophthalmol Vis Sci 2011;52:5009–15.
nents of drusen. PLoS ONE 2010;5:e10329. 174. Spaide RF, Curcio CA. Drusen characterization with multimodal imaging.
144. Guyton JR, Klemp KF. The lipid-rich core region of human atherosclerotic Retina 2010;30:1441–54.
fibrous plaques. Prevalence of small lipid droplets and vesicles by electron 175. Xu H, Chen M, Manivannan A, et al. Age-dependent accumulation of
microscopy. Am J Pathol 1989;134:705–17. lipofuscin in perivascular and subretinal microglia in experimental mice.
145. Friedman DA, Parker JS, Kimble JA, et al. Fluorescein localization in drusen Aging Cell 2008;7:58–68.
associated with age-related macular degeneration. Retina 2012;32:19–24. 176. Ma W, Zhao L, Fontainhas AM, et al. Microglia in the mouse retina alter the
146. Hageman GS, Mullins RG, Russell SR, et al. Vitronectin is a constituent of structure and function of retinal pigmented epithelial cells: a potential cellular
ocular drusen and the vitronectin gene is expressed in human retinal pig- interaction relevant to AMD. PLoS ONE 2009;4:e7945.
mented epithelial cells. FASEB J 1999;13:477–84. 177. Verhoeff FH, Grossman HP. The pathogenesis of disciform degeneration of
147. Fariss RN, Apte SS, Olsen BR, et al. Tissue inhibitor of metalloproteinases-3 the macula. Trans Am Ophthalmol Soc 1937;35:262–94.
is a component of Bruch’s membrane of the eye. Am J Pathol 1997;150: 178. Friedman E. The role of the atherosclerotic process in the pathogenesis of
323–8. age-related macular degeneration. Am J Ophthalmol 2000;130:658–63.
148. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the 179. Williams KJ, Tabas I. The response-to-retention hypothesis of early atherogen-
complement regulatory gene factor H (HF1/CFH) predisposes individuals to esis. Arterioscler Thromb Vasc Biol 1995;15:551–61.
age-related macular degeneration. Proc Natl Acad Sci U S A 2005;102: 180. Tabas I, Williams KJ, Boren J. Subendothelial lipoprotein retention as the
7227–32. initiating process in atherosclerosis: update and therapeutic implications.
149. Johnson LV, Leitner WP, Rivest AJ, et al. The Alzheimer’s Aβ-peptide is Circulation 2007;116:1832–44.
deposited at sites of complement activation in pathologic deposits associated 181. Grossniklaus HE, Green WR. Choroidal neovascularization. Am J Ophthalmol
with aging and age-related macular degeneration. Proc Natl Acad Sci U S A 2004;137:496–503.
2002;99:11830–5. 182. Yannuzzi LA, Freund KB, Takahashi BS. Review of retinal angiomatous pro-
150. Nakata K, Crabb JW, Hollyfield JG. Crystallin distribution in Bruch’s liferation or type 3 neovascularization. Retina 2008;28:375–84.
membrane–choroid complex from AMD and age-matched donor eyes. 183. Klein ML, Wilson DJ. Clinicopathologic correlation of choroidal and retinal
Exp Eye Res 2005;80:821–6. neovascular lesions in age-related macular degeneration. Am J Ophthalmol
151. Klaver CC, Kliffen M, van Duijn CM, et al. Genetic association of apolipopro- 2011;151:161–9.
tein E with age-related macular degeneration. Am J Hum Genet 1998;63: 184. Schwesinger C, Yee C, Rohan RM, et al. Intrachoroidal neovascularization in
200–6. transgenic mice overexpressing vascular endothelial growth factor in the
152. Anderson DH, Ozaki S, Nealon M, et al. Local cellular sources of apolipopro- retinal pigment epithelium. Am J Pathol 2001;158:1161–72.
tein E in the human retina and retinal pigmented epithelium: implications for 185. Spraul CW, Grossniklaus HE. Characteristics of drusen and Bruch’s
the process of drusen formation. Am J Ophthalmol 2001;131:767–81. membrane in postmortem eyes with age-related macular degeneration.
153. Li C-M, Chung BH, Presley JB, et al. Lipoprotein-like particles and Arch Ophthalmol 1997;115:267–73.
cholesteryl esters in human Bruch’s membrane: initial characterization. Invest 186. Vogt SD, Curcio CA, Wang L, et al. Retinal pigment epithelial expression of
Ophthalmol Vis Sci 2005;46:2576–86. complement regulator CD46 is altered early in the course of geographic
154. Li C-M, Clark ME, Chimento MF, et al. Apolipoprotein localization in isolated atrophy. Exp Eye Res 2011;93:413–23.
drusen and retinal apolipoprotein gene expression. Invest Ophthalmol Vis Sci 187. Spaide RF, Armstrong D, Browne R. Continuing medical education review:
2006;47:3119–28. choroidal neovascularization in age-related macular degeneration – what is
155. Sarks JP, Sarks SH, Killingsworth MC. Evolution of geographic atrophy of the the cause? Retina 2003;23:595–614.
retinal pigment epithelium. Eye 1988;2:552–77. 188. Jampol LM, Acheson R, Eagle RC, et al. Calcification of Bruch’s membrane in
156. Sarks S, Cherepanoff S, Killingsworth M, et al. Relationship of basal laminar angioid streaks with homozygous sickle cell disease. Arch Ophthalmol
deposit and membranous debris to the clinical presentation of early age- 1987;105:93–8.
related macular degeneration. Invest Ophthalmol Vis Sci 2007;48:968–77. 189. Bergen AA, Plomp AS, Schuurman EJ, et al. Mutations in ABCC6 cause pseu-
157. Curcio CA, Presley JB, Millican CL, et al. Basal deposits and drusen in eyes doxanthoma elasticum. Nat Genet 2000;25:228–31.
with age-related maculopathy: evidence for solid lipid particles. Exp Eye Res 190. Zweifel SA, Imamura Y, Freund KB, et al. Multimodal fundus imaging of
2005;80:761–75. pseudoxanthoma elasticum. Retina 2011;31:482–91.
158. Curcio CA, Millican CL. Basal linear deposit and large drusen are specific for 191. Uitto J, Bercovitch L, Terry SF, et al. Pseudoxanthoma elasticum: progress
early age-related maculopathy. Arch Ophthalmol 1999;117:329–39. in diagnostics and research towards treatment. Summary of the 2010 PXE
159. Sarks SH, van Driel D, Maxwell L, et al. Softening of drusen and subretinal International Research Meeting. Am J Med Genet Part A 2011;155:1517–26.
neovascularization. Trans Ophthalmol Soc U K 1980;100:414–22. 192. Runge P, Müller DP, McAllister J, et al. Oral vitamin E supplements can
160. Sarks JP, Sarks SH, Killingsworth MC. Morphology of early choroidal neovas- prevent the retinopathy of abetalipoproteinaemia. Br J Ophthalmol 1986;
cularization in age-related macular degeneration: correlation with activity. 70:166–73.
Eye 1997;11:515–22. 193. Duker JS, Belmont J, Bosley TM. Angioid streaks associated with abetalipo-
161. Messinger JD, Johnson M, Medeiros NE, et al. Transition from lipid wall to proteinemia. Case report. Arch Ophthalmol 1987;105:1173–4.
basal linear deposit in age-related maculopathy (ARM). Invest Ophthalmol 194. Dieckert J, White M, Christmann L, et al. Angioid streaks associated with
Vis Sci 2009; 50:4933. abetalipoproteinemia. Ann Ophthalmol 1989;21:172–5.
162. Lommatzsch A, Hermans P, Müller KD, et al. Are low inflammatory reactions 195. Gorin MB, Paul TO, Rader DJ. Angioid streaks associated with abetalipopro-
involved in exudative age-related macular degeneration? Morphological and teinemia. Ophthalmic Genet 1994;15:151–9.
196. Weber BHF, Vogt G, Pruett RC, et al. Mutations in the tissue inhibitor of 223. Bhutto IA, Uno K, Merges C, et al. Reduction of endogenous angiogenesis
metalloproteinases-3 (TIMP3) in patients with Sorsby’s fundus dystrophy. inhibitors in Bruch’s membrane of the submacular region in eyes with
Nat Genet 1994;8:352–65. age-related macular degeneration. Arch Ophthalmol 2008;126:670–8.
197. Hayward C, Shu X, Cideciyan AV, et al. Mutation in a short-chain collagen 224. Lin MY, Kochounian H, Moore RE, et al. Deposition of exon-skipping splice 481
gene, CTRP5, results in extracellular deposit formation in late-onset retinal isoform of human retinal G protein-coupled receptor from retinal pigment
degeneration: a genetic model for age-related macular degeneration. epithelium into Bruch’s membrane. Mol Vis 2007;13:1203–14.
Hum Mol Genet 2003;12:2657–67. 225. Handa JT, Verzijl N, Matsunaga H, et al. Increase in the advanced glycation

Chapter 20
198. Stone E, Lotery A, Munier F, et al. A single EFEMP1 mutation associated with end product pentosidine in Bruch’s membrane with age. Invest Ophthalmol
both malattia leventinese and Doyne honeycomb retinal dystrophy. Vis Sci 1999;40:775–9.
Nat Genet 1999;22:199–202. 226. Glenn JV, Beattie JR, Barrett L, et al. Confocal Raman microscopy can quantify
199. Jacobson SG, Cideciyan AV, Regunath G, et al. Night blindness in Sorsby’s advanced glycation end product (AGE) modifications in Bruch’s membrane
fundus dystrophy reversed by vitamin A. Nat Genet 1995;11:27–32. leading to accurate, nondestructive prediction of ocular aging. FASEB J
200. Jacobson SG, Cideciyan AV, Wright E, et al. Phenotypic marker for 2007;21:3542–52.
early disease detection in dominant late-onset retinal degeneration. Invest 227. Johnson PT, Betts KE, Radeke MJ, et al. Individuals homozygous for the age-
Ophthalmol Vis Sci 2001;42:1882–90. related macular degeneration risk-conferring variant of complement factor H
201. Michaelides M, Jenkins SA, Brantley Jr MA, et al. Maculopathy due to the have elevated levels of CRP in the choroid. Proc Natl Acad Sci U S A
R345W substitution in fibulin-3: distinct clinical features, disease variability, 2006;103:17456–61.

Structure, Function, and Pathology of Bruch’s Membrane


and extent of retinal dysfunction. Invest Ophthalmol Vis Sci 2006;47:3085–97. 228. Spaide R, Ho-Spaide W, Browne R, et al. Characterization of peroxidized
202. Isashiki Y, Tabata Y, Kamimura K, et al. Sorsby’s fundus dystrophy in two lipids in Bruch’s membrane. Retina 1999;19:141–7.
Japanese families with unusual clinical features. Jpn J Ophthalmol 229. Moreira EF, Larrayoz IM, Lee JW, et al. 7-Ketocholesterol is present in lipid
1999;43:472–80. deposits in the primate retina: potential implication in the induction of VEGF
203. Capon MRC, Marshall J, Krafft JI, et al. Sorsby’s fundus dystrophy. A light and CNV formation. Invest Ophthalmol Vis Sci 2009;50:523–32.
and electron microscopic study. Ophthalmology 1989;96:1769–77. 230. Vranka JA, Johnson E, Zhu X, et al. Discrete expression and distribution
204. Kuntz CA, Jacobson SG, Cideciyan AV, et al. Sub-retinal pigment epithelial pattern of TIMP-3 in the human retina and choroid. Curr Eye Res
deposits in a dominant late-onset retinal degeneration. Invest Ophthalmol Vis 1997;16:102–10.
Sci 1996;37:1772–82. 231. Hughes BA, Miller SS, Machen TE. Effects of cyclic AMP on fluid absorption
205. Milam AH, Curcio CA, Cideciyan AV, et al. Dominant late-onset retinal and ion transport across frog retinal pigment epithelium. Measurements in
degeneration with regional variation of sub-RPE deposits, retinal function, the open-circuit state. J Gen Physiol 1984;83:875–99.
and photoreceptor degeneration. Ophthalmology 2000;107:2256–66. 232. Frambach DA, Weiter JJ, Adler AJ. A photogrammetric method to measure
206. Curcio CA, Mayne R, Ninomiya H. Localization of type IV collagen chains in fluid movement across isolated frog retinal pigment epithelium. Biophys J
AMD eyes. 1999. Unpublished observations. 1985;47:547–52.
207. Knupp C, Chong NH, Munro PM, et al. Analysis of the collagen VI assemblies 233. Negi A, Marmor MF. Quantitative estimation of metabolic transport of
associated with Sorsby’s fundus dystrophy. J Struct Biol 2002;137:31–40. subretinal fluid. Invest Ophthalmol Vis Sci 1986;27:1564–8.
208. Bhutto IA, Kim SY, McLeod DS, et al. Localization of collagen XVIII and the 234. Marmor MF, Negi A. Pharmacologic modification of subretinal fluid absorp-
endostatin portion of collagen XVIII in aged human control eyes and eyes with tion in the rabbit eye. Arch Ophthalmol 1986;104:1674–7.
age-related macular degeneration. Invest Ophthalmol Vis Sci 2004;45:1544–52. 235. Tsuboi S. Measurement of the volume flow and hydraulic conductivity across
209. Marmorstein LY, Munier FL, Arsenijevic Y, et al. Aberrant accumulation of the isolated dog retinal pigment epithelium. Invest Ophthalmol Vis Sci
EFEMP1 underlies drusen formation in malattia leventinese and age-related 1987;28:1776–82.
macular degeneration. Proc Natl Acad Sci U S A 2002;99:13067–72. 236. Cantrill HL, Pederson JE. Experimental retinal detachment: VI. The
210. Das A, Frank RN, Zhang NL, et al. Ultrastructural localization of extracellular permeability of the blood–retinal barrier. Arch Ophthalmol 1984;102:
matrix components in human retinal vessels and Bruch’s membrane. 747–51.
Arch Ophthalmol 1990;108:421–9. 237. Pederson JE, Cantrill HL. Experimental retinal detachment: V. Fluid move-
211. Lin WL, Essner E, McCarthy KJ, et al. Ultrastructural immunocytochemical ment through the retinal hole. Arch Ophthalmol 1984;102:136–9.
localization of chondroitin sulfate proteoglycan in Bruch’s membrane of the 238. Chihara E, Nao-i N. Resorption of subretinal fluid by transepithelial flow of
rat. Invest Ophthalmol Vis Sci 1992;33:2072–5. the retinal pigment epithelium. Graefes Arch Clin Exp Ophthalmol
212. Curcio C, Millican C, Kruth H. Cholesterol accumulates with age in human 1985;223:202–4.
Bruch’s membrane. Invest Ophthalmol Vis Sci 2000;41:S115. 239. Anderson DH, Talaga KC, Rivest AJ, et al. Characterization of beta amyloid
213. Liu A, Chang J, Lin Y, et al. Long-chain and very long-chain polyunsaturated assemblies in drusen: the deposits associated with aging and age-related
fatty acids in ocular aging and age-related macular degeneration. J Lipid Res macular degeneration. Exp Eye Res 2004;78:243–56.
2011;51:3217–29. 240. Malek G, Johnson LV, Mace BE, et al. Apolipoprotein E allele-dependent
214. Tezel TH, Geng L, Lato EB, et al. Synthesis and secretion of hemoglobin by pathogenesis: A model for age-related retinal degeneration. Proc Natl Acad
retinal pigment epithelium. Invest Ophthalmol Vis Sci 2009;50:1911–9. Sci U S A 2005;102:11900–5.
215. Sakaguchi H, Miyagi M, Shadrach KG, et al. Clusterin is present in drusen in 241. Hageman GS, Luthert PJ, Chong NHC, et al. An integrated hypothesis that
age-related macular degeneration. Exp Eye Res 2002;74:547–9. considers drusen as biomarkers of immune-mediated processes at the RPE–
216. Chong NH, Keonin J, Luthert PJ, et al. Decreased thickness and integrity of Bruch’s membrane interface in aging and age-related macular degeneration.
the macular elastic layer of Bruch’s membrane correspond to the distribution Progr Ret Eye Res 2001;20:705–32.
of lesions associated with age-related macular degeneration. Am J Pathol 242. Dentchev T, Milam AH, Lee VM, et al. Amyloid-beta is found in drusen from
2005;166:241–51. some age-related macular degeneration retinas, but not in drusen from
217. Mullins RF, Olvera MA, Clark AF, et al. Fibulin-5 distribution in human eyes: normal retinas. Mol Vis 2003;9:184–90.
relevance to age-related macular degeneration. Exp Eye Res 2007;84:378–80. 243. Anderson DH, Radeke MJ, Gallo NB, et al. The pivotal role of the complement
218. Yamada Y, Tian J, Yang Y, et al. Oxidized low density lipoproteins induce a system in aging and age-related macular degeneration: hypothesis re-visited.
pathologic response by retinal pigmented epithelial cells. J Neurochem Prog Retin Eye Res 2010;29:95–112.
2008;105:1187–97. 244. Kochounian H, Johnson LV, Fong HK. Accumulation of extracellular RGR-d
219. Kamei M, Hollyfield JG. TIMP-3 in Bruch’s membrane: changes during aging in Bruch’s membrane and close association with drusen at intercapillary
and in age-related macular degeneration. Invest Ophthalmol Vis Sci regions. Exp Eye Res 2009;88:1129–36.
1999;40:2367–75. 245. Loeffler KU, Mangini NJ. Immunolocalization of ubiquitin and related
220. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin enzymes in human retina and retinal pigment epithelium. Graefes Arch Clin
Eye Res 2009;28:348–68. Exp Ophthalmol 1997;235:248–54.
221. Glenn JV, Mahaffy H, Wu K, et al. Advanced glycation end product (AGE) 246. Wang AL, Lukas TJ, Yuan M, et al. Autophagy and exosomes in the aged
accumulation on Bruch’s membrane: links to age-related RPE dysfunction. retinal pigment epithelium: possible relevance to drusen formation and
Invest Ophthalmol Vis Sci 2009;50:441–51. age-related macular degeneration. PLoS ONE 2009;4:e4160.
222. Guo L, Hussain AA, Limb GA, et al. Age-dependent variation in metallopro- 247. Gouras P, Braun K, Ivert L, et al. Bestrophin detected in the basal membrane
teinase activity of isolated human Bruch’s membrane and choroid. of the retinal epithelium and drusen of monkeys with drusenoid maculopa-
Invest Ophthalmol Vis Sci 1999;40:2676–82. thy. Graefes Arch Clin Exp Ophthalmol 2009;247:1051–6.
Section 1 Anatomy and Physiology

Chapter Vitreous and Vitreoretinal Interface


21  Jerry Sebag, W. Richard Green (posthumously)†

BIOCHEMISTRY OF VITREOUS minor collagens of vitreous is type XVIII, progenitor of end-


ostatin, a potent inhibitor of angiogenesis.11
Vitreous is 98% water and 2% structural proteins, extracellular
matrix components, and miscellaneous compounds. Hyaluronan
Hyaluronan (HA) was first isolated from bovine vitreous in
Collagen 1937. HA appears after birth, perhaps synthesized by hyalo-
Collagen is the major structural protein, consisting of hetero- cytes,2 the ciliary body, and/or Müller cells. It is a large polyan-
typic fibrils (Fig. 21.1) similar to cartilage.1,2 Following vitrec- ion, which can influence the diffusion of drugs through
tomy, a type II procollagen is secreted in humans,3 but vitreous vitreous.12,13 As a result of HA’s entanglement within the vitre-
is not reformed, since reoperations reveal that the gel state of ous collagen fibril matrix, the mechanical force of HA’s exten-
vitreous is not re-established. sion and contraction can be transmitted to the retina, optic disc,
Type II collagen4 comprises 75% of the total collagen content and neovascular complexes, inducing untoward effects in condi-
in vitreous. There are considerable similarities between vitre- tions with fluctuations in ionic balance and hydration, such as
ous and cartilage collagens,5,6 perhaps explaining why inborn diabetes.14
errors of type II collagen metabolism result in “arthro-
ophthalmopathies,”7 manifesting similar phenotypic expression Chondroitin sulfate
in joints and vitreous. Type IX collagen accounts for up to Most vitreous chondroitin sulfate is in the form of versican,15
15% of vitreous collagen,8 where it always contains a chon- believed to form complexes with HA as well as with microfibril-
droitin sulfate glycosaminoglycan chain9 covalently linked to lar proteins such as fibulin-1 and fibullin-2 and play a crucial role
the α2 (IX) chain at the NC3 domain, enabling the molecule
to assume a proteoglycan form. †
 W. Richard Green, MD has been first author of this chapter for all previous
An important function of vitreous is maintaining transparency editions of this book. His monumental contributions to this field are reflected in
the content of this chapter, which will continue to teach long after his parting on
within the eye (Fig. 21.2).2 Studies10 have shown that one of the July 5, 2010.

Fig. 21.1 Fibrillar structure of human vitreous


Chondroitin sulfate collagen. Schematic diagram of the major
glycosminoglycan chain of heterotypic collagen fibrils of vitreous based
type IX collagen upon the current knowledge of the structure
and biophysical attributes of the constituent
molecules. (Reproduced with permission from
Bishop P. The biochemical structure of
N - propeptide of mammalian vitreous. Eye 1996;10:64.)
type V/XI collagen

Type V/XI collagen

Type IX collagen
N - propeptide of
type II collagen

Type II collagen
in maintaining the molecular morphology of vitreous.16 Muta- Opticin
tions that alter the splicing of the central chondroitin sulfate- A major noncollagenous protein of vitreous is opticin (formerly
bearing domains of versican have been implicated in Wagner vitrican).19 It is bound to the surface of the heterotypic collagen 483
syndrome, a condition with excess vitreous liquefaction.17 fibrils and prevents aggregation of adjacent collagen fibrils into
bundles. Opticin binds heparan and chondroitin sulfates, sug-
Noncollagenous structural proteins

Chapter 21
gesting that opticin may play a role in vitreoretinal adhesion.20,21
Fibrillins Similar to its role in articular cartilage,22 opticin may also stabi-
In Marfan syndrome defects in the gene encoding fibrillin-1 lize vitreous gel structure by binding chondroitin sulfate
(FBN1 on chromosome 15q21) result in both ectopia lentis and proteoglycans.
vitreous liquefaction,18 important in rhegmatogenous retinal
detachment (RD) in these patients. ANATOMY AND HISTOLOGY

Vitreous and Vitreoretinal Interface


Vitreous body
Vitreous is a clear gel-like structure with a volume of 4.0 mL.
During invagination of the optic vesicle the “primary” vitreous
forms between the lens and the internal limiting lamina (ILL on
Fig. 21.3A) of the retina. It is noteworthy that the ILL is continu-
ous with Bruch’s membrane, demonstrating a common embryo-
logic origin with analogous molecular composition and structure,
suggesting important similarities later in life.23 The “secondary”
vitreous begins to develop at the 13-mm stage of embryogenesis
and is derived from the retina and mesoderm of the hyaloid
vascular system (Fig. 21.3B).
Classic depictions of human vitreous structure are shown in
Fig. 21.4. Modern concepts proposed membranous (Fig. 21.5A)24
and cisternal (Fig. 21.5)25 systems. Sebag and Balazs26 performed
dark-field slit microscopy to define the posterior vitreous cortex
as a thin, membranous structure continuous from the ora serrata
to the posterior pole. Two round holes are present in the pre-
papillary and premacular areas (Fig. 21.6). Anteroposterior
Fig. 21.2 Dissected human vitreous. Vitreous body from a 9-month-old
girl dissected from the sclera, choroid, and retina. The vitreous body is fibers (Fig. 21.7) comprised of parallel collagen fibrils (Fig.
attached to the anterior segment and the specimen is placed on a 21.8) arise from the vitreous base (Fig. 21.9A), where Gartner27
surgical towel in room air. The exquisite gel state of the vitreous body found “lateral aggregation” in older individuals. Vitreous base
and the crystal-clear transparency are evident. (Specimen courtesy of
collagen fibers insert anterior to the ora serrata forming the
the New England Eye Bank; originally published as cover photo in
Sebag J. The vitreous: structure, function, and pathobiology. New York: anterior loop (Fig. 21.9B), important in anterior proliferative
Springer-Verlag, 1989.) vitreoretinopathy (PVR).28 In the posterior pole, fibers extend

ILL

s
ch’
Bru

A B

Fig. 21.3 Fetal human vitreous. (A) Human embryo eye stained with immunofluorescent anti-ABA (basement membrane lectin) antibodies revealing
the continuity of the internal limiting lamina (ILL) and Bruch’s membrane, demonstrating a common embryologic origin with an analogous molecular
composition and structure. This suggests similarities during aging and as participants in various disease processes, especially those that feature
cell migration and proliferation: biologic processes that will occur along these interfaces. (Reproduced with permission from Sebag J, Hageman GS.
Interfaces. Eur J Ophthalmol 2000;10:1.) (B) Photomicrograph of human fetal eye aged 13 gestational weeks. A prominent hyaloid artery and vasa
hyaloidea propria can be seen arising from the optic nerve and extending anteriorly towards the lens to anastomose with the tunica vasculosa
lentis. (Hematoxylin and eosin; bar = 100 µm.) (Courtesy of Kenneth M.P. Yee and Fred Ross-Cisneros, University of Southern California.)
Fig. 21.4 Classic depictions of vitreous structure. (A) Schematic
representation of vitreous structure. (Redrawn with permission from
Green WR. Pathology of the vitreous. In: Frayer WC (ed.) Lancaster
Weigert's ligament course in ophthalmic histopathology, unit 8. Philadelphia: FA Davis,
Berger's space 1981.) (B) Schematic diagram of vitreous structure with classic
484 nomenclature of internal and interface structures. (Reproduced with
Cloquet's canal permission from Sang DN. Embryology of the vitreous. Congenital and
developmental abnormalities. In: Schepens CL, Neetens A, editors.
The vitreous and vitreoretinal interface. New York: Springer-Verlag;
Section 1

Vitreous base 1987, p. 20.)


Anatomy and Physiology
Basic Science and Translation to Therapy

Peripapillary

A Fovea

Vitreous base
Berger’s space Pars plicata
(retrolental space
Pars plana
of Erggelet)
Ora serrata
Sclera
Choroid
Retina
Canal of Hannover
Cloquet’s canal

Secondary vitreous

Area of Martegiani

Egger’s line forming Anterior hyaloid


Wieger’s ligament Canal of Petit
B (hyaloideocapsular li.)

OS
LM TP
LC
TM
LR
TC

TR

A B

Fig. 21.5 Vitreous structure. (A) Membranelles, called tractae, course from the area of the ciliary body to the posterior pole. OS, ora serrata; LM,
ligamentum medianum; LC, ligamentum coronarium; LR, ligamentum retrolentalis; TP, tractus preretinalis; TM, tractus medianus; TC, tractus
coronarius; TR, tractus retrolentalis. (Reproduced with permission from Eisner G. Clinical anatomy of the vitreous. In: Duane TD, Jaeger EA,
editors. Biomedical foundations of ophthalmology, vol. 1. Philadelphia: JB Lippincott; 1984, p. 21.) (B) Cisterns are identified by injecting with
colored India ink. Light brown, Cloquet’s canal opening into the prepapillary area of Martegiani; light purple, equatorial cistern opening into the
bursa premacularis. (Reproduced with permission from Jongbloed WL, Worst JGF. The cisternal anatomy of the vitreous body. Doc Ophthalmol
1987;67:183–96.)
Fig. 21.6 The two “holes” in the posterior vitreous cortex correspond to
the prepapillary (smaller, to left) and premacular (larger “hole,” to right)
regions. Vitreous can be seen extruding through these holes. The
bright pinpoint foci of intense light-scattering are hyalocytes embedded 485
in the posterior vitreous cortex. (Reproduced with permission from
Sebag J, Balazs EA. Human vitreous fibres and vitreoretinal disease.
Trans Ophthalmol Soc UK 1984;104:123–8.)

Chapter 21
Vitreous and Vitreoretinal Interface
Fig. 21.7 Human vitreous morphology.
Human vitreous structure visualized by
dark-field slit illumination. All photographs are
oriented with the anterior segment below and
the posterior pole above. (A) Posterior vitreous
in the left eye of a 52-year-old man. The
vitreous body is enclosed by the vitreous
cortex. There is a hole in the prepapillary
A (small, to the left) vitreous cortex. Vitreous
B fibers are oriented toward the premacular
region. (B) Posterior vitreous in a 57-year-old
man. A large bundle of prominent fibers is seen
coursing anteroposteriorly and entering the
retrocortical space by way of the premacular
vitreous cortex. (C) Same photograph as B,
at higher magnification. (D) Posterior vitreous
in the right eye of a 53-year-old woman. There
is posterior extrusion of vitreous out of the
prepapillary hole (to the right) and premacular
(large extrusion to the left) vitreous cortex.
Fibers course anteroposteriorly out into the
retrocortical space. (E) Horizontal optical
section of the same specimen as D, at a
different level. A large fiber courses posteriorly
C D from the central vitreous and inserts into the
premacular vitreous cortex. (F) Same view as
E, at higher magnification. The large fiber has
a curvilinear appearance because of traction
by the vitreous extruding into the retrocortical
space (see D). However, because of its
attachment to the posterior vitreous cortex, the
fiber arcs back to its point of insertion. (G)
Anterior and central vitreous in a 33-year-old
woman. Cloquet’s canal is seen forming the
retrolental space of Berger. (H) Anterior and
peripheral vitreous in a 57-year-old man. The
specimen is tilted forward to enable
visualization of the posterior aspect of the lens
E F and the peripheral anterior vitreous. To the
right of the lens there are fibers coursing
anteroposteriorly that insert into the vitreous
base. These fibers “splay out” to insert anterior
and posterior to the ora serrata. (A, E, and F
reproduced with permission from Sebag J,
Balazs EA. Pathogenesis of CME: anatomic
consideration of vitreoretinal adhesions. Surv
Ophthalmol 1984;28 (Suppl):493. B, C from
Sebag J, Balazs EA. Morphology and
ultrastructure of human vitreous fibers. Invest
Ophthalmol Vis Sci 1989;30:187. D, G, and H
from Sebag J. The vitreous: structure, function,
and pathobiology. New York, Springer-Verlag,
1989. Specimens were courtesy of the New
York Bank for Sight and Restoration, New
G H York, NY.)
through the premacular hole (Figs 21.6 and 21.7A), but a few is thin over the macula. The prepapillary hole can sometimes be
attach to the rim of the hole. Condensed bundles of fibers visualized clinically following posterior vitreous detachment
486 insert into the vitreous cortex in the midperiphery and equator (PVD). If peripapillary tissue is torn away during PVD and
(Fig. 21.10). remains attached around the prepapillary hole, it is called Vogt’s
or Weiss’s ring. Gupta et al.30 demonstrated a lamellar organiza-
Vitreoretinal interface tion of the posterior vitreous cortex (Fig. 21.12), confirmed in
Section 1

The equatorial and posterior vitreoretinal interfaces consist of humans by three-dimensional optical coherence tomography
the posterior vitreous cortex, the ILL of the retina, and an inter- (OCT) (Fig. 21.13).31 During anomalous PVD (APVD)32 these pre-
vening extracellular matrix. dispose to splitting along potential cleavage planes, resulting in
vitreoschisis.33
Posterior vitreous cortex
The posterior vitreous cortex is 100–110 µm thick and consists of Hyalocytes
Anatomy and Physiology
Basic Science and Translation to Therapy

densely packed collagen fibrils29 (Fig. 21.11). There is no vitreous Hyalocytes are mononuclear cells embedded in the posterior
cortex over the optic disc (Figs 21.6 and 21.7A), and the cortex vitreous cortex 20–50  µm from the ILL posteriorly (Figs 21.6

Fig. 21.8 Ultrastructure of human vitreous fiber. The fibers of the


human vitreous visible by dark-field microscopy result from bundles of Fig. 21.10 Fibers condense into bundles and insert into the vitreous
parallel collagen fibrils such as the one shown here in cross-section cortex (arrows). Between these insertions are spaces seemingly
(arrow). (Reproduced with permission from Sebag J, Balazs EA. devoid of structure, but probably filled with liquid vitreous. (Reproduced
Morphology and ultrastructure of human vitreous fibers. Invest with permission from Sebag J, Balazs EA. Human vitreous fibres and
Ophthalmol Vis Sci 1989;30:187.) vitreoretinal disease. Trans Ophthalmol Soc UK 1984;104:123–8.)

A
B

Fig. 21.9A Vitreous base morphology. In this eye of a 58-year-old woman, postmortem dark-field slit microscopy studies showed vitreous fibers
that are continuous from the posterior vitreous cortex (at the top of the photo) to the vitreous base, where they “splay out” to insert into the
vitreous base (arrow). (Reproduced with permission from Sebag J, Balazs EA. Pathogenesis of CME: anatomic consideration of vitreoretinal
adhesions. Surv Ophthalmol 1984;28 (Suppl):493–8.)
9B Anterior loop of the vitreous base. Central, anterior, and peripheral vitreous structure in a 76-year-old man. The posterior aspect of the lens is
seen below. Fibers course anteroposteriorly in the central vitreous and insert into the vitreous base. The “anterior loop” configuration of those
fibers inserting into the vitreous base anterior to the ora serrata is seen on the right side of the specimen (arrow).
and 21.14). The highest density of hyalocytes is in the vitreous
base followed by the posterior pole, with the lowest density
at the equator.34,35 Balazs36 suggested that these cells synthesize 487
vitreous HA,37–40 but Swann5 disagreed. Evidence suggests that
hyalocytes maintain ongoing synthesis and metabolism of gly-

Chapter 21
coproteins41,42 and may also synthesize collagen43 and enzymes.44
The phagocytic capacity of hyalocytes has been described in
vivo45 and demonstrated in vitro.46–48 Hyalocytes become phago-
cytic in response to inducting stimuli and are important in
antigen processing and as initiators of the immune response,
making possible intravitreal inoculation of antigens to promote
systemic immunity.49 HA may have a regulatory effect on hya-

Vitreous and Vitreoretinal Interface


locyte phagocytic activity.50,51 Various constituents of vitreous52,53
may be immunogenic and play a role in ocular inflammatory

Fig. 21.12 Lamellar structure of the posterior vitreous cortex.


Immunohistochemistry with anti-ABA lectin antibodies of the monkey
20 KV X 2000 10 U 003 191 ERI vitreoretinal interface demonstrates lamellae in the posterior vitreous
cortex just above the internal limiting lamina of the retina (intensely
Fig. 21.11 Ultrastructure of human vitreoretinal interface. Scanning staining yellow line). These lamellae represent potential cleavage
electron microscopy of anterior surface of the human retina (top) planes during anomalous posterior vitreous detachment with
and the posterior surface of the posterior vitreous cortex (bottom). vitreoschisis. (Reproduced with permission from Gupta P, Yee KMP,
(Reproduced with permission from Sebag J. The vitreous: structure, Garcia P, et al. Vitreoschisis in macular diseases. Br J Ophthalmol
function, and pathobiology. New York: Springer-Verlag; 1989.) 2011;95:376–80.)

Fig. 21.13 Human posterior vitreous cortex.


Three-dimensional spectral-domain optical
coherence tomography imaging of human
posterior vitreous cortex demonstrates the
lamellar structure that predisposes to
vitreoschisis. (Reproduced with permission
from Sebag J. Vitreous – the resplendent
enigma. Br J Ophthalmol 2009;93:989–91.
Image courtesy of Dr Carl Glittenberg and
Prof. Susanne Binder, Vienna.)
diseases. Sakamoto and Ishibashi have recently published an
excellent review of hyalocytes.54
488 Hyalocytes are important in macular pucker when APVD28
and vitreoschisis26,29 leave these cells on the macula (Fig. 21.15).
Under the influence of cytokines, hyalocytes proliferate55 on the
surface of the retina, resulting in hypercellular membranes. Hya-
Section 1

locytes also recruit cells from the circulation and the retina (glial
cells) via the release of connective tissue growth factor and
induce collagen gel contraction in response to platelet-derived
growth factor and other cytokines,56,57 causing tangential vitreo-
retinal contraction.
Anatomy and Physiology
Basic Science and Translation to Therapy

Internal limiting lamina (ILL) of the retina


A The ILL is a multilaminar structure of variable thickness topo-
graphically. Adjacent to Müller cell foot plates is the lamina
rara externa (0.03–0.06  µm) with no species variations or
changes with topography or age. The lamina densa is thinnest
Mi at the fovea (0.01–0.02  µm) and thicker in the posterior pole
(0.5–3.2  µm) than at the equator or vitreous base, where Foos58
found the ILL to be uniformly thin (51  nm) and the lamina
C rara to be 40  nm wide with traversing fibrils that were denser
at sites corresponding to attachment plaques in Müller cells.
The ILL is very thin over major retinal vessels (Fig. 21.16) where
defects allow glial cells to extend on to the inner retina.59
N Acquired ILL defects are in the foveola, retinal pits, retinal
tufts, and retinal lattice. The ILL progressively thickens poste-
riorly to about 306  nm at the equator and about 1887  nm pos-
teriorly. Müller footplates are less numerous at the equator
C than at the vitreous base. Posteriorly, no Müller footplates were
observed and the inner aspect of the ILL remains smooth, while
the outer aspect is irregular. Peripherally, both inner and outer
surfaces are smooth.2,53
The ILL consists of type IV collagen, associated with glycopro-
M
teins,23,60,61 type VI collagen, which may contribute to vitreoreti-
V nal adhesion, and type XVIII,62 which binds opticin.63 Opticin
binds to heparan sulfate, contributing to vitreoretinal adhesion.64
Type XVIII collagen also prevents cell migration from the retina
into vitreous.65
Retinal sheen dystrophy66
This ILL dystrophy has cystic spaces under the ILL and in
the inner nuclear layer (Fig. 21.17), and numerous areas of
separation of the ILL from the retina with filamentous mate-
rial (Fig. 21.18). Endothelial cell swelling and degeneration,
pericyte degeneration, and basement membrane thickening of
retinal capillaries suggest that this condition is primarily a
retinal vasculopathy with edema, swelling, and degeneration
B of Müller cells. Alternatively, the primary defect could be in
Müller cells.
Fig. 21.14 Human hyalocytes. (A) Phase contrast microscopy of Degenerative remodeling
human hyalocytes in situ. (Reproduced with permission from Sebag J. Foos67 defined a spectrum of changes in the ILL as “degenerative
The vitreous: structure, function, and pathobiology. New York: remodeling.” Features include detachment and discontinuity of
Springer-Verlag; 1989, p. 49.) (B) A mononuclear cell is seen
the ILL with vitreous collagen beneath the ILL (Fig. 21.19), cel-
embedded within the dense collagen fibril (black C) network of the
vitreous cortex. There is a lobulated nucleus (N) with a dense marginal lular debris with macrophages, and absence of Müller cell
chromatin (white C). In the cytoplasm there are mitochondria (M), attachment plaques. In larger lesions, vitreous may insinuate
dense granules (arrows), vacuoles (V), and microvilli (Mi) into degenerative crypts and adhere to the cell membrane of the
(magnification = 11 670×). (Courtesy of JL Craft and DM Albert,
Harvard Medical School.) lining Müller cells that have no basal lamina (Fig. 21.20). In the
peripapillary area, retinal glial cells extend from the optic disc
and are continuous with a glial epipapillary membrane that has
vitreous fiber incarceration. Roth and Foos68 observed nasal epi-
papillary membranes associated with Bergmeister papillae in
27.6% of autopsy eyes.
Fig. 21.15 Vitreoschisis (VS). Transmission
electron microscopy of human hyalocyte (same as
MACULAR PUCKER – VS splits anterior to hyalocytes: in Fig. 21.14B) demonstrating two potential levels
hyalocytes remain attached to ILL & recruit cells (blood & splitting during vitreoschisis. Anomalous posterior 489
retina) hypercellular membranes, highly contractile vitreous detachment that induces vitreoschisis
which splits the posterior vitreous cortex anterior
to the level of hyalocytes (red dashes) leaves

Chapter 21
these cells attached to the macula, resulting in
a hypercellular membrane. The lack of attachment
to the optic disc allows inward (centripetal)
C

tangential traction causing contraction and


macular pucker. If vitreoschisis splits the posterior

M
vitreous cortex posterior to the level of hyalocytes
(blue dashes), the remaining membrane
is thin and hypocellular. If there is also
N

Vitreous and Vitreoretinal Interface


V
C
vitreopapillary attachment, the tangential forces
will be outward (initially nasally), opening a central
dehiscence and inducing a macular hole. ILL,
internal limiting lamina; VS, vitreoschisis; MP,
Mi

macular pucker; for other abbreviations see


Fig. 21.14.

MACULAR HOLE – VS splits posterior to hyalocytes


hypocellular membranes that are thinner than MP

Fig. 21.16 Vitreoretinal vessel interface. A large gap (between arrows)


in the internal limiting lamina (ILL) over and adjacent to a retinal artery
is seen with a thin, fibroglial epiretinal membrane over the ILL
(arrowheads) (periodic acid–Schiff; ×470).
Fig. 21.17 Retinal sheen dystrophy. Area with microcysts (large
arrows) beneath the inner limiting lamina (small arrow) and cystic
edema in inner nuclear layer in area of cell bodies of Müller cells
(periodic acid–Schiff; ×850). (Reproduced with permission from Polk T,
Gass JDM, Green WR, et al. Familial internal limiting membrane
dystrophy: a new sheen retinal dystrophy. Arch Ophthalmol 1997;115:
878–85.)

Fig. 21.18 Retinal sheen dystrophy. Inferior


midperipheral area where the inner limiting
lamina (arrow) is separated from Müller cells
(M), and filaments (asterisk) and cellular


debris (two asterisks) are interposed. The
outer portion of the internal limiting lamina
(arrowhead) is separated and remains
attached to Müller cells with junctional
densities (circles) (×15 000).

∗∗
M
the sites of strong vitreoretinal adhesion such as the vitreous
base and optic disc, forming the rationale for pharmacologic
490 vitreolysis using avidin–biotin complex chondroitinase.
Topographic variations
Strength of vitreoretinal adhesion
Section 1

Vitreous is attached to all contiguous structures, but is most


firmly adherent at the vitreous base,72 which includes the poste-
rior 2 mm of the pars plana and extends 1–4 mm posterior to the
ora serrata, varying with age73 and topography (more posterior
temporally). There are also topographic differences posteriorly,
with greater adhesion at the posterior pole than the equator60
Anatomy and Physiology
Basic Science and Translation to Therapy

(Fig. 21.21).
Peripheral fundus and vitreous base
The vitreous base is a three-dimensional structure that straddles
the ora serrata. There is a high density of collagen fibrils oriented
at right angles to the inner surface of the ciliary epithelium and
peripheral retina. The fibrils attach to the basement membrane
of the nonpigmented epithelium of the pars plana and the ILL
of the peripheral retina,74 intimately interwoven with an inter-
vening extracellular matrix. Within the vitreous base, there are
several anatomic variations where vitreoretinal adhesion is
firm75 and associated with retinal breaks76,77:

Fig. 21.19 Degenerative remodeling. Basal area with vitreous collagen 1. Ora bays70,75,78 (Fig. 21.22)
located between detached inner limiting lamina (arrow) and glial cell
process. (Courtesy of Tsuyoshi Kimura.) 2. Meridional folds77,79 (Fig. 21.23)
3. Meridional complexes77 (Figs 21.22 and 21.24)
4. Peripheral retinal excavations77,80 (Fig. 21.24)
5. Retinal tufts
(a)  Noncystic retinal tufts81–83 (Fig. 21.25)
(b) Cystic retinal tufts76,81,82,84 (Fig. 21.26)
(c) Zonular traction tufts79,85–88 (Fig. 21.27)
6. Spiculate and nodular pigment epithelial hyperplasia87
(Fig. 21.28)
7. Retinal lattice “degeneration”58,84,89–105 (Figs 21.29–21.33)
8. White-with-pressure, white-without-pressure106–113
9. Verruca114 (Fig. 21.34).

Interface along major retinal vessels


Fig. 21.20 Basal zone showing detached and degenerating internal The ILL thins and is sometimes absent over major retinal
limiting lamina under which the attachment plaques are missing in the
vessels115,116 (Fig. 21.35). At such points, vitreous may be incarcer-
surface of Müller cells and vitreous has become incarcerated. Vitreous
in degeneration crypt has become firmly adherent to Müller cell (arrow) ated into retina, directly continuous with perivascular tissue,
(×12 600). (Reproduced with permission from Foos RY. Vitreoretinal attachments called “vitreoretinovascular bands,”117 or “spider-
juncture: topographical variations. Invest Ophthalmol 1972;11:801–8.) like bodies,”118 purported to be vitreous fibrils that traverse the
ILL and coil about retinal blood vessels. It is common for retinal
Vitreoretinal interface vessels to be associated with paravascular rarefaction (cystic
degeneration), retinal pits and tears, and avulsion of retinal
The interface between vitreous and adjacent structures consists
vessels.
of a complex formed by the vitreous cortex and basal laminae
which are firmly attached to their cells.69 The only part of vitre- Vitreomacular interface
ous not adjacent to a basal lamina is the annulus of the anterior Attachment of vitreous to the macula occurs in an irregular,
vitreous cortex, which is directly exposed to the zonules and the annular zone of 3–4 mm diameter,2 generally not visible by clini-
aqueous humor of the posterior chamber, similar to the surface cal examination in normal adults but possibly evident in fetal
of articular cartilage, which is exposed to synovial fluid.48 Zim- and young adult eyes and in pathologic conditions. The poste-
merman and Straatsma70 claimed that there are fibrillar attach- rior vitreous cortex is thinner over the macula in a disc-shaped
ments between the posterior vitreous cortex and the ILL. The area about 5 mm in diameter. Discontinuity of the ILL in the
composition of these fibrillar structures is not known and their fovea may be a site where glial cells extend on to the inner
presence has never been confirmed. surface of the retina (Fig. 21.36).
It is currently believed that an extracellular matrix “glue” of Vitreopapillary interface
fibronectin, laminin, and other extracellular matrix components71 At the rim of the optic disc the ILL ceases, although the base-
exists between vitreous and retina, causing adhesion to be ment membrane continues as the inner limiting membrane of
fascial, as opposed to focal.55,56 Chondroitin sulfate is present at Text continued on page 495
491

Chapter 21
p
c

Vitreous and Vitreoretinal Interface


A B

C D Coll

Fig. 21.21 Vitreomacular interface in youth. (A) Dark-field microscopy of the posterior vitreous from a 14-year-old boy. The sclera, choroid, and
retina were dissected off the vitreous, which remains attached to the anterior segment. In contrast to adults, there is an extra layer of tissue that
remained adherent to the posterior vitreous cortex when the retina was dissected off. The white arrow indicates the location of the fovea. The
circular structure below this is the prepapillary hole in the posterior vitreous cortex. Emanating from this hole are linear, branching structures
(black arrows) that correspond to the location of the retinal vessels. (B) Scanning electron microscopy of the tissue shown in (A) demonstrates
round structures adherent to the posterior aspect of this tissue (bar = 1 µm). (C) Higher magnification of structures shown in (B). (D)
Transmission electron microscopy of this specimen identified this tissue as the internal limiting lamina (ILL) of the retina (arrows) attached to the
posterior vitreous cortex (p). The round structures shown in (B) are identified as the inner portion of Müller (m) cells that remained adherent to
the posterior aspect of the ILL. The hole on the posterior aspect of these round structures is where the anterior portion of the Müller cell was torn
away from the rest of the cell body. At the bottom of the photomicrograph are the collagen fibrils of the vitreous (Coll) (original magnification
×20 800). (Panel C reproduced with permission from Sebag J. Age-related differences in the human vitreoretinal interface. Arch Ophthalmol
1991;109:966–71.)
492
Section 1


Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.22 A meridional complex (arrow) and dentate process


(arrowhead) are located at the margin of a nonenclosed ora bay.
Fig. 21.23 Meridional folds, one in line with a meridional complex
(arrowhead) and the other (arrow) in line with a meridional complex and
enclosed ora bay (asterisk). (Reproduced with permission from Green
WR. Pathology of the retina. In: Frayer WC, editor. Lancaster course in
ophthalmic histopathology, unit 9. Philadelphia: FA Davis; 1981.)

Fig. 21.24 Peripheral retinal excavation (arrowhead) in line with a


meridional complex and an enclosed ora bay. (Reproduced with
permission from Green WR. Pathology of the retina. In: Frayer WC,
editor. Lancaster course in ophthalmic histopathology, unit 9.
Philadelphia: FA Davis; 1981.)

Fig. 21.25 Noncystic peripheral retinal tuft. Most tufts consist of glial
cells, and some have strands of vitreous attached (arrow) (hematoxylin
and eosin, ×215). (Reproduced with permission from Green WR.
Retina. In: Spencer WH, editor. Ophthalmic pathology: an atlas and
textbook, vol. 2. Philadelphia: WB Saunders; 1985.)
493

Chapter 21

Vitreous and Vitreoretinal Interface


A

Fig. 21.26 Cystic retinal tuft. (A) The lesion consists of an area of
retinal thickening caused by cystic changes (asterisks) and glial cell
proliferation (hematoxylin and eosin, ×220). (B) Scanning electron
microscopy (EM) demonstrates that the tuft is a cystoid formation of
fibers, similar to those of the nerve fiber layer, and cells, similar to
those found in the inner plexiform layer of the retina, that are
connected to the internal limiting lamina of the retina. This scanning
EM shows the insertion of the vitreous collagen fibers on the tuft’s
apical surface. (Reproduced with permission from Dunker S, Glinz J,
Faulborn J. Morphologic studies of the peripheral vitreoretinal interface
in humans reveal structures implicated in the pathogenesis of retinal B
tears. Retina 1997;17:124–30.)

A B

Fig. 21.27 Zonular traction tuft. Gross (A) and microscopic


(B) appearances of zonular tractional tuft (arrow in panel A) composed
of a strand of glial cells whose anterior part consists of two layers of
embryonal-like epithelium (arrows in panels B and C; hematoxylin and
eosin, ×65). (C) Higher magnification (hematoxylin and eosin, ×290).
(Panels A and B reproduced with permission from Green WR.
Pathology of the retina. In: Frayer WC, editor. Lancaster course in
ophthalmic histopathology, unit 9. Philadelphia: FA Davis; 1981;
panel C reproduced with permission from Green WR. Retina.
C In: Spencer WH, editor. Ophthalmic pathology: an atlas and textbook,
vol. 2. Philadelphia: WB Saunders; 1985.)
494
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.28 Hyperplasia of pigment epithelium at ora serrata.


Hyperplastic pigment epithelium (arrow) extends into the vitreous
cavity at the ora serrata (periodic acid–Schiff; ×220). (Reproduced with
permission from Green WR. Pathology of the retina. In: Frayer WC,
editor. Lancaster course in ophthalmic histopathology, unit 9. Fig. 21.29 Paravascular retinal lattice. Radial perivascular lattice with
Philadelphia: FA Davis; 1981.) sclerotic vessel and hyperplastic retinal pigment epithelium, with
migration into the retina in a paravascular location.

Fig. 21.31 Histopathology of retinal lattice. Lattice lesion with retinal


thinning, discontinuity of inner limiting membrane (arrows), pocket of
fluid vitreous (asterisk), and glial cell proliferation in vitreous at margin
(arrowheads) (Alcian blue stain; ×145).

Fig. 21.30 Lattice wicker. Gross appearance of retinal lattice, with


sclerotic vessels that have a wicker appearance.

Fig. 21.33 Retinal tear at posterior margin of area of lattice


degeneration.

Fig. 21.32 Trypsin digestion preparation of retina discloses relatively


acellular capillaries in area of lattice degeneration (right) (hematoxylin
and eosin and periodic acid–Schiff stains; ×185).
Elschnig.119 This membrane is 50  nm thick and is believed to out of the visual axis, causing “floaters.” Vitreopapillary adhe-
be the basal lamina of the astroglia in the optic nerve head. At sion (VPA) contributes to macular holes121 and any vitreomacu-
the centralmost portion of the optic disc the membrane thins lopathy that features intraretinal cystoid spaces,122 presumably 495
to 20  nm, follows the irregularities of the underlying cells of due to the influence upon the vectors of tangential traction upon
the optic disc, and is composed only of glycosaminoglycans the macula.

Chapter 21
and no collagen (central meniscus of Kuhnt). Given that the
ILL prevents the passage of cells,47 the thinness and chemical PHYSIOLOGY
composition of the central meniscus of Kuhnt and the membrane
Biochemical
of Elschnig may account for frequent cell proliferation from or
near the optic disc. Vitreous is important in maintaining transparency for maximal
Vitreous attachment to the optic disc may persist even though photon transmission to the retina. Vitreous may also maintain
the vitreous is detached elsewhere120 (Fig. 21.37). This adhesion lens transparency by mitigating the effects of reactive oxygen

Vitreous and Vitreoretinal Interface


may be fortified by epipapillary membranes.68 The entire complex species on lens proteins and thus preventing cataracts.123 This
may subsequently detach, resulting in a ring of tissue composed antioxidant effect is primarily the result of high concentrations
of fibrous astrocytes and collagen (Fig. 21.38) that flutters in and of ascorbate in vitreous, an observation originally made in 1944
by Friedenwald and colleagues.124
Gisladottir et al.125 recently emphasized the influence of vitre-
ous on various physiologic processes and showed that vitrec-
tomy can have considerable effects, both beneficial and
harmful.126 Vitrectomy reduces the risk of retinal neovasculariza-
tion, but increases the risk of iris neovascularization, reduces
macular edema, but stimulates cataract formation.

Biophysical
During ocular saccades, the rotational force of the eye wall is
transmitted to the vitreous body via attachments to adjacent
structures.127 During both acceleration and deceleration phases
of saccades, vitreous movement lags behind the eye wall, result-
ing in markedly reduced acceleration.128 This “slack and lag”

Fig. 21.34 Verruca. Scanning electron microscopy shows cellular Fig. 21.35 Vitreous interface over retinal vessels. There is a large gap
elements resembling cells of the inner plexiform layer near the retinal in the inner limiting lamina (ILL) over and adjacent to retinal artery. A
surface. The “trunk” of the verruca extends from the retina to the vitreous glial preretinal membrane (arrowhead) originates at a defect in the
cortex. The “branches” of the verruca are intertwined with vitreous ILL overlying the retinal vessel. Arrow indicates one end of the
collagen fibers. There is local collagen destruction (arrows) and discontinuous ILL (periodic acid–Schiff stain; ×170). (Reproduced with
interruption of the internal limiting lamina of the retina. (Reproduced with permission from Clarkson JG, Green WR, Massof D. A histopathologic
permission from Dunker S, Glinz J, Faulborn J. Morphologic studies review of 168 cases of preretinal membrane. Am J Ophthalmol
of the peripheral vitreoretinal interface in humans reveal structures 1977;84:1–17.)
implicated in the pathogenesis of retinal tears. Retina 1997;17:124–30.)

Fig. 21.36 Vitreomacular interface. The


internal limiting lamina is discontinuous
(between arrows), at which point glial cells
extend on to the inner surface of the retina
and form a thin, fibroglial premacular
membrane (arrowheads) with no apparent
contraction (periodic acid–Schiff stain; ×100).
496
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.37 Vitreopapillary adhesion. (A) Gross appearance illustrates


partial posterior vitreous detachment with strand of vitreous remaining
attached to the optic nerve head. Light reflexes in photograph taken
with specimen out of fluid to depict the findings more clearly.
(B) Combined optical coherence tomography-scanning laser
ophthalmoscopy (OCT-SLO) imaging of vitreopapillary adhesion. The
A gray-scale image is coronal plane imaging by SLO, and the color
image is the transverse OCT image through the optic disc.

results from vitreous viscoelasticity, dampening the force at any single, large pocket forms, the terms “bursa”24 or “precortical
given internal vitreous attachment. The inferonasal displace- pocket”134 have been employed. This large posterior lacuna is a
ment of the optic disc and the shorter distance between the disc manifestation of age-related liquefaction (Fig. 21.39), and not an
and ora inferiorly and nasally make the relief of torsional strain anatomic entity.135 Balazs and Denlinger130 found evidence of
on equatorial and anterior vitreoretinal attachments greater liquid vitreous after the age of 4 years and observed that, by the
nasally and inferiorly. Accordingly, the greatest torsional strain time the human eye reaches adult size (ages 14–18 years), 20%
on anterior and equatorial vitreous attachments should occur of the total volume is liquid vitreous. By the age of 80–90 years
during lateral saccades, with the point of maximum strain more than half the vitreous is liquid.
located somewhere in the superotemporal quadrant, the site of
Pathogenesis of vitreous liquefaction
most frequent retinal tears.129
Changes in collagen136,137 or the conformation of HA with subse-
AGE-RELATED VITREOUS DEGENERATION quent cross-linking of and aggregation of fibrils into bundles
may result in vitreous liquefaction. Free radicals generated by
Liquefaction (synchysis)130,131 metabolism and/or photons alter vitreous macromolecules and
After age 40 years there is a significant decrease in the gel volume trigger dissociation of collagen from HA, leading to liquefac-
and a concurrent increase in the liquid volume of vitreous, pri- tion.138 Vitreous liquefaction may also result from changes in the
marily centrally.132,133 In the posterior vitreous such changes form minor glycosaminoglycans and chondroitin sulfate profile of
pockets of liquid vitreous, called “lacunae” (Fig. 21.39). When a vitreous.139
Aging changes and vitreous biochemistry vitreous HA concentration increases until about the age of 20,
when adult levels are attained. Thereafter, until 70 years, there
Biochemical studies support the rheologic observations described
above. Total vitreous collagen content does not change after ages
are no changes in the HA concentrations of either the liquid or 497
gel compartments. This necessarily means that there is an
20–30 years. However, collagen concentration in gel vitreous at
increase in the HA content of liquid vitreous and a concomitant
the ages of 70–90 years (0.1 mg/mL) was significantly greater

Chapter 21
decrease in the HA content of gel vitreous, since the amount of
than at the ages of 15–20 years (0.05 mg/mL; P < 0.05).130 Since
liquid vitreous increases and the amount of gel vitreous decreases
the total collagen content does not change, this is likely due to
with age.
the decreased volume of gel vitreous that occurs with aging and
a consequent increase in the concentration of the collagen in the
remaining gel. This concept is supported by the finding that Structural changes
Vitreous body

Vitreous and Vitreoretinal Interface


The aforementioned rheologic and biochemical alterations
induce significant structural changes during aging, consisting
of a transition from a clear gel in youth (Fig. 21.2) to a fibrous
structure in adults (Fig. 21.7A). In old age there is advanced
liquefaction with thickening and tortuosity of vitreous fibers,
and collapse (syneresis) of vitreous (Fig. 21.40). Postmortem
studies140 found syneresis in 70% of subjects in the eighth
decade. Syneresis occurs earlier and is more extensive in
myopic eyes,141 and is accelerated with inflammation, trauma,
and arthro-ophthalmopathies.7,142
Aging changes at the vitreoretinal interface
Teng and Chi73 found that the width (in the radial dimension) of
the vitreous base posterior to the ora serrata increases with age
to over 3.0 mm. There is also posterior migration of the posterior
border of the vitreous base with age,73,103 mostly temporally. In
addition, Gartner143 found “lateral aggregation” of the collagen
fibrils in the vitreous base of older individuals. These changes
play important roles in the pathogenesis of peripheral retinal
breaks and rhegmatogenous RD.

Posterior vitreous detachment


Fig. 21.38 Weiss ring. Surgically removed Weiss ring contains fibrous
astrocytes that have polarity, basement membrane (arrowhead), and Due to inadequate diagnostics,144 PVD is an inaccurate diag-
large bundles of 10-nm filaments (arrows) (×20 000). nosis. PVD begins at the posterior pole, perhaps in the

A B

Fig. 21.39 Morphology of human vitreous liquefaction (synchysis). (A) Gross appearance of central vitreous liquefaction (synchysis).
(B) Dark-field slit microscopy of human vitreous demonstrates the central vitreous has thickened, tortuous fibers. The peripheral vitreous has
regions devoid of any structure which contain liquid vitreous. These regions correspond to so-called lacunae (arrows).
498 Liquefaction without vitreoretinal dehiscence
Section 1

Anomalous
PVD

Partial thickness = Full-thickness


VITREOSCHISIS but only partial PVD
Anatomy and Physiology
Basic Science and Translation to Therapy

Premacular Peripheral separation


membrane Posterior traction

Posterior
separation
Peripheral
VPA & No VPA &
traction
centrifugal centripetal
Fig. 21.40 Gross appearance of posterior vitreous detachment in a (outward) (inward) Macular Optic disc
phakic eye. contraction contraction traction traction

perifoveal region.145 An innocuous PVD is clean separation


between the ILL of the retina and the cortical vitreous.146 Macular Macular VMTS Vitreo- Retinal
Whereas it is widely held that PVD is an “abnormal” event, hole pucker EXUD AMD papillary tears &
it is possible that PVD may be a preprogrammed event that traction detachment
mitigates the risks of an attached vitreous, which in old age
are more dangerous than a PVD.147 Fortunately, PVD is innocu-
ous in most cases. Fig. 21.41 Schematic of anomalous posterior vitreous detachment
(PVD). This diagram demonstrates the various possible manifestations of
Epidemiology anomalous PVD. When gel liquefaction and weakening of vitreoretinal
The incidence of PVD is 66% between the ages of 66 and 86 adhesion occur concurrently, the vitreous separates away from the retina
without sequelae. If the gel liquefies without concurrent dehiscence at the
years,148 and 53% after 50 years.149 Clinical examination,150 ultra- vitreoretinal interface, there can be various untoward consequences,
sonography,151 and monochromatic photography152 have been depending upon where the vitreous is most adherent. If separation of
standard, but nanotechnologies such as dynamic light scatter- vitreous from retina is full-thickness but incomplete, there can be different
ing,153,154 are being developed to improve clinical evaluation. forms of partial PVD (right side of diagram). Posterior separation with
persistent peripheral vitreoretinal attachment can induce retinal breaks
In a postmortem study155 of 786 subjects aged >20 years, an and detachments. Peripheral vitreoretinal separation with persistent
upside-down suspension-in-air technique detected a 41% inci- full-thickness attachment of vitreous to the retina posteriorly can induce
dence of PVD over 65 years of age. Of 62 aphakic eyes, 94% had traction upon the macula, known as the vitreomacular traction syndrome
(VMTS). This phenomenon appears to be highly associated with
partial or complete PVD.
exudative age-related macular degeneration (EXUD AMD). Persistent
Symptomatic PVD attachment to the optic disc can induce vitreopapillopathies and also
contribute to neovascularization and vitreous hemorrhage in ischemic
The sudden onset of “floaters” heralds the onset of PVD. Floaters retinopathies. If, during PVD, the posterior vitreous cortex splits
have a significant negative impact on the quality of life.156 The (vitreoschisis), there can be differences depending upon the level of the
incidence of retinal tears in patients with acute symptomatic split. Vitreoschisis anterior to the level of the hyalocytes leaves a
relatively thick cellular membrane attached to the macula. If there is also
PVD varies from 8 to 15%, to as high as 46%.157 In a study158 of separation from the optic disc (present in 82% of cases), inward
589 patients with “floaters,” diffuse dots, vitreous cells, and (centripetal) contraction of this membrane induces macular pucker. If the
hemorrhage were high-risk factors for a retinal tear, since 93 of split occurs at a level posterior to the hyalocytes, the remaining
176 (52.8%) of eyes with one or more of these risk factors had premacular membrane is relatively thin and hypocellular. Persistent
vitreopapillary adhesion (VPA), present in 87.5% of cases, influences the
retinal tears. Novak and Welch159 reported that, in 172 eyes of vector of force in the tangential plane, resulting in outward (centrifugal)
155 patients with acute symptomatic PVD, 31% had complica- tangential traction (especially nasally), inducing a macular hole.
tions. Of these 155 patients, PVD developed in the fellow eye in (Reproduced with permission from Sebag J. Anomalous PVD – a unifying
concept in vitreoretinal diseases. Graefes Arch Clin Exp Ophthalmol
17 (11%) within 2 years. 2004;242:690–8.)

ANOMALOUS PVD (APVD) Vitreous effects of APVD


APVD results from gel liquefaction without concurrent weaken- An important consequence of APVD is vitreoschisis29,32 (Fig.
ing of vitreoretinal adherence, causing various clinical manifes- 21.42), i.e., splitting of the posterior vitreous cortex with forward
tations based upon where vitreous is most liquefied and where displacement of the anterior portion of the cortex, leaving the
the interface is most firmly adherent (Fig. 21.41). posterior layer attached to the retina. Vitreoschisis has been
499

Chapter 21
Focus: –2.5 Scan angle: 29.7°

Vitreous and Vitreoretinal Interface


A B

SPLIT

Fig. 21.42 Vitreoschisis. (A) Ultrasonography of posterior vitreoschisis demonstrates the two layers of the schisis cavity rejoining into full-
thickness posterior vitreous cortex (below). (Reproduced with permission from Green RL, Byrne SF. Diagnostic ophthalmic ultrasound. In: Ryan
SJ, editor. Retina, 1st edn. St Louis: Mosby; 1989.) (B) Optical coherence tomography-scanning laser ophthalmoscopy imaging of posterior
vitreoschisis demonstrates the two walls of the schisis cavity rejoining into full-thickness posterior vitreous cortex (to left). (Reproduced with
permission from Gupta P, Yee KMP, Garcia P, et al. Vitreoschisis in macular diseases. Br J Ophthalmol 2011;95:376–80.) (C) Histopathology of
vitreoschisis. Surgical specimen removed from a patient with macular pucker demonstrates the split in the posterior vitreous cortex and rejoining
of the two walls into full-thickness posterior vitreous cortex. Hyalocytes (arrows) are seen within the posterior vitreous cortex. Note the lack of
any additional cellularity on the surfaces of the split posterior vitreous cortex. (Periodic acid–Schiff; ×300.) (Reproduced with permission from
Gupta P, Yee KMP, Garcia P, et al. Vitreoschisis in macular diseases. Br J Ophthalmol 2011;95:376–80.)
detected in proliferative diabetic retinopathy,14 macular pucker, patient education while Combs and Welch176 concluded that pro-
and macular holes.160 phylactic treatment of acute horseshoe tears with vitreous trac-
500 tion significantly reduces the incidence of RD. A particularly
Peripheral retinal effects of APVD high-risk group are patients with vitreous hemorrhage that
Retinal breaks obscures fundus visualization. Ultrasound may be an effective
Retinal holes unrelated to PVD were observed in 326 (13.9%) of means of identifying retinal tears in such eyes,177 but misdiagno-
Section 1

2334 autopsy cases by Foos et al.96 Retinal tears (Fig. 21.43) result sis at presentation bodes poorly, since there is a 67% incidence
from fluid movements.161 Vitreous remains attached to the pos- of retinal tears.178
terior margin of the retinal flap, which may be avulsed leaving
Other sequelae
a round or oval hole. The flap of retina remains attached to the
1. Retinal tags81
posterior surface of the detached vitreous (operculum). Large
2. Retinal folds
detached flaps may form a cystic structure (Fig. 21.44).
Anatomy and Physiology
Basic Science and Translation to Therapy

3. Traction retinoschisis (Fig. 21.45)


The clinical incidence of retinal tears162 varies from 7.2%163 to
4. Traction on retinal vessels 115 (Fig. 21.46A)
5.8%,164 with a high of 13.75%79 and a low of 0.59%.165 Postmor-
(a) Cystic degeneration169,170 (Fig. 21.46B)
tem incidences were 3.9%,166 8.6%,167 4.7%,168 8.8%,169 3.7%,170 and
(b) Avulsion of retinal vessels179,180
7.3%.96 Although the role of retinal tears in causing RD is undis-
(c) Retinal pits169 (Fig. 21.47)
puted, management is controversial. Byer171 concluded that pro-
phylactic treatment is not justified for asymptomatic retinal Macular effects of APVD
breaks in phakic eyes. However, in a natural history study of 166
Vitreomacular traction
eyes with retinal breaks, Davis105 observed that 31 (18%) pro-
For vitreomacular traction syndrome,2,28,32 see Fig. 21. 48.
gressed to RD. Neumann and Hyams172 reported that 2% of 153
eyes with retinal breaks developed RD. The incidence of retinal Exudative age-related macular degeneration
tears is much greater than RD, which varies between 9 (0.009%)173 Recent studies181,182 determined that vitreomacular adhesion may
and 24.4 (0.02%) per 100 000 per year.174 Benson175 promoted be a risk factor for exudative age-related macular degeneration.
Several subsequent studies have confirmed these findings.
Cystoid macular edema
For cystoid macular edema,183–189 see Figs 21.49 and 21.50.
Macular cysts190
Macular cysts that result from chronic edema (Fig. 21.51) need
to be distinguished from the cystoid spaces created by vitreous
traction in macular holes (Figs 21.52 and 21.53), and macular
pucker with VPA.121

Fig. 21.43 Retinal pits and tears. Gross appearance of eye with
posterior vitreous detachment, a string of retinal pits along vessels
(arrows), and three horseshoe-shaped retinal tears (arrowheads).
(Reproduced with permission from Green WR. Pathology of the
vitreous. In: Frayer WC, editor. Lancaster course in ophthalmic
histopathology, unit 8. Philadelphia: FA Davis; 1981.)

Fig. 21.45 Traction retinoschisis. Gross appearance of large


retinoschisis caused by vitreous traction. A sheet of vitreous (arrow)
Fig. 21.44 Large retinal tear. Microscopic features include rounded extends across the eye and temporally is attached to the apex of the
anterior (arrowheads) and posterior margins of the hole and coiled-up, tented-up inner layer of the extensive areas of retinoschisis. Posteriorly
detached operculum (arrows) that remains attached to the posterior a hole in the outer layer (arrowhead) is present and allows
surface of the detached vitreous (periodic acid–Schiff stain; ×16). visualization of some interbridging strands of tissue between two
(Reproduced with permission from Green WR. Pathology of the layers of schisis. (Reproduced with permission from Green WR.
vitreous. In: Frayer WC, editor. Lancaster course in ophthalmic Retina. In: Spencer WH, editor. Ophthalmic pathology: an atlas and
histopathology, unit 8. Philadelphia: FA Davis; 1981.) textbook, vol. 2. Philadelphia: WB Saunders; 1985.)
501

Chapter 21
B

Vitreous and Vitreoretinal Interface


A

Fig. 21.46 Anomalous posterior vitreous detachment effects on retinal


vessels. (A) Retinal tear with bridging retinal blood vessel. (B) There is
cystic degeneration of the nerve fiber layer near the retinal vessel
(arrow) (periodic acid–Schiff stain; ×120). (Reproduced with permission
from Green WR. Pathology of the retina. In: Frayer WC, editor.


Lancaster course in ophthalmic histopathology, unit 9. Philadelphia: FA
Davis; 1981.) (C) Section through a venous loop which is parallel to
∗∗ the axis of the vein shows the anterior (asterisk) and posterior (double
asterisks) components of the loop as it extends through a discontinuity
in the inner limiting membrane (between arrows). The wall of the loop
is thin and tented at the points of attachment of thin delicate vitreous
strands (arrowheads) anteriorly and posteriorly (periodic acid–Schiff
C stain; ×100). (Reproduced with permission from Hersh PS, Green WR,
Thomas JV. Am J Ophthalmol 1981;92:661–71.)

Fig. 21.47 Retinal pit. Margin of retinal pit with discontinuity of the
internal limiting lamina (arrow) and a glial cell preretinal membrane
(arrowhead). (Periodic acid–Schiff stain; ×185.) (Reproduced with
permission from Clarkson JG, Green WR, Massof D. A histopathologic
review of 168 cases of preretinal membrane. Am J Ophthalmol 1977;
84:1–17.)

Fig. 21.48 Vitreomacular traction syndrome. This extreme example of


anomalous posterior vitreous detachment with persistent full-thickness
adhesion of the posterior vitreous cortex to the macula has induced
sufficient traction upon the macula to induce prominent elevation of
the central macula. Typically, there is only thickening of the macula
without detachment.
502
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.49 Cystoid macular edema with cystic spaces in the outer
plexiform and inner nuclear layers (hematoxylin and eosin stain; ×45).

Fig. 21.50 Irvine–Gass syndrome. Area near posterior pole showing


edema and retinal phlebitis (arrow) (hematoxylin and eosin stain;
×185). (Reproduced with permission from Martin NF, Green WR,
Martin LW. Retinal phlebitis in the Irvine–Gass syndrome. Am J
Ophthalmol 1977;83:377–86.)

Fig. 21.51 Cystic edema (arrows) and a large cyst (asterisk) are
present in the macula (hematoxylin and eosin, ×40). (Reproduced with
permission from Green WR. Retina. In: Spencer WH, editor.
Ophthalmic pathology: an atlas and textbook, vol. 2. Philadelphia: WB
Saunders; 1985.)

Fig. 21.52 Lamellar macular hole. Combined


optical coherence tomography-scanning laser
ophthalmoscopy imaging of a partial-
thickness macular hole with adjacent cystoid
spaces.

Focus: –1.11 Scan angle: 29.2°


503

Chapter 21
Focus: –0 Scan angle: 29.7°

Vitreous and Vitreoretinal Interface


A

B C

Fig. 21.53 Macular hole. (A) Combined optical coherence tomography-


scanning laser ophthalmoscopy imaging of macular hole demonstrating
anomalous posterior vitreous detachment, a pseudo-operculum,
pericentral intraretinal cystoid spaces, and a thin premacular
membrane, particularly visible to the right. (B) Microscopy shows
macular hole, detached posterior vitreous cortex with adherent
pseudo-operculum, cystoid spaces in outer plexiform and inner nuclear
layers, a small area of surrounding retinal detachment, demarcation
chorioretinal adhesions on both sides of the elevated retina, and a
premacular membrane particularly visible to the right (hematoxylin and
eosin, ×55). (C) Higher-power view of B showing a thin hypocellular
preretinal membrane (arrowhead), prominent cystoid spaces, and
demarcation adhesion (arrow) (hematoxylin and eosin, ×140).
(Reproduced with permission from Frangieh GT, Green WR, Engel HM.
A histopathologic study of macular cysts and holes. Retina 1981;
D 1:311–36.) (D) Detached pseudo-operculum demonstrating lack of
neural retina elements.
Continued
Fig. 21.53 Cont’d (E) Composite low-power
view of operculum. Thicker cellular ends are
connected by a thinner segment. A thin
504 collagenous layer along this region (cortical
vitreous) identified the inner vitreal surface
(arrows) of the operculum. A short segment
of ILL (circle) is present in an area of folding
Section 1

(×550). (Reproduced with permission from


Madreperla SA, McCuen BW II,
Hickingbotham D, et al. Am J Ophthalmol
1995;120:197–207.)
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.54 Idiopathic macular hole with


extensive photoreceptor cell atrophy. A thin,
tapered layer of cortical vitreous with a
hypocellular membrane on its inner surface
(arrowhead) apparently exerts tangential
traction with elevation of the hole margins
(periodic acid–Schiff, ×340). (Reproduced
with permission from Guyer DR, Green WR.
Idiopathic macular holes and precursor
lesions. In: Franklin RM (ed.) Proceedings of
the 1992 New Orleans Academy of
Ophthalmology. New York: Kugler, 1993.)

Macular holes Pathogenesis


Macular holes are surrounded by a gray ring of cystoid spaces There have been various theories of macular hole pathogenesis:
and slightly elevated retina, thinning and depigmentation of trauma, foveal degeneration, vitreous traction, and involutional
the underlying retinal pigment epithelium (RPE), yellow thinning with PVD. It is clear from surgical experience199 that
nodular opacities at the level of the RPE, a preretinal membrane vitreous is the cause of macular hole. Johnson and Gass200 for-
in all cases with eccentric retinal pucker in 40%, and an oper- mulated the tangential traction theory by suggesting that shrink-
culum in 25% of patients (Fig. 21.53). Macular hole occurred age of the prefoveal vitreous induces macular hole formation in
in 8 of 37 (22%) fellow eyes in one study.191 Avila et al.192 four stages. There are three possible mechanisms of tangential
reported axial traction as the cause, while Morgan and Schatz193 vitreous traction: (1) fluid vitreous movements and countercur-
proposed that involutional macular thinning predisposes to a rents; (2) cellular remodeling of cortical vitreous; and (3) contrac-
macular hole. tion of a cellular membrane on the tapered cortical vitreous after
Histopathology and pseudo-operculum vitreoschisis.27,31,32 OCT scanning laser ophthalmoscopy (OCT-
Minor RPE hypertrophy and hyperplasia occur in the area of SLO) imaging found vitreoschisis in half of eyes with macular
lamellar and full-thickness macular hole. At the onset of macular hole29,160 (Fig. 21.56). In an ultrastructural study of epiretinal
hole, there is likely vitreoschisis,32 leaving a thin hypocellular tissue removed during vitrectomy for impending macular holes,
layer of the posterior vitreous cortex attached to the macula147,160,194 Smiddy et al.187 observed cortical vitreous in all eyes. VPA may
(Fig. 21.54). There is little cellular proliferation,195 suggesting that be important, as this is present in 88.2% of macular hole eyes.121,122
fluid countercurrents or vitreoschisis (Fig. 21.55) may be impor- VPA influences the vector of tangential forces on the macula and
tant. Healing of macular hole following surgery196 does, however, induces outward (centrifugal) traction, opening a central dehis-
involve glial cell proliferation197 and Müller cell processes.198 cence. In macular pucker, there is usually no VPA and the vector
Macular hole opercula are rarely composed of retinal tissue (Fig. of tangential traction is inward (centripetal), causing a macular
21.53), hence the name “pseudo-operculum.” pucker.
505

Chapter 21
Vitreous and Vitreoretinal Interface
A B

Fig. 21.55 Membranes removed during macular hole surgery. (A) Tissue removed at surgery for macular hole demonstrates a hypocellular membrane
with spindle and stellate-shaped cells (Millipore filter, modified Papanicolaou; ×340). (B) Internal limiting lamina (ILL) of retina. After peeling off the
retina, this tissue demonstrates the characteristic “scrolled” configuration of the ILL following chromodissection. This does not occur with preretinal
membranes (A), only with the ILL. (Millipore filter, modified Papanicolaou stain; ×340.)

Fig. 21.56 Vitreoschisis in macular hole.


Optical coherence tomography-scanning
laser ophthalmoscopy imaging of
vitreoschisis in a stage III macular hole. The
inner wall of the schisis cavity (white arrow)
is lifted off the retinal surface but remains
adherent to the optic disc. The outer wall of
the schisis cavity places centrifugal (outward)
tangential traction of the macula opening a
central dehiscence and inducing the macular
hole. This layer must be removed at surgery,
often done with chromodissection. Black
arrow indicates the outer layer of the
vitreoschsis split in the posterior vitreous
cortex which is still attached to the retina.

Some surgeons201 have advocated prophylactic surgery in


symptomatic fellow eyes. However, a multicenter trial failed to
demonstrate efficacy for vitrectomy in such eyes,202 and thus
surgery is not routinely performed.

Optic disc effects


Anomalous PVD with persistent adhesion to the optic disc can
cause vitreopapillary traction inducing hemorrhage,203 exacer-
bating neovascularization in proliferative diabetic vitreoretinop-
athy,204 and even inducing gaze-evoked visual disturbances.205
Vitreopapillary adhesion also plays a role in the formation of
macular holes and cysts.121,122

VITREORETINAL CHANGES AFTER


LENS EXTRACTION
Fig. 21.57 Traction retinal detachment in eye of a 23-year-old man
Structural with a previous history of traumatic cataract who underwent surgery
1. Opacification.206 complicated by vitreous incarceration in the wound. A vitreous strand
(arrow) is seen attached to the anterior retina that is under traction and
2. Hemorrhage (APVD) with subsequent liquefaction.
“tented” (arrowhead) (periodic acid–Schiff, ×60). (Reproduced with
3. Vitreous incarceration in the wound and vitreoretinal permission from Smiddy WE, Green WR. Retinal dialysis: pathology
traction207 (Fig. 21.57). and pathogenesis. Retina 1982; 2:94–116.)
Biochemical VITREORETINAL CHANGES
208
Reduction of vitreous HA concentration results in decreased AFTER TRAUMA
506 viscosity209 and shock absorption. This can be prevented by
maintaining an intact posterior capsule. Blunt trauma
Blunt trauma may be transmitted to the retina in a direct and
Section 1

contrecoup fashion, resulting in a variety of rhegmatogenous


PVD210,211 sequelae (Fig. 21.58).213 Dialysis at the anterior border of the
PVD occurred in 84% following intracapsular cataract extraction, vitreous base typically occurs inferonasally. Less common are
76% following extracapsular cataract extraction (ECCE) and sur- avulsion of the vitreous base and retinal dialysis at the posterior
gical capsular discission, and in 40% of eyes following ECCE border of the vitreous base. Concussive forces following blunt
with an intact posterior capsule. The incidence of rhegmatoge- trauma can induce retinal edema (commotio retinae).
Anatomy and Physiology
Basic Science and Translation to Therapy

nous RD increases following YAG capsulotomy.212


Shaken-baby syndrome
Circular macular folds with a sub-ILL schisis cavity containing
Inflammatory serosanguineous material214 and vitreous detachment with ILL
Iridovitreal synechiae have been associated with mild cyclitis, throughout the fundus (Fig. 21.59A), especially at the vitreous
vitritis, retinal phlebitis, and cstoid macular edema.211 base, and peripapillary hemorrhage are typical (Fig. 21.59B).

Fig. 21.58 (A) Retinal lesions caused by blunt trauma transmitted to


retina. A, Dialysis at anterior border of vitreous base. B, Avulsion of
vitreous base. C, Macular hole. D, Horseshoe-shaped tear on posterior
margin of vitreous base. E, Horseshoe-shaped tear at posterior end of
B a meridional fold. F, Horseshoe-shaped tear at equator. G, Tear with
operculum in overlying vitreous. H, Retinal dialysis at posterior border
A
of vitreous base. (Modified with permission from Cox MS, Schepens
D CL, Freeman HM. Retinal detachment due to ocular contusion. Arch
Ophthalmol 1966;76:679–85.) (B) Retinal dialysis at posterior aspect of
E vitreous base with retinal pigment epithelium extending through the
dialysis, along the posterior surface of vitreous (arrowheads), and then
along the anterior surface of the retina (arrows) (hematoxylin and
eosin, ×20). (Reproduced with permission from Clarkson JG, Green
F WR, Massof D. A histopathologic review of 168 cases of preretinal
C membrane. Am J Ophthalmol 1977;84:1–17.)

B
507

Chapter 21
A B

Vitreous and Vitreoretinal Interface


Fig. 21.59 Shaken-baby syndrome. (A) Sub-ILL schisis cavity containing serosanguineous material results from severe trauma in a young
individual with firm vitreoretinal adherence (hematoxylin and eosin, ×457). (B) Extensive subdural hemorrhage that extends from the adjacent
sclera (arrows). A peripapillary retinal hemorrhage (arrowhead) is present (hematoxylin and eosin, ×183).

Posterior penetrating and


perforating trauma215
Wound healing at the perforation site allows fibrocellular pro-
liferation into the eye, inducing traction RD (Fig. 21.60). Histo-
pathologic studies revealed cyclitic and periretinal membranes.216
Intraocular proliferation starts 2–4 days after injury,217 PVD
develops at 1–2 weeks,218 and traction RD occurs at 7–11 weeks.
Poliferation can be prevented by vitrectomy,219 less hazardous
after 2 weeks because of the development of PVD220,221 and more
effective if complete.222 Yet Miller et al.223 found that vitreous
plays a role in normal healing of retinal wounds.

PERIRETINAL PROLIFERATION
Premacular membranes
Primary premacular membranes occur in the absence of associ-
ated conditions and are most likely the result of vitreoschisis,32
where APVD31 splits the posterior vitreous cortex leaving the
outermost layer attached to the macula. The level of this split
can vary, as do the consequences from no effects, such as in the
case of so-called simple epiretinal membranes (Fig. 21.61) which
have no contractile features, to macular pucker.147,160 Smiddy
et al.224 observed the principal cell to be RPE in these cases,
although it is likely that many of these cells are actually hyalo-
cytes (Figs 21.6 and 21.14) and circulating monocytes recruited Fig. 21.60 Posterior perforation with fibrous tissue ingrowth (arrow)
from retinal and choroidal vessels.30,160 Secondary premacular and traction retinal detachment (hematoxylin and eosin, ×25).
membranes occur in association with inflammation, accidental
or surgical trauma, and retinovascular disease. Fibrous astro-
cytes are the predominant cell in secondary preretinal macular Complex membranes
membranes (PMMs). Complex membranes (Fig. 21.64) develop after RD surgery or
Macular pucker (Fig. 21.62) results from PMMs that induce after trauma. Commonly called PVR,227 contraction of this pro-
centripetal tangential (inward to the fovea) traction upon the liferative tissue causes traction RD. Anterior PVR features
macula. Macular pucker can have as many as four separate anterior-loop contraction228 (Fig. 21.9).
centers of retinal contraction225 (Fig. 21.63). Eyes with three or Histopathologically, retinal glial cells gain access to the inner
four contraction centers had significantly more macular thicken- retinal surface via ILL discontinuities, such as at the optic
ing and a higher prevalence of intraretinal cysts than eyes with disc, foveola, along major vessels, and at retinal tufts. Acquired
one or two contraction centers. sites of ILL discontinuity include retinal tags, pits (lamellar
holes), holes, and tears (Fig. 21.65), avulsed retinal vessels,
Retroretinal membranes areas of degenerative remodeling, and retinal lattice. Retinal
These membranes typically occur after RD when RPE grows in glial cells are the principal cells in membranes at the optic
sheets over the undersurface of the retina. Strands of fibrous disc, in simple PMMs, in most secondary membranes associ-
tissue contract and prevent retinal reattachment.226 ated with inflammatory diseases or retinovascular disease, after
Fig. 21.61 Simple premacular membrane. (A)
Section illustrates thin, hypocellular epiretinal
membrane (arrow) and wrinkling of internal
508 limiting lamina (ILL) (hematoxylin and eosin,
×310). (Reproduced with permission from
Clarkson JG, Green WR, Massof D. A
histopathologic review of 168 cases of
Section 1

preretinal membrane. Am J Ophthalmol


1977;84:1–17.) (B) The ILL is discontinuous
(arrow), at which point glial cells extend on to
the inner surface of the retina and form a
thin, fibroglial epiretinal membrane
(arrowhead) with no apparent contraction
(periodic acid–Schiff, ×300).
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.62 Macular pucker. (A) Coronal-plane


optical coherence tomography (color) and
scanning laser ophthalmoscopy (gray-scale)
imaging of macular pucker with prominent
retinal striae in a radial configuration
centered in the fovea. (B) Thick fibroglial
membrane (arrow) that apparently contracted
and produced wrinkling of the internal limiting
lamina (ILL) (arrowheads) and a large gap
(asterisk) in the ILL (periodic acid–Schiff,
×90). (Reproduced with permission from
Clarkson JG, Green WR, Massof D. A
histopathologic review of 168 cases of
preretinal membrane. Am J Ophthalmol
1977;84:1–17.)

A
Focus: 2.93 D Scan angle: 25°

B
509

Chapter 21
Vitreous and Vitreoretinal Interface
A B
Focus: 2.93D Focus:1.91D

C D
Focus: 1.83D Focus: 1.81D
Fig. 21.63 Multifocal macular pucker. Coronal plane optical coherence tomography-scanning laser ophthalmoscopy (OCT-SLO) imaging with
superimposition of OCT (color) on to SLO (gray-scale) images. (A) and (C) are reproduced with permission from Gupta P, Sadun AA, Sebag J.
Multifocal retinal contraction in macular pucker analyzed by combined optical coherence tomography/scanning laser ophthalmoscopy. Retina
2008;28:447–52. (A) Unifocal macular pucker. (B) Bifocal macular pucker with two centers (arrows) of retinal contraction. (C) Trifocal macular
pucker with three centers (arrows) of retinal contraction. (D) Four distinct centers (arrows) of retinal contraction are present.
510
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.64 Complex membranes. Fibrous astrocytes in thick epiretinal membrane after anterior-segment surgery and severe intraocular
inflammation. Membrane contains new collagen (asterisk) and fibrocytes in addition to fibrous astrocytes. Multiple layers of fibrous astrocytes line
inner (vitreal) surface (between arrows). These cells show polarity with basement membrane (circles and bottom right inset), and cells contain
numerous cytoplasmic filaments measuring 8–10 nm (rectangle and bottom left inset). Blood vessels (arrowheads) are also present in membrane
(top left and top right insets). (Main figure ×6600; top insets, paraphenylenediamine hydrochloride stain; top left inset ×160; top middle and top right
insets ×250; bottom left inset ×50 000; bottom right inset ×41 000.) (Reproduced with permission from Michels RG. A clinical and histopathologic
study of epiretinal membranes affecting the macula and removed by vitreous surgery. Trans Am Ophthalmol Soc 1982; 80:580–656.)
511

Chapter 21
Vitreous and Vitreoretinal Interface
A B

Fig. 21.65 Glial cell proliferation. (A) Peripheral retinal hole with glial membrane extending on the inner surface of the retina anteriorly and
posteriorly (periodic acid–Schiff, ×65). (B) Higher-power view of anterior margin of retinal tear, showing glial preretinal membrane (arrow)
extending along the inner surface of the internal limiting lamina (arrowhead) (periodic acid–Schiff; ×190). (Reproduced with permission from
Clarkson JG, Green WR, Massof D. A histopathologic review of 168 cases of preretinal membrane. Am J Ophthalmol 1977;84:1–17.)

Fig. 21.66 Retinal pigment epithelium (RPE)


cells in proliferative vitreoretinopathy.
Pigmented membrane removed from
peripapillary area is composed of RPE
arranged in monolayer. Well-developed
basement membrane (arrows) and basal
laminar infoldings (circle, and top right inset)
are present along retinal surface. Extensive
villous processes are present on apical side
of cells. Numerous apically located
subplasmalemmal microfilaments that
measure 4–5 nm and have fusiform densities
are present (left insets). Occasional
cytoplasmic lipid vacuoles are seen
(arrowhead). (Main figure, ×4500; top left
inset, ×17 000; top right inset, ×17 000;
bottom left inset, ×36 000.) (Reproduced with
permission from Michels RG. A clinical and
histopathologic study of epiretinal
membranes affecting the macula and
removed by vitreous surgery. Trans Am
Ophthalmol Soc 1982;80:580–656.)

photocoagulation or RD surgery, and in retinitis pigmentosa. chemoattractants that stimulate migration of RPE232 and glial
RPE gains access to the vitreous via retinal breaks, the ora cells.233 Incomplete vitrectomy leaves behind hyalocytes (Figs
serrata, and retinal lattice. RPE can also migrate through intact 21.6 and 21.14), which are also the first cells to be exposed to
retina. RPE is the principal cell in PVR (Fig. 21.66), but glial these growth factors and other stimuli. When stimulated, these
cells are present in about 50% of cases. Myofibrocytes (Fig. cells become phagocytic (Fig. 21.68). These resident macrophages
21.67) were observed by electron microscopy in 91% of mem- play an important role in early PVR pathogenesis. Thus, target-
branes studied by Kampik et al.,229 and actin has been observed ing these cells for pharmacotherapy, similar to what has been
in cells of PVR membranes.230 done during vitrectomy for RD,234 may significantly mitigate
Incomplete vitrectomy, intraoperative hemorrhage,217 and PVR. Alternatively, eliminating the role of vitreous via pharma-
excessive cryopexy231 render most cases of PVR an iatrogenic cologic vitreolysis235 will have a great impact in PVR and all
disease. Fibronectin and platelet-derived growth factor are the aforementioned conditions.
Fig. 21.67 Myofibrocyte. There is a
characteristic spindle shape that contains
large aggregates of subplasmalemmal
512 microfilaments measuring 4–5 nm and small
fusiform densities (circle and inset). (Main
figure, ×14 000; inset, ×44 000.) (Reproduced
with permission from Michels RG. A clinical
Section 1

and histopathologic study of epiretinal


membranes affecting the macula and
removed by vitreous surgery. Trans Am
Ophthalmol Soc 1982;80:580–656.)
Anatomy and Physiology
Basic Science and Translation to Therapy

Fig. 21.68 Phagocytic hyalocyte.


Macrophage-like cell with multiple
pleomorphic inclusions contained in
membrane-bound secondary lysosomes
(arrows) is most likely a hyalocyte embedded
within the collagen fibrils of the posterior
vitreous cortex. (Main figure, ×12 000; inset,
×22 600.) (Reproduced with permission from
Michels RG. A clinical and histopathologic
study of epiretinal membranes affecting the
macula and removed by vitreous surgery.
Trans Am Ophthalmol Soc 1982; 80:
580–656.)

8. Bishop PN, Crossman MV, McLeod D, et al. Extraction and characterisation


REFERENCES of the tissue forms of collagen types II and IX from bovine vitreous. Biochem
1. Eyre DR, Apon S, Wu JJ, et al. Collagen type IX: Evidence for covalent linkages J 1994;299:497–505.
to type II collagen in cartilage. Fed Eur Biochem Soc 1987;220:337. 9. Bishop PN, Reardon AJ, McLeod D, et al. Identification of alternatively spliced
2. Sebag J. The vitreous: structure, function, and pathobiology. New York, NY: variants of type II procollagen in vitreous. Biochem Biophys Res Commun
Springer-Verlag; 1989. p. 63–4. 1994;203:289.
3. Itakura H, Kishi S, Kotajima N, et al. Vitreous collagen metabolism before and 10. Bhutto IA, Kim SY, McLeod DS, et al. Localization of collagen XVIII and the
after vitrectomy. Graefes Arch Clin Exp Ophthalmol 2005;243:994–8. endostatin portion of collagen XVIII in aged human control eyes and eyes with
4. Schmut O, Mallinger R, Paschke E. Studies on a distinct fraction of bovine age-related macular degeneration. Invest Ophthalmol Vis Sci 2004;45:1544–52.
vitreous body collagen. Graefes Arch Clin Exp Ophthalmol 1984;221:286. 11. Ohlmann AV, Ohlmann A, Welge-Lussen U, et al. Localization of collagen
5. Swann DA. Chemistry and biology of vitreous body. Int Rev Exp Pathol XVIII and endostatin in the human eye. Curr Eye Res 2005;30:27–34.
1980;22:1. 12. Kim H, Robison SB, Csaky KG. Investigating the movement of intravitreal
6. Snowden JM. The stabilization of in vivo assembled collagen fibrils by human serum albumin nanoparticles in the vitreous and retina. Pharm Res
proteoglycans/glycosaminoglycans. Biochem Biophys Acta 1982;703:21. 2009;26:329–37.
7. Maumenee IH. Vitreoretinal degeneration as a sign of generalized connective 13. Comper WD, Laurent TC. Physiological functions of connective tissue
tissue diseases. Am J Ophthalmol 1979;88:432–49. polysaccharides. Physiol Rev 1978;58:255.
14. Sebag J. Diabetic vitreopathy. Ophthalmology 1996;103:205. 51. Sebag J, Balazs EA, Eakins KE, et al. The effect of Na-hyaluronate on
15. Reardon A, Heinegard D, McLeod D, et al. The large chondroitin sulphate prostaglandin synthesis and phagocytosis by mononuclear phagocytes.
proteoglycans versican in mammalian vitreous. Matrix Biol 1998;17: Invest Ophthalmol Vis Sci 1981;20:33.
325–33. 52. Hultsch E. Vitreous structure and ocular inflammation. In: Silverstein AM, 513
16. Cain SA, Morgan A, Sherratt MJ, et al. Proteomics analysis of fibrillin-rich O’Connor GR, editors. Immunology and immunopathology of the eye. New
microfibrils. Proteomics 2006;6:111–22. York: Masson; 1979.
17. Miyamoto T, Inoue H, Sakamoto Y, et al. Identification of a novel splice site 53. Gartner J. The fine structure of the vitreous base of the human eye and

Chapter 21
mutation of the CSPG2 gene in a Japanese family with Wagner syndrome. pathogenesis of pars planitis. Am J Ophthalmol 1971;71:1317–27.
Invest Ophthalmol Vis Sci 2005;46:2726–35. 54. Sakamoto T, Ishibashi T. Hyalocytes: essential cells of the vitreous cavity in
18. Bishop PN. Structural macromolecules and supramolecular organization of vitreoretinal pathophysiology? Retina 2011;31:222–8.
the vitreous gel. Prog Retinal Eye Res 2000;19:323. 55. Nishitsuka K, Kashiwagi Y, Tojo N, et al. Hyaluronan production regulation
19. Reardon AJ, LeGoff M, Briggs MD, et al. Identification in vitreous and molecu- from porcine hyalocyte cell line by cytokines. Exp Eye Res 2007;85:
lar cloning of opticin, a novel member of the family of leucine-rich repeat 539–45.
proteins of the extracellular matrix. J Biol Chem 2000;275:2123. 56. Kita T, Hata Y, Kano K, et al. Transforming growth factor-beta2 and connec-
20. Sanders EJ, Walter MA, Parker E, et al. Opticin binds retinal growth hormone tive tissue growth factor in proliferative vitreoretinal diseases: Possible
in the embryonic vitreous. Invest Ophthalmol Vis Sci 2003;44:5404–9. involvement of hyalocytes and therapeutic potential of Rho kinase inhibitor.
21. Hindson VJ, Gallagher JT, Halfter W, et al. Opticin binds to heparan and Diabetes 2007;56:231–8.

Vitreous and Vitreoretinal Interface


chondroitin sulphate proteoglycans. Invest Ophthalmol Vis Sci 2005;46: 57. Hiryama K, Hata Y, Noda Y, et al. The involvement of the rho-kinase pathway
4417–23. and its regulation in cytokine-induced collagen gel contraction by hyalocytes.
22. Monfort J, Tardif G, Roughley P, et al. Identification of opticin, a member of Invest Ophthalmol Vis Sci 2004;45:3896–903.
the small leucine-rich repeat proteoglycan family, in human articular tissues. 58. Foos RY. Posterior peripheral retinal tears. Ann Ophthalmol 1974;6:
Osteoarthritis Cartilage 2008;16:749–55. 679–87.
23. Sebag J, Hageman GS. Interfaces. Eur J Ophthalmol 2000;10:1. 59. Kishi S, Numaga T, Yoneya S, et al. Epivascular glia and paravascular holes
24. Eisner G. Biomicroscopy of the peripheral fundus. New York: Springer- in normal human retina. Graefes Arch Clin Exp Ophthalmol 1986;224:
Verlag; 1979. 124–30.
25. Worst JG. Cisternal systems of the fully developed vitreous body in the young 60. Sebag J. Age-related differences in the human vitreoretinal interface. Arch
adult. Trans Ophthalmol Soc UK 1977; 97:550–4.a. Ophthalmol 1991;109:966.
26. Sebag J, Balazs EA. Human vitreous fibres and vitreoretinal disease. Trans 61. Kefalides NA. The biology and chemistry of basement membranes. In:
Ophthalmol Soc UK 1984; 104:123–8. Kefalides NA, editor. Proceedings of the First International Symposium on
27. Gartner J. Electron-microscopic study on the fibrillar network and fibrocyte– the Biology and Chemistry of Basement Membranes. New York: Academic
collagen interactions in the vitreous cortex at the ora serrata of human eyes Press; 1978. p. 215–28.
with special regard to the role of disintegrating cells. Exp Eye Res 1986; 62. Fukai N, Eklund L, Marneros AG, et al. Lack of collagen XVIII/endostatin
42:21–3. results in eye abnormalities. EMBO J 2002;21:1535–44.
28. Sebag J. Surgical anatomy of vitreous and the vitreoretinal interface. 63. Hindson VJ, Gallagher JT, Halfter W, et al. Opticin binds to heparan and
In: Tasman W, Jaeger EA, editors. Duane’s clinical ophthalmology, vol 6. chondroitin sulphate proteoglycans. Invest Ophthalmol Vis Sci 2005;46:
Philadelphia: JB Lippincott; 1994. updated 2008. 4417–23.
29. Streeten BA. Disorders of the vitreous. In: Garner A, Klintworth GK, editors. 64. Ramesh S, Bonshek RE, Bishop PN. Immunolocalisation of opticin in the
Pathobiology of ocular disease – a dynamic approach. New York: Marcel human eye. Br J Ophthalmol 2004;88:697–702.
Dekker; 1982. p. 1381–419. 65. Bhutto IA, Kim SY, McLeod DS, et al. Localization of collagen XVIII and the
30. Gupta P, Yee KMP, Garcia P, et al. Vitreoschisis in macular diseases. Br J endostatin portion of collagen XVIII in aged human control eyes and eyes
Ophthalmol 2011;95:376–80. with age-related macular degeneration. Invest Ophthalmol Vis Sci
31. Sebag J. Vitreous – the resplendent enigma. Br J Ophthalmol 2009;93: 2004;45:1544–52.
989–91. 66. Polk TD, Gass DM, Green WR, et al. Familial internal limiting membrane
32. Sebag J. Anomalous PVD – a unifying concept in vitreoretinal diseases. dystrophy: a new sheen retinal dystrophy. Arch Ophthalmol 1997;115:
Graefes Arch Clin Exp Ophthalmol 2004;242:690–8. 878–85.
33. Sebag J. Vitreoschisis. Graefes Arch Clin Exp Ophthalmol 2008;246:329–32. 67. Foos RY. Vitreoretinal juncture: topographical variations. Invest Ophthalmol
34. Balazs EA, Toth LZ, Eckl EA, et al. Studies on the structure of the vitreous 1972;11:801–8.
body. Exp Eye Res 1964;3:57–71. 68. Roth AM, Foos RY. Surface structure of the optic nerve head. I. Epipapillary
35. Gloor BP. Cellular proliferation on the vitreous surface after photocoagula- membranes. Am J Ophthalmol 1972;74:977–85.
tion. Graefes Arch Clin Exp Ophthalmol 1969;178:99. 69. Cohen AI. Electron microscopic observations of the internal limiting mem-
36. Balazs EA: Structure of vitreous gel. Acta XVII Concilium Ophthalmologicum brane and optic fiber layer of the retina of the rhesus monkey. Am J Anat
1954;11:1019. 1961;108:179.
37. Balazs EA, Sundblad L, Toth LZJ. In vitro formation of hyaluronic 70. Zimmerman LE, Straatsma BR. Anatomic relationships of the retina to the
acid by cells in the vitreous body and by comb tissue. Abstr Red Proc vitreous body and to the pigment epithelium. In: Schepens CL, editor. Impor-
1958;17:184. tance of the vitreous body in retina surgery with special emphasis on reopera-
38. Jacobson B, Osterlin S, Balazs EA. A soluble hyaluronic acid synthesizing tion. St Louis: CV Mosby; 1960. p. 15–28.
system from calf vitreous. Proc Fed Am Soc Exp Biol 1966;25:588. 71. Russell SR, Shepherd JD, Hageman GS. Distribution of glycoconjugates in
39. Osterlin SE. The synthesis of hyaluronic acid in the vitreous. III. In vivo the human internal limiting membrane. Invest Ophthalmol Vis Sci 1991;32:
metabolism in the owl monkey. Exp Eye Res 1968;7:524. 1986.
40. Osterlin SE: The synthesis of hyaluronic acid in the vitreous. IV. Regeneration 72. Fine BS, Tousimus AJ. The structure of the vitreous body and the suspensory
in the owl monkey. Exp Eye Res 1969;8:27. ligaments of the lens. Arch Ophthalmol 1961;65:95–110.
41. Rhodes RH, Mandelbaum SH, Minckler DS, et al. Tritiated glucose incorpora- 73. Teng CC, Chi HH. Vitreous changes and the mechanism of retinal detach-
tion in the vitreous body, lens and zonules of the pigmented rabbit. Exp Eye ment. Am J Ophthalmol 1957;44:335–56.
Res 1982;34:921. 74. Hogan MJ. The vitreous, its structure and relation to the ciliary body and
42. Jacobson B. Identification of sialyl and galactosyl transferase activities in calf retina. Invest Ophthalmol 1963;2:418–45.
vitreous hyalocytes. Curr Eye Res 1984;3:1033. 75. Spencer LM, Foos RY, Straatsma BR. Meridional complexes and associated
43. Newsome DA, Linsemayer TF, Trelstad RJ. Vitreous body collagen. Evidence abnormalities of the peripheral retina. Am J Ophthalmol 1970;70:697–713.
for a dual origin from the neural retina and hyalocytes. J Cell Biol 1976;71: 76. Byer NE. Cystic retinal tufts and their relationship to retinal detachment. Arch
59–67. Ophthalmol 1981;99:1788–90.
44. Hoffmann K, Baurwieg H, Riese K. Uber Gehalt und Verteilung niederund 77. Spencer LM, Straatsma BR, Foos RY. Tractional degenerations of the periph-
hoch molekularer Substanzen in Glaskorper. II. Hock molekulare Substanzen eral retina. In: Transactions of the New Orleans Academy of Ophthalmology,
(LDH, MDH, GOT). Graefes Arch Clin Exp Ophthalmol 1974;191:231. Symposium on Retina and Retinal Surgery. St Louis: Mosby; 1969.
45. Teng CC. An electron microscopic study of cells in the vitreous of the 78. Spencer LM, Foos RY, Straatsma BR. Enclosed bays of the ora serrata: relation-
rabbit eye. Part I. The macrophage. Eye Ear Nose Throat Monthly 1969; ship to retinal tears. Arch Ophthalmol 1970;83:421–5.
48:91. 79. Rutnin U, Schepens CL. Fundus appearance in normal eyes. II. The standard
46. Szirmai JA, Balazs EA. Studies on the structure of the vitreous body. 111. Cells peripheral fundus and developmental variations. Am J Ophthalmol
in the cortical layer. Arch Ophthalmol 1958;59:34. 1967;64:840–52.
47. Grabner G, Baltz G, Forster O. Macrophage-like properties of human 80. Foos RY, Spencer LM, Straatsma BR. Trophic degenerations of the peripheral
hyalocytes. Invest Ophthalmol Vis Sci 1980;19:333. retina. In: Transactions of the New Orleans Academy of Ophthalmology,
48. Balazs EA. Functional anatomy of the vitreous. In: Duane TD, Jaeger EA, Symposium on retina and retinal surgery. St Louis: Mosby; 1969.
editors. Biomedical foundations of ophthalmology, vol 1. Philadelphia: 81. Streeten BW, Bert M. The retinal surface in lattice degeneration of the retina.
Harper and Row; 1982. p. 6–12. Am J Ophthalmol 1972;74:1201–9.
49. Sonoda KH, Sakamoto T, Qiao H, et al. The analysis of systemic tolerance 82. Foos RY, Allen RA. Retinal tears and lesser lesions of the peripheral retina.
elicited by antigen inoculation into the vitreous cavity: vitreous cavity- Am J Ophthalmol 1967;64:643–55.
associated immune deviation. Immunology 2005;116:390–9. 83. Spencer LM, Straatsma BR, Foos RY. Tractional degenerations of the periph-
50. Forrester JV, Balazs EA. Inhibition of phagocytosis by high molecular weight eral retina. In: Transactions of the New Orleans Academy of Ophthalmology,
hyaluronate. lmmunology 1980;40:435. Symposium on Retina and Retinal Surgery. St Louis: Mosby; 1969.
84. Foos RY. Vitreous base, retinal tufts, and retinal tears: pathogenic 122. Sebag J, Wang M, Nguyen D, et al. Vitreopapillary adhesion in macular
relationships. In: Pruett RC, Regan CD, editors. Retina congress. New York: diseases. Trans Am Ophthalmol Soc 2009;107:35–46.
Appleton-Century-Crofts; 1974. 123. Shui Y-B, Holekamp NM, Kramer BC, et al. The gel state of the vitreous and
514 85. Foos RY. Zonular traction tufts of the peripheral retina in cadaver eyes. ascorbate-dependent oxygen consumption – relationship to the etiology of
Arch Ophthalmol 1969;82:620–32. nuclear cataracts. Arch Ophthalmol 2009;127:475–82.
86. Inomata H. Electron microscopic observations of cystoid degeneration in the 124. Friedenwald JS, Bushke W, Michel HO. Role of ascorbic acid in secretion of
peripheral retina. Jpn J Ophthalmol 1966;10:26–40. intraocular fluid. Arch Ophthalmol 1944;29:535–74.
87. Green WR. Retina. In: Ryan SJ, editor. Ophthalmic pathology: an atlas and 125. Gisladottir S, Loftsson T, Stefansson E. Diffusion characteristics of vitreous
Section 1

text, 4th edn. Philadelphia: WB Saunders; 1996. humour and saline solution follow the Stokes Einstein equation. Graefes Arch
88. Bradford JD, Wilkinson CP, Fransen SR. Pseudophakic retinal detachment: Clin Exp Ophthalmol. 2009;247:1677–84.
the relationships between retinal tears and the time following cataract surgery 126. Stefansson E. Physiology of vitreous surgery. Graefes Arch Clin Exp
at which they occur. Retina 1989;9:181–6. Ophthalmol. 2009;247:147–63.
89. Byer NE. Lattice degeneration of the retina. Surv Ophthalmol 1979;23: 127. Balazs EA. Physiology of the vitreous body. In: Schepens CL, editor. Proceed-
213–47. ings of the Second Conference of the Retina Foundation. St Louis: Mosby;
90. Straatsma BR, Zeegen PD, Foos RY, et al. Lattice degeneration of the retina. 1960.
Trans Am Acad Ophthalmol Otolaryngol 1974;78:87–113. 128. Hilding AC. Alterations in the form, movement, and structure of the vitreous
Anatomy and Physiology

91. Semes LP. Lattice degeneration of the retina and retinal detachment. body in aphakic eyes. Arch Ophthalmol 1954;52:699–709.
Basic Science and Translation to Therapy

Optom Clin 1992;2:71–91. 129. Schepens CL. Clinical aspects of pathologic changes of the vitreous body.
92. Burton TC. The influence of refractive error and lattice degeneration on the Am J Ophthalmol 1954;38:8–21.
incidence of retinal detachment. Trans Am Ophthalmol Soc 1990;87: 130. Balazs EA, Denlinger JL. Aging changes in the vitreous. In: Aging and human
143–57. visual function. New York: Alan R Liss; 1982.
93. Parelhoff ES, Wood WJ, Green WR, et al. Radial perivascular lattice degenera- 131. O’Malley P. The pattern of vitreous syneresis: a study of 800 autopsy eyes. In:
tion of the retina. Ann Ophthalmol 1980;12:25–32. Irvine AR, O’Malley C, editors. Advances in vitreous surgery. Springfield, IL:
94. Schepens CL. Diagnostic and prognostic factors as found in pre-operative Charles C Thomas; 1976.
examination. Trans Am Acad Ophthalmol Otolaryngol 1952;56:398–418. 132. Eisner G. Zur Anatomie des Glaskorpers. Graefes Arch Clin Exp Ophthalmol
95. Schepens CL. Subclinical retinal detachments. Arch Ophthalmol 1952;47: 1975;193:33–56.
593–606. 133. Sebag J, Balazs EA. Morphology and ultrastructure of human vitreous fibers.
96. Foos RY, Simons KB, Wheeler NC. Comparison of lesions predisposing to Invest Ophthalmol Vis Sci 1989;30:1867–71.
rhegmatogenous retinal detachment by race and subjects. Am J Ophthalmol 134. Kishi S, Shimizu K. Posterior precortical vitreous pocket. Arch Ophthalmol
1983;96:644–9. 1990;108:979–82.
97. Byer NE. Clinical study of lattice degeneration of the retina. Trans Am Acad 135. Sebag J. Letter to the editor. Arch Ophthalmol 1991;109:1059–60.
Ophthalmol Otolaryngol 1965;69:1064–77. 136. Snowden JM. The stabilization of in vivo assembled collagen fibrils
98. Byer NE. Changes in prognosis of lattice degeneration of the retina. Trans Am by proteoglycans/glycosaminoglycans. Biochem Biophys Acta 1982;703:
Acad Ophthalmol Otolaryngol 1974;78:114–24. 21–5.
99. Foos RY, Simons KB. Vitreous in lattice degeneration of retina. 137. Aguayo J, Glaser B, Mildvan A, et al. Study of vitreous liquefaction by
Ophthalmology 1984;91:452–7. NMR spectroscopy and imaging. Invest Ophthalmol Vis Sci 1985;26:
100. Robinson MR, Streeten BW. The surface morphology of retinal breaks 692.
and lattice retinal degeneration: a scanning electron microscopic study. 138. Ueno N, Sebag J, Hirokawa H, et al. Effects of visible-light irradiation on vitre-
Ophthalmology 1986;93:237–46. ous structure in the presence of a photosensitizer. Exp Eye Res 1987;44:
101. Tillery WV, Lucier AC. Round atrophic holes in lattice degeneration: an 863–70.
important cause of phakic retinal detachment. Trans Am Acad Ophthalmol 139. Kamei A, Totani A. Isolation and characterization of minor glycosaminogly-
Otolaryngol 1976;81:509–18. cans in the rabbit vitreous body. Biochem Biophys Res Commun 1982;109:
102. Morse PH, Scheie HG. Prophylactic cryoretinopexy of retinal breaks. 881–7.
Arch Ophthalmol 1974;92:204–7. 140. Foos RY, Wheeler NC. Vitreoretinal juncture: synchysis senilis and posterior
103. Wang J, McLeod D, Henson DB, et al. Age-dependent changes in the basal vitreous detachment. Ophthalmology 1982;89:1502–12.
retinovitreous adhesion. Invest Ophthalmol Vis Sci. 2003;44:1793–800. 141. Goldmann H. Senile changes of the lens and the vitreous. Am J Ophthalmol
104. Byer NE. Lattice degeneration of the retina. Surv Ophthalmol 1979;23: 1964;57:1–13.
213–47. 142. Takahashi M, Jalkh A, Hoskins J, et al. Biomicroscopic evaluation and
105. Davis MD. Natural history of retinal breaks without detachment. Arch photography of liquefied vitreous in some vitreoretinal disorders. Arch
Ophthalmol 1974;92:183–94. Ophthalmol 1981;99:1555–9.
106. Dobbie G. Discussion. New research on the aetiology and surgery of retinal 143. Gartner J. Electron microscopic observations on the cilio-zonular border of the
detachment. Mod Probl Ophthalmol 1975;15:113. human eye with particular reference to the aging changes. Anat Entwick-
107. Schepens CL. Diagnostic and prognostic factors as found in preoperative lungsgesch 1970;131:263.
examination. Trans Am Acad Ophthalmol Otolaryngol 1952;56:398–418. 144. Sebag J. To see the invisible: the quest of imaging vitreous. Dev Ophthalmol
108. Tolentino FI, Schepens CL, Freeman HM. Vitreoretinal disorders: diagnosis 2008;42:5–28.
and management. Philadelphia: WB Saunders; 1976. 145. Johnson M. Perifoveal vitreous detachment and its macular complications.
109. Byer NE. The peripheral retina in profile: a stereoscopic atlas. Torrence, CA: Trans Am Ophthalmol Soc 2005;103:537–67.
Criterion Press; 1982. 146. Foos RY. Ultrastructural features of posterior vitreous detachment. Graefes
110. Watzke RC. The ophthalmoscopic sign, “white with pressure”: a clinicopatho- Arch Clin Exp Ophthalmol 1975;196:103–11.
logic correlation. Arch Ophthalmol 1961;66:812–23. 147. Sebag J. Vitreous – the resplendent enigma. Br J Ophthalmol 2009;93:
111. Daicker B. Sind die Symptome “weiss mit Druck” und weiss ohne Druck 989–91.
durch die Periphere Netzshantsklerose bedingt? Mod Probl Ophthalmol 148. Favre M, Goldmann H. Zur Genese der hinteren Glaskorperabhebung.
1975;15:82–90. Ophthalmologica 1956;132:86–97.
112. Gartner J. Discussion. New research on the aetiology and surgery of retinal 149. Pischel DK. Detachment of the vitreous as seen with slit-lamp examination.
detachment. Mod Probl Ophthalmol 1975;15:112. Trans Am Ophthalmol Soc 1952;50:329–46.
113. Freeman HM. Fellow eyes of giant retinal breaks. Trans Am Ophthalmol Soc 150. Buzney SM, Weiter JJ, Furukawa H, et al. Examination of the vitreous: a
1978;76:343–82. comparison of biomicroscopy using the Goldmann and El Bayadi-Kajiura
114. Dunker S, Glinz J, Faulborn J. Morphologic studies of the peripheral vitreo- lenses. Ophthalmology 1985;92:1745–8.
retinal interface in humans reveal structures implicated in the pathogenesis 151. Fisher YL, Slakter JS, Friedman RA, et al. Kinetic ultrasound evaluation of the
of retinal tears. Retina 1997;17:124–30. posterior vitreoretinal interface. Ophthalmology 1991;98:1135–8.
115. Foos RY. Vitreoretinal juncture over retinal vessels. Graefes Arch Clin Exp 152. Ortiz RG, Lopez PF, Lambert HM, et al. Examination of macular vitreoretinal
Ophthalmol 1977;204:223–34. interface disorders with monochromatic photography. Am J Ophthalmol
116. Wolter JR. Pores in the internal limiting membrane of the human retina. 1992;113:243–7.
Acta Ophthalmol 1964;42:971–4. 153. Ansari RR, Dunker S, Suh K. Quantitative molecular characterization of
117. Kuwabara T, Cogan DG. Studies of retinal vascular patterns. I. Normal bovine vitreous and lens with non-invasive dynamic light scattering. Exp Eye
architecture. Arch Ophthalmol 1960;64:904–11. Res 2001;73:859–66.
118. Mutlu F, Leopold IH. Structure of the human retinal vascular system. 154. Sebag J, Dunker S, Suh KI, et al. Dynamic light scattering measurements in
Arch Ophthalmol 1964;71:93. vitreous. Invest Ophthalmol Vis Sci 1997;38:662.
119. Heergaard S, Jensen OA, Prause JU: Structure of the vitreal face of the monkey 155. Foos RY. Posterior vitreous detachment. Trans Am Acad Ophthalmol Otolar-
optic disc (Macacca mulatta): SEM on frozen resin-cracked optic nerve heads yngol 1972;76:480–97.
supplemented by TEM and immunohistochemistry. Graefes Arch Clin Exp 156. Sebag J. Floaters and the quality of life. Am J Ophthalmol 2011;152:3–4
Ophthalmol 1988;226:377. 157. Tasman WS. Posterior vitreous detachment and peripheral retinal breaks.
120. Foos RY, Roth AM. Surface structure of the optic nerve head. II. Vitreopapil- Trans Am Acad Ophthalmol Otolaryngol 1968;72:217–24.
lary attachments and posterior vitreous detachment. Am J Ophthalmol 158. Boldrey EE. Risk of retinal tears in patients with vitreous floaters. Am J
1973;76:662–71. Ophthalmol 1983;96:783–7.
121. Wang MY, Nguyen D, Hindoyan N, et al. Vitreopapillary adhesion in macular 159. Novak MA, Welch RB. Complications of acute symptomatic posterior vitreous
hole and macular pucker. Retina 2009;29:644–50. detachment. Am J Ophthalmol 1984;97:308–14.
160. Sebag J, Gupta P, Rosen R, et al. Macular holes and macular pucker: The role 195. Sadda SR, Campochiaro PA, de Juan E, Jr, et al. Histopathological features of
of vitreoschisis as imaged by optical coherence tomography/scanning laser vitreous removed at macular hole surgery. Arch Ophthalmol 1999;117:478–84.
ophthalmoscopy. Trans Am Ophthalmol Soc 2007;105:121–31. 196. Kelly NE, Wendel RT. Vitreous surgery for idiopathic macular holes: results
161. Machemer R. The importance of fluid absorption, traction, intraocular of a pilot study. Arch Ophthalmol 1991;109:654–9. 515
currents, and chorioretinal scars in the therapy of rhegmatogenous retinal 197. Funata M, Wendel RT, de la Cruz Z, et al. Clinicopathologic study of bilateral
detachments. Am J Ophthalmol 1984;98:681–93. macular holes treated with pars plana vitrectomy and gas tamponade. Retina
162. Sigelman J. Vitreous base classification of retinal tears: clinical application. 1992;12:289–98.

Chapter 21
Surv Ophthalmol 1980;25:59–74. 198. Madreperla SA, Geiger GL, Funata M, et al. Clinicopathologic correlation of
163. Halpern JI. Routine screening of the retinal periphery. Am J Ophthalmol a macular hole treated by cortical vitreous peeling and gas tamponade.
1966;62:99–102. Ophthalmology 1994;101:682–6.
164. Byer NE. Clinical study of retinal breaks. Trans Am Acad Ophthalmol 199. Haritoglou C, Reiniger IW, Schaumberger M, et al. Five-year follow-up of
Otolaryngol 1967;71:461–73. macular hole surgery with peeling of the internal limiting membrane: update
165. Smith RE, Ganley JP. Ophthalmic survey of a community. I. Abnormalities of of a prospective study. Retina 2006;26:618–22.
the ocular fundus. Am J Ophthalmol 1972;74:1126–30. 200. Johnson RN, Gass JDM. Idiopathic macular holes: observations, stages of
166. Teng CC, Katzin KM. An anatomic study of the periphery of the retina. formation, and implications for surgical intervention. Ophthalmology 1988;
I. Nonpigmented epithelial cell proliferation and hole formation. Am J 95:917–24.
Ophthalmol 1951;34:1237–48. 201. Jost BF, Hutton WL, Fuller DG, et al. Vitrectomy in eyes at risk for macular

Vitreous and Vitreoretinal Interface


167. Boniuk M, Butler FC. An autopsy study of lattice degeneration, retinal breaks, hole formation. Ophthalmology 1990;97:843–7.
and retinal pits. In: McPherson A, editor. New and controversial aspects of 202. de Bustros S. Vitrectomy for prevention of macular holes. Results of a random-
retinal detachment. New York: Hoeber; 1968. ized multicenter clinical trial. Vitrectomy for Prevention of Macular Hole
168. Spencer LM, Foos RY. Paravascular vitreoretinal attachments: role in retinal Study Group. Ophthalmology 1994;101:1055–9.
tears. Arch Ophthalmol 1970;84:557–64. 203. Cibis GW, Watzke RC, Chua J. Retinal hemorrhages in posterior vitreous
169. Barishak YR, Stein R. Retinal breaks without retinal detachment in autopsy detachment. Am J Ophthalmol. 1975;80:1043–6.
eyes. Acta Ophthalmol 1972;50:147–59. 204. Kroll P, Wiegand W, Schmidt J. Vitreopapillary traction in proliferative
170. Foos RY. Tears of the peripheral retina: pathogenesis, incidence, and classifi- diabetic vitreoretinopathy. Br J Ophthalmol 1999;83:261–4.
cation in autopsy eyes. Mod Probl Ophthalmol 1975;15:68–81. 205. Katz B, Hoyt C. Gaze-evoked amaurosis from vitreopapillary traction. Am J
171. Byer NE. The natural history of asymptomatic retinal breaks. Ophthalmology Ophthalmol 2005;139:631–7.
1982;89:1033–9. 206. Bradford RH, Wilkinson CP. Vitreous opacification after cataract extraction.
172. Neumann E, Hyams S. Conservative management of retinal breaks: a Am J Ophthalmol 1987;103:276–80.
follow-up study of subsequent retinal detachment. Br J Ophthalmol 1972;56: 207. McDonnell PJ, de la Cruz ZC, Green WR. Vitreous incarceration complicating
482–6. cataract surgery: a light and electron microscopic study. Ophthalmology
173. Haimann MH, Burton TC, Brown CK. Epidemiology of retinal detachment. 1986;93:247–53.
Arch Ophthalmol 1982;100:289–92. 208. Osterlin S. Macromolecular composition of the vitreous in the aphakic owl
174. Bohringer HR. Statistiches zu Haufigkeit and Risiko der Netz-hautablosung. monkey eye. Exp Eye Res 1978;26:77–84.
Ophthalmologica 1956;131:331–4. 209. Kawano SI, Honda Y, Negi A. Effects of biological stimuli on the viscosity of
175. Benson WE. Prophylactic therapy of retinal breaks. Surv Ophthalmol the vitreous. Acta Ophthalmol 1982;60:977–91.
1977;22:41–7. 210. Heller MD, Straatsma BR, Foos RY. Detachment of the posterior vitreous
176. Combs JL, Welch RB. Retinal breaks without detachment: natural history, in phakic and aphakic eyes. Mod Probl Ophthalmol 1972;10:23–36.
management, and long-term follow-up. Trans Am Ophthalmol Soc 1982;80: 211. McDonnell PJ, Patel A, Green WR. Comparison of intracapsular and extracap-
64–97. sular cataract surgery: histopathologic study of eyes obtained postmortem.
177. DiBernardo C, Blodi B, Byrne SF. Echographic evaluation of retinal tears in Ophthalmology 1985;92:1208–23.
patients with spontaneous vitreous hemorrhage. Arch Ophthalmol 1992;110: 212. Erie JC, Raecker ME, Baratz KH, et al. Risk of retinal detachment after cataract
511–4. extraction, 1980–2004: a population-based study. Trans Am Ophthalmol Soc
178. Sarrafizadeh R, Hassan T, Ruby AJ. Incidence of retinal detachment and 2006;104:167–75.
visual outcome in eyes presenting with posterior vitreous separation and 213. Cox MS, Schepens CL, Freeman HM. Retinal detachment due to ocular
dense fundus-obscuring vitreous hemorrhage. Ophthalmology. 2001;108: contusion. Arch Ophthalmol 1966;76:678–85.
2273–8. 214. Massicotte SJ, Folberg R, Torczynski E, et al. Vitreoretinal traction and
179. Theodossiadis GP, Velissatopoulos P, Magouritsas N, et al. Behandlung und perimacular retinal folds in the eyes of deliberately traumatized children.
Nachuntersuchung von Netzhautrissen ohne Netz-hautablosung mit den Riss Ophthalmology 1991;98:1124–7.
uberlagerndem abgehobenem Netzhautgefass. Klin Monatsbl Augenheilkd 215. de Juan E, Sternberg P, Michels RG. Penetrating ocular injuries: types of
1977;170:411–5. injuries and visual results. Ophthalmology 1983;90:1318–22.
180. Lincoff H, Kreissig I, Richard G. Treating avulsed vessels with a temporary 216. Winthrop SR, Cleary PE, Minckler DS, et al. Penetrating eye injuries: a histo-
balloon buckle. Am J Ophthalmol 1986;101:90–4. pathological review. Br J Ophthalmol 1980;64:809–17.
181. Krebs I, Brannath W, Glittenberg K, et al. Posterior vitreomacular adhesion: 217. Faulborn J, Topping TM. Proliferations in the vitreous cavity after perforating
a potential risk factor for exudative age-related macular degeneration. Am J injuries: a histopathological study. Graefes Arch Clin Exp Ophthalmol
Ophthalmol 2007;144:741–6. 1978;205:157–66.
182. Robison C, Krebs I, Binder S, et al. Vitreomacular adhesion in active and 218. Cleary PE, Ryan SJ. Histology of wound, vitreous and retina in experimental
end-stage age-related macular degeneration. Am J Ophthalmol 2009;148: posterior penetrating eye injury in the rhesus monkey. Am J Ophthalmol
79–82. 1979;88:221–31.
183. Gass JDM, Norton WD. Cystoid macular edema and papilledema following 219. Abrams GW, Topping TM, Machemer R. Vitrectomy for injury: the effect on
cataract extraction: a fluorescein funduscopic and angiographic study. Arch intraocular proliferation following perforation of the posterior segment of the
Ophthalmol 1966;76:646–61. rabbit eye. Arch Ophthalmol 1979;97:743–8.
184. Irvine AR. Cystoid maculopathy. Surv Ophthalmol 1976;21:1–17. 220. Cleary PE, Ryan SJ. Vitrectomy in penetrating eye injury: results of a con-
185. Jaffe NS. Vitreous traction at the posterior pole of the fundus due to alterations trolled trial of vitrectomy in an experimental posterior penetrating eye injury
in the vitreous posterior. Trans Am Acad Ophthalmol Otolaryngol 1967;71: in the rhesus monkey. Arch Ophthalmol 1981;99:287–92.
642–52. 221. Spalding SC, Sternberg P. Controversies in the management of posterior
186. Sebag J, Balazs EA. Pathogenesis of cystoid macular edema: an anatomic segment ocular trauma. Retina 1990;10:76–82.
consideration of vitreoretinal adhesions. Surv Ophthalmol 1984;28(suppl): 222. Gregor Z, Ryan SJ. Complete and core vitrectomies in the treatment of epireti-
493–8. nal posterior penetrating eye injury in the rhesus monkey. II. Histologic
187. Smiddy WE, Green WR, Michels RG, et al. Ultrastructural studies of vitreo- features. Arch Ophthalmol 1983;101:446–50.
macular traction syndrome. Am J Ophthalmol 1989;107:177–85. 223. Miller B, Miller H, Patterson R, et al. Effect of the vitreous on retinal wound-
188. Patel A, de Bustros S, Michels RG. Pars plana vitrectomy for aphakic cystoid healing. Graefes Arch Clin Exp Ophthalmol 1986;224:576–9.
macular edema. Retina 1985;5:11–5. 224. Smiddy WE, Maguire AM, Green WR, et al. Idiopathic epiretinal mem-
189. Fung WE, Vitrectomy–ACME Study Group. Vitrectomy for chronic aphakic branes: ultrastructural characteristics and clinicopathologic correlation.
cystoid macular edema: results of a national, collaborative, prospective, Ophthalmology 1989;96:811–21.
randomized investigation. Ophthalmology 1985;92:1102–11. 225. Gupta P, Sadun AA, Sebag J. Multifocal retinal contraction in macular
190. McDonnell PJ, Fine SL, Hillis AI. Clinical features of idiopathic macular cysts pucker analyzed by combined optical coherence tomography/scanning laser
and holes. Am J Ophthalmol 1982;93:777–86. ophthalmoscopy. Retina 2008;28:447–52.
191. Bronstein MA, Trempe CL, Freeman HM. Fellow eyes of eyes with macular 226. Wilkes SR, Mansour AM, Green WR. Proliferative vitreoretinopathy: histopa-
holes. Am J Ophthalmol 1981;92:757–61. thology of retroretinal membranes. Retina 1987;7:94–101.
192. Avila MP, Jalkh AE, Murakami K, et al. Biomicroscopic study of the vitreous 227. The Retina Society Terminology Committee. The classification of retinal
in macular breaks. Ophthalmology 1983;90:1277–83. detachment with proliferative vitreoretinopathy. Ophthalmology 1983;90:
193. Morgan CM, Schatz H. Involutional macular thinning: a premacular hole 121–5.
condition. Ophthalmology 1986;93:153–61. 228. Charles S. Vitreous microsurgery. Baltimore: Williams and Wilkins; 1981.
194. Guyer DR, Green WR. Idiopathic macular holes and precursor lesions. In: 229. Kampik A, Kenyon KR, Michels RG, et al. Epiretinal and vitreous mem-
Franklin RM, editor. Symposium on vitreous and retinal disease. Proceedings branes: a comparative study of 56 cases. Arch Ophthalmol 1981;99:
of the 1992 New Orleans Academy of Ophthalmology. New York: Kugler; 1993. 1445–54.
230. Wallow IHL, Stevens TS, Greaser ML, et al. Actin filaments in contracting 233. Harvey AK, Roberge F, Hjelmeland LM. Chemotaxis of rat retinal glia to
preretinal membranes. Arch Ophthalmol 1984;102:1370–5. growth factors found in repairing wounds. Invest Ophthalmol Vis Sci 1987;
231. Campochiaro PA, Gaskin HC, Vinores SA. Retinal cryopexy stimulates 28:1092–9.
516 traction retinal detachment formation in the presence of an ocular wound. 234. Sebag J. Shaken not stirred. Ophthalmology 2001;108:1177–78.
Arch Ophthalmol 1987;105:1567–70. 235. Sebag J. Pharmacologic vitreolysis – premise and promise of the first decade.
232. Campochiaro PA, Glaser BM. Platelet-derived growth factor is chemotactic Retina 2009;29:871–4.
for human retinal pigment epithelial cells. Arch Ophthalmol 1985;103:
576–9.
Section 1
Anatomy and Physiology
Basic Science and Translation to Therapy
Basic Mechanisms of Injury in the Retina Section 2

Mechanisms of Oxidative Stress in Retinal Injury Chapter

Milam A. Brantley Jr, Paul Sternberg Jr


22 
Oxidative stress has been implicated in the development and often not the direct cause of oxidative damage but rather act to
progression of retinal diseases. This chapter will focus on three initiate further oxidative damage by nonradical oxidants. The
forms of retinal pathology that are related to oxidative stress: redox hypothesis describes radical-free oxidative stress in which
age-related macular degeneration (AMD), diabetic retinopathy, disrupted thiol redox circuits interfere with regulation of cellular
and inherited retinal degenerations. We will discuss specific redox status, affecting cell signaling and physiological regula-
mechanisms of oxidative stress that affect the retinal pigment tion. Redox-sensitive thiols include the amino acid cysteine
epithelium (RPE), retinal vasculature, photoreceptors, and mito- (Cys), the Cys-derived disulfide cystine (CySS), the Cys-
chondria. We will also consider the evidence linking oxidative containing tripeptide glutathione (GSH), and glutathione disul-
stress and inflammation in the pathogenesis of retinal disease. fide (GSSG). Sulfur redox couples act as “on/off” switches
Finally, we will explore potential therapies targeting oxidative regulating gene expression and protein function. Because the
stress in the retina. Cys/CySS and GSH/GSSG couples are not in equilibrium, it is
plausible that abnormal levels of nonradical oxidants could be
OVERVIEW OF OXIDATIVE STRESS sufficient to disrupt normal function.6
IN THE RETINA The retina is particularly vulnerable to oxidative stress because
of its high oxygen consumption and high proportion of polyun-
Reactive oxygen species (ROS) are a major source of retinal oxi-
saturated fatty acids (PUFAs).1,2 High light exposure and phago-
dative stress. These highly reactive particles include free radi-
cytosis in the RPE contribute to the oxidative burden of the
cals, peroxides, and singlet oxygen. Free radicals, such as the
retina. The turnover rate of photoreceptors is high, with these
hydroxyl radical (OH•), hydroperoxyl radicals (HO2•), super-
cells shedding about 10% of their outer-segment discs each day.
oxide anion (O2–•), and lipid peroxyl radicals, are strong oxidiz-
The disc membranes, in particular the PUFAs, are subject to
ing agents with an unpaired electron in the outer shell. Peroxides
peroxidation, which is highly damaging to the RPE. Two carot-
(e.g., hydrogen peroxide (H2O2), lipid peroxides) and singlet
enoids, lutein and zeaxanthin, comprise the macular pigment
oxygen (1O2) have a full complement of electrons in an unstable
that protects against ROS in the retina. Lutein and zeaxanthin
state.1
can quench the reactive singlet oxygen and form an optical filter
Under physiologic conditions, the body produces ROS through
that blocks highly damaging blue light from reaching the
normal metabolic processes, such as glycolysis and the Krebs
photoreceptors.1
cycle. Aging and disease may disturb the balance between ROS
generation and clearance, resulting in oxidative damage to mac- RETINAL DISEASES RELATED TO
romolecules.2 The majority of endogenous ROS are produced by
mitochondria through the electron transport chain, which con-
OXIDATIVE STRESS
verts 2–3% of all utilized oxygen into ROS.3 Stimuli such as Oxidative stress contributes to diseases of the retina, including
aging, inflammation, irradiation, air pollutants, and cigarette AMD, diabetic retinopathy, and inherited retinal degenerations
smoke lead to increased ROS, and thus increased cellular oxida- (Table 22.1). To set the stage for a detailed discussion of the
tive injury.1,4,5 underlying mechanisms of oxidative damage to the retina, we
The body’s defense against increasing oxidative stress consists will discuss the pathology and evidence for a role of oxidative
of molecules with antioxidant capacity, including vitamins C stress in each of these diseases.
and E, carotenoids, and other free radical scavengers. Most ROS
are eliminated immediately by antioxidant enzymes, such as Age-related macular degeneration
superoxide dismutase (SOD), glutathione peroxidase, and cata- AMD, the leading cause of irreversible vision loss in older indi-
lase. For example, the superoxide anion, produced by the mito- viduals in the western world, is a complex disease influenced by
chondria during the electron transport stage of cellular factors such as genetics, demographics, and environmental
respiration, is converted to the less noxious hydrogen peroxide exposure. Approximately 1.5% of individuals in the USA over
molecule (H2O2) by SOD.1 Smaller antioxidant molecules (e.g., the age of 40 (about 1.75 million people) develop the sight-
vitamin C (ascorbate), vitamin E (tocopherol), and carotenoids) threatening advanced stages of the disease, and this number is
act on free radicals directly, reducing ROS such as the hydroxyl projected to approach 3 million by 2020.7
radical. AMD can be divided into an early, typically asymptomatic
As most free radical chain reactions are prevented by free form, and a late form that often results in severe central vision
radical-scavenging molecules, it appears that free radicals are loss. In the early stages of AMD, whitish-yellow waste deposits
Table 22.1 Oxidative stress in the pathophysiology of common retinal diseases

518 Disease Supporting evidence

AMD AMD risk factors (e.g., aging, smoking) linked to increased systemic oxidative stress1,2,52,53
Oxidative modifications to proteins and DNA in Bruch’s membrane, drusen, and RPE cells23
Section 2

Light exposure associated with increased generation of ROS and AGEs52,53 and increased risk of AMD1,2
Colocalization of RAGE with AGE deposits and macular disease in AMD retinas55
RAGE-induced secretion of VEGF in RPE cells55
Low macular pigment associated with AMD24–26
Dietary or supplemental intake of antioxidants (e.g., vitamins, carotenoids) and zinc linked to lower risk of AMD
progression20–22
Decreased viability and increased proliferation in cultured RPE and choroidal endothelial cells exposed to the oxidant
t-BHP66
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

Increased proliferation and VEGF upregulation in choroidal endothelial cells exposed to AGEs67

Diabetic Mitochondrial overproduction of superoxide potentially disruptive to multiple pathways implicated in diabetes (e.g.,
retinopathy polyol, AGE, protein kinase C, hexosamine)36
Prevention of early retinal cell death by superoxide inhibition33,34
Involvement of oxidative stress-activated caspases and NF-κB in retinal cell death31,32
Increased expression of oxidative stress markers in endothelial cells and pericytes in high-glucose environment32
Impaired glucose transport in H2O2-exposed retinal endothelial cells65
Link between the oxidant peroxynitrite and upregulated VEGF expression in endothelial cells60–62

Inherited retinal Increased oxygen concentration in outer retina of multiple animal models of retinal degeneration68–70
degenerations Cone damage due to increased retinal oxygen levels71,83,84
Link between thioredoxin antioxidant defense and a rod-derived cone survival factor45,87
Increased ROS and decreased GSH in an in vitro model of photoreceptor apoptosis46
Downregulation of DNA repair mediators in the rd1 mouse retina82
Reduction in cone cell loss and preservation of cone ERGs seen in rd1 mice treated with antioxidants85
Decreased antioxidant defense (e.g., GST and GSHPx) in rd1 mice80,81
AMD, age-related macular degeneration; RPE, retinal pigment epithelium; ROS, reactive oxygen species; AGE, advanced glycation end-product; RAGE, receptor
of AGEs; VEGF, vascular endothelial growth factor; t-BHP, tert-butyl hydroperoxide; NF-κB, nuclear factor-κB; GSH, glutathione; ERGs, electroretinograms;
GST, glutathione-S-transferase; GSHPx, glutathione peroxidase.

called drusen accumulate in the macula, usually localized macular pigment in AMD patients.24–26 Significantly lower levels
between the RPE and Bruch’s membrane. These deposits may be of macular pigment (lutein and zeaxanthin) were found by direct
small and discrete (hard drusen) or larger and more confluent measurement in AMD versus control eyes at autopsy.24 Addi-
(soft drusen). Drusen may also be present between the photore- tionally, dietary supplementation with antioxidants (vitamin C,
ceptors and RPE or within the photoreceptor cell layer (reticular vitamin E, and β-carotene) and zinc was shown to decrease the
drusen).8,9 Histologically, drusen consist of numerous proteins risk of progression to advanced AMD in the Age-Related Eye
(e.g., complement, immunoglobulins, amyloid-β) and lipids Disease Study (AREDS).22 In past observational studies, high
(e.g., phospholipids, cholesterol, apolipoproteins).10,11 Early dry dietary intake of lutein/zeaxanthin and omega-3 fatty acids has
AMD may develop into advanced dry or advanced wet AMD. corresponded to reduced prevalence and incidence of AMD.27
Advanced dry AMD (geographic atrophy) is marked by cell These compounds are now under investigation for the treatment
death in the choriocapillaris and overlying RPE. Advanced wet of AMD in the AREDS2 clinical trial.
AMD is characterized by choroidal neovascularization, in which
abnormal choroidal vessels extend into the subretinal space. Diabetic retinopathy
Risk factors for AMD include older age, high body mass index, Diabetic retinopathy, a common microvascular complication of
and high light exposure.12,13 Smoking, the strongest environmen- both type 1 and type 2 diabetes mellitus (DM), is the leading
tal risk factor for AMD, has been linked to disease onset and cause of vision loss in working-age adults.28 In the USA, approxi-
progression in multiple large, epidemiologic studies.14–19 Addi- mately 26 million people are living with diabetes, and the
tionally, high dietary intake of carotenoids and antioxidant sup- number of diabetic adults with retinopathy is projected to triple
plementation have been linked with lower risk of AMD.20,21 to 16 million by the year 2050.29 The majority of DM (90–95%) is
Multiple lines of evidence point to the role of oxidative stress represented by type 2 diabetes, which involves impaired insulin
in the pathogenesis of AMD. Several AMD risk factors, particu- action at target tissues and impaired insulin release. In contrast,
larly aging, smoking, and light exposure, have been linked to the primary cause of type 1 diabetes is the autoimmune destruc-
increased levels of oxidative stress.1 Additionally, proteome tion of pancreatic β cells.
analysis of human donor eyes revealed oxidative modifications Clinically, diabetic retinopathy is defined as the presence of
to proteins and DNA in Bruch’s membrane, drusen, and RPE.23 specific retinal microvascular signs in an individual with DM.
The higher prevalence of these oxidative changes in AMD The nonproliferative form is characterized by microaneurysms,
patients versus controls23 suggests an increased oxidative state hemorrhages, hard exudates, cotton-wool spots, venous dilation
in the retinas of AMD patients. and beading, and intraretinal microvascular abnormalities. The
Impairment of the retinal antioxidant defense system may also more severe proliferative form is characterized by retinal neo-
play a role in AMD, as some studies have reported decreased vascularization, which can lead to vitreous hemorrhage and
tractional retinal detachment. Intraretinal macular edema, which a proinflammatory transcription factor, and transforming growth
may be present in either form of retinopathy, can also lead to factor-β1 (TGF-β), which can contribute to the occlusion of
severe vision loss. capillaries seen in diabetic retinopathy.36,38 519
Interestingly, retinal capillary cells in diabetics undergo accel- The hexosamine biosynthesis pathway may also mediate
erated apoptosis that precedes the detection of any of the histo- some of the toxic effects of hyperglycemia. The inhibition of

Chapter 22
pathological changes characteristic of diabetic retinopathy.30 GADPH by ROS causes diversion of glycolytic metabolites to
Although the exact mechanism for this accelerated apoptosis the hexosamine pathway, producing uridine diphosphate
remains uncertain, studies have pointed to the involvement of N-acetylglucosamine. N-acetylglucosamine is added to serine
oxidative stress-activated caspases and nuclear factor-κB (NF- and threonine residues of transcription factors, often resulting
κB) in retinal cell death.31,32 Additional experiments revealed that in pathological changes in gene expression. Examples include
inhibition of superoxide accumulation in diabetes prevents this increased expression of TGF-β and plasminogen activator
early retinal cell death.33,34 inhibitor-1.36

Mechanisms of Oxidative Stress in Retinal Injury


Oxidative stress appears to be a major risk factor for the onset Hyperglycemia-induced alterations in each of these
and progression of diabetes. Many of the common DM risk DM-associated pathways can be linked to oxidative stress.
factors, including obesity and age, foster an oxidative environ- Toward that end, Brownlee has proposed a “unifying mecha-
ment that may alter insulin sensitivity by increasing insulin nism” that hypothesizes that all of these pathway changes are a
resistance or impairing glucose tolerance. Hyperglycemia, a result of mitochondrial superoxide overproduction in the endo-
common result of both types of diabetes, contributes in turn to thelial cells of both large and small blood vessels.41 In this model,
the progression and maintenance of an overall oxidative diabetic hyperglycemia causes increased production of ROS by
environment.35 mitochondria. Greater concentration of ROS in the body induces
Hyperglycemia may contribute to oxidative stress by generat- nuclear DNA strand breaks. This DNA damage activates the
ing ROS directly or disrupting the cellular redox balance. This DNA repair enzyme poly(ADP-ribose) polymerase, which
hyperglycemia-induced oxidative stress can occur via several reduces GADPH activity by the addition of ADP ribose poly-
well-studied mechanisms, including increased polyol pathway mers. Decreased GAPDH activity results in a backup of the
flux, increased intracellular formation of advanced glycation end glycolytic pathway and activation of the polyol, AGE, PKC, and
products (AGEs), activation of protein kinase C (PKC), and dis- hexosamine pathways, discussed above.36
turbance of the hexosamine biosynthesis pathway. The overpro-
duction of superoxide by the mitochondrial transport chain has Inherited retinal degenerations
been proposed as a possible root cause for these metabolic Inherited retinal degenerations are a group of disorders char-
changes.36 acterized by primary and progressive loss of photoreceptor
The polyol pathway leads to the reduction of glucose to sor- cells, leading to irreversible vision loss. Over 170 genes and
bitol via aldose reductase in an nicotinamide adenine dinucleo- greater than 200 loci have been associated with inherited retinal
tide phosphate (NADPH)-dependent manner. Sorbitol is then degenerations (www.sph.uth.tmc.edu/retnet). The proteins
oxidized to form fructose by sorbitol dehydrogenase. The polyol encoded by these genes are diverse in function, including not
pathway consumes NADPH, an essential cofactor for regenerat- only proteins required for phototransduction but also structural
ing the intracellular antioxidant GSH. By decreasing the amount proteins, RNA splicing factors, intracellular trafficking mole-
of free GSH, the polyol pathway may increase the susceptibility cules, and proteins involved in the phagocytosis and regulation
of cells to oxidative injury.36 of intracellular pH.42
AGEs are modifications of proteins or lipids that become non- The most common form of these diseases, retinitis pigmentosa
enzymatically glycated and oxidized after contact with aldose (RP), which has an estimated prevalence of around 1 in 4000,43
sugars. Early glycation and oxidation result in the formation of involves a primary insult to rods that initiates rod cell death
Schiff bases and Amadori products. Further glycation of proteins followed by cone cell death (rod–cone dystrophy). Other forms
and lipids causes molecular rearrangements that lead to the of inherited retinal degeneration include cone dystrophy, cone–
generation of AGEs.37 AGEs appear to damage cells by modify- rod dystrophy, and macular dystrophy. The most common
ing intracellular proteins, such as transcription factors, and alter- inherited macular dystrophy is Stargardt macular dystrophy, an
ing extracellular proteins and extracellular matrix molecules. autosomal recessive disease with a carrier frequency of approxi-
These modified proteins can then bind and activate receptor of mately 1 in 50.44
AGEs (RAGE), resulting in the upregulation of the transcription The first symptom of RP is typically night blindness, followed
factor NF-κB and the production of inflammatory cytokines and by progressive loss of the peripheral visual field. Fundoscopic
growth factors.36 exam classically demonstrates a pale optic nerve, attenuated
The activation of PKC leads to multiple characteristics of dia- retinal vessels, and bone spicule pigmentation in the retinal
betic retinopathy, including increased vascular permeability, midperiphery. The widespread retinal degeneration leads to
endothelial cell proliferation and apoptosis, and neovasculariza- diminution or abolishment of the a-waves and b-waves on elec-
tion.38 Hyperglycemia-induced ROS inhibit the key glycolytic troretinograms (ERGs). A variety of molecular defects has been
enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH), identified in individuals with RP, including mutations in genes
leading to an increase in glycolytic pathway precursors. Increased encoding rhodopsin, peripherin, and cGMP phosphodiesterase.
levels of triose phosphate upregulate diacylglycerol, the primary These genes are typically rod-specific, but some also code for
activator of PKC.38,39 Activated PKC can induce expression of more general proteins, such as structural proteins in cilia and
vascular endothelial growth factor (VEGF),40 a major effector their associated transport molecules.
of blood flow and vessel permeability and a primary stimulator The unifying feature in animal models and human cases of RP
of neovascularization. PKC also leads to the synthesis of NF-κB, seems to be the apoptotic cell death of rod photoreceptors,
although the mechanisms that lead to photoreceptor cell death the RAGE axis in RPE cells could contribute to neovascular
in RP remain poorly defined. While most mutations in RP spe- macular disease.55
520 cifically affect rods, both rods and cones die. Several mecha- Further evidence supports the idea that oxidative stress in the
nisms have been proposed for the dependence of cone survival RPE has a significant influence on VEGF regulation. Treatment
on the presence of functioning rods. One theory is that rods of cultured human RPE cells with the oxidant DL-buthionine-
produce a survival factor that is required by cones and that (S,R)-sulfoximine caused a significant decrease in intracellular
Section 2

modulates the thioredoxin antioxidant defense system in the GSH and GSH/GSSG ratios. This change in thiol redox status
retina.45 Oxidative stress or alterations in the cellular redox state was associated with increased VEGF-A secretion as well as sig-
have also been proposed by several groups as a common media- nificant induction of VEGF receptors VEGFR-1 and VEGFR-2.56
tor of photoreceptor cell death.46 Several lines of evidence Both VEGF-A and VEGF-C were upregulated in cultured RPE
support this hypothesis, including the induction of apoptosis by cells treated with t-BHP, with secretion higher to the apical side
oxidants such as H2O2 and the inhibition of apoptosis by than to the basal side,57 suggesting that VEGF may directly affect
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

antioxidants. the photoreceptors. VEGF secretion in RPE cells is regulated, in


part, by the mitogen-activated protein kinases, including c-Jun-
activated kinase, p38, and Erk. Studies in cultured RPE cells
OXIDATIVE INJURY TO THE RETINA
demonstrated that constitutive VEGF secretion is regulated by
Specific mechanisms of oxidative damage to the retina are p38, while oxidative stress-induced VEGF secretion is regulated
unique to the affected retinal cell types. Morphology and func- by both p38 and Erk.58 Finally, it appears that autocrine VEGF-A
tion of the RPE, retinal vasculature, and photoreceptors can be can enhance RPE cell survival under oxidative stress, utilizing
severely impaired by the increased oxidative stress associated the autocrine VEGF-A/VEGFR-2/PI3K/Akt pathway.59
with disease. Growing evidence suggests that the mitochondria
of retinal cells contribute to oxidative stress-related pathology Retinal vasculature
and that inflammatory processes are closely tied to pathologic Increased oxidative stress can lead to alterations in both the
oxidative stress in the retina. retinal and choroidal vasculature. In diabetes, hyperglycemia-
induced ROS may link elevated glucose and multiple metabolic
Retinal pigment epithelium abnormalities associated with the development of retinopathy.41
Oxidative stress can lead to the accumulation of debris and Retinal capillary cell cultures containing endothelial cells and
ultimately cell death in the RPE, as manifest in the drusen pericytes incubated in high-glucose medium exhibited increased
and geographic atrophy of AMD. Abnormally high concentra- oxidative stress markers. In these cells, increased caspase-3 and
tions of oxidized products may also interfere with the ability apoptosis were seen after 5 days in culture.32
of RPE cells to regulate key angiogenic factors, resulting in Oxidative stress has been correlated with increased produc-
retinal neovascularization. tion of the angiogenic growth factor and vascular permeability
Oxidative damage to the RPE can be induced by oxidizing factor VEGF and may be involved in the upregulation of
agents or light. Under normal conditions, phagocytosis of pho- endothelial cell VEGF expression seen in diabetes.60 It appears
toreceptor outer segments by the RPE generates ROS via the that increased VEGF expression in vascular endothelial cells
NADPH oxidase system, leading to increased intracellular H2O2 may result from peroxynitrite, a highly reactive oxidant formed
and increased catalase activity.47 The phosphoinositide 3-kinase by the combination of superoxide anion with nitric oxide.61
(PI3K)-Akt pathway may protect RPE cells from such oxidative Peroxynitrite mediates a variety of biological processes includ-
stress. The oxidant H2O2 was demonstrated to induce PI3K- ing inhibition of key metabolic enzymes, lipid peroxidation,
dependent Akt activation in cultured RPE cells. Akt activation nitration of protein tyrosine residues, and oxidation of thiol
can enhance RPE survival by phosphorylating and thus inacti- pools.62
vating multiple proapoptotic factors.48 Cultured human RPE A hyperglycemic environment may promote the damaging
cells exhibited signs of senescence when treated with the oxidiz- effects of VEGF observed in diabetes. Interestingly, VEGF has
ing agents tert-butyl hydroperoxide (t-BHP)49 or H2O2,50 suggest- been shown to be a potent survival factor for endothelial cells in
ing that oxidative stress may contribute to the RPE senescence vitro and in vivo.63 Despite its neuroprotective properties, the
seen in AMD. Oxidative stress induced by hypoxia/reoxygenation increased levels of VEGF seen in diabetic retinas64 are associated
in cultured human RPE cells led to the accumulation of extracel- with increased retinal endothelial cell death. Cultured endothe-
lular matrix and may be linked to the thickening of Bruch’s lial cells subjected to serum withdrawal-induced apoptosis were
membrane seen in early AMD.51 Additionally, animal and labo- protected by VEGF in normal glucose but not in high-glucose or
ratory studies have shown that blue light damages the RPE peroxynitrite media.62 Further experiments determined that
through ROS generation.52 RPE cells treated with subapoptotic high-glucose treatment blocks the prosurvival effect of VEGF
levels of ultraviolet light showed increases in both ROS and and causes accelerated endothelial cell apoptosis via the action
AGEs.53 of peroxynitrite.62
The AGE-induced activation of RAGE may play a key role in Oxidative stress can also affect retinal endothelial cells’ ability
RPE apoptosis. In human donor eyes, RAGE were found to to transport glucose into the cell. Exposure of endothelial cells
colocalize with AGE deposits and macular disease in AMD to sustained oxidative stress in the form of H2O2 resulted in
retinas, while normal retinas displayed little or no immunolabel- decreased glucose transport activity due to increased internal-
ing for either AGE or RAGE. This study also showed that AGEs ization of GLUT1, the most common retinal glucose transporter.65
stimulate RAGE-mediated activation in cultured RPE cells in a H2O2-induced oxidative stress was also found to increase the rate
dose-dependent fashion.54 RAGE increased secretion of VEGF in of GLUT1 internalization by a proteasome-dependent mecha-
cultured RPE cells, suggesting that AGE-mediated activation of nism involving inactivation of Akt.
Oxidative stress may have direct and indirect effects on the and OGG1 and CREB have both been found to be downregu-
choroidal endothelial cells (CEC), and thus the choroidal neo- lated in the rd1 retina. Since OGG1 can be inactivated by cal-
vascularization seen in AMD. Treatment of cultured RPE and pains, an increase in calpain activity could severely compromise 521
CEC with the oxidant t-BHP resulted in decreased viability a cell’s ability to repair oxidative stress-induced DNA damage.
and increased proliferation of both cell types.66 RPE cells Cones appear to be sensitive to retinal oxygen concentration.

Chapter 22
exposed to t-BHP for 24 hours were found to release basic In multiple animal models of RP, increased levels of oxygen in
fibroblast growth factor, a prominent proangiogenic factor. This the retina result in progressive oxidative damage to cones.71,83,84
finding suggests that oxidative stress may stimulate choroidal Treatment of rd1–/– mice with a cocktail of antioxidants down-
neovascularization via RPE-mediated growth factor release. regulated markers of oxidative damage in cones, reduced cone
Interestingly, CECs exposed to oxidative stress-induced AGEs cell loss, and preserved cone ERGs. These results suggest that
in culture demonstrated increased proliferation as well as oxidative damage is important in cone death regardless of the
upregulation of VEGF, suggesting that AGEs may also promote underlying mechanism that kills rods.85 Most recently, studies in

Mechanisms of Oxidative Stress in Retinal Injury


choroidal neovascularization.67 rd1–/– mice showed that NADPH oxidase plays a critical role in
generation of the oxidative stress that leads to cone death in RP.85
Photoreceptors Initial experiments investigating the progressive death of
The process of retinal degeneration disrupts the physiologic cones after rod cell death showed that grafting of normal pho-
balance of oxidants and antioxidants in the retinal spaces. As a toreceptors (97% rods) into the eye of the rodless rd1–/– mouse
source of ROS, photoreceptors contribute to the oxidative burden before the cones degenerated had a positive effect on the host
of retina, particularly in a pathologic oxidative environment. The cones, even at some distance from the transplant.86 Further
vitality of rods and cones is directly affected by the redox status studies identified a protein that allowed for cone survival,
of surrounding tissue. termed rod-derived cone viability factor (RdCVF).45 RdCVF,
Photoreceptor cell loss or a reduction in energy-demanding encoded by the exon 1 of the nucleoredoxin-like 1 (Nxnl1) gene,
activities like phototransduction can lead to elevated tissue corresponds to a truncated thioredoxin (TRX)-like protein.
oxygen concentrations because choroidal blood vessels are not Because TRX proteins are key to maintaining a reducing intracel-
autoregulated by local oxygen levels. Accordingly, increases in lular environment,87 disruption of TRXs can lead to conditions
outer retinal oxygen concentrations have been confirmed in mul- of increased oxidative stress. Nxnl1 knockout mice have been
tiple animal models.68–70 The inner retinal arterioles, in contrast, shown to be sensitive to oxidative stress, suggesting that RdCVF
do autoregulate, leading to their attenuation in the presence of is part of an endogenous redox-based signaling pathway
high oxygen concentrations.71 This oxygen imbalance induced involved in retinal maintenance.87
by photoreceptor dysfunction is manifest by the thinned retinal
vessels seen clinically in RP. Mitochondria
Alterations in the cellular redox state appear to mediate pho- Cumulative oxidative damage to mitochondria appears to play
toreceptor cell death. An in vitro model of photoreceptor apop- a role in aging and retinal diseases.88 Mitochondria are especially
tosis demonstrated an early and sustained increase in intracellular susceptible to oxidative injury due to the high concentration of
ROS accompanied by a rapid depletion of intracellular GSH.46 ROS produced by the electron transport chain and the limited
Evidence suggests that multiple oxidative stress-related mecha- capacity for mitochondrial DNA (mtDNA) repair.89 RPE cells
nisms underlie the programmed cell death of photoreceptors. treated with ultraviolet light demonstrated conformational
Classically, caspase-dependent apoptotic pathways have been changes in mitochondria,53 and oxidized photoreceptor outer
described.72,73 More recently, it has been shown that programmed segments in in vitro damaged RPE mtDNA.90 Additionally, vari-
cell death can also be accomplished through caspase-independent ations in the mitochondrial genome may modulate ROS produc-
apoptosis,74–76 as well as via autophagy.77 In a photoreceptor cell tion and thus susceptibility to disease.
line treated with the nitric oxide donor sodium nitroprusside, Accumulating evidence, reviewed in Table 22.2, suggests a
cytosolic calcium levels increased during photoreceptor apopto- role for mitochondrial damage in the pathophysiology of both
sis, leading to activation of caspases as well as the calcium- AMD and diabetic retinopathy.88 Abnormalities in the number
dependent proteases calpains.78 Inhibitors of both caspases78 and and structure of mitochondria in the RPE have been reported in
calpains,79 applied independently to the same photoreceptor cell studies of AMD donor eyes.91 Proteomic analyses have sug-
line, failed to prevent apoptosis. These results indicate that, in gested that mitochondrial proteins92 and mtDNA93 are altered in
addition to the classic caspase-dependent pathways of apopto- AMD, and long-extension polymerase chain reaction (PCR) con-
sis, calpain activity may be crucial for apoptosis. firmed that mtDNA is increasingly damaged with AMD pro-
Calpains may lead to retinal oxidative damage by impairing gression.94 Oxidative damage to mitochondria can lead to
DNA repair mechanisms. Studies in retinal degeneration (rd1) apoptosis in human RPE cells,95 and decreased levels of the
mice, which have a mutation in exon 7 of the beta subunit of the mitochondrial antioxidant manganese SOD (MnSOD) led to the
rod photoreceptor PDE6 gene, have demonstrated downregula- development of dry AMD-like lesions in mice.96
tion in defensive mechanisms against oxidative stress (e.g., In the case of diabetes, hyperglycemia causes excess produc-
glutathione-S-transferase and glutathione peroxidase), poten- tion of superoxide by the mitochondrial electron transport chain,
tially allowing an increase in intracellular ROS.80,81 A major inhibiting GAPDH activity.36 As mentioned previously, Brown-
product of ROS reactions in the DNA is the highly mutagenic lee has proposed that this excess of mitochondrial superoxide in
oxidated nucleotide 8-oxo-guanosine (8-oxoG). Mismatches of endothelial cells of both large and small blood vessels leads to
8-oxoG with adenine or cytosine are corrected by the specific an array of complications seen in diabetes, including increased
DNA repair enzyme 8-oxoG DNA glycosylase (OGG1).82 OGG1 activity of the polyol pathway and increased production of
is regulated by cAMP response element-binding protein (CREB), AGEs. Specifically, hyperglycemia causes increased oxidation of
Table 22.2 Evidence of mitochondrial oxidative stress in 4917G polymorphism associated with haplogroup T indepen-
age-related macular degeneration and diabetic retinopathy dently predicted the presence of AMD (OR 2.16, 95% CI
522 1.21–3.91).101 Subsequently, PCR analysis of mtDNA from AMD
Disease Supporting evidence
and control donor retinas found that single-nucleotide poly-
AMD Light-induced conformational changes in
morphisms T16126C and A73G, associated with haplogroups
J and T, were more frequent in AMD retinas than in normal
Section 2

mitochondria of RPE cells53


Damage to RPE mtDNA by oxidized retinas.102 The authors then replicated these associations in blood
photoreceptor outer segments90 DNA from 99 cases and 92 controls.102 The mtDNA haplogroup
Alterations in mitochondrial proteins92 and
mtDNA93,94 in AMD T was associated with risk of retinopathy among type 2 diabet-
Apoptosis in human RPE cells induced by ics.103 As this haplogroup has been associated with multifactorial
oxidative damage to mitochondria95 age-related diseases such as coronary artery disease103 and
Development of dry AMD-like lesions in mice
AMD,101 it seems plausible that carriers of haplogroup T may
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

with decreased levels of MnSOD96


Association of multiple mitochondrial haplogroups be exposed to greater oxidative stress or have greater sus­
(H, J, U, T) with AMD99–102 ceptibility to oxidative damage than individuals with other
haplogroups.
Diabetic Enhanced permeability of the mitochondrial
The A69S polymorphism in the ARMS2 gene, which has been
retinopathy membrane due to hyperglycemia-induced
oxidative stress in retinal endothelial cells and linked to mitochondrial function in some studies, is strongly
pericytes97 associated with AMD.104,105 ARMS2 mRNA encoding a 12-kDa
Protective effects of MnSOD on retinal protein has been detected in human retina. Cell culture experi-
antioxidant capacity98 and vascular pathology33
Association of mtDNA haplogroup T with risk of ments have localized this protein to the mitochondrial outer
retinopathy among type 2 diabetics103 membrane106 and specifically to mitochondria in the ellipsoid
AMD, age-related macular degeneration; RPE, retinal pigment epithelium;
region of the photoreceptors.107 These findings suggest a func-
mtDNA, mitochondrial DNA; MnSOD, manganese superoxide dismutase. tional role for ARMS2 in mitochondrial homeostasis and have
led to an attractive hypothesis linking ARMS2 and oxidative
stress. Such a connection fits well with previous reports of an
glucose, leading to an elevation in voltage gradient across the interaction between ARMS2 A69S and cigarette smoking.108,109
mitochondrial membrane. Once a critical threshold in voltage The A69S variant plus smoking was shown to confer a higher
gradient is reached, electron transfer inside complex III of the risk of AMD than either factor alone, suggesting a link between
electron transport chain is blocked. The electrons accumulate at the ARMS2 protein and smoking-induced oxidative stress. The
coenzyme Q, which donates them to molecular oxygen, creating localization of ARMS2 to the mitochondria has been called into
large amounts of superoxide. Ultimately, the increased number question, however, by a recent immunohistologic study that
of ROS in diabetes can alter the mitochondrial membrane found no colocalization of ARMS2 antibodies and mitochondrial
potential, leading to cell death.36 markers, instead reporting localization of the ARMS2 protein to
Animal studies have demonstrated the effects of the cytosol in cultured RPE cells.110 Due to potential inconsisten-
hyperglycemia-induced impairment of the mitochondrial mem- cies in antibodies and visualization, further investigation is nec-
brane. Increased levels of cytochrome c, a key component of essary to confirm or refute the relationship between ARMS2 and
the electron transport chain and the precursor to capase-9 in the mitochondria.
the apoptotic cascade, were detected in rats after 8 months of
diabetes. These rats also had increased levels of the proapoptotic Oxidative stress and inflammation
Bcl-2-associated X protein (BAX) in the mitochondria. In retinal Growing evidence from both cell culture and animal studies
endothelial cells and pericytes, high concentrations of glucose points to a mechanistic link between oxidative stress and inflam-
enhanced the permeability of the mitochondrial membrane, mation (Table 22.3). Decreased SOD2 and increased superoxide
increasing levels of cytochrome c in the cytosol and BAX in the production were associated with mononuclear phagocyte-
mitochondria.97 These glucose-mediated responses were inhib- induced apoptosis of the RPE in sod2+/– mice.111 In mice with
ited by the antioxidant MnSOD. A subsequent study by the oxygen-induced retinopathy, injections of the serine proteinase
same group showed that increased expression of MnSOD can inhibitor SERPINA3K reduced expression of proinflammatory
prevent the reduction of GSH levels and total antioxidant capac- factors (e.g., VEGF, tumor necrosis factor-α, and intercellular
ity caused by diabetes.98 In nontransgenic diabetic mice, upregu- adhesion molecule-1), decreased ROS production, and increased
lation of MnSOD protected against diabetes-related changes in SOD and GSH levels.112 The anti-inflammatory effect of SERPI-
the mitochondria and reduced vascular pathology.33 NA3K was also demonstrated in cultured retinal cells exposed
Genetic association studies have indicated that polymor- to hypoxia.112
phisms in mtDNA influence the risk of developing AMD and Immunologic and inflammatory processes appear to play a
diabetic retinopathy. In the Blue Mountain Eye Study, mito- major role in AMD, including drusen formation, complement
chondrial haplogroup H was associated with a reduced preva- activation, recruitment of tissue-destructive macrophages, and
lence of any AMD (odds ratio (OR) 0.75, 95% confidence interval microglial activation and accumulation.113 The association of
(CI) 0.58–0.97) and early AMD (OR 0.75, 95% CI 0.57–0.98), multiple complement gene polymorphisms with AMD,114–128 the
whereas haplogroups J and U were associated with early AMD presence of complement proteins in drusen,10,11 and the link
only.99 A US study determined that individuals with the T2 between systemic complement activation and AMD129–131 suggest
haplogroup were 2.5 times more likely to have advanced AMD that innate immunity plays a critical role in AMD pathogenesis.
than those without the haplogroup (OR 2.54, 95% CI 1.36–4.80).100 These discoveries have led to experiments designed to define the
This finding was supported by studies showing that the mtDNA role of oxidative stress in complement activation in the RPE.
Table 22.3 Inflammation and its relationship with oxidative stress in age-related macular degeneration (AMD) and diabetic retinopathy

Evidence in AMD Evidence in diabetic retinopathy 523


10,11
Inflammation Presence of complement proteins in drusen Increased vitreal levels of IL-1β, IL-6, and IL-8
Associations of genetic variants in multiple complement genes increased in patients with PDR136

Chapter 22
(CFH, C2/BF, C3, CFI) with AMD114–128 Link between IL-1β and retinal capillary cell
High blood complement protein levels associated with AMD129,130 apoptosis137
Development of retinal deposits similar to AMD drusen in young Involvement of the inflammatory mediators
patients with systemic complement activation131 COX-2 and PGE2 in the development of
diabetic retinopathy via VEGF modulation138

Link between Decreased CFH mRNA expression in the presence of oxidized Increased superoxide leading to endothelial cell
inflammation and photoreceptor outer segments132 damage, increased microvascular permeability,

Mechanisms of Oxidative Stress in Retinal Injury


oxidative stress Reduced CFH expression in RPE cells in response to H2O2133 and recruitment of neutrophils38
Disruption in RPE function in presence of both H2O2 and Increased ROS leading to activation of the
complement-sufficient serum134 transcription factor NF-κB, which upregulates
Development of AMD-like lesions in mice treated with CEP cytokines, nitric oxide, and prostaglandins41
adducts135
Association of increased superoxide and decreased SOD2 with
mononuclear phagocyte-induced apoptosis of the RPE in
sod2+/– mice111
IL, interleukin; PDR, proliferative diabetic retinopathy; COX-2, cyclooxygenase-2; PGE2, prostaglandin E2; VEGF, vascular endothelial growth factor; RPE, retinal
pigment epithelium; ROS, reactive oxygen species; NF-κB, nuclear factor-κB; CEP, carboxyethylpyrrole; SOD2, superoxide dismutase 2.

Cell culture studies have linked oxidative stress to reduced Oxidative stress-related inflammation may also play a key role
expression of the complement factor H (CFH) gene in RPE cells. in the pathogenesis of diabetic retinopathy. Increased superox-
Investigation of constitutive CFH production in a stable RPE cell ide can promote inflammation by damaging endothelial cells,
line demonstrated that long-term treatment of RPE cells with increasing microvascular permeability, and recruiting neutro-
oxidized photoreceptor outer segments (POS), but not normal phils.38 ROS activate the transcription factor NF-κB, which can
POS, markedly downregulated CFH mRNA expression. Further, increase proinflammatory mediators, such as cytokines, nitric
phagocytosis of both oxidized and normal POS reduced CFH oxide, and prostaglandins. The cytokines interleukin (IL)-1β,
protein expression.132 These results suggest that the phagocytosis IL-6, and IL-8 have been reported to be increased in the vitreous
of POS, particularly if oxidized, impairs synthesis and secretion of patients with proliferative diabetic retinopathy.136 Interest-
of CFH, resulting in reduced regulatory control of complement ingly, it appears that IL-1β increases retinal capillary cell
activation and its proinflammatory effects.132 Additionally, the apoptosis.137 Additionally, IL-1 induces the production of
introduction of H2O2 to RPE cells was shown to reduce the cyclooxygenase-2, a catalyst for prostaglandin E2 formation. The
normal interferon-γ-induced stimulation of CFH expression via inflammatory mediators cyclooxygenase-2 and prostaglandin E2
acetylation of FOXO3, which interrupts STAT1 binding to the are reported to contribute to the development of diabetic reti-
CFH promoter.133 nopathy by modulating VEGF-mediated vascular permeability
The dynamic interplay between oxidative stress and inflam- and angiogenesis.138
mation may contribute to the abnormalities in RPE function
evident in AMD. A combination of H2O2 and complement- RETINAL THERAPIES TARGETING
sufficient serum was shown to disrupt the barrier function of
RPE monolayers in culture and evoke polarized secretion of
OXIDATIVE STRESS
VEGF, although introduction of H2O2 or serum alone did not In this section, we will discuss oxidative stress-related therapies
cause these changes.134 These results suggest that oxidative stress for retinal diseases. We will then explore potential avenues for
and complement together reduce RPE function. They also point the development of future treatments that protect against oxida-
to a common pathway that relates an oxidizing environment tive damage in the retina, including antioxidant therapy, AGE
and complement activation with VEGF production. Such a inhibitors, and genetic modification. These potential therapies
pathway could play a role in the neovascularization seen in are outlined in Table 22.4.
late AMD.
A close link between oxidative stress and inflammation in Supplemental antioxidants
AMD is also supported by recent animal work in which mice Currently, care of patients with dry AMD is centered on anti-
immunized with mouse serum albumin adducted with the lipid oxidant and zinc supplementation, as there are no direct inter-
peroxidation product carboxyethylpyrrole developed AMD-like ventions for drusen or geographic atrophy. AREDS, a
lesions in the retina.135 Specifically, these mice produced antibod- multicenter, randomized clinical trial sponsored by the National
ies to the hapten, fixed complement component 3 in Bruch’s Eye Institute, demonstrated that daily intake of supplemental
membrane, and accumulated drusen below the RPE.135 Taken antioxidants (β-carotene, vitamin C, vitamin E) and zinc reduced
together, these studies demonstrate a molecular link between the risk of progression to advanced AMD by 25% over
oxidative stress and complement in RPE cells and suggest that 5 years.22
the interaction between oxidative stress and inflammatory medi- Supplemental carotenoids have also been associated with
ators plays a key role in the development of AMD. decreased AMD risk. Two smaller studies linked xanthophylls
Table 22.4 Retinal therapies targeting oxidative stress treatment of geographic atrophy. In a small 10-patient study,
patients with bilateral geographic atrophy experienced less
524 Potential avenues visual loss in the eye treated with topical OT-551 compared to
for retinal therapy Specific treatments under investigation
the untreated eye, but no significant differences in geographic
Anti-inflammatory Resveratrol to decrease oxidative
atrophy area, contrast sensitivity, microperimetry measure-
ments, and total drusen area were found.142 The larger phase II
Section 2

agents damage and RPE proliferation via


inhibition of the NF-κB pathway149 OMEGA study, evaluating the ability of OT-551 to stop geo-
Curcumin to decrease expression of graphic atrophy progression and reduce the size of the geo-
inflammatory mediators146 and inhibit
the NF-κB pathway145 graphic atrophy lesions, showed no evidence of efficacy in
reaching the stated endpoint (P Sternberg, personal communica-
Boost in antioxidant Vitamin A palmitate supplementation to tion, 2010). With the lack of promising results in the phase II
defense slow rate of retinal degeneration study, no further clinical trials are planned for this agent to
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

(e.g., RP)143
Supplementation with xanthophylls and explore its potential efficacy for atrophic AMD.
omega-3 fatty acids for treatment of Studies have suggested the benefit of nutritional and drug
AMD in AREDS2 clinical trial27 supplements in preserving photoreceptor function. The most
OT-551 eye drop for geographic
widely recognized supplement for RP patients is vitamin A pal-
atrophy in AMD142
Sulforaphane to protect against retinal mitate, which has been suggested to slow the rate of retinal
oxidative damage via activation of the degeneration by an unknown mechanism.143 These same studies
Nrf2-thioredoxin system152–154 suggested that vitamin E may be associated with an increase in
Curcumin to prevent retinal oxidative
damage148 via activation of the the deterioration rate of the ERG in RP patients,143 possibly by
Nrf2-thioredoxin system148 and reducing the availability of other vitamins in the retina. More
increased antioxidant expression recently, studies have investigated the role of DHA in the treat-
(e.g., GST)147 ment of RP. It was reported that patients initiating vitamin A
Genetic Modulation of SOD2 and CAT gene palmitate supplementation may benefit from the addition of
modification expression to protect against DHA in the first 2 years of treatment.144
oxidative damage to retinal cells158,159

Inhibition of AGEs Pyridoxamine to protect against retinal Dietary antioxidants


vascular lesions in diabetics155
Aminoguanidine to prevent capillary Naturally occurring compounds found in food may protect
closure, microaneurysm formation, against retinal degenerations. Curcumin, from the spice tur-
and NOS depletion156,157 meric, has been shown to inhibit the NF-κB pathway, which is
Inhibition of Quercetin to protect against oxidative involved in immune response,145 and to decrease expression of
apoptotic pathways damage and senescence in the retina inflammatory mediators.146 Curcumin also increases expression
through inhibition of caspase-3 of antioxidants, such as heme oxygenase-1, quinone reductase,
activity150,151 and glutathione-S-transferase.147 A 2-week diet supplemented
RPE, retinal pigment epithelium; NF-κB, nuclear factor-κB; RP, retinitis with curcumin protected rats from light-induced retinal
pigmentosa; AMD, age-related macular degeneration; AREDS2, Age-Related
Eye Disease Study 2; GST, glutathione-S-transferase; SOD2, superoxide damage.148 Similarly, treatment of cultured retinal cells with cur-
dismutase 2; CAT, catalase; AGEs, advanced glycation end-products; cumin protected against H2O2-induced cell death by increasing
NOS, nitric oxide synthase.
production of antioxidant enzymes, such as thioredoxin, and
activating the Nrf2 antioxidant pathway.148
Another member of the plant-derived polyphenol family, res-
veratrol, a compound in red wine, has been shown to decrease
oxidative damage and RPE proliferation in vitro.149 Evidence
(lutein and zeaxanthin) and/or omega-3 fatty acids with suggests that resveratrol may take effect by inhibiting the NF-κB
improved visual acuity139 and central retinal function (as mea- pathway involved in inflammatory response.
sured by multifocal ERG).140 In the Rotterdam Study, high The flavonoid quercetin, found in green tea and red onion, is
dietary intake of zinc, omega-3 fatty acids, β-carotene, and a free radical scavenger and a chelating agent that can reduce
lutein/zeaxanthin corresponded to decreased risk of early AMD iron-driven lipid peroxidation.150 Quercetin was recently
onset.141 The AREDS2 study is now investigating the effect of reported to protect against H2O2-induced oxidative damage and
oral supplementation of xanthophylls and omega-3 fatty acids senescence in cultured RPE cells by inhibiting caspase-3
(docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)) activity.151
on the risk of AMD progression. In this ongoing clinical trial, Sulforaphane, an isothiocyanate found in vegetables such as
scheduled to end in 2013, AMD patients are randomly assigned broccoli, appears to exert neuroprotective effects through activa-
to four treatment groups: (1) xanthophylls only (10 mg lutein tion of the Nrf2-thioredoxin system. The antioxidant transcrip-
and 2 mg zeaxanthin); (2) omega-3 fatty acids only (350 mg tion factor Nrf2 binds to the antioxidant response element (ARE),
DHA and 650 mg EPA); (3) xanthophylls and omega-3 fatty activating the antioxidant defense system in response to oxida-
acids; and (4) placebo.27 The results of this trial may alter the tive stress.152 Thioredoxin has been shown to protect against
recommended regimen for AMD patients currently treated with H2O2-induced damage in photoreceptor cells and light damage
antioxidant and zinc supplements. in mice.153 Sulforaphane induces thioredoxin in light-exposed
The topical application of a disubstituted hydroxylamine with RPE cells via the Nrf2-ARE pathway,154 thus protecting against
antioxidant properties, called OT-551, has been studied for the photo-oxidative damage to the retina.
Fig. 22.1 Oxidative stress, brought on by
factors such as aging, smoking, light
OXIDATIVE STRESS exposure, and hyperglycemia, leads to an
Aging Smoking oxidized cellular environment that is marked 525
by the accumulation of reactive oxygen
Light Hyperglycemia species (ROS) and advanced glycation
end-products (AGEs). This oxidized

Chapter 22
environment triggers stress responses, such
Vitamin A as damage to mitochondrial (mt) DNA,
AREDS OXIDIZED ENVIRONMENT vascular changes, inflammatory modulation,
AREDS2 ROS and apoptosis. Concurrently, the cellular
AGEs Pyridoxamine antioxidant defense system, comprised of
Aminoguanidine both antioxidants and antioxidant enzymes, is
activated, mitigating the damage of oxidative
ANTIOXIDANT DEFENSE

ANTIOXIDANTS
stress. The mechanisms of current and

Mechanisms of Oxidative Stress in Retinal Injury


Vitamins C + E Thiols potential treatments for retinal diseases are
Carotenes Zinc depicted above, with green representing
Xanthophylls activation and red representing inhibition.
Sulforaphane AREDS, Age-Related Eye Disease Study;
CAT, catalase; SOD, superoxide dismutase;
ANTIOXIDANT ENZYMES
GPx, glutathione peroxidase; GRx,
CAT GPx PON1 glutathione reductase; PON1, paraoxonase 1.
SOD GRx

STRESS RESPONSE
Gene Therapy mtDNA Vascular Inflammatory
Curcumin Apoptosis
Damage Changes Modulation

Bevacizumab Quercetin
Ranibizumab

Anti-advanced glycation of RPE cells, retinal vascular endothelial cells, and photorecep-
tors. Regardless of a condition’s primary insult, oxidative stress
end-product treatment
often contributes to retinal cell death via DNA damage, modula-
One possible avenue for treatment of diabetic retinopathy targets tion of alternative metabolic pathways, and stimulation of pro-
the accumulation of AGEs, which can disturb retinal vascular apoptotic reactions. Because oxidative stress appears to be a
cell function. The AGE inhibitor pyridoxamine protected against common mechanism of retinal cell damage, the development of
diabetes-induced retinal vascular lesions, thickening of the base- therapy that protects against retinal oxidative damage has the
ment membrane, and acellular capillaries in diabetic rats.155 potential to reduce vision loss from retinal diseases significantly
Aminoguanidine, which acts as an inhibitor of inducible nitric (Fig. 22.1).
oxide synthase and AGE crosslinking, may prevent capillary
closure, microaneurysm formation, and nitric oxide synthase REFERENCES
1. Beatty S, Koh H, Phil M, et al. The role of oxidative stress in the pathogenesis
depletion.156,157 The efficacy of pyridoxamine and aminoguani- of age-related macular degeneration. Surv Ophthalmol 2000;45:115–34.
dine has yet to be investigated for the treatment of diabetic reti- 2. Cai J, Nelson KC, Wu M, et al. Oxidative damage and protection of the RPE.
Prog Retin Eye Res 2000;19:205–21.
nopathy in clinical trials. 3. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian
organs. Physiol Rev 1979;59:527–605.
Genetic modification 4. Borish ET, Pryor WA, Venugopal S, et al. DNA synthesis is blocked by
cigarette tar-induced DNA single-strand breaks. Carcinogenesis 1987;8:
Gene therapy is being explored for the treatment of retinal 1517–20.
degeneration. Towards this end, genes involved in the regula- 5. Jones DP, Mody Jr VC, Carlson JL, et al. Redox analysis of human plasma
allows separation of pro-oxidant events of aging from decline in antioxidant
tion of cellular oxidative stress are being targeted in animal defenses. Free Radic Biol Med 2002;33:1290–300.
studies to reduce ROS production, bolster antioxidant defenses, 6. Jones DP. Radical-free biology of oxidative stress. Am J Physiol Cell Physiol
2008;295:C849–68.
and/or increase cellular repair capacity. In mice with ischemia- 7. Friedman DS, O’Colmain BJ, Munoz B, et al. Prevalence of age-related macular
induced retinal injury, plasmids encoding the human SOD2 (Mn degeneration in the United States. Arch Ophthalmol 2004;122:564–72.
SOD) or CAT (catalase) genes significantly decreased ROS gen- 8. Sarks J, Arnold J, Ho IV, et al. Evolution of reticular pseudodrusen. Br J Oph-
thalmol 2011; 95:979–85.
eration and retinal endothelial cell death.158 Another study 9. Zweifel SA, Spaide RF, Curcio CA, et al. Reticular pseudodrusen are subreti-
showed that an adenovirus carrying the CAT gene protected nal drusenoid deposits. Ophthalmology 2010;117:303–12 e1.
10. Mullins RF, Russell SR, Anderson DH, et al. Drusen associated with aging
against H2O2-induced damage to RPE cells and light-induced and age-related macular degeneration contain proteins common to extracel-
damage to RPE and photoreceptors in mice.159 lular deposits associated with atherosclerosis, elastosis, amyloidosis, and
dense deposit disease. FASEB J 2000;14:835–46.
CONCLUSIONS 11. Wang L, Clark ME, Crossman DK, et al. Abundant lipid and protein compo-
nents of drusen. PLoS ONE 2010;5:e10329.
12. Seddon JM, Cote J, Davis N, et al. Progression of age-related macular degenera-
Oxidative stress plays a significant role in the pathogenesis of tion: association with body mass index, waist circumference, and waist–hip
multiple retinal diseases, including AMD, diabetic retinopathy, ratio. Arch Ophthalmol 2003;121:785–92.
13. Clemons TE, Milton RC, Klein R, et al. Risk factors for the incidence of
and inherited retinal degenerations. ROS, derived primarily advanced age-related macular degeneration in the Age-Related Eye Disease
from the mitochondria, can adversely affect health and survival Study (AREDS). AREDS report no. 19. Ophthalmology 2005;112:533–9.
14. Tan JS, Mitchell P, Kifley A, et al. Smoking and the long-term incidence 45. Leveillard T, Mohand-Said S, Lorentz O, et al. Identification and characteriza-
of age-related macular degeneration: the Blue Mountains Eye Study. tion of rod-derived cone viability factor. Nat Genet 2004;36:755–9.
Arch Ophthalmol 2007;125:1089–95. 46. Carmody RJ, McGowan AJ, Cotter TG. Reactive oxygen species as mediators
526 15. Cackett P, Wong TY, Aung T, et al. Smoking, cardiovascular risk factors, and of photoreceptor apoptosis in vitro. Exp Cell Res 1999;248:520–30.
age-related macular degeneration in Asians: the Singapore Malay Eye Study. 47. Miceli MV, Liles MR, Newsome DA. Evaluation of oxidative processes in
Am J Ophthalmol 2008;146:960–7 e1. human pigment epithelial cells associated with retinal outer segment phago-
16. Chakravarthy U, Augood C, Bentham GC, et al. Cigarette smoking and age- cytosis. Exp Cell Res 1994;214:242–9.
related macular degeneration in the EUREYE Study. Ophthalmology 2007; 48. Yang P, Peairs JJ, Tano R, et al. Oxidant-mediated Akt activation in human
Section 2

114:1157–63. RPE cells. Invest Ophthalmol Vis Sci 2006;47:4598–606.


17. Fraser-Bell S, Wu J, Klein R, et al. Smoking, alcohol intake, estrogen use, and 49. Yu AL, Fuchshofer R, Kook D, et al. Subtoxic oxidative stress induces senes-
age-related macular degeneration in Latinos: the Los Angeles Latino Eye cence in retinal pigment epithelial cells via TGF-beta release. Invest Ophthal-
Study. Am J Ophthalmol 2006;141:79–87. mol Vis Sci 2009;50:926–35.
18. Klein R, Knudtson MD, Cruickshanks KJ, et al. Further observations on 50. Glotin AL, Debacq-Chainiaux F, Brossas JY, et al. Prematurely senescent
the association between smoking and the long-term incidence and progres- ARPE-19 cells display features of age-related macular degeneration.
sion of age-related macular degeneration: the Beaver Dam Eye Study. Free Radic Biol Med 2008 ;44:1348–61.
Arch Ophthalmol 2008;126:115–21. 51. Fuchshofer R, Yu AL, Teng HH, et al. Hypoxia/reoxygenation induces CTGF
Basic Mechanisms of Injury in the Retina

19. Tomany SC, Wang JJ, Van Leeuwen R, et al. Risk factors for incident and PAI-1 in cultured human retinal pigment epithelium cells. Exp Eye Res
Basic Science and Translation to Therapy

age-related macular degeneration: pooled findings from 3 continents. 2009;88:889–99.


Ophthalmology 2004;111:1280–7. 52. Winkler BS, Boulton ME, Gottsch JD, et al. Oxidative damage and age-related
20. Seddon JM, Ajani UA, Sperduto RD, et al. Dietary carotenoids, vitamins A, macular degeneration. Mol Vis 1999;5:32.
C, and E, and advanced age-related macular degeneration. Eye Disease 53. Roehlecke C, Schaller A, Knels L, et al. The influence of sublethal blue light
Case-Control Study Group. JAMA 1994;272:1413–20. exposure on human RPE cells. Mol Vis 2009;15:1929–38.
21. van Leeuwen R, Boekhoorn S, Vingerling JR, et al. Dietary intake of antioxi- 54. Howes KA, Liu Y, Dunaief JL, et al. Receptor for advanced glycation end
dants and risk of age-related macular degeneration. JAMA 2005;294:3101–7. products and age-related macular degeneration. Invest Ophthalmol Vis Sci
22. A randomized, placebo-controlled, clinical trial of high-dose supplementation 2004;45:3713–20.
with vitamins C and E, beta carotene, and zinc for age-related macular degen- 55. Ma W, Lee SE, Guo J, et al. RAGE ligand upregulation of VEGF secretion in
eration and vision loss: AREDS report no. 8. Arch Ophthalmol 2001;119: ARPE-19 cells. Invest Ophthalmol Vis Sci 2007;48:1355–61.
1417–36. 56. Sreekumar PG, Kannan R, de Silva AT, et al. Thiol regulation of vascular
23. Crabb JW, Miyagi M, Gu X, et al. Drusen proteome analysis: an approach to endothelial growth factor-A and its receptors in human retinal pigment
the etiology of age-related macular degeneration. Proc Natl Acad Sci U S A epithelial cells. Biochem Biophys Res Commun 2006;346:1200–6.
2002;99:14682–7. 57. Kannan R, Zhang N, Sreekumar PG, et al. Stimulation of apical and basolat-
24. Bone RA, Landrum JT, Mayne ST, et al. Macular pigment in donor eyes with eral VEGF-A and VEGF-C secretion by oxidative stress in polarized retinal
and without AMD: a case-control study. Invest Ophthalmol Vis Sci 2001;42: pigment epithelial cells. Mol Vis 2006;12:1649–59.
235–40. 58. Klettner A, Roider J. Constitutive and oxidative-stress-induced expression of
25. Bernstein PS, Zhao DY, Wintch SW, et al. Resonance Raman measurement of VEGF in the RPE are differently regulated by different Mitogen-activated
macular carotenoids in normal subjects and in age-related macular degenera- protein kinases. Graefes Arch Clin Exp Ophthalmol 2009;247:1487–92.
tion patients. Ophthalmology 2002;109:1780–7. 59. Byeon SH, Lee SC, Choi SH, et al. Vascular endothelial growth factor as an
26. Obana A, Hiramitsu T, Gohto Y, et al. Macular carotenoid levels of normal autocrine survival factor for retinal pigment epithelial cells under oxidative
subjects and age-related maculopathy patients in a Japanese population. stress via the VEGF-R2/PI3K/Akt. Invest Ophthalmol Vis Sci 2010;51:1190–7.
Ophthalmology 2008;115:147–57. 60. Kuroki M, Voest EE, Amano S, et al. Reactive oxygen intermediates increase
27. Krishnadev N, Meleth AD, Chew EY. Nutritional supplements for age-related vascular endothelial growth factor expression in vitro and in vivo. J Clin
macular degeneration. Curr Opin Ophthalmol 2010;21:184–9. Invest 1996;98:1667–75.
28. Cheung N, Mitchell P, Wong TY. Diabetic retinopathy. Lancet 2010;376: 61. Platt DH, Bartoli M, El-Remessy AB, et al. Peroxynitrite increases VEGF
124–36. expression in vascular endothelial cells via STAT3. Free Radic Biol Med
29. Saaddine JB, Honeycutt AA, Narayan KM, et al. Projection of diabetic reti- 2005;39:1353–61.
nopathy and other major eye diseases among people with diabetes mellitus: 62. Abou-Mohamed G, Johnson JA, Jin L, et al. Roles of superoxide, peroxynitrite,
United States, 2005–2050. Arch Ophthalmol 2008;126:1740–7. and protein kinase C in the development of tolerance to nitroglycerin.
30. Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal microvascular J Pharmacol Exp Ther 2004;308:289–99.
cells in human and experimental diabetic retinopathy. J Clin Invest 1996;97: 63. Gerber HP, McMurtrey A, Kowalski J, et al. Vascular endothelial growth
2883–90. factor regulates endothelial cell survival through the phosphatidylinositol
31. Kowluru RA, Koppolu P, Chakrabarti S, et al. Diabetes-induced activation of 3’-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR
nuclear transcriptional factor in the retina, and its inhibition by antioxidants. activation. J Biol Chem 1998;273:30336–43.
Free Radic Res 2003;37:1169–80. 64. Duh E, Aiello LP. Vascular endothelial growth factor and diabetes: the agonist
32. Kowluru RA, Koppolu P. Diabetes-induced activation of caspase-3 in retina: versus antagonist paradox. Diabetes 1999;48:1899–906.
effect of antioxidant therapy. Free Radic Res 2002;36:993–9. 65. Fernandes R, Hosoya K, Pereira P. Reactive oxygen species downregulate
33. Kanwar M, Chan PS, Kern TS, et al. Oxidative damage in the retinal mito- glucose transport system in retinal endothelial cells. Am J Physiol Cell Physiol
chondria of diabetic mice: possible protection by superoxide dismutase. 2011;300:C927–36.
Invest Ophthalmol Vis Sci 2007;48:3805–11. 66. Eichler W, Reiche A, Yafai Y, et al. Growth-related effects of oxidant-induced
34. Kowluru RA, Atasi L, Ho YS. Role of mitochondrial superoxide dismutase in stress on cultured RPE and choroidal endothelial cells. Exp Eye Res 2008;87:
the development of diabetic retinopathy. Invest Ophthalmol Vis Sci 2006;47: 342–8.
1594–9. 67. Hoffmann S, Friedrichs U, Eichler W, et al. Advanced glycation end products
35. Rains JL, Jain SK. Oxidative stress, insulin signaling, and diabetes. Free Radic induce choroidal endothelial cell proliferation, matrix metalloproteinase-2
Biol Med 2011;50:567–75. and VEGF upregulation in vitro. Graefes Arch Clin Exp Ophthalmol
36. Brownlee M. The pathobiology of diabetic complications: a unifying mecha- 2002;240:996–1002.
nism. Diabetes 2005;54:1615–25. 68. Yu DY, Cringle SJ, Su EN, et al. Intraretinal oxygen levels before and after
37. Goldin A, Beckman JA, Schmidt AM, et al. Advanced glycation end products: photoreceptor loss in the RCS rat. Invest Ophthalmol Vis Sci 2000;41:
sparking the development of diabetic vascular injury. Circulation 2006;114: 3999–4006.
597–605. 69. Yu DY, Cringle S, Valter K, et al. Photoreceptor death, trophic factor
38. Kowluru RA, Chan PS. Oxidative stress and diabetic retinopathy. Exp expression, retinal oxygen status, and photoreceptor function in the P23H rat.
Diabetes Res 2007;43603. Invest Ophthalmol Vis Sci 2004;45:2013–9.
39. Shiba T, Inoguchi T, Sportsman JR, et al. Correlation of diacylglycerol level 70. Padnick-Silver L, Kang Derwent JJ, Giuliano E, et al. Retinal oxygenation and
and protein kinase C activity in rat retina to retinal circulation. Am J Physiol oxygen metabolism in Abyssinian cats with a hereditary retinal degeneration.
1993;265:E783–93. Invest Ophthalmol Vis Sci 2006;47:3683–9.
40. Williams B, Gallacher B, Patel H, et al. Glucose-induced protein kinase C 71. Komeima K, Rogers BS, Lu L, et al. Antioxidants reduce cone cell death in a
activation regulates vascular permeability factor mRNA expression and model of retinitis pigmentosa. Proc Natl Acad Sci U S A 2006;103:11300–5.
peptide production by human vascular smooth muscle cells in vitro. Diabetes 72. Chang GQ, Hao Y, Wong F. Apoptosis: final common pathway of photorecep-
1997;46:1497–503. tor death in rd, rds, and rhodopsin mutant mice. Neuron 1993;11:595–605.
41. Brownlee M. Biochemistry and molecular cell biology of diabetic complica- 73. Tso MO, Zhang C, Abler AS, et al. Apoptosis leads to photoreceptor degenera-
tions. Nature 2001;414:813–20. tion in inherited retinal dystrophy of RCS rats. Invest Ophthalmol Vis Sci
42. Bramall AN, Wright AF, Jacobson SG, et al. The genomic, biochemical, and 1994;35:2693–9.
cellular responses of the retina in inherited photoreceptor degenerations and 74. Carmody RJ, Cotter TG. Oxidative stress induces caspase-independent retinal
prospects for the treatment of these disorders. Annu Rev Neurosci 2010;33: apoptosis in vitro. Cell Death Differ 2000;7:282–91.
441–72. 75. Donovan M, Cotter TG. Caspase-independent photoreceptor apoptosis in
43. Ayuso C, Millan JM. Retinitis pigmentosa and allied conditions today: a para- vivo and differential expression of apoptotic protease activating factor-1 and
digm of translational research. Genome Med 2010;2:34. caspase-3 during retinal development. Cell Death Differ 2002;9:1220–31.
44. Allikmets R, Shroyer NF, Singh N, et al. Mutation of the Stargardt disease 76. Doonan F, Donovan M, Cotter TG. Caspase-independent photoreceptor apop-
gene (ABCR) in age-related macular degeneration. Science 1997;277:1805–7. tosis in mouse models of retinal degeneration. J Neurosci 2003;23:5723–31.
77. Kunchithapautham K, Rohrer B. Autophagy is one of the multiple mecha- 108. Schmidt S, Hauser MA, Scott WK, et al. Cigarette smoking strongly modifies
nisms active in photoreceptor degeneration. Autophagy 2007;3:65–6. the association of LOC387715 and age-related macular degeneration. Am J
78. Sanvicens N, Gomez-Vicente V, Masip I, et al. Oxidative stress-induced apop- Hum Genet 2006;78:852–64.
tosis in retinal photoreceptor cells is mediated by calpains and caspases and 109. Schaumberg DA, Hankinson SE, Guo Q, et al. A prospective study of 2 major 527
blocked by the oxygen radical scavenger CR-6. J Biol Chem 2004;279: age-related macular degeneration susceptibility alleles and interactions with
39268–78. modifiable risk factors. Arch Ophthalmol 2007;125:55–62.
79. Doonan F, Donovan M, Cotter TG. Activation of multiple pathways during 110. Wang G, Spencer KL, Court BL, et al. Localization of age-related macular

Chapter 22
photoreceptor apoptosis in the rd mouse. Invest Ophthalmol Vis Sci degeneration-associated ARMS2 in cytosol, not mitochondria. Invest Ophthal-
2005;46:3530–8. mol Vis Sci 2009;50:3084–90.
80. Ahuja P, Caffe AR, Ahuja S, et al. Decreased glutathione transferase levels in 111. Yang D, Elner SG, Lin LR, et al. Association of superoxide anions with retinal
rd1/rd1 mouse retina: replenishment protects photoreceptors in retinal pigment epithelial cell apoptosis induced by mononuclear phagocytes.
explants. Neuroscience 2005;131:935–43. Invest Ophthalmol Vis Sci 2009;50:4998–5005.
81. Ahuja-Jensen P, Johnsen-Soriano S, Ahuja S, et al. Low glutathione peroxidase 112. Zhang B, Hu Y, Ma JX. Anti-inflammatory and antioxidant effects of SERPI-
in rd1 mouse retina increases oxidative stress and proteases. Neuroreport NA3K in the retina. Invest Ophthalmol Vis Sci 2009;50:3943–52.
2007;18:797–801. 113. Patel M, Chan CC. Immunopathological aspects of age-related macular
82. Hill JW, Hu JJ, Evans MK. OGG1 is degraded by calpain following oxidative degeneration. Semin Immunopathol 2008;30:97–110.
stress and cisplatin exposure. DNA Repair (Amst) 2008;7:648–54. 114. Edwards AO, Ritter 3rd R, Abel KJ, et al. Complement factor H polymorphism

Mechanisms of Oxidative Stress in Retinal Injury


83. Shen J, Yang X, Dong A, et al. Oxidative damage is a potential cause of cone and age-related macular degeneration. Science 2005;308:421–4.
cell death in retinitis pigmentosa. J Cell Physiol 2005;203:457–64. 115. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant
84. Komeima K, Rogers BS, Campochiaro PA. Antioxidants slow photoreceptor increases the risk of age-related macular degeneration. Science 2005;308:
cell death in mouse models of retinitis pigmentosa. J Cell Physiol 2007;213: 419–21.
809–15. 116. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in
85. Usui S, Oveson BC, Lee SY, et al. NADPH oxidase plays a central role in cone age-related macular degeneration. Science 2005;308:385–9.
cell death in retinitis pigmentosa. J Neurochem 2009;110:1028–37. 117. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the
86. Leveillard T, Sahel JA. Rod-derived cone viability factor for treating blinding complement regulatory gene factor H (HF1/CFH) predisposes individuals to
diseases: from clinic to redox signaling. Sci Transl Med 2010;2:26ps16. age-related macular degeneration. Proc Natl Acad Sci U S A 2005;102:
87. Cronin T, Raffelsberger W, Lee-Rivera I, et al. The disruption of the rod- 7227–32.
derived cone viability gene leads to photoreceptor dysfunction and suscepti- 118. Zareparsi S, Branham KE, Li M, et al. Strong association of the Y402H variant
bility to oxidative stress. Cell Death Differ 2010;17:1199–210. in complement factor H at 1q32 with susceptibility to age-related macular
88. Jarrett SG, Lewin AS, Boulton ME. The importance of mitochondria in age- degeneration. Am J Hum Genet 2005;77:149–53.
related and inherited eye disorders. Ophthalmic Res 2010;44:179–90. 119. Souied EH, Leveziel N, Richard F, et al. Y402H complement factor H poly-
89. Wallace DC. Mitochondrial diseases in man and mouse. Science 1999;283: morphism associated with exudative age-related macular degeneration in the
1482–8. French population. Mol Vis 2005;11:1135–40.
90. Jin GF, Hurst JS, Godley BF. Rod outer segments mediate mitochondrial DNA 120. Simonelli F, Frisso G, Testa F, et al. Polymorphism p.402Y>H in the comple-
damage and apoptosis in human retinal pigment epithelium. Curr Eye Res ment factor H protein is a risk factor for age related macular degeneration in
2001;23:11–9. an Italian population. Br J Ophthalmol 2006;90:1142–5.
91. Feher J, Kovacs I, Artico M, et al. Mitochondrial alterations of retinal pigment 121. Despriet DD, Klaver CC, Witteman JC, et al. Complement factor H polymor-
epithelium in age-related macular degeneration. Neurobiol Aging 2006;27: phism, complement activators, and risk of age-related macular degeneration.
983–93. JAMA 2006 ;296:301–9.
92. Nordgaard CL, Berg KM, Kapphahn RJ, et al. Proteomics of the retinal 122. Seitsonen S, Lemmela S, Holopainen J, et al. Analysis of variants in the com-
pigment epithelium reveals altered protein expression at progressive stages plement factor H, the elongation of very long chain fatty acids-like 4 and
of age-related macular degeneration. Invest Ophthalmol Vis Sci 2006;47: the hemicentin 1 genes of age-related macular degeneration in the Finnish
815–22. population. Mol Vis 2006;12:796–801.
93. Nordgaard CL, Karunadharma PP, Feng X, et al. Mitochondrial proteomics 123. Gold B, Merriam JE, Zernant J, et al. Variation in factor B (BF) and complement
of the retinal pigment epithelium at progressive stages of age-related macular component 2 (C2) genes is associated with age-related macular degeneration.
degeneration. Invest Ophthalmol Vis Sci 2008;49:2848–55. Nat Genet 2006;38:458–62.
94. Karunadharma PP, Nordgaard CL, Olsen TW, et al. Mitochondrial DNA 124. Yates JR, Sepp T, Matharu BK, et al. Complement C3 variant and the risk of
damage as a potential mechanism for age-related macular degeneration. age-related macular degeneration. N Engl J Med 2007;357:553–61.
Invest Ophthalmol Vis Sci 2010;51:5470–9. 125. Fagerness JA, Maller JB, Neale BM, et al. Variation near complement factor I
95. Jiang S, Moriarty-Craige SE, Orr M, et al. Oxidant-induced apoptosis in is associated with risk of advanced AMD. Eur J Hum Genet 2009;17:100–4.
human retinal pigment epithelial cells: dependence on extracellular redox 126. Ennis S, Gibson J, Cree AJ, et al. Support for the involvement of complement
state. Invest Ophthalmol Vis Sci 2005;46:1054–61. factor I in age-related macular degeneration. Eur J Hum Genet 2010;18:
96. Justilien V, Pang JJ, Renganathan K, et al. SOD2 knockdown mouse model of 15–6.
early AMD. Invest Ophthalmol Vis Sci 2007;48:4407–20. 127. Hageman GS, Hancox LS, Taiber AJ, et al. Extended haplotypes in the comple-
97. Kowluru RA, Abbas SN. Diabetes-induced mitochondrial dysfunction in the ment factor H (CFH) and CFH-related (CFHR) family of genes protect against
retina. Invest Ophthalmol Vis Sci 2003;44:5327–34. age-related macular degeneration: characterization, ethnic distribution and
98. Kowluru RA, Kowluru V, Xiong Y, et al. Overexpression of mitochondrial evolutionary implications. Ann Med 2006;38:592–604.
superoxide dismutase in mice protects the retina from diabetes-induced 128. Hughes AE, Orr N, Esfandiary H, et al. A common CFH haplotype, with
oxidative stress. Free Radic Biol Med 2006;41:1191–6. deletion of CFHR1 and CFHR3, is associated with lower risk of age-related
99. Jones MM, Manwaring N, Wang JJ, et al. Mitochondrial DNA haplogroups macular degeneration. Nat Genet 2006;38:1173–7.
and age-related maculopathy. Arch Ophthalmol 2007;125:1235–40. 129. Scholl HP, Charbel Issa P, Walier M, et al. Systemic complement activation in
100. SanGiovanni JP, Arking DE, Iyengar SK, et al. Mitochondrial DNA variants age-related macular degeneration. PLoS ONE 2008;3:e2593.
of respiratory complex I that uniquely characterize haplogroup T2 are associ- 130. Reynolds R, Hartnett ME, Atkinson JP, et al. Plasma complement components
ated with increased risk of age-related macular degeneration. PLoS ONE and activation fragments: associations with age-related macular degeneration
2009;4:e5508. genotypes and phenotypes. Invest Ophthalmol Vis Sci 2009;50:5818–27.
101. Canter JA, Olson LM, Spencer K, et al. Mitochondrial DNA polymorphism 131. Huang SJ, Costa DL, Gross NE, et al. Peripheral drusen in membranoprolifera-
A4917G is independently associated with age-related macular degeneration. tive glomerulonephritis type II. Retina 2003;23:429–31.
PLoS ONE 2008;3:e2091. 132. Chen M, Forrester JV, Xu H. Synthesis of complement factor H by retinal
102. Udar N, Atilano SR, Memarzadeh M, et al. Mitochondrial DNA haplogroups pigment epithelial cells is down-regulated by oxidized photoreceptor outer
associated with age-related macular degeneration. Invest Ophthalmol Vis Sci segments. Exp Eye Res 2007;84:635–45.
2009;50:2966–74. 133. Wu Z, Lauer TW, Sick A, et al. Oxidative stress modulates complement factor
103. Kofler B, Mueller EE, Eder W, et al. Mitochondrial DNA haplogroup T is H expression in retinal pigmented epithelial cells by acetylation of FOXO3.
associated with coronary artery disease and diabetic retinopathy: a case J Biol Chem 2007;282:22414–25.
control study. BMC Med Genet 2009;10:35. 134. Thurman JM, Renner B, Kunchithapautham K, et al. Oxidative stress renders
104. Rivera A, Fisher SA, Fritsche LG, et al. Hypothetical LOC387715 is a second retinal pigment epithelial cells susceptible to complement-mediated injury.
major susceptibility gene for age-related macular degeneration, contributing J Biol Chem 2009;284:16939–47.
independently of complement factor H to disease risk. Hum Mol Genet 135. Hollyfield JG, Bonilha VL, Rayborn ME, et al. Oxidative damage-induced
2005;14:3227–36. inflammation initiates age-related macular degeneration. Nat Med 2008;14:
105. Jakobsdottir J, Conley YP, Weeks DE, et al. Susceptibility genes for age- 194–8.
related maculopathy on chromosome 10q26. Am J Hum Genet 2005;77: 136. Yuuki T, Kanda T, Kimura Y, et al. Inflammatory cytokines in vitreous fluid
389–407. and serum of patients with diabetic vitreoretinopathy. J Diabetes Complica-
106. Kanda A, Chen W, Othman M, et al. A variant of mitochondrial protein tions 2001;15:257–9.
LOC387715/ARMS2, not HTRA1, is strongly associated with age-related 137. Kowluru RA, Odenbach S. Role of interleukin-1beta in the pathogenesis of
macular degeneration. Proc Natl Acad Sci U S A 2007;104:16227–32. diabetic retinopathy. Br J Ophthalmol 2004;88:1343–7.
107. Fritsche LG, Loenhardt T, Janssen A, et al. Age-related macular degeneration 138. Wilkinson-Berka JL. Vasoactive factors and diabetic retinopathy: vascular
is associated with an unstable ARMS2 (LOC387715) mRNA. Nat Genet endothelial growth factor, cycoloxygenase-2 and nitric oxide. Curr Pharm Des
2008;40:892–6. 2004;10:3331–48.
139. Cangemi FE. TOZAL Study: an open case control study of an oral antioxidant 150. Murota K, Mitsukuni Y, Ichikawa M, et al. Quercetin-4’-glucoside is more
and omega-3 supplement for dry AMD. BMC Ophthalmol 2007;7:3. potent than quercetin-3-glucoside in protection of rat intestinal mucosa
140. Parisi V, Tedeschi M, Gallinaro G, et al. Carotenoids and antioxidants in age- homogenates against iron ion-induced lipid peroxidation. J Agric Food Chem
528 related maculopathy Italian study: multifocal electroretinogram modifications 2004;52:1907–12.
after 1 year. Ophthalmology 2008;115:324–33 e2. 151. Kook D, Wolf AH, Yu AL, et al. The protective effect of quercetin against
141. Ho L, van Leeuwen R, Witteman JC, et al. Reducing the genetic risk of age- oxidative stress in the human RPE in vitro. Invest Ophthalmol Vis Sci 2008;
related macular degeneration with dietary antioxidants, zinc, and ω-3 fatty 49:1712–20.
acids: the Rotterdam study. Arch Ophthalmol 2011;129:758–66. 152. Nguyen T, Sherratt PJ, Pickett CB. Regulatory mechanisms controlling
Section 2

142. Wong WT, Kam W, Cunningham D, et al. Treatment of geographic atrophy gene expression mediated by the antioxidant response element. Annu Rev
by the topical administration of OT-551: results of a phase II clinical trial. Pharmacol Toxicol 2003;43:233–60.
Invest Ophthalmol Vis Sci 2010;51:6131–9. 153. Tanito M, Agbaga MP, Anderson RE. Upregulation of thioredoxin
143. Berson EL, Rosner B, Sandberg MA, et al. A randomized trial of vitamin A system via Nrf2-antioxidant responsive element pathway in adaptive-retinal
and vitamin E supplementation for retinitis pigmentosa. Arch Ophthalmol neuroprotection in vivo and in vitro. Free Radic Biol Med 2007;42:
1993;111:761–72. 1838–50.
144. Berson EL, Rosner B, Sandberg MA, et al. Further evaluation of docosahexae- 154. Tanito M, Masutani H, Kim YC, et al. Sulforaphane induces thioredoxin
noic acid in patients with retinitis pigmentosa receiving vitamin A treatment: through the antioxidant-responsive element and attenuates retinal light
Basic Mechanisms of Injury in the Retina

subgroup analyses. Arch Ophthalmol 2004;122:1306–14. damage in mice. Invest Ophthalmol Vis Sci 2005;46:979–87.
Basic Science and Translation to Therapy

145. Singh S, Aggarwal BB. Activation of transcription factor NF-kappa B is sup- 155. Stitt A, Gardiner TA, Alderson NL, et al. The AGE inhibitor pyridoxamine
pressed by curcumin (diferuloylmethane) [corrected]. J Biol Chem 1995;270: inhibits development of retinopathy in experimental diabetes. Diabetes 2002;
24995–5000. 51:2826–32.
146. Abe Y, Hashimoto S, Horie T. Curcumin inhibition of inflammatory cytokine 156. Edelstein D, Brownlee M. Mechanistic studies of advanced glycosylation end
production by human peripheral blood monocytes and alveolar macrophages. product inhibition by aminoguanidine. Diabetes 1992;41:26–9.
Pharmacol Res 1999;39:41–7. 157. Friedman EA. Advanced glycosylated end products and hyperglycemia in
147. Scapagnini G, Colombrita C, Amadio M, et al. Curcumin activates defensive the pathogenesis of diabetic complications. Diabetes Care 1999;22(Suppl
genes and protects neurons against oxidative stress. Antioxid Redox Signal 2):B65–71.
2006;8:395–403. 158. Chen B, Caballero S, Seo S, et al. Delivery of antioxidant enzyme genes to
148. Mandal MN, Patlolla JM, Zheng L, et al. Curcumin protects retinal cells from protect against ischemia/reperfusion-induced injury to retinal microvascula-
light-and oxidant stress-induced cell death. Free Radic Biol Med 2009;46: ture. Invest Ophthalmol Vis Sci 2009;50:5587–95.
672–9. 159. Rex TS, Tsui I, Hahn P, et al. Adenovirus-mediated delivery of catalase to
149. King RE, Kent KD, Bomser JA. Resveratrol reduces oxidation and prolifera- retinal pigment epithelial cells protects neighboring photoreceptors from
tion of human retinal pigment epithelial cells via extracellular signal-regulated photo-oxidative stress. Hum Gene Ther 2004;15:960–7.
kinase inhibition. Chem Biol Interact 2005;151:143–9.
Basic Mechanisms of Injury in the Retina Section 2

Mechanisms of ER Stress in Retinal Disease Chapter

Sylvia B. Smith
23 
Basic science and clinical investigations aimed at understanding physiological state.2–4 The possible fates for a protein in the ER
mechanisms of retinal disease frequently focus on retinal tissue, are shown in Figure 23.1 and include: (1) proper folding and exit
specific retinal cell types (such as photoreceptor cells, ganglion through the ER; or in the case of misfolded proteins: (2) transla-
cells, Müller cells) and genes/proteins specific to the retina. An tional attenuation whereby protein synthesis halts temporarily
emerging approach to understanding retinal disease examines to prevent accumulation of unfolded proteins; (3) transcriptional
pathogenesis at the level of an intracellular organelle, the endo- induction of ER chaperone genes to increase protein folding
plasmic reticulum (ER). In this chapter, a brief overview of the capacity; (4) transcriptional induction of ER-associated degrada-
field of ER stress and the unfolded protein response is provided tion (ERAD) activity. If these strategies fail and protein aggrega-
followed by information linking these processes to retinal tion is excessive, apoptosis (5) is induced to dispose of cells
disease. injured by ER stress, thereby ensuring survival of the organism.
Recent studies implicate dysregulation of the UPR in neurode-
THE ENDOPLASMIC RETICULUM generative diseases, including retinal diseases.
As its name suggests, the ER is a membrane-bound labyrinth of ER STRESS AND UPR SIGNALING
tubes and sacs, where virtually all plasma membrane and
secreted proteins begin their progression to the cellular surface. The UPR is a complex cellular signaling pathway that attempts
The ER is the largest membrane compartment within eukaryotic to restore cellular homeostasis, but, in the face of prolonged ER
cells, frequently comprising more than half of the total mem- stress, pathways may be activated that lead to cell death.5 The
brane composition. The ER has a number of major physiological UPR involves three signaling pathways governed by three inte-
functions.1 It plays a key role in lipid and protein biosynthesis; gral ER membrane proteins: PERK (protein kinase-like ER
it functions as a Ca2+ storage organelle. Importantly, it is the site kinase, pancreatic ER eukaryotic translation initiation factor
where most secretory and transmembrane proteins fold into (eIF)-2a kinase), IRE-1 (inositol-requiring protein 1), and ATF6
their native conformation. When proteins enter the ER, they may (activating transcription factor 6). Research related to ER stress
undergo formation of disulfide bonds through the action of and retinal disease has focused on investigations of BiP/GRP78
folding enzymes such as protein disulfide isomerase (PDI).2 and these downstream effector proteins.
Proteins that are processed through the ER frequently undergo
glycosylation, a posttranslational modification involving the
Binding protein/glucose-regulated
addition of asparagine-linked oligosaccharides. There are two protein 78
known chaperone systems in the ER, the calnexin/calreticulin BiP/GRP78 is a calcium-dependent resident ER protein that
system and binding protein/glucose-regulated protein 78 (BiP/ facilitates the transfer of proteins into the ER through the ER
GRP78). The calnexin/calreticulin system is particularly relevant translocator.2,3,6 It is a heat shock protein with a molecular
to folding of glycoproteins, although some calnexin/calreticulin weight of 78  kDa. As a key ER stress regulatory protein, it
substrates can also bind BiP/GRP78. Protein folding is essential binds to exposed hydrophobic amino acid sequences of poly-
for protein function; therefore, sophisticated mechanisms have peptides that would normally be buried in the interior of
evolved to ensure that proper folding occurs or that irreversibly correctly folded proteins. Under nonstress conditions, BiP/
misfolded proteins are eliminated. Quality control is an ER sur- GRP78 binds to the luminal domains PERK, IRE1, and ATF6,
veillance mechanism to permit only properly folded proteins to maintaining them in an inactive state (Fig. 23.2). When mis-
exit the ER en route to other organelles. folded proteins accumulate, BiP/GRP78 preferentially binds
When there is perturbation of ER function (such as inhibition to these misfolded proteins and dissociates from the UPR
of glycosylation or disulfide bond formation, disruption of Ca2+ sensor proteins allowing their activation. BiP/GRP78 and other
homeostasis, hypoxia, and infection), unfolded or misfolded UPR gene targets such as GRP94 and calreticulin contain an
proteins accumulate. This situation, termed ER stress, is defined ER stress response element (ERSE, CCAAT(N9)CCACG) that
as an imbalance between the cellular demand for ER function is required and sufficient to activate the UPR. The absolute
and ER capacity.2–4 When the influx of nascent unfolded poly- requirement for BiP/GRP78 for survival is underscored by
peptides exceeds the folding and processing capacity of the ER, evidence that absence of BiP/GRP78 results in perimplantation
the normal physiological state of the ER is perturbed, activating embryonic lethality in mice.7 Upregulation of BiP gene expres-
signaling pathways, termed the ER stress response or the sion is considered by some investigators as a marker for ER
unfolded protein response (UPR), to return the ER to its normal stress induction.
Fig. 23.1 Overview of the unfolded protein
response (UPR). The ribosome sits at the
endoplasmic reticulum (ER) membrane and
530 the nascent protein is translated. If the
protein is folded properly (1), it will typically
exit the ER to the Golgi apparatus for further
modifications before being secreted or
Section 2

inserted into plasma membrane. If the


Golgi complex protein is misfolded, several outcomes are
possible that comprise the UPR: (2) protein
translation may be attenuated, halting protein
synthesis temporarily to prevent further
Endoplasmic reticulum accumulation of unfolded or misfolded
Degradation proteins; (3) transcriptional induction of ER
chaperone genes may occur, which will
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

increase protein folding capacity within the


3 cell; or (4) transcriptional induction of
ER-associated degradation (ERAD) activity
will dispose of the misfolded protein. If these
4 ER chaperones mechanisms are not successful in managing
ERAD
the misfolded protein, genes that trigger
apoptosis (5) may be upregulated, leading
to cellular death. Excessive accumulation
of misfolded proteins is toxic to cells
and incompatible with long-term survival.
(Courtesy of Ms Laura McKie, Division of
Visual and Instructional Design, Georgia
5 Misfolded protein
Health Sciences University.)
Apoptosis
Retinal diseases
(e.g. retinitis
pigmentosa, Aggregation
diabetic 1
retinopathy, Native protein
macular (properly folded)
degeneration)
2
Attenuation of
translation

mRNA Ribosome

PERK IRE1
The signaling events mediated by PERK represent the most IRE1 is a 100-kDa bifunctional protein with kinase and endo­
immediate response to ER stress in metazoan cells. PERK is ribonuclease (RNase) activity. It was the first component in the
an ER-associated transmembrane serine/threonine protein UPR pathway to be identified10 and is evolutionarily the oldest
kinase. When unfolded proteins accumulate in the ER lumen, branch of the UPR. Under nonstress conditions, the protein
BiP/GRP78 dissociates from PERK, which then dimerizes and kinase is maintained in a monomeric form through its interac-
is subsequently autophosphorylated, triggering phosphoryla- tions with BiP/GRP78. IRE1 can bind members of the tumor
tion of eukaryotic translation initiation factor 2 on the alpha necrosis factor (TNF) receptor family and can activate protein
subunit (eIF2α). Phosphorylation of eIF2α attenuates mRNA kinases that are implicated in immunity, inflammation, and
translation, thereby preventing influx of newly synthesized apoptosis. Under conditions of ER stress, when unfolded pro-
polypeptides into the ER lumen of the stressed cell.6 Interest- teins are accumulating, IRE1 is released from BiP/GRP78, dimer-
ingly, although phosphorylation of eIF2α inhibits translation izes, and autophosphorylates to activate its RNase activity.2–4 In
initiation in general, it is required for selective translation of mammals, there are two forms of the protein, IRE1α and IRE1β.
several mRNAs such as ATF4. Activation of ATF4 can increase IRE1α is expressed in most cells and tissues (including retina),
levels of chaperones, restore cellular redox homeostasis, and while IRE1β is primarily in intestinal epithelial cells. Upon acti-
help the ER to fold proteins or degrade them. Lack of the vation of the UPR, IRE1 RNase activity initiates removal of a
gene encoding PERK is not lethal, but does result in increased 26-nucleotide intron from X-box binding protein (XBP1) mRNA.
hypersensitivity to ER stress.8 It has been reported that activa- Spliced XBP1 is a transcriptional activator that activates many
tion of PERK induces transcription of ~1/3 of UPR-dependent UPR target genes through its interaction with ERSE and it can
genes.9 activate genes required for ER-associated degradation (ERAD).
Fig. 23.2 Key regulators of the unfolded
protein response (UPR). When misfolded
BiP/ proteins aggregate, binding protein/glucose-
GRP78 regulated protein 78 (BiP/GRP78) dissociates 531
from the three endoplasmic reticulum (ER)
GR iP/
8
P7

stress receptors, protein kinase-like ER

GRP78
B

BiP/
kinase (PERK), activating transcription factor

Chapter 23
6 (ATF6), and inositol-requiring protein 1
Misfolded protein (IRE1), which permits their activation.
BiP/GRP78 release Activation of PERK blocks general protein
synthesis by phosphorylating eukaryotic
initiation factor 2α (eIF2α), which enables
translation of ATF4 (through an eIF2α-
BiP/ BiP/ BiP/ independent translation pathway). ATF4
GRP78 GRP78 GRP78 translocates to the nucleus and induces the

Mechanisms of ER Stress in Retinal Disease


transcription of genes required to restore ER
homeostasis. When IRE1 is released from
PERK Ire1
ATF6 BiP/GRP78, it dimerizes and
autophosphorylates to activate its RNase
activity, removing a 26-nucleotide intron from
X-box-binding protein (XBP1) mRNA. Spliced
XBP1 activates many UPR target genes and
elF2 can activate genes required for
Endoplasmic reticulum

sXBP1 XBP1
mRNA mRNA ER-associated degradation (ERAD). ATF6 is
ATF6 activated by limited proteolysis after its
translocation from the ER to the Golgi
apparatus. Active ATF6 can regulate the
elF2 expression of ER chaperones and XBP1.
The combined action of these three ER
stress receptors is to attenuate translation to
Cleaved prevent further accumulation of misfolded
proteins, to enhance proper protein folding,
Golgi and to degrade misfolded proteins. CHOP,
ATF4 sXBP1 ATF6 CCAAT-enhancer-binding protein
complex
homologous protein. (Courtesy of Ms Laura
McKie, Division of Visual and Instructional
Design, Georgia Health Sciences University.)

Cleaved

ATF4 sXBP1
ATF6

Gene expression Gene expression Gene expression


Nucleus

ERAD ERAD Chaperones


UPR UPR CHOP
Autophagy P58IPK XBP1
CHOP

IRE1 may be a focal point for the BCL-2 family of proteins that PERK and IRE1α, ATF6α and ATF6β do not undergo oligomer-
regulates cell death. BCL-2-associated X protein (BAX) and ization, rather in the Golgi ATF6 is cleaved by proteases and the
BCL-2 antagonist/killer (BAK) interact physically with IRE1α, resultant cytoplasmic portion translocates to the nucleus, where
modulating the UPR. If either IRE1 or XBP1 is absent in mouse it (like XBP1) binds to ERSE to activate transcription of ER chap-
models, the result is embryonic lethality. erone genes such as BiP/GRP78, GRP94, and calreticulin. Thus,
ATF6 activation can increase ER chaperone activity. If both
ATF6 ATF6α and ATF6β are deleted in mouse, the result is early
ATF6α and ATF6β are bZIP family transcription factors. In the embryonic lethality.
absence of ER stress, BiP/GRP78 binds to the luminal domain
of ATF6, tethering it to the ER membrane. When unfolded pro- ER-associated degradation
teins accumulate, BiP/GRP78 releases ATF6, which then trans- The ER employs ERAD to clear aggregated, misfolded, or unas-
locates to the Golgi apparatus by vesicular transport.3,6 Unlike sembled proteins. Target proteins are selected through the ER
quality control system, retrotranslocated to the cytosol, and To investigate the role of ER stress genes and proteins in
degraded by the ubiquitin proteosome system. The four steps retina, a number of research tools (e.g., antibodies, molecular
532 associated with ERAD are recognition, retrotranslocation, ubi­ probes) are available commercially or have been developed by
quitination, and degradation.3 In the recognition step, glycosyl- individual laboratories. Figure 23.3 shows immunocytochemical
ated proteins are bound to ER degradation-enhancing studies performed in freshly isolated mouse retinal Müller cells
α-mannosidase-like protein that can discriminate folded from to detect two ER proteins. PDI is a known ER-resident protein
Section 2

unfolded proteins. Misfolded proteins destined for the ret- and hence an excellent marker of this organelle (Fig. 23.3A). In
rotranslocation machinery associate with PDI and BiP/GRP78 to this dual-label experiment, a second antibody was used to detect
cleave disulfide bonds and to unfold the partially folded protein. BiP/GRP78, the key ER stress-regulatory protein (Fig. 23.3B).
Proteins are translocated to the cytoplasm where they undergo The final panel shows the merged image of PDI and BiP/GRP78;
ubiquitination by the E1–E2–E3 ubiquitin system. The proteins there is considerable overlap in the expression of these two pro-
are then deglycosylated and degraded by the proteosome. teins (Fig. 23.3C). Immunodetection methods, gene expression
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

analysis, and elegant genetic manipulation methods have formed


Apoptosis-inducing pathways the basis of investigations of the ER stress genes/proteins in
If PERK, ATF6, and IRE1 pathways cannot suppress ER stress various retinal diseases.
and if the ERAD pathway does not rectify the unfolded protein
accumulation, an apoptotic pathway will be triggered so that the Retinitis pigmentosa and other
cell will die, allowing the organism (or tissue) to survive since photoreceptor dystrophies
accumulation of unfolded proteins is toxic to cells. Apoptosis is RP is an inherited retinal dystrophy in which loss of photorecep-
signaled through mitochondrial-dependent and independent tors leads to progressive vision loss. The prevalence of nonsyn-
pathways. In the ER, apoptosis is signaled through several dromic RP is ~1/3500–4000. The most common form of RP is a
mechanisms, the most significant of which is the PERK/eIF2α- rod–cone dystrophy, characterized initially by night blindness,
dependent or ATF6-dependent transcriptional induction of followed by progressive loss in the peripheral visual field in
CCAAT-enhancer-binding protein homologous protein (CHOP). daylight, eventually leading to blindness after several decades.11
CHOP activates transcription of several genes that potentiate To date mutations in over 40 genes have been implicated in RP.12
apoptosis, including GADD34, ER01, DR5, TRB3, carbonic anhy- In some cases, the genes are specific to photoreceptor cells,
drase VI. One mechanism of CHOP-induced apoptosis is to including rhodopsin, rod cGMP phosphodiesterase, peripherin,
suppress the prosurvival protein Bcl-2. Additionally, GADD34 and rod outer-segment membrane protein-1, while others are
(growth arrest and DNA damage-inducible protein-34) is a par- expressed in retinal and nonretinal cells.
ticularly important transcriptional target of CHOP as it can
dephosphorylate eIF2α, restoring global protein translation and Rhodopsin mutations
suppressing ATF4 translation.
Among the genes associated with RP, mutations within the rho-
RETINAL DISEASES ASSOCIATED WITH dopsin gene account for approximately 25% of cases of autoso-
mal dominantly inherited RP (adRP). AdRP is a human protein
ER STRESS folding disease that is frequently caused by a proline-to-histidine
The UPR has been referred to as a “double-edged sword” mutation at position 23 of rhodopsin (P23H rhodopsin) that
because it can restore cellular homeostasis, but if unchecked may leads to its retention in the ER.13,14 In vitro studies have shown
lead to chronic, overwhelming stress that can cause apoptotic that cells transfected with P23H rhodopsin increased expression
cell death. ER stress and the UPR have been implicated in a of BiP mRNA to a level greater than in cells transfected with
number of retinal diseases, including retinitis pigmentosa (RP), wild-type rhodopsin.13,14 Interestingly, activation of the IRE1α
diabetic retinopathy, and macular degeneration. pathway was protective, while prolonged PERK activation was

A B C

Fig. 23.3 Immunocytochemical analysis of protein disulphide isomerase (PDI) and binding protein/glucose-regulated protein 78 (BiP/GRP78) in
retinal Müller cells. Primary Müller cells were isolated from neonatal mouse retina using methods established in the author’s lab and subjected to
immunocytochemical methods to detect two proteins: PDI, a known endoplasmic reticulum (ER) protein, and BiP/GRP78, the major ER stress-
regulatory protein. The anti-PDI antibody was detected with a secondary antibody that fluoresces red (A); the anti-BiP/GRP78 antibody was
detected with a secondary antibody that fluoresces green (B). When the cells were viewed by epifluorescence using filters to detect green and
red fluorescence simultaneously, the areas of colocalization appear yellow in the merged image (C). The cells were also labeled with a dye
4’-6-diamidino-2-phenylindole (DAPI), which forms fluorescent complexes with natural double-stranded DNA, hence the nucleus stains blue in the
merged image (C). (Studies performed by Dr Yonju Ha in the author’s lab.)
associated with cell death. Elegant studies by Gorbatyuk and retinal dystrophy in early childhood.21 Mutations in RPE65 or
colleagues14 provide immunohistochemical data showing that lecithin-retinol acyltransferase (LRAT) disrupt 11-cis-retinal recy-
when HeLa cells are transfected with mutant rhodopsin (P23H), cling, causing this devastating disease. LRAT catalyzes the ester- 533
the protein was not able to traffic to the cell membrane and was ification of all-trans retinol (vitamin A) to all-trans retinyl esters,
localized in the cytoplasm – clear evidence of the retention in the which are the substrate for RPE65, to produce 11-cis retinol. In

Chapter 23
ER of a misfolded protein.14 These cell culture observations have studies of a murine model of LCA (LRAT−/− mouse), large quanti-
been extended to studies in an animal model of adRP (transgenic ties of M and S opsins are mislocalized to the inner regions of
rat expressing P23H rhodopsin at high levels). During retinal cones, creating an extra burden on the cell.22 Interestingly, mis-
development, levels of BiP mRNA were high, but diminished localized M opsin is degraded, whereas S opsin is resistant to
with age; however, levels of CHOP indicative of apoptosis proteasome degradation, resulting in far more toxic aggregation
increased significantly with age as the retinopathy advanced in of S opsin in the ventral and central retina than of M opsin in
the adRP rat model.13 Subsequent experiments in which BiP/ the dorsal retina. Furthermore, aggregation of S opsin leads to

Mechanisms of ER Stress in Retinal Disease


GRP78 was overexpressed using adenoviral vector-mediated CHOP activation. The UPR in cones copes with mislocalized M
delivery in the rat adRP model resulted in improved a- and opsin more effectively than mislocalized S opsin. Thus, M opsin
b-wave amplitudes of the scotopic electroretinogram (ERG) and is degraded by the ERAD pathway, which relieves ER stress.
a reduction in photoreceptor cell loss.14 The field of microribo- S opsin was resistant to ERAD, resulting in aggregation/
nucleic acid (micro-RNA or miRNA) expression profiling and accumulation, which induces apoptosis.
bioinformatics has identified miR-708 in the homeostatic regula- The apparent role of ER stress and UPR in some forms of
tion of ER function in photoreceptor cells, specifically preventing photoreceptor disease has prompted suggestions that therapeu-
excess rhodopsin from entering the ER. Investigators speculate tic approaches, which modulate these pathways, may prove
that miR-708 may function analogously to UPR control proteins beneficial.22–24
such as PERK and IRE1α.15
Diabetic retinopathy
cGMP-PDE mutations Diabetic retinopathy is a major complication of diabetes mellitus,
Mutations of the gene coding for the β subunit of the rod a complex metabolic disorder characterized by deficiency of or
photoreceptor-specific cGMP phosphodiesterase 6 (PDE6-β) insensitivity to insulin. Diabetic retinopathy is a neurovascular
underlie cases of autosomal recessive RP (arRP) and account for disease.25,26 The microvascular characteristics, which are visible
~1–2% of all cases of human RP.16 The rd1 mouse carries a non- clinically, include pericyte dropout, microaneurysms, intrareti-
sense mutation of this gene and has proven to be a very useful nal hemorrhages, capillary nonperfusion, intraretinal micro­
model for understanding the pathogenic mechanisms of this vascular abnormalities, and neovascularization. The neuronal
form of RP. The absence of phosphodiesterase activity leads to component is characterized by death of ganglion cells, loss of
increased accumulation of cGMP in photoreceptors, which leads cells in the inner nuclear layer,27,28 and functional changes
to increased influx of Na+ and Ca2+ through cGMP-gated cation detected, particularly using multifocal ERG.29 ER stress has been
channels. The uncontrolled influx of Ca2+ triggers apoptosis of implicated in diabetic retinopathy and investigations have
photoreceptor cell nuclei so that between postnatal days 10 and examined features of ER stress in retinal vascular cells and
21, rod photoreceptor cells are lost. The number of rows of cells retinal neurons.30
in the retinal outer nuclear layer decreases from ~10–12 to ~1–2, Regarding the vasculature, retinal homeostasis is regulated in
representing remaining cones. Interestingly, as the rod cells are part by the blood–retinal barrier. The outer barrier is comprised
lost there is an increase in expression of BiP/GRP78, phosphory- of the tight junctions between retinal pigment epithelial (RPE)
lated eIF2α, phosphorylated PERK and caspase-12 over post­ cells while the inner blood–retinal barrier is composed of tight
natal days 10–14, but levels decrease by postnatal day 21.17 These junctions between vascular endothelial cells. Breakdown of this
data clearly implicate ER stress in the pathogenesis of RP caused barrier is characteristic of diabetic retinopathy. TNF-α is a major
by mutations of PDE-β. proinflammatory cytokine induced by diabetes that plays a key
role in endothelial cell injury in diabetic retinopathy. Investiga-
Carbonic anhydrase mutations tors have shown that ER stress plays a pathogenic role in retinal
Carbonic anhydrase IV (CA IV) is another gene that, when mutated, inflammation and vascular leakage in diabetic retinopathy.31–33
leads to human adRP that involves ER stress. This form of RP Interestingly, preconditioning human retinal endothelial cells
(RP17) is caused by an arginine-to-tryptophan (R14W) mutation with very low levels of ER stress-inducing agents such as tunica-
in the signal sequence of carbonic anhydrase IV.18–20 This protein mycin actually alleviate TNF-α-induced endothelial adhesion
is a glycosylphosphatidylinositol-anchored membrane protein molecule expression, retinal leukostasis, and vascular leakage in
expressed in the choriocapillaris of the eye and in the renal epi- vitro.34 The beneficial effects of ER stress preconditioning require
thelium. Interestingly, the mutation results in an exclusively that XBP1, a major regulator of the adaptive response to ER,
ocular phenotype. In vitro studies have shown that in cells, must be activated if ER stress is to be protective of endothelial
R14W mutation, which is the RP17 form of adRP, leads to accu- cell function.34 There are reports showing that P58IPK, a 58-kDa
mulation of CAIV as unfolded protein in the ER. This gene defect inhibitor of protein kinase, which has been shown to be impor-
is associated with increased expression of BiP/GRP78, PERK, tant in ER stress, reduced the level of TNF-α in endothelial
and CHOP, leading to cell death.19 cells.35 Thus, the potential role of modulating ER stress may
prove therapeutically useful for the endothelial cell alterations
LRAT mutations observed in diabetic retinopathy.
An inherited retinal dystrophy that preferentially affects cones Vascular endothelial growth factor (VEGF) plays a key role in
before rods is Leber congenital amaurosis (LCA), the most severe the development and progression of diabetic retinopathy.36
Investigators have shown that homocysteine, which induces ER the σR1 ligand (+)-pentazocine showed ER stress gene expres-
stress, increased expression of VEGF along with BiP/GRP78.37 sion levels that were very similar to the age-matched wild-type
534 Incubating RPE cells with homocysteine in vitro caused transient mice (Fig. 23.4). The data suggest that targeting ER stress may
phosphorylation of eIF2α and increased ATF4 protein level. In hold promise in the treatment of diabetic retinopathy.
addition to its effects on vasculature, excess homocysteine
induces death of ganglion cells and, in a mouse model of hyper- Macular degeneration
Section 2

homocysteinemia, diabetes accelerates the loss of these retinal The macula is the cone photoreceptor-dense region of the retina;
neurons.38 dystrophy of this area is collectively termed “macular degenera-
Retinal neurons die in diabetic retinopathy, as evidenced by tion.” Macular degeneration can develop at an early age through
decreased contrast sensitivity,29 decreased blue–yellow color single-gene mutations or can present later in life as the much
sensitivity,39 and reduced electrical responses in full-field and more common multifactorial age-related macular degeneration
multifocal ERG.40 ER stress has been implicated in the death of (AMD).12 Clinically, patients present with progressive loss of
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

retinal neurons, particularly ganglion cells. Studies subjecting visual acuity, abnormal color vision, and central scotomas. ER
the retinal neuronal cell line RGC-5 to oxidative stress, as stress has been implicated in the pathogenesis of both genetically
observed in diabetic retinopathy, demonstrated increased inherited, early-onset macular dystrophies and multifactorial
expression of the ER stress gene BiP/GRP78 as well as PERK, age-related macular degenerations.
IRE1α, ATF6, and the apoptosis gene CHOP.41 Upregulation of
several of these ER stress markers has been detected in neural Early-onset macular dystrophies
retinas of the Ins2Akita/+ mouse.31,41 The Ins2Akita/+ mouse is an Mutations of at least two genes leading to juvenile macular dys-
endogenous model of diabetic retinopathy characterized by trophy are associated with ER stress, ELOVL4 and EFEMP1.
marked apoptosis of retinal neurons, including ganglion cells ELOVL4 encodes a 314-amino-acid ER-bound transmembrane
and cells of the inner nuclear layer and vasculopathy.42,43 Inter- protein associated with the long-chain fatty acid synthesis
estingly, treatment of this mouse with a ligand for sigma receptor machinery. Retinal tissue has a unique fatty acid composition;
1 (σR1), a molecular chaperone that binds BiP/GRP78, provided the lipid environment is critical for normal retinal functions.
marked neuroprotection against neuronal cell death when it While the precise role of ELOVL4 in photoreceptors is not
was administered over a period of several weeks.44 Examination known, mutations of ELOVL4 result in an autosomal dominant
of the ER stress genes in retinas of these mice (using quantita- atrophic macular dystrophy resembling Stargardt macular
tive reverse-transcriptase polymerase chain reaction) showed degeneration; hence the disease is referred to as Stargardt-like
that BiP/GRP78 expression increased in diabetic mice compared macular dystrophy. Wild-type ELOVL4 is localized to the ER;
with wild-type mice, as did PERK, ATF6, IRE1, ATF4, and CHOP however the mutant form of the protein accumulates in the Golgi
(Fig. 23.4). The retinas of Ins2Akita/+ diabetic mice treated with apparatus.45 Investigators have transfected cells with the mutant
forms of ELOVL4, known to cause Stargardt-like macular dys-
trophy, and observed an increase in the expression of BiP/
GRP78 and the UPR apoptosis-associated gene, CHOP.46
WT Ins2Akita/+ Ins2Akita/+ (+)-PTZ
EFEMP1 (epithelial growth factor (EGF)-containing fibulin-
2.5 * * like extracellular matrix protein 1) encodes an extracellular
* matrix protein, fibulin-3. Fibulin-3 is a glycoprotein that typi-
* cally undergoes proper folding in the ER, is transported to the
Relative quantity of mRNA

2
* Golgi, and then secreted. A missense point mutation (arginine-
to-tryptophan (Arg345Trp)) results in an early-onset autosomal
1.5 * dominant maculopathy, known as Doyne honeycomb retinal
dystrophy (also termed malattia leventinese). Mutant forms of
1 the protein accumulate aberrantly in the ER of RPE, hindering
proper secretion to the extracellular milieu.47,48 To determine the
consequences of the Arg345Trp mutation on ER stress, investiga-
0.5
tors transfected the human ARPE-19 cell line with the mutated
EFEMP1 and demonstrated an upregulation of BiP/GRP78;
0 indeed, the level of BiP/GRP78 expression paralleled the intra-
BiP PERK ATF6 IRE1α ATF4 CHOP cellular levels of fibulin-3. Additional evidence that the UPR was
ER stress genes activated by this mutation was increased IRE-1 endonuclease
activity and XBP-2 mRNA processing.48
Fig. 23.4 Quantitative analysis of endoplasmic reticulum (ER) stress
genes in retinas of wild-type (WT) versus diabetic mice. Total RNA
was isolated from neural retinas of WT, C57Bl/6-Ins2Akita/+ mice, and
Age-related macular degeneration
C57Bl/6-Ins2Akita/+ administered (+)-pentazocine, a ligand for the AMD is the leading cause of visual impairment in elderly
molecular chaperone protein sigma receptor 1 (σR1). The expression persons. The macula is the photoreceptor-dense retinal region
of ER stress genes was analyzed by quantitative reverse transcriptase
polymerase chain reaction. There was a significant increase in which, when affected by this disease, results in central vision
expression of these genes in diabetic mice (*), but expression levels loss. The RPE cells sustain photoreceptor cells through myriad
were similar to wild-type mouse retinas when treated with the ligand activities, including the transport of needed vitamins such as
for σR1. (Adapted with permission from Ha Y, Dun Y, Thangaraju M,
vitamin A and folate, removal of waste, and phagocytosis of
et al. Sigma receptor 1 modulates endoplasmic reticulum stress in
retinal neurons. Invest Ophthalmol Vis Sci 2011;52:527–40; copyright shed outer-segment discs. RPE cells are vascularized via the
held by Association for Research in Vision and Ophthalmology.) choriocapillaris. A hallmark of AMD is the accumulation of
lipofuscin and extracellular deposits known as drusen. The branches of the UPR process (i.e., the PERK, IRE1α, or ATF-6
retina and RPE are exposed constantly to oxidative stress through branches) dictates this outcome. Understanding ER stress in
intense light exposure, high metabolic activity, oxygen con- retinal diseases is emerging as an area of intense investigation. 535
sumption, and the high concentration of polyunsaturated fatty It is hoped that the outcome of these studies will lead to the
acids, making them particularly susceptible to ER stress.49 discovery and development of innovative therapeutic interven-

Chapter 23
Clinically, AMD is classified as either atrophic (dry) AMD or tion strategies for retinopathies.
exudative (wet) AMD,50 based upon whether neovascularization
has developed. A major trigger for exudative AMD is upregula- REFERENCES
tion of VEGF expression and ER stress can trigger this upregula- 1. Chen X, Karnovsky A, Sans MD, et al. Molecular characterization of the endo-
plasmic reticulum: insights from proteomic studies. Proteomics 2010;10:
tion.37,48,51 Also implicated in the development of AMD is 4040–52.
smoking52,53; cigarette smoke extract contains benzopyrene, a 2. Malhotra JD, Kaufman RJ. The endoplasmic reticulum and the unfolded
protein response. Semin Cell Dev Biol 2007;18:716–31.
potent inducer of ER stress via the PERK pathway.54 A number

Mechanisms of ER Stress in Retinal Disease


3. Yoshida H. ER stress and diseases. FEBS J 2007;274:630–58.
of studies performed using human ARPE-19 cells have impli- 4. Ni M, Lee AS. ER chaperones in mammalian development and human
cated cigarette smoke in VEGF upregulation and in induction of diseases. FEBS Lett 2007;581:3641–51.
5. Tsai YC, Weissman AM. The unfolded protein response, degradation from
ER stress.55–57 endoplasmic reticulum and cancer. Genes Cancer 2010;1:764–78.
In vivo studies of AMD implicate ER stress in the disease 6. Kaufman RJ. Regulation of mRNA translation by protein folding the endoplas-
mic reticulum. Trends Biochem Sci 2004;29:152–8.
pathogenesis. For example, Dr C. Chan (National Eye Institute, 7. Luo S, Mao C, Lee B, et al. GRP78/BiP is required for cell proliferation and
National Institutes of Health) developed a mouse model of AMD protecting the inner cell mass from apoptosis during early mouse embryonic
development. Mol Cell Biol 2006;26:5688–97.
that has defects in two genes which are involved in immunologic 8. Harding HP, Zhang Y, Bertolotti A, et al. Perk is essential for translational
processes.58 One gene encodes CX3CR1, the receptor for CX3CL1/ regulation and cell survival during the unfolded protein response. Mol Cell
2000;5:897–904.
fractalkine chemokine, which is expressed in RPE, Müller cells, 9. Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by
and microglia. Two single-nucleotide polymorphisms of CX3CR1 an endoplasmic-reticulum-resident kinase. Nature 1999;397:271–4.
coupled with a decreased number of CX3CR1 transcripts and 10. Cox JS, Walter P. A novel mechanism for regulating activity of a transcription
factor that controls the unfolded protein response. Cell 1996;87:391–404.
protein in AMD maculas are associated with AMD.59 The second 11. Hamel C. Retinitis pigmentosa. Orphanet J Rare Dis 2006;1:40.
gene, Ccl2 (MCP-1, a CC chemokine), is thought to play a homeo- 12. Berger W, Kloeckener-Gruissem B, Neidhardt J. The molecular basis of human
retinal and vitreoretinal diseases. Prog Retin Eye Res 2010;29:335–75.
static, immunoregulatory role in AMD pathogenesis.60 Aged 13. Lin JH, Li H, Yasumura D, et al. IRE1 signaling affects cell fate during the
mice with deficient Ccl2 or Ccr2, the corresponding receptor, unfolded protein response. Science 2007;318:944–9.
14. Gorbatyuk MS, Knox T, LaVail MM, et al. Restoration of visual function in
develop many cardinal features of AMD, including drusen for- P23H rhodopsin transgenic rats by gene delivery of BiP/Grp78. Proc Natl Acad
mation, RPE accumulation of lipofuscin and complement factors, Sci U S A 2010;107:5961–6.
and choroidal neovascularization.61 To mimic closely the patho- 15. Behrman S, Acosta-Alvear D, Walter P. A CHOP-regulated microRNA controls
rhodopsin expression. J Cell Biol 2011;192:919–27.
logic features of AMD, mice with mutations in these two genes 16. Bowes C, Li T, Danciger M, et al. Retinal degeneration in the rd mouse is caused
have been crossed to generate Ccl2/Cx3cr1 mice.62 Fundoscopic by a defect in the beta subunit of rod cGMP-phosphodiesterase. Nature
1990;347:677–80.
examination of Ccl2−/−/Cx3cr1−/− mice as early as 4–6 weeks of age 17. Yang LP, Wu LM, Guo XJ, et al. Activation of endoplasmic reticulum stress in
revealed drusen-like lesions that progressed to large, confluent degenerating photoreceptors of the rd1 mouse. Invest Ophthalmol Vis Sci
2007;48:5191–8.
areas of yellow deposits in the deep retina and subretinal space 18. Rebello G, Ramesar R, Vorster A, et al. 2004. Apoptosis inducing
by 4–6 months of age and flattened atrophic areas by 6 months signal sequence mutation in carbonic anhydrase IV identified in patients
with the RP17 form of retinitis pigmentosa. Proc Natl Acad Sci U S A
of age. Studies of the retinas of these mice suggest that the patho- 101:6617–22.
genesis of AMD may be mediated by ER stress and protein 19. Stams T, Nair SK, Okuyama Pandor A, et al. Cell-specific differences in the
misfolding. Indeed, there is decreased expression at the mRNA processing of the R14W CAIV mutant associated with retinitis pigmentosa 17.
J Cell Biochem 2010;111:735–41.
and protein level of ERp29, a molecular chaperone protein.58 20. Datta R, Waheed A, Bonapace G, et al. Pathogenesis of retinitis pigmentosa
Given that AMD is multifactorial and there is no single mutated associated with apoptosis-inducing mutations in carbonic anhydrase IV.
Proc Natl Acad Sci U S A 2009;106:3437–42.
protein to be targeted, Sauer and colleagues hypothesized that 21. den Hollander AI, Roepman R, Koenekoop RK, et al. Leber congenital amau-
enhancing chaperone activity through the use of chemical and/ rosis: Genes, proteins and disease mechanisms. Prog Retin Eye Res 2008;
27:391–419.
or pharmacological chaperone compounds may prove beneficial 22. Zhang T, Zhang N, Baehr W, et al. Cone opsin determines the time course of
to many individuals suffering from this devastating sight- cone photoreceptor degeneration in Leber congenital amaurosis. Proc Natl
threatening disease.49 A promising example of this concerns the Acad Sci U S A 2011;108:8879–84.
23. Mendes CS, Levet C, Chatelain G, et al. ER stress protects from retinal
chaperone protein, αB crystallin, a 20-kDa member of the small degeneration. EMBO J 2009;28:1296–307.
heat shock protein (HSP) family. HSPs prevent aggregation of 24. Farrar GJ, Palfi A, O’Reilly M. Gene therapeutic approaches for dominant
retinopathies. 3. Curr Gene Ther 2010;10:381–8.
folded proteins and facilitate intracellular protein trafficking. αB 25. Gardner TW, Abcouwer SF, Barber AJ, et al. An integrated approach to diabetic
crystallin is secreted from RPE cells and is taken up by adjacent retinopathy research. Arch Ophthalmol 2011;129:230–5.
26. Barber AJ, Gardner TW, Abcouwer SF. The significance of vascular and neural
photoreceptor cells conferring neuroprotection.63 As increased aβ apoptosis to the pathology of diabetic retinopathy. Invest Ophthalmol Vis Sci
crystallin is a biomarker for AMD, it may be fruitful to exploit 2011;52:1156–63.
27. Barber AJ, Lieth E, Khin SA, et al. Neural apoptosis in the retina during experi-
increased secretion toward neuroprotective effects. mental and human diabetes: early onset and effect of insulin. J Clin Invest
In summary, ER stress is implicated in a number of retinal 1998;102:783–91.
diseases, including RP, diabetic retinopathy, early-onset macu- 28. Abu-El-Asrar AM, Dralands L, Missotten L, et al. Expression of apoptosis
markers in the retinas of human subjects with diabetes. Invest Ophthalmol Vis
lopathies, and AMD. It is paradoxical that the UPR, which is Sci 2004;45:2760–6.
triggered by ER stress, can induce cytoprotective effects that 29. Ng JS, Bearse Jr MA, Schneck ME, et al. Local diabetic retinopathy prediction
by multifocal ERG delays over 3 years. Invest Ophthalmol Vis Sci 2008;49:
restore homeostasis but can also induce cell-destructive effects 1622–8.
that promote apoptosis. It is not clear how the UPR integrates 30. Oshitari T, Hata N, Yamamoto S. Endoplasmic reticulum stress and diabetic
retinopathy. Vasc Health Risk Manag 2008;4:115–22.
these opposing outcomes to progress toward survival or death. 31. Li J, Wang JJ, Yu Q, et al. Endoplasmic reticulum stress is implicated in retinal
It has been postulated that the duration of expression of specific inflammation and diabetic retinopathy. FEBS Lett 2009;583:1521–7.
32. Yang H, Liu R, Cui Z, et al. Functional characterization of 58-kilodalton inhibi- protein response and VEGF expression. Invest Ophthalmol Vis Sci 2005;46:
tor of protein kinase in protecting against diabetic retinopathy via the endo- 3973–9.
plasmic reticulum stress pathway. Mol Vis 2011;17:78–84. 49. Sauer T, Patel M, Chan CC, et al. Unfolding the therapeutic potential of chemi-
536 33. Ikesugi K, Mulhern ML, Madson CJ, et al. Induction of endoplasmic reticulum cal chaperones for age-related macular degeneration. Expert Rev Ophthalmol
stress in retinal pericytes by glucose deprivation. Curr Eye Res 2006;31:947–53. 2008;3:29–42.
34. Li J, Wang JJ, Zhang SX. Preconditioning with endoplasmic reticulum stress 50. Jager RD, Mieler WF, Miller JW. Age-related macular degeneration. N Engl J
mitigates retinal endothelial inflammation via activation of X-box binding Med 2008;358:2606–17.
protein 1. J Biol Chem 2011;286:4912–21. 51. Salminen A, Kauppinen A, Hyttinen JM, et al. Endoplasmic reticulum stress in
Section 2

35. Yang H, Liu R, Cui Z, et al. Functional characterization of 58-kilodalton inhibi- age-related macular degeneration: trigger for neovascularization. Mol Med
tor of protein kinase in protecting against diabetic retinopathy via the endo- 2010;16:535–42.
plasmic reticulum stress pathway. Mol Vis 2011;17:78–84. 52. Klein R, Peto T, Bird A, et al. The epidemiology of age-related macular degen-
36. Praidou A, Androudi S, Brazitikos P, et al. Angiogenic growth factors and their eration. Am J Ophthalmol 2004;137:486–95.
inhibitors in diabetic retinopathy. Curr Diabetes Rev 2010;6:304–12. 53. Kabasawa S, Mori K, Horie-Inoue K, et al. Associations of cigarette smoking
37. Roybal CN, Yang S, Sun CW, et al. Homocysteine increases the expression of but not serum fatty acids with age-related macular degeneration in a Japanese
vascular endothelial growth factor by a mechanism involving endoplasmic population. Ophthalmology 2011;118:1082–8.
reticulum stress and transcription factor ATF4. J Biol Chem 2004;279:14844–52. 54. Hengstermann A, Müller T. Endoplasmic reticulum stress induced by aqueous
Basic Mechanisms of Injury in the Retina

38. Ganapathy PS, Roon P, Moister TK, et al. Diabetes accelerates retinal neuronal extracts of cigarette smoke in 3T3 cells activates the unfolded protein-response-
Basic Science and Translation to Therapy

cell death in a mouse model of endogenous hyperhomocysteinemia. dependent PERK pathway of cell survival. Free Rad Biol Med 2008;44:
Ophthalmol Eye Dis 2009;1:3–11. 1097–107.
39. Daley ML, Watzke RC, Riddle MC. Early loss of blue-sensitive color vision in 55. Bertram KM, Baglole CJ, Phipps RP, et al. Molecular regulation of cigarette
patients with type I diabetes. Diabetes Care 1987;10:777–81. smoke induced oxidative stress in human retinal pigment epithelial cells:
40. Fortune B, Schneck ME, Adams AJ. Multifocal electroretinogram delays reveal implications for age-related macular degeneration. Am J Physiol Cell Physiol
local retinal dysfunction in early diabetic retinopathy. Invest Ophthalmol Vis 2009;297:C1200–10.
Sci 1999;40:2638–51. 56. Pons M, Marin-Castaño ME. Cigarette smoke-related hydroquinone dysregu-
41. Ha Y, Dun Y, Thangaraju M, et al. Sigma receptor 1 modulates endoplasmic lates MCP-1, VEGF and PEDF expression in retinal pigment epithelium in vitro
reticulum stress in retinal neurons. Invest Ophthalmol Vis Sci 2011;52: and in vivo. PLoS ONE 2011;6:e16722.
527–40. 57. Pons M, Cousins SW, Csaky KG, et al. Cigarette smoke-related hydroquinone
42. Barber AJ, Antonetti DA, Kern TS, et al. The Ins2Akita mouse as a model of induces filamentous actin reorganization and heat shock protein 27 phosphory-
early retinal complications in diabetes. Invest Ophthalmol Vis Sci 2005;46: lation through p38 and extracellular signal-regulated kinase 1/2 in
2210–18. retinal pigment epithelium: implications for age-related macular degeneration.
43. Gastinger MJ, Kunselman AR, Conboy EE, et al. Dendrite remodeling and other Am J Pathol 2010;177:1198–213.
abnormalities in the retinal ganglion cells of Ins2Akita diabetic mice. Invest 58. Tuo J, Bojanowski CM, Zhou M, et al. Murine ccl2/cx3cr1 deficiency results
Ophthalmol Vis Sci 2008;49:2635–42. in retinal lesions mimicking human age-related macular degeneration.
44. Smith SB, Duplantier JN, Dun Y, et al. In vivo protection against retinal neu- Invest Ophthalmol Vis Sci 2007;48:3827–36.
rodegeneration by the sigma receptor 1 ligand (+)-pentazocine. Invest Ophthal- 59. Tuo J, Smith B, Bojanowski CM, et al. The involvement of sequence variation
mol Vis Sci 2008;49:4154–61. and expression of CX3CR1 in the pathogenesis of age-related macular degen-
45. Ambasudhan R, Wang X, Jablonski MM, et al. Atrophic macular degeneration eration. FASEB J 2004;18:1297–99.
mutations in ELOVL4 result in the intracellular misrouting of the protein. 60. Forrester JV. Macrophages eyed in macular degeneration. Nat Med 2003;9:
Genomics 2004;83:615–25. 1350–51.
46. Karan G, Yang Z, Howes K, et al. Loss of ER retention and sequestration of the 61. Ambati J, Anand A, Fernandez S, et al. An animal model of age-related macular
wild-type ELOVL4 by Stargardt disease dominant negative mutants. Mol Vis degeneration in senescent Ccl-2- or Ccr-2-deficient mice. Nat Med 2003;9:
2005;11:657–64. 1390–7.
47. Marmorstein LY, Munier FL, Arsenijevic Y, et al. Aberrant accumulation of 62. Chan CC, Ross RJ, Shen D, et al. Ccl2/Cx3cr1-deficient mice: an animal model
EFEMP1 underlies drusen formation in malattia leventinese and age-related for age-related macular degeneration. Ophthalmic Res 2008;40:124–8.
macular degeneration. Proc Natl Acad Sci U S A 2002;99:13067–72. 63. Sreekumar PG, Kannan R, Kitamura M, et al. αB crystallin is apically secreted
48. Roybal CN, Marmorstein LY, Vander Jagt DL, et al. Aberrant accumulation of within exosomes by polarized human retinal pigment epithelium and provides
fibulin-3 in the endoplasmic reticulum leads to activation of the unfolded neuroprotection to adjacent cells. PLoS ONE 2010;5:e12578.
Basic Mechanisms of Injury in the Retina Section 2

Cell Death, Apoptosis, and Autophagy Chapter

in Retinal Injury
Michael E. Boulton, Sayak K. Mitter, Haripriya Vittal Rao, William A. Dunn Jr 24 
INTRODUCTION fragmentation, modification of cytoplasmic organelles, plasma
membrane blebbing, and engulfment by neighboring cells (Fig.
Cell death is an inevitable consequence of life! It plays a critical 24.1).1 Apoptosis can be initiated by a variety of stimuli through
role in development by eliminating transitory organs and tissues two distinct pathways.4 The extrinsic pathway is triggered after
(e.g., hyaloid vessels) and tissue remodeling where cell death the interaction of death receptors present on the cell surface with
allows the removal of excess or unwanted cells (e.g., death of their cognate ligands (e.g., Fas ligand, tumor necrosis factor
oligodendrocytes in the optic nerve). During life, dead and (TNF)-α and TNF-related apoptosis-inducing ligand (TRAIL))
damaged cells are constantly being replaced to ensure homeo- and which can start the downstream executioner caspase (cyst­
stasis and normal functioning of multicellular organisms. eine aspartic acid proteases) cascade within seconds of ligand
However, with increasing age, cell death can exceed replace- binding. By contrast, the intrinsic pathway is initiated by a mul-
ment, leading to loss of tissue/organ function. Furthermore, titude of intracellular triggers, collectively called “stress signals,”
injury and disease can lead to excessive cell loss which over- which can include oxidative damage, deoxyribonucleic acid
whelms the organism and leads to functional impairment and (DNA) damage, loss of cell–cell contact, growth factor with-
even death. The majority of cell death in the retina occurs by drawal, hypoxia, and endoplasmic reticulum stress that all target
apoptosis which has the big advantage over necrosis that it is the mitochondria and induce the release of proapoptotic factors
self-contained and does not result in an overt inflammatory to the cytosol that activate caspases. Excessive or deficient apop-
response. In an attempt to maintain homeostasis and prevent tosis is involved in numerous disease states. Readers requiring
excess cellular damage the cells remove damaged organelles and more detail are directed to the work of Green and Reed.2,3
protein aggregates by a process called autophagy. The following
chapter will discuss cell death in the retina in detail in the context Necrosis
of development, aging, and diseases such as glaucoma, diabetic
Necrosis has been defined as a type of uncontrolled cell death
retinopathy (DR), and age-related macular degeneration (AMD).
that can occur in response to infection, toxins, chemicals, injury,
It will further consider the initiating factors in retinal apoptosis
or lack of blood supply.2,5 Morphologically, necrosis is associated
and discuss potential therapeutic strategies for preventing
with cytoplasmic swelling (oncosis), rupture of the plasma mem-
retinal cell death and preserving vision.
brane, swelling of cytoplasmic organelles, and moderate chro-
matin condensation (Fig. 24.1). The big pathophysiological
MODES OF CELL DEATH difference between necrosis and apoptosis is inflammation.
According to the Nomenclature Committee on Cell Death 2009, Necrosis culminates in the uncontrolled release of antigens
“Cell death can be classified according to its morphological which lead to activation of the immune system and inflamma-
appearance (which may be apoptotic, necrotic, autophagic or tion whereas in apoptosis cell-bound bodies are formed which
associated with mitosis), enzymological criteria (with and are phagocytosed by neighboring cells and there is an absence
without the involvement of nucleases or of distinct classes of of inflammation. However, recent studies suggest that there is a
proteases, such as caspases, calpains, cathepsins and transgluta- molecular signaling network that can regulate the necrotic cell
minases), functional aspects (programmed or accidental, physi- death pathway.6
ological or pathological) or immunological characteristics
(immunogenic or non-immunogenic).”1 A cell is normally con- Other
sidered dead once it has passed an irreversible phase in the A number of other cell death pathways have been identified, of
death process. which autophagic cell death has gained some prominence.
Increased autophagy (see below), such as that occurring under
Apoptosis starvation, leads to self-destruction of intracellular organelles for
Apoptosis, or programmed cell death, has received extensive provision of nutrients which, if starvation is not reversed, will
study due to its critical role in development, tissue homeostasis, culminate in self-destruction of cells and tissues. Autophagic cell
and pathology.2,3 Furthermore, apoptosis does not elicit an death is morphologically defined as occurring in the absence of
inflammatory response, thus allowing “physiological” cell chromatin condensation, massive autophagic vacuolization, and
death to take place without pathological consequences. Morpho- little or no uptake by neighboring cells.1 Interestingly, auto­
logical features of apoptosis include rounding up of the cell, phagy is upregulated in a number of neurodegenerative diseases
reduction in cellular and nuclear volume (pyknosis), nuclear and thus may contribute to cell loss associated with these
induces autophagy as well as apoptosis mediated by the Bcl-2
family proteins.6,7 Atg5 is essential for autophagy, but upon
538 cleavage by calpains it has been reported that the truncated Atg5
associates with BclxL to promote cytochrome c release and
caspase-dependent apoptosis.8 For many years, necrosis was
regarded as the result of an accidental and uncontrolled process.
Section 2

However, accumulating evidence now suggests that necrotic cell


death might also be mediated by a specific set of signal transduc-
Necrosis Apoptosis tion pathways and degradative mechanisms. Similar to apop­
Viable cell
tosis, cell death with a necrotic appearance can contribute to
embryonic development and adult tissue homeostasis. Some
gene products, such as TNFR, CD95, TRAIL-R and RIP1, might
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

trigger both apoptosis and necrosis, depending on interaction


with other proteins.5 Moreover, there is cross-talk between these
two cell death modalities. For example, inactivation of caspases
might cause a shift from apoptosis to necrosis, or to a mixture
of these two cell death modes.5 Thus cell death is not as easily
defined as generally believed and there is considerable cross-talk
Cell shrinks between the different cell death mechanisms.
Chromatin condensation
Cell swells Is cell death a bad thing?
Cell death is often considered as a pathological endpoint, but is
this really the case? During development and differentiation we
produce an excess of many cell types and these undergo apop-
tosis. Cells are constantly dying and being replaced as part of
the overall homeostatic process to remove dysfunctional and
damaged cells. Therefore, in this context cell death is a good
thing as it maintains optimal function in multicellular organ-
isms. Provided the balance between loss and replacement
remains constant, all is good; however, this is not always the case
in aging or pathology where cell death increases and cell replace-
“Budding” ment decreases, resulting in net cell loss and an impaired organ-
ism. Our therapeutic approach is to inhibit cell death. While
laudable, this may not always be the best option as keeping alive
Cell becomes leaky damaged, mutated, or dysregulated cells will be to the detriment
membrane blebbing
of the organism.

AUTOPHAGY AND CELL MAINTENANCE


Autophagy is essential for cellular housekeeping and homeosta-
sis through the sequestration and transfer of intracellular com-
ponents (e.g., protein aggregates, organelles) to lysosomes for
degradation.9–11 In mammalian cells, three primary types of
autophagy have been reported: macroautophagy, chaperone-
Phagocytosis mediated autophagy (CMA), and microautophagy (Fig. 24.2).12–16
apototic bodies These different types of autophagy are designed to nonselec-
Lysis and inflammation (no inflammation) tively or selectively degrade different substrates and are regu-
lated by different signaling pathways.
Fig. 24.1 Apoptotic versus necrotic morphology.
Macroautophagy is the best-characterized autophagy pathway
that generally deals with larger substrates such as protein aggre-
gates, intracellular pathogens, and dysfunctional organelles such
conditions. A number of atypical cell death modalities have also as mitochondria (Fig. 24.2).9–11 The process of macroautophagy
been identified and these are reviewed in Kroemer et al.1 involves over 30 autophagy-related proteins (Atg) which regu-
late different stages of the autophagic response (Fig. 24.3).
Cross-talk between cell death pathways Macro­autophagy is initiated by the sequestration of the cytosolic
Until recently, a requirement for gene expression was docu- substrate into double membrane-bound phagophores which
mented only for apoptotic and autophagic cell death. Interest- predominantly originate from the rough endoplasmic reticulum
ingly, certain genes and their products, e.g., p53, Bcl-2 family with possible contributions from the plasma membrane or
proteins, and calpain, are important for both these modes of cell mitochondria.17–20 The resulting autophagosome acquires endo-
death.5–7 Recent work indicates that basal p53 activity suppresses somal and lysosomal proteins, ultimately maturing into a deg-
autophagy, whereas the activation of p53 by some stimuli radative autolysosome. The mTOR kinase complex is considered
Fig. 24.2 The different types of autophagy
Macroautophagy described in the text.
Mitophagy
539

Chapter 24
Parkin PINK

Lysosome

Cell Death, Apoptosis, and Autophagy in Retinal Injury


Autophagosome

ESCRT I

ESCRT II
Chaperone-mediated Microautophagy
autophagy

Hsc70 KFERQ protein

central to the signaling pathway of autophagy and can sense elongation of the phagophore membrane whereas the
regulating conditions such as nutrient abundance, energy state, GABARAP/GATE-16 subfamily is essential for a later stage
and growth factor levels.21,22 The PI3K-III complex consists of in autophagosome maturation.27 The N-termini of LC3 and
Vps34 and p150 and activators such as Beclin-1, Ambra1, ATG14, GATE-16 are required for autophagosome–lysosome fusion.28
and UVRAG. This complex plays a crucial role in the induction Once in the lysosomes, substrates are degraded by the repertoire
of the autophagic process by generating PtdIns(3)P-rich mem- of lysosomal enzymes.29 It has been proposed that the autopha-
branes, which act as platforms for ATG protein recruitment and gic elimination of mitochondria has its own specialized
autophagosome nucleation.23 Antiapoptotic BH3 proteins such pathway.16 Critical to this process are the proteins PINK1, the E3
as BclxL and Bcl-2 bind to Beclin-1, having a negative impact on ubiquitin ligase, Parkin and possibly p62, a protein that binds to
PI3K-III activity and autophagy. Elongation and completion of ubiquitin and LC3. PINK1 binds to uncoupled mitochondria
the phagophore are brought about by two ubiquitin-like conju- which then facilitates the recruitment of Parkin which leads to
gation systems, the Atg12-Atg5-Atg16 system and the Atg8- ubiquitination of mitochondrial surface proteins. The ubiqui-
phosphatidylethanolamine (PE) system. Atg7 functions as an tinated mitochondrion is then sequestered into the autophago-
E1 enzyme in both systems, while Atg10 and Atg3 act as E2 some through the likely actions of p62 and LC3.
enzymes for Atg12 and Atg8, respectively.24 The C-terminus of Autophagy can be activated by nutrient deprivation and envi-
Atg8 is cleaved by Atg4 which primes the protein for conjuga- ronmental stress. For example, amino acid starvation and reac-
tion to PE. The Atg12–Atg5–Atg16 complex recruits Atg8-PE to tive oxygen species can stimulate autophagy.30,31 Recently, a
the elongating phagophore.25,26 At least eight different Atg8 distinction has been made between starvation- and stress-
orthologs belonging to two subfamilies (LC3 and GATE-16/ induced macroautophagy, also referred to as quality control
GABARAP) occur in mammalian cells. LC3s are involved in autophagy. It has been observed that autophagic deficient cells
Fig. 24.3 The regulatory molecules involved
Autophagosome membrane sources in the different steps of the mammalian
macroautophagy pathway. Potential
540 Rough pharmacological inhibitors used to block
endoplasmic autophagy at different steps are also shown.
Mitochondria
reticulum
Phagophore
Section 2

Plasma membrane
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

PI3 Kinase complex PI3 Kinase Induction and


inhibitors complex III nucleation
3-Methyladenine, Beclin-1 UIk1/UIk2
Wortmannin, Vps34 FIP200
LY2949002 Vps15 Atg13
Atg14 Atg101

Expansion
Atg9, Atg7, Atg10, Multivesicular body
Atg12, Atg5, Atg16,
Atg4, Atg3, LC3, Endosome
GABARAP/GATE16

Fusion
Beclin-1-Vps34
Fusion inhibitor -UVRAG-Rubicon
Bafilomycin A1 LAMP-1 & -2, Rab7,
ESCRT III,
v-SNARE,
Microtubules Amphisome

Lysosome

Autolysosome

Lysosome

tend to accumulate p62-rich aggregates, which in turn cause protein type 2A (LAMP-2A) which spans the lysosomal mem-
Nrf2 to be activated after separation from its interacting partner brane leads to translocation of the cargo across the membrane
Keap1, which allows Nrf2 to mount an antioxidant response.32 and into the lysosomal lumen for degradation.12 This pathway
In addition, histone deacetylase (HDAC)6 stands out as a key has been shown to be progressively ineffective with age, because
regulator in the autophagic response to oxidative damage, as of the age-related loss of LAMP-2A.35
HDAC6 is recruited to ubiquitinated autophagic substrates, Microautophagy involves internalization of cytosolic cargo
where it stimulates autophagosome–lysosome fusion by pro- (cytosolic proteins, glycogen, and ribosomes) through invagina-
moting F-actin remodeling in a cortactin-dependent manner.33 tions of the lysosomal membrane,36,37 which resembles the for-
However, HDAC6 and cortactin are dispensable for starvation- mation of multivesicular bodies (Fig. 24.2).13,34,38 Although the
induced autophagy. molecular mechanisms in mammalian cells are poorly under-
CMA differs from the other types of autophagy as it does not stood, a recent study by Sahu et al. proposes that microautoph-
involve vesicle formation but, rather, a direct translocation of a agy relies on endosomal sorting complexes required for transport
specific set of soluble proteins across the lysosomal membrane (ESCRT) I and III, which are necessary for the formation of the
for subsequent degradation (Fig. 24.2).34 CMA cargo substrates vesicles in which the cytosolic cargo is internalized.38 It appears
include enzymes, transcription factors, binding proteins, sub- that this pathway, like CMA, also involves Hsc70 interaction
units of the proteasome and proteins involved in vesicular traf- with a substrate containing a KFERQ-like motif and that mito­
ficking and contain a KFERQ-like motif which is recognized by phagy and CMA may share common upstream pathways.34,38
the cytosolic chaperone, Hsc70. Binding of the chaperone/ As will be discussed below, autophagy plays a critical role in
substrate to the cytosolic tail of lysosome-associated membrane maintaining retinal homeostasis as it is central, together with the
adaptive attempt to compensate for the lost circuitry due to
photoreceptor loss and/or to make up for existing, yet dysfunc-
RGC
tional, synapses. 541
There is an age-related decrease in the density of photorecep-
tor cells in the human retina, with rods appearing to be more

Chapter 24
vulnerable than cones.51,52 In the equatorial retina cones decrease
IPL uniformly at approximately 16 cells/mm2/year while the
decrease in equatorial rods is greatest, 970 cells/mm2/year,
between the second and fourth decades.52 By contrast, cone
density remains relatively constant at the fovea up to the ninth
decade.51–53 It therefore appears that rod photoreceptors are more
vulnerable to loss during aging than cones and that photorecep-

Cell Death, Apoptosis, and Autophagy in Retinal Injury


INL
tors in the fovea are less susceptible to attrition. Furthermore,
compensatory adaptations have been reported following rod cell
degeneration where the space vacated by dying rods is filled by
enlarged rod inner segments from neighboring photoreceptors,
resulting in similar rod coverage at all ages.51,54–56 Evidence that
ONL cones depend on survival factors secreted by rods may explain
this differential vulnerability between rods and cones57–60 but
remains a matter of intense debate.
The loss of photoreceptors appears to precede the loss of
associated neural cells. The retinal nerve fiber layer thickness
decreases dramatically with age40,61 and is associated with a
significant loss of retinal ganglion cells (RGC) by as much as
150/mm2 over a period of 40+ years.41 RGC death at the equato-
RPE
rial regions follows a similar trend as the photoreceptors during
5 µm
aging, thus maintaining a constant photoreceptor-to-RGC ratio.52
Age-associated degeneration of the rod bipolar cells in the inner
Fig. 24.4 Immunolocalization of the autophagy protein LC3 (green) nuclear layer (INL) has been reported.62 Although rod cell death
in normal mouse retina. Nuclei are stained with 4’,6-diamidino-2-
can initiate as early as the second decade of life, the bipolar
phenylindole (DAPI: blue) and blood with agglutinin (red). LC3 is
strongly expressed in the retinal ganglion cell layer (RGC), retinal cells start to degenerate only after the fourth decade and appre-
vessels, a subpopulation of the inner nuclear layer (INL), the outer ciably reduce by the ninth decade.52 So it seems to be a phe-
nuclear layer (ONL) of rods and cones, and the retinal pigment nomenon secondary to rod cell loss. In a more comprehensive
epithelium (RPE) but is only very weakly expressed in the inner
plexifom layer (IPL). (Courtesy of Xiaoping Qi, University of Florida.) study done using multiphoton confocal microscopy to quantify
neuron densities in the RGC layer, INL, and ONL, it was seen
that the greatest neuronal loss occurs in the RGC layer and
ONL in human aging retinas, whereas the INL is relatively pre-
proteosomal system, in the removal of damaged proteins and served.63 It must also be remembered that RGCs are classified
organelles in highly metabolic nondividing cells that exist in a into a number of subtypes56 and so even a seemingly mild loss
pro-oxidative retinal environment. Autophagy proteins are of RGC in the initial phases could imply the loss of a major
strongly expressed in the retina (Fig. 24.4). However, problems subtype of RGC that could start affecting visual perception.
occur when basal levels of autophagy become dysregulated as Despite numerous studies to determine age-related changes in
either a decrease or increase in autophagy flux will have signifi- RPE cell density, outcomes vary and are highly dependent on
cantly detrimental effects on cell function.39 retinal location. Two studies have reported that RPE density
decreases with age in the equatorial retina and is greatest in the
AGE-RELATED RETINAL CELL LOSS periphery,52,64 with an estimated loss of 0.3% per year.64 By con-
It is well recognized that the human retina undergoes numerous trast, no significant age-related decrease in RPE cell density was
age-related changes which result in altered morphology, observed at the foveal center, suggesting that, like foveal cones,
reduced function, and cell loss. Not surprisingly, this is associ- the RPE cells in this region are more resistant to attrition than
ated with a significant reduction of retinal thickness as a function those outside the fovea.52 However, a further study reported that
of age.40–42 Studies report that mean retinal thickness decreases the macular region in aged eyes contained a significant number
by 0.53  µM/year40 and that, in the macula, retinal thickness and of apoptotic cells and these were greatest in the fovea.54 Given
macular volume decrease by around 0.35  µM and 0.01 mm3/ the disparity between studies on age-related changes in RPE cell
year.42 Changes in cell morphology include: nodular excres- density it is not surprising that there is no clear agreement to
cences in rod outer segments43; accumulation of lipofuscin in what extent the RPE-to-photoreceptor ratio changes with age, if
photoreceptor inner segments and the retinal pigment epithe- at all.52,65
lium (RPE)44–46; displacement of nuclei from the outer nuclear Evidence would suggest that age-related cell loss in the retina
layer (ONL)47; and extension of ON-cone bipolar cell and hori- occurs by apoptosis since occasional apoptotic cells are observed
zontal cell processes into the ONL.48 Such evidence of retinal in retinal sections plus the lack of any significant age-related
reorganization and plasticity has also been corroborated by inflammatory response which would occur via necrosis. The
animal studies.49,50 Reorganization of the dendrites could be an stimulus for age-related cell loss is also unclear but since the
majority of cells affected are postmitotic or terminally differenti- demonstrated that autophagy provided a survival mechanism
ated, the accumulation of stochastic damage as occurs with against caspase-dependent death of neonatal RGCs induced by
542 aging in other tissues is plausible. In particular, oxidative axon damage.81
damage is likely to play a significant role since the retina has
high oxygen levels, is exposed to light, and has a number of Diabetic retinopathy
highly metabolically active cell types, making it an ideal envi- It has long been recognized that diabetes leads to a loss of peri-
Section 2

ronment for the generation of reactive oxygen species.66 As cytes and endothelial cells in the retinal vasculature (Fig. 24.5).82,83
already mentioned, the retina undergoes considerable remodel- Development of new techniques to study the vasculature allowed
ing throughout life to adapt to age-related cell loss. In addition, Cogan and colleagues to identify pericyte “ghosts” (intramural
there is likely to be a basal level of limited cellular replacement pockets in the vascular basement membrane lacking normal cell
through resident and bone marrow-derived stem or progenitor contents) as one of the earliest changes in DR.84 Pericyte loss is
cells that have the capacity to differentiate into a number of dif- accompanied by loss of endothelial cells from the retinal vascu-
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

ferent retinal cell types.67 lature, leading to “acellular” capillaries (intact vessel basement
membrane devoid of cells lining the lumen). Cell death in these
RETINAL DAMAGE: DEATH AND REPAIR populations appears to occur predominantly via the intrinsic
apoptotic pathway.85 These characteristic changes have long
Introduction been considered a hallmark of DR. However, studies by Barber
Retinal cell dysfunction and loss are common features of most and others reveal that diabetes is also associated with increased
retinal diseases (e.g., glaucoma, diabetic retinopathy (DR), age- loss of retinal neurons.86,87 STZ-diabetic rat retinas after 30 weeks
related macular degeneration (AMD)) as well as tissue injury of diabetes showed a 22% reduction in the thickness of the inner
(e.g., retinal detachment, light damage). Such cell loss has a plexiform layer, a 10% reduction in RGCs, and a 14% reduction
major negative impact on retinal function and can lead to sig- in the thickness of the INL.88 Clinical studies of diabetics using
nificant visual loss. scanning laser polarimetry and optical coherence tomography
have largely confirmed these findings in patients.89–91 It is also
Glaucoma and ganglion cell loss likely that other neuronal populations such as amacrine cells are
Glaucoma is a heterogeneous group of diseases that leads to also undergoing apoptosis in diabetes.92 The potential initiators
RGC death.68–70 Pathology is associated with “cupping” of the of retinal cell apoptosis are many and include hyperglycemia,
optic disc due to loss of ganglion cell axons. Death of RGCs in oxidative stress, reduced blood flow, ischemia, neuroinflamma-
both postmortem specimens and experimental animal models tion and, specifically in the case of retinal neurons, glutamate
takes place by apoptosis.71–73 Analysis of retinas from human excitotoxicity.85,86
donors suffering primary open-angle glaucoma demonstrated There is surprisingly little information currently available on
greater than 15 times more terminal deoxynucleotidyl transfer- the potential role of autophagy in the pathogenesis of DR,
ase dUTP nick end labeling (TUNEL)-positive apoptotic cells even though there is extensive evidence that: (1) autophagy is
than the controls.74 Interestingly, apoptosis also accounts for the dysregulated in other diabetic tissues93,94 and (2) damaged
selective elimination of about 50% of excess RGCs during devel-
opmental organization of the visual pathway.75 Apoptosis of
RGCs in glaucoma has generally been considered to occur as a
result of mechanical damage due to an increase in intraocular
pressure, but damage to RGCs can also occur in normal-pressure
glaucoma.76 Other insults that have been reported to induce RGC
apoptosis are neurotrophin deprivation, glial activation, isch-
emia, glutamate excitotoxicity, and oxidative stress.70 Confirma-
tory data that RGC apoptosis occurs by the intrinsic pathway
that involves the mitochondria come from backcrossing DBA/2J
mice that exhibit a spontaneous secondary glaucoma with Bax
(one of several BH3 family death proteins) knockout mice that
resulted in a mouse strain in which glaucomatous neurodegen-
eration was reduced.77
Despite the strong association between oxidative stress, mito-
chondrial damage, and RGC death, there have only been limited
studies on the role of autophagy in RGC maintenance and
glaucoma. Rodriguez-Muela and colleagues recently reported
that autophagy promotes survival of RGCs after optic nerve
axotomy in mice.78 Calpain-mediated cleavage of Beclin-1 and
autophagy deregulation have been reported in a rat model of
retinal ischemic injury that recapitulates features of glaucoma.79
Furthermore, blockade of autophagy increased cell death in
cultured RGC, suggesting a prosurvival role of the autophagic
process. By contrast, activation of autophagy in RGCs occurs
Fig. 24.5 A retinal digest showing pericyte dropout and areas of
after optic nerve transection and increased autophagy offers a avascular capillaries in the rat retina (arrows). (Courtesy of Ashay
protective role in cultured RGCs.80 Sternberg and colleagues Bhatwadekar, University of Florida.)
mitochondria are associated with the pathogenesis of DR.85 We Adjacent to active choroidal neovascularization, choriocapillaris
have recently reported that autophagy flux is decreased in the dropout was evident in the absence of RPE atrophy, resulting in
retinal cells of diabetic rats compared to controls.95 a 50% decrease in vascular area. The authors concluded that the 543
The impact of cell death in the diabetic retina and the order in close association observed between degenerating RPE and cho-
which different cell types die during the progression of DR are riocapillaris suggests that, at least in geographic atrophy, RPE

Chapter 24
unclear. For example, pericyte dropout and acellular capillaries atrophy occurs first, followed by choriocapillaris degeneration.
are observed in many diabetic animal models, yet they do not It has largely been considered that photoreceptor cell death
progress to the sight-threatening proliferative stage. Further- occurs as a result of dysfunction or death of the underlying RPE.
more, the duration of diabetes in many patients may be 15 years However, as discussed above in the aging section, there is sig-
or more before clinical abnormalities are observed in the retina, nificant rod photoreceptor loss as a function of age even in the
even though vascular and neuronal cell death will be occurring. presence of an apparently healthy RPE.51 Photoreceptor topog-
A possible explanation for this chronic, rather than acute, attri- raphy in both dry and wet AMD shows preferential loss of rods

Cell Death, Apoptosis, and Autophagy in Retinal Injury


tion in the retina is a low level of cellular replacement from resi- over cones.104 The total number of foveal cones in eyes with large
dent and bone marrow-derived stem or progenitor cells.67 Bone drusen and basal deposits was similar to that of age-matched
marrow-derived progenitor cells have the capacity to differenti- controls and the foveal cone mosaic appeared normal. By con-
ate in a range of retinal vascular and neuronal cell types in trast, cones appeared large and misshapen and few rods
response to retinal injury and in the case of the retinal vascula- remained in the parafovea and, by late-stage AMD, virtually all
ture can repopulate acellular capillaries in rodent models of surviving photoreceptors in the macula were cones.104 In eyes
DR.96,97 However, these bone marrow-derived progenitors are from wet AMD donors, photoreceptors surviving on, or at the
reduced and appear to be dysfunctional in diabetics and thus margins of, disciform scars were largely cones. Subsequent func-
repair potential is attenuated.98,99 tional studies supported the histological evidence for preferen-
tial vulnerability of rods in aging and AMD.105 Although Jackson
Macular degeneration and colleagues concluded from these histological studies that
The challenge in studying retinal cell loss mechanisms in AMD photoreceptor loss is secondary to RPE dysfunction or death in
is to be able to differentiate between the cell loss in the disease AMD, it still remains possible that a photoreceptor abnormality
from that in the normal course of aging. However, reports concur could be the primary effect in AMD and that this leads to sub-
that photoreceptor, RPE, and choroidal cell loss are accelerated sequent loss of the underlying RPE.
in AMD and that this is regional and often focal. Clinically, areas Despite the overwhelming evidence of retinal cell loss in AMD
of geographic atrophy can be observed within the retinal arcades there are surprisingly few reports on the type of cell death, or
in the eyes of patients with dry AMD and these lesions will the initiating insult(s), in human AMD tissue. Most studies have
increase in size with lengthening duration of AMD (Fig. 24.6). relied on cell culture assays and animal models with an AMD
Areas of geographic atrophy show a continuous enlargement phenotype which indicate that cell death occurs primarily by
over time with a median growth rate of between 1 and 13 mm2/ apoptosis. The most detailed report is from Dunaief and col-
year and progression appears to be linear.100,101 The considerable leagues who observed a statistically significant increase in
variability between patients is unexplained but could reflect TUNEL-positive apoptotic cells in the inner choroid, RPE, ONL,
genetic susceptibility, diet, smoking, and light exposure. and INL of AMD donor eyes.106 This increase in the number of
However, there is concordance of disease progression in bilat- apoptotic cells was evident in 5 of 6 eyes with geographic
eral geographic atrophy.102 Loss of RPE is normally associated atrophy and all eyes with exudative AMD. Interestingly, in eyes
with a reduction in the choroidal vasculature. Evidence of the with drusen, only a few TUNEL-positive cells were observed in
degree of choroidal capillary loss and its relationship with RPE each nuclear layer except the RGC layer. From the same study
loss has been elegantly described by McLeod and colleagues.103 TUNEL-positive photoreceptors are also evident over an area of
They observed a linear relationship between the loss of RPE and disorganized RPE near an edge of atrophy. TUNEL-positive RPE
choriocapillaris in geographic atrophy. The vascular area was cells were found most commonly near areas of atrophy and
reduced by 50% in regions of complete RPE atrophy and there occurred more often in AMD eyes than in controls.106 While an
was extreme constriction of remaining viable capillaries. interesting and informative study on cell death in AMD, the

Fig. 24.6 Fundus autofluorescence images


showing geographic atrophy in a 74-year-old
patient. (A) The patient presented with visual
acuity of 20/40 and several areas of retinal
pigment epithelial atrophy. (B) Two years
later the same patient was re-examined:
visual acuity had decreased to 20/200 and
there was extensive geographic atrophy,
including the foveal area. (Courtesy of Erik
Van Kuijk, Ophthalmology and Visual
Sciences, UTMB, Galveston, Texas.)

A B
number of apoptotic cells seems high, especially in the neural still evident at 28 days postdetachment. However, there is some
retina, for a slowly progressive condition. By contrast, Xu et al. debate whether apoptosis following retinal detachment occurs
544 only observed apoptotic cells in the retina of 4 of 16 AMD donor via the intrinsic or extrinsic pathways.124,128,129 Interestingly,
eyes and the overall numbers of apoptotic cells appeared rela- retina RPE separation in rats causes a Fas-dependent activation
tively low.107 Nordgaard and colleagues undertook proteomic of autophagy in injured photoreceptors and, if autophagy is
analysis of the RPE from donor eyes at progressive stages of inhibited, the time course and number of apoptotic cells are
Section 2

AMD.108 Several components of apoptotic signaling pathways accelerated.130 Thus it appears that autophagy is activated to
(αA crystallin, VDAC1, HSP70, GST-π) demonstrated changes in regulate the level of receptor apoptosis.
expression early in AMD or changed linearly with AMD pro-
gression. Surprisingly, information on death mechanisms in the Retinal dystrophies
AMD is scarce but there is evidence from cell culture and animal Retinal dystrophies encompass a heterogeneous group of inher-
models for both intrinsic and extrinsic apoptosis pathways. FAS- ited conditions, with more than 100 genes or loci identified so
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

mediated apoptosis has been observed in retinal cells, which far.131 The most common subtype is retinitis pigmentosa (RP),
could explain the role of inflammation in AMD,109 and a multi- which is characterized by progressive death of retinal rod and
tude of studies have reported oxidative stress-induced apoptosis cone photoreceptors, and the disease proceeds toward reduction
by the classic mitochondrial route.110–113 The recent finding of Alu of peripheral field with tunnel vision and finally loss of sight.131,132
RNA accumulation leading to RPE cytotoxicity in geographic One of the major impediments in the comprehensive identifica-
atrophy and Dicer1 downregulation suggests a completely new tion of the degenerative mechanisms of retinal dystrophies is the
mechanism of RPE degeneration.114 It should also be considered heterogeneity of the disease and involvement of multiple causal
that cellular repair, cellular replacement, and damage control are genes in the pathogenesis of the disease.133 The mode of rod cell
critical in retinal homeostasis and a declining antioxidant system death in several animal models of RP suggests death by apop-
together with increased oxidative damage will play a major etio- tosis, which is in agreement with findings in RP retinas from
logical role in AMD, and that once the threshold for damage is donor eyes134 (reviewed by Travis135). Cone cells usually die as a
reached, multiple cellular processes for repair and regeneration secondary response to rod cell death, possibly because they
will be impacted.115–118 depend on rod-secreted neurotrophic factors for survival.57,60
There is increasing evidence that autophagy flux may be Studies of apoptotic mechanisms in the photoreceptors of RP
dysregulated in the RPE in AMD119,120 and is likely due to models imply the involvement of caspase-dependent as well as
multiple factors that affect the initiation of autophagy and/or independent pathways.136–138 DNA fragmentation was a regular
the fusion of autophagosomes with lysosomes. These will feature encountered in the mouse models, indicating that pho-
include lipofuscin accumulation, susceptibility to oxidative toreceptors in mouse models die of apoptosis. Administration of
stress, mitochondrial damage, and lysosomal dysregulation, all caspase-3 inhibitors inhibited photoreceptor apoptosis in the
of which have a strong association with AMD. To what extent tubby mouse (Usher syndrome model).139 Caspase-independent
changes in autophagy flux reflect alterations in the formation modes of apoptosis may involve calpain and calcium ion excess
or elimination of autophagosomes and are a cause or conse- in RP.140 In recent years it has been suggested that, although the
quence of AMD remains to be determined. Lipid peroxidation primary cell death mode will be apoptosis, other modes of cell
products reduce autophagy flux and increase lipofuscin accu- removal could be involved, including autophagy and
mulation in cultured RPE cells.121 An increase in lysosomal pH complement-mediated lysis.141 Investigation for different cell
which is associated with the lipofuscin constituent A2E122 may death pathways in three independent mouse models of photo-
impair autophagosome–lysosome fusion, as may the accumula- receptor degeneration – the rd/rd mouse, the rds/rds mouse
tion of lipofuscin granules within the lysosomal vacuome. It and the light-damage model in albino mice – shows that, apart
has been reported that drusen formation may reflect an increase from apoptotic cell death, several oxidative stress markers as
in both mitochondrial damage and autophagy in the RPE.123 well as elements of the autophagic and complement pathways
The researchers speculated that increased autophagy and the are upregulated. While the induction of oxidative stress response
release of intracellular proteins via exosomes by the aged RPE genes is early, the induction of autophagy was only seen in
may contribute to the formation of drusen. It is important to damaged retinas when compared to controls, which made the
note that there is substantial cross-talk between autophagy and authors conclude that autophagy specifically removes damaged
proteasomal degradation pathways which may also affect the photoreceptors from the retina.141 However, the data may also
status of the RPE.119 be interpreted as an attempt in the damaged retina to salvage
the photoreceptors from initial stress which, when overwhelm-
Retinal detachment ing, gives rise to autophagic death. Finally, the evidence of
Retinal detachment resulting from full-thickness retinal breaks, upregulation of high-mobility group box 1 (HMGB1) protein in
subretinal exudation, and/or vitreoretinal traction is a common human eyes with retinal detachment suggests that necrosis
cause of photoreceptor loss.124 Cell death is highly dependent on could also be a mode of photoreceptor death.142 It could well be
the area and duration of the detachment. Analysis of tissue this mode of cell death that accounts for some of the caspase-
samples from patients with retinal detachment showed the pres- independent photoreceptor death pathways that have hitherto
ence of significant numbers of apoptotic cells by 24 hours, which been thought to be apoptosis. However the relevance of necrosis
peaked by 2 days and dropped to a low level by 7 days after in early stages of RP still needs to be investigated. Nevertheless,
detachment.125 These observations have been largely supported the heterogeneity of RP and similar retinal dystrophies necessi-
by experimental retinal detachment in cats and rats, both of tates the understanding of the earliest mechanisms of disease
which show a peak of apoptosis between 1 and 3 days post­ inception such that customized treatments may be catered
detachment, which then declines.126,127 Some apoptotic cells were according to the nature of disease pathology in a patient.
Attempts to block cell death by one strategy may prove to be considered the most significant fluorophores implicated in RPE
futile as the protective effect may only be successful for a short damage. Irradiated lipofuscin granules or their constituents can
duration, after which the cell might proceed through another generate high levels of reactive oxygen species.168–170 Several 545
death mode. models of retinal degeneration in vitro and in vivo demonstrate
the role of calcium in inducing cell death by activating degrada-
Light damage

Chapter 24
tive proteases such as calpain.171,172 Adaptor protein 1 (AP-1)
The retina is vulnerable to damage from ultraviolet radiation, signal transduction has also been implicated in bright light-
visible light (400–700  nm), and infrared radiation.66,143,144 The induced photoreceptor death where there is an increase of AP-1
extent and type of damage are highly dependent upon the gene expression.173 Light can damage not only photoreceptor
wavelength, power, duration, area of coverage, cumulative cells and RPE but also RGCs.174 Photosensitive RGCs contain
exposure, and location (e.g., macular versus periphery). Light- melanopsin, suggesting that light damage could impact a spe-
induced retinal damage can occur via at least one of three cific subset of RGCs and contribute to the pathogenesis of glau-

Cell Death, Apoptosis, and Autophagy in Retinal Injury


mechanisms: (1) mechanical (high irradiances of short duration coma.175,176 The role of autophagy in retinal light damage has
that cause sonic shock waves disrupting the tissue irreversibly); only received limited attention but Kunchithapautham and col-
(2) photothermal (when incident energy is trapped, causing a leagues have shown that upregulation of autophagy can protect
rise of temperature 10°C or more); and (3) photochemical against light damage and oxidative stress in the retina.177,178
(shorter-wavelength visible light is absorbed and dissipated
away, causing molecular alterations).66 Retinal photodamage is THERAPEUTIC OPTIONS
highly dependent on the presence of chromophores, of which
the most obvious are the visual pigments, and in most species Neuroprotection
prolonged intense light exposure will lead to significant pho- Neuroprotection offers a promising approach for preventing or
toreceptor cell damage.145 However, other important chromo- slowing retinal cell loss. Neuroprotection is essentially defined
phores include hemoglobin, melanin, lipofuscin, macular as the use of therapeutic agents to prevent, hinder, and in some
pigment, and flavins.66 Photochemical damage has been the most instances, reverse neuronal cell death whatever the primary
extensively studied form of light damage because it can cause injury.179–181 The strategies for neuroprotection vary considerably
retinal damage within the intensity range of ambient visible dependent upon the target cell, the nature of the factors initiat-
light.66,146 There are two well-defined types of retinal light ing cell loss, and the stage of disease. Clearly, the best time for
damage. Class 1 damage has an action spectrum that is identical neuroprotection is early in the time course of the disease before
to the absorption spectrum of the visual pigment and the initial any significant cell death has occurred. Three broad approaches
damage is in the photoreceptors.66,146,147 Class 2 damage has an have evolved: (1) blocking the pathways involved in the cellular
action spectrum that peaks at shorter wavelengths, is generally damage (e.g., antioxidants); (2) inhibiting the cell death path
confined to the RPE, and is often referred to as the blue light directly (e.g., upregulation of Bcl-2 and inhibition of caspases);
hazard.66,146,148 and (3) treatment with neurotrophic factors that suppress the
Most mechanistic studies on retinal photochemical damage intrinsic apoptosis pathway (e.g., brain-derived neurotrophic
have been undertaken in experimental animal models and all factor (BDNF), glial-derived neurotrophic factor (GDNF),
show that cell death occurs via apoptosis. Excess light has been pigment epithelial-derived factor (PEDF)). A summary of poten-
shown to trigger apoptosis in rat retinas.149,150 Low-intensity light tial neuroprotective agents, their retinal cell targets, and their
exposure for long durations as well as intense light exposure proposed mechanisms of action is shown in Table 24.1.
(3000 lux) for short durations up to 2 hours can cause severe Light damage in rodents, which results in extensive photore-
photoreceptor degeneration by apoptosis followed by RPE ceptor and RPE cell loss, has been extensively studied in the
degeneration.151,152 Both types of retinal photodamage, induced search for effective neuroprotective agents.143 Light induction of
by exposure to either low- or high-intensity light, are dependent photoreceptor apoptosis is dependent on the availability of 11-cis
on the presence of, and regeneration of, rhodopsin in the retinal for rhodopsin regeneration.182 Acute bright light-induced
retina.153–157 The extent of photodamage is correlated to the photoreceptor apoptosis involves activation of nitric oxide
amount of rhodopsin present in the retina prior to the light synthase (NOS) and generation of nitric oxide (NO), increased
damage,158,159 although direct evidence of rhodopsin causing intracellular calcium, oxidative damage, and alterations in mito-
damage is absent. Several rhodopsin intermediates, especially chondrial function.143,183 Inhibition of NOS activity by LNAME183,184
all-trans-retinal accumulation in the photoreceptor membrane or application of the calcium channel blockers D-diltiazem, nilva-
due to decreased reduction by retinol dehydrogenase, have been dipine, or nicardipine167 protected photoreceptor cells following
implicated as mediators of photo-oxidation.160 Decrease of intra- cell death in light-damaged retinas. Inhibition of the apoptotic
cellular calcium levels during the phototransduction cycle has cascade by reduction of the proapoptotic Bcl-2 family members
also been suggested to be an aspect of rhodopsin-mediated Bax and Bak also protects the retina against light damage.185 The
injury causing photoreceptor apoptosis.161 Light-induced photo- use of antioxidants (that are reactive oxygen species quenchers)
receptor apoptosis can either be caspase-mediated162–166 or like dimethylthiourea and N-phenyl-2-naphthylamine and the
caspase-independent.167 The most probable reason for difference administration of exogenous neurotrophic factors such as BDNF,
of opinion in mechanisms like caspase involvement and signal ciliary neurotrophic factor (CNTF), basic fibroblast growth factor
transduction in photoreceptor death is that cellular response will (bFGF), erythropoietin (Epo), and PEDF have all been shown to
vary according to the lighting conditions and, in reality, all of be protective against light damage (reviewed by Wenzel et al.143).
the discovered pathways could exist to decide on cell fate bFGF plays an important role in the endogenous defense against
depending on the prevalence and type of light damage during stress of retinal cells. Preconditioning with light186,187 or ischemic
an individual’s lifetime. Melanosomes and lipofuscin are preconditioning188,189 increases the expression of bFGF and
Table 24.1 A summary of potential neuroprotective agents, their cascade to address as the cause of the dystrophy and associated
retinal cell targets, and their proposed mechanisms of action cell death will remain unless also treated. Photoreceptors are the
546 target cells as these are the predominant cell type lost in the
Mechanistic
retinal dystrophies. Studies in animal models with inherited
Target cell target Therapeutic approaches
retinal diseases have used similar strategies to those described
above for light damage. While considerable success has been
Section 2

Retinal ganglion Excitotoxicity Memantine


cells achieved, this has not translated to clinical care and gene therapy
to reverse the mutation, as in Leber’s congenital amaurosis, is
Oxidative Vitamin E
stress
perhaps a preferable option.194 Both PEDF and CNTF have
shown considerable promise in slowing photoreceptor degen-
Mitochondrial Coenzyme Q10 eration, whether provided by intraocular or gene transfer, in rd1,
damage rd2, rds P216L, rhodopsin S334ter and P23H rodent models of
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

inherited degeneration.195–200 CNTF, released by engineered cells,


Neurotrophin BDNF, gene therapy
derivation slowed retinal degeneration in the rcd1 dog model201 and, after
intravitreal injection, in an autosomal dominant feline model of
Photoreceptors Apoptosis Survival and growth rod–cone dystrophy.202 Neurotrophin-3 can induce upregulation
factors (e.g., BDNF, of bFGF, and thereby neuroprotection, by activation of TrkC in
GDNF)
Müller cells.203 Viral delivery of a BDNF transgene delayed
RPE Apoptosis Survival and growth degeneration induced by the Q344ter mutation in rhodopsin204
factors and prolonged release from transgenic cell transplants into the
eye slowed degeneration in RCS rats.205 Capsase-3 inhibition is
Autophagy Rapamycin
protective in the rd1 degeneration by delaying cell death172,206
Oxidative Antioxidants, lutein, and preservation of retinal morphology.207 Caspase-3 inhibitors
damage zeazanthin, zinc also provided protection in two other models of inherited retinal
degeneration: the S334ter rat137 and the tubby mouse.139 Alterna-
Mitochondrial Antioxidants, resveratrol tive approaches which have reduced photoreceptor loss and
damage
slowed the progression of retinal degeneration in animal models
Lipofuscin Reducing the retinoid include: (1) overexpression of the X-linked inhibitor of apoptosis
system cycle using gene therapy208,209; (2) sustained intravitreal delivery of
Glutathione-S-transferase fluocinolone acetonide, which suppresses microglial activation
Lysosomal Elevated pH (A2A
and inflammation210; (3) GDNF either given by subretinal injec-
system adenosine receptor tion or conjugated to nanoparticles211,212; and (4) neuroprotectin
agonist) D1 (NPD1).110
Rodent and dog models of glaucoma have shown that agents
Bruch’s Subthreshold laser, MMP
membrane upregulation, microbial
that block glutamate excitoxicity, prevent mitochondrial dys-
enzymes function, reduce oxidative stress, or enhance neurotrophic
factors all decrease or prevent the loss of RGCs (Table 24.1).213,214
Choroid Endothelial Growth factors (e.g., The N-methyl-D-aspartate receptor antagonist was shown to be
cell loss VEGF)
a highly effective neuroprotective agent in animal models of
Inflammation Alternative Complement pathway RGC death (reviewed by Cheung et al.213 and Danesh-Meyer214).
complement inhibitors However, the outcome of clinical trials, although showing a
pathway trend for improvement, was inconclusive. An alternative agent
BDNF, brain-derived neurotrophic factor; GDNF, glial-derived neurotrophic factor; is brimonidone tartrate, which is a highly selective alpha2-
RPE, retinal pigment epithelium; MMP, matrix metalloproteinase; VEGF, vascular adrenergic agonist that increases RGC survival in animal
endothelial growth factor.
models.215 Coenzyme Q10, which is an essential cofactor of the
electron transport chain, has been shown to have some neuro-
protective effect on RGCs based on its antioxidant properties.216
protects retinal cells against damaging doses of light or increased Treatment with exogenous neurotrophins such as BNDF, nerve
intraocular pressure. The neuroprotective effect could be growth factor, and CNTF have all been shown to slow the loss
enhanced by a combination of bFGF and PEDF.190 In contrast, no of RGCs but not to inhibit RGC death in the long term. To
protection resulted against constant light exposure for 1 week improve outcomes, strategies that researchers have attempted
when bFGF was expressed locally from a transgene delivered by include sustained release of neurotrophic factors via either gene
adeno-associated virus vector191 or simian immunodeficiency therapy217 or encapsulated cell technology, which allows the
virus vector.192 However, intravitreal injection of recombinant intravitreal implantation of a chamber containing live cells pro-
BDNF protein or its release from transgenic cell transplants193 grammed to release CNTF.218 More direct antiapoptotic strate-
protects the retina against 1–2 weeks of constant light gies have included activation of the Bcl-2 pathways using
exposure. cilostazol or 5-S-GAD which increases RGC survival in animal
Inhibition of apoptotic pathways in a light-damaged neuron models.219,220
is relatively straightforward as the cell is otherwise healthy. RPE cell loss is a hallmark feature of dry AMD. In vitro
However, in retinal cells harboring mutations, as is the case in studies have identified a plethora of agents capable of either
retinal dystrophies, the cell has more than just the apoptotic directly inhibiting apoptosis with, for example, caspase
inhibitors, or indirectly by neutralizing the initiating factors specificity of compounds that activate autophagy in an mTOR-
leading to cell death. Macular carotenoids, NPD1, eEpo, res- independent fashion have yet to be established.239,240 Gene
veratrol, and PEDF have all been shown to confer protection therapy is also a possibility and overexpression of Atg7 can 547
against oxidative stress-induced apoptosis.113,221–227 αB crystallin protect against anoxia/reoxygenation injury.242
is apically secreted within exosomes by polarized human RPE Given the realization of the importance of autophagy in

Chapter 24
and provides neuroprotection to adjacent cells.228 An alterna- cancer and neurodegenerative diseases there has been an
tive approach has been to upregulate antioxidants or molecular extensive effort to identify potential therapeutic regulators of
chaperones to cope with the increased oxidative stress.112,229 autophagy. Those under development include HDAC inhibi-
Neurotrophic protection of the RPE has been more difficult tors, mTOR inhibitors, BH3 domain mimetics, glycolytic
to model in vivo as there are no reliable models of geographic inhibitors, inositol-lowering agents, and protein kinase inhibi-
atrophy. However, the Age-Related Eye Disease Study dem- tors.241,243–245 Of particular interest in cancer has been the use
onstrated that oral supplementation with high levels of anti- of the antimalarial drug hydroxychloroquine (HCQ) which

Cell Death, Apoptosis, and Autophagy in Retinal Injury


oxidants and zinc can reduce the risk of disease progression serves to inhibit autophagy by perturbing lysosomal func-
to advanced AMD.230 tion.243,244 HCQ is now being assessed in combination with a
The strategies described above are likely to be equally effective number of chemotherapy agents in phase I and II clinical
in other retinal conditions resulting in cell death. Imai and col- trials.243,244 However, it has long been recognized that HCQ
leagues emphasize the importance of exogenous neurotrophic can cause ocular toxicity, with the most serious being an irre-
factors in reducing cell loss in DR231 while BDNF, bFGF, and versible retinopathy. The dosage parameters associated with
inhibition of the FAS receptor impede apoptosis following retinopathy are still uncertain but it has been suggested that,
retinal detachment.127,128 for doses of HCQ less than 6.5  mg/kg, the incidence of reti-
nopathy is minimal.246 Nevertheless, some clinical trials are
Modulating autophagy testing dosages that exceed these levels. Furthermore, shutting
As discussed earlier, autophagy is dysregulated in a number of down lysosomal functions, whether it be autophagy or endo-
retinal conditions. However, dependent on the type and stage cytosis, can dramatically alter cellular homeostasis and defense.
of the disease, this dysregulation could reflect as decreased or Proteins other than the autophagic pathway proteins have
increased autophagy. A similar trend is also observed in cancer been implicated in autophagy. Caspases and calpains play key
and neurodegenerative diseases. That is, autophagy plays a role roles in cleavage and activation or inactivation of autophagy
in tumor suppression or oncogenesis but can desensitize cells to proteins (summarized by Kaminskyy and Zhivotovsky247).
cytotoxic agents.232 Thus, while enhancing autophagy could Cross-talk between the autophagy and apoptosis pathways is
prevent tumor progression it could also reduce sensitivity to regulated by caspase cleavage of Beclin-1248 and also by p62/
chemotherapeutic agents. Similarly, upregulation of autophagy Sqstm1-Keap1 signaling.249 Similarly, modulation of the Sirt-1-
can protect against Huntington’s disease in mouse models Foxo pathway also modulates autophagy.250,251 Modulating reac-
whereas enhancing autophagosome formation in Alzheimer’s tive oxygen species levels with antioxidant compounds such as
disease can exacerbate the accumulation of Aβ.232 Therefore any resveratrol and curcumin or overexpressing antioxidant enzymes
therapy must ensure the correct balance of autophagy flux is can negatively modulate autophagy. Quenching reactive oxygen
attained for the targeted disease or cell type. species will decrease mitochondrial damage and autophagy ini-
Many autophagy-related proteins and signaling molecules tiation. In addition, a lowering of reactive oxygen species will
have been implicated in a number of events of autophagy, preserve the activity of lysosomal enzymes (reviewed by Scherz-
including signaling, sequestration, maturation, and degradation. Shouval and Elazar252). It is important to keep in mind that, when
The primary regulation of autophagy proceeds by mTor, GCN2, one type of autophagy is altered, the other types will also be
and PI3K-III signaling pathways. Autophagy is negatively regu- affected. Chronic blockage of CMA promotes upregulation of
lated by mTor and Bcl-2/Bclxl and positively regulated by PI3K macroautophagy,253 whereas acute blockage leads to macro­
class III and GCN2/eIF2α. Compounds that inhibit mTor phos- autophagic dysregulation.254 Cells respond to blockage of
phorylation (rapamycin) or Bcl-2 interaction with Beclin-1 macroautophagy by increasing CMA.254 Acute blockage of the
(ABT737) promote autophagy.233,234 PI3K-III can be activated by proteasome upregulates macroautophagy,255 whereas chronic
inhibiting the interactions between Bcl-2 and Beclin/Atg6 or by blockage leads to macroautophagic dysregulation.256 Some sub-
overexpressing Beclin-1 or Beclin-1-BD (Bcl-2 binding defective units of the proteasome are degraded by CMA,257 which may
mutant) and suppression by Vps34 inhibitors, 3-methyladenine, explain why blockage of CMA is associated with proteasome
or wortmannin.235,236 A number of mTOR-dependent and mTOR- dysregulation.258 Interactions of microautophagy with other
independent agents have been identified that stimulate autoph- proteolytic systems remain undiscovered.
agy. Rapamycin is a well-established compound for inducing
autophagy and attenuating neuronal cell death in a number of Cellular replacement
in vitro and in vivo experimental models. Other stimulators of In order to maintain optimal tissue and organ function, dead
autophagy include lithium and trehalose which enhance auto­ cells need to be replaced. Amphibians and fish exhibit robust
phagy via an mTOR-independent mechanism.237,238 Unfortu- retinal regeneration which, unfortunately, is not retained in
nately, despite their potency, rapamycin and lithium have mammals. It has long been known that amphibians can regener-
significant side-effects which lessen enthusiasm for their clinical ate a completely new and functional retina from the adjacent
use in chronic neurodegenerative diseases.14,239,240 In an attempt RPE, which restores vision.259,260 Retinal regeneration in fish is
to address this researchers are screening for small-molecule not from the RPE and instead occurs from an intrinsic progenitor
enhancers of rapamycin (SMERs) and small molecule inhibitors that is derived from the Müller glia cells.261,262 Birds show some
of rapamycin (SMIRs) with less cytotoxicity.241 The efficacy and limited replacement of lost neurons which involves a mitotic
subpopulation of Müller glia.263 Cellular replacement in the CONCLUSIONS
retina of mammals is very minimal, even in response to injury.260
548 A number of endogenous stem/progenitor populations have As has been discussed in detail above, cell death is a major
been reported but many, such as retinal stem cells in the ciliary feature of retinal injury and disease. Preventing this cell loss or
margin zone, remain controversial.260 Studies in rodents have replacing the lost cells is now becoming a realistic option with a
found evidence of progenitor gene expression in Müller glia number of pharmacological agents in clinical trials and Food and
Section 2

after retinal damage but there is no definitive evidence that Drug Administration approval for cell replacement therapies.
they produce differentiated and functional neurons. However, The last 20 years have seen an exponential increase in our knowl-
Ooto et  al.264 did report that these “progenitor-like” Müller cells edge of cell death pathways in the retina and the identification
can express markers for bipolar cells and photoreceptors after of targets for therapeutic intervention. However, while consider-
injury. An alternative source of reparative cells could be derived able improvement has been observed in animal models of retinal
from the bone marrow. Using chimeric mice transplanted with cell loss, translation into the clinic has, to date, only shown
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

bone marrow cells expressing green fluorescent protein, it was modest success. It is likely that different approaches will be
shown that bone marrow-derived cells would home to the site required for different retinal conditions as preserving a cell with
of retinal injury and had the potential to differentiate into astro- a debilitating genetic mutation is likely to be detrimental to the
cytes, macrophages/microglia, endothelial cells, pericytes, and retina, while preventing apoptosis of normal cells following
RPE.96,97 However, recruitment and integration occurred at a retinal detachment or light damage would be beneficial. Simi-
very low level. To overcome this, a recent study infected bone larly, autophagy has been shown to play a protective role in a
marrow-derived cells ex vivo with lentiviral vector expressing number of retinal diseases but the balance is critical as excess
the RPE-specific gene RPE65 and injected these cells into the autophagy will lead to removal of essential organelles and loss
circulation of mice in which the RPE had been destroyed by of cell function while too little autophagy will lead to the buildup
sodium iodate.265 These transplanted cells homed to the neural of damaged organelles. A further problem is that all these thera-
retina Bruch’s membrane interface in large numbers and showed peutic approaches will remain limited if the initiating factors
restoration of a functional RPE layer, with typical RPE pheno- resulting in cell loss are not also addressed. Finally, we have to
type, including coexpression of another RPE-specific marker, address the clinical limitation that intervention is often not until
CRALBP, and photoreceptor outer-segment phagocytosis. Most the late stage of disease when extensive cell death has taken
importantly, retinal degeneration was prevented and visual place and thus we need to consider strategies for treating much
function was restored to levels similar to those found in normal earlier if we are to prevent significant retinal cell loss. Despite
animals.265 these hurdles our ever-increasing understanding of retinal
An alternative approach, with greater clinical application, pathogenesis and cell death together with improved pharmaco-
has been the transplantation of a variety of stem/progenitor logical screening for novel therapeutic agents will almost cer-
cell types. Most focus has been on the RPE as it can be readily tainly result in major advances in clinical treatment over the
generated from embryonic stem cells (ESC) or induced plu- next decade.
ripotent stem (iPS) cells.266–269 These cells have been successfully
REFERENCES
transplanted into a number of animal models of retinal degen- 1. Kroemer G, Galluzzi L, Vandenabeele P, et al. Classification of cell death:
eration and demonstrated recovery of vision, and ESC-derived recommendations of the Nomenclature Committee on Cell Death 2009.
Cell Death Differ 2009;16:3–11.
RPE is now in phase I/II clinical trials. While a promising 2. Green DR. Means to an end: apoptosis and other cell death mechanisms.
strategy, there remain concerns that: (1) ESC may be prone to New York: Cold Spring Harbor Laboratory Press; 2011.
teratoma formation270; (2) iPS cells contain protein-coding point 3. Green DR, Reed JC. Apoptosis: physiology and pathology. Cambridge:
Cambridge University Press; 2011.
mutations271; (3) success with RPE transplantation in humans 4. Pereira WO, Amarante-Mendes GP. Apoptosis: a programme of cell death or
has been modest272,273; and (4) transplantation is normally into cell disposal? Scand J Immunol 2011;73:401–7.
5. Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition.
a severely degenerated retina with late-stage disease.272,273 Trends Biochem Sci 2007;32:37–43.
Despite these limitations, considerable progress has been made 6. Hitomi J, Christofferson DE, Ng A, et al. Identification of a molecular
signaling network that regulates a cellular necrotic cell death pathway.
in the field of RPE replacement in the last decade. Repair of Cell 2008;135:1311–23.
the neural retina is more complicated due to the need to form 7. Zhivotovsky B, Orrenius S. Cell death mechanisms: cross-talk and role in
the requisite neural connections to perform vision. The most disease. Exp Cell Res 2010;316:1374–83.
8. Yousefi S, et al. Calpain-mediated cleavage of Atg5 switches autophagy to
cited study is that of MacLaren and colleagues, who showed apoptosis. Nat Cell Biol 2006;8:1124–32.
that donor cells can integrate into the adult or degenerating 9. Chen Y, Klionsky DJ. The regulation of autophagy – unanswered questions.
J Cell Sci 2011;124:161–70.
mouse retina provided the donor cells are derived from the 10. Hubbard VM, Valdor R, Macian F, et al. Selective autophagy in the mainten­
developing retina at a time coincident with the peak of rod ance of cellular homeostasis in aging organisms. Biogerontology 2011 Apr 3.
[Epub ahead of print].
genesis.274 Importantly, these transplanted cells integrated and 11. Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases.
differentiated into functional rod photoreceptors that formed Nat Neurosci 2010;13:805–11.
12. Arias E, Cuervo AM. Chaperone-mediated autophagy in protein quality
synaptic connections, and improved visual function in the host control. Curr Opin Cell Biol 2011;23:184–9.
animals. Human ESC or iPS have been shown, under the right 13. Mijaljica D, Prescott M, Devenish RJ. Microautophagy in mammalian cells:
conditions, to be able to differentiate into rods and cones275–278 Revisiting a 40-year-old conundrum. Autophagy 2011;7:673–82.
14. Mizushima N, Levine B, Cuervo AM, et al. Autophagy fights disease through
and when transplanted into the adult mouse retina can dif- cellular self-digestion. Nature 2008;451:1069–75.
ferentiate into photoreceptors and restore light responses in 15. Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and
signaling regulation. Curr Opin Cell Biol 2010;22:124–31.
Crx-deficient mice.279 Since it is not the intention of this chapter 16. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol
to provide a detailed review of regenerative medicine for retinal 2011;12:9–14.
17. Hailey DW, Rambold AS, Satpute-Krishnan P, et al. Mitochondria supply
repair, the reader is pointed to the following reviews and membranes for autophagosome biogenesis during starvation. Cell 2010;141:
Chapter 35, Stem cells and cellular therapy.260,275,280–283 656–67.
18. Ravikumar B, Moreau K, Jahreiss L, et al. Plasma membrane contributes to 52. Gao H, Hollyfield JG. Aging of the human retina. Differential loss of neurons
the formation of pre-autophagosomal structures. Nat Cell Biol 2010;12: and retinal pigment epithelial cells. Invest Ophthalmol Vis Sci 1992;33:1–17.
747–57. 53. Curcio CA, Owsley C, Jackson GR. Spare the rods, save the cones in aging
19. Ravikumar B, Moreau K, Rubinsztein DC. Plasma membrane helps autopha- and age-related maculopathy. Invest Ophthalmol Vis Sci 2000;41:2015–8. 549
gosomes grow. Autophagy 2010;6:1184–6. 54. Sanyal S, Hawkins RK, Jansen HG, et al. Compensatory synaptic growth in
20. Dunn Jr WA. Studies on the mechanisms of autophagy: formation of the the rod terminals as a sequel to partial photoreceptor cell loss in the retina of
autophagic vacuole. J Cell Biol 1990;110:1923–33. chimaeric mice. Development 1992;114:797–803.

Chapter 24
21. Nobukuni T, Joaquin M, Roccio M, et al. Amino acids mediate mTOR/raptor 55. Jansen HG, Sanyal S. Synaptic plasticity in the rod terminals after partial
signaling through activation of class 3 phosphatidylinositol 3OH-kinase. photoreceptor cell loss in the heterozygous rds mutant mouse. J Comp Neurol
Proc Natl Acad Sci U S A 2005;102:14238–43. 1992;316:117–25.
22. Ravikumar B, Vacher C, Berger Z, et al. Inhibition of mTOR induces autoph- 56. Spear PD. Neural bases of visual deficits during aging. Vision Res 1993;33:
agy and reduces toxicity of polyglutamine expansions in fly and mouse 2589–609.
models of Huntington disease. Nat Genet 2004;36:585–95. 57. Leveillard T, Mohand-Said S, Fintz AC, et al. The search for rod-dependent
23. Fimia GM, Di Bartolomeo S, Piacentini M, et al. Unleashing the Ambra1-Beclin cone viability factors, secreted factors promoting cone viability. Novartis
1 complex from dynein chains: Ulk1 sets Ambra1 free to induce autophagy. Found Symp 2004;255:117–27; discussion 127–30, 177–8.
Autophagy 2011;7:115–7. 58. Chalmel F, Leveillard T, Jaillard C, et al. Rod-derived cone viability factor-2
24. Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like conjugation systems is a novel bifunctional-thioredoxin-like protein with therapeutic potential.

Cell Death, Apoptosis, and Autophagy in Retinal Injury


in macroautophagy. “Protein modifications: beyond the usual suspects” BMC Mol Biol 2007;8:74.
review series. EMBO Rep 2008;9:859–64. 59. Fridlich R, Delalande F, Jaillard C, et al. The thioredoxin-like protein rod-
25. Hanada T, Noda NN, Satomi Y, et al. The Atg12-Atg5 conjugate has a novel derived cone viability factor (RdCVFL) interacts with TAU and inhibits its
E3-like activity for protein lipidation in autophagy. J Biol Chem 2007;282: phosphorylation in the retina. Mol Cell Proteomics 2009;8:1206–18.
37298–302. 60. Yang Y, Mohand-Said S, Danan A, et al. Functional cone rescue by RdCVF
26. Fujita N, Itoh T, Omori H, et al. The Atg16L complex specifies the site of LC3 protein in a dominant model of retinitis pigmentosa. Mol Ther 2009;
lipidation for membrane biogenesis in autophagy. Mol Biol Cell 2008;19: 17:787–95.
2092–100. 61. Feuer WJ, Budenz DL, Anderson DR, et al. Topographic differences in the
27. Weidberg H, Shvets E, Shpilka T, et al. LC3 and GATE-16/GABARAP sub- age-related changes in the retinal nerve fiber layer of normal eyes measured
families are both essential yet act differently in autophagosome biogenesis. by Stratus optical coherence tomography. J Glaucoma 2011;20:133–8.
EMBO J 2010;29:1792–802. 62. Aggarwal P, Nag TC, Wadhwa S. Age-related decrease in rod bipolar cell
28. Weidberg H, Shpilka T, Shvets E, et al. LC3 and GATE-16 N termini mediate density of the human retina: an immunohistochemical study. J Biosci
membrane fusion processes required for autophagosome biogenesis. Dev Cell 2007;32:293–8.
2011;20:444–54. 63. Lei Y, Garrahan N, Hermann B, et al. Transretinal degeneration in ageing
29. Saftig P. Physiology of the lysosome. In: Mehta A, Beck M, Sunder-Plassmann human retina: a multiphoton microscopy analysis. Br J Ophthalmol
G, editors. Fabry disease: perspectives from 5 years of FOS. Oxford: Oxford 2011;95:727–30.
PharmaGenesis; 2006. Chapter 3. 64. Panda-Jonas S, Jonas JB, Jakobczyk-Zmija M. Retinal pigment epithelial cell
30. Mortimore GE, Poso AR. Intracellular protein catabolism and its control count, distribution, and correlations in normal human eyes. Am J Ophthalmol
during nutrient deprivation and supply. Annu Rev Nutr 1987;7:539–64. 1996;121:181–9.
31. Scherz-Shouval R, Shvets E, Fass E, et al. Reactive oxygen species are essential 65. Dorey CK, Wu G, Ebenstein D, et al. Cell loss in the aging retina. Relationship
for autophagy and specifically regulate the activity of Atg4. EMBO J to lipofuscin accumulation and macular degeneration. Invest Ophthalmol Vis
2007;26:1749–60. Sci 1989;30:1691–9.
32. Komatsu M, Kurokawa H, Waguri S, et al. The selective autophagy substrate 66. Boulton M, Rozanowska M, Rozanowski B. Retinal photodamage. J Photochem
p62 activates the stress responsive transcription factor Nrf2 through inactiva- Photobiol B 2001;64:144–61.
tion of Keap1. Nat Cell Biol 2010;12:213–23. 67. Boulton M, Albon J. Stem cells in the eye. Int J Biochem Cell Biol 2004;
33. Lee JY, Koga H, Kawaguchi Y, et al. HDAC6 controls autophagosome matura- 36:643–57.
tion essential for ubiquitin-selective quality-control autophagy. EMBO J 68. Agarwal R, Gupta SK, Agarwal P, et al. Current concepts in the pathophysiol-
2010;29:969–80. ogy of glaucoma. Ind J Ophthalmol 2009;57:257–66.
34. Shpilka T, Elazar Z. Shedding light on mammalian microautophagy. Dev Cell 69. Kisiswa L, Dervan AG, Albon J, et al. Retinal ganglion cell death postponed:
2011;20:1–2. giving apoptosis a break? Ophthalmic Res 2010;43:61–78.
35. Cuervo AM, Dice JF. Age-related decline in chaperone-mediated autophagy. 70. Qu J, Wang D, Grosskreutz CL. Mechanisms of retinal ganglion cell injury
J Biol Chem 2000;275:31505–13. and defense in glaucoma. Exp Eye Res 2010;91:48–53.
36. Farre JC, Krick R, Subramani S, et al. Turnover of organelles by autophagy in 71. Pease ME, McKinnon SJ, Quigley HA, et al. Obstructed axonal transport of
yeast. Curr Opin Cell Biol 2009;21:522–30. BDNF and its receptor TrkB in experimental glaucoma. Invest Ophthalmol
37. Marzella L, Ahlberg J, Glaumann H. Autophagy, heterophagy, microauto­ Vis Sci 2000;41:764–74.
phagy and crinophagy as the means for intracellular degradation. Virchows 72. Quigley HA. Neuronal death in glaucoma. Prog Retin Eye Res 1999;18:
Arch B Cell Pathol Incl Mol Pathol 1981;36:219–34. 39–57.
38. Sahu R, Kaushik S, Clement CC, et al. Microautophagy of cytosolic proteins 73. Quigley HA, Nickells RW, Kerrigan LA, et al. Retinal ganglion cell death
by late endosomes. Dev Cell 2011;20:131–9. in experimental glaucoma and after axotomy occurs by apoptosis. Invest
39. Banerjee R, Beal MF, Thomas B. Autophagy in neurodegenerative disorders: Ophthalmol Vis Sci 1995;36:774–86.
pathogenic roles and therapeutic implications. Trends Neurosci 2010;33: 74. Kerrigan LA, Zack DJ, Quigley HA, et al. TUNEL-positive ganglion cells in
541–9. human primary open-angle glaucoma. Arch Ophthalmol 1997;115:1031–5.
40. Alamouti B, Funk J. Retinal thickness decreases with age: an OCT study. 75. Levin LA. Mechanisms of optic neuropathy. Curr Opin Ophthalmol 1997;8:
Br J Ophthalmol 2003;87:899–901. 9–15.
41. Cavallotti C, Artico M, Pescosolido N, et al. Age-related changes in the human 76. Baltmr A, Duggan J, Nizari S, et al. Neuroprotection in glaucoma – Is there a
retina. Can J Ophthalmol 2004;39:61–8. future role? Exp Eye Res 2010;91:554–66.
42. Eriksson U, Alm A. Macular thickness decreases with age in normal eyes: 77. Libby RT, Li Y, Savinova O, et al. Susceptibility to neurodegeneration in a
a study on the macular thickness map protocol in the Stratus OCT. Br J glaucoma is modified by Bax gene dosage. PLoS Genet 2005;1:17–26.
Ophthalmol 2009;93:1448–52. 78. Rodriguez-Muela N, Germain F, Marino G, et al. Autophagy promotes sur-
43. Marshall J, Grindle J, Ansell PL, et al. Convolution in human rods: an ageing vival of retinal ganglion cells after optic nerve axotomy in mice. Cell Death
process. Br J Ophthalmol 1979;63:181–7. Differ 2011;19:162–9.
44. Tucker GS. Refractile bodies in the inner segments of cones in the aging 79. Russo R, Berliocchi L, Adornetto A, et al. Calpain-mediated cleavage of
human retina. Invest Ophthalmol Vis Sci 1986;27:708–15. Beclin-1 and autophagy deregulation following retinal ischemic injury in vivo.
45. Iwasaki M, Inomata H. Lipofuscin granules in human photoreceptor cells. Cell Death Dis 2011;2:e144.
Invest Ophthalmol Vis Sci 1988;29:671–9. 80. Kim SH, Munemasa Y, Kwong JM, et al. Activation of autophagy in retinal
46. Boulton ME. Lipofuscin of the retinal pigment epithelium. In: Lois N, ganglion cells. J Neurosci Res 2008;86:2943–51.
Forrester JV, editors. Fundus autofluorescence. Philadelphia: Wolters 81. Sternberg C, Benchimol M, Linden R. Caspase dependence of the death of
Kluwer/Lipincott Williams & Wilkins; 2009. neonatal retinal ganglion cells induced by axon damage and induction of
47. Gartner S, Henkind P. Aging and degeneration of the human macula. 1. Outer autophagy as a survival mechanism. Braz J Med Biol Res 2010;43:950–6.
nuclear layer and photoreceptors. Br J Ophthalmol 1981;65:23–8. 82. Hammes HP, Lin J, Renner O, et al. Pericytes and the pathogenesis of diabetic
48. Eliasieh K, Liets LC, Chalupa LM. Cellular reorganization in the human retina retinopathy. Diabetes 2002;51:3107–12.
during normal aging. Invest Ophthalmol Vis Sci 2007;48:2824–30. 83. Porta M. The importance of endothelial damage: Diabetic retinopathy.
49. Mansergh F, Orton NC, Vessey JP, et al. Mutation of the calcium channel Adv Stud Med 2005;5:S150–8.
gene Cacna1f disrupts calcium signaling, synaptic transmission and cellular 84. Cogan DG, Toussaint D, Kuwabara T. Retinal vascular patterns. IV. Diabetic
organization in mouse retina. Hum Mol Genet 2005;14:3035–46. retinopathy. Arch Ophthalmol 1961;66:366–78.
50. Liets LC, Eliasieh K, van der List DA, et al. Dendrites of rod bipolar cells 85. Kowluru RA. Diabetic retinopathy: mitochondrial dysfunction and retinal
sprout in normal aging retina. Proc Natl Acad Sci U S A 2006;103:12156–60. capillary cell death. Antioxid Redox Signal 2005;7:1581–87.
51. Curcio CA, Millican CL, Allen KA, et al. Aging of the human photoreceptor 86. Barber AJ, Gardner TW, Abcouwer SF. The significance of vascular and neural
mosaic: evidence for selective vulnerability of rods in central retina. Invest apoptosis to the pathology of diabetic retinopathy. Invest Ophthalmol Vis Sci
Ophthalmol Vis Sci 1993;34:3278–96. 2011;52:1156–63.
87. El-Remessy AB, Al-Shabrawey M, Khalifa Y, et al. Neuroprotective 120. Mitter SK, Rao HV, Qi X, et al. Autophagy in the retina: A potential role in
and blood–retinal barrier-preserving effects of cannabidiol in experimental age-related macular degeneration. Adv Exp Med Biol 2012;723:83–90.
diabetes. Am J Pathol 2006;168:235–44. 121. Krohne TU, Stratmann NK, Kopitz J, et al. Effects of lipid peroxidation prod-
550 88. Barber AJ, Lieth E, Khin SA, et al. Neural apoptosis in the retina during ucts on lipofuscinogenesis and autophagy in human retinal pigment epithelial
experimental and human diabetes. Early onset and effect of insulin. J Clin cells. Exp Eye Res 2010;90:465–71.
Invest 1998;102:783–91. 122. Liu J, Lu W, Reigada D, et al. Restoration of lysosomal pH in RPE cells
89. Lopes de Faria JM, Russ H, Costa VP. Retinal nerve fibre layer loss in patients from cultured human and ABCA4(–/–) mice: pharmacologic approaches and
with type 1 diabetes mellitus without retinopathy. Br J Ophthalmol functional recovery. Invest Ophthalmol Vis Sci 2008;49:772–80.
Section 2

2002;86:725–8. 123. Wang AL, Lukas TJ, Yuan M, et al. Autophagy and exosomes in the
90. van Dijk HW, Kok PH, Garvin M, et al. Selective loss of inner retinal aged retinal pigment epithelium: possible relevance to drusen formation and
layer thickness in type 1 diabetic patients with minimal diabetic retinopathy. age-related macular degeneration. PLoS ONE 2009;4:e4160.
Invest Ophthalmol Vis Sci 2009;50:3404–9. 124. Yang L, Bula D, Arroyo JG, et al. Preventing retinal detachment-associated
91. van Dijk HW, Verbraak FD, Kok PH, et al. Decreased retinal ganglion cell photoreceptor cell loss in Bax-deficient mice. Invest Ophthalmol Vis Sci
layer thickness in patients with type 1 diabetes. Invest Ophthalmol Vis Sci 2004;45:648–54.
2010;51:3660–5. 125. Arroyo JG, Yang L, Bula D, et al. Photoreceptor apoptosis in human retinal
92. Gastinger MJ, Singh RS, Barber AJ. Loss of cholinergic and dopaminergic detachment. Am J Ophthalmol 2005;139:605–10.
Basic Mechanisms of Injury in the Retina

amacrine cells in streptozotocin-diabetic rat and Ins2Akita-diabetic mouse 126. Cook B, Lewis GP, Fisher SK, et al. Apoptotic photoreceptor degeneration in
Basic Science and Translation to Therapy

retinas. Invest Ophthalmol Vis Sci 2006;47:3143–50. experimental retinal detachment. Invest Ophthalmol Vis Sci 1995;36:990–6.
93. Chen ZF, Li YB, Han JY, et al. The double-edged effect of autophagy in 127. Hisatomi T, Sakamoto T, Goto Y, et al. Critical role of photoreceptor apoptosis
pancreatic beta cells and diabetes. Autophagy 2011;7:12–6. in functional damage after retinal detachment. Curr Eye Res 2002;24:161–72.
94. Gonzalez CD, Lee MS, Marchetti P, et al. The emerging role of autophagy in 128. Besirli CG, Chinskey ND, Zheng QD, et al. Inhibition of retinal detachment-
the pathophysiology of diabetes mellitus. Autophagy 2011;7:2–11. induced apoptosis in photoreceptors by a small peptide inhibitor of the fas
95. Qi X, Cai J, Dunn WR, et al. Circadian rhythmicity in the expression of receptor. Invest Ophthalmol Vis Sci 2010;51:2177–84.
autophagy proteins in normal and diabetic retinas. Invest Ophthalmol Vis Sci 129. Dong K, Sun X. Targeting death receptor induced apoptosis and necroptosis:
2010;51:ARVO E-Abstract 5614. A novel therapeutic strategy to prevent neuronal damage in retinal detach-
96. Chan-Ling T, Baxter L, Afzal A, et al. Hematopoietic stem cells provide repair ment. Med Hypotheses 2011;77:144–6.
functions after laser-induced Bruch’s membrane rupture model of choroidal 130. Besirli CG, Chinskey ND, Zheng QD, et al. Autophagy activation in the
neovascularization. Am J Pathol 2006;168:1031–44. injured photoreceptor inhibits fas-mediated apoptosis. Invest Ophthalmol Vis
97. Grant MB, May WS, Caballero S, et al. Adult hematopoietic stem cells provide Sci 2011;52:4193–9.
functional hemangioblast activity during retinal neovascularization. Nat Med 131. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet 2006;368:
2002;8:607–12. 1795–809.
98. Busik JV, Tikhonenko M, Bhatwadekar A, et al. Diabetic retinopathy is 132. Marigo V. Programmed cell death in retinal degeneration: targeting apoptosis
associated with bone marrow neuropathy and a depressed peripheral clock. in photoreceptors as potential therapy for retinal degeneration. Cell Cycle
J Exp Med 2009;206:2897–906. 2007;6:652–5.
99. Caballero S, Sengupta N, Afzal A, et al. Ischemic vascular damage can be 133. Ferrari S, Di Iorio E, Barbaro V, et al. Retinitis pigmentosa: genes and disease
repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes mechanisms. Curr Genomics 2011;12:238–49.
2007;56:960–7. 134. Portera-Cailliau C, Sung CH, Nathans J, et al. Apoptotic photoreceptor cell
100. Holz FG, Bindewald-Wittich A, Fleckenstein M, et al. Progression of death in mouse models of retinitis pigmentosa. Proc Natl Acad Sci U S A
geographic atrophy and impact of fundus autofluorescence patterns in 1994;91:974–8.
age-related macular degeneration. Am J Ophthalmol 2007;143:463–72. 135. Travis GH. Mechanisms of cell death in the inherited retinal degenerations.
101. Schatz H, McDonald HR. Atrophic macular degeneration. Rate of spread of Am J Hum Genet 1998;62:503–8.
geographic atrophy and visual loss. Ophthalmology 1989;96:1541–51. 136. Cottet S, Schorderet DF. Mechanisms of apoptosis in retinitis pigmentosa.
102. Fleckenstein M, Adrion C, Schmitz-Valckenberg S, et al. Concordance Curr Mol Med 2009;9:375–83.
of disease progression in bilateral geographic atrophy due to AMD. 137. Liu C, Li Y, Peng M, et al. Activation of caspase-3 in the retina of transgenic
Invest Ophthalmol Vis Sci 2010;51:637–42. rats with the rhodopsin mutation s334ter during photoreceptor degeneration.
103. McLeod DS, Grebe R, Bhutto I, et al. Relationship between RPE and chorio- J Neurosci 1999;19:4778–85.
capillaris in age-related macular degeneration. Invest Ophthalmol Vis Sci 138. Chang GQ, Hao Y, Wong F. Apoptosis: final common pathway of photorecep-
2009;50:4982–91. tor death in rd, rds, and rhodopsin mutant mice. Neuron 1993;11:595–605.
104. Curcio CA, Medeiros NE, Millican CL. Photoreceptor loss in age-related 139. Bode C, Wolfrum U. Caspase-3 inhibitor reduces apototic photoreceptor cell
macular degeneration. Invest Ophthalmol Vis Sci 1996;37:1236–49. death during inherited retinal degeneration in tubby mice. Mol Vis 2003;9:
105. Jackson GR, Owsley C, Curcio CA. Photoreceptor degeneration and dysfunc- 144–50.
tion in aging and age-related maculopathy. Ageing Res Rev 2002;1:381–96. 140. Doonan F, Donovan M, Cotter TG. Caspase-independent photoreceptor apop-
106. Dunaief JL, Dentchev T, Ying GS, et al. The role of apoptosis in age-related tosis in mouse models of retinal degeneration. J Neurosci 2003;23:5723–31.
macular degeneration. Arch Ophthalmol 2002;120:1435–42. 141. Lohr HR, Kuntchithapautham K, Sharma AK, et al. Multiple, parallel cellular
107. Xu GZ, Li WW, Tso MO. Apoptosis in human retinal degenerations. suicide mechanisms participate in photoreceptor cell death. Exp Eye Res
Trans Am Ophthalmol Soc 1996;94:411–30; discussion 430–1. 2006;83:380–9.
108. Nordgaard CL, Berg KM, Kapphahn RJ, et al. Proteomics of the retinal 142. Arimura N, Ki-i Y, Hashiguchi T, et al. Intraocular expression and release of
pigment epithelium reveals altered protein expression at progressive stages high-mobility group box 1 protein in retinal detachment. Lab Invest 2009;
of age-related macular degeneration. Invest Ophthalmol Vis Sci 2006;47: 89:278–89.
815–22. 143. Wenzel A, Grimm C, Samardzija M, et al. Molecular mechanisms of light-
109. Jiang S, Wu MW, Sternberg P, et al. Fas mediates apoptosis and oxidant- induced photoreceptor apoptosis and neuroprotection for retinal degenera-
induced cell death in cultured hRPE cells. Invest Ophthalmol Vis Sci 2000;41: tion. Prog Retin Eye Res 2005;24:275–306.
645–55. 144. Mainster MA, Boulton ME. Retinal toxicity. In: Albert DM, Jakobiec FA, Azar
110. Bazan NG. Cell survival matters: docosahexaenoic acid signaling, neuropro- DT, et al., editors. Principles and practice of ophthalmology. London: Elsevier;
tection and photoreceptors. Trends Neurosci 2006;29:263–71. 2008. p. 2195–205.
111. Cai J, Nelson KC, Wu M, et al. Oxidative damage and protection of the RPE. 145. Organisciak DT, Vaughan DK. Retinal light damage: mechanisms and
Prog Retin Eye Res 2000;19:205–21. protection. Prog Retin Eye Res 2010;29:113–34.
112. Kasahara E, Lin LR, Ho YS, et al. SOD2 protects against oxidation-induced 146. Wu J, Seregard S, Algvere PV. Photochemical damage of the retina.
apoptosis in mouse retinal pigment epithelium: implications for age-related Surv Ophthalmol 2006;51:461–81.
macular degeneration. Invest Ophthalmol Vis Sci 2005;46:3426–34. 147. Noell WK, Walker VS, Kang BS, et al. Retinal damage by light in rats.
113. Mansoor S, Gupta N, Patil AJ, et al. Inhibition of apoptosis in human retinal Invest Ophthalmol 1966;5:450–73.
pigment epithelial cells treated with benzo(e)pyrene, a toxic component of 148. Ham Jr WT, Mueller HA, Ruffolo Jr JJ, et al. Action spectrum for retinal injury
cigarette smoke. Invest Ophthalmol Vis Sci 2010;51:2601–7. from near-ultraviolet radiation in the aphakic monkey. Am J Ophthalmol
114. Kaneko H, Dridi S, Tarallo V, et al. DICER1 deficit induces Alu RNA toxicity 1982;93:299–306.
in age-related macular degeneration. Nature 2011;471:325–30. 149. Abler AS, Chang CJ, Ful J, et al. Photic injury triggers apoptosis of photorecep-
115. Shamsi FA, Boulton M. Inhibition of RPE lysosomal and antioxidant activity tor cells. Res Commun Mol Pathol Pharmacol 1996;92:177–89.
by the age pigment lipofuscin. Invest Ophthalmol Vis Sci 2001;42:3041–6. 150. Zhang SR, Li SH, Abler A, et al. Tissue transglutaminase in apoptosis
116. Jarrett SG, Boulton ME. Antioxidant up-regulation and increased nuclear of photoreceptor cells in rat retina. Invest Ophthalmol Vis Sci 1996;37:
DNA protection play key roles in adaptation to oxidative stress in epithelial 1793–9.
cells. Free Radic Biol Med 2005;38:1382–91. 151. Hafezi F, Marti A, Munz K, et al. Light-induced apoptosis: differential timing
117. Liang FQ, Godley BF. Oxidative stress-induced mitochondrial DNA damage in the retina and pigment epithelium. Exp Eye Res 1997;64:963–70.
in human retinal pigment epithelial cells: a possible mechanism for RPE aging 152. Hafezi F, Steinbach JP, Marti A, et al. The absence of c-fos prevents light-
and age-related macular degeneration. Exp Eye Res 2003;76:397–403. induced apoptotic cell death of photoreceptors in retinal degeneration in vivo.
118. Winkler BS, Boulton ME, Gottsch JD, et al. Oxidative damage and age-related Nat Med 1997;3:346–9.
macular degeneration. Mol Vis 1999;5:32. 153. Grimm C, Wenzel A, Hafezi F, et al. Protection of Rpe65-deficient mice identi-
119. Kaarniranta K. Autophagy – hot topic in AMD. Acta Ophthalmol 2010;88: fies rhodopsin as a mediator of light-induced retinal degeneration. Nat Genet
387–8. 2000;25:63–6.
154. Grimm C, Wenzel A, Williams T, et al. Rhodopsin-mediated blue- 187. Li F, Cao W, Anderson RE. Alleviation of constant-light-induced photorecep-
light damage to the rat retina: effect of photoreversal of bleaching. tor degeneration by adaptation of adult albino rat to bright cyclic light.
Invest Ophthalmol Vis Sci 2001;42:497–505. Invest Ophthalmol Vis Sci 2003;44:4968–75.
155. Keller C, Grimm C, Wenzel A, et al. Protective effect of halothane anesthesia 188. Casson RJ, Chidlow G, Wood JP, et al. The effect of retinal ganglion cell injury 551
on retinal light damage: inhibition of metabolic rhodopsin regeneration. on light-induced photoreceptor degeneration. Invest Ophthalmol Vis Sci
Invest Ophthalmol Vis Sci 2001;42:476–80. 2004;45:685–93.
156. Wenzel A, Reme CE, Williams TP, et al. The Rpe65 Leu450Met variation 189. Casson RJ, Wood JP, Melena J, et al. The effect of ischemic preconditioning

Chapter 24
increases retinal resistance against light-induced degeneration by slowing on light-induced photoreceptor injury. Invest Ophthalmol Vis Sci 2003;44:
rhodopsin regeneration. J Neurosci 2001;21:53–8. 1348–54.
157. Rozanowska M, Sarna T. Light-induced damage to the retina: role of rhodop- 190. Cao W, Tombran-Tink J, Elias R, et al. In vivo protection of photoreceptors
sin chromophore revisited. Photochem Photobiol 2005;81:1305–30. from light damage by pigment epithelium-derived factor. Invest Ophthalmol
158. Farber DB, Danciger JS, Organisciak DT. Levels of mRNA encoding proteins Vis Sci 2001;42:1646–52.
of the cGMP cascade as a function of light environment. Exp Eye Res 1991;53: 191. Lau D, Flannery J. Viral-mediated FGF-2 treatment of the constant light
781–6. damage model of photoreceptor degeneration. Doc Ophthalmol 2003;106:
159. Organisciak DT, Xie A, Wang HM, et al. Adaptive changes in visual cell 89–98.
transduction protein levels: effect of light. Exp Eye Res 1991;53:773–9. 192. Spencer B, Agarwala S, Gentry L, et al. HSV-1 vector-delivered FGF2 to the
160. Kennedy MJ, Lee KA, Niemi GA, et al. Multiple phosphorylation of rhodopsin retina is neuroprotective but does not preserve functional responses. Mol Ther

Cell Death, Apoptosis, and Autophagy in Retinal Injury


and the in vivo chemistry underlying rod photoreceptor dark adaptation. 2001;3:746–56.
Neuron 2001;31:87–101. 193. Kano T, Abe T, Tomita H, et al. Protective effect against ischemia and light
161. Fain GL, Lisman JE. Light, Ca2+, and photoreceptor death: new evidence damage of iris pigment epithelial cells transfected with the BDNF gene.
for the equivalent-light hypothesis from arrestin knockout mice. Invest Invest Ophthalmol Vis Sci 2002;43:3744–53.
Ophthalmol Vis Sci 1999;40:2770–2. 194. Bainbridge JW, Ali RR. Success in sight: The eyes have it! Ocular gene therapy
162. Wu J, Seregard S, Spangberg B, et al. Blue light induced apoptosis in rat retina. trials for LCA look promising. Gene Ther 2008;15:1191–2.
Eye (Lond) 1999;13:577–83. 195. Bok D, Yasumura D, Matthes MT, et al. Effects of adeno-associated virus-
163. Wu J, Gorman A, Zhou X, et al. Involvement of caspase-3 in photoreceptor vectored ciliary neurotrophic factor on retinal structure and function in mice
cell apoptosis induced by in vivo blue light exposure. Invest Ophthalmol Vis with a P216L rds/peripherin mutation. Exp Eye Res 2002;74:719–35.
Sci 2002;43:3349–54. 196. Cayouette M, Smith SB, Becerra SP, et al. Pigment epithelium-derived factor
164. Westlund BS, Cai B, Zhou J, et al. Involvement of c-Abl, p53 and the MAP delays the death of photoreceptors in mouse models of inherited retinal
kinase JNK in the cell death program initiated in A2E-laden ARPE-19 cells by degenerations. Neurobiol Dis 1999;6:523–32.
exposure to blue light. Apoptosis 2009;14:31–41. 197. Huang SP, Lin PK, Liu JH, et al. Intraocular gene transfer of ciliary neuro-
165. Tezel G, Kolker AE, Kass MA, et al. Parapapillary chorioretinal atrophy in trophic factor rescues photoreceptor degeneration in RCS rats. J Biomed Sci
patients with ocular hypertension. I. An evaluation as a predictive factor for 2004;11:37–48.
the development of glaucomatous damage. Arch Ophthalmol 1997;115: 198. Liang FQ, Aleman TS, Dejneka NS, et al. Long-term protection of retinal
1503–8. structure but not function using RAAV.CNTF in animal models of retinitis
166. Tezel G, Kolker AE, Wax MB, et al. Parapapillary chorioretinal atrophy in pigmentosa. Mol Ther 2001;4:461–72.
patients with ocular hypertension. II. An evaluation of progressive changes. 199. Schlichtenbrede FC, MacNeil A, Bainbridge JW, et al. Intraocular gene deliv-
Arch Ophthalmol 1997;115:1509–14. ery of ciliary neurotrophic factor results in significant loss of retinal function
167. Donovan M, Cotter TG. Caspase-independent photoreceptor apoptosis in in normal mice and in the Prph2Rd2/Rd2 model of retinal degeneration.
vivo and differential expression of apoptotic protease activating factor-1 and Gene Ther 2003;10:523–7.
caspase-3 during retinal development. Cell Death Differ 2002;9:1220–31. 200. Miyazaki M, Ikeda Y, Yonemitsu Y, et al. Simian lentiviral vector-mediated
168. Gaillard ER, Atherton SJ, Eldred G, et al. Photophysical studies on human retinal gene transfer of pigment epithelium-derived factor protects retinal
retinal lipofuscin. Photochem Photobiol 1995;61:448–53. degeneration and electrical defect in Royal College of Surgeons rats.
169. Rozanowska M, Jarvis-Evans J, Korytowski W, et al. Blue light-induced reac- Gene Ther 2003;10:1503–11.
tivity of retinal age pigment. In vitro generation of oxygen-reactive species. 201. Tao W, Wen R, Goddard MB, et al. Encapsulated cell-based delivery of CNTF
J Biol Chem 1995;270:18825–30. reduces photoreceptor degeneration in animal models of retinitis pigmentosa.
170. Rozanowska M, Wessels J, Boulton M, et al. Blue light-induced singlet oxygen Invest Ophthalmol Vis Sci 2002;43:3292–8.
generation by retinal lipofuscin in non-polar media. Free Radic Biol Med 202. Chong NH, Alexander RA, Waters L, et al. Repeated injections of a ciliary neu-
1998;24:1107–12. rotrophic factor analogue leading to long-term photoreceptor survival in
171. Paquet-Durand F, Azadi S, Hauck SM, et al. Calpain is activated in degenerat- hereditary retinal degeneration. Invest Ophthalmol Vis Sci 1999;40:1298–305.
ing photoreceptors in the rd1 mouse. J Neurochem 2006;96:802–14. 203. Harada T, Harada C, Nakayama N, et al. Modification of glial-neuronal cell
172. Sharma AK, Rohrer B. Calcium-induced calpain mediates apoptosis via interactions prevents photoreceptor apoptosis during light-induced retinal
caspase-3 in a mouse photoreceptor cell line. J Biol Chem 2004;279:35564–72. degeneration. Neuron 2000;26:533–41.
173. Hafezi F, Marti A, Grimm C, et al. Differential DNA binding activities of the 204. Okoye G, Zimmer J, Sung J, et al. Increased expression of brain-derived
transcription factors AP-1 and Oct-1 during light-induced apoptosis of neurotrophic factor preserves retinal function and slows cell death from
photoreceptors. Vision Res 1999;39:2511–8. rhodopsin mutation or oxidative damage. J Neurosci 2003;23:4164–72.
174. Sang A, Cheng Y, Lu H, et al. Light-induced retinal ganglion cell damage in 205. Lawrence JM, Keegan DJ, Muir EM, et al. Transplantation of Schwann cell line
vivo involves Dexras1. Mol Vis 2011;17:134–43. clones secreting GDNF or BDNF into the retinas of dystrophic Royal College
175. Dacey DM, Liao HW, Peterson BB, et al. Melanopsin-expressing ganglion of Surgeons rats. Invest Ophthalmol Vis Sci 2004;45:267–74.
cells in primate retina signal colour and irradiance and project to the LGN. 206. Yoshizawa K, Kiuchi K, Nambu H, et al. Caspase-3 inhibitor transiently delays
Nature 2005;433:749–54. inherited retinal degeneration in C3H mice carrying the rd gene. Graefes Arch
176. Osborne NN, Li GY, Ji D, et al. Light affects mitochondria to cause apoptosis Clin Exp Ophthalmol 2002;240:214–9.
to cultured cells: possible relevance to ganglion cell death in certain optic 207. Zeiss CJ, Neal J, Johnson EA. Caspase-3 in postnatal retinal development and
neuropathies. J Neurochem 2008;105:2013–28. degeneration. Invest Ophthalmol Vis Sci 2004;45:964–70.
177. Kunchithapautham K, Coughlin B, Lemasters JJ, et al. Differential effects of 208. Leonard KC, Petrin D, Coupland SG, et al. XIAP protection of photoreceptors
rapamycin on rods and cones during light-induced stress in albino mice. in animal models of retinitis pigmentosa. PLoS ONE 2007;2:e314.
Invest Ophthalmol Vis Sci 2011;52:2967–75. 209. Renwick J, Narang MA, Coupland SG, et al. XIAP-mediated neuroprotection
178. Kunchithapautham K, Rohrer B. Apoptosis and autophagy in photoreceptors in retinal ischemia. Gene Ther 2006;13:339–47.
exposed to oxidative stress. Autophagy 2007;3:433–41. 210. Glybina IV, Kennedy A, Ashton P, et al. Photoreceptor neuroprotection in
179. Bahr M. Neuroprotection: models, mechanisms, and therapies. Weinheim, RCS rats via low-dose intravitreal sustained-delivery of fluocinolone aceton-
Germany: Wiley-VCH, 2004. ide. Invest Ophthalmol Vis Sci 2009;50:4847–57.
180. Faden AI, Stoica B. Neuroprotection: challenges and opportunities. Arch 211. Delyfer MN, Simonutti M, Neveux N, et al. Does GDNF exert its neuroprotec-
Neurol 2007;64:794–800. tive effects on photoreceptors in the rd1 retina through the glial glutamate
181. Whitcup SM. Clinical trials in neuroprotection. Prog Brain Res 2008;173: transporter GLAST? Mol Vis 2005;11:677–87.
323–35. 212. Read SP, Cashman SM, Kumar-Singh R. POD nanoparticles expressing GDNF
182. Ishizawa Y, Sharp R, Liebman PA, et al. Halothane binding to a G provide structural and functional rescue of light-induced retinal degeneration
protein coupled receptor in retinal membranes by photoaffinity labeling. in an adult mouse. Mol Ther 2010;18:1917–26.
Biochemistry 2000;39:8497–502. 213. Cheung W, Guo L, Cordeiro MF. Neuroprotection in glaucoma: drug-based
183. Donovan M, Carmody RJ, Cotter TG. Light-induced photoreceptor apoptosis approaches. Optom Vis Sci 2008;85:406–16.
in vivo requires neuronal nitric-oxide synthase and guanylate cyclase activity 214. Danesh-Meyer HV. Neuroprotection in glaucoma: recent and future
and is caspase-3-independent. J Biol Chem 2001;276:23000–8. directions. Curr Opin Ophthalmol 2011;22:78–86.
184. Goureau O, Jeanny JC, Becquet F, et al. Protection against light-induced retinal 215. Dong CJ, Guo Y, Agey P, et al. Alpha2 adrenergic modulation of NMDA
degeneration by an inhibitor of NO synthase. Neuroreport 1993;5:233–6. receptor function as a major mechanism of RGC protection in experimental
185. Hahn P, Lindsten T, Lyubarsky A, et al. Deficiency of Bax and Bak protects glaucoma and retinal excitotoxicity. Invest Ophthalmol Vis Sci 2008;49:
photoreceptors from light damage in vivo. Cell Death Differ 2004;11:1192–7. 4515–22.
186. Liu C, Peng M, Laties AM, et al. Preconditioning with bright light evokes a 216. Russo R, Cavaliere F, Rombola L, et al. Rational basis for the development of
protective response against light damage in the rat retina. J Neurosci coenzyme Q10 as a neurotherapeutic agent for retinal protection. Prog Brain
1998;18:1337–44. Res 2008;173:575–82.
217. Pease ME, Zack DJ, Berlinicke C, et al. Effect of CNTF on retinal ganglion cell 249. Stepkowski TM, Kruszewski MK. Molecular cross-talk between the NRF2/
survival in experimental glaucoma. Invest Ophthalmol Vis Sci 2009;50: KEAP1 signaling pathway, autophagy, and apoptosis. Free Radic Biol Med
2194–200. 2011;50:1186–95.
552 218. Tao W. Application of encapsulated cell technology for retinal degenerative 250. Goligorsky MS. SIRTing out the link between autophagy and ageing. Nephrol
diseases. Expert Opin Biol Ther 2006;6:717–26. Dial Transplant 2010;25:2434–6.
219. Kashimoto R, Kurimoto T, Miyoshi T, et al. Cilostazol promotes survival of 251. Hariharan N, Maejima Y, Nakae J, et al. Deacetylation of FoxO by Sirt1 plays
axotomized retinal ganglion cells in adult rats. Neurosci Lett 2008;436: an essential role in mediating starvation-induced autophagy in cardiac myo-
116–9. cytes. Circ Res 2010;107:1470–82.
Section 2

220. Koriyama Y, Tanii H, Ohno M, et al. A novel neuroprotective role of a small 252. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: physiology and
peptide from flesh fly, 5-S-GAD in the rat retina in vivo. Brain Res 2008; pathology. Trends Biochem Sci 2011;36:30–8.
1240:196–203. 253. Massey AC, Kaushik S, Sovak G, et al. Consequences of the selective blockage
221. Bazan NG. Neurotrophins induce neuroprotective signaling in the retinal of chaperone-mediated autophagy. Proc Natl Acad Sci U S A 2006;103:
pigment epithelial cell by activating the synthesis of the anti-inflammatory 5805–10.
and anti-apoptotic neuroprotectin D1. Adv Exp Med Biol 2008;613:39–44. 254. Kaushik S, Massey AC, Mizushima N, et al. Constitutive activation of
222. Mukherjee PK, Marcheselli VL, Barreiro S, et al. Neurotrophins enhance chaperone-mediated autophagy in cells with impaired macroautophagy. Mol
retinal pigment epithelial cell survival through neuroprotectin D1 signaling. Biol Cell 2008;19:2179–92.
Basic Mechanisms of Injury in the Retina

Proc Natl Acad Sci U S A 2007;104:13152–7. 255. Iwata A, Christianson JC, Bucci M, et al. Increased susceptibility of cytoplas-
Basic Science and Translation to Therapy

223. Wang ZY, Shen LJ, Tu L, et al. Erythropoietin protects retinal pigment epithe- mic over nuclear polyglutamine aggregates to autophagic degradation.
lial cells from oxidative damage. Free Radic Biol Med 2009;46:1032–41. Proc Natl Acad Sci U S A 2005;102:13135–40.
224. Wrona M, Rozanowska M, Sarna T. Zeaxanthin in combination with ascorbic 256. Ding Q, Dimayuga E, Martin S, et al. Characterization of chronic low-level
acid or alpha-tocopherol protects ARPE-19 cells against photosensitized proteasome inhibition on neural homeostasis. J Neurochem 2003;86:489–97.
peroxidation of lipids. Free Radic Biol Med 2004;36:1094–101. 257. Cuervo AM, Palmer A, Rivett AJ, et al. Degradation of proteasomes by
225. Bryckaert M, Guillonneau X, Hecquet C, et al. Both FGF1 and bcl-x synthesis lysosomes in rat liver. Eur J Biochem 1995;227:792–800.
are necessary for the reduction of apoptosis in retinal pigmented epithelial 258. Massey AC, Follenzi A, Kiffin R, et al. Early cellular changes after blockage
cells by FGF2: role of the extracellular signal-regulated kinase 2. Oncogene of chaperone-mediated autophagy. Autophagy 2008;4:442–56.
1999;18:7584–93. 259. Araki M. Regeneration of the amphibian retina: role of tissue interaction and
226. Elner SG, Yoshida A, Bian ZM, et al. Human RPE cell apoptosis induced related signaling molecules on RPE transdifferentiation. Dev Growth Differ
by activated monocytes is mediated by caspase-3 activation. Trans Am 2007;49:109–20.
Ophthalmol Soc 2003;101:77–89; discussion 89–91. 260. Karl MO, Reh TA. Regenerative medicine for retinal diseases: activating
227. Ferrington DA, Tran TN, Lew KL, et al. Different death stimuli evoke apop- endogenous repair mechanisms. Trends Mol Med 2010;16:193–202.
tosis via multiple pathways in retinal pigment epithelial cells. Exp Eye Res 261. Bernardos RL, Barthel LK, Meyers JR, et al. Late-stage neuronal progenitors
2006;83:638–50. in the retina are radial Müller glia that function as retinal stem cells. J Neurosci
228. Sreekumar PG, Kannan R, Kitamura M, et al. alphaB crystallin is apically 2007;27:7028–40.
secreted within exosomes by polarized human retinal pigment epithelium 262. Raymond PA, Hitchcock PF. How the neural retina regenerates. Results Probl
and provides neuroprotection to adjacent cells. PLoS ONE 2010;5:e12578. Cell Differ 2000;31:197–218.
229. Alge CS, Priglinger SG, Neubauer AS, et al. Retinal pigment epithelium is 263. Fischer AJ, Reh TA. Müller glia are a potential source of neural regeneration
protected against apoptosis by alphaB-crystallin. Invest Ophthalmol Vis Sci in the postnatal chicken retina. Nat Neurosci 2001;4:247–52.
2002;43:3575–82. 264. Ooto S, Akagi T, Kageyama R, et al. Potential for neural regeneration after
230. AREDS. A randomized, placebo-controlled, clinical trial of high-dose supple- neurotoxic injury in the adult mammalian retina. Proc Natl Acad Sci U S A
mentation with vitamins C and E, beta carotene, and zinc for age-related 2004;101:13654–9
macular degeneration and vision loss: AREDS report no. 8. Arch Ophthalmol 265. Sengupta N, Caballero S, Sullivan SM, et al. Regulation of adult hematopoietic
2001;119:1417–36. stem cells fate for enhanced tissue-specific repair. Mol Ther 2009;17:
231. Imai H, Singh RS, Fort PE, et al. Neuroprotection for diabetic retinopathy. 1594–604.
Dev Ophthalmol 2009;44:56–68. 266. Carr AJ, Vugler AA, Hikita ST, et al. Protective effects of human iPS-derived
232. Bao XH, Naomoto Y, Hao HF, et al. Autophagy: Can it become a potential retinal pigment epithelium cell transplantation in the retinal dystrophic rat.
therapeutic target? Int J Mol Med 2010;25:493–503. PLoS ONE 2009;4:e8152.
233. Codogno P, Meijer AJ. Autophagy and signaling: their role in cell survival 267. Du H, Lim SL, Grob S, et al. Induced pluripotent stem cell therapies for geo-
and cell death. Cell Death Differ 2005;12(Suppl 2):1509–18. graphic atrophy of age-related macular degeneration. Semin Ophthalmol
234. Maiuri MC, Le Toumelin G, Criollo A, et al. Functional and physical interac- 2011;26:216–24.
tion between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J 2007;26: 268. Vugler A, Lawrence J, Walsh J, et al. Embryonic stem cells and retinal repair.
2527–39. Mech Dev 2007;124:807–29.
235. Pattingre S, Tassa A, Qu X, et al. Bcl-2 antiapoptotic proteins inhibit Beclin 269. Lu B, Malcuit C, Wang S, et al. Long-term safety and function of RPE from
1-dependent autophagy. Cell 2005;122:927–39. human embryonic stem cells in preclinical models of macular degeneration.
236. Petiot A, Ogier-Denis E, Blommaart EF, et al. Distinct classes of phospha- Stem Cells 2009;27:2126–35.
tidylinositol 3’-kinases are involved in signaling pathways that control 270. Fong CY, Gauthaman K, Bongso A. Teratomas from pluripotent stem cells:
macroautophagy in HT-29 cells. J Biol Chem 2000;275:992–8. A clinical hurdle. J Cell Biochem 2010;111:769–81.
237. Sarkar S, Rubinsztein DC. Inositol and IP3 levels regulate autophagy: biology 271. Gore A, Li Z, Fung HL, et al. Somatic coding mutations in human induced
and therapeutic speculations. Autophagy 2006;2:132–4. pluripotent stem cells. Nature 2011;471:63–7.
238. Sarkar S, Davies JE, Huang Z, et al. Trehalose, a novel mTOR-independent 272. Binder S, Stanzel BV, Krebs I, et al. Transplantation of the RPE in AMD.
autophagy enhancer, accelerates the clearance of mutant huntingtin and Prog Retin Eye Res 2007;26:516–54.
alpha-synuclein. J Biol Chem 2007;282:5641–52. 273. da Cruz L, Chen FK, Ahmado A, et al. RPE transplantation and its role in
239. Shacka JJ, Roth KA, Zhang J. The autophagy-lysosomal degradation pathway: retinal disease. Prog Retin Eye Res 2007;26:598–635.
role in neurodegenerative disease and therapy. Front Biosci 2008;13:718–36. 274. MacLaren RE, Pearson RA, MacNeil A, et al. Retinal repair by transplantation
240. Winslow AR, Rubinsztein DC. Autophagy in neurodegeneration and devel- of photoreceptor precursors. Nature 2006;444:203–7.
opment. Biochim Biophys Acta 2008;1782:723–9. 275. Bi YY, Feng DF, Pan. DC. Stem/progenitor cells: a potential source of retina-
241. Sarkar S, Rubinsztein DC. Small molecule enhancers of autophagy for neuro- specific cells for retinal repair. Neurosci Res 2009;65:215–21.
degenerative diseases. Mol Biosyst 2008;4:895–901. 276. Ikeda H, Osakada F, Watanabe K, et al. Generation of Rx+/Pax6+ neural
242. Kim JS, Nitta T, Mohuczy D, et al. Impaired autophagy: A mechanism of retinal precursors from embryonic stem cells. Proc Natl Acad Sci U S A 2005;
mitochondrial dysfunction in anoxic rat hepatocytes. Hepatology 2008;47: 102:11331–6.
1725–36. 277. Osakada F, Ikeda H, Sasai Y, et al. Stepwise differentiation of pluripotent stem
243. Amaravadi RK, Lippincott-Schwartz J, Yin XM, et al. Principles and current cells into retinal cells. Nat Protoc 2009;4:811–24.
strategies for targeting autophagy for cancer treatment. Clin Cancer Res 278. Osakada F, Ooto S, Akagi T, et al. Wnt signaling promotes regeneration in the
2011;17:654–66. retina of adult mammals. J Neurosci 2007;27:4210–9.
244. Carew JS, Nawrocki ST, Giles FJ, et al. Targeting autophagy: a novel antican- 279. Lamba DA, Gust J, Reh TA. Transplantation of human embryonic stem cell-
cer strategy with therapeutic implications for imatinib resistance. Biologics derived photoreceptors restores some visual function in Crx-deficient mice.
2008;2:201–4. Cell Stem Cell 2009;4:73–9.
245. Dalby KN, Tekedereli I, Lopez-Berestein G et al. Targeting the prodeath and 280. Bull ND, Martin KR. Using stem cells to mend the retina in ocular disease.
prosurvival functions of autophagy as novel therapeutic strategies in cancer. Regen Med 2009;4:855–64.
Autophagy 2010;6:322–9. 281. Enzmann V, Yolcu E, Kaplan HJ, et al. Stem cells as tools in regenerative
246. Spalton DJ. Retinopathy and antimalarial drugs – the British experience. therapy for retinal degeneration. Arch Ophthalmol 2009;127:563–71.
Lupus 1996;5(Suppl 1):S70–2. 282. Machalinska A, Baumert B, Kuprjanowicz L, et al. Potential application of
247. Kaminskyy V, Zhivotovsky B. Proteases in autophagy. Biochim Biophys Acta adult stem cells in retinal repair – challenge for regenerative medicine.
2011;1824:44–50. Curr Eye Res 2009;34:748–60.
248. Kang R, Zeh HJ, Lotze MT, et al. The Beclin 1 network regulates autophagy 283. Lamba DA, Karl MO, Reh TA. Strategies for retinal repair: cell replacement
and apoptosis. Cell Death Differ 2011;18:571–80. and regeneration. Prog Brain Res 2009;175:23–31.
Basic Mechanisms of Injury in the Retina Section 2

Inflammatory Response and Mediators Chapter

in Retinal Injury
Chi-Chao Chan, Wendy M. Smith 25 
INTRODUCTION
Host defense against injury depends upon inflammatory
responses to remove damaged cells and stimulate tissue repair.
Uncontrollable and/or unresolved inflammatory responses in
the retina are associated with many retinal disorders, including * *
inflammatory diseases such as infectious and noninfectious reti-
nitis, macular retinopathies such as age-related macular degen-
eration (AMD), and vasculopathies such as diabetic retinopathy
and retinal vein occlusion. The heterogeneity of the root causes
of these diseases belies their multiple common, though unre-
lated, signaling pathways. The purpose of this chapter is to give
an overview of the role of inflammation in retinal injury due to
ischemia, oxidative stress, and trauma.

RETINAL INJURY
Ischemia–hypoxia
The retina consumes oxygen more rapidly than any other tissue
Fig. 25.1 Diabetic retinopathy. Inflammatory cells (arrows) are
in the body in part to fuel the constant renewal and replacement
surrounding a small microaneurysm (open arrow) and located in the
of photoreceptor outer segments.1 The presence of abundant cotton-wool spots (asterisks) of the retina with diabetic retinopathy.
mitochondria in the photoreceptor inner segments is evidence of (Hematoxylin and eosin, ×200.)
this high energy demand.2 In addition, the choroid receives a
high rate of blood flow to supply oxygen and glucose to the
retinal pigment epithelium (RPE).3 Inadequate blood supply
decreases oxygen delivery to the retina (hypoxia) and may result
in ischemic injury. The subsequent ischemia can induce neuronal
apoptosis and neovascularization. Classical examples of the
retinal diseases associated with hypoxia and ischemia are dia-
betic retinopathy and retinal vein occlusion.
Inflammation often occurs in hypoxic-ischemic conditions.
Diabetic retinopathy is characterized by retinal vascular nonper-
fusion and leakage. In an experimental rat model of diabetes
mellitus (DM) induced by streptozotocin, retinovascular leuko­
stasis develops within 1 week, followed by a progressive break-
down of the blood–retinal barrier. After 1 week, areas of
nonperfusion and reperfusion develop, and extravascular leuko-
cytes are observed in the retina.4 On a microscopic level in
human diabetic retinopathy, the inflammation is demonstrated
by vascular dilatation, exudation and transudation of fluids, and
leukocyte (macrophages, neutrophils, and T lymphocytes) infil- Fig. 25.2 Retinal vein occlusion. Many inflammatory cells (asterisk)
infiltrate an occluded branch retinal vein. (Hematoxylin and eosin, ×200.)
tration in the retina (Fig. 25.1).5–7 Retinal vein occlusion is another
common condition associated with hypoxia and ischemia. In a
histopathological study of 29 cases with central vein occlusion, plentiful growth factors, including vascular endothelial growth
lymphocytes and macrophages were observed at the occlusion factor (VEGF). Hypoxia and ischemia also result in upregulation
site in 47% of cases (Fig. 25.2).8 of retinal VEGF, which can induce expression of adhesion mol-
Macrophages are often present in the diabetic retina as well as ecules and further leukostasis. Ultimately, retinal neovascular-
in nondiabetic retinal ischemic lesions. Macrophages can produce ization or proliferative diabetic retinopathy may result. Retinal
hypoxia further contributes to the breakdown of the retinal– permeability,21 and activate microglia22 in experimental diabetic
blood barrier with resultant retinal edema and tissue damage. rats. Treatments that target factors related to retinal–blood
554 Further evidence of the involvement of inflammatory pro- barrier breakdown have also been used in retinal vein
cesses in diabetic retinopathy and retinal vein occlusion is based occlusion.
on the upregulation of several inflammatory genes and the As corticosteroids have anti-inflammatory, antiangiogenic,
detection of inflammatory cytokines and chemokines under and antivascular permeability properties, the administration of
Section 2

hypoxic-ischemic conditions. Tumor necrosis factor (TNF)-α, intravitreal corticosteroids (triamcinolone) has shown promising
interleukin (IL)-1β, monocyte chemotactic protein 1 (MCP-1/ results for diabetic macular edema in a number of randomized
CCL2), and macrophage inflammatory protein (MIP)/CCL3 clinical trials.9,23 However, in a carefully designed, prospective,
transcripts have been detected in the ischemic-hypoxic retina. randomized trial, the Diabetic Retinopathy Clinical Research
These proinflammatory cytokines, particularly TNF-α and IL-1β, Network showed that focal or grid laser photocoagulation was
are thought to play a major role in the breakdown of the blood– more effective and had fewer side-effects than intravitreal triam-
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

retinal barrier and the degeneration of retinal capillaries.9 CCL2 cinolone treatment of diabetic macular edema.24 Corticosteroids
and RANTES (regulated upon activation, normal T-cell expressed have also been used to treat both central and branch retinal vein
and secreted)/CCL5 are significantly elevated in sera of patients occlusions.25 Case series have suggested that intravitreal injec-
with severe nonproliferative diabetic retinopathy compared tion of triamcinolone may be useful for the treatment of macular
with those who have less severe retinopathy.7 Increased edema in patients with branch retinal vein occlusion.26 Nonethe-
C-reactive protein (CRP), IL-6, and TNF-α, and especially inter- less, due to a high rate of adverse events, the use of this
cellular adhesion molecule (ICAM)-1, vascular cell adhesion treatment was not supported by the Standard care versus Corti-
molecule-1, and E-selectin are associated with nephropathy, reti- costeroid for Retinal vein occlusion (SCORE) study, a random-
nopathy, and cardiovascular disease in both type 1 and type 2 ized trial which included 411 patients with branch retinal vein
diabetes.10 In proliferative diabetic retinopathy, vitreous cyto- occlusion and vision loss from macular edema.27 In another
kine levels of IL-6, IL-8, and CCL2 are strongly correlated with study of 437 patients with central retinal vein occlusion and 830
elevated VEGF.11 Patients with central retinal vein occlusion with branch vein occlusion, the longer-term delivery of intravit-
have significantly higher levels of aqueous and vitreous VEGF real corticosteroids through a dexamethasone implant was
and IL-6 versus control subjects with nonischemic ocular reported to be associated with a shorter time to achieve a gain
disease.12,13 in visual acuity.28 Elevation of intraocular pressure and cataract
Type 1 DM is now considered a common autoimmune disor- progression are among the potential significant adverse effects
der with multiple genetic susceptibility loci as well as environ- for patients receiving intravitreal corticosteroids. Further devel-
mental risk factors. Recently, a genomewide association study of opment of noncorticosteroid agents that target the inflammatory
type 1 DM was combined in a meta-analysis with two other components underlying retinovascular disease pathogenesis
studies to examine a total of 7514 cases and 9045 reference may provide future therapeutic options.
samples. Several genetic regions, including those containing the
immunoregulatory cytokine genes IL-10, IL-19, and IL-20, Oxidative stress
showed significant associations with type 1 DM, suggesting pos- Oxidative processes occur through the removal of electrons from
sible functional relevance of these genes to the disease pathogen- molecules. In biologic systems, energy is released when lipids,
esis.14 The Wellcome Trust Case Consortium genomewide proteins, and carbohydrates are oxidized to form carbon dioxide
association study of type 1 diabetic families identified a genetic and water. Oxidative reactions may also result in the production
susceptibility region which is close to the inflammatory genes of reactive oxygen intermediates (ROIs), such as free radicals,
Toll-like receptor (TLR)7 and TLR8.15 In another study, a custom hydrogen peroxide, and singlet oxygen. ROIs can damage other
panel of 1536 single-nucleotide polymorphisms (SNPs) located molecules and increase under conditions of irradiation, aging,
in 193 candidate genes was used to genotype 437 African Ameri- reperfusion, inflammation, increased partial pressure of oxygen,
cans with type 1 DM, 128 with and 309 without severe diabetic cigarette smoke, and air pollution.29 The biologic mechanisms to
retinopathy. The results indicated an association between genes prevent the detrimental effects of ROIs include cellular compart-
involved in inflammation, such as ANGPT1 (angiopoietin 1), mentalization, repair of DNA and proteins, and neutralization
FLT1 (fms-related tyrosine kinase 1), PLA2G4A (cytosolic phos- by antioxidant compounds. The retina is an ideal environment
pholipase A2), OLR1 (oxidized low-density lipoprotein lectin- for the generation of ROIs for several reasons: (1) high oxygen
like receptor 1), and PPARG (peroxisome proliferator-activated consumption; (2) high levels of cumulative irradiation; (3) RPE
receptor-γ), and the progression of diabetic retinopathy.16 In con- phagocytosis, which is an oxidative stress that produces
trast, a study of 315 patients and 335 age- and gender-matched ROIs; (4) high levels of polyunsaturated fatty acids in the
control subjects showed no significant genetic association photoreceptor outer-segment membranes; and (5) abundant
between central retinal vein occlusion and variants in many photosensitizers in the neurosensory retina and RPE.29
inflammation-related genes, including TNF-α, IL-1β, IL-6, IL-8, Oxidative stress results when there is an imbalance between
IL-10, and CCL2.17 pro-oxidants and antioxidants, leading to molecular damage
The concept that diabetic retinopathy is a persistent low-grade and/or a disruption of redox signaling.30 Inflammation and oxi-
inflammatory response to ischemia-induced retinal neovascular- dative stress are closely interrelated: inflammatory cells can gen-
ization and damage has led to the clinical application of anti- erate ROIs, and oxidative stress can induce inflammation
inflammatory treatments such as corticosteroids and anti-TNF-α through NF-κβ-mediated inflammatory gene expression.30 Oxi-
agents.5,18 In addition to being a potent inflammatory molecule, dative stress plays a role in the pathogenesis of several retinal
TNF-α can also promote apoptosis and loss of retinal vascular disorders, including AMD and retinopathy of prematurity
pericytes and endothelial cells,19,20 increase retinal endothelial (ROP).
Multiple changes are associated with the aging eye, including
thickening of Bruch’s membrane, accumulation of lipofuscin in
the RPE, and loss of parafoveal rods. In a model outlined by 555
Curcio et al.31 and others, the RPE and Bruch’s membrane are
modified or damaged by oxidative stress and enzymatic pro-

Chapter 25
cesses over time. The resultant damage slows the normal flow
of materials such as lipids, protein, and lipofuscin from the outer
retina and RPE through Bruch’s membrane. However, simple
accumulation of material does not lead to AMD. The retained
materials, including lipoproteins, may be modified by oxidative
stress and become stimuli for inflammation.31 This model of
Bruch’s membrane lipoprotein accumulation and modification

Inflammatory Response and Mediators in Retinal Injury


in a chronic proinflammatory environment is similar to the
current understanding of the pathogenesis of atherosclerosis.

Oxidative stress and inflammation in AMD


Several studies have provided evidence that oxidative stress is
involved in AMD pathogenesis. Crabb et  al. performed a pro-
teomic analysis of drusen from normal and AMD donor eyes.32 Fig. 25.3 Age-related macular degeneration (AMD). Scattered
They identified multiple proteins modified by oxidation, includ- inflammatory cells (arrows) are found in an AMD lesion. (Hematoxylin
ing cross-linked molecules of carboxyethyl pyrrole (CEP) protein and eosin, ×200.)
adducts, tissue metalloproteinase inhibitor 3, and vitronectin.
Carboxymethyl lysine, an advanced glycation end product pro-
duced through oxidation of carbohydrate, was also isolated. that activates the classical complement pathway and enhances
These oxidation-related products were identified more fre- phagocytosis via phagocytic complement receptors.40,41 In this
quently in drusen from AMD eyes. In addition, drusen in AMD manner, oxidative damage to the retina may lead to macrophage
donor eyes were also more likely to contain crystallins, which infiltration and activation via the innate immune response.
are proteins upregulated during cellular stress such as oxidation. An immunohistochemical study of surgically excised AMD-
In another proteomics study, Yuan et  al. isolated the Bruch’s associated choroidal neovascular (CNV) membranes provides
membrane–choroid complex from postmortem AMD and in vivo evidence to support this hypothesis.42 Oxidized lipo-
control eyes.33 They found elevated levels of retinoid-processing proteins were detected in RPE cells as well as on macrophages
proteins in the early and midstage AMD eyes. These retinoids associated with the CNV membranes. Scavenger receptors for
are highly susceptible to oxidative damage, can accumulate in oxidized lipoproteins were also present on RPE cells, macro-
RPE lipofuscin granules, and activate complement. They also phages, and some endothelial cells in the CNV membranes,
found significant elevation of an oxidation product-related although the type and amount of scavenger receptor differed
receptor in eyes with advanced dry AMD. In the study by Yuan for each cell type. These data suggest macrophages accumulate
et  al.,33 approximately 60% of the elevated proteins were in AMD lesions in order to phagocytize oxidized lipoproteins
involved in immune response or host defense, including comple- via scavenger receptors.
ment factors C5 and C7, α-crystallin A, α-crystallin B, and major Hollyfield et  al. have tied together earlier in vitro experiments
histocompatibility complex (MHC) class II molecule DRα. In and in vivo observations by producing AMD-like lesions in
contrast, several proteins classified as damage-associated molec- mice exposed to an oxidative product.43,44 They immunized mice
ular patterns (DAMPs) were present at similar levels in AMD with CEP adducts formed by the covalent interaction with an
and control eyes. Damaged cells release DAMPs which can bind oxidation fragment of docosahexanoic acid (DHA), and found
receptors in the complement system, Toll-like receptors, and higher levels of antibodies against CEP in these mice. The
receptors for advanced glycation end products. These data authors hypothesized that the mouse immune system responds
suggest that both normal and AMD eyes express proteins in to the CEP antibodies by depositing complement below the
response to aging-associated damage; however, in AMD eyes RPE. In support of this idea, they detected complement C3d
the response to injury may evolve into a chronic proinflamma- in Bruch’s membrane and on RPE cells in the CEP-immunized
tory environment. mice but not controls. Furthermore, there was a close relation-
Numerous histopathologic studies of AMD eyes have docu- ship between the level of CEP-specific antibodies and the sever-
mented the presence of macrophages and multinucleated giant ity of RPE pathology. An intact immune response was required
cells, mainly associated with vascular channels and breaks in for CEP-induced RPE changes since Rag-deficient mice which
Bruch’s membrane (Fig. 25.3).34–39 It is unclear whether the mac- lack mature T and B cells did not develop RPE pathology after
rophages contribute to the development of AMD lesions or if CEP immunization. Monocytes and macrophages were observed
they accumulate as an adaptive response to injury associated in the interphotoreceptor matrix. Hollyfield et  al. suggested
with AMD. Some evidence suggests the relationship between the macrophages probably did not initiate the pathology since
oxidative stress and inflammation in AMD may be mediated many lesions were observed without associated macrophages.
in part by macrophages similar to the pathogenesis of athero- They hypothesized that macrophages could be attracted by the
sclerotic plaques. Oxidation of lipoproteins exposes phosphor- release of cytokines from lysed cells and participate in debris
ylcholine (PC), which is also present on the membranes of removal, as supported by the presence of melanin-laden mac-
apoptotic cells. PC can be bound by CRP, an opsonic molecule rophages in the RPE lesions.
noted an age-dependent accumulation of lipofuscin in subretinal
Microglia and AMD microglia, possibly related to phagocytosis of photoreceptor
556 Microglia are specialized tissue macrophages which constitute outer segments. Shen et al. reported that naloxone, a potent
the main resident immune cells in the neuroretina. In concert microglial inhibitor, ameliorated AMD-like retinal lesions in
with the RPE cells, choroidal macrophages, and dendritic cells, Ccl2/Cx3cr1 double deficient with rd8 background mice via sup-
the microglia play an important role in maintaining normal pression of proinflammatory molecules of nitric oxide (NO),
Section 2

retinal immune function as well as supporting the surrounding TNF-α, and IL-β.53 Under experimentally induced photo-
cells composing the neuroretina. In their quiescent state, oxidative stress in rats exposed to blue light, Rutar et al. used
microglia are inconspicuous stellate cells that produce anti- microarray expression analyses to show that light-induced
inflammatory IL-10 in response to RPE-derived cytokines. injury induced differential expression of complement-related
Microglia can rapidly transform into enlarged, activated microg- genes, including upregulation of complement activators and
lia in response to microenvironment signals of infection, neuro- receptors.54 They also detected complement C3-expressing
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

nal injury, ischemia, or oxidative stress.45–49 Quiescent and microglia in the outer nuclear layer and in the subretinal spaces
activated microglia secrete polypeptide neurotrophic factors, adjacent to areas of injury.
including brain-derived neurotrophic factor, ciliary neurotrophic
factor (CNTF), nerve growth factor, neurotrophin-3, and basic Other inflammatory-related molecules and pathways
fibroblast growth factor which influence neuron physiology and The inflammatory response to oxidative stress involves many
survival.47 other molecules and pathways which may represent future ther-
The sub-RPE space is immunologically protected by the outer apeutic targests. Neuroprotectin D1 (NPD1) is an endogenous
blood–retinal barrier and normally devoid of immune cells. It is anti-inflammatory mediator which is derived from the essential
hypothesized that as lipofuscin and oxidized material accumu- omega-3 polyunsaturated fatty acid, DHA.55,56 Multiple studies
late in the RPE and Bruch’s membrane of the aging eye, the RPE have shown the RPE generates NPD1 in response to oxidative
may be stimulated to produce inflammatory cytokines and che- stress.57–60 NPD1 promotes RPE survival by upregulating anti-
mokines which activate microglia and stimulate sub-RPE migra- apoptotic genes and downregulating antiapoptotic genes57,58,61
tion.48,50 Under conditions of chronic inflammation and activation, and also inhibits cytokine-mediated proinflammatory gene
advanced glycation end products and oxidized lipids bind induction.57,62
microglia and promote further inflammatory response. The The PPARs are transcription factors which are activated by
microglia may have a detrimental effect in the neuroretina as fatty acids. They exist in three forms, PPAR-γ, -α, and -δ (also
they perpetuate the inflammation and injury cycle by secreting known as -β, NUC-1, or FAAR), which differ in tissue distri-
molecules that kill neighboring cells. Ma et al. cocultured acti- bution and ligand specificity.63 PPAR-γ is expressed by normal
vated retinal microglia with primary cultures of mouse RPE cells human RPE and upregulated in response to oxidative stress,
and measured lower levels of junctional proteins and RPE-65 as including in AMD lesions.64 PPAR-γ activation may protect
well as prominent structural disorganization and increased pro- against oxidative injury through upregulation of antioxidative
liferation of the RPE cells in comparison to controls.51 They also enzymes,65,66 downregulation of chemokines such as CXCR4
found increased RPE expression and secretion of multiple pro- and CCL2,67,68 and modulation of microglial activation.69 Phar-
inflammatory cytokines, including IL-1β, TNF-α, IL-6, and macologic agonists for both PPAR-α (fibrates) and -γ (thiazoli-
granulocyte–macrophage colony-stimulating factor; the anti- dinediones) have been investigated as potential treatments
inflammatory cytokine IL-10 was not markedly changed. Cocul- for ocular neovascularization through inhibition of the VEGF
ture with activated microglia also fostered a proangiogenic pathway63,70–72; however, due to the overlap between many
environment with increased mRNA levels of the matrix metal- components of the pathways of angiogenesis and inflamma-
loproteinases MMP1 and MMP9 as well as protein levels of tion, further clinical studies are warranted.73
MMP1, MMP2, MMP9, and VEGF. Finally, Ma et al. evaluated Nuclear erythroid-2-related factor (NRF2) is another transcrip-
the in vivo effects of subretinal activated microglia by trans- tion factor which plays an important role in the cellular response
planting them into the subretinal space of adult wild-type mice. to oxidative stress and may be regulated by PPAR-γ.68 NRF2
When the animals were euthanized and examined 4 days after regulates antioxidant gene expression and complement activa-
the subretinal injections, there were large and prominent choroi- tion to prevent further inflammation-mediated oxidative stress.74
dal neovascular membranes in these mice. To date, most studies have focused on the role of NRF2 in lung
Both human and mouse in vivo studies provide evidence of disease (modulation of macrophage activation in response to
microglial migration in conditions of aging or stress. Gupta et al. cigarette smoke)75 and neuroinflammation (downmodulation of
examined three AMD eyes with geographic atrophy and two activated microglia).76,77
normal donor eyes.52 They found quiescent microglia in the inner Heat shock proteins (Hsps) prevent harmful protein aggrega-
layers of the normal retinas; in contrast, the microglia were tion by serving as molecular chaperones that bind damaged
balloon-shaped and present in the inner and outer nuclear intracellular proteins and assist in the processes of repair or
layers, often in association with degenerate rods in the AMD removal and degradation.78 Hsps participate in chaperone-
eyes. The activated microglia were also observed in the subreti- mediated autophagy which is an oxidative stress-activated
nal space of the AMD eyes; their rhodopsin-positive cytoplasmic lysosomal pathway that results in degradation of cytoplasmic
inclusions were evidence of their role in phagocytizing photore- material and organelles. Some Hsps also have cytoprotective
ceptor debris. The findings suggest microglial migration occurs effects through modulation of apoptosis and stabilization of
in AMD. cytoskeletal proteins.79,80 Age-related decreases in Hsp activity
Xu et al. studied the mouse retina and observed that the and autophagy may contribute to the accumulation of aggrega-
numbers of subretinal microglia increased with age.50 They also tions of oxidized proteins and lipids (lipofuscin) in RPE cells.81–83
Oxidative stress is known to increase the expression of increased oxidative stress.112,113 Oxidative injury causes endothe-
many of the Hsps, especially Hsp27 (also known as HspB1) lial cell apoptosis and death with resultant obliteration of retinal
and Hsp 70.80,84,85 Several studies have measured higher levels vessels.114 In addition to the inherent pro-oxidative characteris- 557
of Hsps, including Hsp27, αA- and αB-crystallin in drusen, tics of the retina discussed earlier in this section, relative to the
Bruch’s membrane, and choroid in AMD eyes compared to adult, the neonatal retina contains more free iron to catalyze

Chapter 25
normal controls.86,87 It is hypothesized that AMD eyes have oxidizing reactions and fewer antioxidants. In the neonatal
higher levels of Hsps as an attempt to adapt to increased oxi- period, the cytochrome C oxidase and NO synthase inflamma-
dative stress. However, the Hsp αB-crystallin may actually tory pathways are highly active, leading to production of ROIs
contribute to angiogenesis through modulation of VEGF-A, as and prostaglandins. ROIs react with NO to produce reactive
suggested by the work of Kase et  al.88 nitrogen species which also contribute to retinal vascular
degeneration.115–118 Lipid peroxidation of endothelial cell mem-
Genes and inflammation in AMD
branes and polyunsaturated fatty acids in the retina results in

Inflammatory Response and Mediators in Retinal Injury


Multiple genes related to inflammation have been linked to
further damage. Peroxidation of arachidonic acid produces iso-
AMD risk and may provide evidence for the roles of oxidative
prostanes that have indirect cytotoxic effects on the retinal vas-
stress and inflammation.89 Variations in the complement factor
cular endothelium.119 Lipid peroxidation also produces several
H gene (CFH) were the first genetic changes associated with
proinflammatory mediators including lysophosphatidic acid,
AMD.90–93 CFH is a regulatory protein which suppresses forma-
platelet-activated factor, and the eicosanoids derived from
tion of the C3 cleavage enzyme and inhibits the alternative
omega-3 and omega-6 long-chain polyunsaturated fatty acids.120
pathway of complement activation. Variations in CFH may limit
Oxidative stress and ROIs may also play an important role in
the protein’s ability to inhibit complement activation and con-
neovascularization, one of the major complications of ROP, by
tribute to the development of a chronic inflammatory state. A
triggering angiogenesis through VEGF and other signaling path-
SNP in the DNA repair gene ERCC6 has a modest association
ways, including Janus kinase-signal transducer and activator of
with AMD risk; however, when present in combination with a
transcription (JAK/STAT).121–124
SNP in the CFH intron there is a strong correlation with AMD.94
Polymorphisms in other complement genes, including factors C3 Anti-inflammatory and antioxidant therapies in AMD
and I, have also been linked with increased risk of AMD.95–97 and ROP
Conversely, variants associated with factors B, C2, and C7, as As research studies continue to clarify and expand our knowl-
well as SERPING1, which encodes complement component 1 edge of the roles of oxidative stress and inflammation in the
inhibitor, may be protective against AMD.98–100 pathogenesis of AMD and ROP, the rationale for therapies tar-
Multiple chemokine and cytokine gene variants have also been geting these pathways grows. A large randomized and con-
associated with increased risk of AMD. CX3CR1 is a chemokine trolled clinical trial, the Age-Related Eye Disease Study (AREDS),
receptor found on macrophages, microglia, T cells, and astro- demonstrated the benefit of vitamin and mineral supplementa-
cytes which mediates leukocyte migration and activation.101,102 tion in preventing progression from early to advanced AMD.125
SNPs in CX3CR1 have been associated with AMD risk. Com- Although the exact mechanism is unknown, it is hypothesized
badière et al. showed that monocytes from AMD patients who that the AREDS vitamin formulation counteracts oxidative
were homozygous for a SNP in CX3CR1 had impaired chemo- stresses in the retina. Several studies suggested supplementation
taxis and suggested a possible mechanism for the subretinal with vitamin E, a potent scavenger of ROIs, suppressed the
accumulation of microglia frequently observed in AMD.103 Other development of severe ROP disease in premature infants;
inflammation-related genes that may be linked to AMD risk however, subsequent trials have not reproduced these results.112
include IL-8, TLR3, and TLR4.104–106 Dietary enrichment with anti-inflammatory lipids is another
Finally, data from several studies suggest variants in two potential direction for disease prevention in both AMD and
additional genes in an AMD susceptibility locus on chromo- ROP.112,126 In AMD, multiple areas of evidence support the use
some 10q26 may play a role in AMD risk. ARMS2 has been of systemic and local anti-inflammatory therapies for disease
localized to mitochondria which are a site of ROI generation.107 prevention or in combination with anti-VEGF medications as
Multiple studies have shown correlations between an ARMS2 treatment for neovascular disease. Several AMD clinical trials
variant and advanced AMD.89,108,109 Of note, Wang et  al. dem- have demonstrated the benefits of immunomodulatory medica-
onstrated an especially strong risk in AMD patients with ele- tions such as methotrexate, sirolimus, and daclizumab; ongoing
vated serum levels of the inflammatory markers CRP, IL-6, studies are now investigating complement component inhibi-
and soluble ICAM-1.110 Another gene on chromosome 10q26, tors.127 In the future, treatment and prevention strategies for
HtrA1, is a stress-inducible member of the family of heat shock retinal diseases should be multifactorial in order to address
serine proteases. Multiple AMD-associated SNPs have been the complex and interacting pathways associated with aging,
described, although there is some controversy whether HtrA1 oxidative stress, and inflammation.
or ARMS2 is the true gene associated with AMD.89,111
Oxidative stress and inflammation in retinopathy TRAUMA
of prematurity Inflammation also plays a role in physical retinal injury such as
Oxidative stress also plays a role in ROP. Development of the rhegmatogenous retinal detachment (RRD) and intraocular
human retinal vasculature continues through the 40th week of foreign body. Retinal vasculitis may develop after RRD (Fig.
gestation; therefore, premature infants are born with an incom- 25.4). Inflammatory cells, including foreign-body giant cells, are
plete retinal blood supply. Inability to autoregulate the blood often present surrounding an intraocular foreign body. Separa-
flow to the retina and choroid combined with external oxygen tion of the neuroretina from the RPE disrupts the normal homeo-
supplementation leads to exaggerated retinal oxygenation and stasis of the retina. Studies of experimental RRD animal models
eliminates photoreceptor apoptosis perhaps by preventing
macrophage/microglia generation of ROIs.142 In another experi-
558 V R ment, Nakazawa et al. administered intraperitoneal dexametha-
sone and suppressed upregulation of TNF-α and photoreceptor
R
degeneration after experimental RRD.143
Photoreceptor loss can lead to irreversible vision loss even
Section 2

after retinal reattachment; however, significant numbers of pho-


toreceptors may survive for days to weeks after RRD. In neural
systems, IL-6 could actually enhance cell survival by preventing
apoptosis and therefore reducing photoreceptor death after
* RRD. Zacks et al. showed that genes in the IL-6 pathway are
upregulated within 24 hours of experimental RRD.144 Transcript
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

and protein levels of IL-6 as well as CNTF, a member of the IL-6


family with neuroprotective effects, were increased in experi-
mentally detached retina.144 They also measured increased tran-
SR
scription of two pathways that might have beneficial effects on
photoreceptor survival, transforming growth factor-β (antiapop-
tosis) and aryl hydrocarbon receptor (antioxidative stress).144 In
Fig. 25.4 Traumatic retinal detachment. Inflammatory cells (arrows) another study, Chong et al. showed that absence of IL-6 function
are surrounding a small retinal vessel in a totally detached retina (R) resulted in increased photoreceptor apoptosis after experimental
and the vitreous (V) due to trauma. Fresh hemorrhages (asterisk) are RRD in both IL-6 double-knockout as well as in wild-type mice
seen in the vitreous. The subretinal space (SR) contains serous
material. (Hematoxylin and eosin, ×25.) treated with anti-IL-6 antibody.145 Subretinal injection of IL-6
immediately or up to 2 weeks after creation of the experimental
RD in wild-type Norway rats resulted in preservation of photo-
receptors 4 weeks later, but this effect had disappeared at 8
and human surgical specimens provide insight into the cellular weeks. Finally, intraperitoneal injections of minocycline, a semi-
and molecular processes that occur in the detached neuroretina. synthetic tetracycline antibiotic with antiapoptotic and anti-
In the earliest phase after detachment, the photoreceptor outer inflammatory effects, resulted in preservation of the photoreceptor
segments become disorganized and slough off in the subretinal inner and outer segments, decreased photoreceptor apoptosis,
space where they are phagocytized and removed by infiltrating and decreased glial activation.146
macrophages and RPE cells.128–130 Subsequently, some photore- Among the complications of RRD, PVR is associated with an
ceptor cells become apoptotic131–133 and are phagocytized by mac- increased risk of recurrent detachment and worse visual
rophages and activated microglia infiltrating the subretinal outcome. Chronic or poorly regulated inflammation may be cor-
space.128,134 Other changes associated with RRD include prolifera- related with the development of PVR. Multiple studies of vitre-
tion of Müller cells, RPE, astrocytes, microglia, pericytes, and ous and subretinal fluid specimens from human RRD patients
capillary endothelial cells,135 as well as Müller cell hypertrophy have demonstrated increased levels of inflammatory cytokines
and retinal gliosis. RPE cells frequently undergo hypertrophy, and related molecules (i.e., IL-1, IL-6, TNF-α, interferon-γ,
hyperplasia, hypotrophy or atrophy, and migrate to the subreti- ICAM-1) and growth factors (i.e., VEGF, fibroblast growth factor,
nal space.130,136,137 Over time, unless the retina is reattached, and hepatocyte growth factor) in association with PVR.147–159
further photoreceptor atrophy accompanies glial scarring, Examination of PVR membranes also has revealed macrophages,
subretinal fibrosis, proliferative vitreoretinopathy (PVR), and lymphocytes, MHC class II positive cells, as well as deposits of
epiretinal membrane formation.138,139 immunoglobulins, complement factors, and cellular adhesion
Several studies provide evidence of an early inflammatory molecules.160 In an attempt to decrease the incidence of PVR,
response to RRD. As early as 1 hour after experimental RRD, multiple adjunctive treatments have been investigated. Two
Nakazawa et al. measured increased mRNA and protein levels antiproliferative agents, 5-fluorouracil and daunorubicin, were
of TNF-α, IL-1β, and CCL2.140 Hollborn et al. analyzed mRNA administered intravitreally during or at the completion of vitrec-
levels 24 hours after experimental RRD and identified upregula- tomy for PVR with varied success depending on the outcomes
tion of genes related to inflammation and immune response, measured.160,161 Despite several clinical trials, neither adjunctive
antioxidants and metal homeostasis, intracellular proteolysis, agent is currently in wide use.
and blood coagulation/fibrinolysis.141 The inflammation-related An intraocular foreign body may occur as a result of penetrat-
genes included CCL2, MHC class I and II molecules, interferon- ing ocular trauma or through an iatrogenic mechanism such as
regulatory factors, TLR-2, TLR-4, and IL-18. In another gene the use of a silicone oil tamponade for repair of a retinal detach-
expression study 7 days after experimental RRD, Zacks reported ment. Although some intraocular foreign bodies such as acrylic
multiple upregulated genes, including those involved in inflam- plastic may be tolerated with minimal inflammation, others such
mation (i.e., interferon pathway-related), immune complexes as organic material may elicit immediate and intense inflamma-
(i.e., CFH), antigen presentation, acute stress response (i.e., tion. Surgical specimens are rarely available for pathologic
crystallins), and metabolism.129 examination; however, a few case reports in the literature have
High levels of CCL2 are produced by Müller cells after shed light on the intraocular inflammatory responses to foreign
RRD.140,142 CCL2 mediates photoreceptor apoptosis and the bodies. Lee et al. described a focal posttraumatic choroidal gran-
macrophage/microglial response to RRD. Blocking or deleting ulomatous inflammation that presented as a subretinal mass 19
expression of CCL2 in an experimental model of RRD nearly weeks after the eye sustained a penetrating injury with a metal
projectile.162 The surgically excised mass consisted of fibrovascu- 11. Yoshimura T, Sonoda KH, Sugahara M, et al. Comprehensive analysis of
inflammatory immune mediators in vitreoretinal diseases. PLoS ONE 2009;4:
lar tissue with reactive vascularization, histiocytes, lymphocytes, e8158.
and occasional giant cells. Several cases were previously 12. Noma H, Funatsu H, Mimura T, et al. Vitreous levels of interleukin-6 and 559
vascular endothelial growth factor in macular edema with central retinal vein
described in the literature; all were associated with penetrating occlusion. Ophthalmology 2009;116:87–93.
trauma and several had identifiable foreign material.163 They 13. Noma H, Funatsu H, Mimura T, et al. Aqueous humor levels of vasoactive

Chapter 25
differ from sympathetic ophthalmia due to the absence of inflam- molecules correlate with vitreous levels and macular edema in central retinal
vein occlusion. Eur J Ophthalmol 2010;20:402–9.
mation in the noninjured eye. 14. Barrett JC, Clayton DG, Concannon P, et al. Genome-wide association study
Wickham et al. examined enucleated globes that had a history and meta-analysis find that over 40 loci affect risk of type 1 diabetes.
Nat Genet 2009;41:703–7.
of retinal detachment surgery with silicone oil.164 They reported 15. Cooper JD, Walker NM, Smyth DJ, et al. Follow-up of 1715 SNPs from the
the presence of silicone oil in the iris, ciliary body, retina, optic Wellcome Trust Case Control Consortium genome-wide association study in
type 1 diabetes families. Genes Immun 2009;10(Suppl 1):S85–94.
nerve, trabecular meshwork, and epiretinal membranes. Macro- 16. Roy MS, Hallman DM, Fu YP, et al. Assessment of 193 candidate genes for
phages were observed in close association with the silicone oil, retinopathy in African Americans with type 1 diabetes. Arch Ophthalmol

Inflammatory Response and Mediators in Retinal Injury


2009;127:605–12.
and the number of macrophages generally reflected the degree 17. Maier R, Steinbrugger I, Haas A, et al. Role of inflammation-related gene poly-
of silicone oil distribution. The macrophages often contained morphisms in patients with central retinal vein occlusion. Ophthalmology
phagocytized silicone oil. Focal areas of retinal infiltration by 2011;119:1125–9.
18. Sfikakis PP, Grigoropoulos V, Emfietzoglou I, et al. Infliximab for diabetic
macrophages occurred at retinectomy sites and were associated macular edema refractory to laser photocoagulation: a randomized, double-
with subretinal silicone oil. T and B lymphocytes were located blind, placebo-controlled, crossover, 32-week study. Diabetes Care 2010;33:
1523–8.
only in the inflammatory membranes (i.e., cyclitic, PVR, epireti- 19. Behl Y, Krothapalli P, Desta T, et al. Diabetes-enhanced tumor necrosis factor-
nal, and glaucomatous) and among the perivascular inflamma- alpha production promotes apoptosis and the loss of retinal microvascular
cells in type 1 and type 2 models of diabetic retinopathy. Am J Pathol
tion. In general, the distribution and number of lymphocytes did 2008;172:1411–8.
not differ from the control eyes with a history of RRD or trauma. 20. Joussen AM, Doehmen S, Le ML, et al. TNF-alpha mediated apoptosis
The authors concluded that intraocular silicone oil elicits a local- plays an important role in the development of early diabetic retinopathy and
long-term histopathological alterations. Mol Vis 2009;15:1418–28.
ized inflammatory response mediated by macrophages. 21. Aveleira CA, Lin CM, Abcouwer SF, et al. TNF-alpha signals through
PKCzeta/NF-kappaB to alter the tight junction complex and increase retinal
CONCLUSION endothelial cell permeability. Diabetes 2010;59:2872–82.
22. Ibrahim AS, El-Shishtawy MM, Pena A, Jr., et al. Genistein attenuates retinal
inflammation associated with diabetes by targeting of microglial activation.
Acute and chronic inflammatory processes are intricately Mol Vis 2010;16:2033–42.
involved in ischemia, oxidative stress, and trauma in the retina. 23. Bandello F, Iacono P, Battaglia Parodi M. Treatment options for diffuse
The inflammatory response may represent a double-edged diabetic macular edema. Eur J Ophthalmol 2010;21:45–50.
24. Diabetic Retinopathy Clinical Research Network. A randomized trial compar-
sword with both harmful and beneficial effects.165 Acute inflam- ing intravitreal triamcinolone acetonide and focal/grid photocoagulation for
mation is often an essential and helpful response to maintain diabetic macular edema. Ophthalmology 2008;115:1447–9, 1449. e1–10.
25. Wong TY, Scott IU. Clinical practice. Retinal-vein occlusion. N Engl J Med
normal function; for example, macrophages can remove necrotic 2010;363:2135–44.
or apoptotic cells and promote wound healing. However, pro- 26. Jonas JB, Akkoyun I, Kamppeter B, et al. Branch retinal vein occlusion treated
by intravitreal triamcinolone acetonide. Eye (Lond) 2005;19:65–71.
longed or dysregulated chronic inflammation can magnify and 27. Scott IU, Ip MS, VanVeldhuisen PC, et al. A randomized trial comparing the
perpetuate damage of the retina, causing atrophic chorioretinal efficacy and safety of intravitreal triamcinolone with standard care to treat
vision loss associated with macular edema secondary to branch retinal vein
scarring or PVR. Since inflammation is only one component of occlusion: the Standard care vs Corticosteroid for Retinal Vein occlusion
most retinal diseases, current anti-inflammatory therapies (SCORE) study report 6. Arch Ophthalmol 2009;127:1115–28.
merely provide palliative treatment. As we better understand 28. Haller JA, Bandello F, Belfort Jr R, et al. Randomized, sham-controlled trial of
dexamethasone intravitreal implant in patients with macular edema due to
the inflammatory pathways and mechanisms, we will develop retinal vein occlusion. Ophthalmology 2010;117:1134–46 e3.
more effective prevention and treatment strategies to manage all 29. Beatty S, Koh H, Phil M, et al. The role of oxidative stress in the pathogenesis
of age-related macular degeneration. Surv Ophthalmol 2000;45:115–34.
retinal diseases. 30. Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB
signaling. Cell Res 2011;21:103–15.
REFERENCES 31. Curcio CA, Johnson M, Huang JD, et al. Apolipoprotein B-containing lipopro-
1. Cohen LH, Noell WK. Relationships between visual function and metabolism. teins in retinal aging and age-related macular degeneration. J Lipid Res
In: Graymore CN, editor. Biochemistry of the retina. New York: Academic 2010;51:451–67.
Press; 1965. 32. Crabb JW, Miyagi M, Gu X, et al. Drusen proteome analysis: an approach to
2. Young RW, Bok D. Participation of the retinal pigment epithelium in the rod the etiology of age-related macular degeneration. Proc Natl Acad Sci U S A
outer segment renewal process. J Cell Biol 1969;42:392–403. 2002; 99:14682–7.
3. Wilson TM, Strang R, Wallace J, et al. The measurement of the choroidal blood 33. Yuan X, Gu X, Crabb JS, et al. Quantitative proteomics: comparison of
flow in the rabbit using 85-krypton. Exp Eye Res 1973;16:421–5. the macular Bruch membrane/choroid complex from age-related macular
4. Miyamoto K, Khosrof S, Bursell SE, et al. Prevention of leukostasis and degeneration and normal eyes. Mol Cell Proteomics 2010;9:1031–46.
vascular leakage in streptozotocin-induced diabetic retinopathy via intercel- 34. Killingsworth MC, Sarks JP, Sarks SH. Macrophages related to Bruch’s
lular adhesion molecule-1 inhibition. Proc Natl Acad Sci U S A 1999;96: membrane in age-related macular degeneration. Eye (Lond) 1990;4:613–21.
10836–41. 35. Green WR, Enger C. Age-related macular degeneration histopathologic
5. Adamis AP, Berman AJ. Immunological mechanisms in the pathogenesis of studies. The 1992 Lorenz E. Zimmerman Lecture. Ophthalmology 1993;100:
diabetic retinopathy. Semin Immunopathol 2008;30:65–84. 1519–35.
6. Cusick M, Chew EY, Chan CC, et al. Histopathology and regression of retinal 36. Dastgheib K, Green WR. Granulomatous reaction to Bruch’s membrane in
hard exudates in diabetic retinopathy after reduction of elevated serum lipid age-related macular degeneration. Arch Ophthalmol 1994;112:813–8.
levels. Ophthalmology 2003;110:2126–33. 37. Grossniklaus HE, Cingle KA, Yoon YD, et al. Correlation of histologic
7. Meleth AD, Agron E, Chan CC, et al. Serum inflammatory markers in diabetic 2-dimensional reconstruction and confocal scanning laser microscopic
retinopathy. Invest Ophthalmol Vis Sci 2005;46:4295–301. imaging of choroidal neovascularization in eyes with age-related maculopa-
8. Green WR, Chan CC, Hutchins GM, et al. Central retinal vein occlusion: a thy. Arch Ophthalmol 2000;118:625–9.
prospective histopathologic study of 29 eyes in 28 cases. Retina 1981;1: 38. Grossniklaus HE, Miskala PH, Green WR, et al. Histopathologic and ultra-
27–55. structural features of surgically excised subfoveal choroidal neovascular
9. Kaur C, Foulds WS, Ling EA. Blood–retinal barrier in hypoxic ischaemic lesions: submacular surgery trials report no. 7. Arch Ophthalmol 2005;123:
conditions: basic concepts, clinical features and management. Prog Retin Eye 914–21.
Res 2008;27:622–47. 39. Cao X, Shen D, Patel MM, et al. Macrophage polarization in the maculae of
10. Goldberg RB. Cytokine and cytokine-like inflammation markers, endothelial age-related macular degeneration: a pilot study. Pathol Int 2011;61:528–35.
dysfunction, and imbalanced coagulation in development of diabetes and its 40. Chang MK, Binder CJ, Torzewski M, et al. C-reactive protein binds to both
complications. J Clin Endocrinol Metab 2009;94:3171–82. oxidized LDL and apoptotic cells through recognition of a common ligand:
phosphorylcholine of oxidized phospholipids. Proc Natl Acad Sci U S A 71. Del V, Cano M, Gehlbach PL. PPAR-alpha ligands as potential therapeutic
2002;99:13043–8. agents for wet age-related macular degeneration. PPAR Res 2008;2008:821592.
41. Hazen SL, Chisolm GM. Oxidized phosphatidylcholines: pattern recognition 72. Pershadsingh HA, Moore DM. PPARgamma agonists: potential as therapeu-
560 ligands for multiple pathways of the innate immune response. Proc Natl Acad tics for neovascular retinopathies. PPAR Res 2008;2008:164273.
Sci U S A 2002;99:12515–7. 73. Herzlich AA, Tuo J, Chan CC. Peroxisome proliferator-activated receptor and
42. Kamei M, Yoneda K, Kume N, et al. Scavenger receptors for oxidized lipo- age-related macular degeneration. PPAR Res 2008;2008:389507.
protein in age-related macular degeneration. Invest Ophthalmol Vis Sci 2007; 74. Cano M, Thimmalappula R, Fujihara M, et al. Cigarette smoking, oxidative
48:1801–7. stress, the anti-oxidant response through Nrf2 signaling, and age-related
Section 2

43. Hollyfield JG, Bonilha VL, Rayborn ME, et al. Oxidative damage-induced macular degeneration. Vision Res 2010;50:652–64.
inflammation initiates age-related macular degeneration. Nat Med 2008;14: 75. Rangasamy T, Cho CY, Thimmulappa RK, et al. Genetic ablation of
194–8. Nrf2 enhances susceptibility to cigarette smoke-induced emphysema in mice.
44. Hollyfield JG, Perez VL, Salomon RG. A hapten generated from an oxidation J Clin Invest 2004;114:1248–59.
fragment of docosahexaenoic acid is sufficient to initiate age-related macular 76. Innamorato NG, Rojo AI, García-Yagüe AJ, et al. The transcription factor Nrf2
degeneration. Mol Neurobiol 2010;41:290–8. is a therapeutic target against brain inflammation. J Immunol 2008;181:
45. Ng TF, Streilein JW. Light-induced migration of retinal microglia into the 680–9.
subretinal space. Invest Ophthalmol Vis Sci 2001;42:3301–10. 77. Rojo AI, Innamorato NG, Martín-Moreno AM, et al. Nrf2 regulates microglial
Basic Mechanisms of Injury in the Retina

46. Chen L, Yang P, Kijlstra A. Distribution, markers, and functions of retinal dynamics and neuroinflammation in experimental Parkinson’s disease.
Basic Science and Translation to Therapy

microglia. Ocul Immunol Inflamm 2002;10:27–39. Glia 2010;58:588–98.


47. Langmann T. Microglia activation in retinal degeneration. J Leukoc Biol 78. Kaarniranta K, Salminen A, Eskelinen EL, et al. Heat shock proteins as
2007;81:1345–51. gatekeepers of proteolytic pathways – implications for age-related macular
48. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin degeneration (AMD). Ageing Res Rev 2009;8:128–39.
Eye Res 2009;28:348–68. 79. Alge CS, Priglinger SG, Neubauer AS, et al. Retinal pigment epithelium is
49. Karlstetter M, Ebert S, Langmann T. Microglia in the healthy and degenerat- protected against apoptosis by alphaB-crystallin. Invest Ophthalmol Vis Sci
ing retina: insights from novel mouse models. Immunobiology 2010;215: 2002;43:3575–82.
685–91. 80. O’Reilly AM, Currie RW, Clarke DB. HspB1 (Hsp 27) expression and
50. Xu H, Chen M, Manivannan A, et al. Age-dependent accumulation of neuroprotection in the retina. Mol Neurobiol 2010;42:124–32.
lipofuscin in perivascular and subretinal microglia in experimental mice. 81. Louie JL, Kapphahn RJ, Ferrington DA. Proteasome function and protein
Aging Cell 2008;7:58–68. oxidation in the aged retina. Exp Eye Res 2002;75:271–84.
51. Ma W, Zhao L, Fontainhas AM, et al. Microglia in the mouse retina alter the 82. Kapphahn RJ, Bigelow EJ, Ferrington DA. Age-dependent inhibition of pro-
structure and function of retinal pigmented epithelial cells: a potential cellular teasome chymotrypsin-like activity in the retina. Exp Eye Res 2007;84:
interaction relevant to AMD. PLoS ONE 2009;4:e7945. 646–54.
52. Gupta N, Brown KE, Milam AH. Activated microglia in human retinitis 83. Jung T, Bader N, Grune T. Lipofuscin: formation, distribution, and metabolic
pigmentosa, late-onset retinal degeneration, and age-related macular degen- consequences. Ann N Y Acad Sci 2007;1119:97–111.
eration. Exp Eye Res 2003;76:463–71. 84. Strunnikova N, Baffi J, Gonzalez A, et al. Regulated heat shock protein 27
53. Shen D, Cao X, Zhao L, et al. Naloxone ameliorates retinal lesions in Ccl2/ expression in human retinal pigment epithelium. Invest Ophthalmol Vis Sci
Cx3cr1 double-deficient mice via modulation of microglia. Invest Ophthalmol 2001;42:2130–8.
Vis Sci 2011;52:2897–904. 85. Yaung J, Jin M, Barron E, et al. alpha-Crystallin distribution in retinal pigment
54. Rutar M, Natoli R, Kozulin P, et al. Analysis of complement expression in epithelium and effect of gene knockouts on sensitivity to oxidative stress.
light-induced retinal degeneration: Synthesis and deposition of C3 by Mol Vis 2007;13:566–77.
microglia/macrophages is associated with focal photoreceptor degeneration. 86. Nakata K, Crabb JW, Hollyfield JG. Crystallin distribution in Bruch’s
Invest Ophthalmol Vis Sci 2011;52:5347–58. membrane–choroid complex from AMD and age-matched donor eyes.
55. Bazan NG. Homeostatic regulation of photoreceptor cell integrity: signifi- Exp Eye Res 2005;80(6):821–6.
cance of the potent mediator neuroprotectin D1 biosynthesized from docosa- 87. Pons M, Cousins SW, Csaky KG, et al. Cigarette smoke-related hydroquinone
hexaenoic acid: the Proctor Lecture. Invest Ophthalmol Vis Sci 2007;48: induces filamentous actin reorganization and heat shock protein 27 phos-
4866–81. phorylation through p38 and extracellular signal-regulated kinase 1/2 in
56. Serhan CN, Yacoubian S, Yang R. Anti-inflammatory and proresolving lipid retinal pigment epithelium: implications for age-related macular degenera-
mediators. Annu Rev Pathol 2008;3:279–312. tion. Am J Pathol 2010;177:1198–213.
57. Mukherjee PK, Marcheselli VL, Serhan CN, et al. Neuroprotectin D1: a doco- 88. Kase S, He S, Sonoda S, et al. alphaB-crystallin regulation of angiogenesis by
sahexaenoic acid-derived docosatriene protects human retinal pigment epi- modulation of VEGF. Blood 2010;115:3398–406.
thelial cells from oxidative stress. Proc Natl Acad Sci U S A 2004;101:8491–6. 89. Ding X, Patel M, Chan CC. Molecular pathology of age-related macular
58. Mukherjee PK, Marcheselli VL, Barreiro S, et al. Neurotrophins enhance degeneration. Prog Retin Eye Res 2009;28:1–18.
retinal pigment epithelial cell survival through neuroprotectin D1 signaling. 90. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the
Proc Natl Acad Sci U S A 2007;104:13152–7. complement regulatory gene factor H (HF1/CFH) predisposes individuals to
59. Mukherjee PK, Marcheselli VL, de Rivero Vaccari JC, et al. Photoreceptor age-related macular degeneration. Proc Natl Acad Sci U S A 2005;102:
outer segment phagocytosis attenuates oxidative stress-induced apoptosis 7227–32.
with concomitant neuroprotectin D1 synthesis. Proc Natl Acad Sci U S A 91. Edwards AO, Ritter 3rd R, Abel KG, et al. Complement factor H polymor-
2007;104:13158–63. phism and age-related macular degeneration. Science 2005;308:421–4.
60. Bazan NG. Neurotrophins induce neuroprotective signaling in the retinal 92. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant
pigment epithelial cell by activating the synthesis of the anti-inflammatory increases the risk of age-related macular degeneration. Science 2005;308:
and anti-apoptotic neuroprotectin D1. Adv Exp Med Biol 2008;613:39–44. 419–21.
61. Antony R, Lukiw WJ, Bazan NG. Neuroprotectin D1 induces dephosphoryla- 93. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in
tion of Bcl-xL in a PP2A-dependent manner during oxidative stress and pro- age-related macular degeneration. Science 2005;308:385–9.
motes retinal pigment epithelial cell survival. J Biol Chem 2010;285:18301–8. 94. Tuo J, Ning B, Bojanowski CM, et al. Synergic effect of polymorphisms in
62. Marcheselli VL, Hong S, Lukiw WJ, et al. Novel docosanoids inhibit brain ERCC6 5’ flanking region and complement factor H on age-related macular
ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory degeneration predisposition. Proc Natl Acad Sci U S A 2006;103:9256–61.
gene expression. J Biol Chem 2003;278:43807–17. 95. Yates JR, Sepp T, Matharu BK, et al. Complement C3 variant and the risk of
63. Malchiodi-Albedi F, Matteucci A, Bernardo A, et al. PPAR-gamma, microglial age-related macular degeneration. N Engl J Med 2007;357:553–61.
cells, and ocular inflammation: new venues for potential therapeutic 96. Fagerness JA, Maller JB, Neale BM, et al. Variation near complement factor
approaches. PPAR Res 2008;2008:295784. I is associated with risk of advanced AMD. Eur J Hum Genet 2009;17:
64. Herzlich AA, Ding X, Shen D, et al. Peroxisome proliferator-activated receptor 100–4.
expression in murine models and humans with age-related macular degenera- 97. Thakkinstian A, McKay GJ, McEvoy M, et al. Systematic review and meta-
tion. Open Biol J 2009;2:141–8. analysis of the association between complement component 3 and age-related
65. Garg TK, Chang JY. Oxidative stress causes ERK phosphorylation and cell macular degeneration: a huge review and meta-analysis. Am J Epidemiol
death in cultured retinal pigment epithelium: prevention of cell death by 2011;173:1365–79.
AG126 and 15-deoxy-delta 12, 14-PGJ2. BMC Ophthalmol 2003;3:5. 98. Gold B, Merriam JE, Zernant J, et al. Variation in factor B (BF) and complement
66. Qin S, McLaughlin AP, De Vries GW. Protection of RPE cells from oxidative component 2 (C2) genes is associated with age-related macular degeneration.
injury by 15-deoxy-delta12,14-prostaglandin J2 by augmenting GSH and Nat Genet 2006;38:458–62.
activating MAPK. Invest Ophthalmol Vis Sci 2006;47:5098–105. 99. Dinu V, Miller PL, Zhao H. Evidence for association between multiple
67. Kostadinova R, Wahli W, Michalik L. PPARs in diseases: control mechanisms complement pathway genes and AMD. Genet Epidemiol 2007;31:224–37.
of inflammation. Curr Med Chem 2005;12:2995–3009. 100. Ennis S, Jomary C, Mullins R, et al. Association between the SERPING1
68. Rodrigues GA, Maurier-Mahé F, Shurland DL, et al. Differential effects of gene and age-related macular degeneration: a two-stage case-control study.
PPARgamma ligands on oxidative stress-induced death of retinal pigmented Lancet 2008;372:1828–34.
epithelial cells. Invest Ophthalmol Vis Sci 2011;52:890–903. 101. Chan CC, Tuo J, Bojanowski CM, et al. Detection of CX3CR1 single nucleotide
69. Bernardo A, Minghetti L. PPAR-gamma agonists as regulators of microglial polymorphism and expression on archived eyes with age-related macular
activation and brain inflammation. Curr Pharm Des 2006;12:93–109. degeneration. Histol Histopathol 2005;20:857–63.
70. Murata T, He S, Hangai M, et al. Peroxisome proliferator-activated receptor- 102. Tuo J, Smith BC, Bojanowski CM, et al. The involvement of sequence variation
gamma ligands inhibit choroidal neovascularization. Invest Ophthalmol Vis and expression of CX3CR1 in the pathogenesis of age-related macular
Sci 2000;41:2309–17. degeneration. FASEB J 2004;18:1297–9.
103. Combadière C, Feumi C, Raoul W, et al. CX3CR1-dependent subretinal macrophage-mediated phagocytosis via phosphatidylserine receptor and
microglia cell accumulation is associated with cardinal features of age-related integrin alphavbeta3. Am J Pathol 2003;162:1869–79.
macular degeneration. J Clin Invest 2007;117:2920–8. 135. Geller SF, Lewis GP, Anderson DH, et al. Use of the MIB-1 antibody for detect-
104. Goverdhan SV, Ennis S, Hannan SR, et al. Interleukin-8 promoter polymor- ing proliferating cells in the retina. Invest Ophthalmol Vis Sci 1995;36: 561
phism -251A/T is a risk factor for age-related macular degeneration. Br J 737–44.
Ophthalmol 2008;92:537–40. 136. Charteris DG, Sethi CS, Lewis GP, et al. Proliferative vitreoretinopathy-
105. Zareparsi S, Buraczynska M, Branham KE, et al. Toll-like receptor 4 variant developments in adjunctive treatment and retinal pathology. Eye (Lond)

Chapter 25
D299G is associated with susceptibility to age-related macular degeneration. 2002;16:369–74.
Hum Mol Genet 2005;14:1449–55. 137. Iandiev I, Uckermann O, Pannicke T, et al. Glial cell reactivity in a
106. Yang Z, Stratton C, Francis PJ, et al. Toll-like receptor 3 and geographic porcine model of retinal detachment. Invest Ophthalmol Vis Sci 2006;47:
atrophy in age-related macular degeneration. N Engl J Med 2008;359: 2161–71.
1456–63. 138. Wickham L, Sethi CS, Lewis GP, et al. Glial and neural response in short-term
107. Fritsche LG, Loenhardt T, Janssen A, et al. Age-related macular degeneration human retinal detachment. Arch Ophthalmol 2006;124:1779–82.
is associated with an unstable ARMS2 (LOC387715) mRNA. Nat Genet 139. Lewis GP, Chapin EA, Luna G, et al. The fate of Müller’s glia following
2008;40:892–6. experimental retinal detachment: nuclear migration, cell division, and
108. Jakobsdottir J, Conley YP, Weeks DE, et al. Susceptibility genes for age-related subretinal glial scar formation. Mol Vis 2010;16:1361–72.
maculopathy on chromosome 10q26. Am J Hum Genet 2005;77:389–407. 140. Nakazawa T, Matsubara A, Noda K, et al. Characterization of cytokine

Inflammatory Response and Mediators in Retinal Injury


109. Schmidt S, Hauser MA, Scott WK, et al. Cigarette smoking strongly responses to retinal detachment in rats. Mol Vis 2006;12:867–78.
modifies the association of LOC387715 and age-related macular degeneration. 141. Hollborn M, Francke M, Iandiev I, et al. Early activation of inflammation- and
Am J Hum Genet 2006;78:852–64. immune response-related genes after experimental detachment of the porcine
110. Wang JJ, Ross RJ, Tuo J, et al. The LOC387715 polymorphism, inflammatory retina. Invest Ophthalmol Vis Sci 2008;49:1262–73.
markers, smoking, and age-related macular degeneration. A population- 142. Nakazawa T, Hisatomi T, Nakazawa C, et al. Monocyte chemoattractant
based case-control study. Ophthalmology 2008;115:693–9. protein 1 mediates retinal detachment-induced photoreceptor apoptosis.
111. Friedrich U, Myers CA, Fritsche LG, et al. Risk- and non-risk-associated vari- Proc Natl Acad Sci U S A 2007;104:2425–30.
ants at the 10q26 AMD locus influence ARMS2 mRNA expression but exclude 143. Nakazawa T, Kayama M, Ryu M, et al. Tumor necrosis factor-alpha medi-
pathogenic effects due to protein deficiency. Hum Mol Genet 2011;20: ates photoreceptor death in a rodent model of retinal detachment.
1387–99. Invest Ophthalmol Vis Sci 2011;52:1384–91.
112. Sapieha P, Joyal JS, Rivera JC, et al. Retinopathy of prematurity: understand- 144. Zacks DN, Han Y, Zeng Y, et al. Activation of signaling pathways and
ing ischemic retinal vasculopathies at an extreme of life. J Clin Invest stress-response genes in an experimental model of retinal detachment.
2010;120:3022–32. Invest Ophthalmol Vis Sci 2006;47:1691–5.
113. Hartnett ME. The effects of oxygen stresses on the development of features 145. Chong DY, Boehlke CS, Zheng QD, et al. Interleukin-6 as a photoreceptor
of severe retinopathy of prematurity: knowledge from the 50/10 OIR model. neuroprotectant in an experimental model of retinal detachment. Invest
Doc Ophthalmol 2010;120:25–39. Ophthalmol Vis Sci 2008;49:3193–200.
114. Saito Y, Geisen P, Uppal A, et al. Inhibition of NAD(P)H oxidase reduces 146. Yang L, Kim JH, Kovacs KD, et al. Minocycline inhibition of photoreceptor
apoptosis and avascular retina in an animal model of retinopathy of prema- degeneration. Arch Ophthalmol 2009;127:1475–80.
turity. Mol Vis 2007;13:840–53. 147. Kauffmann DJ, van Meurs JC, Mertens DA, et al. Cytokines in vitreous humor:
115. Hardy P, Dumont I, Bhattacharya M, et al. Oxidants, nitric oxide and pros- interleukin-6 is elevated in proliferative vitreoretinopathy. Invest Ophthalmol
tanoids in the developing ocular vasculature: a basis for ischemic retinopathy. Vis Sci 1994;35:900–6.
Cardiovasc Res 2000;47:489–509. 148. Kon CH, Occleston NL, Aylward GW, et al. Expression of vitreous cytokines
116. Beauchamp MH, Sennlaub F, Speranza G, et al. Redox-dependent effects of in proliferative vitreoretinopathy: a prospective study. Invest Ophthalmol Vis
nitric oxide on microvascular integrity in oxygen-induced retinopathy. Sci 1999;40:705–12.
Free Radic Biol Med 2004;37:1885–94. 149. Limb GA, Chignell AH. Vitreous levels of intercellular adhesion molecule
117. Gu X, El-Remessy AB, Brooks SE, et al. Hyperoxia induces retinal vascular 1 (ICAM-1) as a risk indicator of proliferative vitreoretinopathy. Br J
endothelial cell apoptosis through formation of peroxynitrite. Am J Physiol Ophthalmol 1999;83:953–6.
Cell Physiol 2003;285:C546–54. 150. Mitamura Y, Takeuchi S, Matsuda A, et al. Hepatocyte growth factor
118. Balazy M. Trans-arachidonic acids: new mediators of inflammation. J Physiol levels in the vitreous of patients with proliferative vitreoretinopathy. Am J
Pharmacol 2000;51:597–607. Ophthalmol 2000;129:678–80.
119. Brault S, Martinez-Bermudez AK, Marrache AM, et al. Selective neuromicro- 151. El-Ghrably IA, Dua HS, Orr GM, et al. Intravitreal invading cells contribute
vascular endothelial cell death by 8-Iso-prostaglandin F2alpha: possible role to vitreal cytokine milieu in proliferative vitreoretinopathy. Br J Ophthalmol
in ischemic brain injury. Stroke 2003;34:776–82. 2001;85:461–70.
120. Hardy P, Beauchamp M, Sennlaub F, et al. New insights into the retinal 152. La Heij EC, van de Waarenburg MP, Blaauwgeers HG, et al. Basic fibroblast
circulation: inflammatory lipid mediators in ischemic retinopathy. Prostaglan- growth factor, glutamine synthetase, and interleukin-6 in vitreous fluid from
dins Leukot Essent Fatty Acids 2005;72:301–25. eyes with retinal detachment complicated by proliferative vitreoretinopathy.
121. Ushio-Fukai M. VEGF signaling through NADPH oxidase-derived ROS. Am J Ophthalmol 2002;134:367–75.
Antioxid Redox Signal 2007;9:731–9. 153. Ogata N, Nishikawa M, Nishimura T, et al. Inverse levels of pigment
122. Saito Y, Uppal A, Byfield G, et al. Activated NAD(P)H oxidase from epithelium-derived factor and vascular endothelial growth factor in the
supplemental oxygen induces neovascularization independent of VEGF in vitreous of eyes with rhegmatogenous retinal detachment and proliferative
retinopathy of prematurity model. Invest Ophthalmol Vis Sci 2008;49: vitreoretinopathy. Am J Ophthalmol 2002;133:851–2.
1591–8. 154. Dieudonné SC, La Heij EC, Diederen R, et al. High TGF-beta2 levels during
123. Liu T, Castro S, Brasier AR, et al. Reactive oxygen species mediate virus- primary retinal detachment may protect against proliferative vitreoretino­
induced STAT activation: role of tyrosine phosphatases. J Biol Chem pathy. Invest Ophthalmol Vis Sci 2004;45:4113–8.
2004;279:2461–9. 155. Banerjee S, Savant V, Scott RA, et al. Multiplex bead analysis of vitreous
124. Al-Shabrawey M, Bartoli M, El-Remessy AB, et al. Inhibition of NAD(P)H humor of patients with vitreoretinal disorders. Invest Ophthalmol Vis Sci
oxidase activity blocks vascular endothelial growth factor overexpression and 2007;48:2203–7.
neovascularization during ischemic retinopathy. Am J Pathol 2005;167: 156. Dieudonné SC, La Heij EC, Diederen RM, et al. Balance of vascular endothelial
599–607. growth factor and pigment epithelial growth factor prior to development of
125. Krishnadev N, Meleth AD, Chew EY. Nutritional supplements for age-related proliferative vitreoretinopathy. Ophthalmic Res 2007;39:148–54.
macular degeneration. Curr Opin Ophthalmol 2010;21:184–9. 157. Yoshimura T, Sonoda KH, Sugahara M, et al. Comprehensive analysis of
126. Stahl A, Krohne TU, Sapieha P, et al. Lipid metabolites in the pathogenesis inflammatory immune mediators in vitreoretinal diseases. PLoS ONE 2009;4:
and treatment of neovascular eye disease. Br J Ophthalmol 2011;95: e8158.
1496–501. 158. Rasier R, Gormus U, Artunay O, et al. Vitreous levels of VEGF, IL-8, and
127. Wang Y, Wang VM, Chan CC. The role of anti-inflammatory agents in TNF-alpha in retinal detachment. Curr Eye Res 2010;35:505–9.
age-related macular degeneration (AMD) treatment. Eye (Lond) 2011;25: 159. Ricker LJ, Kijlstra A, Kessels AG, et al. Interleukin and growth factor levels
127–39. in subretinal fluid in rhegmatogenous retinal detachment: a case-control
128. Lewis GP, Sethi CS, Carter KM, et al. Microglial cell activation following study. PLoS ONE 2011;6:e19141.
retinal detachment: a comparison between species. Mol Vis 2005;11:491–500. 160. Charteris DG. Proliferative vitreoretinopathy: pathobiology, surgical manage-
129. Zacks DN. Gene transcription profile of the detached retina (an AOS thesis). ment, and adjunctive treatment. Br J Ophthalmol 1995;79:953–60.
Trans Am Ophthalmol Soc 2009;107:343–82. 161. Charteris DG, Sethi CS, Lewis GP, et al. Proliferative vitreoretinopathy-
130. Wickham L, Charteris DG. Glial cell changes of the human retina in prolifera- developments in adjunctive treatment and retinal pathology. Eye (Lond)
tive vitreoretinopathy. Dev Ophthalmol 2009;44:37–45. 2002;16:369–74.
131. Chang CJ, Lai WW, Edward DP, et al. Apoptotic photoreceptor cell death after 162. Lee SJ, Aaberg Sr TM, Grossniklaus HE. Surgically excised focal posttraumatic
traumatic retinal detachment in humans. Arch Ophthalmol 1995;113:880–6. choroidal granulomatous inflammation. Retina 2003;23:719–20.
132. Cook B, Lewis GP, Fisher SK, et al. Apoptotic photoreceptor degeneration in 163. Wilson MW, Grossniklaus HE, Heathcote JG. Focal posttraumatic choroidal
experimental retinal detachment. Invest Ophthalmol Vis Sci 1995;36:990–6. granulomatous inflammation. Am J Ophthalmol 1996;121:397–404.
133. Arroyo JG, Yang L, Bula D, et al. Photoreceptor apoptosis in human retinal 164. Wickham L, Asaria RH, Alexander R, et al. Immunopathology of intraocular
detachment. Am J Ophthalmol 2005;139:605–10. silicone oil: enucleated eyes. Br J Ophthalmol 2007;91:253–7.
134. Hisatomi T, Sakamoto T, Sonoda KH, et al. Clearance of apoptotic photo- 165. Gronert K. Resolution, the grail for healthy ocular inflammation. Exp Eye Res
receptors: elimination of apoptotic debris into the subretinal space and 2010;91:478–85.
Section 2 Basic Mechanisms of Injury in the Retina

Chapter Basic Mechanisms of Pathological Retinal and


26  Choroidal Angiogenesis
Demetrios G. Vavvas, Joan W. Miller

INTRODUCTION progenitor cells, which migrate and contribute to sites of


choroidal and retinal neovascularization.3–6
The vasculature of the eye is completely formed shortly after Hypoxia is a primary stimulus for angiogenesis. In early
birth and is normally quiescent and nonproliferating in adults.1 studies of retinal development, growing vessels were noted
An active balance of pro- and antiangiogenic influences is to invade the developing retina, giving rise to the hypothesis
required to maintain this state of homeostasis. Neovasculariza- that a diffusible hypoxia-induced growth factor initiated this
tion, or angiogenesis, occurs when environmental changes result blood vessel growth.7,8 In DR and other adult vasculopathies,
in tissue expression of molecules that favor the growth of neovascularization was associated with retinal capillary
abnormal vessels prone to leak, bleed, and cause fibrous pro- nonperfusion.9–11 Experimental laser vein occlusion in the min-
liferation. In the retina, neovascularization causes vitreous iature pig and monkey models have demonstrated this.12,13
hemorrhage and traction retinal detachment in diseases such Hypoxia prevents the ubiquitin proteosome system degrada-
as diabetic retinopathy (DR) and retinopathy of prematurity tion of the transcription factor hypoxia-inducible factor 1-alpha
(ROP). Choroidal neovascularization (CNV) denotes the patho- (HIF-1α). Stable HIF-1α binds to the promoter region of its
logic growth of new blood vessels from pre-existing choroidal major target gene, vascular endothelial growth factor (VEGF),
vessels into the subretinal space. The newly formed vessels resulting in transcriptional upregulation of VEGF and causing
lie between the choroid and the retinal pigment epithelium angiogenesis (Fig. 26.2).14–16
(RPE) or between the native RPE and the neurosensory retina; Response to tissue injury also plays a key role in angio-
thus, CNV is also referred to as subretinal neovascularization. genesis. CNV occurs both experimentally and clinically after
In some patients neovascularization forms in the neurosensory injury to the outer retina, iatrogenic laser injury, traumatic
retina, and at later stages may extend to the choroidal space. choroidal ruptures, and outer retinal choroidopathies.17,18 Sub-
This is also known as retinal angiomatous proliferation or lethal cellular injury may release growth factors such as basic
retinochoroidal anastomosis. Neovascularization leads to fibroblast growth factor (bFGF or FGF-2).19 Growth factor-
destruction of the retina and formation of a disciform scar in secreting monocytes may be recruited to the site of tissue
diseases such as neovascular age-related macular degeneration injury, which may increase the expression of growth factors
(AMD) (Fig. 26.1). causing angiogenesis.20
Neovascularization is the final common pathway in DR, AMD, Free radicals in the form of reactive oxygen species (ROS) or
vascular occlusive disease, and ROP, all of which contribute to intermediates have been implicated in the pathogenesis of
the major causes of blindness in developed countries. Our numerous vascular diseases. ROS initiate the repair process by
knowledge of neovascularization continues to grow as new mol- stimulating an angiogenic response. There is evidence that, over
ecules are discovered, new interactions between molecules are time, continual ROS stress depletes tissue of its antioxidant capa-
elucidated, and our understanding of the pathogenesis of spe- bilities. Oxidative stress and inflammation may play a role in the
cific retinal diseases evolves. Our expanding discovery in this pathogenesis of AMD.21 Increase in free radicals is believed to
field is essential to the development of new strategies for result in an overexpression of growth factors by the RPE, causing
improved therapeutic targeting. a proinflammatory state, damage to Bruch’s membrane, and
ultimately CNV.22 The Age-Related Eye Disease Study (AREDS),
PATHOGENESIS a multicentered randomized clinical trial, showed a clinical
In angiogenesis, capillary endothelial cells proliferate from pre- benefit of high-dose antioxidants in reducing visual loss
existing blood vessels. Activated endothelial cells secrete proteo- associated with macular degeneration.23
lytic enzymes that dissolve basement membrane around the It is clear that the process of neovascularization is complex,
parent vessel and align themselves to form a new capillary involving inflammatory, angiogenic, apoptotic, and stress-
sprout. By curving and elongating, the sprouts form tubes with induced interactions. We will now examine the pathogenesis of
lumens which anastomose to form loops. Mesenchymal cells are angiogenesis associated with several important eye diseases,
recruited to form smooth-muscle cells, or pericytes, and new Subsequently, we will discuss specific key growth factors
basement membrane is deposited.2 More recently it has been involved in pro- and antiangiogenic responses. Examining their
demonstrated that hematopoietic stem cells may potentially dif- influences in the tissue environment may provide further insight
ferentiate into various cellular lineages, including endothelial to the processes involved in ocular neovascularization.
Retinal neovascularization Ischemic retina
A B Retinal hemorrhage
Retinal fibrosis
563

GCL

Chapter 26
INL

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


POS

RPE
BM
CC

Normal retina Retinal neovascularization

C D

GCL

INL

POS

RPE
BM
CC

Choroidal neovascularization Retinal angiomatous proliferation

Fig. 26.1 Forms of abnormal angiogenesis encountered in ocular neovascular disorders. (A) The vascular network of the retina consists of
the superficial and intraretinal vascular plexus, which provide oxygen and nutrients to the inner two-thirds of the retina. The outer one-third
of the retina (photoreceptors and retinal pigment epithelium (RPE)) receives its blood supply from the choroidal circulation. (B) Retinal
neovascularization involves the proliferation of new vessels from the superficial retina at the vitreoretinal interface. These fragile and tortuous
neovessels often result in hemorrhage, fibrosis, and traction of the retina. (C) In choroidal neovascularization, new vessels arising from the
choroidal circulation grow through breaks in Bruch’s membrane (BM) under the RPE, neurosensory retina, or a combination of both. (D) In some
cases, neovascularization forms in the neurosensory retina and at later stages may form anastomoses with the choroidal circulation (CC), which
is also known as retinal angiomatous proliferation (RAP) or retinochoroidal anastomosis (RCA). GCL, ganglion cell layer; INL, inner nuclear layer;
POS, photoreceptor outer segments. (Courtesy of Aristomenis Thanos, MD.)
Fig. 26.2 The hypoxia-inducible factor (HIF)
Normoxia Hypoxia pathway. Simplified representation of HIF
regulation under normoxia and hypoxia.
564 Under normoxic conditions (left), HIF is
-OH Hydroxylation of HIF-1
degraded by the proteasome through a
HIF-α by proline and asparagine HIF-α process involving hydroxylation and then
-OH hydroxylase ubiquitination of HIF molecules by von
Section 2

No hydroxylation
Hippel–Lindau (VHL) protein complex. Under
of HIF-α hypoxic conditions, HIF-1α accumulates in
HIF-1 polyubiquitination the cytoplasm and dimerizes with HIF-1β.
pVHL pVHL The dimer then translocates to the nucleus,
by VHL protien complex
upregulating a number of proangiogenic
molecules, including VEGF, vascular
Ub endothelial growth factor; PDGF-β, platelet-
Ub derived growth factor-β; TGF-α, transforming
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

-OH Ub
Ub HIF-α growth factor-α; and EPO, erythropoietin.
HIF-α Degradation
(Courtesy of Aristomenis Thanos, MD.)
of HIF-α
-OH HIF-β

Nucleus
Proteosome Cytoplasm translocation

Nucleus

HIF-α
HIF-β p300

Hypoxia-inducible genes

VEGF PDGF-β TGF-α EPO

CNV associated with AMD (Fig. 26.3) collagenous zone of the Bruch’s membrane) also have important
associations with CNV.20 Therefore, abnormal deposits that
Aging and senescence of the RPE
occur with diffuse distribution pattern between the RPE layer
The incidence and progression of nearly every feature of AMD,
and Bruch’s membrane are predisposing features of CNV. It is
including CNV, relate to age.24 Lipofuscin, a byproduct of pho-
hypothesized that deposits between the RPE layer and Bruch’s
toreceptor outer-segment digestion by lysosomes, increases with
membrane may block the diffusion of oxygen and nutrients from
age in RPE as lysosomal activity decreases in RPE with aging.
choriocapillaris to the RPE monolayer and photoreceptors.33 This
Progressive accumulation of lipofuscin is thought to result in
speculative localized cellular hypoxia could result in overexpres-
disturbance of RPE function. Aged RPE cells may decrease pro-
sion of angiogenic growth factors such as VEGF, which, in turn,
duction of antiangiogenic molecules such as pigment epithelium-
induce neovascularization from the choroidal vasculature.
derived growth factor (PEDF) as successive passage of cultured
Additionally, the deposits are known to contain components
RPE cells results in diminished production of PEDF.25–27
of the immune response, and thus may act as initiators of
inflammation.34–37 The deposits may also serve as a reservoir for
Drusen, basal laminar/linear deposit formation
sequestration of factors such as advanced glycation end prod-
Soft drusen, unlike hard drusen, appear to be an important asso-
ucts (AGE)38,39 that may affect the function of adjacent RPE and
ciated and predisposing feature of CNV. It is thought that mem-
choroidal endothelial cells.
branous accumulation of debris as part of a diffuse disturbance
of the RPE, softening of hard drusen, and cleavage in basal Enzymatic and mechanical disruption
laminar/linear deposits may aid in the formation of soft of Bruch’s membrane
drusen.28–32 In CNV, activated endothelial cells migrate through Bruch’s
Histopathologic studies also reveal that basal laminar deposits membrane; this process occurs by degradation of an intact
(accumulating between the plasma and basement membrane of Bruch’s membrane, or growth through an existing Bruch’s
the RPE) and basal linear deposits (with a thickening of the inner membrane break. Clinicopathologic studies suggest that classic
A B
565

Chapter 26
VEGF MMPs
Soft drusen Soft drusen

MMPs MMPs

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


Macrophage Macrophage

C D

bFGF
VEGF
Soft drusen Soft drusen

E F

VEGF/bFGF

Soft drusen

Fig. 26.3 Choroidal neovascularization (CNV). Presumed pathologic stages required for the development and progression of CNV. (A) Low-grade
inflammation and tissue hypoxia at the sites of soft drusen trigger the secretion of vascular endothelial growth factor (VEGF) by retinal pigment
epithelium (RPE) and photoreceptor cells. Macrophages are attracted to the area in response to locally secreted chemoattractants or drusen
components (e.g., monocyte chemoattractant protein-1, complement factor H). (B) Rupture of Bruch’s membrane as a prerequisite for the
development of CNV, which is thought to occur due to the local secretion of matrix metalloproteinases (MMPs) by RPE, choroidal endothelial
cells, or macrophages. This proteolytic degradation of Bruch’s membrane activates the wound-healing response at the choroidal–RPE interface.
(C) Angiogenic cytokines such as VEGF and basic fibroblast growth factor (bFGF) induce the activation and proliferation of choroidal endothelial
cells through the breaks in Bruch’s membrane. Macrophages migrate into the subretinal space and promote the angiogenic response. (D) The
newly formed vessels lack pericytes, are fragile, and often result in subretinal hemorrhage. Growth factors secreted locally induce the
transdifferentiation of RPE cells and its further migration from the monolayer into the stroma of the lesion. (E) Maturation of the neovascular
membrane with recruitment of fibroblasts, transdifferentiated RPE cells, and inflammatory cells. New blood vessel formation continues in
response to continued expression of angiogenic growth factors and hemorrhage. (F) The endpoint of this wound-healing response is the
formation of a disciform lesion, characterized by aberrant fibrous tissue proliferation severely affecting the function of overlying photoreceptors.
(Courtesy of Aristomenis Thanos, MD.)

CNVs are predominantly subretinal in location, whereas occult collagenase),41 MMP-2 (72-kDa gelatinase),41,42 MMP-3 (stromo-
CNVs are predominantly sub-RPE.40 Bruch’s membrane may lysin), and MMP-9 (92-kDa gelatinase),41 as well as TIMP-1,42,43
be disrupted when the balance between proteolytic enzymes TIMP-2, and TIMP-3.43 Thus, proteolysis of Bruch’s membrane
such as matrix metalloproteinases (MMPs) and their inhibitors, may potentially result from reduced expression of TIMPs or
the tissue inhibitors of metalloproteinases (TIMPs), favors a increased expression of MMPs. MMPs also play an important
proteolytic environment. RPE cells express MMP-1 (interstitial role in degrading the extracellular matrix at the leading edge
of neovascular fronds. It has been shown that decreases in debris may lead to further inflammation.63 CX3CR1 is a cytokine
thickness and integrity of the elastic layer of Bruch’s membrane produced by MC, and an association between AMD and CX3CR1
566 are seen in the macula of eyes with AMD but not in controls.44 has been reported.64–66 A double knockout of MCP1 and CX3CR1
Lysyl oxidase-like (LOXL) protein 1 has been shown to guide in an animal model leads to an AMD phenotype with spontane-
the spatially defined deposition of elastin and is essential for ous CNV formation.67,68
the maintenance of elastic fibers. LOXL1-deficient mice have The contribution of bone marrow-derived endothelial cells
Section 2

been shown to develop increased neovascularization after laser to CNV has been evaluated by inducing CNV in irradiated
injury.45 Impairment in other components of the Bruch’s mem- mice that have received bone marrow transplants from green
brane such as collagen XVIII has been shown to affect CNV fluorescent protein (GFP)-expressing mice. These studies show
formation in animals. Collagen XVIII knockout mice develop that GFP+ endothelial cells are incorporated into the laser-
normal choroidal vasculature; however, they demonstrate induced CNV, suggesting that bone marrow-derived progenitor
increased size and leakage of laser-induced CNV.46 cells provide an additional source of endothelial cells in
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

Transgenic mice with an intact Bruch’s membrane that over- CNV.6,69,70


express VEGF in photoreceptors develop subretinal neovascu-
CNV membrane formation
larization; however, the subretinal vessels extend from retinal
In a series of 760 eyes with AMD from 450 patients; 310 eyes
vessels rather than the choroidal vasculature.47 In contrast, trans-
(~40%) demonstrated a disciform membrane.20 The mean diam-
genic mice that overexpress VEGF in RPE cells show intra­
eter of these membranes was 3.73 mm, and the mean thickness
choroidal CNV.48 These findings support the notion that CNV
was 0.27 mm. Preservation of photoreceptor cells was seen when
requires both the expression of an angiogenic factor and a break
the thickness of the disciform scars was 0.2 mm or less. Histo-
in Bruch’s membrane (by proteolysis, physical disruption, or
logic studies of highly vascular CNV show that they contain
pre-existing break).
endothelial cell-lined channels, RPE cells, and macrophages
Macrophages are an alternative source of enzymes (such as
within the extracellular matrix-rich stroma. The stromal RPE are
MMPs) that could cause focal disruption of Bruch’s membrane.49
immunoreactive for smooth-muscle actin, indicating a transdif-
Histopathologic studies reveal that macrophages accumulate
ferentiated phenotype. The transdifferentiated RPE often
near thinned segments of Bruch’s membrane.50 In AMD, the RPE
expresses VEGF, suggesting that these RPE have a proangio-
shows increased expression of monocyte chemoattractant
genic role, and that VEGF is a mediator of AMD-related CNV.71
protein-1 (MCP-1), a factor critical for macrophage recruitment.51
CNV likely occurs when there is an imbalance between pro-
Macrophages in the choroid may subsequently degrade Bruch’s
angiogenic and antiangiogenic growth factors. Angiogenic
membrane, thus forming a passage that can be used by activated
growth factors, such as VEGF, angiopoietins (Ang 1, Ang 2), and
choroidal endothelial cells to gain entrance to the sub-RPE space.
bFGF, released from RPE cells and/or other retinal cells, promote
Complement, AMD, and CNV CNV. Choroidal angiogenesis is inhibited by antiangiogenic
Recent studies suggest that complement dysregulation may growth factors such as thrombospondin-1 and PEDF72 and by
mediate AMD pathogenesis. Complement components have Fas-mediated killing of new vessels by Fas-ligand-positive RPE
been detected in drusen29,30,34–37 and an animal model of CD59 or leukocytes.73
knockout leads to increased CNV.52 Most recently, a Y402H poly- CNV lesions are leaky and often show evidence of hemor-
morphism in complement factor H (CFH) has been associated rhage. In fact, the evolution of the CNV often involves repeated
with increased risk of AMD and likely wet AMD.53–56 This par- episodes of serous leakage and hemorrhage. In response to these
ticular allele of CFH leads to decrease in its activity and thus an changes, fibrovascular membrane formation occurs around the
increased complement-mediated inflammation since CFH is a CNV, resulting in a disciform lesion. The presence of hemosid-
negative regulator of the alternative pathway. Other human erin within the disciform CNV suggests that fibrovascular orga-
studies have identified protective haplotypes in two other genes nization of hemorrhage is important in its evolution. Over time,
encoding complement proteins, factor B (BF) and complement the newly formed vessels mature and show reduced leakage that
component 2 (C2).57 may result from the perivascular growth of RPE.
Studies of human donor eyes have shown increased levels of
C-reactive protein and decreased CFH in the RPE/Bruch’s Cicatricial membrane formation
membrane/choriocapillaris region of eyes with AMD compared
Cellular and highly vascularized membranes gradually evolve
to controls, suggesting a role of complement and inflammation
into paucicellular cicatricial membranes. The loss of cellularity
in the progression of the disease.58
is most likely due to cell death of stromal cells.74 Surgically
Inflammation, bone marrow-derived cells, and CNV excised AMD-related CNV contains apoptotic stromal RPE,
Macrophages are an additional source of angiogenic growth endothelial cells, and macrophages. Cell death may be associ-
factors that may promote the development of CNV. Activated ated with a local decrease in the expression of angiogenic
macrophages show increased expression of inflammatory cyto- growth factors that promote survival of activated cells. Fas
kines such as tumor necrosis factor-α (TNF-α) and interleukin-1β and Fas ligand expression may also be involved in the induc-
(IL-1β), which may promote angiogenesis by stimulating VEGF tion of apoptosis in these cells.75 Little is known about the
expression in RPE.59 Depletion of macrophages diminishes the mediators of collagenous scar formation in CNV. Studies have
lesion size and severity in experimental laser-induced CNV.60,61 shown that connective tissue growth factor (CTGF), a proangi­
Ccr-2 knockout mice, characterized by hampered macrophage ogenic and profibrotic growth factor, is expressed in stromal
recruitment, also show inhibition of laser-induced CNV.62 RPE cells in surgically excised CNV.76 The development of a
Microglial cells (MC) are involved in phagocytosis of injured cicatricial disciform lesion promotes overlying photoreceptor
and dead cells. Dysfunction of phagocytosis of accumulating cell loss.77
However, since Epo has neuroprotective effects, its inhibition for
Neovascularization associated neovascularization may come with significant collateral damage.
with diabetic retinopathy In addition to elevation of proangiogenic factors, there is also 567
Neovascularization in DR can be thought of as a two-step an imbalance of antiangiogenic factors. PEDF, now known as
process. An initial vascular dropout/occlusion phase is followed serpin peptidase inhibitor clade F member 1(SERPINF1), is con-

Chapter 26
by abnormal vascular growth. Chronic hyperglycemia leads to sidered to be the most potent inhibitor of neovascularization,
vascular injury with basement membrane thickening, pericyte inhibiting endothelial cell migration with a median effective
loss, microaneurysms, and vascular leakage.78,79 Many biochemi- dose of 0.4 nM.102 It specifically interferes with neovasculature
cal pathways (protein kinase C, nuclear factor (NF)-κB, mitogen and not with established vessels due to its mechanism of action
activated protein kinase) have been involved in the pathogenesis through Fas/FasL and its cooperation with angiogenic factors.
of early DR but the exact mechanism remains ill defined. Oxida- SERPINF1 upregulates FasL on endothelial cells, whereas angio-
tive injury, microthrombi formation, inflammatory mediator genic factors induce its essential partner Fas receptor on neoves-

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


upregulation, and leukostasis have been observed. Proinflam- sels but not on established vessels.103 In PDR, it has been found
matory cytokines IL-1β and TNF-α are elevated in animal models that PEDF levels are downregulated compared to controls.104
of DR, recruiting leukocytes and accelerating vascular dropout PEDF also has neuroprotective effects, and thus seems to be an
by promoting further inflammation as well as activating apop- ideal candidate for therapeutic intervention.
tosis directly or indirectly.80–82 This leads to vascular leakage as Inflammation is another important player in the pathogenesis
well as microvascular occlusion. As a consequence of microvas- of proliferative DR. Inflammatory cytokines such as TNF-α,
cular occlusion there is inadequate supply of oxygen and nutri- IL-1β, intercellular adhesion molecule (ICAM), inducible nitric
ents for proper cellular metabolism. This leads neurons and oxide synthase (iNOS), and IL-6 are elevated in patients with
supporting astrocytes to secrete proangiogenic factors such as PDR.105–108 Localized inflammation is thought to lead to leukosta-
VEGF and insulin-like growth factor (IGF)-1. Clinical studies sis through the interaction of ICAM and CD18.81,109–111 This leu-
have substantiated the role of VEGF in DR.83,84 VEGF levels are kostasis leads to local vaso-occlusion, nonperfusion, and
elevated in patients with proliferative DR (PDR), and although ischemia, which leads to upregulation of angiogenic factors.
recent reports have not been able to show decreases of VEGF IL-1β can promote endothelial cell proliferation, propagate
levels in the short term after panretinal photocoagulation,85,86 inflammation, and upregulate HIF-1α.112,113 TNF-α can promote
successful laser retinal ablation decreases them over the long angiogenesis, and macrophage-induced angiogenesis is medi-
term.84 In addition, several studies have shown the beneficial ated through TNF-α.114,115 TNF-receptor p55-deficient animals
effects of anti-VEGF intervention in DR.87–90 Before the discovery are protected from retinal neovascularization in animal models.116
of VEGF and its important role in DR, hypophysectomy studies Inhibitors of inflammation such as cyclo-oxygenase 2 inhibitors
had revealed a role of growth hormone (GH) and its associated and aspirin have been shown to curtail vascular pathology and
factors in DR. IGF-1 levels have been shown to be elevated in neovascularization.81,117,118
the vitreous of patients with PDR relative to control eyes.91–93 The proinflammatory environment is characterized by ele-
Although there may be a role for IGF-1 in retinal neovasculariza- vated MMPs, which degrade the extracellular matrix, a step
tion, our understanding of IGF-1 in DR remains unclear.94 IGF-1 necessary for angiogenesis. Additionally, MMP-9 not only is
may act indirectly via VEGF. Studies of cultured RPE cells dem- induced by IL-8119 but also activates IL-8,120 which recruits more
onstrated that adding IGF-1 in vitro increased VEGF mRNA and inflammatory cells, feeding a destructive positive-feedback loop.
secreted protein.95 The accumulation of AGE after long-term Insulin121 and PEDF122 are degraded by MMP-9, leading to a state
exposure to hyperglycemia caused an increase in IGF-1 synthesis of more insulin resistance and neuronal peril. Beránek et al. in
in human monocytes, suggesting a role for inflammatory 2008 showed MMP2 and MMP9 to be elevated in patients
pathways.96 with PDR.123
Angiopoietins are molecules that bind endothelial Tie recep-
tors and are involved in angiogenesis during development.
Neovascularization associated
Vitreous level of Ang 2 and VEGF were significantly higher with retinopathy of prematurity
in patients with PDR than in controls or those with inactive Retinal vasculature development begins at about the fourth
PDR and the levels of Ang 2 correlated significantly with that month of gestation and is complete at about 40 weeks. When
of VEGF, suggesting an association of Ang 2 and VEGF with an infant is born prematurely, it moves from a hypoxic
angiogenic activity in PDR.97 A nuclease-resistant RNA aptamer (PaO2 ~30 mmHg) to relative hyperoxic (PaO2 ~ 60–100 mmHg)
that binds and inhibits Ang 2 but not the related Tie2 agonist, environment.124,125 The increase in oxygen tension, coupled with
Ang 1, inhibited bFGF-mediated neovascularization in the rat weak antioxidant defenses, leads to production of toxic ROS
corneal micropocket angiogenesis assay, demonstrating that and decreased HIF. The combination of these effects leads to
a specific inhibitor of Ang 2 can act as an antiangiogenic damage of the vascular endothelial cells and decreased pro-
agent.98 duction of VEGF and Epo, with resultant cessation of normal
More recently, a role of erythropoietin (Epo) in DR has been vascular development. As the peripheral avascular retina con-
discovered.99 Epo is involved in the generation of red blood cells tinues to develop in the absence of a developing vascular bed,
but has been shown to be synthesized in astrocytes and its recep- it becomes relatively hypoxic and secretes VEGF and Epo at
tors have been detected in photoreceptors. Epo has been shown levels above normal physiologic values. Accordingly, the new
to have neuroprotective effects in various models.100,101 It has also vascular growth becomes exaggerated, misguided, and abnor-
been shown to be proangiogenic. Epo levels are higher in the mal, leading to physical traction and potential retinal detach-
vitreous of patients with PDR, and inhibition of both Epo ment. It is important to know that there is no direct evidence
and VEGF leads to greater inhibition of neovascularization.99 of the above-described processes from eyes of premature babies,
but it is rather an extrapolation from animal data and other twins.141 Family members of individuals with AMD are more
vascular eye diseases. However, the importance of VEGF likely to develop the disease.
568 upregulation in the proliferative phase of this disease has been The protein ApoE is involved in the regulation of blood
shown in humans, since blockade of VEGF with anti-VEGF lipids, and has been found to be involved in AMD. The ε2
antibodies (bevacizumab) showed significant efficacy in infants allele of the APOE gene may increase the risk, whereas the
with stage 3+ zone I disease.126 ε4 allele may be protective.142 However, other studies find
Section 2

Oxygen level changes cannot fully account for disease pro- opposite or no association between ApoE and AMD.143–145
gression and prematurity is the strongest risk factor for ROP The chemokine receptor CX3CR1 and the Toll-like receptor 4
development. Growth factors such as IGF-1 have been impli- (TLR4) have been implicated in several, but not all, studies as
cated by epidemiological and animal studies. Babies with risk factors for progression of the disease.146–149 Conflicting data
decreased levels of IGF-1 have slower vascular development in exist as well on TLR3 as a risk factor for AMD progression.150–153
the first phase of the disease and animals that lack IGF-1 have Convincing genetic association data exist for a locus of
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

substantial reduction in neovascularization in the proliferative chromosome 10q26, which harbors the pleckstrin homology
phase of the disease.127 domain-containing protein 1 (PLEKHA1), the ARMS2 gene
product of unknown function, and the trypsin-like protease
Neovascularization in vascular occlusions HTRA1. This locus is associated with a seven- to tenfold
Ischemia and subsequent elevation of angiogenic factors are increased risk of the disease.154–156 In addition, very strong evi-
thought to be major and important contributors in neovascular- dence exists for the CFH gene, whereas the single nucleotide
ization after central retinal vein occlusion (CRVO). The role of polymorphism Y402H can increase the risk of the disease three-
VEGF in particular has been well established. Aiello et al. in to sevenfold.157–159 However, the variation in these genomic
1994 showed increased VEGF levels in the vitreous of patients regions alone is unable to predict disease development with
with active neovascular CRVO;84 in the same year, Miller et al. high accuracy.
showed evidence that VEGF is temporally and spatially corre- Several other studies have implicated various other compon­
lated with ocular angiogenesis in a primate model of CRVO and ents of the complement pathway in disease progression. Varia-
that blockade of VEGF could inhibit neovascularization.128 Obvi- tions in complement 2, complement factor B, and CFH-related
ously, VEGF is not the only cytokine elevated in CRVO, and protein 1 and 3 are thought to have protective odds ratios
elevated levels of VEGF can be seen without neovascularization. whereas complement 3 may be associated with increased
Several interleukins have been shown to be elevated in CRVO susceptibility.57,160–163
as well as MCP-1 and ICAM-1.129–131 Additionally, antiangio- Mutations in other genes have been associated with macular
genic factors such as PEDF have been shown to be downregu- degenerations other than AMD, such as the ABCR gene, a rod-
lated in CRVO.132 The alterations of cytokines seen in CRVO not specific ATP-binding cassette (ABC) transporter seen in Star-
only lead to potential neovascularization but are major contrib- gardt disease.164,165 Mutations in TIMP-3, an inhibitor of
utors to vascular leakage and macular edema. Thus far, only proteolysis found within Bruch’s membrane, are seen in Sorsby
anti-VEGF blockade has been studied as a targeted therapy in fundus dystrophy, an autosomal dominant disorder with histo-
human patients with CRVO, and several studies have shown logic changes similar to those of neovascular AMD.166–169
significant (but not absolute) success of such a strategy, suggest- Mutation in the vitelliform macular dystrophy (VMD)2 gene
ing that combination therapies may be needed for improved that encodes the protein bestrophin has been found in Best
outcomes.133 macular dystrophy, and mutation in the epidermal growth
factor-containing, fibrillin-like extracellular matrix protein
Neovascularization in uveitis (EFEMP1) gene has been seen in patients with malattia levan-
Retinal neovascularization and CNV in uveitis are well recog- tinese and Doyne honeycomb retinal dystrophy, which are
nized, although uncommon. Ischemia from nonperfusion and disorders associated with drusen formation.170,171
inflammation may contribute to the formation of new vessels.
Peripheral ischemia and the extent of neovascularization seem Diabetic retinopathy
to correlate in predominantly ischemic vasculitides such as Eales Although the Diabetes Control and Complications Trial and
disease.134 However, neovascularization of the disc has been seen the UK Prospective Diabetes Study demonstrate the beneficial
in other uveitic cases without apparent ischemia.135 Given the effects of tight glucose and blood pressure control, patients
fact that neovascularization can regress with corticosteroid and with good glucose control still develop DR. The recent
other immune-suppressive treatments, it seems that inflamma- ADVANCE study reported that intensive glucose control to
tion alone may be sufficient for retinal neovascularization and reduce A1c to 6.5% or less had no effect on retinopathy rates.172,173
CNV in this heterogeneous group of diseases.136–138 Nevertheless, In addition, it is apparent that some patients with poor control
VEGF is still a key molecule in uveitis-associated neovascular- of their disease may not develop DR even over long periods
ization, and several studies have shown benefit (albeit partial) of time, while others develop DR in a short time despite good
of anti-VEGF therapy.139,140 disease control. This is exemplified in the Joslin Medalist study,
which found that a significant number of elderly patients with
GENETIC ASPECTS OF type 1 diabetes had no evidence of retinopathy despite surviv-
NEOVASCULARIZATION ing over 50 years with diabetes.174,175 These results suggest
that genetic factors may play a role in the progression of this
Age-related macular degeneration disease.
AMD has a significant genetic component, as there is higher Candidate gene approaches have shown fairly consistent
concordance among monozygotic twins compared to dizygotic associations with genes encoding aldose reductase (ALR2),
VEGF, and receptor for AGE (RAGE) in the progression of DR Retinopathy of prematurity
(reviewed by Liew et  al.176). Aldose reductase is involved in
Besides early birth and reduced birth weight, other factors have
the metabolism of glucose into sorbitol inside the cells. Sorbitol
been shown to contribute to development of ROP. Excessively
569
accumulation leads to osmotic stress and cell injury. Unfortu-
high levels of oxygen in incubators, used to save the lives of
nately, clinical trials with aldose reductase inhibitors failed to
premature infants, led to an ROP epidemic and the realization

Chapter 26
show benefit. It is important to note that in these trials no
that reducing the level of oxygen given to premature babies
stratification on ALR2 gene polymorphism status was under-
reduces the incidence of ROP.204,205 Although oxygen levels and
taken.177,178 Polymorphism in the VEGF promoter region has
ROP are linked, the exact mechanism is not clear. It is possible
been shown to be associated with DR progression and there
that the rate of change of oxygenation rather than the absolute
may be ethnic variation in the “at-risk” haplotype (reviewed
level may be more important for disease progression, and there
by Liew et  al.176).
have been reports that return to high oxygen levels for pro-

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


Retinopathy of prematurity longed time with gradual decline to normal oxygen levels may
halt and reverse the disease.206–210
ROP is a multifactorial disease that has many phenotypic simi-
Light exposure has also been investigated, but the LIGHT-
larities with a genetic disease called familial exudative vitreo-
ROP study concluded that a reduction in ambient light exposure
retinopathy (FEVR). At least four genes have been implicated in
does not alter the incidence of ROP.211
FEVR (norrin–FZD4–LRP5–TSPAN12 signaling pathway), and a
The diet of premature babies is usually poorer in omega-3
candidate gene approach has shown that three of them (NDP,
fatty acids compared to mother’s milk. Omega-3 fatty acid
FZD4, and LRP5) are mutated in a small percentage of severe
supplementation in animal models of ROP has shown reduc-
ROP patients.179 Association between VEGF, IGF-1 receptor,
tion in neovascularization.212 Human studies with docosahexa­
and angiotensin-converting enzyme gene polymorphisms have
enoic acid supplementation of infant formula at 0.32% of total
been implicated in some (but not all) studies.180–191 Other sug-
fatty acids showed improved visual acuity as measured by
gestive associations have been reported between ROP and
visual-evoked potential, but this study did not investigate
several candidate genes such as angiotensin II type I receptor,
neovascularization.213
Indian hedgehog, T-box 5, glycoprotein Ibα polypeptide, and
cholesterol ester transfer protein, but confirmation is still
needed.192 ANGIOGENIC AND ANTIANGIOGENIC
FACTORS IN NEOVACULARIZATION
ENVIRONMENTAL FACTORS
Vascular endothelial growth factor
Age-related macular degeneration VEGF was first identified in tumor models as a vasopermeability
Several modifiable risk factors have been associated with pro- factor and initiator of angiogenesis upregulated by hypoxia. In
gression of AMD. Amongst them, smoking has been the most addition to being the strongest endothelial cell mitogen, VEGF
consistently reported risk factor, elevating the risk of disease has been shown to induce the expression of plasminogen activa-
progression by about twofold.193–195 Additionally, a health- tors in microvascular endothelial cells, which is important in the
promoting diet (rich in seafood, vegetables, and fruits), physical extracellular proteolysis necessary for capillary formation.214
activity, and a nonsmoker status are thought to lead to a 70% VEGF induces the expression of endothelial cell α1β1 and α2β1
reduction in the risk of developing AMD.196 The AREDS study integrins, which are important in migration.215 There is also in
showed that supplementation with vitamins A and C, zinc, and vitro evidence that VEGF upregulates endothelial cell fenestra-
copper in patients with moderate-severity AMD reduces the tions in kidney glomerulus, choroid plexus, and even the cho-
risk of progression by about 25%.197 Unfortunately, this was riocapillaris.216 Leukocyte adhesion has been shown to be
not one of the a priori questions set to be answered by the important in vascular leakage. VEGF increases the expression of
study, so the benefits of this supplementation are not universally ICAM-1 on endothelial cells, resulting in increased leukostasis,
accepted. More recently, the Women’s Antioxidant and Folic which mediates the breakdown of the blood–retinal barrier.217,218
Acid Cardiovascular Study discovered that supplementation VEGF, also known as VEGF-A or VEGF-1, is expressed as five
with folate, vitamins B6 and B12 reduced the risk of advanced mRNA splice variants in humans: isoforms 121, 145, 165, 189,
AMD by 30–40% in women with high-risk cardiovascular and 206.219 VEGF is a heparin-binding dimeric glycoprotein with
factors.198 disulfide-linked subunits, which share significant sequence
homology with the A and B chains of platelet-derived growth
Diabetic retinopathy factor (PDGF).220 VEGF 121 is entirely soluble and unbound to
Smoking, hypercholesterolemia, and hypertension are known extracellular matrix. VEGF 165 is intermediate and binds some-
factors that worsen disease progression. Smoking increases what to extracellular matrix. VEGF 189 is almost entirely seques-
oxidative stress, and may increase TNF levels as well as alter tered to extracellular matrix and cell surface sites.221,222 VEGF 165
high-density lipoprotein and low-density lipoprotein levels.199,200 is the predominantly expressed isoform when human cDNA
Smoking has also been shown to elevate VEGF levels acutely libraries are screened, and is optimal for bioavailability and bio-
in about one-quarter of diabetic patients.201 Several studies have logic potency. It is the critical isoform for both developmental
shown the link between high blood pressure and cholesterol and pathologic retinal angiogenesis.222–224 Two other related
in the progression of DR. Recent studies show that there is endothelial growth factors, VEGF-B and VEGF-C, have struc-
greater benefit in reducing the progression of the disease by tural homology to VEGF and appear to play roles in tumor
lowering cholesterol levels compared to lowering high blood angiogenesis and in the development of the lymphatic system,
pressure.202,203 as does VEGF-D.225 VEGF-C, also known as VEGF-2, has been
shown to promote angiogenesis in the rabbit ischemic hind limb model, dominant-negative VEGF receptors and VEGF antisense
model.226 VEGF-E has similar angiogenic activity to VEGF-A, oligonucleotides substantially decreased retinal neovasculariza-
570 primarily through VEGF receptor 2.227 tion.254,255 This work has confirmed the central role of VEGF in
Both high- and low-affinity VEGF receptors have been identi- pathologic retinal neovascularization.
fied on not only endothelial cells, but also on bone marrow- VEGF also plays an important role in the development of
derived and retinal epithelial cells.228–230 They belong to the CNV. Immunostaining of surgical specimens of choroidal neo-
Section 2

family of tyrosine kinases requiring phosphorylation to be acti- vascular membranes showed increased VEGF expression.71,256
vated upon ligand binding. VEGFR-1 (FMS-like tyrosine kinase In situ hybridization studies have demonstrated a correlation
or FLT-1 in human) and VEGFR-2 (fetal liver kinase 1 or Flk-1 between VEGF expression and the development of CNV in laser
in the mouse; TKR-C in the rat; kinase insert domain receptor or injury models in the rat and monkey.257 Oxidative stress may
KDR in the human) are expressed on endothelial cells, whereas stimulate the overexpression of growth factors from the RPE, a
VEGFR-3 (FLT4) is primarily found on lymphatic endothelial possible inflammatory state, and subsequent damage and thick-
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

cells.231 Although VEGF binds to both VEGFR-1 and VEGFR-2, ening of Bruch’s membrane from recruited macrophages.21,22 It
the latter is primarily responsible for endothelial cell mitogene- has been suggested that the abnormally thickened Bruch’s mem-
sis, survival, and permeability.232 VEGFR-1 may be important in brane may interfere with polarized RPE secretion of VEGF nec-
development by sequestering VEGF, preventing it from interact- essary for maintenance of the choriocapillaris. Atrophy of the
ing with VEGFR-2.233 VEGFR-1 has an established role in mono- choriocapillaris, often seen clinically, may result in a state of
cyte chemotaxis.234,235 VEGF-C has also been shown to be a ligand outer retinal hypoxia, stimulating VEGF-induced angiogen­
for VEGFR-2 and VEGFR-3.226 Synergism between FGF-2 and esis.258 Studies have also shown that despite the absence of
VEGF has been demonstrated by the finding that FGF-2 increases hypoxia in the laser injury models, specific compounds that bind
VEGFR-2 expression in microvascular endothelial and aortic VEGF and its receptors virtually eliminate CNV.259,260 These data
endothelial cells.236 In addition to the receptor tyrosine kinases, have important clinical implications, as specific pharmacologic
VEGF also interacts with a family of coreceptors, the neuropilins, inhibitors of VEGF have been approved for the treatment of
which may enhance its angiogenic function.237 neovascular AMD and shown to be effective.261–264
VEGF is expressed by retinal cells in vitro and is upregulated
by hypoxia.238,239 Other modulators of VEGF expression are Insulin-like growth factor-1
hypoglycemia,234 beta-estradiol, and ROS.240–242 Several growth The role of GH and its associated factors in DR was first sug-
factors cause upregulation of VEGF gene expression, including gested by the clinical observation of regression of neovascular-
epidermal growth factor, transforming growth factor (TGF)-α ization in the retina following infarction of the pituitary during
and β, IGF-1, FGF, and PDGF, implicating both paracrine and pregnancy.265 This was followed by experimental and clinical
autocrine release of these factors in the hypoxic regulation of observations that hypophysectomy led to remission of DR.266,267
VEGF.228,243 The complications of hypophysectomy were frequent, severe,
In vivo work has demonstrated VEGF to be spatially and and often lethal; with the advent of laser photocoagulation, the
temporally correlated with iris neovascularization in a monkey therapy was largely abandoned. Subsequently, it was demon-
model of ischemic retinopathy and iris neovascularization.128 strated that GH mediated its effects through IGF-1 and IGF-2,
VEGF protein levels in serial aqueous samples have been shown and investigations were directed towards these factors. The role
to correlate with the severity of induced retinal ischemia and iris of IGF-1 in ROP is more developed. Studies of transgenic mice
neovascularization. In situ hybridization identified the inner have shown that IGF-1 is important for normal retinal vessel
retina as the source of VEGF.128,244 Work in models of ROP also development, and allows for VEGF to enhance endothelial cell
implicates the importance of VEGF in the development of retinal survival and promote early vascular development.268 Transgenic
neovascularization.245–247 mice expressing a GH antagonist demonstrated decreased retinal
In vitro, VEGF has been shown to be sufficient to produce neovascularization when subjected to hyperoxia/normoxia in
neovascularization. VEGF results in neovascularization in the the murine model of ischemic retinopathy. Adding exogenous
corneal micropocket and in chick chorioallantoic membrane IGF-1 completely restored the amount of retinal neovasculariza-
(CAM) bioassay.248,249 A single intra-arterial bolus of VEGF was tion seen in controls. Inhibition of retinal neovascularization in
sufficient to stimulate angiogenesis and collateralization in a these mice was inversely proportional to serum levels of GH and
rabbit ischemic hind limb model, leading to studies of the thera- IGF-1.269 It has been shown that a low serum level of IGF-1 is a
peutic use of VEGF in peripheral vascular and coronary artery predictor for ROP in infants.268,270
disease.250 Injections of recombinant human VEGF in normal
monkey eyes led to iris neovascularization and neovascular Fibroblast growth factor-2
glaucoma, as well as many of the changes of DR, including Acidic and basic FGF are prototype members of the FGF family.
vessel dilation, tortuosity, microaneurysm formation, hemor- Basic FGF or FGF-2 is a fibroblast mitogen, and one of the first
rhage, edema, capillary dropout, and intraretinal neovascular- angiogenic factors identified and suspected in ocular neovascu-
ization.251,252 However, VEGF alone may not be sufficient in vivo larization. FGF-2 has been called “the stored growth factor”
to induce retinal neovascularization. because much of the cell-associated FGF-2 is found in the extra-
To demonstrate the causal role of VEGF in neovascularization cellular matrix.271
secondary to ischemia, VEGF activity was specifically blocked FGF-2 has been implicated in various aspects of angiogenesis.
using anti-VEGF antibodies, soluble VEGF receptors, or anti- It stimulates endothelial cell proliferation and migration and
sense oligonucleotides to VEGF. Intravitreous injection of anti- induces the production of proteases.272 FGF-2 is angiogenic in
VEGF antibodies completely prevented the development of iris vivo in the chick CAM and corneal micropocket bioassays at
neovascularization in the monkey model.253 In a mouse ROP very low levels.273,274 FGF-2 has been isolated from many normal
tissues and tumors of mesodermal and neuroectodermal origin, rabbit cornea micropocket model, neither Ang-1 or Ang-2 alone
as well as in CNV membranes.275–278 induced corneal neovascularization, but either Ang-1 or Ang-2
One of the arguments against FGF-2’s role in pathologic angio- added to VEGF promoted neovascularization, with Ang-2 571
genesis is its lack of a definitive signal peptide for secretion.279 having the more potent effect.298 Transgenic mice overexpressing
However, a number of alternative pathways for FGF-2 release Ang-2 demonstrate disruption of blood vessel formation, and

Chapter 26
have been postulated, including the ATP-binding cassette (ABC) are similar in phenotype to Ang-1-deficient mice.297 Additional
transport proteins, selective exocytosis, and cell death or studies are needed to understand better the role of the Ang and
injury.19,280–282 During sublethal injury, cells may transiently Tie2 in ocular angiogenesis.
release FGF-2. Using cultured aortic endothelial cells, McNeil
and colleagues have demonstrated that mechanical wounding Pigment epithelium-derived factor
by scraping leads to efficient release of FGF-2 from injured cells19 PEDF is a 50-kDa serpin protease that was first discovered as a
(also reviewed by D’Amore281). In an experimental model of secreted protein from human fetal RPE cells.299 PEDF is the most

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


optic nerve crush in the mouse, FGF-2 immunostaining is dra- potent antiangiogenic growth factor identified to date.300 PEDF
matically increased in the retinal photoreceptor layer.283 Finally, has been demonstrated to promote apoptosis in proliferating
injury to the corneal epithelium leads to release of FGF-2, which endothelial cells through increasing the interaction of Fas ligand
binds to basement membrane.284 These and other observations with its receptor Fas. This seems to be the only cell type where
suggest that FGF-2 may act as a “wound hormone” both in PEDF promotes apoptosis.301 PEDF has also been shown to
maintenance of tissue integrity and repair after injury. signal cellular differentiation of retinoblastoma cells in vitro.299
Furthermore, a neuroprotective role for PEDF has been identi-
Integrins fied in neural cerebellar granule cells where PEDF caused the
The integrins are a family of cell adhesion proteins that are expression of antiapoptotic genes through the activation of
heterodimer combinations of 15 α and 8 β subunits, and are NF-κB.302 In retinal degeneration slow (RDS) mutant mice,
important regulators of angiogenesis. They mediate endothelial PEDF was shown to protect photoreceptors from undergoing
cell adhesion to the extracellular matrix, which facilitates pro- apoptosis.303 Thus, the signaling cascades that are activated by
liferation, migration, and response to prosurvival or apoptotic PEDF lead to cell-specific actions to promote both survival and
signals in the formation of new vessels.285 Different integrins cell death.
can bind to the same ligand, but initiate different intracellular Immunohistochemical studies have shown PEDF to be
signaling pathways. Also, one integrin can bind to multiple expressed in the RPE cells, the interphotoreceptor matrix, gan-
ligands; in addition to extracellular matrix components, ICAMs glion cells, and in the ciliary neuroepithelium.304,305 PEDF is
important for leukocyte adhesion can also be bound to also bound to components of the extracellular matrix. In vitro,
integrins.286 hypoxia causes a decrease in PEDF expression while VEGF
αvβ3 has been demonstrated to inhibit endothelial cell apopto- is increased.300 In vivo, levels of PEDF were shown to be
sis in newly sprouting blood vessels through the regulation of decreased in the vitreous of patients with proliferative DR
NF-κB.287 αvβ3 and αvβ5 are highly expressed on angiogenic endo- and wet AMD, such that PEDF levels correlated inversely
thelial cells and have been demonstrated in neovascular tissue with neovascularization.306–308
of experimental models and in clinical specimens.288,289 Fried- PEDF may have therapeutic potential as both a neuroprotec-
lander and colleagues found both αvβ3 and αvβ5 expressed in tive and antiangiogenic agent. The fact that it is both
neovascular tissue from eyes with proliferative DR, whereas differentiation-promoting and antiangiogenic has popularized
only αvβ3 was expressed in neovascular tissue from AMD and its study in tumor research. Using gene therapy approaches to
ocular histoplasmosis syndrome.289 Blocking the integrins halts administer PEDF may also be important in treating ocular neo-
angiogenesis in the chick CAM and corneal neovascularization vascularization. Both intravitreal and subretinal adenoviral-
models, and integrin binding also partially suppresses retinal associated PEDF have been used to treat retinal ischemia and
neovascularization.290,291 CNV in mouse models.309,310

Ang and Tie2 Matrix metalloproteinases


Ang-1 is a 70-kDa glycoprotein that is chemotactic for endothe- The extracellular matrix of the microvasculature is a highly
lial cells, and is postulated to play a role in the assembly of dynamic structure containing collagens, laminins, fibronectins,
nonendothelial cell components and the formation of capillary proteoglycans, and other proteins. Growth factors such as FGF-2
sprouts.292,293 Ang-1 binds to Tie2, a tyrosine kinase receptor, are localized there and integrins mediate interactions between
which is expressed on endothelial cells and early hematopoietic cells and the extracellular matrix. The extracellular matrix is
cells.294,295 Knockouts of the gene encoding Ang-1 or its receptor remodeled during development and angiogenesis, with degra-
Tie2 are embryologically lethal, with failure to recruit smooth- dation of existing matrix and synthesis of new matrix material
muscle and pericyte precursors. Tie2 has also been demonstrated permitting migration and proliferation of endothelial cells. Deg-
in quiescent and angiogenic vasculature in adults, and may play radation is accomplished by 16 known MMPs and plasmin.
a role in vascular maintenance.295,296 Ang-2 also binds strongly to Plasmin is synthesized as a latent proenzyme, plasminogen,
Tie2 but does not result in phosphorylation of the receptor; which requires proteolytic activation by enzymes such as
instead, it acts as a competitive inhibitor preventing Ang-1 urokinase-type plasminogen activator. This activation can be
binding.297 It has been postulated that Ang-2 binding mediates inhibited by plasminogen activator inhibitors. The MMPs are
endothelial cell survival signals by making them more respon- likewise synthesized as latent proenzymes and require proteo-
sive to VEGF, resulting in neovascularization. However, with lytic activation. The active MMPs can be inhibited by four known
VEGF inhibition, Ang-2 binding results in apoptosis.297 In the specific tissue inhibitors (TIMPS) and α-macroglobulin. For a
review of the MMPs and their inhibitors, see Hadler-Olsen the effects of other growth factors such as bFGF and VEGF.275
et al.311 This hypothesis is supported by the fact that TGF-β and IL-1
572 VEGF has been shown to induce tissue factor and MMP induce VEGF expression in cultured choroidal fibroblasts.327
production in endothelial cells, and MMP production in CTGF is a proangiogenic and profibrotic growth factor that is
smooth-muscle cells via Flt-1.312,313 AGE also caused increased expressed in stromal cells in human CNV.76 It also plays a role
MMP-2 mRNA in choroidal endothelial cells in vitro.314 Another in mediating and modulating the effects of other growth factors;
Section 2

interaction has been demonstrated between MMP-2 and the CTGF is upregulated in vitro by VEGF in choroidal endothelial
integrin αvβ3, which are functionally associated on the surface cells and by TGF-β in RPE cells. However, the relative impor-
of angiogenic blood vessels; upon their binding, collagenolytic tance of these growth factors, in comparison with VEGF, has not
activity is increased. A naturally occurring fragment of MMP-2, been determined.
termed PEX, has also been shown to prevent binding of MMP-2
to αvβ3, acting to decrease the proteolytic activity.315 Thus, CONCLUSIONS
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

endogenous MMP fragments and TIMPs serve to regulate the


Neovascularization is a tightly regulated cascade of events that
invasive new vessels. MMP-9 is one of the major inducible
includes molecular signaling of vascular assembly and matura-
MMPs. Its levels in the basal state seem to be suppressed
tion in the recruitment of pericytes, smooth muscle, and endo-
by the energy sensor AMP-activated protein kinase (AMPK).316
thelial cells.
Human neutrophils have been shown uniquely to release
A better understanding of the processes that regulate intra­
TIMP-free MMP-9 to provide a potent catalytic stimulator of
ocular neovascularization in retinal vascular diseases will help
angiogenesis.317
direct strategies for therapeutic intervention. There are numer-
Angiostatin and endostatin ous pro- and antiangiogenic molecules, and the interaction and
influences that regulate the cellular response to external stimuli
Angiostatin is a 38-kDa peptide identified in a murine model of
are complex. Among the angiogenic factors, VEGF may be par-
Lewis lung carcinoma, where it inhibited tumor metastasis at
ticularly significant. Among the antiangiogenic molecules, PEDF
distant sites.318 It was found to be an internal fragment of plas-
may play an important role.
minogen. This fragment contains kringle structures, which based
External stimuli may influence not only angiogenic pathways
on their disulfide architecture, have been shown to inhibit angio-
but also are likely to affect apoptosis, immune responses, cell
genesis. Angiostatin contains four out of the five plasminogen
cycle and proliferation, and alterations in free radical signaling.
kringle structures. Once angiostatin is cleaved from the parent
All of these influences are likely to have a complex interaction
plasminogen molecule, it has been shown specifically to inhibit
and role in new blood vessel growth.
endothelial cell proliferation.318 It inhibited neovascularization
We are just beginning to understand the roles of various medi-
in the chick CAM assay and inhibited neovascularization and
ators of angiogenesis. Maintaining homeostasis is a dynamically
growth of metastases in a mouse tumor model.
regulated process involving many signaling pathways and inter-
Endostatin is a 20-kDa peptide isolated from murine heman-
actions between pathways, where the same molecules may have
gioendothelioma, and has been identified as the C-terminal frag-
different roles under different conditions.
ment of collagen XVIII.319 Endostatin inhibits endothelial cell
In summary, ocular neovascularization is complex. The effects
proliferation and angiogenesis in vivo, including corneal neo-
may be tissue-, organ-, and cellular-specific. It is important to
vascularization. Mice that lack collagen XVIII form larger and
establish responses in ocular models as the environment may
leakier laser-induced CNV.46 Finally, circulating endostatin has
not necessarily mimic other tissues or even diseases.
been identified in human plasma.320 Both angiostatin and end-
ostatin have been proposed as potential therapeutic agents to REFERENCES
treat ocular neovascularization; however, up to now they have 1. Denekamp J. Vascular endothelium as the vulnerable element in tumours.
Acta Radiol Oncol 1984;23:217–25.
not found success in clinical use. 2. Ausprunk DH, Folkman J. Migration and proliferation of endothelial cells in
preformed and newly formed blood vessels during tumor angiogenesis.
Advanced glycosolation end products Microvasc Res 1977;14:53–65.
3. Grant MB, Caballero S, Brown GA, et al. The contribution of adult hemato-
AGEs are formed by nonenzymatic protein glycation. They have poietic stem cells to retinal neovascularization. Adv Exp Med Biol 2003;522:
been localized in basal laminar/linear deposits or soft drusen in 37–45.
4. Grant MB, May WS, Caballero S, et al. Adult hematopoietic stem cells provide
AMD.39 In surgically excised CNV, AGE deposition is spatially functional hemangioblast activity during retinal neovascularization. Nat Med
associated with RPE that express VEGF.39 AGEs induce VEGF 2002;8:607–12.
5. Tomita M, Adachi Y, Yamada H, et al. Bone marrow-derived stem cells can
expression by RPE cells in vitro,321 and growth of RPE on AGE differentiate into retinal cells in injured rat retina. Stem Cells 2002;20:
leads to the upregulation of genes associated with RPE aging322 279–83.
6. Tomita M, Yamada H, Adachi Y, et al. Choroidal neovascularization is pro-
and downregulation of cathepsin D expression.323 AGEs are also vided by bone marrow cells. Stem Cells 2004;22:21–6.
involved in lipofuscin formation in RPE cells.324 Reactive oxygen 7. Ashton N. Retinal vascularization in health and disease. Am J Ophthalmol
1957;44:7–24.
intermediates are generated in parallel with AGEs either directly 8. Michaelson I. The mode of development of the vascular system of the
or through AGE–RAGE interaction.325 Consistent with the ability retina, with some observations on its significance for certain retinal disease.
of AGEs to induce VEGF expression in vitro,321 AGEs can also Trans Ophthalmol Soc UK 1948;68:137–80.
9. Gartner S, Henkind N. Neovascularization of the iris (rubeosis iridis).
induce angiogenesis in vivo.326 Surv Ophthalmol 1978;22:291–312.
10. Henkind P. Ocular neovascularization. The Krill memorial lecture. Am J
Other growth factors Ophthalmol 1978;85:287–301.
11. Wise G. Retinal neovascularization. Trans Ophthalmol Soc UK 1956;54:
PDGF expression has been reported in the outer nuclear layer of 729–826.
12. Pournaras CJ, Miller JW, Gragoudas ES, et al. Systemic hyperoxia decreases
the macula from patients with AMD.77 RPE cells in CNV are vascular endothelial growth factor gene expression in ischemic primate retina.
strongly immunoreactive for TGF-β, and may act by modulating Arch Ophthalmol 1997;115:1553–8.
13. Pournaras CJ, Tsacopoulos M, Strommer K, et al. Experimental retinal branch 43. Vranka JA, Johnson E, Zhu X, et al. Discrete expression and distribution
vein occlusion in miniature pigs induces local tissue hypoxia and vasoprolif- pattern of TIMP-3 in the human retina and choroid. Curr Eye Res 1997;16:
erative microangiopathy. Ophthalmology 1990;97:1321–8. 102–10.
14. Liu Y, Cox SR, Morita T, et al. Hypoxia regulates vascular endothelial growth 44. Chong NH, Keonin J, Luthert PJ, et al. Decreased thickness and integrity of 573
factor gene expression in endothelial cells. Identification of a 5’ enhancer. the macular elastic layer of Bruch’s membrane correspond to the distribution
Circ Res 1995;77:638–43. of lesions associated with age-related macular degeneration. Am J Pathol
15. Maxwell PH, Dachs GU, Gleadle JM, et al. Hypoxia-inducible factor-1 modu- 2005;166:241–51.

Chapter 26
lates gene expression in solid tumors and influences both angiogenesis and 45. Yu HG, Liu X, Kiss S, Connolly E, et al. Increased choroidal neovasculariza-
tumor growth. Proc Natl Acad Sci USA 1997;94:8104–9. tion following laser induction in mice lacking lysyl oxidase-like 1.
16. Tsuzuki Y, Fukumura D, Oosthuyse B, et al. Vascular endothelial growth Invest Ophthalmol Vis Sci 2008;49:2599–605.
factor (VEGF) modulation by targeting hypoxia-inducible factor-1alpha – 46. Marneros AG, She H, Zambarakji H, et al. Endogenous endostatin inhibits
hypoxia response element – VEGF cascade differentially regulates vascular choroidal neovascularization. FASEB J 2007;21:3809–18.
response and growth rate in tumors. Cancer Res 2000;60:6248–52. 47. Okamoto N, Tobe T, Hackett SF, et al. Transgenic mice with increased expres-
17. Ryan SJ. Subretinal neovascularization after argon laser photocoagulation. sion of vascular endothelial growth factor in the retina: a new model of
Albrecht Von Graefes Arch Klin Exp Ophthalmol 1980;215:29–42. intraretinal and subretinal neovascularization. Am J Pathol 1997;151:281–91.
18. Wallow I, Johns K, Chandra BS, et al. Chorioretinal and choriovitreal neovas- 48. Schwesinger C, Yee C, Rohan RM, et al. Intrachoroidal neovascularization in
cularization after photocoagulation for proliferative diabetic retinopathy. A transgenic mice overexpressing vascular endothelial growth factor in the

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


clinicopathologic correlation. Ophthalmology 1985;92:523–32. retinal pigment epithelium. Am J Pathol 2001;158:1161–72.
19. McNeil PL, Muthukrishnan L, Warder E, et al. Growth factors are released by 49. Goetzl EJ, Banda MJ, Leppert D. Matrix metalloproteinases in immunity.
mechanically wounded endothelial cells. J Cell Biol 1989;109:811–22. J Immunol 1996;156:1–4.
20. Green WR, Enger C. Age-related macular degeneration histopathologic 50. Killingsworth MC, Sarks JP, Sarks SH. Macrophages related to Bruch’s mem-
studies. The 1992 Lorenz E. Zimmerman lecture. Ophthalmology 1993;100: brane in age-related macular degeneration. Eye 1990;4:613–21.
1519–35. 51. Grossniklaus HE, Ling JX, Wallace TM, et al. Macrophage and retinal pigment
21. Beatty S, Koh H, Phil M, et al. The role of oxidative stress in the pathogenesis epithelium expression of angiogenic cytokines in choroidal neovasculariza-
of age-related macular degeneration. Surv Ophthalmol 2000;45:115–34. tion. Mol Vis 2002;8:119–26.
22. Penfold PL, Madigan MC, Gillies MC, et al. Immunological and aetiological 52. Bora NS, Kaliappan S, Jha P, et al. CD59, a complement regulatory protein,
aspects of macular degeneration. Prog Retin Eye Res 2001;20:385–414. controls choroidal neovascularization in a mouse model of wet-type age-
23. AREDS. A randomized, placebo-controlled, clinical trial of high-dose supple- related macular degeneration. J Immunol 2007;178:1783–90.
mentation with vitamins C and E, beta carotene, and zinc for age-related 53. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in
macular degeneration and vision loss: AREDS report no. 8. Arch Ophthalmol age-related macular degeneration. Science 2005;308:385–9.
2001;119:1417–36. 54. Edwards AO, Ritter R, Abel KJ, et al. Complement factor H polymorphism
24. Klein R, Klein BE, Jensen SC, et al. The five-year incidence and progression and age-related macular degeneration. Science 2005;308:421–4.
of age-related maculopathy: the Beaver Dam Eye Study [see comments]. 55. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the
Ophthalmology 1997;104:7–21. complement regulatory gene factor H (HF1/CFH) predisposes individuals to
25. Wilcox DK. Vectorial accumulation of cathepsin D in retinal pigmented age-related macular degeneration. Proc Natl Acad Sci U S A 2005;102:7227–32
epithelium: effects of age. Invest Ophthalmol Vis Sci 1988;29:1205–12. 56. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant
26. Kennedy CJ, Rakoczy PE, Constable IJ. Lipofuscin of the retinal pigment increases the risk of age-related macular degeneration. Science 2005;308:
epithelium: a review. Eye 1995;9:763–71. 419–21.
27. Tombran-Tink J, Shivaram SM, Chader GJ, et al. Expression, secretion, and 57. Gold B, Merriam JE, Zernant J, et al. Variation in factor B (BF) and complement
age-related downregulation of pigment epithelium-derived factor, a serpin component 2 (C2) genes is associated with age-related macular degeneration.
with neurotrophic activity. J Neurosci 1995;15:4992–5003. Nat Genet 2006;38:458–62
28. Rakoczy PE, Zhang D, Robertson T, et al. Progressive age-related changes 58. Johnson PT, Betts KE, Radeke MJ, et al. Individuals homozygous for the age-
similar to age-related macular degeneration in a transgenic mouse model. related macular degeneration risk-conferring variant of complement factor H
Am J Pathol 2002;161:1515–24. have elevated levels of CRP in the choroid. Proc Natl Acad Sci U S A 2006;
29. Hageman GS, Mullins RF. Molecular composition of drusen as related to 103:17456–61
substructural phenotype. Mol Vis 1999;5:28. 59. Oh H, Takagi H, Takagi C, et al. The potential angiogenic role of macrophages
30. Mullins RF, Russell SR, Anderson DH, et al. Drusen associated with aging in the formation of choroidal neovascular membranes. Invest Ophthalmol Vis
and age-related macular degeneration contain proteins common to extracel- Sci 1999;40:1891–8.
lular deposits associated with atherosclerosis, elastosis, amyloidosis, and 60. Espinosa-Heidmann DG, Suner IJ, Hernandez EP, et al. Macrophage depletion
dense deposit disease. FASEB J 2000;14:835–46. diminishes lesion size and severity in experimental choroidal neovasculariza-
31. Sarks JP, Sarks SH, Killingsworth MC. Evolution of soft drusen in age-related tion. Invest Ophthalmol Vis Sci 2003;44:3586–92.
macular degeneration. Eye 1994;8:269–83. 61. Sakurai E, Anand A, Ambati BK, et al. Macrophage depletion inhibits experi-
32. Abdelsalam A, Del Priore L, Zarbin MA. Drusen in age-related macular mental choroidal neovascularization. Invest Ophthalmol Vis Sci 2003;44:
degeneration: pathogenesis, natural course, and laser photocoagulation- 3578–85.
induced regression. Surv Ophthalmol 1999;44:1–29. 62. Tsutsumi C, Sonoda KH, Egashira K, et al. The critical role of ocular-infiltrating
33. Starita C, Hussain AA, Patmore A, et al. Localization of the site of major macrophages in the development of choroidal neovascularization. J Leukoc
resistance to fluid transport in Bruch’s membrane. Invest Ophthalmol Vis Sci Biol 2003;74:25–32.
1997;38:762–7. 63. Gupta N, Brown KE, Milam AH. Activated microglia in human retinitis
34. Russell SR, Mullins RF, Schneider BL, et al. Location, substructure, and com- pigmentosa, late-onset retinal degeneration, and age-related macular degen-
position of basal laminar drusen compared with drusen associated with aging eration. Exp Eye Res 2003;76:463–71.
and age-related macular degeneration. Am J Ophthalmol 2000;129:205–14. 64. Tuo J, Smith B, Bojanowski CM, et al. The involvement of sequence variation
35. Mullins RF, Aptsiauri N, Hageman GS. Structure and composition of drusen and expression of CX3CR1 in the pathogenesis of age-related macular degen-
associated with glomerulonephritis: implications for the role of complement eration. FASEB J 2004;18:1297–9.
activation in drusen biogenesis. Eye (Lond) 2001;15:390–5. 65. Chan CC, Tuo J, Bojanowski CM, et al. Detection of CX3CR1 single nucleotide
36. Hageman GS, Luthert PJ, Victor Chong NH, et al. An integrated hypothesis polymorphism and expression on archived eyes with age-related macular
that considers drusen as biomarkers of immune-mediated processes at degeneration. Histol Histopathol 2005;20:857–63.
the RPE-Bruch’s membrane interface in aging and age-related macular 66. Combadière C, Feumi C, Raoul W, et al. CX3CR1-dependent subretinal
degeneration. Prog Retin Eye Res 2001;20:705–32. microglia cell accumulation is associated with cardinal features of age-related
37. Anderson DH, Mullins RF, Hageman GS, et al. A role for local inflammation macular degeneration. J Clin Invest 2007;117:2920–8.
in the formation of drusen in the aging eye. Am J Ophthalmol 2002;134: 67. Tuo J, Bojanowski CM, Zhou M, et al. Murine ccl2/cx3cr1 deficiency results
411–31. in retinal lesions mimicking human age-related macular degeneration.
38. Lutty GA, McLeod DS, Merges C, et al. Localization of vascular endothelial Invest Ophthalmol Vis Sci 2007;48:3827–36.
growth factor in human retina and choroid. Arch Ophthalmol 1996;114: 68. Chan C-C, Ross RJ, Shen D, et al. Ccl2/Cx3cr1-deficient mice: an animal model
971–7. for age-related macular degeneration. Ophthalmic Res 2008;40:124–8.
39. Ishibashi T, Murata T, Hangai M, et al. Advanced glycation end products in 69. Espinosa-Heidmann DG, Caicedo A, Hernandez EP, et al. Bone marrow-
age-related macular degeneration. Arch Ophthalmol 1998;116:1629–32. derived progenitor cells contribute to experimental choroidal neovasculariza-
40. Lafaut BA, Bartz-Schmidt KU, Vanden Broecke C, et al. Clinicopathological tion. Invest Ophthalmol Vis Sci 2003;44:4914–9.
correlation in exudative age related macular degeneration: histological dif- 70. Sengupta N, Caballero S, Mames RN, et al. The role of adult bone marrow-
ferentiation between classic and occult choroidal neovascularisation. Br J derived stem cells in choroidal neovascularization. Invest Ophthalmol Vis Sci
Ophthalmol 2000;84:239–43. 2003;44:4908–13.
41. Alexander JP, Bradley JM, Gabourel JD, et al. Expression of matrix metallo- 71. Lopez PF, Sippy BD, Lambert HM, et al. Transdifferentiated retinal pigment
proteinases and inhibitor by human retinal pigment epithelium. Invest epithelial cells are immunoreactive for vascular endothelial growth factor
Ophthalmol Vis Sci 1990;31:2520–8. in surgically excised age-related macular degeneration-related choroidal
42. Padgett LC, Lui GM, Werb Z, et al. Matrix metalloproteinase-2 and tissue neovascular membranes. Invest Ophthalmol Vis Sci 1996;37:855–68.
inhibitor of metalloproteinase-1 in the retinal pigment epithelium and inter- 72. Miyajima-Uchida H, Hayashi H, Beppu R, et al. Production and accumulation
photoreceptor matrix: vectorial secretion and regulation. Exp Eye Res of thrombospondin-1 in human retinal pigment epithelial cells. Invest
1997;64:927–38. Ophthalmol Vis Sci 2000;41:561–7.
73. Kaplan HJ, Leibole MA, Tezel T, et al. Fas ligand (CD95 ligand) controls 106. Abu el Asrar AM, Maimone D, Morse PH, et al. Cytokines in the vitreous of
angiogenesis beneath the retina. Nat Med 1999;5:292–7. patients with proliferative diabetic retinopathy. Am J Ophthalmol 1992;114:
74. Wyllie AH. Glucocorticoid-induced thymocyte apoptosis is associated with 731–6.
574 endogenous endonuclease activation. Nature 1980;284:555–6. 107. Funatsu H, Yamashita H, Shimizu E, et al. Relationship between vascular
75. Hinton DR, He S, Lopez PF. Apoptosis in surgically excised choroidal neo- endothelial growth factor and interleukin-6 in diabetic retinopathy. Retina
vascular membranes in age-related macular degeneration. Arch Ophthalmol 2001;21:469–77.
1998;116:203–9. 108. Meleth AD, Agron E, Chan CC, et al. Serum inflammatory markers in diabetic
76. He S, Jin ML, Worpel V, et al. Connective tissue growth factor and its role in retinopathy. Invest Ophthalmol Vis Sci 2005;46:4295–301.
Section 2

the pathogenesis of choroidal neovascularization. Arch Ophthalmol 109. Miyamoto K, Khosrof S, Bursell SE, et al. Prevention of leukostasis and vas-
2003;121:1283–8. cular leakage in streptozotocin-induced diabetic retinopathy via intercellular
77. Kliffen M, Sharma HS, Mooy CM, et al. Increased expression of angiogenic adhesion molecule-1 inhibition. Proc Natl Acad Sci U S A 1999;96:10836–41.
growth factors in age-related maculopathy. Br J Ophthalmol 1997;81:154–62. 110. Kern TS. Contributions of inflammatory processes to the development of the
78. Engerman RL. Pathogenesis of diabetic retinopathy. Diabetes 1989;38: early stages of diabetic retinopathy. Exp Diabetes Res 2007;2007:95103.
1203–6. 111. Iliaki E, Poulaki V, Mitsiades N, et al. Role of α4 integrin (cd49d) in the
79. Engerman RL, Kern TS. Retinopathy in animal models of diabetes. Diabetes pathogenesis of diabetic retinopathy. Invest Ophthalmol Vis Sci 2009;50:
Metab Rev 1995;11:109–20. 4890–904.
Basic Mechanisms of Injury in the Retina

80. Kowluru RA, Odenbach S. Role of interleukin-1beta in the pathogenesis of 112. Dinarello CA. Biologic basis for interleukin-1 in disease. Blood 1996;87:
Basic Science and Translation to Therapy

diabetic retinopathy. Br J Ophthalmol 2004;88:1343–7. 2095–147.


81. Joussen AM, Poulaki V, Mitsiades N, et al. Nonsteroidal anti-inflammatory 113. Jung YJ, Isaacs JS, Lee S, et al. IL-1beta-mediated up-regulation of HIF-1alpha
drugs prevent early diabetic retinopathy via TNF-alpha suppression. FASEB via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link between
J 2002;16:438–40. inflammation and oncogenesis. FASEB J 2003;17:2115–7.
82. Adamis AP, Berman AJ. Immunological mechanisms in the pathogenesis of 114. Frater-Schroder M, Risau W, Hallmann R, et al. Tumor necrosis factor type,
diabetic retinopathy. Semin Immunopathol 2008;30:65–84. a potent inhibitor of endothelial cell growth in vitro, is angiogenic in vivo.
83. Adamis AP, Miller JW, Bernal MT, et al. Increased vascular endothelial Proc Natl Acad Sci U S A 1987;84:5277–81.
growth factor levels in the vitreous of eyes with proliferative diabetic reti- 115. Leibovich SJ, Polverini PJ, Shepard HM, et al. Macrophage-induced angiogen-
nopathy. Am J Ophthalmol 1994;118:445–50. esis is mediated by tumour necrosis factor-alpha. Nature 1987;329:630–2.
84. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in 116. Kociok N, Radetzky S, Krohne TU, et al. Pathological but not physiological
ocular fluid of patients with diabetic retinopathy and other retinal disorders. retinal neovascularization is altered in TNF-Rp55-receptor-deficient mice.
N Engl J Med 1994;331:1480–7. Invest Ophthalmol Vis Sci 2006;47:5057–65.
85. Shimura M, Yasuda K, Nakazawa T, et al. Panretinal-photocoagulation before 117. Joussen AM, Poulaki V, Qin W, et al. Retinal vascular endothelial growth
pars plana vitrectomy influences vitreous level of interleukin-6 but not factor induces intercellular adhesion molecule-1 and endothelial nitric oxide
of vascular endothelial growth factor in patients with diabetic retinopathy. synthase expression and initiates early diabetic retinal leukocyte adhesion in
Int J Biomed Sci 2007;3:31–7. vivo. Am J Pathol 2002;160:501–9.
86. Shimura M, Yasuda K, Nakazawa K, et al. Panretinal photocoagulation induces 118. Joussen AM, Poulaki V, Le ML, et al. A central role for inflammation in the
pro-inflammatory cytokines and macular thickening in high-risk proliferative pathogenesis of diabetic retinopathy. FASEB J 2004;18:1450–2.
diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2009;247:1617–24. 119. Pruijt JFM, Fibbe WE, Laterveer L, et al. Prevention of interleukin-8-induced
87. Moradian S, Ahmadieh H, Malihi M, et al. Intravitreal bevacizumab in mobilization of hematopoietic progenitor cells in rhesus monkeys by inhibi-
active progressive proliferative diabetic retinopathy. Graefes Arch Clin Exp tory antibodies against the metalloproteinase gelatinase B (MMP-9). Proc Natl
Ophthalmol 2008;246:1699–705. Acad Sci U S A 1999;96:10863–8.
88. Cho WB, Oh SB, Moon JW, et al. Panretinal photocoagulation combined with 120. Van den Steen PE, Proost P, Wuyts A, et al. Neutrophil gelatinase B potenti-
intravitreal bevacizumab in high-risk proliferative diabetic retinopathy. ates interleukin-8 tenfold by aminoterminal processing, whereas it degrades
Retina 2009;29:516–22. CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood
89. Tonello M, Costa RA, Almeida FP, et al. Panretinal photocoagulation versus 2000;96:2673–81.
PRP plus intravitreal bevacizumab for high-risk proliferative diabetic 121. Descamps FJ, Van den Steen PE, Martens E, et al. Gelatinase B is diabetogenic
retinopathy (IBeHi study). Acta Ophthalmol 2008;86:385–9. in acute and chronic pancreatitis by cleaving insulin. FASEB J 2003;17:887–9.
90. Jorge R, Costa RA, Calucci D, et al. Intravitreal bevacizumab (Avastin) for 122. Notari L, Miller A, Martínez A, et al. Pigment epithelium-derived factor is a
persistent new vessels in diabetic retinopathy (IBEPE study). Retina 2006;26: substrate for matrix metalloproteinase type 2 and type 9: implications for
1006–13. downregulation in hypoxia. Invest Ophthalmol Vis Sci 2005;46:2736–47.
91. Boulton M, Gregor Z, McLeod D, et al. Intravitreal growth factors in prolifera- 123. Beránek M, Kolar P, Tschoplova S, et al. Genetic variations and plasma
tive diabetic retinopathy: correlation with neovascular activity and glycaemic levels of gelatinase A (matrix metalloproteinase-2) and atinase B (matrix
management. Br J Ophthalmol 1997;81:228–33. metalloproteinase-9) in proliferative diabetic retinopathy. Mol Vis 2008;14:
92. Dills DG, Moss SE, Klein R, et al. Association of elevated IGF-I levels with 1114–21.
increased retinopathy in late-onset diabetes. Diabetes 1991;40:1725–30. 124. Bell EF, Klein JM. Comments on oxygen toxicity and retinopathy (ROP) in the
93. Grant M, Russell B, Fitzgerald C, et al. Insulin-like growth factors in vitreous. premature infant. In: Bell EF, Segar JL, editors. Iowa neonatology handbook:
Studies in control and diabetic subjects with neovascularization. Diabetes pulmonary. Iowa: University of Iowa, Children’s Hospital, Department of
1986;35:416–20. Pediatrics; 1994.
94. Wang Q, Dills DG, Klein R, et al. Does insulin-like growth factor I predict 125. Ashton N. Oxygen and the growth and development of retinal vessels. In vivo
incidence and progression of diabetic retinopathy? Diabetes 1995;44:161–4. and in vitro studies. The XX Francis I. Proctor Lecture. Am J Ophthalmol
95. Punglia RS, Lu M, Hsu J, et al. Regulation of vascular endothelial growth 1966;62:412–35.
factor expression by insulin-like growth factor I. Diabetes 1997;46:1619–26. 126. Mintz-Hittner HA, Kennedy KA, Chuang AZ, et al. Efficacy of intravitreal
96. Kirstein M, Aston C, Hintz R, et al. Receptor-specific induction of insulin-like bevacizumab for stage 3+ retinopathy of prematurity. N Engl J Med
growth factor I in human monocytes by advanced glycosylation end product- 2011;364:603–15.
modified proteins. J Clin Invest 1992;90:439–46. 127. Hellstrom A, Perruzzi C, Ju M, et al. Low IGF-I suppresses VEGF-survival
97. Watanabe D, Suzuma K, Suzuma I, et al. Vitreous levels of angiopoietin 2 signaling in retinal endothelial cells: direct correlation with clinical retinopa-
and vascular endothelial growth factor in patients with proliferative diabetic thy of prematurity. Proc Natl Acad Sci U S A 2001;98:5804–8.
retinopathy. Am J Ophthalmol 2005;139:476–81. 128. Miller JW, Adamis A, Shima DT, et al. Vascular endothelial growth factor/
98. White RR, Shan S, Rusconi CP, et al. Inhibition of rat corneal angiogenesis by vascular permeability factor is temporally and spatially correlated with ocular
a nuclease-resistant RNA aptamer specific for angiopoietin-2. Proc Natl Acad angiogenesis in a primate model. Am J Pathol 1994;145:574–84.
Sci U S A 2003;100:5028–33. 129. Noma H, Funatsu H, Mimura T, et al. Vitreous levels of interleukin-6 and
99. Watanabe D, Suzuma K, Matsui S, et al. Erythropoietin as a retinal angiogenic vascular endothelial growth factor in macular edema with central retinal vein
factor in proliferative diabetic retinopathy. N Engl J Med 2005;353:782–92. occlusion. Ophthalmology 2009;116:87–93.
100. Sakanaka M, Wen T-C, Matsuda S, et al. In vivo evidence that erythropoietin 130. Suzuki Y, Nakazawa M, Suzuki K, et al. Expression profiles of cytokines and
protects neurons from ischemic damage. Proc Nat Acad Sci U S A 1998;95: chemokines in vitreous fluid in diabetic retinopathy and central retinal vein
4635–40. occlusion. Jpn J Ophthalmol 2011;55:256–63.
101. Becerra SP, Amaral J. Erythropoietin – an endogenous retinal survival factor. 131. Noma H, Funatsu H, Harino S, et al. Vitreous inflammatory factors in macular
N Engl J Med 2002;347:1968–70. edema with central retinal vein occlusion. Jpn J Ophthalmol 2011;55:248–55.
102. Dawson DW, Volpert OV, Gillis P, et al. Pigment epithelium-derived factor: 132. Noma H, Funatsu H, Mimura T, et al. Vitreous levels of pigment epithelium-
a potent inhibitor of angiogenesis. Science 1999;285:245–8. derived factor and vascular endothelial growth factor in macular edema with
103. Volpert OV, Zaichuk T, Zhou W, et al. Inducer-stimulated Fas targets acti- central retinal vein occlusion. Curr Eye Res 2011;36:256–63.
vated endothelium for destruction by anti-angiogenic thrombospondin-1 and 133. Brown DM, Campochiaro PA, Singh RP, et al. Ranibizumab for macular
pigment epithelium-derived factor. Nat Med 2002;8:349–57. edema following central retinal vein occlusion: six-month primary end point
104. Ogata N, Matsuoka M, Matsuyama K, et al. Plasma concentration of results of a phase III study. Ophthalmology 2010;117:1124–33.
pigment epithelium-derived factor in patients with diabetic retinopathy. 134. Brockhurst R, Schepens C, Okamura I. Uveitis. II. Peripheral uveitis: clinical
J Clin Endocrinol Metab 2007;92:1176–9. description, complications and differential diagnosis. Am J Ophthalmol
105. Spranger J, Meyer-Schwickerath R, Klein M, et al. TNF-alpha level in the 1960;49:1257–66.
vitreous body. Increase in neovascular eye diseases and proliferative diabetic 135. Shorb S, Irvine A, Kimura S. Optic disc neovascularization associated with
retinopathy. Med Klin (Munich) 1995;90:134–7. chronic uveitis. Am J Ophthalmol 1976;82:175–8.
136. Dees C, Arnold JJ, Forrester JV, et al. Immunosuppressive treatment of 166. Weber BH, Vogt G, Pruett RC, et al. Mutations in the tissue inhibitor of
choroidal neovascularization associated with endogenous posterior uveitis. metalloproteinases-3 (TIMP3) in patients with Sorsby’s fundus dystrophy.
Arch Ophthalmol 1998;116:1456–61. Nat Genet 1994;8:352–6.
137. Flaxel CJ, Owens SL, Mulholland B, et al. The use of corticosteroids for cho- 167. Fariss RN, Apte SS, Olsen BR, et al. Tissue inhibitor of metalloproteinases-3 is 575
roidal neovascularisation in young patients. Eye 1998;12:266–72. a component of Bruch’s membrane of the eye. Am J Pathol 1997;150:323–8.
138. Martidis A, Miller DG, Ciulla TA, et al. Corticosteroids as an antiangiogenic 168. Felbor U, Doepner D, Schneider U, et al. Evaluation of the gene encoding
agent for histoplasmosis-related subfoveal choroidal neovascularisation. the tissue inhibitor of metalloproteinases-3 in various maculopathies.

Chapter 26
J Ocul Pharmacol Ther 1999;15:425–8. Invest Ophthalmol Vis Sci 1997;38:1054–9.
139. Adan A, Mateo C, Navarro R, et al. Intravitreal bevacizumab (Avastin) injec- 169. De La Paz MA, Pericak-Vance MA, Lennon F, et al. Exclusion of TIMP3 as a
tion as primary treatment of inflammatory choroidal neovascularisation. candidate locus in age-related macular degeneration. Invest Ophthalmol Vis
Retina 2007;27:1180–6. Sci 1997;38:1060–5.
140. Chan WM, Lai TM, Liu DT, et al. Intravitreal bevacizumab (Avastin) for 170. Petrukhin K, Koisti MJ, Bakall B, et al. Identification of the gene responsible
choroidal neovascularisation secondary to central serous retinopathy, sec- for Best macular dystrophy. Nat Genet 1998;19:241–7.
ondary to punctuate inner choroidopathy, or of idiopathic origin. Am J 171. Marmorstein LY, Munier FL, Arsenijevic Y, et al. Aberrant accumulation of
Ophthalmol 2007;143:977–83. EFEMP1 underlies drusen formation in malattia leventinese and age-related
141. Hammond CJ, Webster AR, Snieder H, et al. Genetic influence on early age- macular degeneration. Proc Natl Acad Sci USA 2002;99:13067–72.
related maculopathy: a twin study. Ophthalmology 2002;109:730–6. 172. Heller SR; ADVANCE Collaborative Group. A summary of the ADVANCE

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


142. Baird PN, Guida E, Chu DT, et al. The epsilon2 and epsilon4 alleles of the Trial. Diabetes Care 2009;32(Suppl 2):S357–61.
apolipoprotein gene are associated with age-related macular degeneration. 173. Beulens JWJ, Patel A, Vingerling JR, et al. Effects of blood pressure lowering
Invest Ophthalmol Vis Sci 2004;45:1311–5. and intensive glucose control on incidence and progression of retinopathy
143. Asensio-Sanchez VM, Rodriguez-Martin T, Gala-Molina I, et al. Age- in patients with type 2 diabetes mellitus: a randomised controlled trial.
related macular degeneration: its association with the epsilon4 allele of the Diabetologia 2009;52:2027–36.
apolipoprotein E gene. Arch Soc Esp Oftalmol 2006;81:9–12. 174. Sun JK, Keenan HA, Cavallerano JD, et al. Protection from retinopathy and
144. Utheim OA, Ritland JS, Utheim TP, et al. Apolipoprotein E genotype and other complications in patients with type 1 diabetes of extreme duration: the
risk for development of cataract and age-related macular degeneration. Joslin 50-year Medalist study. Diabetes Care 2011;34:968–74.
Acta Ophthalmol 2008;86:401–3. 175. Keenan HA, Costacou T, Sun JK, et al. Clinical factors associated with resis-
145. Losonczy G, Fekete Á, Vokó Z, et al. Analysis of complement factor H Y402H, tance to microvascular complications in diabetic patients of extreme disease
LOC387715, HTRA1 polymorphisms and ApoE alleles with susceptibility to duration: the 50-year Medalist study. Diabetes Care 2007;30:1995–7.
age-related macular degeneration in Hungarian patients. Acta Ophthalmol 176. Liew G, Klein R, Wong TY. The role of genetics in susceptibility to diabetic
2011;89:255–62. retinopathy. Int Ophthalmol Clin 2009;49:35–52.
146. Brión M, Sanchez-Salorio M, Cortón M, et al. Genetic association study 177. Sorbinil Retinopathy Trial Research Group. A randomized trial of sorbinil, an
of age-related macular degeneration in the Spanish population. Acta aldose reductase inhibitor, in diabetic retinopathy. Arch Ophthalmol
Ophthalmol 2011;89:e12–22. 1990;108:1234–44.
147. Yang X, Hu J, Zhang J, et al. Polymorphisms in CFH, HTRA1 and CX3CR1 178. Lorenzi M. The polyol pathway as a mechanism for diabetic retinopathy:
confer risk to exudative age-related macular degeneration in Han Chinese. attractive, elusive, and resilient. Exp diabetes res 2007;2007:61038.
Br J Ophthalmol 2010;94:1211–4. 179. Ells A, Guemsey DL, Wallace K, et al. Severe retinopathy of prematurity
148. Zerbib J, Puche N, Richard F, et al. No association between the T280M poly- associated with FZD4 mutations. Ophthalmic Genet 2010;31:37–43.
morphism of the CX3CR1 gene and exudative AMD. Exp Eye Res 2011;93: 180. Haider MZ, Devarajan LV, Al-Essa M, et al. Angiotensin-converting enzyme
382–6. gene insertion/deletion polymorphism in Kuwaiti children with retinopathy
149. Liu MM, Agrón E, Chew E, et al. Copy number variations in candidate genes of prematurity. Biol Neonate 2002;82:84–8.
in neovascular age-related macular degeneration. Invest Ophthalmol Vis Sci 181. Cooke RW, Drury JA, Mountford R, et al. Genetic polymorphisms and
2011;52:3129–35. retinopathy of prematurity. Invest Ophthalmol Vis Sci 2004;45:1712–5.
150. Yang Z, Stratton C, Francis P, et al. Toll-like receptor 3 and geographic 182. Vannay AV, Dunai G, Banyasz I, et al. Association of genetic polymorphisms
atrophy in age-related macular degeneration. N Engl J Med 2008;359: of vascular endothelial growth factor and risk of proliferative retinopathy of
1456–63. prematurity. Pediatr Res 2005;57:396–8.
151. Sng CC, Cackett PD, Yeo IY, et al. Toll-like receptor 3 polymorphism rs3775291 183. Shastry BS, Qu X. Lack of association of the VEGF gene promoter (-634 G to
is not associated with choroidal neovascularization or polypoidal choroidal C and -460 C to T) polymorphism and the risk of advanced retinopathy of
vasculopathy in Chinese subjects. Ophthalmic Res 2011;45:191–6. prematurity. Graefes Arch Clin Exp Ophthalmol 2007;245:741–3.
152. Klein ML, Ferris 3rd FL, Francis PJ, et al. Progression of geographic atrophy 184. Shastry BS. Lack of association of VEGF (-2578 C to A) and ANG2 (-35 G to
and genotype in age-related macular degeneration. Ophthalmology C) gene polymorphisms with the progression of retinopathy of prematurity.
2010;117:1554–9, 1559.e1. Graefes Arch Clin Exp Ophthalmol 2009;247:859–60.
153. Edwards AO, Chen D, Fridley BL, et al. Toll-like receptor polymorphisms 185. Kwinta P, Bik-Multanowski M, Mitkowska Z, et al. The clinical role of vascu-
and age-related macular degeneration. Invest Ophthalmol Vis Sci 2008;49: lar endothelial growth factor (VEGF) system in the pathogenesis of retinopa-
1652–9. thy of prematurity. Graefes Arch Clin Exp Ophthalmol 2008;246:1467–75.
154. Gotoh N, Yamashiro K, Nakanishi H, et al. Haplotype analysis of the ARMS2/ 186. Banyasz I, Bokodl G, Vannay A, et al. Genetic polymorphisms of vascular
HTRA1 region in Japanese patients with typical neovascular age-related endothelial growth factor and angiopoietin 2 in retinopathy of prematurity.
macular degeneration or polypoidal choroidal vasculopathy. Jpn J Ophthalmol Curr Eye Res 2006;31:685–90.
2010;54:609–14. 187. Dunai G, Vasarhelyi B, Szabo M, et al. Published genetic variants in retinopa-
155. Friedrich U, Myers CA, Fritsche LG, et al. Risk- and non-risk-associated thy of prematurity: random forest analysis suggests a negligible contribution
variants at the 10q26 AMD locus influence ARMS2 mRNA expression but to risk and severity. Curr Eye Res 2008;33:501–5.
exclude pathogenic effects due to protein deficiency. Hum Mol Genet 2011;20: 188. Shastry BS. Assessment of the contribution of insulin-like growth factor -I
1387–99. receptor 3174 G to A polymorphism to the progression of advanced retinopa-
156. Hadley D, Orlin A, Brown G, et al. Analysis of six genetic risk factors highly thy of prematurity. Eur J Ophthalmol 2007;17:950–3.
associated with AMD in the region surrounding ARMS2 and HTRA1 on 189. Balogh A, Derzbach L, Vannay A, et al. Lack of association between insulin-
chromosome 10, region q26. Invest Ophthalmol Vis Sci 2010;51:2191–6. like growth factor – I receptor G3174A polymorphism and retinopathy of
157. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant prematurity. Graefes Arch Clin Exp Ophthalmol 2006;244:1035–8.
increases the risk of age-related macular degeneration. Science 2005;308: 190. Spiegler J, Gilhaus A, Konig IR, et al. Polymorphism in the renin–angiotensin
419–21. system and outcome of very low birth weight infants. Neonatology 2009;97:
158. Edwards AO, Ritter III R, Abel KJ, et al. Complement factor H polymorphism 10–4.
and age-related macular degeneration. Science 2005;308:421–4. 191. John BR, Loggins J, Yanamandra K. Angiotensin converting enzyme insertion/
159. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in deletion polymorphism does not alter sepsis outcome in ventilated very low
age-related macular degeneration. Science 2005;308:385–9. birth weight infants. J Perinatol 2005;25:205–9.
160. Francis PJ, Hamon SC, Ott J, et al. Polymorphisms in C2, CFB and C3 are 192. Mohamed S, Schaa K, Cooper ME, et al. Genetic contributions to the develop-
associated with progression to advanced age related macular degeneration ment of retinopathy of prematurity. Pediatr Res 2009;65:193–7.
associated with visual loss. J Med Genet 2009;46:300–7. 193. Christen WG, Glynn RJ, Manson JE, et al. A prospective study of cigarette
161. Yates JR, Sepp T, Matharu BK, et al. Complement C3 variant and the risk of smoking and risk of age-related macular degeneration in men. JAMA 1996;276:
age-related macular degeneration. N Engl J Med 2007;357:553–61. 1147–51.
162. Maller JB, Fagerness JA, Reynolds RC, et al. Variation in complement factor 194. Seddon JM, Willett WC, Speizer FE, et al. A prospective study of cigarette
3 is associated with risk of age-related macular degeneration. Nat Genet smoking and age-related macular degeneration in women. JAMA 1996;276:
2007;39:1200–1. 1141–6.
163. Spencer KL, Hauser MA, Olson LM, et al. Deletion of CFHR3 and CFHR1 195. Klein R, Klein BE, Moss SE. Relation of smoking to the incidence of age-related
genes in age-related macular degeneration. Hum Mol Genet 2008 1;17:971–7. maculopathy: the Beaver Dam Eye Study. Am J Epidemiol 1998;147:103–10.
164. Allikmets R, Singh N, Sun H, et al. A photoreceptor cell-specific ATP-binding 196. Mares JA, Voland RP, Sondel SA, et al. Healthy lifestyles related to subsequent
transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy prevalence of age-related macular degeneration. Arch Ophthalmol 2011;129:
[see comments]. Nat Genet 1997;15:236–46. 470–80.
165. Webster AR, Heon E, Lotery AJ, et al. An analysis of allelic variation in the 197. Age-Related Eye Disease Study Research Group. A randomized, placebo-
ABCA4 gene. Invest Ophthalmol Vis Sci 2001;42:1179–89. controlled, clinical trial of high-dose supplementation with vitamins C and E,
beta carotene, and zinc for age-related macular degeneration and vision loss: 228. Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor.
AREDS report No. 8. Arch Ophthalmol 2001;119:1417–36. Endocr Rev 1997;18:4–25.
198. Christen WG, Glynn RJ, Chew EY, et al. Folic acid, pyridoxine, and cyano­ 229. Jakeman LB, Winer J, Bennett GL, et al. Binding sites for vascular endothelial
576 cobalamin combination treatment and age-related macular degeneration in growth factor are localized on endothelial cells in adult rat tissues. J Clin
women: the Women’s Antioxidant and Folic Acid Cardiovascular Study. Invest 1992;89:244–53.
Arch Intern Med 2009;169:335–41. 230. Kim I, Ryan AM, Rohan R, et al. Constitutive expression of VEGF, VEGFR-1,
199. Zoppini G, Faccini G, Muggeo M, et al. Elevated plasma levels of soluble and VEGFR-2 in normal eyes. Invest Ophthalmol Vis Sci 1999;40:2115–21.
receptors of TNF-alpha and their association with smoking and microvascular 231. Ferrara N, Gerber H, LeCouter J. The biology of VEGF and its receptors.
Section 2

complications in young adults with type 1 diabetes. J Clin Endocrinol Metab Nat Med 2003;9:669–76.
2001;86:3805–8. 232. Matsumoto T, Claesson-Welsh L. VEGF receptor signal transduction.
200. Mol MJ, de Rijke YB, Demacker PN, et al. Plasma levels of lipid and cholesterol Sci STKE 2001;2001:RE21.
oxidation products and cytokines in diabetes mellitus and cigarette smoking: 233. Park JE, Chen HH, Winer J, et al. Placenta growth factor. Potentiation of
effects of vitamin E treatment. Atherosclerosis 1997;129:169–76. vascular endothelial growth factor bioactivity, in vitro and in vivo, and high
201. Wasada T, Kawahara R, Katsumori K, et al. Plasma concentration of immu- affinity binding to Flt-1 but not to Flk-1/KDR. J Biol Chem 1994;269:
noreactive vascular endothelial growth factor and its relation to smoking. 25646–54.
Metabolism 1998;47:27–30. 234. Barleon B, Sozzani S, Zhou D, et al. Migration of human monocytes in
Basic Mechanisms of Injury in the Retina

202. Keech AC, Mitchell P, Summanen PA, et al. Effect of fenofibrate on the need response to vascular endothelial growth factor (VEGF) is mediated via the
Basic Science and Translation to Therapy

for laser treatment for diabetic retinopathy (FIELD study): a randomised VEGF receptor Flt-1. Blood 1996;87:3336–43.
controlled trial. Lancet 2007;370:1687–97. 235. Hiratsuka S, Minowa O, Kuno J, et al. Flt-1 lacking the tyrosine kinase domain
203. ACCORD Study Group, et al. Effects of medical therapies on retinopathy is sufficient for normal development and angiogenesis in mice. Proc Natl
progression in type 2 diabetes. N Engl J Med 2010;363:233–44. Acad Sci U S A 1998;95:9349–54.
204. Forrester RM, Jefferson E, Naunton WJ. Oxygen and retrolental fibroplasia; 236. Pepper MS, Mandriota SJ. Regulation of vascular endothelial growth factor
a seven-year survey. Lancet 1954;ii:258–60. receptor-2 (Flk-1) expression in vascular endothelial cells. Exp Cell Res
205. Editorial. Oxygen restriction and retinopathy of prematurity. Lancet 1992;339: 1998;241:414–25.
961–2. 237. Lee P, Goishi K, Davidson AJ, et al. Neuropilin-1 is required for vascular
206. Szewczyk TS. Retrolental fibroplasia: etiology and prophylaxis. Am J development and is a mediator of VEGF-dependent angiogenesis in zebrafish.
Ophthalmol;1951;34:1609. Proc Natl Acad Sci U S A 2002;99:10470–5.
207. Bedrossian RH, Carmichael P, Ritter J. Retinopathy of prematurity (retrolental 238. Shima DT, Adamis A, Ferrara N, et al. Hypoxic induction of endothelial
fibroplasia) and oxygen. Am J Ophthalmol 1954;37:78. cell growth factors in retinal cells: identification and characterization of
208. Kinsey VE. Retrolental fibroplasia. Co-operative study of retrolental fibropla- vascular endothelial growth factor (VEGF) as the mitogen. Mol Med 1995;1:
sia and the use of oxygen. Arch Ophthalmol 1955;59:481–542. 182–93.
209. Forrester RM. Oxygen cerebral palsy and retrolental fibroplasia. Dev Med 239. Shweiki D, Itin A, Soffer D, et al. Vascular endothelial growth factor induced
Child Neurol 1964;6:648–50. by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 1992;359:
210. Swart-Van Der Hoeven JT, Mak TMB. Effects of oxygen on retrolental 843–5.
fibroplasia in premature infants, with report of two cases. Maadschr 240. Huang JC, Liu DY, Dawood MY. The expression of vascular endothelial
Kindergeneesk 1952;20:276. growth factor isoforms in cultured human endometrial stromal cells and its
211. Reynolds JD, Hardy RJ, Kennedy KA, et al. Lack of efficacy of light reduction regulation by 17beta-oestradiol. Mol Hum Reprod 1998;4:603–7.
in preventing retinopathy of prematurity. Light Reduction in Retinopathy of 241. Kuroki M, Voest EE, Amano S, et al. Reactive oxygen intermediates increase
Prematurity (LIGHT-ROP) Cooperative Group. N Engl J Med 1998;338: vascular endothelial growth factor expression in vitro and in vivo. J Clin
1572–6. Invest 1996;98:1667–75.
212. Connor KM, SanGiovanni JP, Lofqvist C, et al. Increased dietary intake of ω3 242. Satake S, Kuzuya M, Miura H, et al. Up-regulation of vascular endothelial
polyun- saturated fatty acids reduces pathological retinal angiogenesis. Nat growth factor in response to glucose deprivation. Biol Cell 1998;90:161–8.
Med 2007;7:868–73. 243. Neufeld G, Cohen T, Gengrinovitch S, et al. Vascular endothelial growth
213. Birch EE, Carlson SE, Hoffman DR, et al. The DIAMOND (DHA Intake And factor (VEGF) and its receptors. FASEB J 1999;13:9–22.
Measurement Of Neural Development) study: a double-masked, randomized 244. Shima DT, Gougos A, Miller JW, et al. Cloning and mRNA expression of
controlled clinical trial of the maturation of infant visual acuity as a function vascular endothelial growth factor in ischemic retinas of Macaca fascicularis.
of the dietary level of docosahexaenoic acid. Am J Clin Nutr 2010;91:848–59. Invest Ophthalmol Vis Sci 1996;37:1334–40.
214. Pepper MS, Ferrara N, Orci L, et al. Vascular endothelial growth factor (VEGF) 245. Dorey CK, Aouididi S, Reynaud X, et al. Correlation of vascular permeability
induces plasminogen activators and plasminogen activator inhibitor-1 in factor/vascular endothelial growth factor with extraretinal neovasculariza-
microvascular endothelial cells. Biochem Biophys Res Commun 1991;181: tion in the rat. Arch Ophthalmol 1996;114:1210–7.
902–6. 246. Pierce EA, Avery RL, Foley ED, et al. Vascular endothelial growth factor/
215. Senger DR, Claffey K, Benes JE, et al. Angiogenesis promoted by vascular vascular permeability factor expression in a mouse model of retinal
endothelial growth factor: regulation through alpha1beta1 and alpha2beta1 neovascularization. Proc Natl Acad Sci U S A 1995;92:905–9.
integrins. Proc Natl Acad Sci U S A 1997;94:13612–7. 247. Stone J, Chan-Ling T, Pe’er J, et al. Roles of vascular endothelial growth factor
216. Esser S, Wolburg K, Wolburg H, et al. Vascular endothelial growth factor and astrocyte degeneration in the genesis of retinopathy of prematurity.
induces endothelial fenestrations in vitro. J Cell Biol 1998;140:947–59. Invest Ophthalmol Vis Sci 1996;37:290–9.
217. Lu M, Perez VL, Ma N, et al. VEGF increases retinal vascular ICAM-1 248. Connolly DT, Heuvelman DM, Nelson R, et al. Tumor vascular permeability
expression in vivo. Invest Ophthalmol Vis Sci 1999;40:1808–12. factor stimulates endothelial cell growth and angiogenesis. J Clin Invest
218. Miyamoto K, Khosrof S, Bursell SE, et al. Vascular endothelial growth factor 1989;84:1470–8.
(VEGF)-induced retinal vascular permeability is mediated by intercellular 249. Plouet J, Schilling J, Gospodarowicz D. Isolation and characterization of a
adhesion molecule-1 (ICAM-1). Am J Pathol 2000;156:1733–9. newly identified endothelial cell mitogen produced by AtT-20 cells. EMBO J
219. Hutchings H, Ortega N, Plouet J. Extracellular matrix-bound vascular 1989;8:3801–6.
endothelial growth factor promotes endothelial cell adhesion, migration, and 250. Takeshita S, Zheng L, Brogi E, et al. Therapeutic angiogenesis. A single
survival through integrin ligation. FASEB J 2003;17:1520–2. intraarterial bolus of vascular endothelial growth factor augments revas-
220. Tischer E, Gospodarowicz D, Mitchell R, et al. Vascular endothelial growth cularization in a rabbit ischemic hind limb model. J Clin Invest 1994;93:
factor: a new member of the platelet-derived growth factor gene family. 662–70.
Biochem Biophys Res Commun 1989;165:1198–206. 251. Tolentino MJ, Miller JW, Gragoudas ES, et al. Vascular endothelial growth
221. Ferrara N, Houck K, Jakeman L, et al. Molecular and biological properties factor is sufficient to produce iris neovascularization and neovascular glau-
of the vascular endothelial growth factor family of proteins. Endocr Rev coma in a nonhuman primate. Arch Ophthalmol 1996;114:964–70.
1992;13:18–32. 252. Tolentino MJ, Miller JW, Gragoudas ES, et al. Intravitreous injections of vas-
222. Houck KA, Leung DW, Rowland AM, et al. Dual regulation of vascular cular endothelial growth factor produce retinal ischemia and microangi­
endothelial growth factor bioavailability by genetic and proteolytic mecha- opathy in an adult primate. Ophthalmology 1996;103:1820–8.
nisms. J Biol Chem 1992;267:26031–7. 253. Adamis AP, Shima DT, Tolentino MJ, et al. Inhibition of vascular endothelial
223. Carmeliet P, Ng YS, Nuyens D, et al. Impaired myocardial angiogenesis and growth factor prevents retinal ischemia-associated iris neovascularization in
ischemic cardiomyopathy in mice lacking the vascular endothelial growth a nonhuman primate. Arch Ophthalmol 1996;114:66–71.
factor isoforms VEGF164 and VEGF188. Nat Med 1999;5:495–502. 254. Aiello LP, Pierce EA, Foley ED, et al. Suppression of retinal neovascularization
224. Stalmans I, Ng YS, Rohan R, et al. Arteriolar and venular patterning in retinas in vivo by inhibition of vascular endothelial growth factor (VEGF) using
of mice selectively expressing VEGF isoforms. J Clin Invest 2002;109:327–36. soluble VEGF-receptor chimeric proteins. Proc Natl Acad Sci U S A 1995;92:
225. Salven P, Lymboussaki A, Heikkila H, et al. Vascular endothelial growth 10457–61.
factors VEGF-B and VEGF-C are expressed in human tumors. Am J Pathol 255. Robinson GS, Pierce EA, Rook SL, et al. Oligodeoxynucleotides inhibit retinal
1998;153:103–8. neovascularization in a murine model of proliferative retinopathy. Proc Natl
226. Witzenbichler B, Asahara T, Murohara T, et al. Vascular endothelial growth Acad Sci U S A 1996;93:4851–6.
factor-C (VEGF-C/VEGF-2) promotes angiogenesis in the setting of tissue 256. Kvanta A, Algvere PV, Berglin L, et al. Subfoveal fibrovascular membranes
ischemia. Am J Pathol 1998;153:381–94. in age-related macular degeneration express vascular endothelial growth
227. Meyer M, Clauss M, Lepple-Wienhues A, et al. A novel vascular endothelial factor. Invest Ophthalmol Vis Sci 1996;37:1929–34.
growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via sig- 257. Ishibashi T, Hata Y, Yoshikawa H, et al. Expression of vascular endothelial
nalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine growth factor in experimental choroidal neovascularization. Graefes Arch
kinases. EMBO J 1999;18:363–74. Clin Exp Ophthalmol 1997;235:159–67.
258. Schlingemann RO. Role of growth factors and the wound healing response in 292. Folkman J, D’Amore PA. Blood vessel formation: what is its molecular basis?
age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol 2004; Cell 1996;87:1153–5.
242:92–101. 293. Koblizek TI, Weiss C, Yancopoulos GD, et al. Angiopoietin-1 induces
259. Kwak N, Okamoto N, Wood JM, et al. VEGF is major stimulator in model of sprouting angiogenesis in vitro. Curr Biol 1998;8:529–32. 577
choroidal neovascularization. Invest Ophthalmol Vis Sci 2000;41:3158–64. 294. Davis S, Aldrich TH, Jones PF, et al. Isolation of angiopoietin-1, a ligand
260. Seo MS, Kwak N, Ozaki H, et al. Dramatic inhibition of retinal and choroidal for the TIE2 receptor, by secretion-trap expression cloning. Cell 1996;87:
neovascularization by oral administration of a kinase inhibitor. Am J Pathol 1161–9.

Chapter 26
1999;154:1743–53. 295. Witzenbichler B, Maisonpierre PC, Jones GD, et al. Chemotactic properties of
261. Gragoudas ES, Adamis AP, Cunningham Jr ET, et al. Pegaptanib for neovas- angiopoietin-1 and -2, ligands for the endothelial-specific receptor tyrosine
cular age-related macular degeneration. N Engl J Med 2004;351:2805–16. kinase Tie2. J Biol Chem 1998;273:18514–21.
262. CATT Research Group. Ranibizumab and bevacizumab for neovascular 296. Wong AL, Haroon ZA, Werner S, et al. Tie2 expression and phosphorylation
age-related macular degeneration. N Engl J Med 2011;364:897–908. in angiogenic and quiescent adult tissues. Circ Res 1997;81:567–74.
263. Rosenfeld PJ, Brown DM, Heier JS, et al. Ranibizumab for neovascular 297. Maisonpierre PC, Suri C, Jones PF, et al. Angiopoietin-2, a natural antagonist
age-related macular degeneration.N Engl J Med 2006;355:1419–31. for Tie2 that disrupts in vivo angiogenesis. Science 1997;277:55–60.
264. Brown DM, Kaiser PK, Michels M, et al. Ranibizumab versus verteporfin for 298. Asahara T, Chen D, Takahashi T, et al. Tie2 receptor ligands, angiopoietin-1
neovascular age-related macular degeneration. N Engl J Med 2006;355: and angiopoietin-2, modulate VEGF-induced postnatal neovascularization.
1432–44. Circ Res 1998;83:233–40.

Basic Mechanisms of Pathological Retinal and Choroidal Angiogenesis


265. Poulsen J. The Houssay phenomenon in man: recovery from retinopathy in a 299. Tombran-Tink J, Johnson LV. Neuronal differentiation of retinoblastoma cells
case of diabetes with Simmonds’ disease. Diabetes 1953;2:7–12. induced by medium conditioned by human RPE cells. Invest Ophthalmol Vis
266. Luft R, Olivecrona H, Ikkos D. Hypophysectomy in man: further experiences Sci 1989;30:1700–7.
in severe diabetes mellitus. Br Med J 1955;2:752–6. 300. Dawson DW, Volpert OV, Gillis SE, et al. Pigment epithelium-derived factor:
267. Sharp PS, Fallon TJ, Brazier OJ, et al. Long-term follow-up of patients who a potent inhibitor of angiogenesis. Science 1999;285:245–8.
underwent yttrium-90 pituitary implantation for treatment of proliferative 301. Volpert OV, Zaichuk T, Zhou W, et al. Inducer-stimulated Fas targets acti-
diabetic retinopathy. Diabetologia 1987;30:199–207. vated endothelium for destruction by anti-angiogenic thrombospondin-1 and
268. Hellstrom A, Perruzzi C, Ju M, et al. Low IGF-I suppresses VEGF-survival pigment epithelium-derived factor. Nat Med 2002;8:349–57.
signaling in retinal endothelial cells: direct correlation with clinical retin­ 302. Yabe T, Wilson D, Schwartz JN, Fkappa B. Activation is required for the
opathy of prematurity. Proc Natl Acad Sci U S A 2001;98:5804–8. neuroprotective effects of pigment epithelium-derived factor (PEDF) on
269. Smith LE, Kopchick JJ, Chen W, et al. Essential role of growth hormone in cerebellar granule neurons. J Biol Chem 2001;276:43313–9.
ischemia-induced retinal neovascularization. Science 1997;276:1706–9. 303. Cayouette M, Smith SB, Becerra S, et al. Pigment epithelium-derived factor
270. Hellstrom A, Engstrom E, Hard AL, et al. Postnatal serum insulin-like growth delays the death of photoreceptors in mouse models of inherited retinal
factor I deficiency is associated with retinopathy of prematurity and other degenerations. Neurobiol Dis 1999;6:523–32.
complications of premature birth. Pediatrics 2003;112:1016–20. 304. Karakousis PC, John SK, Behling KC, et al. Localization of pigment epithelium
271. Vlodavsky I, Folkman J, Sullivan R, et al. Endothelial cell-derived basic fibro- derived factor (PEDF) in developing and adult human ocular tissues. Mol Vis
blast growth factor: synthesis and deposition into subendothelial extracellular 2001;7:154–63.
matrix. Proc Natl Acad Sci U S A 1987;84:2292–6. 305. Ogata N, Wada M, Otsuji T, et al. Expression of pigment epithelium-derived
272. Mignatti P, Tsuboi R, Robbins E, et al. In vitro angiogenesis on the human factor in normal adult rat eye and experimental choroidal neovascularization.
amniotic membrane: requirement for basic fibroblast growth factor-induced Invest Ophthalmol Vis Sci 2002;43:1168–75.
proteinases. J Cell Biol 1989;108:671–82. 306. Holekamp NM, Bouck N, Volpert O. Pigment epithelium-derived factor is
273. Lobb RR, Alderman EM, Fett JW. Induction of angiogenesis by bovine brain deficient in the vitreous of patients with choroidal neovascularization due to
derived class 1 heparin-binding growth factor. Biochemistry 1985;24: age-related macular degeneration. Am J Ophthalmol 2002;134:220–7.
4969–73. 307. Ogata N, Nishikawa M, Nishimura T, et al. Inverse levels of pigment
274. Shing Y, Folkman J, Haudenschild C, et al. Angiogenesis is stimulated by epithelium-derived factor and vascular endothelial growth factor in the
a tumor-derived endothelial cell growth factor. J Cell Biochem 1985;29: vitreous of eyes with rhegmatogenous retinal detachment and proliferative
275–87. vitreoretinopathy. Am J Ophthalmol 2002;133:851–2.
275. Amin R, Puklin JE, Frank RN. Growth factor localization in choroidal neovas- 308. Ogata N, Tombran-Tink J, Nishikawa M, et al. Pigment epithelium-derived
cular membranes of age-related macular degeneration. Invest Ophthalmol Vis factor in the vitreous is low in diabetic retinopathy and high in rhegmatoge-
Sci 1994;35:3178–88. nous retinal detachment. Am J Ophthalmol 2001;132:378–82.
276. Baird A. Fibroblast growth factors. In: Roberts A, editor. Handbook of experi- 309. Mori K, Gehlbach S, Yamamoto E, et al. AAV-mediated gene transfer of
mental pharmacology: peptide growth factors and their receptors. Berlin: pigment epithelium-derived factor inhibits choroidal neovascularization.
Springer-Verlag; 1990. Invest Ophthalmol Vis Sci 2002;43:1994–2000.
277. Gospodarowicz D. Fibroblast growth factors. In: Pimental E, Perucho M, 310. Raisler BJ, Berns KI, Grant MB, et al. Adeno-associated virus type-2 expression
editors. Critical reviews in oncogenesis. Boca Raton, FL: CRC Press; 1989. of pigmented epithelium-derived factor or Kringles 1–3 of angiostatin reduce
278. Kitaoka T, Morse LS, Schneeberger S, et al. Expression of FGF5 in choroidal retinal neovascularization. Proc Natl Acad Sci U S A 2002;99:8909–14.
neovascular membranes associated with ARMD. Curr Eye Res 1997;16: 311. Hadler-Olsen E, Fadnes B, Sylte I, et al. Regulation of matrix metalloprotein-
396–9. ase activity in health and disease. FEBS J 2011;278:28–45.
279. Abraham JA, Mergia A, Whang JL, et al. Nucleotide sequence of a bovine 312. Wang H, Keiser JA. Vascular endothelial growth factor upregulates the
clone encoding the angiogenic protein, basic fibroblast growth factor. Science expression of matrix metalloproteinases in vascular smooth muscle cells: role
1986;233:545–8. of flt-1. Circ Res 1998;83:832–40.
280. Cooper DN, Barondes SH. Evidence for export of a muscle lectin from cytosol 313. Zucker S, Mirza H, Conner CE, et al. Vascular endothelial growth factor
to extracellular matrix and for a novel secretory mechanism. J Cell Biol induces tissue factor and matrix metalloproteinase production in endothelial
1990;110:1681–91. cells: conversion of prothrombin to thrombin results in progelatinase A
281. D’Amore PA. Modes of FGF release in vivo and in vitro. Cancer Metastasis activation and cell proliferation. Int J Cancer 1998;75:780–6.
Rev 1990;9:227–38. 314. Hoffmann S, Friedrichs U, Eichler W, et al. Advanced glycation end products
282. Kuchler K, Dohlman HG, Thorner J. The a-factor transporter (STE6 gene induce choroidal endothelial cell proliferation, matrix metalloproteinase-2
product) and cell polarity in the yeast Saccharomyces cerevisiae. J Cell Biol and VEGF upregulation in vitro. Graefes Arch Clin Exp Ophthalmol
1993;120:1203–15. 2002;240:996–1002.
283. Kostyk SK, D’Amore PA, Herman IM, et al. Optic nerve injury alters basic 315. Brooks PC, Silletti S, von Schalscha TL, et al. Disruption of angiogenesis by
fibroblast growth factor localization in the retina and optic tract. J Neurosci PEX, a noncatalytic metalloproteinase fragment with integrin binding activity.
1994;14:1441–9. Cell 1998;92:391–400.
284. Adamis AP, Meklir B, Joyce NC. In situ injury-induced release of basic- 316. Morizane Y, Thanos A, Takeuchi K, et al. AMP-activated protein kinase
fibroblast growth factor from corneal epithelial cells. Am J Pathol suppresses matrix metalloproteinase-9 expression in mouse embryonic
1991;139:961–7. fibroblasts. J Biol Chem 2011;286:16030–8.
285. Joseph-Silverstein J, Silverstein RL. Cell adhesion molecules: an overview. 317. Ardi VC, Kupriyanova TA, Deryugina EI, et al. Human neutrophils uniquely
Cancer Invest 1998;16:176–82. release TIMP-free MMP-9 to provide a potent catalytic stimulator of angio-
286. McEver RP. Adhesive interactions of leukocytes, platelets, and the vessel wall genesis. Proc Natl Acad Sci U S A 2007;104:20262–7.
during hemostasis and inflammation. Thromb Haemost 2001;86:746–56. 318. O’Reilly MS, Holmgren L, Shing Y, et al. Angiostatin novel angiogenesis
287. Scatena M, Almeida M, Chaisson ML, et al. NF-kappaB mediates alphavbeta3 inhibitor that mediates the suppression of metastases by a Lewis lung
integrin-induced endothelial cell survival. J Cell Biol 1998;141:1083–93. carcinoma. Cell 1994;79:315–28.
288. Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v 319. O’Reilly MS, Boehm T, Shing Y, et al. Endostatin: an endogenous inhibitor of
beta 3 for angiogenesis. Science 1994;264:569–71. angiogenesis and tumor growth. Cell 1997;88:277–85.
289. Friedlander M, Theesfeld CL, Sugita M, et al. Involvement of integrins alpha 320. Standker L, Schrader M, Kanse SM, et al. Isolation and characterization of the
v beta 3 and alpha v beta 5 in ocular neovascular diseases. Proc Natl Acad Sci circulating form of human endostatin. FEBS Lett 1997;420:129–33.
U S A 1996;93:9764–9. 321. Lu M, Kuroki M, Amano S, et al. Advanced glycation end products increase
290. Friedlander M, Brooks PC, Shaffer RW, et al. Definition of two angiogenic retinal vascular endothelial growth factor expression. J Clin Invest 1998;101:
pathways by distinct alpha v integrins. Science 1995;270:1500–2. 1219–24.
291. Hammes HP, Brownlee M, Jonczyk A, et al. Subcutaneous injection of a 322. Honda S, Farboud B, Hjelmeland LM, et al. Induction of an aging mRNA
cyclic peptide antagonist of vitronectin receptor-type integrins inhibits retinal retinal pigment epithelial cell phenotype by matrix-containing advanced gly-
neovascularization. Nat Med 1996;2:529–33. cation end products in vitro. Invest Ophthalmol Vis Sci 2001;42:2419–25.
323. McFarlane S, McMullen CB, Henry DN. Advanced glycation end products 325. Yan SD, Schmidt AM, Anderson GM, et al. Enhanced cellular oxidant stress
(AGEs) modulate cathepsin D expression in retinal pigment epithelium (RPE): by the interaction of advanced glycation end products with their receptors/
implications for age-related macular dysfunction. Invest Ophthalmol Vis Sci binding proteins. J Biol Chem 1994;269:9889–97.
578 2001;42:S414. 326. Okamoto T, Tanaka S, Stan AC, et al. Advanced glycation end products
324. Schutt F, Bergmann M, Holz FG, et al. Proteins modified by malondialdehyde, induce angiogenesis in vivo. Microvasc Res 2002;63:186–95.
4-hydroxynonenal, or advanced glycation end products in lipofuscin of 327. Kvanta A. Expression and regulation of vascular endothelial growth factor in
human retinal pigment epithelium. Invest Ophthalmol Vis Sci 2003;44: choroidal fibroblasts. Curr Eye Res 1995;14:1015–20.
3663–8.
Section 2
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy
Basic Mechanisms of Injury in the Retina Section 2

Blood–Retinal Barrier, Immune Privilege, Chapter

and Autoimmunity
Robert Katamay, Robert B. Nussenblatt 27 

INTRODUCTION BASIC CONCEPTS OF IMMUNOLOGY


More than 100 years ago a fascinating discovery was made. Innate immunity
Different tissue samples were placed into the anterior chamber
There are two systems of immunity, innate and adaptive, coex-
of dogs or rabbits and were then observed for 4 months. Sur-
isting to keep pathogens out. Innate immunity, phlyogenetically
prisingly, the induced intraocular inflammation was very
earlier, is nonspecific and immediate in response. It is the first
limited. Most of the tissue samples were not rejected as they
immune response mounted against invading pathogens. The
were known to occur from other parts of the body. At this
key components of innate immunity are a cadre of leukocytes
time, the eye as one of the first sites of immune privilege was
and plasma proteins that are capable of detecting and destroy-
recognized.1
ing pathogens. These leukocytes include polymorphonuclear
Today, we know that the eye is a unique and complex organ
cells, monocytes, macrophages, natural killer (NK) cells, eosino-
that has developed multiple mechanisms to protect itself against
phils, and basophils and the plasma proteins belong to families
frivolous immunological attacks and inflammation in order to
of the complement cascade, clotting cascade, and acute-phase
maintain its delicate structure and function. The blood–ocular
reactants.
barrier system is formed by the blood–aqueous barrier and the
inner and outer blood–retinal barrier. It restrictively limits the
passage of ions and proteins to maintain homeostasis and to Adaptive immunity
keep tissue-specific antigens sequestrated. Furthermore, many In contrast, adaptive immunity, as the name implies, is antigen-
active factors have been discovered in the eye that contribute to specific, adaptive, transferable, and has other cardinal features
a downregulatory immune environment (DIE).2 The blood– like immunologic memory and tolerance. Adaptive immunity
ocular barrier and the DIE are mechanisms that depend on each has evolved and developed novel molecules like the antibody
other’s proper function. They are central to preserving the and the T-cell receptor (TCR), which come in almost limitless
immune privilege of the eye. Immune privilege has served well varieties to recognize most molecules of biological interest,
to curb excessive inflammation and preserve function. However, including both self and nonself. These molecules, developed in
this privilege may be lost under certain conditions and situa- the absence of exogenous stimulation, have unique configura-
tions, resulting in retinal autoimmunity and ocular inflamma- tions that confer specificity in antigen recognition. The key
tion. A dysfunction of the inner blood–retinal barrier initiates cellular components of adaptive immunity are the T and B lym-
diabetic retinopathy.3,4 A breakdown of the blood–retinal barrier phocytes which possess unique receptors for recognizing bil-
can also be found in cystoid macular edema, while a loss of the lions of different antigenic epitopes.8 Each clone of lymphocyte
DIE is essential to many forms of uveitis and in all probability expresses molecularly identical receptors on the surface; hence,
to the development of age-related macular degeneration (AMD).5 in order to recognize billions of different epitopes, there are
For a long time, retinal autoimmunity has been perceived as billions of unique lymphocytes. Upon maturation in the thymus
pathogenic, and active suppression of retinal immunity was pre- (T cells) and the bone marrow (B cells), cells remain quiescent in
sumed necessary to maintain the health of the eye. However, G0 of the cell cycle until they encounter the complementary
studies have shown the presence of retinal autoantibodies in antigen of sufficient affinity to their receptors. Binding with the
normal controls,6 and constitutive expression of proinflamma- receptors in the presence of appropriate costimulation is the
tory ligands have been found in the normal retina in high con- initiating event to trigger an immune response targeting elimina-
centration (B Kim, M Pillai, Z Li, unpublished data). Moreover, tion of the complementary molecule. If this molecule is pathogen-
animal optic nerve injury studies revealed possible beneficial derived, activation will lead to the elimination of the pathogen,
roles of retinal autoimmunity in controlling collateral damage to but if the molecule is self-derived then activation will potentially
the retinal ganglion cells.7 Thus, retinal autoimmunity can be lead to autoimmunity, tissue injury, disease, and the destruction
viewed as a “double-edged sword,” with both protective and of host tissue. Activation of naive lymphocytes results in the
destructive effects. To understand retinal autoimmunity, we first emergence of clones of lymphocytes through proliferation, each
have to understand basic concepts in immunology and how with a unique receptor to recognize one epitope of the inciting
some of these immunological components are at work to main- antigen. The cytokine environment during activation of naive T
tain the eye’s immune privilege. cells is responsible for differentiation into three major types of
effector T cells. Interferon (IFN)-γ leads to Th1 cell production, an array of cytokines, including IL-2, IL-3, granulocyte–
while interleukin (IL)-4 is responsible for Th2 production. Some macrophage colony-stimulating factor, IFN-γ, and IL-4. These
580 Th1 and Th2 cell clones will become memory cells which are cytokines serve as growth and stimulation factors for the lym-
responsible for the enhanced immune response if the antigen is phocytes and APCs, hence amplifying the proliferation process.
re-encountered. The rest of Th1 will secrete cytokines targeted In addition, IL-2, IFN-γ, and IL-4 also promote the differentiation
at macrophages and other cells mediating cellular immunity. By of CD8+ T lymphocytes to mature cytotoxic T lymphoctes (CTLs)
Section 2

contrast, Th2 will stimulate B cells into proliferation and anti- which recognize antigen-derived peptides presented on class I
body production, mediating humoral immunity. Th17 are a MHC molecules. Upon activation, these CD8+ CTLs function as
recently discovered family of T helper cells that are capable of effectors to lyse the targeted cells and also produce proinflam-
producing IL-17. IL-1 and IL-23 seem to be needed for human matory cytokines, especially IFN-γ. The cytokine production by
Th17 differentiation.9 Th17 play an important role in combatting CD4+ T cells also promotes activation and differentiation of B
various bacterial and fungal species by producing IL-23. In con- lymphocytes. IFN-γ and IL-2 stimulate B cells to produce
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

trast, large amounts of Th17 can be found in autoimmune dis- complement-fixing immunoglobulin G (IgG) antibodies, while
eases like uveitis, multiple sclerosis, and rheumatoid arthritis.10 IL-4, IL-5, IL-6, and IL-10 result in the production of
Both immune and accessory cells, including those of the innate noncomplement-fixing IgG, IgE, or IgA antibodies. The final
immunity system, are activated to eliminate pathogens. In this products after the central processing in the lymphoid tissues are
way, adaptive immunity serves to enhance immune protection immune effectors. These are the CD4+, CD8+ T cells and B cells
through better-coordinated and more specific attacks using that have receptors specific for the inciting antigen. They are
innate immunity mechanisms. Within the genome lies the ability transported, predominantly via a hematogenous route, to the
to create antibodies and TCR for antigens that bind to self anti- site of the inciting antigens to execute their effector function,
gens with high affinity.11 Hence, the threat of autoimmunity is thereby completing the efferent loop of the immune response.
inborn and regulation is crucial. The ability to distinguish At the target sites, sites of infection and inflammation, vessels
between foreign and self, and the ability to regulate autoimmu- are leaky, the vascular endothelial cells display ligands that bind
nity, are critical for survival. to receptors on the immune cells, and chemokines secreted by
The immune response can be seen as analogous to the neural the local inflammatory cells serve to attract more immune effec-
reflex arc, which has an afferent limb, a central process, and an tor cells to the site. While the engagement and activation of B
efferent limb. In the immune afferent limb, antigens are cap- cells can be direct, activation of the T cells still needs the APCs
tured, processed, transported, and finally presented to the lym- for antigen recognition. The activated CD4+ T cells secrete cyto-
phocytes. As the naive lymphocytes can only be activated in the kines such as IFN-γ and tumor necrosis factor (TNF)-α which in
organized lymph tissues in secondary lymphoid organs such as turn attract and recruit the cells of the innate immune system,
lymph nodes, spleen, tonsils, and Peyer’s patches, it needs spe- such as monocytes, macrophages, and NK cells, to the site to
cialized cells to execute the immune afferent limb. These cells effect the actual destruction of the antigen or pathogen through
are professional antigen-presenting cells (APCs). Dendritic cells generation of cytotoxic products and phagocytosis of the patho-
and macrophages, both bone marrow-derived, have APC func- gens. B cells secrete antibodies specific for an antigen which will
tions serving to capture antigens through phagocytosis and result in direct killing/lysis of the target and also recruit poly-
endocytosis, process the antigens, and present the processed morphonuclear cells via complement activation and the comple-
antigen in conjunction with special major histocompatibility ment activation products respectively. In this way, the adaptive
complex (MHC) molecules on their cell surface. In addition, the immune system serves to direct the innate immune system
APCs provide the necessary costimulation needed for lympho- components to target the inciting antigen or pathogen.
cyte activation by upregulating an array of surface molecules
(CD80, CD86, intercellular adhesion molecule-1, lymphocyte Immune regulation
function-associated molecule-3, and CD40) that function as The immune system can be regulated at any part of the immune
ligands for receptors expressed by the lymphocytes. They also response loop, i.e., afferent limb, central process, and efferent
secrete cytokines such as IL-12, IL-6, IL-10, and IL-1β serving limb. Regulation of the autoimmune response can be via any of
similar costimulation functions. Costimulation is required as the the following mechanisms: central tolerance – clonal deletion
second signal for full activation of the lymphocytes, independent and peripheral tolerance – clonal anergy, T-cell suppression,
of the first signal, which is the antigen and the lymphocyte recep- immune deviation, immunologic ignorance, and antigen seques-
tor engagement.12 There are differences in antigen presentation tration. Since the genome has the capability to generate both
to B and T lymphocytes. For B lymphocytes, the receptor, which self- and nonself-recognizing antibodies and TCRs, mechanisms
is a surface-bound antibody, can engage a naive antigen directly must be in place to contain and prevent the activation of self-
while TCRs can only recognize peptide fragments presented on reactive T cells. During lymphocyte development, both T and B
special surface molecules (MHC classes I and II). MHC class I, cells with receptors that recognize self molecules with high
which is present in most cells, presents peptides derived from affinity are clonally deleted via apoptosis (central tolerance).13
protein degradation in the cytoplasm (intracellular antigens However the process is not foolproof, as autoreactive T and B
such as viral and intracellular microbe products), while MHC cells in the periphery exist even in normal individuals. Many
class II molecules, which are present in APCs and lymphocytes, tissue-specific antigens, like the eye-restricted molecules, may
present peptides from phagocytic vesicles (extracellular antigens not be expressed in the thymus or cells may have escaped the
from the microenvironment). selection process by the central tolerance mechanism in the
Upon activation, the first cells to respond are the CD4+ T cells thymus without encountering the specific self antigen. Hence,
specific for the presented peptide on the MHC class II molecules the potential for induction and expression of autoimmunity still
of the APCs. The CD4+ lymphocytes will proliferate and secrete exists, and mechanisms to contain these autoreactive immune
cells are crucial to prevent autoimmunity. Evidence for periph- Foxp3 results in hyperactivation of T cells and severe autoim-
eral tolerance can be found in all phases of the immune response, mune disease.23 Natural and induced Tregs maintain peripheral
i.e., afferent limb, central process, and efferent limb.14,15 The tolerance by modifying the functions of other T cells, both CD4+ 581
afferent limb depends heavily on the functional properties of and CD8+ populations, either directly through T-cell to T-cell
the APCs which offer many opportunities for modulation. First, interactions, or indirectly through APCs.24 Hence they are crucial

Chapter 27
the antigen can be sequestrated and prevented from contact for maintaining immune homeostasis and controlling autoim-
with the APCs by physical barriers or by rendering APCs inca- munity in order to keep the immune system healthy.
pable of antigen capture in the microenvironment (sequestra-
tion). Second, the ability of the APC to degrade, process, and BLOOD–OCULAR BARRIER
express the antigens can be inhibited. Third, the antigen-bearing At the end of the 19th century, the German bacteriologist and
APCs may be prevented from migrating to the lymphoid tissues. immunologist Paul Ehrlich experimented with staining tissues
Lastly, the ability of the APCs to generate the costimulatory

Blood–Retinal Barrier, Immune Privilege, and Autoimmunity


in animal models. When he injected dyes into the bloodstream,
signal can be inhibited or different costimulatory signals may be almost all tissues were colored intensively. However, the brain
produced (immune deviation). remained unstained.25 A few years later, one of Ehrlich’s stu-
Regulation of APC function can be seen as a form of dents, Edwin Goldmann, performed the experiment the other
immunologic ignorance where reactive autoimmune cells are way around, by injecting dyes into the cerebrospinal fluid. This
present but never have the opportunity to encounter its antigen time, the brain became stained and the rest of the body remained
in a proper immunogenic form due to a deficiency in antigen dye-free. Goldmann correctly concluded that there was a res­
presentation. In the central processing phase of the immune trictive barrier mechanism inhibiting the passage of solubles
response, two signals are required for full activation of the between the two compartments, the blood and the cerebrospinal
lymphocyte. The absence of costimulation in the presence of fluid.26 In the same year, in 1913, Schnaudigel found a similar
antigen-specific stimulation renders the T cells unresponsive barrier for trypan blue in the retina of rabbits.27
to further antigen stimulation, even in the presence of costimu- Today, we know that the eye consists of several anatomical
latory signals (anergy).16 On the other hand, too much costimu- structures that limit flux between the blood and most parts of
lation in the presence of high antigen dose may lead to profound the eye itself.28 The blood–ocular barrier is represented by two
cell activation and apoptosis. Both mechanisms will result in main components: the blood–aqueous barrier and the blood–
tolerance. retinal barrier. The blood–aqueous barrier is formed by the non-
It is also known that APCs from certain immune-privileged pigmented layer of the ciliary body epithelium and by the
sites such as the eye, and APCs in the gut processing orally endothelium of the iridal capillaries. The blood–retinal barrier is
ingested antigens, may cause some immune cells to develop into located at two levels. The inner barrier is the nonfenestrated
immunoregulatory cells like IL-10-secreting Tr1 cells or trans- capillaries of the retinal vessels, whereas the outer barrier is the
forming growth factor-β (TGF-β) secreting Th3 cells. These cells tight junctions of the RPE, enabling a high degree of control of
have in common that they are adaptively regulatory. They fluid permeability.29
receive a suppressive potential following specific antigen stimu- Limiting the passage of ions enables the eye to maintain
lation in a particular cytokine milieu.17 homeostasis in the aqueous humor and in the retina while the
In the efferent limb, immune regulation can occur via targeting barrier also keeps small and large molecules like drugs from
at the level of antigen-specific immune effector cells. One entering the eye. Specially hydrophilic substances show a low
such mechanism would be through Fas ligand – Fas-induced permeability across the blood–ocular barrier.30 Furthermore,
apoptosis of lymphocytes.18,19 The immune-reactive lymphocytes the barrier has an essential immunologic function: the seques-
express the CD95 receptor (Fas receptor). On encountering the tration of tissue-specific antigens is crucial to preserve the
Fas ligand, which is expressed in immuno-privileged eye tissues immune privilege of the eye, and, therefore, to avoid intraocu-
such as the cornea and retina, these immune cells undergo apop- lar inflammation. On the other hand, inflammation can cause
tosis. Other molecular mechanisms, such as TNF-α production breakdown of the blood–retinal barrier, leading to autoimmu-
by immune and accessory cells, such as Müller cells and retinal nity and inflammation, and allowing drugs and other mole-
pigment epithelial (RPE) cells during antigen encounter, can also cules to penetrate into the eye. Considering its essential
result in apoptosis, thereby deleting these immune-reactive cells. function it is not surprising that many of the most common eye
Similar to the central process, effector cells need costimulatory diseases are directly associated with alterations of the blood–
signals to be activated and the expression of inappropriate ocular barrier.
costimulatory signals such as IL-10 or TGF-β by APCs can lead
to anergy or reprogramming of lymphocytes into immunoregu-
latory cells secreting immunosuppressive cytokines, thereby
Blood–retinal barrier
further suppressing autoimmunity.20,21 in diabetic retinopathy
An important regulator of immune responses is the CD4+ The initial disorder causing both nonproliferative and prolif-
CD25high forkhead box protein 3 (Foxp3)+ regulatory T cell erative diabetic retinopathy is chronic hyperglycemia leading
(Treg). There are two groups of Treg in humans: the naturally to retinal hypoxia. Compensatory mechanisms like vasodilata-
occuring Treg and the induced Treg. The natural Treg is devel- tion are a regulatory attempt to increase retinal blood flow.
opmentally determined in the thymus as a distinct T-cell sub- After exhaustion of autoregulation, a disruption of endothelial
population that is specialized for suppressive function.17 It is tight junctions in the retinal vasculature can occur.4 Further-
distinguished from the induced Treg by a demethylated pro- more, chronic hyperglycemia seems to be able to induce a
moter region for Foxp3.22 Foxp3 seems to act as a repressor of hypoxia-mediated expression of growth factors and cytokines
transcription that regulates T-cell activation. A dysfunction of in the retinal endothelium, such as insulin-like growth
factor-1, vas­cular endothelial growth factor-A, angiopoietin-2 Table 27.1 Components contributing to ocular immune privilege
and TNF-α, leading to leukocyte adhesion, endothelial cell
582 injury, increased permeability of endothelial cells, and finally Passive
angiogenesis.31–33 The resulting plasma leakage from the inner
Blood–retinal barrier Nonfenestrated vascular endothelium of
blood–retinal barrier may cause diabetic macular edema and the retinal vessels and tight junctions
consecutive vision loss.
Section 2

among the retinal pigment epithelium

Blood–retinal barrier in cystoid Lack of lymphatic Absence of lymphatics in the retina – it


macular edema drainage is present in the choroid

Cystoid macular edema is a major reason for vision loss in Tissue fluid Tissue fluid drains via the
various ocular diseases. Different conditions such as trauma, drainage hematogenous route
Basic Mechanisms of Injury in the Retina

inflammation, or vascular degeneration lead to an increased


Basic Science and Translation to Therapy

Reduced Antigen presentation is reduced by


permeability of the blood–retinal barrier. The inner and outer expression of MHC reduction of MHC class I and II
ple­xiform layers of the retina act as diffusion barriers against molecules and APCs. The APCs are
continuous fluid distribution. Therefore, a breakdown of the Reduced APCs in altered by the microenvironment to
the retina promote immune regulation
inner blood–retinal barrier causes cysts mainly in the inner
nuclear layer, whereas serum leakage from the outer blood–
APCs with altered
retinal barrier pools into cystic spaces in the Henle fiber function
layer.34
Active
THE EYE AS AN IMMUNE-PRIVILEGED SITE Immunosuppressive TGF-β, α-MSH, VIP, CGRP, MIF, IL-1
The eye has been recognized as an immune-privileged site for receptor antagonist and free cortisol
microenvironment
more than 100 years. In the 1940s Medawar demonstrated this
by showing prolonged, often indefinite, survival of organs or Factors expressed CD95 ligand, CD59, CD55, CD46
tissue grafts in the anterior chamber of the eye.35 It is now on cell surface
known that this immune privilege is a dynamic process in which MHC, major histocompatibility complex; APC, antigen-presenting cell; TGF,
immunoregulatory mechanisms combined with anatomical transforming growth factor; MSH, melanocyte-stimulating hormone; VIP,
vasoactive intestinal peptide; CGRP, calcitonin gene-related peptide; MIF,
factors maintain the vitality of grafts in privileged sites and of migration inhibitory factor; IL, interleukin; CD, nomenclature for surface proteins
privileged organs and tissues as grafts. Various tissues, includ- of human leukocytes.
ing allogenic skin grafts, thyroid tissues, neuronal retinal tissue,
and allogenic tumor cells, have been shown to survive in the
anterior chamber of the eye for prolonged periods.36 However, Downregulatory immune environment
this privilege is influenced by the immunogenic strength of the The DIE is an evolutionary adaptation of the eye to protect
antigens expressed by the cells. Tumors expressing MHC- itself against excessive inflammation. A small focus of inflam-
encoded alloantigens had only a transient extension of survival mation in the eye has a far greater impact than a similar one
compared to those that expressed weaker transplantation anti- in the kidney. The posterior segment seems to have several
gens.37 Although most studies were done placing tissue in the active factors that help to keep the local immunoenvironmental
anterior chamber of the eye, a number of studies demonstrated balance in a downregulatory state. Active factors include the
a similar immune privilege when allogenic tumor cells, foreign downregulatory effect of Müller cells in cell-to-cell contact with
neuronal retinal tissue, and RPE were placed in the vitreous lymphocytes.44 Furthermore, there are many other ocular cells
cavity and the subretinal space.38,39 Certain eye tissues are that affect DIE (NK T cells, RPE , microglia, F4/80+ macro-
known to be immune-privileged, i.e., demonstrating an altered phages), as well as cell surface and soluble factors in the ocular
immune response and less rejection. Ocular tissues include the microenvironment (e.g., CD95 ligand, CD55, CD59, CD46, IL-10,
cornea, RPE, and probably the retina. When transplanted IL-11, TGF-β, α-melanocyte-stimulating hormone, vasoactive
beneath the capsule of the kidney, a nonimmune-privileged site, intestinal peptide, calcitonin gene-related peptide, macrophage
cornea demonstrated extended survival compared with other migration-inhibitory factor, IL-1-receptor antagonist, and free
ocular tissues like the conjunctiva.40 Both passive factors, such cortisol). All of these factors contribute to active immunoregula-
as the blood–retinal barrier, and active factors, such as the DIE, tion in an attempt to prevent any autoimmunity.2
are essential to establish and maintain the ocular immune privi-
lege (Table 27.1). Downregulatory immune environment in
age-related macular degeneration
Transportation of antigens Several findings in patients with AMD (e.g., variants of CFH,
The blood–retinal barrier serves to keep tissue-specific antigens HTRA-1, PLEKHA1 and Toll-like receptors, activated macro-
sequestrated. There is an absence of lymphatics in the retina, phages in the blood, autoantibodies to retinal components) allow
although there is evidence that antigen may still be able to be it to be considered as an immune-mediated disease.5 Addition-
transported to the lymph nodes.41 Tissue drainage via the hema- ally, there is a masked randomized pilot study indicating that
togenous route may alter the APC function.42 There is also a anti-inflammatory therapy was beneficial in treating wet AMD.45
reduced expression of MHC class I and II molecules and an Not all these associations may be directly related to AMD, but
absence of bone marrow-derived cells that function within rather to the underlying DIE, weakening it and bringing it out
tissues as APCs in the normal retina.43 of balance.46
Anterior-chamber-associated revealed that ACAID is dependent on the presence of Fas
ligands, as these mice are unable to induce ACAID.53 Hence,
immune deviation
Fas–Fas ligand interaction may be more complex than just induc- 583
Anterior-chamber-associated immune deviation (ACAID) is the ing apoptosis of activated immune cells, as initially presumed.
best-studied immune-privilege phenomenon in the eye.47 Although there are active soluble factors in the aqueous
Although its name implies that it is an anterior-chamber phe-

Chapter 27
humor that can inhibit T-cell activation in vitro, decrease the
nomenon, there is much information to show that the same ability of NK cells to lyse their target,54 and block comple-
mechanisms are at work in the vitreous and subretinal space.39 ment activation,55,56 the eye is not left defenseless against
Therefore understanding ACAID could further our knowledge pathogens. It still retains certain immune functions to deter
of retinal autoimmunity. There are a large number of bone pathogenic invasion. Antibodies in the aqueous are capable
marrow-derived APCs in the iris, trabecular meshwork, and of neutralizing viruses and cytotoxic T cells (terminally dif-
ciliary body. Studies have demonstrated that antigen captured ferentiated) can bind and kill their target cells as elsewhere

Blood–Retinal Barrier, Immune Privilege, and Autoimmunity


in the anterior chamber by these APCs is processed in the in the body.57
spleen.48 These APCs reach the spleen via the hematogenous
route, bypassing the afferent lymphatics and the lymph nodes.
The result is a reduction of the antigen’s capacity to activate
Retinal antigens and experimental
cell-mediated immune mechanisms. Functionally, these APCs autoimmune uveoretinitis
are different. If transferred, these APCs induce ACAID rather Several animal models for autoimmune posterior uveitis have
than inducing conventional immune responses. The microenvi- been described (Table 27.2). EAU is one of the most studied of
ronment of the eye also contains factors such as TGF-β2 that are these models since its development in 1968 by Wacker and
able to reprogram the APCs from conventional sites, if exposed Lipton.58 (It is a good animal model for human ocular autoim-
to the eye microenvironment, to secrete less IL-2, express less munity.) EAU is mostly self-limited and requires the use of
CD40 (a costimulatory signal for T-cell activation), and produce adjuvant for disease induction. The EAU model has been instru-
more mature TGF-β. These TGF-β2-treated APCs activate naive mental in eliciting immune mechanisms, the identification of
T cell to secrete IL-4 and trace amounts of IL-2, but not IFN-γ.49 pathogenic epitopes of autoantigens in the eye in animals, and
The spleen is the critical lymphoid organ for the induction of the evaluation of therapeutic strategies with clinical relevance.
ACAID. In the spleen, a unique population of CD4+ lympho- Several retinal antigens, including S-antigen (arrestin), inter-
cytes, which secrete IL-2 and TGF-β and develop a Th2 immune photoreceptor retinoid-binding protein (IRBP), rhodopsin,
response upon antigen exposure, is activated. The B-cell recoverin, and phosducin, have been found to have uveitogenic
response, occurring upon re-exposure to antigen with adjuvant, properties. Immunization with these antigens or their fragments
produces IgG1 isotype antibodies instead of IgG2a, IgG2b, and can induce ocular inflammation in susceptible strains of guinea
IgG3, consistent with a Th2 response.50 There appears to be a pigs, rats, mice, and in rabbits and monkeys. The disease
concerted effort to deviate the immune response from the usual that is induced resembles various human uveitic conditions
Th1 pathway to a Th2 pathway, one that is seen in allergic such as ocular sarcoidosis (Fig. 27.1), Vogt–Koyanagi–Harada
responses. The role of CD95 and its ligand, Fas ligand, is still disease, sympathetic ophthalmia, and Behçet disease (Fig. 27.2).
unclear. It is tempting to speculate that Fas ligand acts by Using advanced techniques, more novel eye autoantigens have
causing active immune cells to apoptose upon engagement, been identified, such as uveal autoantigen with coiled coil
thereby resulting in the elimination of the active immune cells.51 domains and ankyrin repeats in patients with Vogt–Koyanagi–
One would expect an upregulation of an inflammatory response Harada disease,59 and an unknown retinal autoantigen targeting
if Fas ligand were absent in the eye. Contrary to the predictions, the connecting cilium region of photoreceptors in a patient with
Fas ligand-deficient mice developed less severe inflammation Waldenström’s macroglobulinemia, and retinal dysfunction
during experimental autoimmune uveoretinitis (EAU) than wild similar to cancer-associated retinopathy.60 Much of our under-
types.52 Studies using Fas ligand-deficient mice have also standing of retinal autoantigens is derived from EAU models

Table 27.2 Animal models for autoimmune posterior uveitis

Model Antigen Target Clinical disease

Experimental autoimmune S-antigen Photoreceptor layer Posterior uveitis and retinal vasculitis-like sympathetic
uveoretinitis (EAU) IRBP of the retina ophthalmia, VKH, and Behçet disease
Rhodopsin
Recoverin
Phosduscin

Experimental melanin Melanin proteins


protein-induced uveitis (EMIU) Tyrosinase-related Choroid Chronic panuveitis and posterior uveitis like SO and
proteins 1 and 2 VKH

Experimental autoimmune Retinal pigment Retinal pigment Posterior uveitis


pigment epithelial uveitis epithelium membrane epithelial cells
(EAPU) protein-induced
VKH, Vogt–Koyanagi–Harada; IRBP, interphotoreceptor retinoid-binding protein; SO, sympathetic ophthalmia.
584
Section 2
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

A B C

Fig. 27.1 Photomicrograph of (A) normal retina in monkey; (B) retinal vasculitis (arrow) in experimental autoimmune uveoretinitis in monkey;
(C) retinal vasculitis (arrow) in patient with sarcoidosis. (Courtesy of Dr Chi-Chao Chan, Laboratory of Immunology, National Eye Institute.)

A B

Fig. 27.2 (A) Color fundus photograph of experimental autoimmune uveoretinitis in nonhuman primate. (Courtesy of Dr Chi-Chao Chan,
Laboratory of Immunology, National Eye Institute.) (B) Color fundus photograph of retinal vasculitis in patient with Behçet disease. (Courtesy of
Associate Professor Soon-Phaik Chee, Singapore National Eye Center.)

using S-antigen, IRBP (peptides derived from these proteins), identified as targets of the T-cell epitopes. One major uveitogenic
and, to a lesser extent, rhodopsin, recoverin, and phosducin. peptide is M (sequence 303–320). Some of the fragments are
species-specific in their uveitogenicity.64 Although the demon-
S-antigen stration of uveitogenic properties of S-antigen has helped in the
S-antigen, a 48-kDa protein also referred to as arrestin, is one understanding of uveitis mechanisms, the relevance to human
of the antigens most commonly used to induce EAU.61 It was uveitis needs to be more fully explored. S-antigen has also been
first identified in the soluble fraction of retinal extracts. As it is implicated in the pathogenesis of uveitis through molecular
the first autoretinal antigen to be implicated in the pathogenesis mimicry. It has been shown that several exogenous (baker’s
of uveitis, the sequence and its role in phototransduction have yeast, Escherichia coli, hepatitis B virus, streptococcal M5 protein,
been well characterized.62,63 It is a highly conserved protein Moloney murine sarcoma virus, and baboon endogenous virus)
found in the retinal photoreceptor cells and in the pinealocyte. and endogenous (human leukocyte antigen (HLA) B-derived
The main function of arrestin is to block the interaction of rho- peptide, tropomyosin) antigens share sequence homology with
dopsin with the G-protein transducin in the phototransduction peptide M of S-antigen.65 Some have been shown to be uveito-
cascade. Immunization of susceptible animals (such as Lewis genic in the EAU models, and lymphocytes from animals immu-
rats but not mice) with S-antigen induces a predominantly CD4+ nized with M peptide cross-reacted and proliferated when
T-cell-mediated inflammatory response in the retina, uveal tract, stimulated with peptides derived from some of these exogenous
and pineal gland. Six peptide fragments of S-antigen have been or endogenous antigens. Antistreptococcal monoclonal antibod-
identified as uveitogenic. There are six regions of S-antigen ies were found to recognize several uveitogenic peptides of
S-antigen, providing further evidence that immunological retinopathy.77 High levels of antirecoverin antibodies were
mimicry between self and exogenous antigens from an infectious detected in some patients with occult cancer and retinal dysfunc-
agent may be a potential mechanism in the pathogenesis of tion. More recently, these autoantibodies have been demon- 585
uveitis in humans.66 strated to induce apoptosis of photoreceptors, supporting its
pathogenic role in retinal autoimmunity.78
Interphotoreceptor retinoid-binding protein

Chapter 27
IRBP can be used to induce EAU in both mouse and rat.67 It is a Phosducin
major protein (1264 amino acid residues) of the interphotorecep- Phosducin is a 33-kDa protein, a cytosolic regulator of G-protein-
tor matrix, functioning as a transporter of retinoids between the mediated signaling, found in the retina and also in nonretinal
retina and RPE. Similar to S-antigen, it is also found in both the tissues such as liver, lung, heart, and brain.79 Immunization with
eye and the pineal gland, and induction of EAU with IRBP will phosducin in Lewis rats produces mild to moderate EAU char-
lead to disease in both locations. Depending on the dose of acterized by late onset, low-grade severity, and predominantly

Blood–Retinal Barrier, Immune Privilege, and Autoimmunity


antigen used and the species of animals, a spectrum of disease involving the posterior segment.80 Its role in human disease is
ranging from hyperacute to chronic relapsing disease could be unclear.
induced. The inflammation is targeted at the photoreceptor
layer, producing histopathology similar to human lesions seen RETINAL AUTOIMMUNITY
in uveitis, such as retinal vasculitis, granuloma, focal serous
detachments, loss of photoreceptors, and formation of sub-RPE Autoimmunity in human uveitis
infiltrates similar to Dalen–Fuchs nodules, seen in patients with More than four decades of work on EAU models using these
Vogt–Koyanagi–Harada disease and sympathetic ophthalmia.68 retinal autoantigens have passed. However, it is still unclear
The disease activity shows two peaks at the 5th and 10th weeks whether there is an association between the occurrence of eye-
after immunization, simulating the relapsing course in human specific autoantibodies and lymphocyte proliferation response
uveitis.69 The relatively long duration of disease activity in the and clinical disease, and exactly what their roles are in the patho-
murine IRBP EAU model makes it a good model for evaluation genesis of human uveitis and other potentially immune-mediated
of therapeutic strategies in established disease. diseases like AMD. Lymphocytes reactive to retinal antigens
Rhodopsin have been demonstrated in normal individuals4 and those with
Immunization with the particulate fraction of the retinal extract, retinal disease.81 In normal individuals, the presence of circulat-
fraction P, was shown to induce EAU. Later, rhodopsin was ing autoreactive cells suggests that immunoregulatory mecha-
identified to be the uveitogenic antigen in fraction P. nisms must exist to prevent retinal autoimmunity resulting in
Rhodopsin is the visual pigment of rod photoreceptor cells, disease; in the latter it is tempting to hypothesize the possible
belonging to the larger family of G-protein-coupled receptors.70 roles of these cells in the induction of retinal autoimmunity. Both
It is a membrane-bound protein comprising a polypeptide chain, the humoral and cellular responses against S-antigen and IRBP
the opsin, and a covalently bound chromophore, 11 cis-retinal. have been investigated. T-lymphocyte responses to S-antigen,
The function of rhodopsin is to capture photons and trigger the IRBP, and peptides derived from patients with uveitis were
phototransduction cascade. The uveitis induced in susceptible investigated in a few clinical studies. An increase in T-lymphocyte
animals such as guinea pigs, rabbits, rats, and monkeys is typi- responses was reported in patients with Behçet disease during
cally a retinitis with multifocal destruction of photoreceptor periods of active ocular inflammation.82 In another similar study,
cells, showing dense mononuclear and polymorphonuclear cell a large proportion of the patients had lymphocytes which
infiltrates in the retina and anterior uvea in severe disease.71 responded to stimulation with S-antigen, IRBP, and several
There are three distinct immunopathogenic sites on the mole- uveitogenic peptides such as S-antigen peptide M, but no differ-
cule: sequence 230–250 (extracellular loop V-VI) is the most ence was reported in the cellular response between Behçet
uveitogenic, comparable to S-antigen and IRBP.72,73 The role of disease patients with and without uveitis.83 Similar cellular
rhodopsin autoantigens in human diseases is still unclear. responses to S-antigen, S-antigen-derived peptides (peptides M
Studies in patients with normal-pressure glaucoma have dem- and G), and uveitogenic peptide derived from IRBP (R16) have
onstrated an elevated antirhodopsin antibody titer, indicating been reported for patients with idiopathic uveitis.84
the possibility of its role in the optic neuropathy observed in Lymphocyte proliferation responses to 20 peptides spanning
these patients.74 the entire sequence of S-antigen were investigated in patients
with uveitis in one study. Half of the patients with idiopathic
Recoverin uveitis responded to at least one peptide whereas there was no
Recoverin is a 23-kDa calcium-binding protein present in response to the peptides in the normal controls. The peptide
photoreceptor cells and functioning as a calcium sensor that fragments found to elicit more lymphocyte proliferation were
regulates rhodopsin phosphorylation through inhibition of rho- 61–80, 81–100, 145–160, 161–180, 221–240, and 241–260.85 Other
dopsin kinase. It is also found in bipolar cells, pinealocytes, and fragments of IRBP and S-antigen were studied in another similar
certain tumor cells. The function in these nonphotoreceptor cells study involving 82 patients with uveitis. Patients who responded
is unknown as they do not contain rhodopsin kinase.75 Immuni- to IRBP or S-antigen also responded to one or two of the frag-
zation of Lewis rats with high doses of recoverin produced EAU ments but to lesser magnitude. This suggests that these frag-
similar in severity, histology, and duration to other retinal auto- ments are probably not the primary mediators of the disease.86
antigens such as S-antigen.76 The major immunopathogenic Isolation of S-antigen-specific T cells from the eyes of uveitis
epitope, sequence 62–81, has been characterized in Lewis rats. It patients, which were expanded to antigen-specific T-cell lines,
has been identified as an autoantigen in paraneoplastic and was reported.87 A study evaluating oral tolerance by feeding
degenerative diseases of the retina such as cancer-associated S-antigen to uveitis patients showed a good clinical response and
CNS leads not only to the degeneration of the affected axon but
also the neighboring axon through self-destructive compounds
586 released by the degenerating axon into the microenvironment.94
Studies have revealed that autoreactive T cells directed against
myelin antigens have beneficial effects on CNS myelinated axons
after a mechanical crush injury95 and it has been proposed that
Section 2

this neuroprotective activity exhibited by autoimmune T cells


may be a physiological process which the body develops to cope
with stressful conditions.96 The observation seen in the CNS
could be extrapolated to the retina. Supporting this are the find-
ings of a study demonstrating that vaccination with peptides
derived from IRBP resulted in protection of retinal ganglion cells
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

from glutamate-induced death or death as a consequence of


optic nerve injury.7 It appears that the immune system not only
protects the body against invading pathogens but also protects
Fig. 27.3 Photomicrograph showing antiretinal antibodies in the serum it from toxic substances released by self tissues during trauma
of a patient with Vogt–Koyanagi–Harada disease reacting and stress. Therefore the autoreactive cells that induce neuropro-
immunohistochemically to the photoreceptor layer of the monkey retina tection and those that induce autoimmune disease may share the
(arrows). (Courtesy of Dr Chi-Chao Chan, Laboratory of Immunology, same specificity and phenotype, indicating their potential to be
National Eye Institute.)
protective and destructive at the same time.97 The final outcome
depends on how they are regulated. Correlation between the
ability to manifest an autoimmune response with a beneficial
the patients required less immunosuppression.88 Together, these outcome and the ability to resist the development of an autoim-
studies provided further support for the possible pathogenic mune disease in animals has been reported.98 From these find-
roles of retinal autoantigen in uveitis. ings, it seems clear that the ability to protect the eye from
Although both, EAU in animals and uveitis in most humans inflammation and injury does not solely depend on mechanisms
can be a CD4+ Th1 or a Th17 effector response driving autoim- conferring immune privilege but rather a precise regulation of
munity, there are differences in the T-cell specificities in antigen autoimmunity.
response.89 Patients with uveitis, due to epitope spread, can
respond to more than one peptide, i.e., several epitopes are rec- Role of retinal autoimmunity in infection
ognized in the same patient, while animals with EAU tend to The role of infection in the pathogenesis of eye autoimmunity
respond only to the inciting antigen. There is no common pattern has long been suspected but there is no conclusive study to
of lymphocyte response in clinical studies reported so far. Early demonstrate it. The possible causative relationship between
clinical studies on the humoral response showed the presence of infections and ocular autoimmunity such as streptococcus, her-
antiretinal autoantibodies, including anti-S-antigen antibodies in pesvirus, and Behçet disease,99 Gram-negative bacteria, HLA
the sera of some uveitis patients90,91 (Fig. 27.3), but there was no B27-associated uveitis,100 herpesviruses, and serpiginous choroi-
good correlation between presence or levels of autoantibodies ditis101 has been reported. Infection as an exogenous trigger for
and clinical disease. Moreover, anti-IRBP antibody was reported inflammation in the eye is an enticing possibility as infections
to occur in the same frequency in the sera of patients with uveitis have been linked to the pathogenesis of many autoimmune dis-
and normal controls. The only difference has been the stronger eases such as rheumatic fever,102 inflammatory bowel diseases,103
affinity of the antibodies in patients with uveitis.92 and sarcoidosis.104 Antiretinal antibodies in the serum of patients
The relationship between humoral and cellular response to with toxoplasmosis retinochoroiditis,105 antiretinal and anti-RPE
these autoantigens was studied in a group of patients with antibodies in murine coronavirus retinopathy and degenera-
endogenous uveitis such as birdshot retinochoroidopathy, tion106 were reported. These reports provided support for a role
retinal vasculitis, and Behçet disease. The antibody titers to of infection in retinal autoimmunity. Even in the current EAU
S-antigen and IRBP were found to be decreased during active models in animals, bacteria-derived products like mycobacteria
inflammation periods and the lymphocyte responses to S-antigen in complete Freund’s adjuvant, lipopolysaccharides, and pertus-
were found to be most significant in the period preceding a sis toxin are used to promote the development of autoimmune
relapse of ocular inflammation, supporting the pathogenic roles disease.107 An infection could possibly play a similar role, in
of these lymphocytes in retinal autoimmunity.93 The role of sensitizing the immune system to eye-restricted antigens, which
humoral and cellular responses to retinal autoantigens remains are “sequestrated” or expressed in low levels below the immune
unclear and it is possible that different subsets of patients with activation threshold. The mechanisms involved in the pathogen-
uveitis, genetically determined, respond to different epitopes esis of uveitis may be similar to that of the adjuvant in EAU,
and antigens, resulting in the wide spectrum of clinical disease including changes in the blood–tissue barrier to allow infiltra-
encountered. tion of inflammatory cells,108 promotion of Th1 response,109 stim-
ulatory effects on the APCs,110 and enhancement of innate
Role of retinal autoimmunity in protection immune response.111 On the other hand, there is evidence that
Retinal autoimmunity may not always be pathogenic. Studies on immunologic mimicry may play a role in pathogenesis as molec-
central nervous system (CNS) injury and its reparative process ular homology exists between certain retinal autoantigens and
have revealed a possible beneficial role of autoimmunity in limit- peptides derived from microbes, such as the M peptide derived
ing self-injury during insults to the CNS. Axonal injury in the from S-antigen and the streptococcal cell wall peptide, the yeast’s
histone peptide, and certain viral agents.66,112,113 Molecular 5. Nussenblatt RB, Liu B, Li Z. Age-related macular degeneration: an immuno-
logically driven disease. Curr Opin Invest Drugs 2009;10:434–42.
mimicry has also been suspected of playing a role in conferring 6. Yamamoto JH, Minami M, Inaba G, et al. Cellular autoimmunity to retinal
disease susceptibility in patients with certain HLA haplotypes, specific antigens in patients with Behçet’s disease. Br J Ophthalmol 1993;77: 587
584–9.
like HLA B27 for acute anterior uveitis,114 HLA B51 for Behçet’s 7. Mizrahi T, Hauben E, Schwartz M. The tissue-specific self-pathogen is the
disease,115 and HLA-A29 for birdshot retinochoroidopathy.116,117 protective self-antigen: the case of uveitis. J Immunol 2002;169:5971–7.

Chapter 27
Synthetic peptides derived from HLA-B molecules such as B27 8. Davis MM. T cell receptor gene diversity and selection. Annu Rev Biochem
1990;59:475–96.
and B51 with amino acid sequence homologous with S-antigen 9. Romagnani S, Maggi E, Liotta F, et al. Properties and origin of human Th17
have been reported to induce EAU in animals.118 Lymphocytes cells. Mol Immunol 2009;47:3–7.
10. Amadi-Obi A, Yu CR, Liu X, et al. TH17 cells contribute to uveitis and scleritis
from patients with uveitis, especially Behçet’s disease, respond and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 2007;
to both peptides derived from HLA-B molecules and peptides 13:711–8.
11. Croft M, Dubey C. Accessory molecule and co-stimulation requirements for
derived from S-antigen, demonstrating possible immunologic CD4 T cell response. Crit Rev Immunol 1997;17:89–118.
mimicry.119 In addition, further studies also showed sequence 12. Schatz DG, Oettinger MA, Schlissel MS. V(D)J recombination. Annu Rev

Blood–Retinal Barrier, Immune Privilege, and Autoimmunity


Immunol 1992;10:359–83.
homology between these peptides, rotavirus, and casein protein, 13. Sprent J. Central tolerance of T cells. Int Rev Immunol 1995;13:95–105.
a protein found in milk, and cross-reaction in cellular immunity 14. Arnold B, Schönrich G, Hämmerling GJ. Multiple levels of peripheral
between these peptides was found in animal studies.120 Together tolerance. Immunol Today 1993;14:12–4.
15. Moudgil KD, Ametani A, Grewal IS, et al. Processing of self-proteins and
these studies bring forward the possibility of immunologic its impact on shaping the T cell repertoire, autoimmunity and immune
mimicry for exogenous factors like infection or foreign proteins regulation. Int Rev Immunol 1993;10:365–77.
16. Sinclair NR, Anderson CC. Co-stimulation and co-inhibition: equal partners
breaking down tolerance and inducing autoimmunity. in regulation. Scand J Immunol 1996;43:597–603.
The guardian that prevents pathologic autoimmunity is 17. Sakaguchi S, Miyara M, Costantino CM, et al. FOXP3+ regulatory T cells in
the human immune system. Nat Rev Immunol 2010;10:490–500.
immune tolerance. Immune tolerance, usually maintained by 18. Van Parijs L, Abbas AK. Role of Fas-mediated cell death in the regulation of
mechanisms described earlier, could be lost or modulated. This immune responses. Curr Opin Immunol 1996;8:355–61.
was demonstrated in a study showing that heat shock protein 19. Griffith TS, Ferguson TA. The role of FasL-induced apoptosis in immune
privilege. Immunol Today 1997;18:240–4.
(HSP)91 can modulate APC function, resulting in loss of tolerance 20. De Vries JE. Immunosuppressive and antiinflammatory properties of
in an animal model.121 HSPs, especially microbial, have been interleukin 10. Ann Med 1995;27:537–41.
21. Sosroseno W, Herminajeng E. The immunoregulatory roles of transforming
implicated in the pathogenesis of Behçet’s disease122 and demon- growth factor beta. Br J Biomed Sci 1995;52:142–8.
strated to induce inflammation in the eye in animal studies.123 22. Baron U, Floess S, Wieczorek G, et al. DNA demethylation in the human
FOXP3 locus discriminates regulatory T cells from activated FOXP3(+)
Therefore, one may speculate that an infection and the subse- conventional T cells. Eur J Immunol 2007;37:2378–89.
quent release of microbial HSPs could be the initial triggering 23. Tommasini A, Ferrari S, Moratto D, et al. X-chromosome inactivation analy-
sis in a female carrier of FOXP3 mutation. Clin Exp Immunol 2002;130:
event, resulting in changes of APC function and a subsequent 127–30.
loss of tolerance, leading to autoimmunity. These notions remain 24. Sakaguchi S, Sakaguchi N, Asano M, et al. Immunologic self-tolerance main-
as hypotheses for uveitis since it is a heterogeneous disease, and tained by activated T cells expressing IL-2 receptor alpha-chains (CD25).
Breakdown of a single mechanism of self-tolerance causes various autoim-
evidence of the initial triggering event is usually difficult to mune diseases. J Immunol 1995;155:1151–64.
identify due to the protracted course and the delay between 25. Ehrlich P. Das Sauerstoffbedürfnis des Organismus, eine farbenanalytische
Studie. Berlin: Hirschwald; 1885.
trigger and disease presentation. Furthermore, a disease could 26. Goldmann EE. Vitalfärbung am Zentralnervensystem. Abh Preuss Akd Wiss
have multiple triggering mechanisms and different triggers Phys – Math Kl / 1 1913;1–3.
27. Schnaudigel O. Die Vitalfarbung mit Trypanblau am Auge. Graefes Arch
may lead to the same disease, making clinical data difficult to Ophthalmol 1913;86:93–7.
interpret. 28. Cunha-Vaz JG, Skakib M, Ashton N. Studies on the permeability of the blood–
retinal barrier. I. On the existence, development, and site of a blood–retinal
barrier. Br J Ophthalmol 1966;50:441–53.
CONCLUSION 29. Runkle EA, Antonetti DA. The blood–retinal barrier: structure and functional
significance. Methods Mol Biol 2011;686:133–48.
30. Toda R, Kawazu K, Oyabu M, et al. Comparison of drug permeabilities across
Retinal autoimmunity may be a physiologic process rather than the blood–retinal barrier, blood–aqueous humor barrier, and blood–brain
a pathologic one, given its role in organ- and tissue-specific barrier. J Pharm Sci 2011;100:3904–11.
31. Haurigot V, Villacampa P, Ribera A, et al. Increased intraocular insulin-like
protection from damage from injury and stress in the normal growth factor-I triggers blood–retinal barrier breakdown. J Biol Chem
retina. That may account for the evidence of retinal autoimmu- 2009;284:22961–9.
nity detected in normal subjects, but there is still much that is 32. Rangasamy S, Srinivasan R, Maestas J, et al. A potential role for angiopoietin
2 in the regulation of the blood–retinal barrier in diabetic retinopathy. Invest
unknown about its regulation and the maintenance of the fine Ophthalmol Vis Sci 2011;52(6):3784–91.
balance between protection and destruction given that these 33. Joussen AM, Poulaki V, Mitsiades N, et al. Nonsteroidal anti-inflammatory
drugs prevent early diabetic retinopathy via TNF-alpha suppression. FASEB
paradoxical roles reside in the same group of cells. With rapid J 2002;16(3):438–40.
technological advances in molecular immunology, new tools to 34. Bringman A, Reichenbach A, Wiedemann P. Pathomechanisms of cystoid
macular edema. Ophthalmic Res 2004;36:241–9.
unravel the roles of retina autoimmunity in both the physiologi- 35. Medawar P. Immunity to homologous grafted skin. III. The date of skin
cal and pathological states of the eye will be available and novel homografts transplanted to the brain, to subcutaneous tissue and to the
anterior chamber of the eye. Br J Exp Pathol 1948;29:58–69.
targeted therapeutic strategies could be developed for the man- 36. Niederkorn J, Streilein JW, Shadduck JA. Deviant immune responses to
agement of retinal autoimmunity-induced diseases in the near allogeneic tumors injected intracamerally and subcutaneously in mice.
future. Invest Ophthalmol Vis Sci 1980;20:355–63.
37. Niederkorn JY, Streilein JW. Immunogenetics basis for immunologic privilege
in the anterior chamber of the eye. Immunogenetics 1981;13:27–236.
REFERENCES 38. Wenkel H, Streilein JW. Analysis of immune deviation elicited by antigens
1. Van Dooremaal JC. Die Entwicklung der in fremden Grund versetzten leben- injected into the sub-retinal space. Invest Ophthalmol Vis Sci 1998;39:
den Gewebe. Graefes Arch Ophthalmol 1873;19:359–73. 1823–34.
2. Taylor AW. Ocular immunosuppressive microenvironment. Chem Immunol 39. Jiang LQ, Jorquera M, Streilein JW. Subretinal space and vitreous cavity as
1999;73:72–89. immunologically privileged sites for retinal allografts. Invest Ophthalmol Vis
3. Cunha-Vaz J, Faria de Abreu JR, Campos AJ. Early breakdown of the Sci 1993;34:3347–54.
blood–retinal barrier in diabetes. Br J Opthalmol 1975;59:649–56. 40. Streilein JW. Immune privilege and the cornea. In: Pleyes U, Hartmann C,
4. Wallow IH, Engerman RL. Permeability and patency of retinal blood vessels Sterry W, editors. Proceedings of symposium on bullous oculo-muco-
in experimental diabetes. Invest Ophthalmol Vis Sci 1977;16:447–61. cutaneous disorders. Buren: Aeolus Press; 1997. p. 43–52.
41. Egan RM, Yorkey C, Black R, et al. Peptide-specific T cell clonal expansion in 71. Adamus G, Arendt A, Hargrave PA. Genetic control of antibody response
vivo following immunization in the eye, an immune-privileged site. to bovine rhodopsin in mice: epitope mapping of rhodopsin structure.
J Immunol 1996;157:2262–71. J Neuroimmunol 1991;34:89–97.
588 42. McMenamin PG, Forrester JV. Dendritic cells in the central nervous system 72. Adamus G, Schmied JL, Hargrave PA, et al. Induction of experimental auto-
and eye and their associated supporting tissues. In: Lotze MY, Thomson AW, immune uveitis with rhodopsin synthetic peptides in Lewis rats. Curr Eye
editors. Dendritic cells: biology and clinical applications. San Diego: Res 1992;11:657–67.
Academic Press; 1999. p. 205–54. 73. Adamus G, Ortega H, Witkowska D, et al. Recoverin: a potent uveitogen for
43. Zhang J, Wu GS, Ishimoto S, et al. Expression of major histocompatibility the induction of photoreceptor degeneration in Lewis rats. Exp Eye Res
Section 2

complex molecules in rodent retina: immunohistochemical study. Invest 1994;59:447–55.


Ophthalmol Vis Sci 1997;38:1848–57. 74. Romano C, Barett DA, Li Z, et al. Antirhodopsin antibodies in sera from
44. Caspi RR, Roberge FG, Nussenblatt RB. Organ-resident, nonlymphoid cells patients with normal-pressure glaucoma. Invest Ophthalmol Vis Sci 1995;36:
suppress proliferation of autoimmune T-helper lymphocytes. Science 1968–75.
1987;237:1029–32. 75. Kawamura S, Cox JA, Nef P. Inhibition of rhodopsin phosphorylation by
45. Nussenblatt RB, Byrnes G, Sen HN, et al. A randomized pilot study of non-myristoylated recombinant recoverin. Biochem Biophys Res Commun
systemic immunosuppression in the treatment of age-related macular 1994;203:121–7.
degeneration with choroidal neovascularization. Retina 2010;30:1579–87. 76. Gery I, Chanaud III NP, Anglade E. Recoverin is highly uveitogenic in Lewis
Basic Mechanisms of Injury in the Retina

46. Nussenblatt RB, Ferris 3rd F. Age-related macular degeneration and the rats. Invest Ophthalmol Vis Sci 1994;35:3342–5.
Basic Science and Translation to Therapy

immune response: implications for therapy. Am J Ophthalmol 2007;144: 77. Adamus G, Guy J, Schmied JL, et al. Role of anti-recoverin autoantibodies in
618–26. cancer-associated retinopathy. Invest Ophthalmol Vis Sci 1993;34:2626–33.
47. Stein-Streilein J, Streilein JW. Anterior chamber associated immune deviation 78. Adamus G. Autoantibody-induced apoptosis as a possible mechanism of
(ACAID): regulation, biological relevance, and implications for therapy. autoimmune retinopathy. Autoimmun Rev 2003;2:63–8.
Int Rev Immunol 2002;21:123–52. 79. Danner S, Lohse MJ. Phosducin is a ubiquitous G-protein regulator. Proc Natl
48. Kaplan HJ, Streilein JW. Do immunologically privileged sites require a func- Acad Sci U S A 1996;93:10145–50.
tioning spleen? Nature 1974;251:553–4. 80. Dua HS, Lee RH, Loeely RN, et al. Induction of experimental autoimmune
49. Takeuchi M, Alard P, Streilein JW. TGF-beta promotes immune deviation by uveitis by the retinal photoreceptor cell protein, phosducin. Curr Eye Res
altering accessory signals of antigen-presenting cells. J Immunol 1998;160: 1992;11(Suppl.):107–11.
1589–97. 81. Nussenblatt RB, Gery I, Ballintine EJ, et al. Cellular immune responsiveness
50. Wilbanks GA, Streilein JW. Distinctive humoral immune responses following of uveitis patients to retinal S-antigen. Am J Ophthalmol 1980;89:173–9.
anterior chamber and intravenous administration of soluble antigen. Evidence 82. de Smet MD, Dayan M. Prospective determination of T-cell responses to
for active suppression of IgG2-secreting B lymphocytes. Immunology 1990; S-antigen in Behçet’s disease patients and controls. Invest Ophthalmol Vis Sci
71:566–72. 2000;41:3480–4.
51. Ferguson TA, Stuart PM, Herndon JM, et al. Apoptosis, tolerance, and regula- 83. Yamamoto JH, Minami M, Inaba G, et al. Cellular autoimmunity to retinal
tory T cells – old wine, new wineskins. Immunol Rev 2003;193:111–23. specific antigens in patients with Behçet’s disease. Br J Ophthalmol 1993;77:
52. Wahlsten JL, Gitchell HL, Chan CC, et al. Fas and Fas ligand expressed on 584–9.
cells of the immune system, not on the target tissue, control induction of 84. Rajasingh J, Singh VK, Singh V, et al. Cellular immune response to retinal
experimental autoimmune uveitis. J Immunol 2000;165:5480–6. S-antigen and interphotoreceptor retinoid binding protein fragments in
53. Kawashima H, Yamagami S, Tsuru T, et al. Anterior chamber inoculation of idiopathic human uveitis. Ind J Med Res 1996;103:222–6.
splenocytes without Fas/Fas-ligand interaction primes for a delayed-type 85. Rai G, Saxena S, Kumar H, et al. Human retinal S-antigen: T cell epitope
hypersensitivity response rather than inducing anterior chamber-associated mapping in posterior uveitis patients. Exp Mol Pathol 2001;70:140–5.
immune deviation. Eur J Immunol 1997;27:2490–4. 86. de Smet MD, Yamamoto JH, Mochizuki M, et al. Cellular immune
54. Apte RS, Niederkorn JY. Isolation and characterization of a unique natural responses of patients with uveitis to retinal antigens and their fragments.
killer cell inhibitory factor present in the anterior chamber of the eye. Am J Ophthalmol 1990;110:135–42.
J Immunol 1996;156:2667–73. 87. Nussenblatt RB, Palestine AG, El-Saied M, et al. Long-term antigen specific
55. Sohn JH, Kaplan HJ, Suk HJ, et al. Chronic low level complement activation and non-specific T-cell lines and clones in uveitis. Curr Eye Res 1984;3:
within the eye is controlled by intraocular complement regulatory proteins. 299–305.
Invest Ophthalmol Vis Sci 2000;41:3492–502. 88. Nussenblatt RB, Gery I, Weiner HL, et al. Treatment of uveitis by oral admin-
56. Sohn JH, Kaplan HJ, Suk HJ, et al. Complement regulatory activity of istration of retinal antigens: results of a phase I/II randomized masked trial.
normal human intraocular fluid is mediated by MCP, DAF, and CD59. Am J Ophthalmol 1997;123:583–92.
Invest Ophthalmol Vis Sci 2000;41:4195–202. 89. Luger D, Silver PB, Tang J, et al. Either a Th17 or a Th1 effector response can
57. Koevary SB, Beaudry K. Effect of anterior chamber-associated immune devia- drive autoimmunity: conditions of disease induction affect dominant effector
tion (ACAID) on rat islet allograft rejection. Ocul Immunol Inflamm 2000;8: category. J Exp Med 2008;205:799–810.
39–47. 90. Chan CC, Palestine AG, Nussenblatt RB, et al. Anti-retinal auto-antibodies
58. Wacker WB, Lipton MM. Experimental allergic uveitis. II. Serologic and in Vogt–Koyanagi–Harada syndrome, Behçet’s disease, and sympathetic
hypersensitive responses of the guinea pig following immunization with ophthalmia. Ophthalmology 1985;92:1025–8.
homologous retina. J Immunol 1968;101:157–65. 91. Doekes G, Luyendijk L, Gerritsen MJ, et al. Anti-retinal S-antigen antibodies
59. Nishimura Y, Yamada K, Senju S, et al. Negi identification of a novel autoan- in human sera: a comparison of reactivity in ELISA with human or bovine
tigen UACA in patients with panuveitis. Biochem Biophys Res Commun S-antigen. Int Ophthalmol 1992;16:147–52.
2001;280:1169–76. 92. Hoekzema R, Hwan SB, Rothova A, et al. Serum antibody response to human
60. Sen NH, Chan CC, Caruso RC, et al. Waldenström’s macroglobulinemia- and bovine IRBP in uveitis. Curr Eye Res 1990;9:1177–83.
associated retinopathy. Ophthalmology 2004;111:535–9. 93. Jobin D, Thillaye B, de Kozak Y, et al. Severe retinochoroidopathy: variations
61. Singh VK, Nussenblatt RB, Donoso LA, et al. Identification of a uveitopatho- of humoral and cellular immunity to S-antigen in a longitudinal study.
genic and lymphocyte proliferation site in bovine S-antigen. Cell Immunol Curr Eye Res 1990;9(Suppl.):91–6.
1988;115:413–9. 94. Liu D, Yang R, Yan X, et al. Hydroxyl radicals generated in vivo kill neurons
62. Hirose S, Singh VK, Donoso LA, et al. An 18-mer peptide derived from the in the rat spinal cord: electrophysiological, histological, and neurochemical
retinal S antigen induces uveitis and pinealitis in primates. Clin Exp Immunol results. J Neurochem 1994;62:37–44.
1989;77:106–11. 95. Hauben E, Nevo U, Yoles E, et al. Autoimmune T cells as potential neuropro-
63. Merryman CF, Donoso LA, Zhang XM, et al. Characterization of a new, tective therapy for spinal cord injury. Lancet 2000;355:286–7.
potent, immunopathogenic epitope in S-antigen that elicits T cells expressing 96. Yoles E, Hauben E, Palgi O, et al. Protective autoimmunity is a physiological
V beta 8 and V alpha 2-like genes. J Immunol 1991;146:75–80. response to CNS trauma. J Neurosci 2001;21:3740–8.
64. Gregerson DS, Fling SP, Obritsch WF, et al. A new perspective of S-antigen 97. Kipnis J, Mizrahi T, Yoles E, et al. Myelin specific Th1 cells are necessary for
from immunochemical analysis. Curr Eye Res 1990;9(Suppl.):145–53. post-traumatic protective autoimmunity. J Neuroimmunol 2002;130:78–85.
65. Shinohara T, Singh VK, Tsuda M, et al. S-antigen: from gene to autoimmune 98. Kipnis J, Yoles E, Schori H, et al. Neuronal survival after CNS insult is deter-
uveitis. Exp Eye Res 1990;50:751–7. mined by a genetically encoded autoimmune response. J Neurosci 2001;21:
66. Lerner MP, Donoso LA, Nordquist RE, et al. Immunological mimicry between 4564–71.
retinal S-antigen and group A streptococcal M proteins. Autoimmunity 99. Zouboulis CC, May T. Pathogenesis of Adamantiades–Behçet’s disease.
1995;22:95–106. Med Microbiol Immunol (Berl) 2003;192:149–55.
67. Broekhuyse RM, Winkens HJ, Kuhlmann ED. Induction of experimental auto- 100. Sahly H, Podschun R, Kekow J, et al. Humoral immune response to Klebsiella
immune uveoretinitis and pinealitis by IRBP. Comparison to uveoretinitis capsular polysaccharides in HLA-B27-positive patients with acute anterior
induced by S-antigen and opsin. Curr Eye Res 1986;5:231–40. uveitis and ankylosing spondylitis. Autoimmunity 1998;28:209–15.
68. Caspi RR, Roberge FG, Chan CC, et al. A new model of autoimmune disease. 101. Priya K, Madhavan HN, Reiser BJ, et al. Association of herpesviruses in the
Experimental autoimmune uveoretinitis induced in mice with two different aqueous humor of patients with serpiginous choroiditis: a polymerase chain
retinal antigens. J Immunol 1988;140:1490–5. reaction-based study. Ocul Immunol Inflamm 2002;10:253–61.
69. Chan CC, Caspi RR, Ni M, et al. Pathology of experimental autoimmune 102. Guilherme L, Kalil J. Rheumatic fever: the T cell response leading to
uveoretinitis in mice. J Autoimmun 1990;3:247–55. autoimmune aggression in the heart. Autoimmun Rev 2002;1:261–6.
70. Hargrave PA, McDowell JH. Rhodopsin and phototransduction. Int Rev Cytol 103. Cumming JH, Macfarlane GT, Macfarlane S. Intestinal bacteria and ulcerative
1992;137B:49–97. colitis. Curr Issues Intest Microbiol 2003;4:9–20.
104. Ishige I, Usui Y, Takemura T, et al. Quantitative PCR of mycobacterial and 113. Singh VK, Kalra HK, Yamaki K, et al. Molecular mimicry between a uveito-
propionibacterial DNA in lymph nodes of Japanese patients with sarcoidosis. pathogenic site of S-antigen and viral peptides: induction of experimental
Lancet 1999;354:120–3. autoimmune uveitis in Lewis rats. J Immunol 1990;144:1282–7.
105. Whittle RM, Wallace GR, Whiston RA, et al. Human antiretinal antibodies in 114. Suhler EB, Martin TM, Rosenbaum JT. HLA-B27-associated uveitis: overview 589
toxoplasma retinochoroiditis. Br J Ophthalmol 1998;82:1017–21. and current perspectives. Curr Opin Ophthalmol 2003;14:378–83.
106. Hooks JJ, Percopo C, Wang Y, et al. Retina and retinal pigment epithelial cell 115. Mizuki N, Inoko H, Mizuki N, et al. Human leukocyte antigen serologic
autoantibodies are produced during murine coronavirus retinopathy. and DNA typing of Behçet’s disease and its primary association with B51.

Chapter 27
J Immunol 1993;151:3381–9. Invest Ophthalmol Vis Sci 1992;33:3332–40.
107. Sasamoto Y, Kawano YI, Bouligny R, et al. Immunomodulation of experimen- 116. LeHoang P, Ozdemir N, Benhamou A, et al. HLA-A29.2 subtype associated
tal autoimmune uveoretinitis by intravenous injection of uveitogenic with birdshot retinochoroidopathy. Am J Ophthalmol 1992;113:33–5.
peptides. Invest Ophthalmol Vis Sci 1992;33:2641–9. 117. Barnett LA, Fujinami RS. Molecular mimicry: a mechanism for autoimmune
108. Linthicum DS, Munoz JJ, Blaskett A. Acute experimental autoimmune injury. FASEB J 1992;6:840–4.
encephalomyelitis in mice. I. Adjuvant action of Bordetella pertussis is due to 118. Wildner G, Thurau SR. Cross-reactivity between an HLA-B27-derived peptide
vasoactive amine sensitization and increased vascular permeability of the and a retinal autoantigen peptide: a clue to major histocompatibility complex
central nervous system. Cell Immunol 1982;73:299–310. association with autoimmune disease. Eur J Immunol 1994;24:2579–85.
109. Silver PB, Chan CC, Wiggert B, et al. The requirement for pertussis to induce 119. Wildner G, Diedrichs-Mohring M, Thurau SR. Induction of arthritis and
EAU is strain-dependent: B10.RIII, but not B10.A mice, develop EAU and Th1 uveitis in Lewis rats by antigenic mimicry of peptides from HLA-B27 and

Blood–Retinal Barrier, Immune Privilege, and Autoimmunity


responses to IRBP without pertussis treatment. Invest Ophthalmol Vis Sci cytokeratin. Eur J Immunol 2002;32:299–306.
1999;40:2898–905. 120. Wildner G, Diedrichs-Mohring M. Autoimmune uveitis induced by molecular
110. Hofstetter HH, Shive CL, Forsthuber TG. Pertussis toxin modulates the mimicry of peptides from rotavirus, bovine casein and retinal S-antigen.
immune response to neuroantigens injected in incomplete Freund’s adjuvant: Eur J Immunol 2003;33:2577–87.
induction of Th1 cells and experimental autoimmune encephalomyelitis in the 121. Millar DG, Garza KM, Odermatt B, et al. Hsp70 promotes antigen-presenting
presence of high frequencies of Th2 cells. J Immunol 2002;169:117–25. cell function and converts T-cell tolerance to autoimmunity in vivo. Nat Med
111. Agarwal RK, Sun SH, Su SB, et al. Pertussis toxin alters the innate and the 2003;9:1469–76.
adaptive immune responses in a pertussis-dependent model of autoimmu- 122. Zierhut M, Mizuki N, Ohno S, et al. Immunology and functional genomics of
nity. J Neuroimmunol 2002;129:133–40. Behçet’s disease. Cell Mol Life Sci 2003;60:1903–22.
112. Lerner MP, Donoso LA, Nordquist RE, et al. Immunological mimicry between 123. Lehner T, Bergmeier LA, Wang Y, et al. Heat shock proteins generate
retinal S-antigen and group A streptococcal M proteins. Autoimmunity beta-chemokines which function as innate adjuvants enhancing adaptive
1995;22:95–106. immunity. Eur J Immunol 2000;30:594–603.
Section 2 Basic Mechanisms of Injury in the Retina
For additional online content visit http://www.expertconsult.com

Chapter Mechanisms of Macular Edema and


28  Therapeutic Approaches
Antonia M. Joussen, Thomas J. Wolfensberger

INTRODUCTION following conditions: diabetes, vascular occlusion, hypertensive


retinopathy, epiretinal membranes, intraocular tumors (e.g.,
Macular edema is a common phenomenon in various diseases melanoma, choroidal hemangioma), intraocular inflammation
where fluid accumulates in between the retinal cells. The fluid (e.g., pars planitis), macroaneurysm, retinitis pigmentosa, cho­
originates from the intravascular compartment. The focal, dif­ roidal neovascularization, and radiation retinopathy.
fuse, and cystic forms are all characterized by extracellular accu­ Given the heterogeneous etiology of macular edema, its effec­
mulation of fluid, specifically in Henle’s layer and the inner tive treatment depends upon a better understanding of its patho­
nuclear layer of the retina. The compartmentalization of the genesis. In general, formation of macular edema is related to
accumulated fluid is likely to be due in part to the relative barrier metabolic changes, ischemia, hydrostatic forces, inflammatory
properties of the inner and outer plexiform layers. and toxic mechanisms, or mechanical forces that occur to various
The breakdown of the blood–retinal barrier (BRB), modulated degrees in different conditions (Table 28.1).
via different growth factors, results from a disturbance of the Metabolic alterations have a causal role in diabetic maculopa­
integrity of the tight junctions.1 Starling’s law predicts that thy, but also in inherited diseases such as the autosomal domi­
macular edema will develop if the hydrostatic pressure gradient nant form of macular edema or macular edema in retinitis
between capillary and retinal tissue is increased. That can occur, pigmentosa. Furthermore, ischemia of the inner or outer BRB
for example, in the presence of elevated blood pressure, or if the leads to formation of a macular edema. Decreased perfusion
osmotic pressure gradient is decreased by excessive protein of the retinal capillaries is seen, e.g., in vein occlusion and
accumulation in the extracellular space within the retina.2 diabetic retinopathy, whereas ischemia plus decreased perfusion
of the choroid with associated serous retinal detachment occurs
MACULAR EDEMA AS A RESULT OF
in severe hypertensive retinopathy, in eclampsia, or in rheu­
VARIOUS DISEASE MECHANISMS matoid disorders. Following retinal vascular occlusion the intra­
Causes of macular edema vascular pressure increases and leads to dysfunction of the BRB.
Similarly, hydrostatic forces are effective in arterial hypertension
The classic pattern of cystoid macular edema (CME) with the
or in eyes with low intraocular pressure and may cause fluid
petaloid appearance originating from the fluorescein leakage
accumulation in the macula.
from perifoveal capillaries may be seen in cases of advanced
Mechanical traction such as in epiretinal membranes or in
edema of various origins (Fig. 28.1, panel B online). This includes
vitreomacular traction syndrome promotes macular edema by
postsurgical CME as well as CME associated with one of the
physical forces.
Inflammation is important in the pathogenesis of macular
edema in conditions such as intermediate uveitis, postoperative
CME (Irvine–Gass syndrome), diabetic macular edema (DME)
and various forms of choroidal inflammatory disease, including
Vogt–Koyanagi–Harada syndrome and birdshot retinocho­
roidopathy. All prostaglandin-like pharmacological agents, even
if applied topically, can induce macular edema via a cytokine
response similar to inflammatory conditions.
The knowledge of the basic mechanisms involved in vascular
leakage is essential for the development of an effective clinical
treatment. Development of optimal strategies for treating retinal
edema may depend on determining the ratio of the contribution
of intra- and extracellular mechanisms to edema and measuring
how this ratio changes between patients, between different
retinopathies, and during disease progression.
A
With the growing understanding of the pathophysiology of
the macular edema, the therapeutic thinking is likely to change
Fig. 28.1 Cystoid macular edema. (A) Histological section of the from a merely symptomatic treatment (either surgical or medical)
fovea area demonstrating cystoid macular edema with large cystic
spaces in the outer nuclear and plexiform layer as well as in the to a treatment that targets specifically the causal factors involved
inner nuclear layer. Panel B, online. in its formation (e.g., cytokine or growth factor inhibition).
590.e1

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
B
Fig. 28.1, online (B) Schematic drawing of the central retina with large
cysts (top), finally resulting in a pseudohole formation (bottom).
Table 28.1 Causes of macular edema in relation to of techniques measuring accumulation of material from plasma
underlying disorders in the neural retina have been investigated to assess permeabil­
ity. Such accumulation seems diffuse in nature and focal defects 591
Disease group Disorder Pathogenesis
have not been reproducibly described in diabetic mice; as well,
Metabolic Diabetes Abnormal glucose
interpretations of techniques involving tracer accumulation have

Chapter 28
alterations metabolism not been validated in terms of “gold standard” permeability
Aldose reductase surface area product.3 Interestingly, edema has not been demon­
strated in the retina of diabetic mice based on retinal thickness
Retinitis pigmentosa CME: leakage at the
level of RPE
measurements despite the indication of increased permeability.
The BRB consists of the retinal pigment epithelium (RPE) layer
Inherited CME Müller cell disease: (outer BRB), and the vascular endothelium (inner BRB), that
(autosomal- leakage from prohibit the passage of macromolecules and circulating cells

Mechanisms of Macular Edema and Therapeutic Approaches


dominant) perifoveolar
capillaries
from the vascular compartment to the extracellular compart­
ment and therefore intraretinal space.4 Intracellular edema (or
Ischemia Vein occlusion Inner BRB (retinal cytotoxic edema) is defined as cellular swelling that occurs
Diabetic retinopathy capillary without opening of the BRB. Extracellular (or vasogenic) edema
hypoperfusion)
is characterized by retinal thickening in association with loss of
Severe hypertensive Outer BRB (ischemic BRB integrity (Fig. 28.2). While for diabetes and ischemic reti­
retinopathy hypoperfusion of the nopathies the inner BRB was found to play a dominant role in
HELLP syndrome choroid: serous vascular leakage, the importance of the outer BRB has recently
Vasculitis, detachment)
been supported.5 The outer BRB separates the neural retina from
collagenosis
the choroidal vasculature, which is responsible for approxi­
Hydrostatic Retinal vascular Increased mately 80% of the blood supply in the eye. The outer BRB-
forces occlusions intravascular specific leakage of fluorescent macromolecules can be visualized
Venous occlusion pressure
in diabetic and ischemic rodents and substantial leakage of mac­
Arterial hypertension Failure of the BRB
Low IOP romolecules through the outer BRB can be detected.
The breakdown of the inner BRB may occur to a variable
Mechanical Vitreous traction on Epiretinal extent via dysfunction of intercellular junctions, increased trans­
forces the macula membranes with
cellular transport, or increased endothelial cell destruction, and
tangential traction
Vitreomacular result in an increase in vascular permeability (Fig. 28.3B, online).
traction syndrome The initial site of damage that results in the increased vascular
permeability is controversial to date. Although impairment of
Inflammation Intermediate uveitis Mediated by
the perivascular supporting cells such as pericytes and glial cells
prostaglandins
CME is considered might play a role, endothelial cell dysfunction and injury seems
an indication for more likely to be the first pathogenetic step towards the break­
treatment down of the BRB early in the course of the disease. In order to
dissect the molecular and pathophysiologic mechanisms that
Postoperative CME Perivascular
leukocytic infiltrates lead to the accumulation of fluid in the macular area, we have
chosen DME as a model.
DME Diabetic leukostasis
mediates vascular Cell-to-cell junctions and vascular permeability
leakage by
endothelial cell Fluid homeostasis and endothelial permeability are mostly regu­
apoptosis lated by intercellular junctions in the nondiseased retina. Inter­
cellular junctions are complex structures formed by the assembly
Choroidal Vogt–Koyanagi– of a transmembrane and cytoplasmic/cytoskeletal protein com­
inflammatory Harada syndrome
diseases Birdshot ponents. At least four different types of endothelial junctions
retinochoroidopathy have been described: tight junctions, gap junctions, adherence
junctions, and syndesmos. Tight junctions are the most apical
Pharmacotoxic e.g., Mostly via component of the intercellular cleft (Fig. 28.4, online).
effects Epinephrine prostaglandins
(in aphakia) Although the molecular structure of tight junctions generally
Betaxolol appears to be similar in all barrier systems, there are some dif­
Latanoprost ferences between epithelial and endothelial tight junctions, and
CME, cystoid macular edema; RPE, retinal pigment epithelium; BRB, blood– between tight junctions of peripheral and retinal endothelial
retinal barrier; HELLP syndrome, hemolytic anemia, elevated liver enzymes, and cells.6 Expression of selected endothelial cell tight junction genes
low platelet count; IOP, intraocular pressure; DME, diabetic macular edema.
and particularly that of occludin and claudin-5 is reduced in
the diabetic retina.7 In contrast to tight junctions in epithelial
Molecular and cellular alterations leading to systems, structural and functional characteristics of tight junc­
macular edema tions in endothelial cells respond promptly to ambient factors.
Much of the knowledge on the pathophysiology of macular It is likely that inflammatory agents increase permeability by
edema has been determined from extensive experimental studies binding to specific receptors that transduce intercellular signals,
on diabetic retinopathy and diabetic vascular leakage. A variety which in turn cause cytoskeletal reorganization and widening
591.e1

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
A
Fig. 28.3, online (A) In general, water outflow through vessels is
possible via three major routes: paracellular via dysfunction of tight
junctions, transcellular via increased transport, e.g., mediated via Fig. 28.4, online Intercellular junctions in endothelial cells. Endothelial
growth factors, and finally directly via endothelial gaps after cell death. cells are connected and communicate with each other by tight
junctions and adherens junctions. Tight junctions resemble a major
part of the inner blood–retinal barrier. They are built by different
proteins, including occludin, ZO-1, and the claudin family.
Fig. 28.2 Blood–retinal barrier (BRB) and
Vitreous body breakdown in vascular disease. (A) Normal
retinal tissue: dotted blue line marks the
592 outer BRB at the level of the retinal pigment
epithelial cells. The inner BRB is at the level
of the endothelial cells of retinal vessels
(dashed red line). Further structural guidance
Section 2

is given by Müller cells. (B) Diabetic


retinopathy: note the large caveolae of fluid
and accumulation of blood cells in the retinal
tissue, resulting from a breakdown of the
inner BRB. (Courtesy of Sarah Coupland,
MD, Liverpool.)
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

Choroid A B

Fig. 28.3 Pathogenesis of macular edema


and vascular leakage. (B) Vascular leakage
is increased by a variety of factors, including
Increased via: ILM Reduced via inhibition of:
growth factors and inflammatory cytokines.
Leakage in turn is decreased by steroids,
• VEGF • Protein kinase C inhibition of tight junction formation, or
• TGF-β • Myosin light chain kinase antibodies to factors inducing leakage. ILM,
• Adenosine • Tyrosin phosphorylation inner limiting membrane; ELM, external
• Histamine ELM of TJAPs such as: limiting membrane; RPE, retinal pigment
• Prostaglandins • ZO-1, ZO-2 epithelium; VEGF, vascular endothelial
• Interleukins • Occludin, Cingulin growth factor; TGF-β, transforming
RPE growth factor-beta; TNF-α, tumor necrosis
• TNF-α • Cadherin/Cathenin
• IGF-1 • Via: factor-alpha; IGF-1, insulin-like growth
• Glucose • Steroids factor-1; TJAPs, tight junction proteins;
• NSAIDs NSAIDs, nonsteroidal anti-inflammatory
• Anti-VEGF antibodies drugs. Panel A, online.
B • Other agents

of the interendothelial clefts. For example, tumor necrosis factor- altered leukocytes and the endothelial cells and the subsequent
alpha (TNF-α) signals through protein kinase C (PKC)ζ/nuclear endothelial damage represents a crucial pathogenic step9,11,12
factor-kappa B (NF-κB) to alter the tight junction complex and (Fig. 28.5).
increase retinal endothelial cell permeability.8 Endothelial junc­ Inflammatory cytokines such as TNF-α decrease the protein
tions also regulate leukocyte extravasation. Once leukocytes and mRNA content of the tight junction proteins zonula
have adhered to the endothelium, a coordinated opening of occludens (ZO)-1 and claudin-5.8 TNF-α and interleukin-1
interendothelial cell junctions occurs. beta (IL-1β) are elevated in the vitreous of diabetic patients
Similar to the breakdown of the inner BRB, the breakdown of and in the retina of diabetic rats associated with increased
the outer BRB is associated with a significant depletion of the retinal vascular permeability and leukostasis13,14 (Fig. 28.6,
occludin in the RPE of ischemic and diabetic rodents. online). Furthermore, TNF-α is involved in ischemic vascular
changes.15
Inflammation and vascular permeability
Leukocytic infiltration of the retinal tissue characterizes many Growth factors, vasoactive factors, and
inflammatory diseases such as diabetes, pars planitis, or choroi­ vascular permeability
dal inflammatory diseases. The disruption of endothelial integrity leads to retinal ischemia
In diabetes, activated leukocytes adhere to the retinal vascular and vascular endothelial growth factor (VEGF)-mediated iris
endothelium.9,10 Increased leukostasis is one of the first histologic and retinal neovascularization.9,16,17 VEGF is 50 000 times more
changes in diabetic retinopathy and occurs prior to any apparent potent than histamine in causing vascular permeability.18–20 Pre­
clinical pathology. vious work has shown that retinal VEGF levels correlate with
Adherent leukocytes play a crucial role in diabetic retinopathy diabetic BRB breakdown in rodents21 and humans.22 Flt-1(1–3 Ig)
by directly inducing endothelial cell death in capillaries,11 Fc, a soluble VEGF receptor, reverses early diabetic BRB break­
causing vascular obstruction and vascular leakage. Endothelial down and diabetic leukostasis in a dose-dependent manner.17
cell death precedes the formation of acellular capillaries.10 With Early BRB breakdown localizes, in part, to retinal venules and
time, however, acellular capillaries prevail and become wide­ capillaries of the superficial inner retinal circulation23 and can be
spread. Although the mechanism of this destructive process sufficiently reduced by VEGF inhibition (Fig. 28.7, online).
remains elusive, it is clear that the interaction between the Although VEGF is only one of the cytokines involved in the
592.e1

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
Fig. 28.7, online Vascular endothelial growth factor (VEGF) is the key
mediator of vascular damage and finally proliferation in the diabetic
retina. AGE, advanced glycation end-products; IGF, insulin-like growth
factor.
Fig. 28.6, online Inflammatory mediators are involved in leukocyte–
endothelial interaction that, via reduction of tight junction protein
expression and induction of apoptosis, results in vascular leakage.
MCP-1, monocyte chemoattractant protein; Il-8, interleukin-8; MIB-1,
macrophage-inhibitory factor; TNF-a, tumor necrosis factor-alpha;
ZO-1, zona occludens-1.
593

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
non-diabetic 24 months hyperhexosemia

24 months hyperhexosemic ICAM-1-/- mice 24 months hyperhexosemic CD18-/- mice


Fig. 28.5 Inhibition of retinal pathology in long-term hyperhexosemic intercellular adhesion molecule-1 (ICAM-1) and CD18-deficient mice.
Trypsin digests demonstrating a large destruction of the capillary network comparable to nonproliferative diabetic retinopathy with acellular
capillaries and microaneurysms in 24-month hyperhexosemic mice. The capillary network in mice with a “noninflammatory” phenotype (CD-18- or
ICAM-1-deficient) demonstrates almost normal capillaries. (Reproduced with permission from Joussen AM, Poulaki V, Le ML, et al. A central role
for inflammation in the pathogenesis of diabetic retinopathy. FASEB J 2004;18:1450–2.)

pathogenesis of the vascular leakage, it is likely to be one of the To determine the significance of Müller-cell-derived VEGF in
most effective therapeutic targets. diabetic retinopathy, VEGF expression in Müller cells was dis­
On a cellular level, VEGF has been implicated in many differ­ rupted with an inducible Cre/lox system and diabetes-induced
ent mechanisms, which lead to macular edema. VEGF has, for retinal inflammation and vascular leakage was examined in
example, been shown to decrease the proteins responsible for these conditional VEGF knockout (KO) mice. Diabetic condi­
the tightness of the intercellular junctions and induces rapid tional VEGF KO mice exhibited significantly reduced leukosta­
phosphorylation of the tight junction proteins occludin and sis, reduced expression of inflammatory biomarkers, depletion
ZO-1, resulting in breakdown of the BRB.24 VEGF-induced BRB of tight junction proteins, reduced numbers of acellular capil­
breakdown appears to be effected via nitric oxide.17 VEGF also laries, and reduced vascular leakage compared to diabetic
increases paracellular transport without altering the solvent control mice.
drag reflection coefficient.25 Furthermore, VEGF activation of Investigations on cell–cell interactions by D’Amore and
PKC stimulates occludin phosphorylation and contributes to coworkers demonstrated an inhibitory effect of TGF-β secreted
endothelial permeability.26 by pericytes on endothelial cell growth. In diabetic retinopathy
There are tight connections between inflammation and VEGF formation of sorbitol via aldose reductase leads to PKC activa­
expression.17 Recently, Müller-cell-derived VEGF was shown tion, resulting in a loss of the inhibitory balance28 (Fig. 28.8).
to be essential for diabetes-induced retinal inflammation and There are several other vasoactive factors and biochemical
vascular leakage.27 pathways affected by sustained hyperglycemia and known to be
Fig. 28.8 Transforming growth factor-β
Pericyte–endothelial cell interaction Balance of TGF-β (TGF-β) is involved in an inhibitory balance
inhibition of endothelial cells between pericytes and endothelial cells.
594
Glucose

Aldose- NADPH TGF-β


Section 2

Reductase
NADP

Sorbitol
Osmotic effect
TGF-β
Protein kinase C
Others
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

Loss of inhibitory
balance via TGF-β
Degeneration Receptive endothelial cell
of pericytes

Basement membrane
thickening

involved in DME, which are discussed in more detail in other and nutrition die and only acellular basement membranes per­
chapters. sists. A reduction in intraocular pressure may cause macular
High glucose concentration leads to increased diacylglycerol edema with cystoid degenerative changes and secondary atro­
(DAG) by two pathways: de novo synthesis and through dehy­ phic alterations at the outer retina. Similarly, a reduction in
drogenation of phosphatidylcholine. Increased levels of DAG retinal perfusion pressure, often linked to carotid/ophthalmic
mediate PKC activation. Several studies have shown that a artery insufficiency, can have similar retinal manifestations and
decrease in retinal blood flow occurs with PKC activation. Con­ in extreme circumstances there may be retrograde filling of
versely, inhibition of PKC with LY333531 (Eli Lilly, Indianapolis, arteries from fellow veins. Stasis of the blood flow in capillaries
IN) normalized decreased retinal blood flow in diabetic rats.29,30 after venous or arterial occlusions results in rapid apoptosis of
PKC activation causes vasoconstriction by increasing the endothelial cells.38
expression of endothelins (ET), especially ET-1. The expression Similarly, in diabetes, retinal barrier breakdown is at least
of ETs can be induced by a variety of growth factors and cyto­ in part due to endothelial cell damage and apoptosis. The pro­
kines, including thrombin, TNF-α, TGF-β, insulin, and vaso­ apoptotic molecule Fas-ligand (FasL) induces apoptosis in cells
active substances including: angiotensin II, vasopressin, and that carry its receptor Fas (CD 95).39 There is evidence that
bradykinin. FasL is expressed on vascular endothelium where it functions
Furthermore, retinal vascular endothelial cells are very sen­ to inhibit leukocyte extravasation. The expression of FasL on
sitive to histamine. Several studies have documented increased vascular endothelial cells might thus prevent detrimental
vascular histamine synthesis in diabetic rats and humans.31–33 inflammation by inducing apoptosis in leukocytes as they
The administration of histamine reduces ZO-1 protein expres­ attempt to enter the vessel. In fact, during inflammation and
sion and thus correlates with vascular permeability. The H1 ensuing TNF-α release, the retinal endothelium upregulates
receptor stimulates PKC that has been implicated in increased several adhesion molecules40 that mediate the adherence of the
retinal vascular permeability.34 Interestingly, Aiello and leukocytes, but also downregulates FasL thus allowing leuko­
coworkers showed that administration of LY333531, a PKC-β cyte survival and migration to active sites of inflammation. In
isoform-selective inhibitor, does not significantly decrease experimental diabetic retinopathy, inhibition of Fas-mediated
histamine-induced permeability but instead VEGF-induced apoptotic cell death reduces vascular leakage.41 The cumulative
permeability. In contrast, administration of nonisoform- endothelial cell death during the course of diabetes plays a
selective PKC inhibitors did significantly suppress histamine- causal role in the pathogenesis of the diabetic vascular leakage
induced permeability.35 and maculopathy.
Furthermore, in vascular endothelial cells, advanced glycation
Extracellular matrix alterations and
end-products (AGE) may affect the gene expression of ET-1 and
vascular permeability
modify VEGF expression. The AGE-stimulated increased VEGF
Degradation of the extracellular matrix affects endothelial cell
expression is dose- and time-dependent and additive to
function at many levels causing endothelial cell lability which is
hypoxia.36,37
required for cellular invasion and proliferation, or influencing
Endothelial cell death and vascular permeability the cellular resistance and therefore the vascular permeability.
Where intraluminal pressure falls below a critical closing pres­ The degradation and modulation of the extracellular matrix are
sure the tone of the arteriolar wall cannot be maintained and the exerted by matrix metalloproteinases (MMPs), a family of zinc-
downstream capillary bed collapses and endothelial cells become binding, calcium-dependent enzymes.42 Elevation of MMP-9 and
“fibrin-locked.” Endothelial cells deprived of their circulation MMP-2 expression has been shown in diabetic neovascular
membranes,43,44 although a direct effect of glucose on MMP-9 retina, substrates reach Müller cells and photoreceptors from the
expression in vascular endothelial cells could not be shown.45 It choroid via the RPE.52 Microglia associate intimately with
is probable that MMPs participate at various stages during the neurons that express molecules, such as CX3CL1 (fractalkine) 595
course of the BRB dysfunction and breakdown. Their actions and CD20, that negatively regulate microglial activation through
include early changes of the endothelial cell resistance with their respective receptors. As such, perturbation of expression of

Chapter 28
influence on intercellular junction formation and function46 to ligand or receptor during stress would activate microglia to
active participation in endothelial and pericyte cell death47 that produce proinflammatory cytokines and acquire an activated
occurs late in the course of the disease. morphology. Activated microglia produce chemokines such as
monocyte chemoattractant protein-1, inducing expression of
Transcellular transport and vascular permeability
adhesion molecules, which can promote the leukostasis of neu­
Disruption of the BRB is an early phenomenon in preclinical
trophils on endothelium, and potentially inducing the extravasa­
diabetic retinopathy. Two vascular permeability pathways may
tion of inflammatory macrophages.52,54 Induction of glial fibrillary

Mechanisms of Macular Edema and Therapeutic Approaches


be affected, the paracellular pathway involving endothelial cell
acidic protein (GFAP) is a marker of glial activation and increased
tight junctions, and the endothelial transcellular pathway medi­
expression of this protein occurs in Müller cells from the retinas
ated by endocytotic vesicles (caveolae). Despite the fact that
of diabetic patients, but also after ischemic injury.
pinocytic transport is critically involved in the transepithelial
fluid exchange, its role in the pathogenesis of increased vascular Mechanical factors involved in
leakage in diabetes is just emerging.48,49 The importance of the the formation of macular edema
regulation of fluid homeostasis by active cellular transport of
Clinical and anatomic evidence indicates that abnormalities in
nutrients and fluid via pinocytosis is underlined by recent data
the structure of the vitreoretinal interface may play an important
suggesting a transient induction of the paracellular pathway and
role in the pathogenesis of DME.55–57 It was suggested that vit­
prolonged involvement of transcellular endothelial transport
reoretinal adhesions in diabetic eyes are stronger than the shear
mechanisms in the increased permeability of retinal capillaries
forces of traction from vitreous shrinkage and this in turn may
in diabetes.7
lead to the development of vitreomacular traction and subse­
It is currently known that one of the factors involved in the
quently to macular edema.58 Nevertheless, the risk of developing
regulation of pinocytic transport is VEGF. VEGF increases vas­
diffuse macular edema was 3.4-fold lower in the group of eyes
cular permeability not only by disrupting the intercellular tight
with complete posterior vitreous attachment or complete vitreo­
junctions between the retinal endothelial cells but also by induc­
retinal separation compared to the eyes with vitreomacular
ing the formation of fenestrations and vesiculovacuolar organ­
adhesion.59
elles. The role of VEGF in the disruption of the pinocytic transport
The vitreous humor is a gel-like structure composed mostly of
that is translated into increased vascular permeability in disease
water (99%), hyaluronic acid, and collagen. A structural barrier
states is still controversial.50 Whereas, in higly permeable blood
between the vitreous cavity and the retina is formed by the inner
vessels the number of pinocytotic vesicles at the endothelial
limiting membrane (ILM), which is localized between the inner­
luminal membrane transporting plasma immunoglobulin G is
most layer of the retina and the outer boundary of the vitreous.
significantly increased, no fenestrations or vesicles were found
The ILM shows typical ultrastructural characteristics of a basal
in the endothelial cells of the VEGF-affected eyes when exam­
lamina, is found in close contact with the foot processes of Müller
ined by electron microscopy.
cells, and contains proteins that are typically found in basal
Neuronal involvement in the formation of laminae such as collagen type IV and laminin.60 Striated collagen
macular edema fibrils of the vitreous cortex insert into the inner portion of the
Recent research on DME emphasizes the role of neuronal cells ILM,61 which is also known as the hyaloid membrane of the
in the diabetic retinal damage. The retina consists of a network vitreous. Detachment of the posterior hyaloid membrane with
of neurons and glia (astrocytes, Müller cells, and microglial cells) aging or pathology results in a condensation of the posterior
that comprise approximately 95% of the tissue, with blood vitreous surface (membrana hyaloidea posterior). In youth, there
vessels representing less than 5% of the retinal mass.51 As the is adhesion between the vitreous cortex and the ILM that is
network of retinal neurons and glia is intimately linked, there is stronger than Müller cells themselves and Müller cell foot pro­
no doubt that the neural (photoreceptors, bipolar cells, horizon­ cesses become separated from their main cell body and remain
tal cells, amacrine cells, and ganglion cells) and vascular compo­ connected to the posterior aspect of the ILM when this is separ­
nents of the retina are closely associated by metabolic synergy ated from the retinal surface.62
and paracrine communication.52,53 Neuroglial cells are involved There has been a controversial discussion regarding the
in vision, and blood vessels provide nutrients to facilitate the embryonic origin of the ILM, which can be demonstrated as
process.54 early as 4 weeks after gestation in the human eye.63,64 Tradition­
The functional integration of blood vessels with the neurosen­ ally, the ILM has been considered to be synthesized by Müller
sory retina is clinically evident during autoregulation in which cells. This concept has been challenged by data presented from
retinal arterioles and venules constrict in response to hyperten­ Sarthy, who investigated the expression of collagen type IV
sion and hyperoxia and dilate in response to hypercapnia. Like­ during development of the mouse eye.65 ILM proteins appear to
wise, disorders of the neurosensory retina and retinal vasculature originate largely from lens and ciliary body, although a contribu­
are integrally linked, and understanding of the neurodegenera­ tion of retinal glial cells in ILM synthesis cannot be excluded. In
tive component of retinal damage is of importance for treat­ support of this are data which show that also other ILM proteins
ments of macular edema. such as perlecan, laminin-1, nidogen, and collagen XVIII are
In the inner retina, metabolic substrates, such as glucose, flow expressed predominantly in lens and ciliary body, but are not
from vascular endothelium to astrocytes to neurons. In the outer detected in the retina.66
Diffuse DME has been found in association with an attached, The RPE cells may respond to the injury in several ways: if the
thickened, and taut posterior hyaloid.67 As immunocytochemical lesion is relatively small, the RPE defect can be filled by cell
596 staining for cytokeratin (found in RPE) and GFAP protein (found spreading; if the defect is relatively large, the cells can proliferate
in astrocytes and Müller cells) demonstrated the existence of to resurface the area, and the RPE can produce cytokines (e.g.,
cells in the premacular posterior hyaloids, suggesting a possible TGF-β) that antagonize the permeabilizing effects of VEGF.80,81
role for cell infiltration in the development or maintenance of Laser therapy is well established in diabetic retinopathy as
Section 2

macular edema. It remains to be elucidated whether these cells well as in diseases with peripheral retinal ischemia. The Early
in the posterior vitreous cause macular edema physiologically Treatment Diabetic Retinopathy Study (ETDRS) was designed
rather than mechanically through the production of cytokines. to evaluate the effects of argon laser photocoagulation for DME
in a prospective, randomized, multicenter clinical trial. Among
TREATMENT OF MACULAR EDEMA the subgroup of eyes with mild to moderate nonproliferative
diabetic retinopathy with DME, visual acuity improved in 16%,
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

The current therapy for macular edema targets conditions remained unchanged in 77%, and worsened in 7% of treated
where mechanical traction, hydrostatic force, or inflammation, eyes, whereas visual acuity improved in 11%, remained
plays a pathogenetic role. Laser coagulation, pharmacological unchanged in 73%, and worsened in 16% of untreated eyes after
approaches, and surgical measures are the most frequently used 2 years of follow-up. After 3 years of follow-up, vision worsened
therapies. in 12% of treated eyes compared to 24% of untreated eyes. There
was a statistically significant benefit of laser photocoagulation in
Laser treatment this group of eyes. Nevertheless, a current literature review indi­
Many studies have demonstrated a beneficial effect of photo­ cates that, at least in selected groups, a beneficial effect of pan­
coagulation therapy for DME.68–73 The exact mechanism of action retinal photocoagulation with respect to DME can be identified.
of laser photocoagulation-induced resolution of DME is The exact relationship between peripheral ischemia and DME
unknown. In short, a laser-induced destruction of oxygen- and thus the relevance of peripheral panretinal photocoagula­
consuming photoreceptors has been discussed as well as cell tion in these patients as a treatment for DME still remains to be
death and scarring (involving gliosis and RPE hyperplasia) determined.
induced by the temporary rise in tissue temperature. Oxygen While focal laser coagulation reduces hypoxic areas and
that normally diffuses from the choriocapillaris into the outer directly occludes leaky microaneurysms, the rationale for grid
retina can now diffuse through the laser scar to the inner retina, laser treatment in DME is not yet well established. Potentially,
thus relieving inner retinal hypoxia.74,75 There are contrasting grid laser may have its beneficial effect by thinning the retina,
data whether an increased preretinal oxygen partial pressure is bringing retinal vessels closer to choroidal vessels, permitting
involved and allows for microvascular repair in the treated the retinal vessels to constrict by autoregulation, thereby decreas­
areas.76,77 ing retinal blood flow and consequently decreasing edema
When studying the diameter of retinal arterioles, venules, and formation.82
their macular branches before and after macular laser photoco­ Potential side-effects are choroidal neovascularization induc­
agulation in eyes with DME, the macular arteriolar branches tion (Fig. 28.9) or lack of effect in cases of diffuse edema
were found to be constricted by 20.2% and the venular branches (Fig. 28.10).
13.8%. This was attributed to an improved retinal oxygenation
caused by the laser treatment leading to autoregulatory vasocon­ Medical treatment
striction, improving the DME.78 General aspects of systemic and topical
According to another theory, the beneficial effect of laser pho­ medical therapy
tocoagulation is due to an enhanced proliferation of RPE and In many cases, macular edema is caused by a generalized health
endothelial cells leading to repair and restoration of the BRB.79 problem such as diabetes, high blood pressure, or inflammatory

A B C

Fig. 28.9 Complications after grid laser coagulation. (A) Enlarged central retinal pigment epithelial scars of the left eye, no obvious macula
edema. (B) Early-phase fluorescein angiography demonstrates choroidal neovascularization lesion growing from earlier photocoagulation
scars at the right eye. (C) Late phase with increased leakage of fluorescein.
597

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
A B

Fig. 28.10 (A, B) Grid laser treatment in persistent diffuse diabetic macular edema has limited effects. The optical coherence tomography
imaging demonstrates no difference in retinal thickness prior to and 3 months after grid laser treatment.

conditions.83 These generalized diseases need to be treated prior


to any other measures. There have been several reports in the H 3 Na K
literature that such treatments – particularly in diabetes, high
blood pressure, and inflammatory diseases – can cure macular
edema without any additional specific ocular treatment.84
Na 2 K K/Na 2 Cl Na 2 HCO3
Modification of the systemic blood flow, including hyperbaric
Cl Na Na
oxygen treatment, is thought to alter blood flow via vasocon­
striction, and facilitates the reformation of damaged junctional
complexes in the vessel wall. However, despite an improvement
in visual acuity in patients with chronic CME after cataract
HCO3 HCO K Cl
extraction, there was no correlation to macular edema.85 In
uveitis-associated CME hyperbaric oxygen had no signifcant Carbonic anhydrase NSAIDs
effect. Other rheological treatments such as plasma membrane inhibitors
filtration demonstrated good effects in initial studies; however
these treatments did not enter large-scale prospective studies.86 Fig. 28.11 (A) Schematic diagram showing selected ion channels in
Still, there is no evidence that rheological measures have any the retinal pigment epithelium. Note that carbonic anhydrase inhibitors
act on the bicarbonate/chloride exchange channel in the basal
effect on inflammatory or DME. membrane, and nonsteroidal inflammatory drugs have been shown
Medical treatment of macular edema so far is best established to act on the chloride channel in the basal membrane of the retinal
in postsurgical and predominantly inflammatory edema, e.g., in pigment epithelium. Both channels are associated with transcellular
fluid transport. (Panel B online.)
uveitis, but gaining importance in the treatment of DME. The
majority of the therapeutic strategies inhibit the release of inflam­
matory mediators and therefore target the pathogenetic factors
responsible for the altered vascular permeability.
activity of cells and may act as a bicarbonate channel.89,90 The
Medical treatment for macular edema consists of therapeutic
CA activity in the RPE shows a clearcut polarized distribution
agents that are collectively categorized into three groups: cor­
with a large amount of enzyme on the apical surface of the cell,
ticosteroids, cyclooxygenase (COX) inhibitors, and carbonic
whereas there is less CA activity on the basolateral portion of
anhydrase (CA) inhibitors.
the cell membrane.91 Further immunohistochemical differentia­
Carbonic anhydrase inhibitors and nonsteroidal tion has shown the isozyme IV is responsible for apical CA
anti-inflammatory drugs activity in the RPE.89 Intravenous injection of acetazolamide has
CA inhibitors have been used clinically for over 20 years in the been shown to decrease the pH in the subretinal space in both
treatment of macular edema. The initial observation on its thera­ chicks and cats.89,92 This acidification was followed immediately
peutic efficacy was reported in 1988 in a study of 41 patients with by a reduction of the subretinal volume, and it has been postu­
CME of various etiologies.87 The rationale of CA inhibitors as a lated that it is the acidification that induces changes in ion and
therapeutic agent in the treatment of macular edema is to consequent fluid transport through the RPE91 (Fig. 28.11, panel
improve the ability of the RPE to pump fluid out of the retina B online).
by modulating the polarized distribution of CA at the level of Under normal conditions, roughly 70% of the subretinal fluid
the RPE.88 is removed by metabolic transport to the choroid. In an in vivo
In the retina, CA is found in the cytoplasm of red/green cones rabbit model, this fluid transport, which is driven to a large
(albeit not in rods) and especially inside Müller cells. The RPE, extent by active ion transport through the RPE, can be enhanced
however, appears to contain almost exclusively the membrane- by acetazolamide93,94 Furthermore, experiments in an animal
bound form of CA.89 The latter appears to regulate and modu­ model of iatrogenically induced retinal detachments showed
late the extracellular pH gradients created by the metabolic that the disappearance of fluorescein through the RPE increased
A
597.e1

Chapter 28
Mechanisms of Macular Edema and Therapeutic Approaches
B

Fig. 28.11, online (B) Membrane-bound carbonic anhydrase (CA IV)


55 kDa. NSAIDs, nonsteroidal anti-inflammatory drugs. (Adapted with
permission from Wolfensberger TJ, Mahieu I, Jarvis-Evans J, et al.
Membrane-bound carbonic anhydrase in human retinal pigment
epithelium. Invest Ophtholmol Vis Sci 1994;35:3401–7.)

Fig. 28.13, online (A) Action of steroids on arachidonic acid. (B)


Action of steroids on growth factor release. COX, cyclooxygenase;
NSAIDs, nonsteroidal anti-inflammatory drugs; PG, prostaglandin; TX,
thromboxane; LT, leukotriene; VEGF, vascular endothelial growth
factor; PDGF, platelet-derived growth factor; PAF, platelet-activating
factor; BRB, blood–retinal barrier.
Fig. 28.12, online Nonsteroidal anti-inflammatory drugs (NSAIDs).
Interaction of NSAID with vascular endothelial growth factor (VEGF)
and inflammatory cytokines and their signaling pathways. COX-1,
cyclooxygenase 1; PGs, prostaglandins; TXs, thromboxanes;
TNF-a, tumor necrosis factor-alpha; NF-kB, nuclear factor-kappa B;
eNOS, endothelial nitric oxide synthase; NO, nitric oxide;
ICAM-1, intercellular adhesion molecule 1; MEK, miogen-activated
protein kinase.
by 25% after intravenous injection of acetazolamide.95 The same Corticosteroids
authors also observed a marked increase in resorption of sub­ Steroids are currently regaining attention by the growing use
598 retinal fluid at a higher dosage of 50–65 mg/kg body weight. of intravitreal triamcinolone. Corticosteroids block the release
Further studies on the frog RPE demonstrated that active chlo­ of arachidonic acid from cell membranes by inhibiting the
ride and bicarbonate transport probably occurs at the basal enzyme COX and thus reduce the synthesis of prostaglandins,
surface, which faces the choroidal blood supply and it was pos­ but they also have a multitude of other anti-inflammatory
Section 2

tulated that subretinal fluid absorption occurs at this level.90 effects by acting, among others, on IL-1 and by reducing vas­
Currently, there are no randomized studies available that cular permeability109 (Fig. 28.13A, online). Their additive anti-
confirm a beneficial effect of CA inhibitors in the treatment of inflammatory effect to NSAIDs has been shown to be useful in
macular edema. Nonrandomized observations demonstrated the treatment of various postoperative inflammatory condi­
improved visual function in patients with postsurgical macular tions. One potential mode of action is the increased resorption
edema, e.g., after cataract surgery or buckling procedures.87,96 of fluid through the RPE, although the exact mechanism of this
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

The effect lasts only as long as the patient takes the drug (on–off is not as yet clear. Steroids specifically stabilize endothelial tight
effect, tachyphylaxis).96 The favorable reports that were described junctions and increase their numbers.110,111 Another action of
at first regarding the application of CA inhibition in patients steroids is the downregulation of the production of the VEGF,
with macular edema secondary to retinitis pigmentosa are not which, in turn, renders the BRB tighter (Fig. 28.13B, online).
supported by the long-term observation. With the continuous Steroids also downregulate VEGF production,112 specifically in
use of methazolamide a rebound phenomenon is observed.97 the retina,113,114 and this explains the clinical observation that the
Contrary to tenacious clinical habit, the use of CA inhibition application of steroids both intravitreally and into the subtenon
for intraretinal macular edema is not based upon scientific space can reduce macular edema considerably. Furthermore,
evidence to date. The use of CA inhibitors for DME is not steroids have been shown to prevent the induction of VEGF
recommended. production by platelet-activating factor and platelet-derived
growth factor.115 Triamcinolone also inhibits IL-6- and VEGF-
Nonsteroidal anti-inflammatory drugs (NSAIDs) induced angiogenesis downstream of the IL-6 and VEGF recep­
As COX inhibitors block the synthesis and release of prostaglan­ tors.116 Leukocyte adhesion plays an important role in macular
dins, nonsteroidal drugs have been investigated in the prophy­ edema – particularly so in diabetic maculopathy.11,12 The endo­
laxis and therapy of postsurgical CME. The action is based on thelial damage resulting from this leukocyte adherence to vessel
the inhibition of the enzyme COX, which in turn inhibits the walls is mediated by nitric oxide, adhesion molecules, and other
production of prostaglandins, a degradation product of arachi­ inflammatory mediators.17,117 Subtenon triamcinolone inhibits
donic acid in the eye.98 Diclofenac sodium in high doses inhibits leukocyte–endothelium interactions in the retina and down­
the formation of leukotrienes, which amplify cellular infiltration regulates adhesion molecules of retinal vascular endothelium118
during an inflammatory reaction. Other NSAIDs have been and thus decreases the retinal thickness.
shown to modulate chloride movement, and, as a consequence, The Müller cells represent a further site of action of steroids.119
fluid movement through the RPE.99 The effect of COX inhibitors Macular edema is thought to be partly linked to the downregula­
on inflammatory aspects of DME was only demonstrated in tion of the Müller cell protein Kir4.1. The resultant increase in
preclinical studies13 (Fig. 28.12, online). intracellular K+ leads to the uptake of proteins and osmotic
Thus, NSAIDS target the inflammatory mediators that are swelling of the Müller cells via aquaporin 4 channels. The
responsible for the edema formation and, although they may not administration of triamcinolone reduces the production of
be an optimal standalone treatment, they can be used as steroid- VEGF, arachidonic acid, and prostaglandins, allowing the reac­
sparing agents. Topical NSAIDs have become the mainstay in tivation of fluid clearance by Müller cells via endogenous ade­
the treatment of inflammatory CME.100 The clinical efficacy of nosine and an increase in TWIK-related acid-sensitive potassium
topical NSAIDs has been shown to be of value both in the (TASK) channels. These processes lead to an efflux of potassium,
prevention101–103 and in the treatment104,105 of inflammatory CME, thus correcting the downregulation of the Kir4.1 protein.119
particularly when related to cataract surgery. Two double- Corticosteroids have different potency levels depending on
masked, placebo-controlled studies in which corticosteroids their chemical composition120 and the newer synthetically pro­
were not used demonstrated that ketorolac 0.5% ophthalmic duced compounds show an up to 25-fold increase in activity, as
solution, administered for up to 3 months, improves vision in compared to cortisone. These new agents, such as triamcinolone,
some patients with chronic CME after cataract surgery.106,107 A dexamethasone, and fluocinolone acetonide, have fluor at the 9α
meta-analysis of the results from several different randomized position, which increases corticosteroid receptor binding. Routes
controlled trials suggests that NSAIDs are beneficial as a medical of clinical administrations are manifold, including topical,
prophylaxis for aphakic and pseudophakic CME and as medical periocular, intraocular, oral, and intravenous routes. Subtenon
treatment for chronic CME.108 injections of corticosteroids are widely used in patients with
On the basis of these findings, it has been suggested to asymmetric or unilateral uveitis. The advantages of the periocu­
employ topical NSAIDs in the treatment of inflammatory CME, lar injections are high concentrations of corticosteroids in the
especially when related to ocular surgery. posterior eye, and reduction of the adverse effects compared to
There may be several explanations for why NSAIDs cannot systemic administration. Intraocular levels of corticosteroids are
improve vision in DME, such as chronic edema, inflammation, identical between subtenon and retrobulbar administration.121
and ischemia that induce permanent structural alterations. For oral administration, the initial high dose (1–1.5 mg/kg) is
Although effects on diabetic vascular leakage were achieved in subsequently decreased according to clinical effect.122
preclinical studies,13 there is so far no clinical evidence for an Recent publications suggest that the intravitreal application of
effect of NSAIDs in DME. triamcinolone seems to be a promising therapeutic method for
macular edema that fails to respond to conventional treat­ edema of different origins.133–135 Ranibizumab (Lucentis) and
ment.123,124 Martidis et al. published a prospective, noncompara­ bevacizumab (Avastin) are antibodies with high affinity for
tive, interventional case series to determine if intravitreal VEGF, which bind to all VEGF isoforms. 599
injection of triamcinolone acetonide is safe and effective in treat­ The intravitreal injection of bevacizumab potentially reduces
ing DME unresponsive to prior laser photocoagulation.124 Most not only VEGF but also stromal cell-derived factor 1α. This sug­

Chapter 28
recent data of a randomized clinical trial demonstrated that, over gests that intravitreal bevacizumab may influence intraocular
a 2-year period, focal/grid photocoagulation is more effective mediators other than VEGF.136
and has fewer side-effects than intravitreal triamcinolone, sup­ Clinical studies have proven the effect of ranibizumab in
porting that focal/grid photocoagulation currently should be the several forms of macular edema; it is approved for the treat­
benchmark against which other treatments are compared in ment of DME and macular edema after vein occlusion in the
clinical trials of DME.125 In comparison to VEGF inhibitors tri­ USA and Europe (Fig. 28.14). Other VEGF inhibitors such as
amcinolone has a lesser long-term effect. Cataract and secondary VEGF trap or small-molecule inhibitors are currently being

Mechanisms of Macular Edema and Therapeutic Approaches


glaucoma formation have to be taken into consideration. investigated in clinical trials. The VEGF aptamer pegaptanib
Similar studies were performed for patients with uveitic (modified RNA oligonucleotide, which binds and inactivates
macular edema, central vein occlusion, and CME after cataract VEGF165 only) has been shown in animal models to restore
surgery.123,124 In most published reports, complications do not the BRB in diabetic retinopathy.137 Similar to the developments
appear to be prohibitive. in AMD, lesser injections and slow-release systems are in
Reviewing the published data on intravitreal injections of tri­ demand as well as better knowledge about the combination of
amcinolone acetonide, the therapeutic window seems very wide. VEGF inhibitors with systemic treatment of the underlying
The dose range of intravitreally injected triamcinolone acetonide disease.
varies from 2 mg125 to 4 mg124 and even 25 mg in a single report.126
Interestingly, reaccumulation of fluid in cystoid spaces occurs Other medical treatments
between 6 weeks and 3 months after injection, and this does not Steroid-sparing immunosuppressive drugs are frequently used
seem to be dose-dependent. Repeated injections at intervals as additional second-line agents, particularly in patients with
ranging from 10 weeks127 to more than 6 months show a variable severe intraocular inflammation and CME.1 The rationale for
treatment response. these treatments relies on the inhibition of several different pro­
There are currently no data on the pharmacokinetic profile of inflammatory cytokines, which are specifically involved in
intravitreal triamcinolone, which might be altered from a previ­ causing macular edema by breaking down the BRB in intraocu­
ous vitrectomy. Physiological intravitreal cortisol levels are lar inflammatory disorders.
reported to be 5.1 ng/mL, vitreous levels after peribulbar injec­ Apart from the well-known agents such as VEGF, prostaglan­
tions are in the range of 13 mg/mL. The effective dose of the dins, and leukotrienes, these cytokines also include insulin-like
triamcinolone acetonide is further influenced by the mandatory growth factor 1, IL-6, stromal cell-derived factor 1, and hepato­
washes of the widely used stabilizing agent benzylethanol cyte growth factor. Particularly elevated levels of intraocular
during the preparation of the injection that, even if standardized, VEGF and IL-6 have been correlated with the severity of uveitic
they alter the remaining amounts of the drug in the solution. macular edema138 and treatments directed specifically against
Additionally, an inhibitory effect of the stabilizing agent on the these factors have been proposed.
drug cannot be excluded. Promising results have also been reported using interferon
Jaffe and coworkers128,129 constructed a fluocinolone acetonide α2139 as a treatment for long-standing refractory CME in uveitis.
drug delivery device that releases fluocinolone acetonide in a In addition, a beneficial effect of interferon on inflammatory
linear manner over an extended period. A clinical phase III study CME was noted in a retrospective study of patients with mul­
by Bausch & Lomb investigated the efficacy of 0.5 mg (slow- tiple sclerosis-associated intermediate uveitis.140 Others reported
release) fluocinolone acetonide in 80 patients with diffuse DME. comparable efficacy of cyclosporine A to prednisolone in the
Patients receiving the implant showed a statistically significant treatment of macular edema in patients with endogenous
regression of retinal thickness after 6 months in comparison to uveitis.141 Anti-TNF therapy has also been demonstrated as a
the control group. promising therapy for uveitic macular edema.142 Somatostatin
The safety and efficacy of dexamethasone intravitreal implant analogs such as octreotide may also be effective in the treat­
(DEX implant; Ozurdex, Allergan, Irvine, CA) compared with ment of CME by blocking the local and systemic production
sham in eyes with vision loss due to macular edema associated of growth hormone, insulin-like growth factor, and VEGF.143
with branch retinal vein occlusion or central retinal vein occlu­ Treatment with octreotide resulted in marked improvement,
sion as well as posterior uveitis was assessed. The device was or even complete resolution, of CME in uveitic patients.144
found to reduce the risk of vision loss in these conditions, while Similarly, calcium dobesilate prevents the BRB breakdown
the vitreoretinal pharmacokinetic profiles were, in contrast to induced by diabetes, by restoring tight junction protein levels
triamcinolone, similar between nonvitrectomized and vitrecto­ and organization and decreasing leukocyte adhesion to retinal
mized eyes.130–132 vessels. The protective effects of calcium dobesilate likely
involve the inhibition of p38 mitogen-activated protein kinase
Antiangiogenic treatment and NF-κB activation, possibly through the inhibition of
Anti-VEGF agents oxidative/nitrosative stress.
As discussed above, there are several reports which support the
notion that corticosteroids act as indirect anti-VEGF agents. Surgical approaches
However, more recently, directly acting anti-VEGF agents have Vitrectomy with or without peeling of posterior hyaloid
come to the forefront as promising treatment options for macular membrane may be beneficial for the treatment of DME in eyes
A B
600
Section 2
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

Fig. 28.14 Optical coherence tomography imaging of diabetic macular


edema before and after injection of ranibizumab. (A) Prior to injection:
cystoid spaces and macula thickening. (B) Resolution of the cystic
spaces 3 weeks after injection. (C) Four weeks after second injection,
the macula remains “normal.”

that are resistant to laser photocoagulation and/or steroid


injection.
There is clinical evidence that traction forces at the vitreo­
retinal interface may play an important role in the pathogenesis
of macular edema. Several authors have studied vitrectomy
for persistent macular edema and have suggested that release
of the tractional forces at the vitreomacular interface may
improve resolution of the macular edema and restore visual
acuity (Fig. 28.15).
Although pars plana vitrectomy may be considered as a very
simple surgical procedure, its manifold effects on a cellular level A
are becoming more and more understood.2
Tractional origin of macular edema and
surgical aspects
The initial rationale for using vitrectomy in cases of macular
edema was entirely structural, i.e., aimed at the removal of vitre­
ous traction on the macula.55,145 The effect of traction on retinal
structures becomes more understandable using Newton’s third
law: to any action there is always an equal reaction in the oppo­
site direction. The force of vitreoretinal traction will thus be met
by an equal and opposite force in the retina, resulting in the
retinal tissues being pulled apart. Eventually this results in the
lowering of the tissue pressure within the retina, which in turn
B
increases the difference between the hydrostatic pressure in the
blood vessels and the tissue and contributes thus to edema for­
mation (Starling’s law). Releasing the traction will increase tissue Fig. 28.15 Release of traction after spontaneous posterior vitreous
detachment in a 34-year-old diabetic patient. (A) Visual acuity 20/200
pressure and lower the hydrostatic pressure gradient, reducing with cystoid edema and vitreomacular traction. (B) Spontaneous
the water flux from blood vessels into retinal tissue.2 resolution of the traction with vitreous detachment. Visual acuity 20/30.
Vitreoretinal traction associated with macular edema has (2–8%), development of hard exudates (4%), macular ischemia
been identified in diabetic retinopathy, following complicated (11%), and neovascular glaucoma (4–8%).
cataract surgery (Irvine–Gass syndrome) and in several other 601
disease entities. The removal of such traction by vitreoretinal
surgery has been found to be beneficial.55,145,146 Peeling of the ILM
DISCUSSION AND CONCLUSION

Chapter 28
of the retina ensures complete release of tractional forces, Unfortunately, even the currently available surgical and phar­
removes a potential diffusional barrier, and inhibits reprolifera­ macological treatments have suboptimal results in many cases.
tion of fibrous astrocytes.147 Therefore, there is an obvious need for the development of a
The rational for vitrectomy (removal of the hyaloid) plus more effective and targeted treatment that can be satisfied only
peeling of the ILM is the postulated improvement of fluid diffu­ by a better understanding of the pathophysiology. There is still
sion from the retina to the vitreous cavity and is explained in a need for discussion of functional endpoints for clinical studies
more detail in Chapter 118, CME and vitreomacular traction. (e.g., microperimetry or near visual acuity) and of clinical moni­

Mechanisms of Macular Edema and Therapeutic Approaches


However, vitrectomy may not only be beneficial in the pres­ toring (decrease in retinal thickness on optical coherence tomog­
ence of macular traction, but also in cases where no particular raphy, or a “dry macula” on angiography). Further, clinically
deformation of the macula can be identified. feasible retreatment criteria need to be determined for the abso­
This is particularly true for macular edema of vascular origin, lute and relative thickness variations as well as visual acuity
such as diabetes or retinal vein occlusion. The beneficial effect measures.
of vitrectomy is thought to be based, at least in part, on two Ischemic maculopathy remains untreatable in the area of
mechanisms. First, it has been found, for example, that oxygen laser coagulation. A foveal avascular zone of more than 500  µm
transport between the anterior and posterior segments of the eye should be considered ischemic and thus, to the current recom­
is increased in the vitrectomized lentectomized eyes.148,149 Others mendations, cannot be treated. The value of treatment for
have shown that pharmacologic vitreolysis also improves ischemic maculopathy requires further investigation when
oxygen diffusion within the vitreous cavity.150 This means that determined upon angiography.
following vitrectomy and/or posterior vitreous detachment, the Early intervention in macular edema is undoubtedly advanta­
transport of molecules to and from the retina is increased. geous, as the risk of ultrastructural alterations induced by a
Second, it has been shown that several growth factors such as persistent macular edema increases with time. Late treatment
VEGF, IL-6, platelet-derived growth factor, and others are risks transition to untreatable ischemic forms of macular edema.
secreted in large amounts into the vitreous during proliferative To date, however, most surgical approaches will only be consid­
vasculopathies such as diabetic retinopathy or retinal vein occlu­ ered for persistent macular edema that is not responsive to laser
sion151,152 and it is conceivable that a complete vitrectomy will treatment or pharmacological approaches. Earlier intervention
remove this excess of growth factors mechanically with the could be possibly more advantageous.
desired effect of a restitution of the BRB. The rapid clearance of The different treatment approaches are likely to affect the
VEGF and other cytokines may thus help to prevent macular clinical course of macular edema at variable time points and
edema and retinal neovascularization in ischemic retinopathies, for different time periods. While the effect of intravitreal ste­
such as diabetic retinopathy and retinal vein occlusions. Vitre­ roids is known to deteriorate with time, similar fluctuations
ous clearance of growth factors may indeed have the same effect are likely for other treatments. Especially for the surgical options
as the presence of, for example, VEGF antibodies in the vitreous long-term data are currently unavailable. Thus, any treatment
cavity.2,153,154 option should be evaluated for the duration of the anticipated
If judging the final benefit, the complications encountered beneficial effect and beyond. As macular edema requiring treat­
with pars plana vitrectomy for DME should be considered, and ment appears to be mostly chronic, a follow-up of even 1 year
include cataract formation (up to 63%), retinal detachment (10%), is not sufficient.
retinal tear (14–21%), tractional rhegmatogenous retinal detach­ In the future, when more specific treatments become available,
ment (5–8%), vitreous hemorrhage (12–16%), epiretinal mem­ study design and also routine clinical follow-up will require
brane formation (8–12%), fibrinoid syndrome (8%), glaucoma adjustment.

Bonus images for this chapter can be found online at http://www.expertconsult.com


Fig. 28.1 (B) Schematic drawing of the central retina with large Fig. 28.6 Inflammatory mediators are involved in leukocyte–
cysts (top), finally resulting in a pseudohole formation endothelial interaction that, via reduction of tight junction
(bottom). protein expression and induction of apoptosis, results
Fig. 28.3 (A) In general, water outflow through vessels in vascular leakage. MCP-1, monocyte chemoattractant
is possible via three major routes: paracellular via protein; IL-8, interleukin-8; MIB-1, macrophage-inhibitory
dysfunction of tight junctions, transcellular via increased factor; TNF-α, tumor necrosis factor-alpha; ZO-1, zona
transport, e.g., mediated via growth factors, and finally occludens-1.
directly via endothelial gaps after cell death. Fig. 28.7 Vascular endothelial growth factor (VEGF) is the key
Fig. 28.4 Intercellular junctions in endothelial cells. Endothelial cells mediator of vascular damage and finally proliferation in
are connected and communicate with each other by tight the diabetic retina. AGE, advanced glycation end-
junctions and adherens junctions. Tight junctions products; IGF, insulin-like growth factor.
resemble a major part of the inner blood–retinal barrier. Fig. 28.11 (B) Membrane-bound carbonic anhydrase (CA IV) 55
They are built by different proteins, including occludin, kDa. NSAIDs, nonsteroidal anti-inflammatory drugs.
ZO-1, and the claudin family. (Adapted with permission from Wolfensberger TJ, Mahieu
I, Jarvis-Evans J, et al. Membrane-bound carbonic synthase; NO, nitric oxide; ICAM-1, intercellular adhesion
anhydrase in human retinal pigment epithelium. Invest molecule 1; MEK, miogen-activated protein kinase.
602 Ophtholmol Vis Sci 1994;35:3401–7.) Fig. 28.13 (A) Action of steroids on arachidonic acid.
Fig. 28.12 Nonsteroidal anti-inflammatory drugs (NSAIDs). Interaction (B) Action of steroids on growth factor release. COX,
of NSAID with vascular endothelial growth factor (VEGF) cyclooxygenase; NSAIDs, nonsteroidal anti-inflammatory
and inflammatory cytokines and their signaling pathways. drugs; PG, prostaglandin; TX, thromboxane; LT,
Section 2

COX-1, cyclooxygenase 1; PGs, prostaglandins; TXs, leukotriene; VEGF, vascular endothelial growth factor;
thromboxanes; TNF-α, tumor necrosis factor-α; NF-κB, PDGF, platelet-derived growth factor; PAF, platelet-
nuclear factor-kappa B; eNOS, endothelial nitric oxide activating factor; BRB, blood–retinal barrier.

REFERENCES 28. Antonelli-Orlidge A, Saunders KB, Smith SR, et al. An activated form of
1. Tranos PG, Wickremasinghe SS, Stangos NT, et al. Macular edema. Surv
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

transforming growth factor beta is produced by cocultures of endothelial cells


Ophthalmol 2004;49:470–90. and pericytes. Proc Natl Acad Sci U S A 1989;86:4544–8.
2. Stefansson E. Physiology of vitreous surgery. Graefes Arch Clin Exp 29. Ishii H, Jirousek MR, Koya D, et al. Amelioration of vascular dysfunction in
Ophthalmol 2009;247:147–63. diabetic rats by an oral PKC beta inhibitor. Science 1996;272:728–31
3. Kern TS, Tang J, Berkowitz BA. Validation of structural and functional lesions 30. Whiteside C, Dlugosz J. Mesangial cell protein kinase C isozyme activation in
of diabetic retinopathy in mice. Mol Vis 2010;16:2121–31. diabetic milieu. Am J Physiol Renal Physiol 2002; 282:F975–80.
4. Antcliff RJ, Marshall J. The pathogenesis of edema in diabetic maculopathy. 31. Orlidge A, Hollis TM. Aortic endothelial and smooth muscle histamine
Semin Ophthalmol 1999;14:223–32. metabolism in experimental diabetes. Atherosclerosis 1982;2:142–50.
5. Xu HZ, Le YZ. Significance of outer blood–retina barrier breakdown in 32. Gilbert RE, Kelly DJ, Cox AJ, et al. Angiotensin converting enzyme inhibi­
diabetes and ischemia. Invest Ophthalmol Vis Sci 2011;52:2160–4. tion reduces retinal overexpression of vascular endothelial growth factor
6. Schulze C, Firth JA. Immunohistochemical localization of adherens junction and hyperpermiability in experimental diabetes. Diabetologia 2000;43:
components in blood–brain barrier microvessels of the rat. J Cell Sci 1360–7.
1993;104:773–82. 33. Gill DS, Barradas MA, Fonseca VA, et al. Plasma histamine concentrations are
7. Klaassen I, Hughes JM, Vogels IM, et al. Altered expression of genes related elevated in patients with diabetes mellitus and peripheral vascular disease.
to blood–retina barrier disruption in streptozotocin-induced diabetes. Metabolism 1989;38:243–7.
Exp Eye Res 2009;89:4–15. 34. Gardner T. Histamine, ZO-1 and increased blood retinal permeability in dia­
8. Aveleira CA, Lin CM, Abcouwer SF, et al. TNF-α signals through PKCζ/ betic retinopathy. Trans Am Ophthalmol Soc 1995;93:583–621.
NF-κB to alter the tight junction complex and increase retinal endothelial cell 35. Aiello LP, Bursell S-E, Clermont A, et al. Vascular endothelial growth factor-
permeability. Diabetes 2010;59:2872–82. induced retinal permeability is mediated by protein kinase C in vivo and
9. Miyamoto K, Khosrof S, Bursell S-E, et al. Prevention of leukostasis and vas­ suppressed by an orally effective beta isoform selective inhibitor. Diabetes
cular leakage in streptozotocin-induced diabetic retinopathy via intercellular 1997;46:1473–80.
adhesion molecule-1 inhibition. Proc Natl Acad Sci U S A 1999; 96:10836–41. 36. Vlassara H. Recent progress in advanced glycation end products and diabetic
10. Schröder S, Palinski W, Schmidt-Schönbein GW. Activated monocytes and complications. Diabetes 1997;46:S19–25.
granulocytes, capillary nonperfusion, and neovascularization in diabetic 37. Lu M, Kuroki M, Amano S, et al. Advanced glycation end products increase
retinopathy. Am J Pathol 1991;139:81–100. retinal vascular endothelial growth factor expression. J Clin Invest 1998;101:
11. Joussen AM, Murata T, Tsujikawa A, et al. Leukocyte-mediated endothelial 1219–24.
cell injury and death in the diabetic retina. Am J Pathol 2001; 158:147–52. 38. Donati G, Kapetanios A, Dubois-Dauphin M, et al. Caspase-related apoptosis
12. Joussen AM, Poulaki V, Le ML, et al. A central role for inflammation in the in chronic ischaemic microangiopathy following experimental vein occlusion
pathogenesis of diabetic retinopathy. FASEB J 2004;18:1450–2. in mini-pigs. Acta Ophthalmol 2008;86:302–6.
13. Joussen AM, Poulaki V, Mitsiades N, et al. Potential use of nonsteroidal anti- 39. Cardier JE, Schulte T, Kammer H, et al. Fas (CD95-Apo-1) antigen expression
inflammatory drugs for prevention of diabetic vascular changes: aspirin pre­ and function in nurine liver endothelial cells: implications for the regulation
vents diabetic leakage and leukocyte adhesion through inhibition of TNF-α. of apoptosis in liver endothelial cells. FASEB J 1999;13:1950–60.
FASEB J 2002;16:438–40. 40. Walsh K, Sata M. Is extravasation a Fas-regulated process? Mol Med Today
14. Joussen AM, Doehmen S, Le ML, et al. TNF-alpha mediated apoptosis 1999;5:61–7.
plays an important role in the development of early diabetic retinopathy and 41. Joussen AM, Poulaki V, Mitsiades N, et al. Suppression of Fas-FasL-induced
long-term histopathological alterations. Mol Vis 2009;15:1418–28. endothelial cell apoptosis prevents diabetic blood–retinal barrier breakdown
15. Kociok N, Radetzky S, Krohne TU, et al. Pathological but not physiological in a model of streptozotocin-induced diabetes. FASEB J 2003;17:76–8.
retinal neovascularization is altered in TNF-Rp55-receptor-deficient mice. 42. Matrisian LM. The matrix-degrading metalloproteinases. Bio Assays
Invest Ophthalmol Vis Sci 2006;47:5057–65. 1992;14:455–63.
16. Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal microvascular 43. Das A, McGuire PG, Eriqat C, et al. Human diabetic neovascular membranes
cells in human and experimental diabetic retinopathy. J Clin Invest 1996;97: contain high levels of urokinase and metalloproteinase enzymes. Invest
2883–90. Ophthalmol Vis Sci 1999;40:809–13.
17. Joussen AM, Qin W, Poulaki V, et al. Endogenous VEGF induces retinal 44. Salzmann J, Limb GA, Khaw PT, et al. Matrix metalloproteinases and their
ICAM-1 and eNOS expression and initiates early diabetic retinal leukostasis. natural inhibitors in fibrovascular membranes of proliferative diabetic
Am J Pathol 2002;160:501–9. retinopathy. Br J Ophthalmol 2000;84:1091–6.
18. Senger DR, Van De Water L, Brown LF, et al. Vascular permeability factor 45. Grant MB, Caballero S, Tarnuzzer RT, et al. Matrix metalloproteinases expres­
(VPF, VEGF) in tumor biology. Cancer Metastas Rev 1993;12:303–24. sion in human retinal microvascular cells. Diabetes 1998;47:1311–7.
19. Keck PJ, Hauser SD, Krivi G, et al. Vascular permeability factor, an endothelial 46. Fernandez-Patron C, Zouki C, Whittal R, et al. Matrix metalloprotein­
cell mitogen related to PDGF. Science 1989; 246:1309–12. ases regulate neutrophil-endothelial cell adhesion through generation of
20. Ferrara N, Houck K, Jakeman L, et al. Molecular and biological properties of endothelin-1. FASEB J 2001:15:2230–40.
the vascular endothelial growth factor family of proteins. Endocr Rev 1992; 47. Behzadian MA, Wang XL, Windsor LJ, et al. TGF-beta increases retinal
13: 18–32 endothelial cell permeability by increasing MMP-9: possible role of glial
21. Engerman RL, Kern TS. Retinopathy and tissue hexose in drug-treated cells in endothelial barrier function. Invest Ophthalmol Vis Sci 2001;42:
animals. Arch Ophthalmol 1998;116:543–4. 853–9.
22. Amin RH, Frank RN, Kennedy A, et al. Vascular endothelial growth factor is 48. Abrass CK. Measurement of the rates of basal pinocytosis of horseradish
present in glial cells of the retina and optic nerve of human subjects with non­ peroxidase and internalization of heat-aggregated IgG by macrophages from
proliferative diabetic retinopathy. Invest Ophthalmol Vis Sci 1999;38:36–47. normal and streptozotocin-induced diabetic rats. Immunology 1998;65:
23. Qaum T, Xu Q, Joussen AM, et al. Early diabetic blood–retinal barrier break­ 411–5.
down is VEGF-dependent. Invest Ophthalmol Vis Sci 2001;42:2408–13. 49. Fitzgerald ME, Caldwell RB. The retinal microvasculature of spontaneously
24. Antonetti DA, Barber AJ, Khin S, et al. Vascular permeability in experimental diabetic BB rats: structure and luminal surface properties. Microvasc Res
diabetes is associated with reduced endothelial occludin content: vascular 1990;39:15–27.
endothelial growth factor decreases occludin in retinal endothelial cells. Penn 50. Hofman P, Blauwegers HG, Tolentino MJ, et al. VEGF-A induced hyperper­
State Retina Research Group. Diabetes 1998;47:1953–9. meability of blood–retinal barrier endothelium in vivo is predominantly asso­
25. DeMaio L, Antonetti DA, Scaduto RC Jr, et al. VEGF increases paracellular ciated with pinocytotic vesicular transport and not with formation of
transport without altering the solvent-drag reflection coefficient. Microvasc fenestrations. Vascular endothelial growth factor-A. Curr Eye Res 2000;21:
Res 2004;68:295–302. 637–45.
26. Harhaj NS, Felinski EA, Wolpert EB, et al. VEGF activation of protein kinase 51. Gardner TW, Abcouwer SF, Barber AJ, et al. An integrated approach to dia­
C stimulates occludin phosphorylation and contributes to endothelial perme­ betic retinopathy research. Arch Ophthalmol 2011;129:230–5.
ability. Invest Ophthalmol Vis Sci 2006;47:5106–15. 52. Antonetti DA, Barber AJ, Bronson SK, et al. Diabetic retinopathy: seeing
27. Wang J, Xu X, Elliott MH, et al. Müller cell-derived VEGF is essential for beyond glucose-induced microvascular disease. Diabetes 2006;55:2401–11.
diabetes-induced retinal inflammation and vascular leakage. Diabetes 2010; 53. Masland RH. The fundamental plan of the retina. Nat Neurosci 2001;
59:2297–305. 4:877–86.
54. Müther PS, Semkova I, Schmidt K, et al. Conditions of retinal glial and inflam­ 88. Marmor MF, Maak T. Enhancement of retinal adhesion and subretinal fluid
matory cell activation after irradiation in a GFP-chimeric mouse model. Invest absorbtion by acetazolamide. Invest Ophthalmol Vis Sci 1982;23:121–4.
Ophthalmol Vis Sci 2010;51:4831–9. 89. Wolfensberger TJ, Dmitriev AV, Govardovskii VI. Inhibition of mem­
55. Lewis H, Abrams GW, Blumenkranz MS, et al. Vitrectomy for diabetic branebound carbonic anhydrase decreases subretinal pH and volume. 603
macular traction and edema associated with posterior hyaloidal traction. Doc Ophthalmol 1999;97:261–71.
Ophthalmology 1992;99:753–9. 90. Miller SS, Steinberg RH. Active transport of ions across frog retinal pigment
56. Harbour JW, Smiddy WE, Flynn HW Jr, et al. Vitrectomy for diabetic macular epithelium. Exp Eye Res 1977;25:235.

Chapter 28
edema associated with a thickened and taut posterior hyaloid membrane. 91. Wolfensberger TJ, Gregor ZJ. Macular edema – rationale for therapy.
Am J Ophthalmol 1996;121:405–413 Dev Ophthalmol 2010;47:49–58.
57. Tachi N, Ogino N. Vitrectomy for diffuse macular edema in cases of diabetic 92. Yamamoto F, Steinberg RH. Effects of intravenous acetazolamide on retinal
retinopathy. Am J Ophthalmol 1996;8:258–60. pH in the cat. Exp Eye Res 1992;54:711– 8.
58. Tagawa H, McMeel JW, Furukawa H et al. Role of the vitreous in diabetic 93. Wolfensberger TJ, Chiang RK, Takeuchi A, et al. Inhibition of membrane-
retinopathy. I. Vitreous changes in diabetic retinopathy and in physiologic bound carbonic anhydrase enhances subretinal fluid absorption and retinal
aging. Ophthalmology 1986;93:596–601. adhesiveness. Graefes Arch Clin Exp Ophthalmol 2000;238:76–80.
59. Lopes de Faria JM, Jalkh AE, Trempe CL, et al. Diabetic macular edema: risk 94. Marmor MF, Negi A. Pharmacologic modification of subretinal fluid absorp­
factors and concomitants. Acta Ophthalmol Scand 1999;77:170–5. tion in the rabbit eye. Arch Ophthalmol 1986;104:1674–7.
60. Bron AJ, Tripathi RC, Tripathi BJ. The inner limiting membrane. In: Wolff’s 95. Tsuboi S, Pederson JE. Experimental retinal detachment. 10. Effect of acetazol­

Mechanisms of Macular Edema and Therapeutic Approaches


Anatomy of the eye. London: Chapman & Hall; 1997. Ch. 14, p. 488. amide on vitreous fluorescein disappearance. Arch Ophthalmol 1985;103:
61. Hogan MJ, Alvaroda JA, Weddell JE. Retina. In: Histology of the human eye. 1557–8.
Philadelphia: W.B. Saunders; 1971. p. 393–522. 96. Weene LE. Cystoid macular edema after scleral buckling responsive to acet­
62. Sebag J. Age-related differences in the human vitreoretinal interface. azolamide. Ann Ophthalmol 1992;24:423–4.
Arch Ophthalmol 1991;109:966–71. 97. Fishman GA, Gilbert LD, Anderson RJ, et al. Effects of methazolamidine on
63. Rhodes RH. A light microscopic study of the developing human neural retina. chronic macular edema in patients with retinitis pigmentosa. Ophthalmology
Am J Anat 1979;154:195–209. 1994;101:687–93.
64. Spira AW, Hollenberg MJ. Human retinal development: ultrastructure of the 98. Colin J. The role of NSAIDs in the management of postoperative ophthalmic
inner retinal layers. Dev Biol 1973;31:1–21. inflammation. Drugs 2007;67:1291–308.
65. Sarthy V. Collagen IV mRNA expression during development of the mouse 99. Bialek S, Quong JN, Yu K, et al. Nonsteroidal anti-inflammatory drugs alter
retina: an in situ hybridization study. Invest Ophthalmol Vis Sci 1993;34: chloride and fluid transport in bovine retinal pigment epithelium. Am J
145–52. Physiol 1996;270:C1175–89.
66. Halfter W, Dong S, Schurer B, et al. Embryonic synthesis of the inner limit­ 100. Wolfensberger TJ, Herbort CP. Treatment of cystoid macular edema with
ing membrane and vitreous body. Invest Ophthalmol Vis Sci 2005;46: non-steroidal anti-inflammatory drugs and corticosteroids. Doc Ophthalmol
2202–9. 1999;97:381–6.
67. Jumper JM, Embabi SN, Toth CA, et al. Electron immunocytochemical analy­ 101. Flach AJ, Stegman RC, Graham J, et al. Prophylaxis of aphakic cystoid macular
sis of posterior hyaloid associated with diabetic macular edema. Retina edema without corticosteroids. A paired-comparison, placebo-controlled
2000;20:63–8. double-masked study. Ophthalmology 1990;97:1253–8.
68. Blankenship GW. Diabetic macular edema and argon laser photocoagulation: 102. Almeida DR, Johnson D, Hollands H, et al. Effect of prophylactic nonsteroidal
a prospective randomized study. Ophthalmology 1979;86:69–76. antiinflammatory drugs on cystoid macular edema assessed using optical
69. Early Treatment Diabetic Retinopathy Study research group. Photocoagula­ coherence tomography quantification of total macular volume after cataract
tion for diabetic macular edema. Early Treatment Diabetic Retinopathy Study surgery. J Cataract Refract Surg 2008;34:64–9.
report number 1. Arch Ophthalmol 1985;103:1796–1806. 103. DeCroos FC, Afshari NA. Perioperative antibiotics and anti-inflammatory
70. Early Treatment Diabetic Retinopathy Study research group. Treatment tech­ agents in cataract surgery. Curr Opin Ophthalmol 2008;19:22–26.
niques and clinical guidelines for photocoagulation of diabetic macular 104. Nelson ML, Martidis A. Managing cystoid macular edema after cataract
edema. Early Treatment Diabetic Retinopathy Study report number 2. Oph­ surgery. Curr Opin Ophthalmol 2003;14: 39–43.
thalmology 1987;94:761–74. 105. Sivaprasad S, Bunce C, Patel N. Non-steroidal anti-inflammatory agents
71. Early Treatment Diabetic Retinopathy Study research group. Techniques for for treating cystoid macular oedema following cataract surgery. Cochrane
scatter and local photocoagulation treatment of diabetic retinopathy: Early Database Syst Rev 2005;1:CD004239.
Treatment Diabetic Retinopathy Study report no. 3. Int Ophthalmol Clin 106. Flach AJ, Dolan BJ, Irvine AR. Effectiveness of ketorolac tromethamine 0.5%
1987;27:254–64. ophthalmic solution for chronic aphakic and pseudophakic cystoid macular
72. Early Treatment Diabetic Retinopathy Study research group. Photocoagula­ edema. Am J Ophthalmol 1987;103:479–86.
tion for diabetic macular edema: Early Treatment Diabetic Retinopathy Study 107. Flach AJ, Jampol LM, Weinberg D, et al. Improvement in visual acuity in
report no. 4. Int Ophthalmol Clin 1987;27:265–72. chronic aphakic and pseudophakic cystoid macular edema after treatment
73. Early Treatment Diabetic Retinopathy Study research group. Early photoco­ with topical 0.5% ketorolac tromethamine. Am J Ophthalmol 1991;112:514–9.
agulation for diabetic retinopathy. ETDRS report number 9. Ophthalmology 108. Rossetti L, Chaudhuri J, Dickersin K. Medical prophylaxis and treatment of
1991;98:766–85. cystoid macular edema after cataract surgery. The results of a meta-analysis.
74. Weiter JJ, Zuckerman R. The influence of the photoreceptor–RPE complex on Ophthalmology 1998;105:397–405.
the inner retina. An explanation for the beneficial effects of photocoagulation. 109. Nehme A, Edelman J. Dexamethasone inhibits high glucose-, TNF-α-, and
Ophthalmology 1980;87:1133–9. IL-1β-induced secretion of inflammatory and angiogenic mediators from
75. Bresnick GH. Diabetic maculopathy. A critical review highlighting diffuse retinal microvascular pericytes. Invest Ophthalmol Vis Sci 2008;49:2030–8.
macular edema. Ophthalmology 1983; 90:1301–17. 110. Romero IA, Radewicz K, Jubin E, et al. Changes in cytoskeletal and tight
76. Molnar I, Poitry S, Tsacopoulos M, et al. Effect of laser photocoagulation on junctional proteins correlate with decreased permeability induced by dexa­
oxygenation of the retina in miniature pigs. Invest Ophthalmol Vis Sci methasone in cultured rat brain endothelial cells. Neurosci Lett 2003;334:
1985;26:1410–4. 112–6.
77. Perry DD, Risco JM. Choroidal microvascular repair after argon photocoagu­ 111. Antonetti DA, Wolpert EB, DeMaio L, et al. Hydrocortisone decreases retinal
lation. Am J Ophthalmol 1982;93:787–93. endothelial cell water and solute flux coincident with increased content and
78. Gottfredsdottir MS, Stefansson E, Jonasson F, et al. Retinal vasoconstriction decreased phosphorylation of occludin. J Neurochem 2002;80:667–77.
after laser treatment for diabetic macular edema. Am J Ophthalmol 112. Edelman JL, Lutz D, Castro MR. Corticosteroids inhibit VEGF-induced vas­
1993;115:64–7. cular leakage in a rabbit model of blood–retinal and blood–aqueous barrier
79. Wallow IH, Sponsel WE, Stevens TS. Clinicopathologic correlation of diode breakdown. Exp Eye Res 2005;80:249–58.
laser burns in monkeys. Arch Ophthalmol 1991;109:648–53. 113. Wang K, Wang Y, Gao L, et al. Dexamethasone inhibits leukocyte accumula­
80. Glaser BM, Campochiaro PA, Davis JL Jr, et al. Retinal pigment epithelial cells tion and vascular permeability in retina of streptozotocin-induced diabetic
release inhibitors of neovascularization. Ophthalmology 1987;94:780–4. rats via reducing vascular endothelial growth factor and intercellular
81. Boulton ME, Xiao M, Khaki A. Changes in growth factor expression in pig adhesion molecule-1 expression. Biol Pharm Bull 2008;31:1541–6.
eyes following scatter laser photocoagulation. Invest Ophthalmol Vis Sci 114. Zhang X, Bao S, Lai D, et al. Intravitreal triamcinolone acetonide inhibits
1995;36(suppl.):95. breakdown of the blood–retinal barrier through differential regulation of
82. Wilson D, Finkelstein D, Quingley H, et al. Macular grid photocoagulation: VEGF-A and its receptors in early diabetic rat retinas. Diabetes 2008;57:
an experimental animal study on the primate retina. Arch Ophthalmol 1026–33.
1988;106:100–5. 115. Nauck M, Roth M, Tamm M, et al. Induction of vascular endothelial growth
83. Gardner TW, Gabbay RA. Diabetes and obesity: a challenge for every factor by platelet-activating factor and platelet-derived growth factor is down­
ophthalmologist. Arch Ophthalmol 2009;127:328–9. regulated by corticosteroids. Am J Respir Cell Mol Biol 1997;16:398–406.
84. Liew G, Mitchell P, Wong TY. Systemic management of diabetic retinopathy. 116. Ebrahem Q, Minamoto A, Hoppe G, et al. Triamcinolone acetonide inhibits
Br Med J 2009;338:b441. IL-6- and VEGF-induced angiogenesis downstream of the IL-6 and VEGF
85. Pfoff DS, Thom SR. Preliminary report on the effect of hyperbaric oxygen on receptors. Invest Ophthalmol Vis Sci 2006;47:4935–41.
cystoid macular edema. J Cataract Refract Surg 1987;13:136–40. 117. Leal EC, Manivannan A, Hosoya K, et al. Inducible nitric oxide synthase
86. Widder RA, Brunner R, Walter P, et al. Improvement of visual acuity in isoform is a key mediator of leukostasis and blood-retinal barrier breakdown
patients suffering from diabetic retinopathy after membrane differential in diabetic retinopathy. Invest Ophthalmol Vis Sci 2007;48:5257–65.
filtration: a pilot study. Transfus Sci 1999;21:201–6. 118. Mizuno S, Nishiwaki A, Morita H, et al. Effects of periocular administration
87. Cox SN, Hay E, Bird AC. Treatment of chronic macular edema with acetazol­ of triamcinolone acetonide on leukocyte–endothelium interactions in the
amide. Arch Ophthalmol 1988;106:1190–5. ischemic retina. Invest Ophthalmol Vis Sci 2007;48:2831–6.
119. Reichenbach A, Wurm A, Pannicke T, et al. Muller cells as players in retinal 137. Starita C, Patel M, Katz B, et al. Vascular endothelial growth factor and the
degeneration and edema. Graefes Arch Clin Exp Ophthalmol 2007;245: potential therapeutic use of pegaptanib (macugen) in diabetic retinopathy.
627–36. Dev Ophthalmol 2007;39:122–48.
604 120. Haynes RC Jr, Murad F. Adrenocorticotropic hormone: adrenocortical ste­ 138. van Kooij B, Rothova A, Rijkers GT, et al. Distinct cytokine and chemokine
roids and their synthetic analogs: inhibitors of adrenocortical steroid biosyn­ profiles in the aqueous of patients with uveitis and cystoid macular edema.
thesis. In: Gilman AG, Goodman LS, Rall TW, et al., editors. Goodman and Am J Ophthalmol 2006;142:192–4.
Gilman’s The pharmacological basis of therapeutics. New York: Macmillan; 139. Deuter CM, Koetter I, Guenaydin I, et al. Interferon alfa-2a: a new treatment
1985. p. 1459–89. option for long lasting refractory cystoid macular edema in uveitis? A pilot
Section 2

121. Thach AB, Dugel PU, Flindall RJ, et al. A comparison of retrobulbar versus study. Retina 2006;26:786–91.
subtenon’s corticosteroid therapy for cystoid macular edema refractory to 140. Becker MD, Heiligenhaus A, Hudde T, et al. Interferon as a treatment
topical medications. Ophthalmology 1997;104:2003–8. for uveitis associated with multiple sclerosis. Br J Ophthalmol 2005;89:
122. Freeman G. Cystoid macular oedema in uveitis: an unsolved problem. Eye 1254–7.
2001;15:12–7. 141. Nussenblatt RB, Palestine AG, Chan CC, et al. Randomized, double-masked
123. Jonas, JB, Sofker A. Intraocular injection of crystalline cortisone as adjunctive study of cyclosporine compared to prednisolone in the treatment of endoge­
treatment of diabetic macular edema. Am J Ophthalmol 2001;132:425–7. nous uveitis. Am J Ophthalmol 1991;112:138–46.
124. Martidis A, Duker JS, Greenberg PB, et al. Intravitreal triamcinolone for 142. Theodossiadis PG, Markomichelakis NN, Sfikakis PP. Tumor necrosis factor
Basic Mechanisms of Injury in the Retina

refractory diabetic macular edema. Ophthalmology 2002;109:920–7. antagonists: preliminary evidence for an emerging approach in the treatment
Basic Science and Translation to Therapy

125. Diabetic Retinopathy Clinical Research Network. A randomized trial compar­ of ocular inflammation. Retina 2007;27:399–413.
ing intravitreal triamcinolone acetonide and focal/grid photocoagulation for 143. Rothova A. Inflammatory cystoid macular edema. Curr Opin Ophthalmol
diabetic macular edema. Ophthalmology 2008 ;115:1447–9, 1449. 2007;18:487–92.
126. Jonas JB, Kreissig I, Degenring RF. Intravitreal triamcinolone acetonide as 144. Kafkala C, Choi JY, Choopong P, et al. Octreotide as a treatment for uveitic
treatment of macular edema in central retinal vein occlusion. Graefes Arch cystoid macular edema. Arch Ophthalmol 2006;124:1353–5.
Clin Exp Ophthalmol 2002;240:782–3. 145. Fung WE. Vitrectomy for chronic aphakic cystoid macular edema.
127. Jonas JB, Kreissig I, Sofker A, et al. Intravitreal injection of triamcinolone for Results of a national, collaborative, prospective, randomized investigation.
diffuse diabetic macular edema. Arch Ophthalmol 2003;121:57–61. Ophthalmology 1985;92:1102–11.
128. Jaffe GJ, Yang CH, Guo H, et al. Safety and pharmacokinetics of an intraocular 146. Margherio RR, Trese MT, Margherio AR, et al. Surgical management of
fluocinolone acetonide sustained delivery device. Invest Ophthalmol Vis Sci vitreomacular traction syndromes. Ophthalmology 1989;96:1437–1445.
2000;41:3569–75. 147. Gandorfer A, Messmer EM, Ulbig MW, et al. Resolution of diabetic macular
129. Jaffe GJ, Ben-Nun J, Guo H, et al. Fluocinolone acetonide sustained drug edema after surgical removal of the posterior hyaloid and the inner limiting
delivery device to treat severe uveitis. Ophthalmology 2000;107:2024–33. membrane. Retina 2000;20:126–33.
130. Haller JA, Bandello F, Belfort R Jr, et al. Randomized, sham-controlled trial of 148. Stefansson E, Novack RL, Hatchell DL. Vitrectomy prevents retinal hypoxia
dexamethasone intravitreal implant in patients with macular edema due to in branch retinal vein occlusion. Invest Ophthalmol Vis Sci 1990;31:284–9.
retinal vein occlusion. Ophthalmology 2010;117:1134–46.e3. 149. Holekamp NM, Shui YB, Beebe DC. Vitrectomy surgery increases oxygen
131. Lowder C, Belfort R Jr, Lightman S, et al. Dexamethasone intravitreal implant exposure to the lens: a possible mechanism for nuclear cataract formation.
for noninfectious intermediate or posterior uveitis. Arch Ophthalmol Am J Ophthalmol 2005;139:302–10.
2011;129:545–53. 150. Giblin FJ, Quiram PA, Leverenz VR, et al. Enzyme induced posterior vitreous
132. Boyer DS, Faber D, Gupta S, et al. Dexametasone intravitreal implant for detachment in the rat produces increased lens nuclear pO2 levels. Exp Eye Res
treatment of diabetic macular edema in vitrectomized patients. Retina 2011; 2009;88:286–92.
31:915–23 151. Noma H, Funatsu H, Mimura T, et al. Vitreous levels of interleukin-6 and
133. Cordero Coma M, Sobrin L, Onal S, et al. Intravitreal bevacizumab for vascular endothelial growth factor in macular edema with central retinal vein
treatment of uveitic macular edema. Ophthalmology 2007;114: 1574–9. occlusion. Ophthalmology 2009;116:87–93.
134. Mason JO 3rd, Albert MA Jr, Vail R. Intravitreal bevacizumab (Avastin) for 152. Praidou A, Klangas I, Papakonstantinou E, et al. Vitreous and serum
refractory pseudophakic cystoid macular edema. Retina 2006;26:356–7. levels of platelet-derived growth factor and their correlation in patients with
135. Spaide RF, Chang LK, Klancnik JM, et al. Prospective study of intravitreal proliferative diabetic retinopathy. Curr Eye Res 2009;34:152–61.
ranibizumab as a treatment for decreased visual acuity secondary to central 153. Stefansson E. The therapeutic effects of retinal laser treatment and vitrectomy.
retinal vein occlusion. Am J Ophthalmol 2009;147:298–306. A theory based on oxygen and vascular physiology. Acta Ophthalmol Scand
136. Arimura N, Otsuka H, Yamakiri K, et al. Vitreous mediators after intravitreal 2001;79:435–40.
bevacizumab or triamcinolone acetonide in eyes with proliferative diabetic 154. Stefansson E. Ocular oxygenation and the treatment of diabetic retinopathy.
retinopathy. Ophthalmology 2009;116:921–6. Surv Ophthalmol 2006;4:364–80.
Basic Mechanisms of Injury in the Retina Section 2

Cellular Effects of Detachment and Reattachment Chapter

on the Neural Retina and the Retinal


Pigment Epithelium 29 
Louisa Wickham, Geoffrey P. Lewis, David G. Charteris, Steven K. Fisher

INTRODUCTION no intraretinal vasculature, with the inner retina being supplied


by vessels that lie on the vitreal surface. The rabbit retina has
In retinal detachment the separation of the neural retinal from proved to be a more difficult animal model in long-term experi-
the retinal pigment epithelium (RPE) initiates a complex series ments because the retina tends to degenerate very rapidly fol-
of cellular and molecular changes.1 Left untreated, retinal detach- lowing detachment; for short-term studies, however, (i.e., 3–7
ment results in permanent visual loss; however, early interven- days) rabbits continue to be a very useful model.
tion may be associated with good visual outcomes, suggesting Ideally the characteristics of an experimental detachment
that some of these molecular changes may be arrested or should closely mimic those found in humans while allowing for
reversed.2,3 precise control over the extent of separation between the two
By studying the cellular and molecular changes that occur layers (detachment height), the location of the detachment, its
after detachment and/or reattachment clinicians may gain a surface area, and the onset of detachment (or reattachment). A
more precise understanding of the degenerative processes number of methods have been used to simulate human retinal
within the retina that lead to visual impairment and the mecha- detachment in animal models. These range from creating large
nisms underlying the serious complications of detachment, such retinal tears to subretinal injections of fluid or viscous sub-
as proliferative vitreoretinopathy (PVR). In addition, these stances. Experiments where retinal detachment induction is
insights may aid the development of future treatment strategies standardized with a micropipette provide a very controlled
and adjunctive therapies aimed at improving visual outcomes. environment for analysis; however they differ from the clinical
This chapter reviews the many changes that occur in retinal pattern of events in which acute retinal tears of variable size are
cells following retinal detachment and the ensuing process of induced by vitreoretinal traction at the time of posterior vitreous
morphologic recovery following reattachment, as revealed by detachment. It is possible that retinal tearing may act as a more
human case series and studies of experimental retinal detach- potent stimulus for cellular disorganization, loss, and remodel-
ment and reattachment in animal models. ing, leading more rapidly to the advanced pathology usually
seen following longer periods of retinal detachment in animal
USE AND LIMITATIONS OF ANIMAL models. Although experiments involving animal models may
MODELS IN THE STUDY OF differ in methodology, species used, and outcome measures,
RETINAL DETACHMENT they have yielded similar results to give a relatively detailed
profile of the changes that occur after detachment. Retinal tissue
Human studies of cellular changes following acute retinal
removed from human postmortem specimens and from patients
detachment are limited to isolated case reports as surgical man-
undergoing retinal detachment surgery has demonstrated
agement does not routinely involve removal of retinal tissue.
changes similar to those seen in animal models.6–8
More recently data have become available from patients under-
Animal models have proved invaluable in providing insight
going macular translocation surgery in which the retina is
into the regenerative capacity of photoreceptor cells and the
detached as part of the procedure, allowing sampling of the
ability of reattachment to slow, stop, or reverse changes induced
retina as early as 1 hour following detachment.4 In patients with
by detachment. They also continue to provide opportunities to
advanced stages of retinal detachment and PVR, surgical man-
test adjunctive agents targeting neuroprotection and wound
agement may involve excising areas of scarred retinal tissue
healing before progressing to human surgical trials. Further, in
allowing histopathological analysis. However, the data from
mouse models the large variety of genetic mutations that exist
human studies is still limited by small numbers, the challenges
provides additional scope to study retinal detachment and
of sampling and analyzing small retinal specimens, and an
potential gene therapies.
inability to study cellular recovery following reattachment.4–7
This difficulty in obtaining retinal tissue from patients with CELLULAR CHANGES IN RESPONSE TO
retinal detachments has led to the use of animal models. Animal
models have been developed in a variety of mammalian species
RETINAL DETACHMENT
from rodents to primates, most commonly in rabbits and cats, Descriptions of the cellular response to retinal detachment are
and more recently in mice. The feline retina is rod-dominant, and frequently divided into those observed in the early stages of
has an intraretinal circulation that is excluded from the photo- retinal detachment and those observed in more chronic cases
receptor layer and a choroidal circulation that supplies the pho- and in PVR. There is, however, a continuous progression of
toreceptor layer. The rabbit retina is also rod-dominant but has pathological change.
Acute retinal detachment into the subretinal space, and the nucleus becomes displaced to
a more apical position10,14 (Fig. 29.2).
Acute retinal detachment, i.e., changes occurring in the first 3
606 days of detachment, has been extensively documented.
In the feline model, experiments using 3H-thymidine have
shown that within 72 hours of retinal detachment the RPE has
A rapid response to retinal detachment has been shown to
begun to proliferate and may be observed as areas of hyperplasia
occur within 15 minutes, including phosphorylation of fibroblast
within the RPE monolayer.10 This proliferative response trans-
Section 2

growth factor receptor (FGFR-1) and increased expression by


forms the RPE’s uniform monolayer into a heterogeneous mor-
RPE and Müller cells of extracellular signal-regulated kinase and
phology in which strands of cells extend from the original
activator protein transcription factor.9 This initiates a cascade of
monolayer into the subretinal space or result in the formation of
events that leads to a number of molecular and cellular changes
multiple layers of cells whose polarity does not necessarily
within the retina and RPE.
match that of the original monolayer (Fig. 29.3). This effect is
limited to the region of detachment; in attached regions the RPE
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

RPE–photoreceptor interface
remains mitotically quiet, suggesting that attachment of the RPE
The earliest structural effects of retinal detachment are seen
to the neural retina acts to keep the RPE mitotically inactive and
at the interface of photoreceptor outer segments and the RPE.10
its apical surface highly differentiated.15–17 The proliferative
The mature RPE is a polarized monolayer of neuroepithelial
response of the RPE cells also appears to be self-limiting with
cells that rests on Bruch’s membrane, between the choriocapil-
only low levels of proliferation observed after long detachment
laris and the neural retina.11 The relationship of the apical
intervals (e.g., 12–14 months) in owl, monkey, and cat retinas10,16
surface of the RPE to differentiated photoreceptors is anatomi-
(Box 29.1 and Fig. 29.4).
cally complex. There are no actual cellular junctions between
The subretinal space is usually free of cells; however within
the two layers in the mature eye, but the two are adherent,
24 hours of retinal detachment a number of cell types (polymor-
with the degree of adhesion varying among species.12 With
phonuclear neutrophils, monocytes, and macrophages) migrate
the onset of retinal detachment changes to this interface include
into this space from the choroidal and retinal capillaries.10,18 Free
alterations in the RPE apical surface, proliferation of RPE cells,
RPE cells are also seen in the subretinal space within 72 hours
migration of cells into the subretinal space, degeneration of
of retinal detachment and frequently contain outer-segment
photoreceptor outer segments, and changes in photoreceptor
fragments, indicating that they may play a role in phagocytosis
outer-segment renewal.1
of cellular debris.10,18
Within a few hours of retinal detachment, the long and elab­
orate sheet-like and villous processes that normally ensheath the Photoreceptors
outer segments are lost and replaced by a “fringe” of short Within 12 hours of experimental retinal detachment, photore-
microvilli (Fig. 29.1).13 At the same time, the overall surface mor- ceptor outer segments show evidence of structural damage. Ini-
phology of the RPE cells changes into a rounded contour, as tially, the distal end of the outer segment becomes vacuolated or
cytoplasm protrudes past the normal limits of the apical surface distorted and by 24–72 hours all rod and cone outer segments

MV

SRS

Fig. 29.1 Electron micrograph of the retinal pigment epithelium (RPE)


1 day after production of a retinal detachment. Compared with normal
RPE cells, the apical surface is mounded. The sheet-like apical Fig. 29.2 Scanning electron micrograph of the apical surface of the
projections that normally ensheath the outer segments have been retinal pigment epithelium 6 weeks after production of an experimental
replaced by a homogeneous fringe of short, microvillous processes detachment, demonstrating the pronounced mounding response of the
(MV). In this particular cell, the nucleus (N) is displaced into the epithelial cells. (Reproduced with permission from Anderson DH, Stern
mounded region. The cells’ lateral junctions are indicated by arrows. WH, Fisher SK, et al. Retinal detachment in the cat: The pigment
SRS, subretinal space. (Reproduced with permission from Anderson epithelial–photoreceptor interface. Invest Ophthalmol Vis Sci
DH, Stern WH, Fisher SK, et al. Retinal detachment in the cat: The 1983;24:910.)
pigment epithelial–photoreceptor interface. Invest Ophthalmol Vis Sci
1983;24:909.)
Fig. 29.3 Light micrograph of an area
of retinal pigment epithelium (RPE) cell
proliferation in a cat retina detached for
14 days and reattached for 30 days. Three 607
layers of RPE cells are present (L1-L3),
each displaying different surface polarity. The
apical surfaces of L1 and L2 face each other,

Chapter 29
as do the basal surfaces of L2 and L3. The
basal lamina of L2 is clearly evident (arrow).
IS Only outer-segment fragments (asterisk)
appear near the inner segment (IS)
tips (×800). ONL, outer nuclear layer.
(Reproduced with permission from Anderson
ONL DH, Guerin CJ, Erickson PA, et al.
Morphological recovery in the reattached

Cellular Effects of Detachment and Reattachment on the Neural Retina and the Retinal Pigment Epithelium
retina. Invest Ophthalmol Vis Sci
1986;27:174.)

persisting for just 1 week following retinal detachment, after


which their expression is downregulated.21
During the first day of a detachment the inner segments
appear essentially normal, but between the first and third days
they begin to show signs of degeneration: most commonly swell-
ing, disruption, and loss of mitochondria (and loss of anticyto-
chrome oxidase labeling) in the ellipsoid region,3,22 an overall
disruption of the organized rough endoplasmic reticulum and
Golgi apparatus in the myoid region, and, within a few days, an
overall size reduction of the inner segment. It is interesting to
note that the connecting cilium, which is essential for production
of the outer segment, is retained even in severely affected inner
segments in long-term detachments. This is crucial as its loss
would prevent regeneration of outer segments following reat-
tachment. Similarly, the loss of mitochondria also has the poten-
Fig. 29.4 Color fundus photograph of a chronic retinal detachment with
a pigmented demarcation line indicating the interface between tial to affect the photoreceptors’ ability to regenerate significantly,
attached and detached retina. because the metabolic rate in these cells is among the highest of
any in the body.
The outer nuclear layer contains the cell bodies of the photo-
Box 29.1 Clinical correlates
receptor cells. These cell bodies extend a process toward the
Retinal pigment epithelial (RPE) proliferation can be seen as outer plexiform layer, where they form synapses with second-
subretinal pigment deposition in chronic retinal detachments. It is order neurons. Rods and cones have characteristic synaptic ter-
likely that the demarcation lines noted in human retinal
detachments represent zones of proliferated RPE occurring at
minals called spherules and pedicles respectively.23 The outer
transitions between detached and attached regions of the eye plexiform layer also contains the processes of second-order
(Fig. 29.4). neurons, the cell bodies of which lie in the inner nuclear layer.
These processes synapse with each other and with the photore-
ceptors. The photoreceptor cell bodies and synaptic terminals
are significantly shorter and distorted with disoriented discs.19 show a rapid response to detachment with extensive vacuoliza-
The degeneration of outer segments may proceed until those in tion, degeneration of mitochondria, and disorganization of the
the zone of detachment appear only as empty sacs of membrane microtubules and actin filaments. Cell death via the apoptotic
attached to the connecting cilium.10 pathway peaks at day 3 following retinal detachment but con-
Outer-segment debris is shed into the subretinal space where tinues at low levels for as long as the retina is detached, a process
it is phagocytosed by RPE cells and macrophages which have that appears to be mediated via caspases 3, 7, and 9.24,25 Recent
migrated into the area.10,18 studies have shown that, when caspase pathways are blocked,
Although retinal detachment interrupts the process of disc receptor interacting protein (RIP) kinases promote necrosis and
production and shedding, outer-segment specific proteins con- overcome apoptosis inhibition. Therefore, targeting of both
tinue to be produced but localize to abnormal cellular locations. caspase and RIP kinase pathways is required for effective pho-
Opsin, normally concentrated in the outer segment, begins to toreceptor protection26 (Box 29.2).
accumulate in the plasma membrane vitread to the outer segment Following cell death some photoreceptors are extruded into
within a day following experimental retinal detachment (Fig. the subretinal space where they are phagocytosed by macro-
29.5).19 Peripherin/rds, another outer-segment protein specific to phages while others appear to undergo degeneration and phago-
the disc rims, is also redistributed and begins to appear in cyto- cytosis within the outer nuclear layer.28
plasmic vesicles.20 Cone outer-segment proteins appear to be Not all photoreceptors degenerate at the same rate; areas of
more susceptible to damage, with redistributed cone opsins extensive degeneration coexist with areas of relatively intact
Fig. 29.5 Laser scanning confocal images
of normal (A), 7-day (B), and 28-day (C) cat
retinas, as well as a human detached retina
608 OS (D), labeled with isolectin B4 (green), antiglial
fibrillary acidic protein (anti-GFAP) (blue),
and antirod opsin (red). In the normal retina,
isolectin B4 labels microglia in the inner
Section 2

plexiform layer (IPL) (and blood vessels),


anti-GFAP labels astrocytes and Müller cell
ONL endfeet, and antirod opsin labels rod outer
segments (OS) (A). Following 7 days of
detachment, isolectin B4 labeling illustrates
an overall increase in the number of
microglia and their presence in the outer
retina, rod opsin becomes mislocalized to
Basic Mechanisms of Injury in the Retina
Basic Science and Translation to Therapy

the photoreceptor cell bodies, and GFAP


increases in Müller cells (B). At 28 days,
INL numerous microglia can be observed
throughout the retina, photoreceptor
cell bodies are extruded out of the retina,
IPL and Müller cell processes extend into the
subretinal space (C). A similar pattern of
increased numbers of microglia, rod opsin
GCL mislocalization, and increased GFAP labeling
is observed in human detached retinas (D).
Note rod axons extending into the inner
A B retina in (D), a common phenomenon
in cat retinas following detachment and
reattachment. ONL, outer nuclear layer; INL,
inner nuclear layer; GCL, ganglion cell layer.

ONL

INL

IPL

GCL
C D

Box 29.2 Clinical correlates of detachment. These synaptic terminals are normally filled with
synaptic vesicles and contain one or two large presynaptic
More recently, photoreceptor apoptosis has also been ribbons. When the retina has been detached for 3 days, many of
demonstrated in human retinal specimens, with a peak at day 2
following retinal detachment.27 these terminals appear depleted of vesicles, except for a few that
remain as a halo around a greatly truncated ribbon.29 Many
terminals appear as if they have “retracted” into the cell body,
and some synaptic structures generally associated with the outer
plexiform layer now occur within the outer nuclear layer (Fig.
photoreceptors.19 It does appear that rod cell bodies appear to 29.6).29,30 As with the cone and rod photoreceptor cell bodies, the
degenerate quicker than cones following retinal detachment.21 In cone synaptic terminals seem to survive the early effect

You might also like