You are on page 1of 546

Advances in Experimental Medicine and Biology 1415

John D. Ash · Eric Pierce · Robert E. Anderson ·


Catherine Bowes Rickman · Joe G. Hollyfield ·
Christian Grimm Editors

Retinal
Degenerative
Diseases XIX
Mechanisms and Experimental Therapy
Advances in Experimental Medicine
and Biology

Volume 1415

Series Editors
Wim E. Crusio, Institut de Neurosciences Cognitives et Intégratives
d’Aquitaine, CNRS and University of Bordeaux, Pessac Cedex, France
Haidong Dong, Departments of Urology and Immunology, Mayo Clinic
Rochester, MN, USA
Heinfried H. Radeke, Institute of Pharmacology and Toxicology, Clinic of
the Goethe University Frankfurt Main, Frankfurt am Main, Hessen,
Germany
Nima Rezaei, Research Center for Immunodeficiencies, Children’s Medical
Center, Tehran University of Medical Sciences, Tehran, Iran
Ortrud Steinlein, Institute of Human Genetics, LMU University Hospital
Munich, Germany
Junjie Xiao, Cardiac Regeneration and Ageing Lab, Institute of
Cardiovascular Sciences, School of Life Science, Shanghai University
Shanghai, China
Advances in Experimental Medicine and Biology provides a platform for
scientific contributions in the main disciplines of the biomedicine and the life
sciences. This series publishes thematic volumes on contemporary research in
the areas of microbiology, immunology, neurosciences, biochemistry,
biomedical engineering, genetics, physiology, and cancer research. Covering
emerging topics and techniques in basic and clinical science, it brings together
clinicians and researchers from various fields.
Advances in Experimental Medicine and Biology has been publishing
exceptional works in the field for over 40 years, and is indexed in SCOPUS,
Medline (PubMed), EMBASE, BIOSIS, Reaxys, EMBiology, the Chemical
Abstracts Service (CAS), and Pathway Studio.
2021 Impact Factor: 3.650 (no longer indexed in SCIE as of 2022)
John D. Ash • Eric Pierce
Robert E. Anderson
Catherine Bowes Rickman
Joe G. Hollyfield • Christian Grimm
Editors

Retinal Degenerative
Diseases XIX
Mechanisms and Experimental
Therapy
Editors
John D. Ash Eric Pierce
Department of Ophthalmology Ocular Genomics Institute
University of Pittsburgh School Department of Ophthalmology
of Medicine Massachusetts Eye and Ear Infirmary
Pittsburgh, PA, USA Harvard Medical School
Boston, MA, USA
Robert E. Anderson
Health Sciences Center Catherine Bowes Rickman
University of Oklahoma Health Department of Ophthalmology
Sciences Center Duke Medical Center
Oklahoma City, OK, USA Durham, NC, USA

Joe G. Hollyfield Christian Grimm


Department of Ophthalmology Laboratory for Retinal Cell Biology
Cleveland Clinic Lerner College Department of Ophthalmology
of Medicine University Hospital Zurich
Cleveland, OH, USA University of Zurich
Schlieren, Switzerland

ISSN 0065-2598     ISSN 2214-8019 (electronic)


Advances in Experimental Medicine and Biology
ISBN 978-3-031-27680-4    ISBN 978-3-031-27681-1 (eBook)
https://doi.org/10.1007/978-3-031-27681-1

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher,
whether the whole or part of the material is concerned, specifically the rights of translation,
reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any
other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, expressed or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
The editors are pleased dedicate this publication to the memory
of our long-time friend and colleague, Alan M. Laties. Except
for the most recent years, Alan attended each of these biennial
retinal degeneration meetings since they began in 1984. Early
on Alan recognized the importance of our attempt to provide a
continuing international platform for discussions and scientific
exchange to take place among investigators focused on retinal
degeneration research. Through his scientific leadership at the
Foundation Fighting Blindness (formerly the Retinitis
Pigmentosa Foundation), we received the first meeting grant to
partially cover some of the expenses of the RD meeting held in
San Francisco in 1988. The Foundation has provided
continuing support for each of the subsequent meetings in the
form of travel grant support for young investigators.
Born in Beverly, Massachusetts, the son of Russian immigrants,
he attended Harvard College (BA, 1954) and completed
medical school at Baylor College of Medicine (MD, 1959),
followed by a residency in ophthalmology in the Hospital of the
University of Pennsylvania (1961–63). A United States Public
Health Service Special Research Fellowship supported his
vi 

research training in the Institute of Neurological Sciences at


the University of Pennsylvania (1963–64). He joined the faculty
at the University of Pennsylvania in 1965 where he moved
through the academic ranks until retiring as Emeritus
Professor of Ophthalmology at the Perelman School of
Medicine in 2020. He held joint appointments in
Ophthalmology and Neurology where he was the Irene Heinz
Given and John LaPorte Given Research Professor and the
Harold G. Scheie Research Professor in Ophthalmology. He
served as neuro-ophthalmologist at the Hospital of the
University of Pennsylvania while pursuing basic research on
the autonomic innervation of the eye, eye growth, and
therapeutic approaches to eye diseases. He has published 140
original research papers, 30 review articles, and presented
numerous invited lectures at major university medical centers
around the world on a variety of topics critical to the treatment
of diseases of the eye. He was an inventor holding multiple
patents in the area of ophthalmology.
In the early 1970s, Alan was approached by the Retinitis
Pigmentosa Foundation to help them develop a scientific plan
to support targeted research that would lead to an
understanding of the causes of retinitis pigmentosa. At the time,
it was recognized that these diseases were inherited, but only in
a very limited way (autosomal dominant, recessive or
X-linked). At the time, no mutations causing RP had been
identified and the Human Genome Project would not be
initiated for another 20 years. Alan agreed and organized the
first Scientific Advisory Board for this Foundation and served
as Chairman. In this leadership role, Alan helped identify and
direct funding to the first laboratory focused on degenerative
retinal disease research, the Berman-Gund Laboratory at the
Massachusetts Eye and Ear Infirmary, Harvard University.
Research Centers focused on retinal degeneration would later
be expanded to many medical centers in North America,
England, and Europe. Alan recognized the importance and
need for animal models with these inherited retinal diseases
and directed funds from the Foundation to support the
development of the dog models with RP identified by Dr.
Gustavo Aguirre at the College of Veterinary Medicine. In the
early 1980s, Alan initiated a scientific plan for the Foundation
to identify the major genes responsible for RP. This led in 1989
to the discovery of a mutation in the rhodopsin gene
 vii

responsible for an autosomal dominant form of retinitis


pigmentosa. Discovery of mutations in other genes causing
retinitis pigmentosa quickly followed. With the discovery of
RP-65, a gene that causes a recessive form of RP, gene therapy
in a dog model with this recessive disorder could be quickly
initiated because of Dr. Laties’ early support from the
Foundation of these dog model lines. Dr. Laties’ early
leadership was hugely important to gene therapy clinical trials
and a number of other therapies related to these inherited
retinal diseases. To honor Dr. Laties, the Foundation Fighting
Blindness named their physicians’ and physician-scientists’
career development award the Alan Laties Career Development
Program and honored him with the inaugural Llura Liggett
Gund Lifetime Achievement Award.
Dr. Laties was a gifted scientist, outstanding leader, and
compassionate human who enriched the lives of his
contemporaries. He played a key role in nurturing and
expanding research in inherited retinal diseases. He is survived
by his wife Deena Gu, a distinguished artist, daughter Jane
Laties, sons Alex P. Laties and Nicholas P. Robinson, and a
brother, David.
Preface

The XIX International Symposium on Retinal Degeneration was held from


September 26 to October 2, 2021. The symposium was initially planned for
October of 2020 in Mendoza, Argentina. However, the global pandemic made
this meeting impossible. With the availability of vaccines, we decided in
March of 2021 that it would be possible to organize the meeting for late
September of 2021. From the beginning, we planned the meeting as an in-­
person meeting with the capability of switching to a hybrid or fully online
meeting depending on the state of the pandemic, and we moved the in-person
meeting to the United States to reduce travel complications for most attend-
ees. As the delta variant began to surge in the weeks leading up to the meet-
ing, we had to activate the hybrid meeting. The meeting platform we
established allowed both in-person and virtual platform talks as well as both
in-person and virtual attendance. The platform was organized so that all pre-
sentations were live and all participants were able to ask questions. All pre-
sentations, including posters, were recorded and made available 4 months
after the meeting. The in-person sessions were held in the Sonesta Nashville
Airport Hotel in Nashville, TN. Because of COVID concerns, the in-person
attendance was small (118 scientists) compared to previous meetings (~250
scientists), but the overall attendance increased to 344 attendees. The virtual
option was the main driver for the increase in attendance. The meeting pro-
gram included four outstanding keynote presentations from Michael Chiang,
Director of the National Eye Institute on Artificial intelligence for clinical
care and research; Douglas Wallace, National academy of Science member
and Professor at the University of Pennsylvania on Mitochondria and the eti-
ology of disease; David Gamm, Professor at the University of Wisconsin-­
Madison on Ultrathin micromolded 3D scaffolds for outer retina
reconstruction; Valeria Canto-Soler, Professor at the University of Colorado
on Human iPSC-derived 3D retinal tissue for stem cell-based therapies for
retinal degenerative diseases. Drs Chiang and Wallace presented via the vir-
tual platform, while Drs Gamm and Canto-Soler presented from the podium.
The program also included 41 platform talks, with 28 presented in person
from the podium and another 13 presented virtually. In addition, 143 posters
were presented as short talks on the virtual platform. Seventy-three of the
posters were also presented in person during two well-attended poster ses-
sions. New and important data were presented at the meeting, and we were
mentioned in a written article published on NPR, and several attendees were
interviewed by reporters from Science and other journals.

ix
x Preface

The RD2021 Travel award competition was highly successful at attracting


qualified applicants. We received a 35% increase in TA applications for a total
of 196. The applications were reviewed by a panel of 14 expert reviewers,
including 6 women, 8 men, and sceintists from a recognized underrepre-
sented minority (URM). Since funding from European sources is dedicated to
European early career scientists, we included three reviewers from Europe.
Many of the panel members have been prior travel awardees. Each applica-
tion was assigned four reviewers, and reviewers independently scored appli-
cations on a 1–9 scale. Based on scores, the applications are ranked and
slotted into funding sources based on funding agency criteria. We were able
to support full travel awards for 60 in-person early career scientists and
another 41 virtual early-career scientists. This is the largest pool of awardees
at an RD meeting. The awards were balanced between men and women. In
addition, we implemented a new diversity and inclusion policy and dedicated
a minimum of six awards to underrepresented minorities (URM). In the end,
we were able to fund 11 URMs to attend the RD meeting.
Although the pandemic made the RD2021 meeting more complex and
more challenging to organize, the RD2021 meeting was, by all accounts, a
terrific success.

Pittsburgh, PA, USA John D. Ash


Boston, MA, USA Eric Pierce
Oklahoma City, OK, USA Robert E. Anderson
Durham, NC, USA Catherine Bowes Rickman
Cleveland, OH, USA Joe G. Hollyfield
Schlieren, Switzerland Christian Grimm
Contents

Part I Age-related Macular Degeneration

High-Resolution Imaging Mass Spectrometry of Human


Donor Eye: Photoreceptors Cells and Basal Laminar
Deposit of Age-­Related Macular Degeneration������������������������������������   3
David M. G. Anderson, Ankita Kotnala, Jeffrey D. Messinger,
Nathan Heath Patterson, Jeffrey M. Spraggins, Christine A. Curcio,
Richard M. Caprioli, and Kevin L. Schey
The Noncanonical Role of Complement Factor H
in Retinal Pigment Epithelium (RPE) Cells and Implications
for Age-Related Macular Degeneration (AMD)������������������������������������   9
Angela Armento, David Adrian Merle, and Marius Ueffing
 acular Pigment Carotenoids and Bisretinoid A2E���������������������������� 15
M
Ranganathan Arunkumar and Paul S. Bernstein
Disturbed Matrix Metalloproteinases Activity in Age-Related
Macular Degeneration ���������������������������������������������������������������������������� 21
Beatriz Martins and Rosa Fernandes
Current Views on Chr10q26 Contribution to Age-Related
Macular Degeneration ���������������������������������������������������������������������������� 27
Navdeep Gogna, Lillian F. Hyde, Gayle B. Collin, Lisa Stone,
Jurgen K. Naggert, and Patsy M. Nishina
Untargeted Lipidomic Profiling of Aged Human Retina
With and Without Age-Related Macular Degeneration (AMD)���������� 37
Ankita Kotnala, David M. G. Anderson, Jeffrey D. Messinger,
Christine A. Curcio, and Kevin L. Schey
Decoding Race and Age-Related Macular Degeneration:
GPR 143 Activity Is the Key�������������������������������������������������������������������� 43
Dorothy Tung and Brian S. McKay
 eroxisome Proliferator-Activated Receptor Gamma
P
Coactivator-­1Alpha (PGC-1α): A Transcriptional Regulator at
the Interface of Aging and Age-Related Macular Degeneration? ������ 49
Freya M. Mowat

xi
xii Contents

Regulation of ABCA1 by miR-33 and miR-34a


in the Aging Eye �������������������������������������������������������������������������������������� 55
Florian Peters and Christian Grimm
 he Role of Gene Expression Regulation on Genetic Risk
T
of Age-­Related Macular Degeneration�������������������������������������������������� 61
Rinki Ratnapriya
Elastin Layer in Bruch’s Membrane as a Target
for Immunization or Tolerization to Modulate Pathology
in the Mouse Model of Smoke-Induced Ocular Injury������������������������ 67
Bärbel Rohrer, Nathaniel Parsons, Balasubramaniam Annamalai,
Crystal Nicholson, Elisabeth Obert, Bryan Jones,
and Andrew D. Dick
Repurposing Drugs for Treatment of Age-Related
Macular Degeneration ���������������������������������������������������������������������������� 73
Sarah G. Francisco and Sheldon Rowan

Part II Extracellular Vesicles

Extracellular Vesicle RNA Contents as Biomarkers


for Ocular Diseases���������������������������������������������������������������������������������� 81
Heran Getachew and Eric Pierce
Proteomics of Retinal Extracellular Vesicles: A Review
into an Unexplored Mechanism in Retinal Health
and AMD Pathogenesis���������������������������������������������������������������������������� 87
Adrian V. Cioanca, Riccardo Natoli, and Yvette Wooff

Part III Gene Editing

Prime Editing Strategy to Install the PRPH2


c.828+1G>A Mutation ���������������������������������������������������������������������������� 97
Salvatore Marco Caruso, Yi-Ting Tsai, Bruna Lopes da Costa,
Masha Kolesnikova, Laura A. Jenny, Stephen H. Tsang,
and Peter M. J. Quinn
Analysis of CRB1 Pathogenic Variants Correctable
with CRISPR Base and Prime Editing�������������������������������������������������� 103
Bruna Lopes da Costa, Laura A. Jenny, Irene H. Maumenee,
Stephen H. Tsang, and Peter M. J. Quinn
Generation of an Avian Myeloblastosis Virus (AMV)
Reverse Transcriptase Prime Editor������������������������������������������������������ 109
Yi-Ting Tsai, Bruna Lopes da Costa, Salvatore Marco Caruso,
Nicolas D. Nolan, Sarah R. Levi, Stephen H. Tsang,
and Peter M. J. Quinn
Contents xiii

Part IV Gene Therapy

Preexisting Neutralizing Antibodies against Different


Adeno-Associated Virus Serotypes in Humans
and Large Animal Models for Gene Therapy���������������������������������������� 117
Divya Ail and Deniz Dalkara
Optimization of Capillary-Based Western Blotting
for MYO7A ���������������������������������������������������������������������������������������������� 125
Kaitlyn R. Calabro, Sanford L. Boye, and Shannon E. Boye
AAV Serotypes and Their Suitability for Retinal
Gene Therapy ������������������������������������������������������������������������������������������ 131
Lynn J. A. Ebner and Christian Grimm
Gene Augmentation for Autosomal Dominant CRX-Associated
Retinopathies�������������������������������������������������������������������������������������������� 135
Chi Sun and Shiming Chen
 xnip Gene Therapy of Retinitis Pigmentosa Improves
T
Cone Health���������������������������������������������������������������������������������������������� 143
Yunlu Xue

Part V Human Retinal Degeneration

Factors Affecting Readthrough of Natural Versus


Premature Termination Codons ������������������������������������������������������������ 149
Avigail Beryozkin, Kerstin Nagel-Wolfum, Eyal Banin,
and Dror Sharon
Integrating Computational Approaches to Predict
the Effect of Genetic Variants on Protein Stability
in Retinal Degenerative Disease�������������������������������������������������������������� 157
Michelle Grunin, Ellen Palmer, Sarah de Jong, Bowen Jin,
David Rinker, Christopher Moth, John A. Capra,
Jonathan L. Haines, William S. Bush, and Anneke I. den Hollander
 etwork Biology and Medicine to Rescue: Applications
N
for Retinal Disease Mechanisms and Therapy�������������������������������������� 165
Anupam K. Mondal and Anand Swaroop
Non-syndromic Retinal Degeneration Caused by Pathogenic
Variants in Joubert Syndrome Genes���������������������������������������������������� 173
Riccardo Sangermano, Egle Galdikaité-Braziené,
and Kinga M. Bujakowska
Exonic Variants that Affect Splicing – An Opportunity
for “Hidden” Mutations Causing Inherited Retinal Diseases�������������� 183
Yogapriya Sundaresan, Eyal Banin, and Dror Sharon
 nhanced S-cone Syndrome, a Mini-review������������������������������������������ 189
E
Yiyi Wang, Jessica Wong, Jacque L. Duncan, Austin Roorda,
and William S. Tuten
xiv Contents

Part VI Inflammation

 he Role of Microglia in Inherited Retinal Diseases���������������������������� 197


T
Asha Kumari and Shyamanga Borooah
CD68: Potential Contributor to Inflammation and RPE
Cell Dystrophy������������������������������������������������������������������������������������������ 207
Mayur Choudhary and Goldis Malek
 ene Expression of Clusterin, Tissue Inhibitor
G
of Metalloproteinase-1, and Their Receptors in Retinal
Pigment Epithelial Cells and Müller Glial Cells Is
Modulated by Inflammatory Stresses���������������������������������������������������� 215
Mengmei Zheng, Eun-Jin Lee, Shinwu Jeong,
and Cheryl Mae Craft

Part VII Mechanisms of Degeneration

 xonal Transport Defects in Retinal Ganglion Cell Diseases�������������� 223


A
Iskalen Cansu Topcu Okan, Fatma Ozdemir, and Cavit Agca
 onnexins Biology in the Pathophysiology of Retinal Diseases���������� 229
C
Alejandro Ponce-Mora, Andrea Yuste, Giuliana Perini-Villanueva,
María Miranda, and Eloy Bejarano
 ole of Nuclear NAD+ in Retinal Homeostasis�������������������������������������� 235
R
Emily E. Brown, Michael J. Scandura, and Eric Pierce
Retinal Pigmented Epithelium-­Derived Ectopic Norrin Does
Not Promote Intraretinal Angiogenesis in Transgenic Mice���������������� 241
Andrea E. Dillinger and Ernst R. Tamm
Caveolin-1 in Müller Glia Exists as Heat-Resistant, High
Molecular Weight Complexes ���������������������������������������������������������������� 249
Eric N. Enyong, Jami Gurley, Virginie Sjoelung,
and Michael H. Elliott
 ole of VLC-PUFAs in Retinal and Macular Degeneration���������������� 257
R
Aruna Gorusupudi, Uzoamaka Nwagbo, and Paul S. Bernstein
Ocular Amyloid, Condensates, and Aggregates – Higher-Order
Protein Assemblies Participate in Both Retinal Degeneration
and Function�������������������������������������������������������������������������������������������� 263
Michael H. Hayes, DaNae R. Woodard, and John D. Hulleman
 hotoreceptor Ion Channels in Signaling and Disease������������������������ 269
P
Shivangi M. Inamdar, Colten K. Lankford, and Sheila A. Baker
The Role of Peripherin-2/ROM1 Complexes in Photoreceptor
Outer Segment Disc Morphogenesis������������������������������������������������������ 277
Tylor R. Lewis, Muayyad R. Al-Ubaidi, Muna I. Naash,
and Vadim Y. Arshavsky
Contents xv

Human Mutations in Arl3, a Small GTPase Involved


in Lipidated Cargo Delivery to the Cilia, Cause
Retinal Dystrophy������������������������������������������������������������������������������������ 283
Amanda M. Travis and Jillian N. Pearring
Genotype–Phenotype Association in ABCA4-Associated
Retinopathy���������������������������������������������������������������������������������������������� 289
Maximilian Pfau, Wadih M. Zein, Laryssa A. Huryn,
Catherine A. Cukras, Brett G. Jeffrey, Robert B. Hufnagel,
and Brian P. Brooks
Retinal Pathoconnectomics: A Window into
Neurodegeneration���������������������������������������������������������������������������������� 297
Rebecca L. Pfeiffer and Bryan W. Jones
The Role of Ceramide in Inherited Retinal
Disease Pathology������������������������������������������������������������������������������������ 303
Xinye Qian, Tanmay Srinivasan, Jessica He, and Rui Chen
 xtracellular Matrix: The Unexplored Aspects
E
of Retinal Pathologies and Regeneration���������������������������������������������� 309
Dmitri Serjanov and David R. Hyde
Role of TFEB in Diseases Associated with
Lysosomal Dysfunction���������������������������������������������������������������������������� 319
Hsuan-Yeh Pan and Mallika Valapala
Retinoic Acid Receptor-Related Orphan Receptors
(RORs) in Eye Development and Disease���������������������������������������������� 327
Felix Yemanyi, Kiran Bora, Alexandra K. Blomfield,
and Jing Chen

Part VIII Mechanisms of Degeneration – Animal Models

A Novel Mouse Model for Late-­Onset Retinal Degeneration


(L-ORD) Develops RPE Abnormalities Due
to the Loss of C1qtnf5/Ctrp5�������������������������������������������������������������������� 335
Shyamanga Borooah, Anil Chekuri, Shikha Pachauri,
Bhubananda Sahu, Marina Vorochikhina, John J. Suk,
Dirk-­Uwe Bartsch, Venkata R. M. Chavali, Monica M. Jablonski,
and Radha Ayyagari
 omparison of Mouse Models of Autosomal Dominant Retinitis
C
Pigmentosa Due to the P23H Mutation of Rhodopsin�������������������������� 341
Shannon R. Barwick and Sylvia B. Smith
Compensatory Cone-Mediated Mechanisms in Inherited
Retinal Degeneration Mouse Models: A Functional
and Gene Expression Analysis���������������������������������������������������������������� 347
Alicia A. Brunet, David M. Hunt, Carla Mellough,
Alan R. Harvey, and Livia S. Carvalho
xvi Contents

Inhibition of Ryanodine Receptor 1 Reduces Endoplasmic


Reticulum (ER) Stress and Promotes ER Protein
Degradation in Cyclic Nucleotide-Gated
Channel Deficiency���������������������������������������������������������������������������������� 353
Fan Yang, Hongwei Ma, Rekha Garg, Alfred Lewin,
and Xi-Qin Ding
 ouse Choroid Proteome Revisited: Focus on Aging�������������������������� 359
M
Donita Garland, James Harnly, and Radha Ayyagari
Morphological and Functional Comparison
of Mice Models for Retinitis Pigmentosa ���������������������������������������������� 365
Prakadeeswari Gopalakrishnan, Avigail Beryozkin,
Eyal Banin, and Dror Sharon
 urrent Advancements in Mouse Models of Retinal Disease�������������� 371
C
T. J. Hollingsworth, Xiangdi Wang, Raven N. Simpson,
William A. White, Robert W. Williams,
and Monica M. Jablonski
Single-Cell Transcriptomic Profiling of Müller
Glia in the rd10 Retina���������������������������������������������������������������������������� 377
Duygu Sigurdsson and Christian Grimm

Methods for In Vivo Characterization of Proteostasis
in the Mouse Retina �������������������������������������������������������������������������������� 383
Yixiao Wang and Ekaterina S. Lobanova
Absence of PRCD Leads to Dysregulation in Lipid
Homeostasis Resulting in Disorganization
of Photoreceptor Outer Segment Structure������������������������������������������ 389
Sree I. Motipally and Saravanan Kolandaivelu

Expansion Microscopy of Mouse Photoreceptor Cilia ������������������������ 395
Abigail R. Moyel, Michael A. Robichaux, and Theodore Wensel
Rod Photoreceptor-Specific Ablation of Metformin Target,
AMPK, in a Preclinical Model of Autosomal
Recessive Retinitis Pigmentosa �������������������������������������������������������������� 403
Nicholas D. Nolan, Laura A. Jenny, Stephen H. Tsang,
and Xuan Cui
TLR2 Is Highly Overexpressed in Retinal Myeloid
Cells in the rd10 Mouse Model of Retinitis Pigmentosa ���������������������� 409
Alonso Sánchez-Cruz, Enrique J. de la Rosa,
and Catalina Hernández-Sánchez
Environmental Light Has an Essential Effect
on the Disease Expression in a Dominant RPE65 Mutation���������������� 415
Wenjing Wu, Yusuke Takahashi, Xiang Ma, Gennadiy Moiseyev,
and Jian-Xing Ma
Contents xvii

Microglia Preserve Visual Function in a Mouse Model


of Retinitis Pigmentosa with Rhodopsin-P23H Mutant ���������������������� 421
Chen Yu and Daniel R. Saban

Part IX Mechanisms of Degeneration – Metabolism

Measuring the Release of Lactate from Wild-Type


and rd1 Mouse Retina������������������������������������������������������������������������������ 429
Yiyi Chen, Laimdota Zizmare, Christoph Trautwein,
and François Paquet-Durand
Aerobic Glycolysis in Photoreceptors Supports Energy
Demand in the Absence of Mitochondrial Coupling���������������������������� 435
Daniel T. Hass, Celia M. Bisbach, Martin Sadilek,
Ian R. Sweet, and James B. Hurley

Redox Status in Retinitis Pigmentosa���������������������������������������������������� 443
L. Olivares-González, S. Velasco, I. Campillo, J. M. Millán,
and R. Rodrigo
Perspectives on Retinal Dolichol Metabolism,
and Visual Deficits in Dolichol Metabolism-Associated
Inherited Disorders���������������������������������������������������������������������������������� 449
Sriganesh Ramachandra Rao, Steven J. Pittler,
and Steven J. Fliesler
Retinal Metabolic Profile on IMPG2 Deficiency Mice
with Subretinal Lesions �������������������������������������������������������������������������� 457
Rong Xu, Yekai Wang, Jianhai Du, and Ezequiel M. Salido

Part X Neuroprotection

Glutathione Coating of Liposomes Enhances the Delivery


of Hydrophilic Cargo to the Inner Nuclear Layer
in Retinal Cultures ���������������������������������������������������������������������������������� 467
Gustav Christensen and François Paquet-Durand
Modification of Müller Glial Cell Fate and Proliferation
with the Use of Small Molecules ������������������������������������������������������������ 473
Marcus J. Hooper
A Potential Neuroprotective Role for Pyruvate Kinase
2 in Retinal Degeneration������������������������������������������������������������������������ 479
Jiaming Zhou, Michel Rasmussen, and Per Ekström

Part XI Photoreceptors

Critical Role of VEGF as a Direct Regulator


of Photoreceptor Function���������������������������������������������������������������������� 487
Jianyan Hu, Meili Zhu, Dai Li, Qiang Wu, and Yun-Zheng Le
xviii Contents


Lysine Ubiquitylation Drives Rhodopsin Protein Turnover���������������� 493
Allen P. F. Chen, Leon Chea, Eun-Jin Lee,
and Jonathan H. Lin
 Silico Prediction of MYO1C-­Rhodopsin Interactions
In
and Its Significance in Protein Localization
and Visual Function �������������������������������������������������������������������������������� 499
Glenn P. Lobo, Rakesh Radhakrishnan, Matthias Leung,
Andrew Gruesen, Hans-­Joachim Knölker, Frederik J. van Kuijk,
and Sandra R. Montezuma
A Ciliary Branched Actin Network Drives
Photoreceptor Disc Morphogenesis�������������������������������������������������������� 507
William J. Spencer and Vadim Y. Arshavsky

Part XII RPE


Revisiting the Daily Timing of POS Phagocytosis�������������������������������� 515
Antonio E. Paniagua, Harjas S. Sabharwal, Kausalya Kethu,
Andrew W. Chang, and David S. Williams
Inhibition of Bacterial Peptidoglycan Cytopathy
by Retina Pigment Epithelial PGRP2 Amidase������������������������������������ 521
Marlyn P. Langford, Laura A. Perilloux-Lyons,
and A. Scott Kavanaugh
Understanding Ischemic Retinopathies: The Role
of Succinate and Its Receptor in Retinal
Pigment Epithelium �������������������������������������������������������������������������������� 527
Bilge Esin Ozturk

The Amphipathic Helix in Visual Cycle Proteins: A Review���������������� 533
Sheetal Uppal, Eugenia Poliakov, Susan Gentleman,
and T. Michael Redmond

The Retinal Pigment Epithelium: Cells That Know the Beat!������������ 539
Elora M. Vanoni and Emeline F. Nandrot

Part XIII Stem Cell Models and Therapies

Retinal Organoids: A Human Model System


for Development, Diseases, and Therapies�������������������������������������������� 549
Sangeetha Kandoi and Deepak A. Lamba

Modeling Retinitis Pigmentosa with Patient-Derived iPSCs �������������� 555
Yeh Chwan Leong and Jane C. Sowden

Primary Retinal Cell Cultures as a Model to Study
Retina Biology������������������������������������������������������������������������������������������ 565
Germán A. Michelis, Luis E. Politi, and S. Patricia Becerra
Contents xix

Generation of CRB1 RP Patient-­Derived iPSCs


and a CRISPR/Cas9-­Mediated Homology-Directed
Repair Strategy for the CRB1 c.2480G>T Mutation���������������������������� 571
Bruna Lopes da Costa, Yao Li, Sarah R. Levi, Stephen H. Tsang,
and Peter M. J. Quinn
Inducing Neural Regeneration from Glia Using Proneural
bHLH Transcription Factors������������������������������������������������������������������ 577
Levi Todd

Index�������������������������������������������������������������������������������������������������������� 583
Part I
Age-related Macular Degeneration
High-Resolution Imaging Mass
Spectrometry of Human Donor Eye:
Photoreceptors Cells and Basal
Laminar Deposit of Age-­Related
Macular Degeneration

David M. G. Anderson, Ankita Kotnala,


Jeffrey D. Messinger, Nathan Heath Patterson,
Jeffrey M. Spraggins, Christine A. Curcio,
Richard M. Caprioli, and Kevin L. Schey

Abstract nohistochemistry. Although both techniques


provide valuable information on prognosis
Pathologies of the retina are clinically visual- and disease state, a comprehensive method for
ized in vivo with OCT and ex vivo with immu- fully elucidating molecular constituents pres-
ent in locations of interest is desirable. The
D. M. G. Anderson · N. H. Patterson · R. M. Caprioli purpose of this work was to use multimodal
· K. L. Schey (*) imaging technologies to localize the vast num-
Department of Biochemistry, Vanderbilt University, ber of molecular species observed with
Nashville, TN, USA
matrix-assisted laser desorption ionization
Mass Spectrometry Research Center, Vanderbilt imaging mass spectrometry (MALDI IMS) in
University School of Medicine, Nashville, TN, USA
aged and diseased retinal tissues. Herein,
e-mail: k.schey@vanderbilt.edu
MALDI IMS was utilized to observe molecu-
A. Kotnala
lar species that reside in photoreceptor cells
Department of Biochemistry, Vanderbilt University,
Nashville, TN, USA and also a basal laminar deposit from two
human donor eyes. The molecular species
Department of Ophthalmology and Visual Sciences,
University of Alabama at Birmingham, observed to accumulate in these discrete
Birmingham, AL, USA regions can be further identified and studied to
J. D. Messinger · C. A. Curcio attempt to gain a greater understanding of bio-
Department of Ophthalmology and Visual Sciences, logical processes occurring in debilitating eye
University of Alabama at Birmingham, diseases such as age-related macular degener-
Birmingham, AL, USA
ation (AMD).
J. M. Spraggins
Mass Spectrometry Research Center, Vanderbilt
Keywords
University School of Medicine, Nashville, TN, USA
Department of Ophthalmology and Visual Sciences, Age-related macular degeneration · Macula ·
University of Alabama at Birmingham, Retinal pigment epithelium · Photoreceptors ·
Birmingham, AL, USA Basal lamina deposit · MALDI IMS
Department of Cell and Developmental Biology,
Vanderbilt University School of Medicine,
Nashville, TN, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 3


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_1
4 D. M. G. Anderson et al.

1 Introduction 2.1 Tissue Acquisition


and Characterization
Matrix-assisted laser desorption ionization
imaging mass spectrometry (MALDI IMS) can Whole eyes were obtained from deceased human
localize and display the tissue distributions of donors via Advancing Sight Network (formerly
hundreds to thousands of molecules, at cellular the Alabama Eye Bank) by the UAB authors.
resolution, without the need for antibodies or
radioisotopes [1]. With effective co-registration
to multimodal optical imaging and optical 2.2 Tissue Handling and Ex Vivo
coherence topography (OCT) microscopy, these Imaging
distributions can be accurately correlated to
very small histological features of the neural Methods were optimized for multimodal ex vivo
retina and retinal pigment epithelium (RPE). clinical imaging of the ocular fundus [13]. Globes
MALDI IMS methods have been used to exam- with lens and iris in place were immersed in buff-
ine eye tissues including the retina [2–4], optic ered 4% paraformaldehyde overnight. Iris and
nerve [4–6], lens [7, 8], and cornea [9]. Distinct lens were removed before imaging.
cell and synaptic layers of the retina have unique For imaging with OCT and scanning laser
layer-specific lipid and metabolite signatures ophthalmoscopy, globes were immersed in buffer
distinguished by IMS [4, 10, 11]. By applying facing frontward within a custom-built chamber
multimodal optical imaging technologies with with a 60-diopter lens [13]. Spectral domain
accurate registration and incorporating data-rich OCT images were captured with a Spectralis
IMS images [12], cellular and subcellular local- (HRA&OCT, HRA2; Heidelberg Engineering).
ization of specific molecules informative to cell- Tissues were embedded in 2.5% carboxy-
specific biochemistry can be observed. Human methyl cellulose (Sigma C9481), and serial
retinal lipid composition studies have been per- 10 μm cryosections were collected on Superfrost
formed in the past. The results, while valuable, glass slides and on large, 45 × 45 mm in-house,
provide limited information on cellular origin, polylysine-coated indium-tin-oxide (ITO) slides
as experiments require dissections followed by (Delta Technologies Loveland, CO, USA).
solvent extractions. MALDI IMS offers a
“molecular microscope” that localizes tissue
components in situ by molecular weights [11], 2.3 MALDI IMS Analysis
simultaneously providing hundreds to thou-
sands of spatially resolved signals. In this study, The matrices 2,5-dihydroxyacetophenone (DHA)
we used a newly developed method of high- and 1,5-diaminonaphthalene (DAN) (Sigma
accuracy registration [12] to co-­ register high Aldrich, St. Louis, MO, USA) were applied to
spatial resolution IMS images with OCT auto- tissue sections by sublimation [14]. MALDI IMS
fluorescence and histological images of the data were acquired with a laser spot size of
same tissue to examine subcellular localizations 10–15 μm in full scan mode using a Bruker
and molecular features of photoreceptors and SolariX 9.4T FTICR mass spectrometer (Bruker
AMD pathology. Daltonics Billerica, MA, USA). Following data
acquisition, an advanced image registration
workflow [12] was performed. More detailed
2 Methods information of the image registration process can
be found in publications by Patterson et al. [12]
This section has been summarized from Anderson and Anderson et al. [3]. Molecular identifications
et al. [3]; for detailed explanations, please see were made using LC-MS of chloroform-­methanol
this reference. extracts from adjacent tissue sections.
High-Resolution Imaging Mass Spectrometry of Human Donor Eye: Photoreceptors Cells and Basal… 5

3 Results a cardiolipin CL(70:5). Cardiolipins are highly


abundant in mitochondria, which are abundant in
3.1 Signals Specific the ellipsoid portion of photoreceptor inner seg-
to Photoreceptors and RPE ments. At the distal part of the photoreceptor
cells, outer segments are highly interleaved with
Figure 1a shows MALDI IMS and optical micros- apical processes of RPE cells. A DHA-containing
copy focusing on photoreceptors and their PE(18:0_22:6) is observed at m/z 790.539 (yel-
­support cells. The RPE sends delicate processes low) in panel D which can be observed with high
in the apical direction to contact photoreceptor abundance in the outer segments, while a signal
outer segments, near the RPE cell body for rods observed at m/z 728.596 (green) is localized
and 10–15 μm above the cell body, to contact above and within the RPE.
cone outer segments, which are shorter. Figure 1a
is color-coded depiction of photoreceptor and
RPE compartments associated with IMS signals 3.2 Signals Specific to Basal
in Fig. 1b (blue ONL, red inner segments, yellow Laminar Deposit
outer segments, green RPE).
Figure 1b shows MALDI IMS images over- Figure 2 shows multimodal imaging of a 93-year-­
laid with H&E images from this donor. The pho- old donor tissue with the imaging modalities
toreceptors on the left side of the image are separated into panels. Figure 2a displays ex vivo
attached to the RPE and detached from the RPE OCT hyperreflective foci (yellow arrowhead) and
on the right side, a common artifact which can an RPE elevation (green arrowhead) near the
occur during sample preparation. In Fig. 1a, the fovea. Figure 2b shows that retinal layers are vis-
signal at m/z 818.575 was observed with high ible in H&E-stained sections after IMS data
abundance in the ONL and was identified as acquisition. The inset magnifies BLamD (PASH
PE(20:0_22:6) (blue). This region is comprised staining of an alternate section), clearly indicat-
of the photoreceptor cell bodies and processes of ing thickened extracellular matrix between the
Müller (radial) glia. A highly localized signal can RPE plasma membrane and its native basal lam-
be observed with high abundance along a narrow ina. Figure 2c shows autofluorescence of the ele-
band aligned with photoreceptor inner segments vated RPE layer and anteriorly migrated RPE
at m/z 1426.0 (red). This signal was identified as cells, which account for high-risk-indicating

Fig. 1 MALDI IMS signals consistent with localization MALDI IMS images and H&E-stained tissue images
to photoreceptor and RPE compartments. (a) Schematic overlaid in perifoveal retina displaying signals from mul-
diagram of outer retina, excerpted from Fig. 1a. Blue, tiple lipid classes that localize to subcellular compart-
pink, yellow, and green bands indicate layers formed by ments of the photoreceptor cells. (b) Overlay showing
highly compartmentalized and vertically aligned photore- four separate signals. (c) Localized to ONL. (d) Localized
ceptors and RPE cells in panels b, c. Layers: OPL outer to photoreceptor inner and outer segments. (e) Localized
plexiform layer, ONL outer nuclear layer, ELM external to mitochondria-rich photoreceptor inner segments. (f)
limiting membrane, RPE retinal pigment epithelium, BrM Localized to RPE apical processes
Bruch’s membrane, R rod, C cone photoreceptors. (b–f)
6 D. M. G. Anderson et al.

Fig. 2 Imaging mass spectrometry (IMS) for molecularly pigmented debris (yellow) and dysmorphic RPE overly-
informed optical coherence tomography (OCT) and ing BLamD. Insert, BLamD with basal mound (arrow).
tissue-­level target discovery. Asterisk, foveal pit; RPE, Basal mounds contain soft druse material. Layers: GCL
retinal pigment epithelium. Color-coded arrowheads rep- ganglion cell, INL inner nuclear, HFL Henle fiber, ONL
resent corresponding structures across modalities, in a outer nuclear. (c) Autofluorescent, pigmented debris (yel-
93-year-old human donor eye. (a) OCT B-scan shows low) and dysmorphic RPE (green). (d) IMS reveals an m/z
subretinal hyperreflective material (yellow) and an RPE signal at 799.673, restricted to BLamD and not detected in
elevation (green). (b) H&E stained cryosection shows the RPE

hyperreflective foci of clinical OCT. Figure 2d 4 Conclusions


shows that a sphingomyelin-related lipid
(PE-Cer-NMe2(42:1)) at m/z 799.671 [15] is MALDI IMS combined with multimodal imag-
highly abundant and localizes to BLamD and ing methods and ex vivo OCT provides a power-
RPE, building on previous histochemical and ful tool to elucidate the molecular composition
chromatography findings of lipids in this deposit and localization of molecular species in key
[16, 17]. regions and pathology associated with ocular dis-
High-Resolution Imaging Mass Spectrometry of Human Donor Eye: Photoreceptors Cells and Basal… 7

ease. Understanding molecular processes occur- 9. Chen Y, Jester JV, Anderson DM, Marchitti SA, Schey
KL, Thompson DC, et al. Corneal haze phenotype
ring in BLamD in early AMD is important as in Aldh3a1-null mice: in vivo confocal microscopy
they are early-identified histologic risk factors and tissue imaging mass spectrometry. Chem Biol
for AMD progression [18] and are just now being Interact. 2017;276:9–14.
recognized clinically [19, 20]. 10. Anderson DM, Ablonczy Z, Koutalos Y, Spraggins
J, Crouch RK, Caprioli RM, et al. High reso-
lution MALDI imaging mass spectrometry of
Acknowledgments This project was supported by the retinal tissue lipids. J Am Soc Mass Spectrom.
National Institutes of Health P41 GM103391 (R.M.C.) 2014;25(8):1394–403.
and R01 EY027948 (C.A.C.). Support was also received 11. Anderson DMG, Ablonczy Z, Koutalos Y,
by a Research to Prevent Blindness Catalyst Award for Hanneken AM, Spraggins JM, Calcutt MW, et al.
Innovative Research Approaches for Age-Related Macular Bis(monoacylglycero)phosphate lipids in the retinal
Degeneration to K.L.S. pigment epithelium implicate lysosomal/endosomal
dysfunction in a model of Stargardt disease and
human retinas. Sci Rep. 2017;7(1):17352.
12. Patterson NH, Tuck M, Van de Plas R, Caprioli
References RM. Advanced registration and analysis of
MALDI imaging mass spectrometry measurements
1. Caprioli RM, Farmer TB, Gile J. Molecular imag- through autofluorescence microscopy. Anal Chem.
ing of biological samples: localization of peptides 2018;90(21):12395–403.
and proteins using MALDI-TOF MS. Anal Chem. 13. Pang CE, Messinger JD, Zanzottera EC, Freund KB,
1997;69(23):4751–60. Curcio CA. The onion sign in neovascular age-related
2. Bowrey HE, Anderson DM, Pallitto P, Gutierrez DB, macular degeneration represents cholesterol crystals.
Fan J, Crouch RK, et al. Imaging mass spectrometry Ophthalmology. 2015;122(11):2316–26.
of the visual system: advancing the molecular under- 14. Hankin J, Barkley R, Murphy R. Sublimation as
standing of retina degenerations. Proteomics Clin a method of matrix application for mass spec-
Appl. 2016;10(4):391–402. trometric imaging. J Am Soc Mass Spectrom.
3. Anderson DMG, Messinger JD, Patterson NH, Rivera 2007;18(9):1646–52.
ES, Kotnala A, Spraggins JM, et al. Lipid landscape 15. Liu A, Chang J, Lin Y, Shen Z, Bernstein PS. Long-­
of the human retina and supporting tissues revealed chain and very long-chain polyunsaturated fatty acids
by high resolution imaging mass spectrometry. bioR in ocular aging and age-related macular degeneration.
xiv:2020:2020.04.08.029538. J Lipid Res. 2011;51:3217–29.
4. Zemski Berry KA, Gordon WC, Murphy RC, Bazan 16. Curcio CA, Presley JB, Malek G, Medeiros NE,
NG. Spatial organization of lipids in the human retina Avery DV, Kruth HS. Esterified and unesteri-
and optic nerve by MALDI imaging mass spectrom- fied cholesterol in drusen and basal deposits of
etry. J Lipid Res. 2014;55(3):504–15. eyes with age-related maculopathy. Exp Eye Res.
5. Anderson DM, Spraggins JM, Rose KL, Schey 2005;81(6):731–41.
KL. High spatial resolution imaging mass spectrom- 17. Wang L, Li C-M, Rudolf M, Belyaeva OV, Chung
etry of human optic nerve lipids and proteins. J Am BH, Messinger JD, et al. Lipoprotein particles of
Soc Mass Spectrom. 2015;26(6):940–7. intraocular origin in human Bruch membrane: an
6. Stark DT, Anderson DMG, Kwong JMK, Patterson unusual lipid profile. Invest Ophthalmol Vis Sci.
NH, Schey KL, Caprioli RM, et al. Optic nerve regen- 2009;50(2):870–7.
eration after crush remodels the injury site: molecu- 18. Sarks SH. Ageing and degeneration in the macular
lar insights from imaging mass spectrometryoptic region: a clinico-pathological study. Br J Ophthalmol.
nerve regeneration imaging mass spectrometry. Invest 1976;60(5):324–41.
Ophthalmol Vis Sci. 2018;59(1):212–22. 19. Tan ACS, Astroz P, Dansingani KK, Slakter JS,
7. Grey AC, Schey KL. Age-related changes in the spa- Yannuzzi LA, Curcio CA, et al. The plateau, an
tial distribution of human lens alpha-crystallin prod- optical coherence tomographic signature of geo-
ucts by MALDI imaging mass spectrometry. Invest graphic atrophy: evolution, multimodal imaging,
Ophthalmol Vis Sci. 2009;50(9):4319–29. and candidate histology. Invest Ophthalmol Vis Sci.
8. Stella DR, Floyd KA, Grey AC, Renfrow MB, 2017;58(4):2349–58.
Schey KL, Barnes S. Tissue localization and solu- 20. Sura AA, Chen L, Messinger JD, Swain TA, McGwin
bilities of alphaA-crystallin and its numerous G Jr, Freund KB, et al. Measuring the contributions of
C-terminal truncation products in pre- and postcata- basal laminar deposit and Bruch’s membrane in age-­
ractous ICR/f rat lenses. Invest Ophthalmol Vis Sci. related macular degeneration. Invest Ophthalmol Vis
2010;51(10):5153–61. Sci. 2020;61(13):19.
The Noncanonical Role
of Complement Factor H in Retinal
Pigment Epithelium (RPE) Cells
and Implications for Age-Related
Macular Degeneration (AMD)

Angela Armento, David Adrian Merle,


and Marius Ueffing

Abstract pathway of the complement system, and the


FH Y402H variant leads to increased comple-
Age-related macular degeneration (AMD) is a ment activation, which is detectable in AMD
complex degenerative disease of the retina. patients. For this reason, various therapeutic
Dysfunction of the retinal pigment epithelium approaches targeting the complement system
(RPE) occurs in early stages of AMD, and have been developed, however, so far with
progressive RPE atrophy leads to photorecep- very limited or no efficacy. Interestingly,
tor death and visual impairments that ulti- recent studies suggest roles for FH beyond
mately manifest as geographic atrophy (GA), complement regulation. Here, we will discuss
one of the late-stage forms of AMD. AMD is the noncanonical functions of FH in RPE cells
caused by a combination of risk factors includ- and highlight the potential implications of
ing aging, lifestyle choices, and genetic pre- those functions for future therapeutic
disposition. A gene variant in the complement approaches.
factor H gene (CFH) that leads to the Y402H
polymorphism in the factor H protein (FH) Keywords
confers the second highest risk for the devel-
opment and progression of AMD. FH is a Age-related macular degeneration (AMD) ·
major negative regulator of the alternative Retinal pigment epithelium (RPE) cells ·
Intracellular complement factor H (CFH)

A. Armento (*) · M. Ueffing


Institute for Ophthalmic Research, Department for
Ophthalmology, Eberhard Karls University of 1 Introduction
Tübingen, Tübingen, Germany
e-mail: angela.armento@uni-tuebingen.de;
marius.ueffing@uni-tuebingen.de Age-related macular degeneration (AMD) is a
D. A. Merle complex and progressive disease of the macula,
Institute for Ophthalmic Research, Department for which affects mainly the elderly population,
Ophthalmology, Eberhard Karls University of especially in the developed countries [1, 2]. Due
Tübingen, Tübingen, Germany to demographic changes in western societies,
Department of Ophthalmology, Medical University of AMD incidence is constantly rising, and the con-
Graz, Graz, Austria comitant visual impairment threatens not only
e-mail: david.merle@medunigraz.at

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 9


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_2
10 A. Armento et al.

the individual’s quality of life but also poses a 402H of FH causes uncontrolled complement
significant burden on healthcare systems world- activation in vitro and in vivo [9]. Moreover,
wide [3]. The early stages of the disease are char- complement factors accumulate in drusen [10],
acterized by the presence of enlarging drusen in and increased complement system activation has
the retina. With disease progression, early AMD been detected in AMD patients, independent of
advance to late stages: wet or dry AMD. Wet their genotype [11].
AMD is defined by the presence of neovascular- Due to the strong implications of complement
ization in the retina, while dry AMD is limited to system activation in AMD pathology, various
progressive RPE and retinal atrophy, called geo- complement inhibitors have been tested in clini-
graphic atrophy (GA), without newly formed cal trials. However, most agents that entered
vessels. Around 10% of the cases constitute wet phase II were discontinued due to a reported lack
AMD, and intravitreal anti-VEGF is effective in of efficacy. Interestingly, in some clinical trials, a
treating most cases. However, only recently, group of subjects showed improvements, while
one approved treatment has become available for others did not [12]. Apparently, one of the sus-
dry AMD in the US [2, 8]. pected reasons for the observed lack of efficacy is
The main pathogenetic driver for AMD is a lack of patient’s stratification and identification
advanced age. Specific genetic factors can pre- of those patients that are likely to benefit from
dispose for AMD or protect from it. Unhealthy complement inhibition. Moreover, several stud-
lifestyle habits, like smoking or malnutrition, ies point to a lack of understanding of the role of
can further increase the risk for AMD. The com- FH in AMD pathology, especially in RPE cells,
bination of those risk factors leads to pathologi- which are degenerating in AMD.
cal changes in the retina, involving a plethora of
cellular mechanisms promoting disease pro-
gression [4]. 3 Noncanonical Role
of Complement Factor H
in RPE Cells
2 Complement System in AMD
RPE cells exert several functions vital for retinal
Among the processes involved in AMD, the com- homeostasis, which are impaired in AMD [13].
plement system is associated to AMD at different First of all, RPE cells are responsible for phago-
levels. First of all, many single nucleotide poly- cytosis of shed photoreceptor outer segments
morphisms (SNPs) associated with AMD cluster (POS) and recycling of visual pigments, needed
into genes of the alternative pathway of the com- for a proper visual cycle. This process is energy
plement system with the Complement Factor H intensive, requiring constant mitochondrial res-
(CFH) gene carrying the second highest risk for piration and glycolysis within RPE cells. Due to
AMD [5]. The most common risk haplotype a lack of the inner retina vasculature in the mac-
causes an amino acid substitution from tyrosine ula, RPE cells are vital to ascertain nutrient sup-
to histidine at position 402 (called Y402H) in ply from the choriocapillaris. Any pathological
proteins coded by CFH, which are full length FH disruption or perturbance of metabolic activity
and a truncated version, FHL-1 [6, 7]. The com- of RPE cells would affect the amount of nutri-
plement system is a part of the innate immune ents that can reach the neuroretina. Indeed, the
system designed to recognize and mediate the so-­called bioenergetics crisis of RPE cells has
removal of pathogens or waste material [8]. The been described for AMD pathology [14].
eye is an immune-privileged organ, meaning that Moreover, RPE cells quench short UV light as
inflammation needs to be strictly limited, and well as the continuous influx of peroxidized lip-
therefore uncontrolled complement activation is ids from phagocytosed photoreceptor outer seg-
deleterious [8]. FH is a major negative regulator ments and thus need a high degree of antioxidant
of the complement system, and the AMD variant capacity as well as perfect functioning of their
The Noncanonical Role of Complement Factor H in Retinal Pigment Epithelium (RPE) Cells and… 11

mitochondria to protect the retina from exces- [15] or in the presence of the CFH 402H variant
sive oxidative stress. [20]. Moreover, accumulation of lipids was
Our recent work demonstrates that FH is shown also in cfh Y402H mouse models of AMD,
endogenously produced by RPE cells. Within especially when subjected to high cholesterol
RPE cells, FH is acting locally supporting retinal diet [21].
homeostasis [15, 16]. This function of FH is con- The mechanism of action of intracellular FH
text dependent, and we suggest that locally pro- is not fully elucidated. Based on the data gener-
duced FH exerts specific functions at the ated in iPSC-RPE cells carrying the CFH 402H
RPE/retina interface. Most of these newly dis- variant, it has been speculated that more than one
covered functions (summarized in Fig. 1) affect a signaling pathway mediates the effects of FH
balanced regulation of immune competence and [19]. Specifically, it has been postulated that the
surveillance as well as regulating oxidative stress NF-kB pathway and mTOR pathway are likely
response and energy metabolism, mechanisms involved, in concomitance with autophagy and
which are part of AMD pathology [8]. proteasome activity dysregulation, which ulti-
FH plays a protective role against oxidative mately lead to mitochondrial damage [19].
stress in RPE cells. Exogenously applied recom- In our model of CFH-silenced hTERT-RPE1
binant FH protects RPE cells from damage cells, we could verify these hypotheses. Indeed,
induced by the exposure to lipid oxidation prod- we found that in absence of endogenous FH, both
ucts [17, 18]. In parallel, loss of endogenous FH the NF-kB and mTOR pathway are upregulated,
in RPE cells, via CFH silencing, also led to with NF-kB triggering pro-inflammatory cyto-
increased vulnerability to oxidative stress, kine expression [22] and the mTOR pathway
increased levels of oxidized lipids, and reduced modulating mitochondrial respiration, but not
viability [15]. Interestingly, addition of recombi- glycolysis [23]. The mTOR pathway is a major
nant FH in CFH-silenced RPE cells did not cause regulator of autophagy [24]. Dysfunction in the
any rescuing effects [15]. This finding indicates autophagy-lysosomal axis in iPSC-RPE cells
that, although exogenous FH is protective, a with CFH high risk was reported but is not regu-
proper intracellular FH function must be present lated by this pathway. Its dysfunction rather
to ascertain physiological balance and resilience depends on the accumulation of complement
to stress. This phenomenon may be explained by activation products [25]. Based on our analyses
impaired mitochondrial function seen in iPSC-­ of the intracellular FH protein interactions that
RPE cells carrying the CFH Y402H polymor- form a functional interactome in RPE cells, we
phism [19] and CFH-silenced hTERT-RPE1 cells suggest a regulatory role of FH in interaction
[15]. In both models, major processes involved in with the ubiquitin proteasome system (UPS) and
energy production, glycolysis, and mitochondrial RB1/E2F signaling, adding a new level of com-
respiration are impaired. In addition, a misbal- plexity in the understanding of FH mechanism of
ance in the degree of mitochondria turnover action [23]. Interestingly, and based on studies in
seems to be involved, with an exaggerated other cell types than RPE, it has already been
increase in mitophagy [15, 19]. Moreover, it has suggested that FH has a noncanonical intracellu-
been shown in iPSC-RPE carrying CFH 402H lar function, independent from complement acti-
variant that mitochondria are enlarged and accu- vation. In clear renal cell carcinoma cells and
mulating [20]. Mitochondria are essential for a kidney endothelial cells, FH knockdown impairs
correct response to oxidative stress; therefore, cellular homeostasis, at least partially via NF-kB
damage or dysfunction of these organelles is del- activation [26, 27].
eterious in such an oxidative milieu like the ret- The synopsis of these findings indicates that
ina. Accumulation of oxidized lipids has been understanding the function of FH in a specific
reported in RPE cells under FH dysregulation cell and tissue context is key. In the context of
and induced oxidative stress, when FH function AMD, it is important to advance our understand-
was impaired as a consequence of CFH silencing ing on how RPE cells interact with the neuroret-
12 A. Armento et al.

Fig. 1 Schematic representation of the noncanonical functions of FH in RPE cells

ina once intracellular FH dysregulation or a CFH ment pathway inhibitor strategy to treat AMD
402H high risk variant perturbs intracellular progression may not suffice. Rational re-­balancing
homeostasis within these cells. In this regard, we strategies in a form of combinatorial therapies
have recently established a novel coculture model may be needed to target several drivers of AMD
of RPE and neuroretina. Using this system, we molecular pathology in a systems-­oriented fash-
show that CFH-silenced RPE cells promote reti- ion simultaneously. More specifically, the increase
nal degeneration, which could not be rescued by of alternative complement pathway activity at the
the addition of exogenous FH. Moreover, we Bruch’s membrane/RPE interface may have to be
showed that the damage occurred at the level of considered concomitantly with pro-inflammatory,
mitochondrial activity and lipid oxidation in the oxidative stress-­ related perturbation and meta-
photoreceptors, rather than a canonical inflam- bolic disbalance of RPE and neuroretina to
mation or complement activation [16]. achieve effective future therapeutic regimes for
dry AMD. Last but not least, a very thorough
stratification of patient groups may be needed to
4 Future Perspective move to an individualized and rational therapy for
dry AMD of the future.
Recent works of several groups, including ours,
emphasize a noncanonical and intracellular role
of FH in RPE cells. Besides their reduced capac- References
ity to dampen alternative complement activity,
CFH risk polymorphisms such as Y402H impair 1. Ferris FL 3rd, Wilkinson CP, Bird A, Chakravarthy
U, Chew E, Csaky K, et al. Clinical classification of
RPE homeostasis and are likely contributing to age-related macular degeneration. Ophthalmology.
promote AMD. In consequence, a simple comple- 2013;120(4):844–51.
The Noncanonical Role of Complement Factor H in Retinal Pigment Epithelium (RPE) Cells and… 13

2. van Lookeren Campagne M, LeCouter J, Yaspan BL, porcine retinal explant co-culture model. bioR
Ye W. Mechanisms of age-related macular degen- xiv:2021:2021.07.26.453778.
eration and therapeutic opportunities. J Pathol. 17. Borras C, Canonica J, Jorieux S, Abache T, El
2014;232(2):151–64. Sanharawi M, Klein C, et al. CFH exerts anti-oxidant
3. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng effects on retinal pigment epithelial cells indepen-
CY, et al. Global prevalence of age-related macular dently from protecting against membrane attack com-
degeneration and disease burden projection for 2020 plex. Sci Rep. 2019;9(1):13873.
and 2040: a systematic review and meta-analysis. 18. Krilis M, Qi M, Qi J, Wong JWH, Guymer R,
Lancet Glob Health. 2014;2(2):e106–16. Liew G, et al. Dual roles of different redox forms
4. Armstrong RA, Mousavi M. Overview of risk factors of complement factor H in protecting against age
for age-related macular degeneration (AMD). J Stem related macular degeneration. Free Radic Biol Med.
Cells. 2015;10(3):171–91. 2018;129:237–46.
5. Fritsche LG, Fariss RN, Stambolian D, Abecasis GR, 19. Ebeling MC, Geng Z, Kapphahn RJ, Roehrich H,
Curcio CA, Swaroop A. Age-related macular degen- Montezuma SR, Dutton JR, et al. Impaired mitochon-
eration: genetics and biology coming together. Annu drial function in iPSC-retinal pigment epithelium
Rev Genomics Hum Genet. 2014;15:151–71. with the complement factor H polymorphism for age-­
6. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, related macular degeneration. Cell. 2021;10(4):789.
Haynes C, et al. Complement factor H polymor- 20. Hallam D, Collin J, Bojic S, Chichagova V, Buskin A,
phism in age-related macular degeneration. Science. Xu Y, et al. An induced pluripotent stem cell patient
2005;308(5720):385–9. specific model of complement factor H (Y402H)
7. Day AJ, Willis AC, Ripoche J, Sim RB. Sequence polymorphism displays characteristic features of
polymorphism of human complement factor age-related macular degeneration and indicates a
H. Immunogenetics. 1988;27(3):211–4. beneficial role for UV light exposure. Stem Cells.
8. Armento A, Ueffing M, Clark SJ. The complement 2017;35(11):2305–20.
system in age-related macular degeneration. Cell Mol 21. Ding JD, Kelly U, Groelle M, Christenbury JG, Zhang
Life Sci. 2021;78(10):4487–505. W, Bowes Rickman C. The role of complement dys-
9. Skerka C, Lauer N, Weinberger AA, Keilhauer regulation in AMD mouse models. Adv Exp Med
CN, Suhnel J, Smith R, et al. Defective comple- Biol. 2014;801:213–9.
ment control of factor H (Y402H) and FHL-1 in 22. Armento A, Schmidt TL, Sonntag I, Merle DA,
age-related macular degeneration. Mol Immunol. Jarboui MA, Kilger E, et al. CFH loss in human RPE
2007;44(13):3398–406. cells leads to inflammation and complement system
10. Mullins RF, Russell SR, Anderson DH, Hageman dysregulation via the NF-kappaB pathway. Int J Mol
GS. Drusen associated with aging and age-related Sci. 2021;22(16):8727.
macular degeneration contain proteins common to 23. Merle D, Provenzano F, Jarboui MA, Kilger E, Clark
extracellular deposits associated with atherosclero- S, Deleidi M, et al. mTOR inhibition via Rapamycin
sis, elastosis, amyloidosis, and dense deposit disease. treatment partially reverts the deficit in energy
FASEB J. 2000;14(7):835–46. metabolism caused by FH loss in RPE cells. bioR
11. Heesterbeek TJ, Lechanteur YTE, Lores-Motta L, xiv:2021:2021.10.29.466270.
Schick T, Daha MR, Altay L, et al. Complement 24. Wang Y, Zhang H. Regulation of autophagy by mTOR
activation levels are related to disease stage in signaling pathway. In: Qin Z-H, editor. Autophagy:
AMD. Invest Ophthalmol Vis Sci. 2020;61(3):18. biology and diseases: basic science. Singapore:
12. Park DH, Connor KM, Lambris JD. The challenges Springer; 2019. p. 67–83.
and promise of complement therapeutics for ocular 25. Cerniauskas E, Kurzawa-Akanbi M, Xie L, Hallam D,
diseases. Front Immunol. 2019;10:1007. Moya-Molina M, White K, et al. Complement modu-
13. Strauss O. The retinal pigment epithelium in visual lation reverses pathology in Y402H-retinal pigment
function. Physiol Rev. 2005;85(3):845–81. epithelium cell model of age-related macular degen-
14. Fisher CR, Ferrington DA. Perspective on AMD eration by restoring lysosomal function. Stem Cells
pathobiology: a bioenergetic crisis in the RPE. Invest Transl Med. 2020;9(12):1585–603.
Ophthalmol Vis Sci. 2018;59(4):AMD41–AMD7. 26. Daugan MV, Revel M, Thouenon R, Dragon-Durey
15. Armento A, Honisch S, Panagiotakopoulou V, MA, Robe-Rybkine T, Torset C, et al. Intracellular
Sonntag I, Jacob A, Bolz S, et al. Loss of comple- factor H drives tumor progression independently
ment factor H impairs antioxidant capacity and of the complement cascade. Cancer Immunol Res.
energy metabolism of human RPE cells. Sci Rep. 2021;9:909–25.
2020;10(1):10320. 27. Mahajan S, Jacob A, Kelkar A, Chang A, McSkimming
16. Armento A, Murali A, Marzi J, Arrango-Gonzalez D, Neelamegham S, et al. Local complement factor H
B, Kilger E, Clark SJ, et al. FH loss in RPE cells protects kidney endothelial cell structure and func-
causes retinal degeneration in a human RPE-­ tion. Kidney Int. 2021;100(4):824–36.
Macular Pigment Carotenoids
and Bisretinoid A2E

Ranganathan Arunkumar and Paul S. Bernstein

Abstract Keywords

Lutein (L), zeaxanthin (Z), and meso-­ Abca4 · A2E · Antioxidant · Bisretinoids ·
zeaxanthin (MZ) are the three macular pig- Carotenoids · Lipofuscin · Macular pigments
ments (MP) carotenoids that uniquely · Photo-oxidation · Retina · Retinal pigment
accumulate in the macula lutea region of the epithelium
human retina. L and Z are obtained by humans
through dietary intake. The third MP, MZ, is
rarely present in diet, and its abundance in the 1 Introduction
human fovea is due to the metabolic conver-
sion of dietary L by the retinal pigment epithe- Carotenoids are natural pigments synthesized by
lium’s RPE65 enzyme. The major functions of photosynthetic bacteria, algae, yeast, and plants.
MP in ocular health are to filter high-intensity, L, Z, and MZ are oxygenated carotenoids (xan-
phototoxic blue light and to act as effective thophylls) obtained by humans through dietary
antioxidants for scavenging free radicals. The intake of leafy greens, fruits, and vegetables.
pyridinium bisretinoid, N-retinylidene-N-­ Among the 15 major dietary carotenoids detected
retinylethanolamine (A2E), contributes to in human serum, these three xanthophyll carot-
drusen formation in dry age-related macular enoids selectively accumulate with a unique dis-
degeneration (AMD) and to the autofluores- tribution in the macular region of human retina
cent flecks in autosomal recessive Stargardt [1]. The MP’s total concentration is about 1 mM
disease (STGD1). Retinal carotenoids attenu- at the fovea, and its concentration declines to less
ate A2E formation and can directly and indi- than 10 μM in the peripheral retina [2]. The ratio
rectly alleviate A2E-mediated oxidative of L:Z:MZ in the peripheral retina is about 3:1:0,
damage. In this chapter, we review these more and the concentration of total carotenoids rises
recently recognized interconnections between 100-fold in the macula lutea with a change in the
MP carotenoids and A2E bisretinoids. ratio of L:Z:MZ of about 1:1:1, as measured by
high-performance liquid chromatography
(HPLC) [3]. More recently, high-resolution con-
R. Arunkumar · P. S. Bernstein (*) focal resonance Raman microscopy imaging of
Department of Ophthalmology and Visual Sciences, the human retina from our laboratory has showed
John A. Moran Eye Center, University of Utah that the ratio of Z + MZ:L can be greater than 9:1
School of Medicine, Salt Lake City, UT, USA at the foveal center, while L is more diffusely dis-
e-mail: paul.bernstein@hsc.utah.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 15


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_3
16 R. Arunkumar and P. S. Bernstein

tributed across the macula at a relatively lower aided by conjugated double-bond structure with
concentration [4]. Retinal carotenoids mainly the hydrophilic group on the ionone ring [2]. The
localize to the outer plexiform (Henle fiber) layer absorption maximum of L is around 445 nm, and
and the inner plexiform layer, with axial exten- Z is around 450 nm, corresponding with the haz-
sion from the inner limiting membrane to the ardous blue light range of 430–500 nm.
outer limiting membrane. The specificity in the Depending on the retinal carotenoid concentra-
distribution of retinal carotenoids in the human tion, they are estimated to absorb 40–90% of
retina may be due to the selective uptake of reti- incident short-wavelength, high-energy photo-
nal carotenoids by carotenoid binding proteins toxic blue light [8].
StARD3 (L binding protein) and GSTP1 (Z and Retinal carotenoids with conjugated double
MZ binding protein). The other major factors that bond (C=C) structures are associated with greater
influence the MP carotenoid distributions and singlet oxygen quenching. Z has 11 conjugated
concentrations between individuals are the double bonds and is 50% better at quenching sin-
amount of oral intake of carotenoids and their glet oxygen relative to L with its 10-conjugated
bioavailability [5]. L is present in higher concen- double bonds [9]. MZ, a stereoisomer of Z, with
trations in the diet, but it is converted to MZ by an identical 11-conjugated double-bond structure
the RPE65 enzyme, and the preferential accumu- would be expected to possess the same antioxi-
lation of Z and MZ at the foveal center may be dant properties as Z, but it exhibited better singlet
due to their higher antioxidant potential relative oxygen quenching properties relative to L and Z
to L. The fovea is prone to photo-damage caused [9]. Furthermore, the antioxidant activity of all
by light-induced oxidative stress from reactive three retinal carotenoids as a mixture showed bet-
oxygen species, and singlet oxygen can be gener- ter singlet oxygen quenching ability than any of
ated by photo-oxidation of A2E and other bisreti- the individual MP carotenoids. Retinal carot-
noid components of lipofuscin [6]. A2E acts as a enoids not only quench singlet oxygen, but they
photosensitizer in the presence of short-­ also quench superoxide anion radicals and
wavelength blue light and oxygen that generate hydroxyl radicals, the major causative agents of
reactive oxygen caused by photo-oxidation and lipid peroxidation and light-induced damage [10,
photo-degradation, resulting in retinal degenera- 11]. Generally, retinal carotenoids are better
tion and apoptosis of photoreceptor and RPE hydroxyl radical scavengers than superoxide
cells [7]. Supplementation with retinal carot- anion scavengers, and Z exhibits better hydroxyl
enoids potentially attenuates harmful blue light radical scavenging activity than lutein [10]. MP
and effectively scavenges singlet oxygen and carotenoids protect the retina, which is vulnera-
reactive free radicals in ocular tissues and in bio- ble to oxidative damage caused by exposure to
chemical assays. light, high concentration of oxygen, abundant
photosensitizers, and high concentrations of
polyunsaturated fatty acids (PUFAs).
2 Structure and Function
of the Retinal Carotenoids
3 A2E Bisretinoids
Retinal carotenoids are characterized by the pres-
ence of hydroxyl (O-H) functional groups In the visual cycle, the activation of rhodopsin by
attached at the 3 and 3′ positions of terminal ion- a photon of light during phototransduction results
one rings connected by an isoprenoid backbone in the isomerization of 11-cis-retinal to all-trans-­
structure (Fig. 1). The presence of hydroxyl retinal in photoreceptor outer segments and the
groups and the conjugated double bonds struc- release of all-trans-retinal, which is subsequently
ture determine the light-absorbing properties and reduced to all-trans-retinol and transported to the
antioxidant activities of retinal carotenoids. The RPE cells. In the RPE, the all-trans-retinol is
light filtering function of retinal carotenoids is either stored as a fatty acid ester or is converted
Macular Pigment Carotenoids and Bisretinoid A2E 17

OH

HO

Lutein
OH

HO
Zeaxanthin OH
N
OH

A2E
HO

Meso-Zeaxanthin

Fig. 1 Structures of the macular carotenoids: lutein (L), zeaxanthin (Z), meso-zeaxanthin (MZ), and N-retinylidene-N-­
retinylethanolamine (A2E)

back to an 11-cis-retinoid that is transported back RPE, A2PE is further hydrolyzed to A2E by
to the photoreceptor outer segment, which can phospholipase D inside RPE phagosomes.
bind to opsin to produce a regenerated visual pig- Mutation or dysfunction in the ABCA4 gene
ment. Some all-trans-retinal in the photoreceptor leads to excessive accumulation of A2E and other
outer segments reacts with phosphatidylethanol- bisretinoids in RPE lipofuscin, resulting in reti-
amine (PE) to form N-retinylidene-PE (NRPE), nal cell death and vision loss. A2E is reported to
which is transported or “flipped” by the ATP-­ accumulate with aging and dry AMD and in
binding cassette, subfamily A, member 4 STGD1 [13].
(ABCA4) protein present in photoreceptor outer A2E (C42H58ON; molecular weight, 592) is a
segments. ABCA4 translocates NRPE across the well-characterized component of lipofuscin. It
lipid bilayer from the luminal side to the cyto- exhibits absorbance in both the UV and visible
plasmic side of the disc membrane. NRPE regions of the spectrum [A2E: absorbance max-
released on the cytoplasmic side is hydrolyzed ima (λmax) are 440 and 340 nm, and iso-A2E’s
into all-trans-retinal and reduced to all-trans-­ λmax are 430 and 340 nm]. A2E’s hydrophobic
retinol by retinol dehydrogenases (RDHs) [12]. If side arms and positively charged hydrophilic
ABCA4 does not effectively translocate NRPE, it head group confer an amphiphilic structure
reacts with one more molecule of all-trans-­retinal (Fig. 1). A2E has a detergent-like structure that
to form a toxic bisretinoid, dihydropyridinium-­ may influence membrane properties and inhibit
A2PE. Dihydropyridinium-A2PE is further the lysosomal degradation of lipids. A2E induces
oxidized either to A2-dihydropyridine-PE
­ loss of membrane integrity and perturbs mem-
(A2-DHP-PE) by eliminating one hydrogen or to brane stability, and it inhibits cytochrome C oxy-
phosphatidylethanolamine-pyridinium bisreti- genase and the ATP-driven proton pump. A2E
noid (A2PE) by eliminating two hydrogens in the photo-oxidation products can activate the com-
acidic and oxidizing environment (Fig. 2). During plement system and cause inflammation and
phagocytosis of photoreceptor outer segments by DNA damage [12, 13].
18 R. Arunkumar and P. S. Bernstein

Fig. 2 The visual cycle and bisretinoid formation in pho- hydrogens generates A2PE, and loss of one hydrogen gen-
toreceptor outer segments and RPE. When a photon of erates A2-DHP-PE; phosphate hydrolysis of the latter
light is captured by the visual chromophore, its 11-cis-­ produces A2-DHP-E. A2E, lysoA2PE, and A2-GPE are
retinal (11-cis-RAL) Schiff base chromophore photoi- produced from A2PE. Reaction of the all-trans-RAL
somerizes to all-trans-retinal (all-trans-RAL). The dimer with PE with the formation of a Schiff base linkage
released all-trans-RAL from opsin is reduced to all-trans-­ generates all-trans-retinal dimer-PE (atRALdi-PE), and
retinol (all-trans-ROL) by retinol dehydrogenases phosphate hydrolysis of the latter yields all-trans-retinal
(RDHs). Alternatively, some all-trans-RAL reacts with dimer-ethanolamine (atRALdi-E). All-trans-ROL is
phosphatidylethanolamine (PE) in the photoreceptor outer transported into RPE cells where all-trans-ROL is esteri-
segments to form N-retinylidene-PE (NRPE), which is fied to fatty acids to make all-trans retinyl esters (all-­
transported by ABCA4. The bisretinoid synthesis path- trans-­RE) by the enzyme lecithin retinol acyl transferase
way (red) is initiated when NRPE, rather than hydrolyzing (LRAT). All-trans-RE is converted to 11-cis retinol
to all-trans-ROL and PE, reacts with a second molecule of (11-cis-ROL) by the RPE65 isomerohydrolase. 11-cis
retinaldehyde. A multistep pathway leads to the formation retinol dehydrogenase (11cRDH) oxidizes 11-cis-ROL to
of the intermediate dihydropyridinium-A2PE. Auto-­ 11-cis-RAL which enters the photoreceptor outer seg-
oxidation of dihydropyridinium-A2PE with loss of two ments for the regeneration of opsin

4 Retinal Carotenoids showed additional peaks at m/z 608, 624, 640,


Attenuate Bisretinoids 656, 672, and 688, as well as 1239, 1255, 1271,
in Various Systems and 1286. The additional higher m/z peaks are
photo-oxidation products of A2E and A2PE
4.1 In Vitro formed by the insertion of oxygen atoms at the
C=C bonds along the side arms of A2E and
A2E and its precursor A2PE can be irradiated A2PE. When A2E and A2PE are irradiated at
with 430 nm blue light and analyzed by fast atom 430 nm in the presence of L and Z, the additional
bombardment mass spectroscopy (FAB-MS). photo-oxidized peaks are absent [14]. Singlet
The FAB-MS spectra after irradiation not only oxygen quenching abilities of retinal carotenoids
showed a molecular ion peak at m/z 592 and 1223 were studied in the presence of endoperoxide
attributable to A2E and A2PE, but they also 1,4-dimethylnapthalene (1 mM), which releases
Macular Pigment Carotenoids and Bisretinoid A2E 19

singlet oxygen with a half-life time of 5 h. @ 3.1-fold in the zeaxanthin-supplemented


25 °C. The endoperoxide was incubated with group. A2E levels increased sixfold in the
A2E (500 μM) with and without retinal carot- unsupplemented control group, whereas there
enoids (500 μM to 4 mM). A2E oxidation was was minimal increase in A2E in the MP carot-
measured using HPLC, and it was found that reti- enoid supplemented group, and their A2E lev-
nal carotenoids effectively quench singlet oxygen els were significantly lower than the control
products in a concentration-dependent manner. group [16].
Furthermore, Z was observed to be a better sin-
glet oxygen quencher than L [14].
4.5 Rodent Model

4.2 Cell Culture Until recently, the protective effects of macular


carotenoids against bisretinoids had not been
The protective effects of retinal carotenoids on studied in small laboratory mammals because
photo-oxidation of A2E were studied in ARPE-­ non-primates do not accumulate substantial
19 cells grown in Dulbecco’s Modified Eagle amounts of carotenoids in ocular tissues due to
Medium (DMEM) medium with 10 μM A2E for the presence of very active carotenoid cleavage
10 days. The cells accumulated A2E and were enzymes (Bco1 and Bco2). This led us to cross
incubated with L or Z (10 μM), and they were our Bco2−/− “macular pigment mice” that accu-
then exposed to blue light (430 nm). Cells incu- mulate dietary MP carotenoids in their retinas
bated with retinal carotenoids completely attenu- with a mouse model of STGD that accumulates
ated A2E photo-oxidation and inhibited A2E with increasing age. Abca4−/−/Bco2−/− mice
proteasome inactivation [15]. fed with L or Z have lower levels of A2E and iso-­
A2E compared to the control group with no
carotenoid supplementation, and there was a sta-
4.3 Human Donor Eyes tistically significant inverse correlation between
retinal carotenoids and A2E and iso-A2E levels
A2E concentrations in human cadaver eyes in RPE/choroid (Fig. 3). Furthermore, L and Z
increase with age in both the macula and in the supplementation improved visual performance in
peripheral retina, and A2E levels are threefold Abca4−/−/Bco2−/− mice compared to the control
lower in the macula region compared to the group [13].
peripheral retina despite excess light exposure
and high metabolic activity. Moreover, A2E con-
centrations are inversely related to retinal carot- 5 Summary
enoid concentrations [16]. Lipofuscin and A2E
levels are significantly lower in the fovea region The macular pigment carotenoids are distinct
where retinal carotenoids are present in abun- from other nutrients due to their selective accu-
dance [17]. mulation and unique spatial distribution in the
fovea, the region most vulnerable to excess light
and high metabolic oxygen. While it is widely
4.4 Avian Model appreciated that the beneficial properties of MP
carotenoids are in part mediated by their blue
The association between retinal carotenoids light-absorbing and antioxidant properties, it is
and A2E was studied in Japanese quail, whose now becoming increasingly understood that
retinal carotenoids are present as fatty acid attenuation of A2E formation and oxidation by L,
esters in distinct photoreceptor oil drops. After Z, and MZ are important protective effects as
16 weeks of feeding, total carotenoids rose well, further emphasizing their valuable role as
1.6-fold in the lutein-supplemented group and safe and cost-effective nutritional interventions
20 R. Arunkumar and P. S. Bernstein

lifespan and in prenatal supplementation. J Lipid Res.


2021;62:100038.
6. Kim HJ, Montenegro D, Zhao J, Sparrow
JR. Bisretinoids of the retina: photo-oxidation, iron-­
catalyzed oxidation, and disease consequences.
Antioxidants. 2021;10(9):1382.
7. Ueda K, Zhao J, Kim HJ, Sparrow
JR. Photodegradation of retinal bisretinoids in
mouse models and implications for macular degen-
eration. Proc Natl Acad Sci. 2016;113(25):6904–9.
8. Arunkumar R, Calvo CM, Conrady CD, Bernstein
PS. What do we know about the macular pigment
in AMD: the past, the present, and the future. Eye.
Fig. 3 Distribution of RPE/choroid A2E + iso-A2E levels 2018;32(5):992–1004.
in relation to retinal carotenoids in Abca4−/−/Bco2−/− mice. 9. Li B, Ahmed F, Bernstein PS. Studies on the singlet
There was a statistically significant inverse correlation oxygen scavenging mechanism of human macular
between retinal carotenoids and A2E and iso-A2E levels pigment. Arch Biochem Biophys. 2010;504(1):56–60.
in RPE/choroid (p values: *p < 0.05). Red line refers to L 10. Boehm F, Edge R, Truscott TG. Anti-and pro-­oxidative
with R2 = 0.815, blue line refers to Z with R2 = 0.825, and mechanisms comparing the macular carotenoids zea-
the orange dotted line refers to combined regression plot xanthin and lutein with other dietary carotenoids–a
of L, Z, and placebo group with R2 = 0.904. [13] singlet oxygen, free-radical in vitro and ex vivo study.
Photochem Photobiol Sci. 2020;19(8):1001–8.
11. Widomska J, Gruszecki WI, Subczynski WK. Factors
differentiating the antioxidant activity of macular
against visual loss from age-related and inherited xanthophylls in the human eye retina. Antioxidants.
macular diseases. 2021;10(4):601.
12. Kim HJ, Sparrow JR. Bisretinoid phospholipid and
vitamin A aldehyde: shining a light. J Lipid Res.
Conflict of Interest No authors have any conflicts of 2021;62:100042.
interest. 13. Arunkumar R, Gorusupudi A, Li B, Blount JD,
Grant Support EY-11600 and EY-14800; Research to Nwagbo U, Kim HJ, Sparrow JR, Bernstein
Prevent Blindness. PS. Lutein and zeaxanthin reduce A2E and iso-A2E
levels and improve visual performance in Abca4−/−/
bco2−/− double knockout mice. Exp Eye Res.
2021;209:108680.
References 14. Kim SR, Nakanishi K, Itagaki Y, Sparrow
JR. Photooxidation of A2-PE, a photoreceptor outer
1. Arunkumar R, Gorusupudi A, Bernstein PS. The segment fluorophore, and protection by lutein and
macular carotenoids: a biochemical overview. zeaxanthin. Exp Eye Res. 2006;82(5):828–39.
Biochim Biophys Acta (BBA) Mol Cell Biol Lipids. 15. Bian Q, Gao S, Zhou J, Qin J, Taylor A, Johnson EJ,
2020;1865(11):158617. Tang G, Sparrow JR, Gierhart D, Shang F. Lutein
2. Krinsky NI, Landrum JT, Bone RA. Biologic mecha- and zeaxanthin supplementation reduces photo-
nisms of the protective role of lutein and zeaxanthin in oxidative damage and modulates the expression of
the eye. Annu Rev Nutr. 2003;23(1):171–201. inflammation-­related genes in retinal pigment epithe-
3. Bone RA, Landrum JT, Friedes LM, Gomez lial cells. Free Radic Biol Med. 2012;53(6):1298–307.
CM, Kilburn MD, Menendez E, Vidal I, Wang 16. Bhosale P, Serban B, Bernstein PS. Retinal carot-
W. Distribution of lutein and zeaxanthin stereoisomers enoids can attenuate formation of A2E in the reti-
in the human retina. Exp Eye Res. 1997;64(2):211–8. nal pigment epithelium. Arch Biochem Biophys.
4. Li B, George EW, Rognon GT, Gorusupudi A, 2009;483(2):175–81.
Ranganathan A, Chang FY, Shi L, Frederick JM, 17. Adler L, Boyer NP, Anderson DM, Spraggins JM,
Bernstein PS. Imaging lutein and zeaxanthin in the Schey KL, Hanneken A, Ablonczy Z, Crouch RK,
human retina with confocal resonance Raman micros- Koutalos Y. Determination of N-retinylidene-N-­
copy. Proc Natl Acad Sci. 2020;117(22):12352–8. retinylethanolamine (A2E) levels in central and
5. Bernstein PS, Ranganathan A. The emerging roles peripheral areas of human retinal pigment epithelium.
of the macular pigment carotenoids throughout the Photochem Photobiol Sci. 2015;14(11):1983–90.
Disturbed Matrix
Metalloproteinases Activity
in Age-Related Macular
Degeneration

Beatriz Martins and Rosa Fernandes

Abstract ulation and TIMPs’ significant regulatory


functions.
Matrix metalloproteinases (MMPs) are a
tightly regulated family of proteolytic Keywords
enzymes that break down extracellular matrix
(ECM) and basement membrane components. Age-related macular degeneration · Matrix
Because it is associated with development, metalloproteinases · Extracellular matrix ·
morphogenesis, tissue remodeling, and repair, Bruch’s membrane · Geographic atrophy ·
ECM remodeling is an important mechanism. Choroidal neovascularization
MMPs are thought to act as a double-edged
sword, as they contribute to maintaining pho-
toreceptors/retinal pigment epithelium (RPE)/ 1 Introduction
Bruch’s membrane (BM)/choroid complex
homeostasis and also contribute to the onset Matrix metalloproteinases (MMPs), also known
and progression of age-related macular degen- as matrixins, belong to an important class of
eration (AMD). Polymorphisms and/or altered zinc-dependent endopeptidases, the metzincins
expression in MMPs and their tissue inhibitors superfamily [1]. This superfamily includes 21
(TIMPs) are associated with age-related mac- human disintegrins and metalloproteinase
ular degeneration (AMD). Here, we review domain (ADAMs) and 19 secreted disintegrin
the evidence for MMPs’ role in the onset and and metalloproteinase thrombospondin domain
progression of AMD via addressing their reg- (ADAMTSs), in addition to 23 MMPs expressed

B. Martins R. Fernandes (*)


Faculty of Medicine, Coimbra Institute for Clinical Faculty of Medicine, Coimbra Institute for Clinical
and Biomedical Research (iCBR), University of and Biomedical Research (iCBR), University of
Coimbra, Coimbra, Portugal Coimbra, Coimbra, Portugal
Institute of Pharmacology and Experimental Institute of Pharmacology and Experimental
Therapeutics, Faculty of Medicine, University of Therapeutics, Faculty of Medicine, University of
Coimbra, Coimbra, Portugal Coimbra, Coimbra, Portugal
Association for Innovation and Biomedical Research
on Light and Image (AIBILI), Coimbra, Portugal
e-mail: rcfernandes@fmed.uc.pt

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 21


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_4
22 B. Martins and R. Fernandes

in human tissue [2]. MMPs typically have an The specific interplay of pathophysiological
80-amino acid propeptide, a 170-amino acid cat- events that converge to AMD and related degen-
alytic metalloproteinase domain, a variable-­ eration mechanisms remain to be fully elucidated
length linker peptide (hinge region), and a [11]. Nevertheless, several studies have been
200-amino acid hemopexin (Hpx) domain. A highlighting the alterations in the regulation of
detailed description of MMPs structure and an ECM, by dysregulation of the activity of MMPs
overview of their substrate preferences and their and TIMPs, as one of the main mechanisms
association with extracellular matrix (ECM) com- involved in the development of AMD [12]. This
ponents and inhibitors can be found in other modulation of ECM turnover is associated with
excellent reports [2–4]. the other molecular mechanisms involved in the
The MMPs are usually secreted as zymogens pathophysiology of the disease since some stud-
by a variety of cells into the extracellular space ies have shown that both oxidative stress and acti-
[3] or stay tethered to their plasma membrane [5]. vation of the complement system can modulate
In humans, there are six membrane-type (MT)- the activity of ECM components [13, 14].
MMPs [5]. In the generation of active MMPs, Additionally, the formation of drusen, the hall-
enzymatic cleavages of the zymogens are needed. mark of AMD, is also believed to be associated
Active MMP-2, for example, is made from pro-­ with a dysregulation of the ECM components,
MMP-­2 by MMP-14 cleavage, while pro-MMP-9 especially in the RPE and Bruch’s membrane
is cleaved by MMP-3 [5, 6]. (BM) [15]. These alterations in ECM modulation
MMPs are mainly classified based on their caused by dysregulation of MMP/TIMP complex
substrate preference and domain organization, can also be influenced not only by environmental
being grouped into gelatinases (MMP-2, MMP-­ but also by genetic factors. For example, poly-
9), collagenases (MMP-1, MMP-8, MMP-13), morphisms in MMP-2 (rs243865) and in MMP-9
stromelysins (MMP-3, MMP-10), MT-MMPs (rs3918424 and rs3918241) are associated with
(MMP-14, MMP-15, MMP-16, MMP-17), and increased risk of AMD, mainly in younger
others [7]. Under normal conditions, both MMPs patients (<65 years) [16–18]. Other MMP-2
and the tissue inhibitors of metalloproteinases polymorphisms (rs243865, rs243866, and
(TIMPs) have an important role in the regulation rs2287074) are associated with a lower likeli-
of the turnover of ECM [2, 8]. A dysregulation of hood of AMD development [19, 20]. There are
these components can both contribute to patho- also some TIMPs polymorphisms associated
logical ECM remodeling and serve as a nexus for with AMD. Despite some polymorphisms in
a variety of pathways involved in age-related TIMP-2 (rs817909) and in TIMP-3 (rs5754227)
macular degeneration (AMD) pathogenesis. appear to have a protective effect, other polymor-
phisms in TIMP-3 (rs713685 and rs743751) are
more frequent in AMD patients [18, 21, 22]. As a
2 MMPs Are Implicated in AMD result, these findings highlight evidence that
changes in ECM turnover are essential mecha-
AMD is a retinal degenerative disease that is a nisms implicated in the development of AMD
leading cause of central vision loss in the elderly. and may help explain the susceptibility to the dis-
This disease, in its early stages, is characterized ease’s late stages.
by the accumulation of drusen that causes pro-
gressive degeneration of the photoreceptors and
retinal pigment epithelium (RPE). The disease 2.1 MMPs in Dry AMD
can progress to geographic atrophy (GA), an
advanced form of dry AMD, and/or choroidal BM is a five-layered ECM located at the inter-
neovascularization (CNV), also known as wet face of the retina and choriocapillaris. BM plays
AMD [9, 10]. an important role in cell-cell communication and
Disturbed Matrix Metalloproteinases Activity in Age-Related Macular Degeneration 23

regulates the exchange of oxygen and nutrients oxygen supply to the outer retina. The oxidative
between the choroid and the outer retina [23]. and inflammatory processes that occur in late
However, BM undergoes a number of age-related stages of AMD might promote a pro-angiogenic
alterations that may have a role in AMD patho- environment, resulting in CNV, which is the hall-
genesis. Especially noteworthy, the accumulation mark of wet AMD. Neovessels from choroid,
of cellular debris and ECM proteins leads BM to also referred as CNV, develop beneath the RPE
thicken and to the formation of drusen [24]. or penetrate into the subretinal space in patients
Several studies have been demonstrated that with wet AMD [38, 39].
aging causes structural and functional changes in Similar to GA and BM thickness, in this case
the BM due to a decrease in the activity of the also dysregulation of ECM turnover appears to
gelatinase components of the MMP system. In have a close relation with CNV. Increased activ-
this case, increased levels of high molecular ity of MMPs can degrade some components of
weight components of the MMPs pathway BM, essential to the growth of new vessels [40,
(HMW1 and HMW2) reduce the pool of the acti- 41]. Several studies have shown that patients with
vated gelatinases MMP-2 and MMP-9 and con- wet AMD present increased levels of MMP-9 in
tribute to reduced matrix degradation and aqueous humor, vitreous, and plasma [42–44].
thickness of BM [25–27]. Another study revealed This increase in the levels of MMP-9 contributes
that higher TIMP-3 concentrations in the drusen to CNV since this MMP can increase VEGF lev-
block MMPs activity, lowering proteolytic activ- els (pro-angiogenic factor) by decreasing PEDF
ity within the drusen [28]. Oxidative stress levels (anti-angiogenic factor), both secreted by
reduces MMP-2 activity and leads to the forma- the RPE [45]. MMP-2, MMP-7, and MMP-13
tion of sub-RPE deposits that can be involved in have likewise shown a rise in their expression and
the formation of drusen during AMD develop- levels in patients with CNV lesions [18, 46, 47].
ment [29]. Moreover, cellular and animal models of CNV
Nevertheless, several other studies also lesions reported increased expression of MMP-2
detected increased levels of other MMPs in and MMP-9, which were involved in the forma-
patients with GA. MMP-7 levels were found to tion of experimental CNV. Additionally, the inhi-
be lower in these patients, but MMP-9 and bition of these two gelatinases appears to be a
TIMP-1 levels were higher [30, 31]. In vitro and good strategy to treat CNV, since their elimina-
in vivo models of dry AMD led to increased tion is associated with decreased CNV lesion size
expression, secretion, and activation of MMP-1, [48–50]. Some studies have also been underlin-
MMP-3, and MMP-9 and are associated with ing the alterations in TIMPs activity as an impor-
decreased RPE cell viability [32–34]. Moreover, tant mechanism underlying CNV. On both wet
MMP-9’s role in the breakdown of the RPE bar- AMD patients and animal models, decreased lev-
rier has been highlighted, as silencing of this els of TIMPs (TIMP-2 and TIMP-3) contribute to
MMP can improve the barrier integrity [35, 36]. CNV susceptibility [18, 51].
Regarding genetic alterations, only a few stud- Regarding genetic alterations, several studies
ies have related polymorphisms specifically with identified various polymorphisms on both MMPs
dry AMD. Liutkeviciene et al. detected that a and TIMPs that are related to CNV and wet
polymorphism in the MMP-2 gene (rd243865) is AMD. Microsatellites in the promoter of MMP-­
associated with the development of hard drusen 9, along with other polymorphisms on the
in AMD patients [37]. MMP-9 gene (rs142450006, rs3918424), have
been associated with increased risk of CNV pro-
gression and wet AMD development [17, 52, 53].
2.2 MMPs in Wet AMD Also polymorphisms on MMP-1 (rs1799750),
MMP-2 (rs243865), MMP-7 (rs11568818), and
The choroid is a network of blood vessels, located MMP-20 (rs10895322) are associated with
below the BM that is critical for nutrients and increased CNV lesion size and development of
24 B. Martins and R. Fernandes

wet AMD [54–56]. Finally, a polymorphism in 8. Agren MS, Auf dem Keller U. Matrix metallo-
proteinases: how much can they do? Int J Mol Sci.
the TIMP-3 gene (rs9621532) has a protective 2020;21(8):2678.
effect and is associated with decreased risk of 9. Mitchell P, Liew G, Gopinath B, Wong
wet AMD [57]. TY. Age-related macular degeneration. Lancet.
2018;392(10153):1147–59.
10. Al-Zamil WM, Yassin SA. Recent developments
in age-related macular degeneration: a review. Clin
3 Concluding Remarks Interv Aging. 2017;12:1313–30.
11. Ambati J, Fowler BJ. Mechanisms of age-related
Due to their ability to break down ECM constitu- macular degeneration. Neuron. 2012;75(1):26–39.
12. Peng H, Hulleman JD. Prospective application of
ents, MMPs are relevant components in many activity-based proteomic profiling in vision research-­
pathological processes of AMD. Specifically, potential unique insights into ocular protease biology
they play an important role in the accumulation and pathology. Int J Mol Sci. 2019;20(16):3855.
of drusen and degeneration of photoreceptors/RPE 13. Klettner A. Oxidative stress induced cellular sig-
naling in RPE cells. Front Biosci (Schol Ed).
in dry AMD and pathological vessel growth in 2012;4:392–411.
wet AMD. TIMPs control changes in their 14. Fernandez-Godino R, Pierce EA. C3a triggers for-
expression and activity. To better understand the mation of sub-retinal pigment epithelium depos-
role of each MMP in AMD pathogenesis, further its via the ubiquitin proteasome pathway. Sci Rep.
2018;8(1):9679.
research is needed. 15. Luibl V, Isas JM, Kayed R, Glabe CG, Langen R,
Chen J. Drusen deposits associated with aging and
Acknowledgments Funding provided by the Global age-related macular degeneration contain nonfibrillar
Ophthalmology Awards Program (GOAP), a Bayer spon- amyloid oligomers. J Clin Invest. 2006;116(2):378–85.
sored initiative committed to supporting ophthalmic 16. Liutkeviciene R, Lesauskaite V, Zaliaduonyte-­
research across the world. Thanks are also due to FCT Peksiene D, et al. Role of MMP-2 (-1306 C/T)
(Portugal) and Strategic Projects UIDB/04539/2020 and polymorphism in age-related macular degeneration.
UIDP/04539/2020 (CIBB), COMPETE-FEDER (POCI- Ophthalmic Genet. 2016;37(2):170–6.
01-0145-FEDER-007440), and Centro 2020 Regional 17. Liutkeviciene R, Lesauskaite V, Sinkunaite-­
Operational Program: BRAINHEALTH 2020 Marsalkiene G, et al. The role of matrix metallo-
(CENTRO-01-0145- FEDER-­000008). The authors also proteinases polymorphisms in age-related macular
acknowledge FCT for the doctoral research grant degeneration. Ophthalmic Genet. 2015;36(2):149–55.
2020.04811.BD (to B.M.). 18. Oszajca K, Szemraj M, Szemraj J, Jurowski
P. Association analysis of genetic polymorphisms and
expression levels of selected genes involved in extra-
cellular matrix turnover and angiogenesis with the
References risk of age-related macular degeneration. Ophthalmic
Genet. 2018;39(6):684–98.
1. Goodwin L. A closer look at metalloproteinases. 19. Cheng J, Hao X, Zhang Z. Risk of macular degen-
Hauppauge: Nova Science Publisher Inc; 2019. 310 p eration affected by polymorphisms in Matrix
2. Laronha H, Caldeira J. Structure and function of human metalloproteinase-2: a case-control study in
matrix metalloproteinases. Cell. 2020;9(5):1076. Chinese Han population. Medicine (Baltimore).
3. Cui N, Hu M, Khalil RA. Biochemical and biological 2017;96(47):e8190.
attributes of matrix metalloproteinases. Prog Mol Biol 20. Seitzman RL, Mahajan VB, Mangione C, et al.
Transl Sci. 2017;147:1–73. Estrogen receptor alpha and matrix metallo-
4. Maskos K. Crystal structures of MMPs in complex proteinase 2 polymorphisms and age-related
with physiological and pharmacological inhibitors. maculopathy in older women. Am J Epidemiol.
Biochimie. 2005;87(3-4):249–63. 2008;167(10):1217–25.
5. Itoh Y. Membrane-type matrix metalloprotein- 21. Fritsche LG, Igl W, Bailey JN, et al. A large genome-­
ases: their functions and regulations. Matrix Biol. wide association study of age-related macular degen-
2015;44–46:207–23. eration highlights contributions of rare and common
6. Wilcock DM, Morgan D, Gordon MN, et al. Activation variants. Nat Genet. 2016;48(2):134–43.
of matrix metalloproteinases following anti-Abeta 22. Kaur I, Rathi S, Chakrabarti S. Variations in TIMP3
immunotherapy; implications for microhemorrhage are associated with age-related macular degeneration.
occurrence. J Neuroinflammation. 2011;8:115. Proc Natl Acad Sci U S A. 2010;107(28):E112–3.
7. Nagase H, Visse R, Murphy G. Structure and function 23. Booij JC, Baas DC, Beisekeeva J, Gorgels TG, Bergen
of matrix metalloproteinases and TIMPs. Cardiovasc AA. The dynamic nature of Bruch’s membrane. Prog
Res. 2006;69(3):562–73. Retin Eye Res. 2010;29(1):1–18.
Disturbed Matrix Metalloproteinases Activity in Age-Related Macular Degeneration 25

24. Murali A, Krishnakumar S, Subramanian A, 37. Liutkeviciene R, Lesauskaite V, Sinkunaite-­


Parameswaran S. Bruch’s membrane pathol- Marsalkiene G, et al. MMP-2 Rs24386 (C--
ogy: a mechanistic perspective. Eur J Ophthalmol. >T) gene polymorphism and the phenotype of
2020;30(6):1195–206. age-related macular degeneration. Int J Ophthalmol.
25. Hussain AA, Lee Y, Marshall J. Understanding the 2017;10(9):1349–53.
complexity of the matrix metalloproteinase system 38. Kur J, Newman EA, Chan-Ling T. Cellular and physi-
and its relevance to age-related diseases: age-related ological mechanisms underlying blood flow regula-
macular degeneration and Alzheimer’s disease. Prog tion in the retina and choroid in health and disease.
Retin Eye Res. 2020;74:100775. Prog Retin Eye Res. 2012;31(5):377–406.
26. Hussain AA, Lee Y, Marshall J. High molecu- 39. Grossniklaus HE, Green WR. Choroidal neovascular-
lar-weight gelatinase species of human Bruch’s ization. Am J Ophthalmol. 2004;137(3):496–503.
membrane: compositional analyses and age- 40. Cabral T, Mello LGM, Lima LH, et al. Retinal and
related changes. Invest Ophthalmol Vis Sci. choroidal angiogenesis: a review of new targets. Int J
2010;51(5):2363–71. Retina Vitreous. 2017;3:31.
27. Hussain AA, Lee Y, Zhang JJ, Marshall J. Disturbed 41. Zeng J, Jiang D, Liu X, Zhu X, Tang L. Matrix metal-
matrix metalloproteinase activity of Bruch’s mem- loproteinases expression in choroidal neovascular
brane in age-related macular degeneration. Invest membranes. Yan Ke Xue Bao. 2004;20(3):191–3.
Ophthalmol Vis Sci. 2011;52(7):4459–66. 42. Kim J, Kim JH, Do JY, et al. Key role of microglial
28. Leu ST, Batni S, Radeke MJ, Johnson LV, Anderson matrix metalloproteinases in choroidal neovascular-
DH, Clegg DO. Drusen are cold spots for proteolysis: ization. Front Cell Neurosci. 2021;15:638098.
expression of matrix metalloproteinases and their tis- 43. Ecker SM, Pfahler SM, Hines JC, Lovelace AS,
sue inhibitor proteins in age-related macular degen- Glaser BM. Sequential in-office vitreous aspirates
eration. Exp Eye Res. 2002;74(1):141–54. demonstrate vitreous matrix metalloproteinase 9 lev-
29. Marin-Castano ME, Csaky KG, Cousins els correlate with the amount of subretinal fluid in
SW. Nonlethal oxidant injury to human retinal pig- eyes with wet age-related macular degeneration. Mol
ment epithelium cells causes cell membrane blebbing Vis. 2012;18:1658–67.
but decreased MMP-2 activity. Invest Ophthalmol Vis 44. Chau KY, Sivaprasad S, Patel N, Donaldson TA,
Sci. 2005;46(9):3331–40. Luthert PJ, Chong NV. Plasma levels of matrix
30. Lashkari K, Teague GC, Beattie U, et al. Plasma bio- metalloproteinase-2 and -9 (MMP-2 and MMP-9)
markers of the amyloid pathway are associated with in age-related macular degeneration. Eye (Lond).
geographic atrophy secondary to age-related macular 2008;22(6):855–9.
degeneration. PLoS One. 2020;15(8):e0236283. 45. Pouw AE, Greiner MA, Coussa RG, et al. Cell-matrix
31. Krogh Nielsen M, Subhi Y, Rue Molbech C, Nilsson interactions in the eye: from cornea to choroid. Cell.
LL, Nissen MH, Sorensen TL. Imbalances in tissue 2021;10(3):687.
inhibitors of metalloproteinases differentiate choroi- 46. Lecomte J, Louis K, Detry B, et al. Bone marrow-­
dal neovascularization from geographic atrophy. Acta derived mesenchymal cells and MMP13 contribute to
Ophthalmol. 2019;97(1):84–90. experimental choroidal neovascularization. Cell Mol
32. Di Marco S, Carnicelli V, Franceschini N, et al. Life Sci. 2011;68(4):677–86.
Saffron: a multitask neuroprotective agent for reti- 47. Yazama F, Kadonosono K, Itoh N, Ohno S. Role of
nal degenerative diseases. Antioxidants (Basel). matrix metalloproteinase-7 in angiogenesis associated
2019;8(7):224. with age-related macular degeneration. J Electron
33. Kernt M, Walch A, Neubauer AS, et al. Filtering blue Microsc. 2002;51(2):127–31.
light reduces light-induced oxidative stress, senes- 48. Lambert V, Wielockx B, Munaut C, et al. MMP-2 and
cence and accumulation of extracellular matrix pro- MMP-9 synergize in promoting choroidal neovascu-
teins in human retinal pigment epithelium cells. Clin larization. FASEB J. 2003;17(15):2290–2.
Exp Ophthalmol. 2012;40(1):e87–97. 49. Berglin L, Sarman S, van der Ploeg I, et al. Reduced
34. Sanchez-Ramos C, Vega JA, del Valle ME, Fernandez-­ choroidal neovascular membrane formation in
Balbuena A, Bonnin-Arias C, Benitez-del Castillo matrix metalloproteinase-2-deficient mice. Invest
JM. Role of metalloproteases in retinal degeneration Ophthalmol Vis Sci. 2003;44(1):403–8.
induced by violet and blue light. Adv Exp Med Biol. 50. Lambert V, Munaut C, Jost M, et al. Matrix metallo-
2010;664:159–64. proteinase-­9 contributes to choroidal neovasculariza-
35. Cao L, Wang H, Wang F, Xu D, Liu F, Liu tion. Am J Pathol. 2002;161(4):1247–53.
C. Abeta-­ induced senescent retinal pigment epi- 51. Ebrahem Q, Qi JH, Sugimoto M, et al. Increased
thelial cells create a proinflammatory microen- neovascularization in mice lacking tissue inhibitor
vironment in AMD. Invest Ophthalmol Vis Sci. of metalloproteinases-3. Invest Ophthalmol Vis Sci.
2013;54(5):3738–50. 2011;52(9):6117–23.
36. Cao L, Wang H, Wang F. Amyloid-beta-induced 52. Fiotti N, Pedio M, Battaglia Parodi M, et al. MMP-9
matrix metalloproteinase-9 secretion is associated microsatellite polymorphism and susceptibility to
with retinal pigment epithelial barrier disruption. Int exudative form of age-related macular degeneration.
J Mol Med. 2013;31(5):1105–12. Genet Med. 2005;7(4):272–7.
26 B. Martins and R. Fernandes

53. Lauwen S, Lefeber DJ, Fauser S, Hoyng CB, den neovascular lesion size in age-related macular
Hollander AI. Increased pro-MMP9 plasma levels degeneration. Ophthalmology. 2015;122(11):2295–
are associated with neovascular age-related macular 302 e2.
degeneration and with the risk allele of rs142450006 56. Liutkeviciene R, Vilkeviciute A, Borisovaite D,
near MMP9. Mol Vis. 2021;27:142–50. Miniauskiene G. Association of exudative age-related
54. Budiene B, Liutkeviciene R, Gustiene O, et al. The macular degeneration with matrix metalloproteinases-
association of matrix metalloproteinases poly- ­2 (-1306 C/T) rs243865 gene polymorphism. Indian J
morphisms and interleukins in advanced age-­ Ophthalmol. 2018;66(4):551–7.
related macular degeneration. Ophthalmic Genet. 57. Ardeljan D, Meyerle CB, Agron E, et al. Influence of
2018;39(4):463–72. TIMP3/SYN3 polymorphisms on the phenotypic pre-
55. Akagi-Kurashige Y, Yamashiro K, Gotoh N, et al. sentation of age-related macular degeneration. Eur J
MMP20 and ARMS2/HTRA1 are associated with Hum Genet. 2013;21(10):1152–7.
Current Views on Chr10q26
Contribution to Age-Related
Macular Degeneration

Navdeep Gogna, Lillian F. Hyde, Gayle B. Collin,


Lisa Stone, Jurgen K. Naggert,
and Patsy M. Nishina

Abstract 1 Introduction

Age-related macular degeneration (AMD) is Age-related macular degeneration (AMD) is the


the leading cause of blindness in the global leading cause of irreversible central vision loss in
aging population. Familial aggregation and aging populations [1, 2]. By 2050, an estimated
genome-wide association (GWA) studies have 22 million people in the USA [3], and, by 2040,
identified gene variants associated with AMD, 288 million people worldwide [4], will be
implying a strong genetic contribution to affected with AMD. Early stages of AMD are
AMD development. Two loci, on human Chr characterized by the presence of few (<20)
1q31 and 10q26, respectively, represent the medium-sized drusen and/or retinal pigment epi-
most influential of all genetic factors. While thelium (RPE) abnormalities [5–7]. Drusen,
the role of CFH at Chr 1q31 is well estab- lipid-rich, protein-containing deposits that accu-
lished, uncertainty remains about the genes mulate between the RPE and Bruch’s membrane,
ARMS2 and HTRA1, at the Chr 10q26 locus. are considered a “hallmark” of AMD [8].
Since both genes are in strong linkage disequi- Intermediate AMD exhibits at least one large
librium, assigning individual gene effects is druse, multiple medium-size drusen, or geo-
difficult. In this chapter, we review current lit- graphic atrophy (GA) that does not extend to the
erature about ARMS2 and HTRA1 and their center of the macula [5]. Advanced or late AMD
relevance to AMD risk. Future studies will be is either non-neovascular (dry, atrophic, or non-
necessary to unravel the mechanisms by which exudative) or neovascular (wet or exudative) [9].
they contribute to AMD. Dry AMD, characterized by drusen and GA,
extends to macula’s center and affects the chorio-
Keywords capillaris, RPE cells, and photoreceptors (PRs)
without leakage of blood or serum into the retina
AMD · ARMS2 · HTRA1 · Linkage disequi-
and macula [10]. In contrast, wet AMD is associ-
librium · Genetics
ated with RPE detachment and choroidal neovas-
cularization, leading to leakage and fibrovascular
scarring [11].
AMD is a complex, multifactorial disease
N. Gogna (*) · L. F. Hyde · G. B. Collin · L. Stone · [12–24]. Risk factors identified from case-­
J. K. Naggert · P. M. Nishina control, cross-sectional, and prospective cohort
The Jackson Laboratory, Bar Harbor, ME, USA studies [10, 25–34] include age [35], family his-
e-mail: navdeep.gogna@jax.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 27


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_5
28 N. Gogna et al.

tory [28, 36], dietary choices [30, 37–40], race 2.1 Reported Tissue, Cellular
and ethnicity [41–43], gender [21], and smoking and Subcellular Expression
[25, 44–46]. In addition, genetic factors explain of ARMS2/ARMS2
46–71% of the variation in overall disease sever-
ity [47]. To date, a total of 52 independently asso- The expression of ARMS2 was detected in pla-
ciated common and rare gene variants cental tissue [55], at low levels in retinal samples
(p < 5 × 10−8), distributed across 34 loci, have [56, 73] and later, ubiquitously, in human tissues
been associated with AMD risk [48–51]. [77]. The Bgee gene expression database (v14.2)
Genome-wide association (GWA) and family-­ currently lists 62 distinct human tissues with
based linkage studies identified genetic variants varying ARMS2 mRNA levels [78].
in the complement factor H (CFH) gene on Kanda et al. expressed human retinal ARMS2
Chr1q32 and two tightly linked genes—ARMS2 mRNA in COS-1, ARPE-19, and JEG cells.
and HTRA1—on Chr10q26 as major contributors Subcellular fractionation, followed by co-­staining
to AMD risk [52–65]. Together, these increase with MitoTracker and cytochrome C oxidase
AMD predisposition by more than 40-fold [66]. subunit IV, located ARMS2 to the mitochondrial
While significant progress has been made toward (Mt) outer membrane [73]. Subsequently, another
understanding the role of CFH in AMD risk [67– group co-stained ARMS2 with retinoschisin (an
70], uncertainty remains regarding the gene(s) extracellular protein that stains PR inner seg-
responsible at the Chr10q26 locus. Strong link- ments) and Mt cytochrome c oxidase subunit II in
age disequilibrium between ARMS2 and HTRA1 human retinal sections. ARMS2 localized in the
genes has made it difficult to distinguish individ- Mt-enriched ellipsoid region of the inner seg-
ual gene effects by statistical methods [71]. ments of both rod and cone PR cells [79].
Moreover, PLEKHA1 is also found in linkage However, conflicting results were presented by
disequilibrium with ARMS2 [55]. Several studies Wang et al. [80]. Using immunofluorescence
suggest that ARMS2/HTRA1 genetic variations (IF), verified by immunoblotting, endogenous
are more strongly associated with AMD risk than ARMS2 in ARPE-19 cells and overexpressed
PLEKHA1 [56, 72, 73]. In this chapter, we review GFP-tagged and HIS-tagged exogenous ARMS2
existing information about ARMS2 and HTRA1 in COS7 cells were found to localize in the cyto-
and their association with AMD. sol and not in Mt or any other organelle [80]. The
group further performed in silico analyses to
identify the protein features required for Mt tar-
2 
ARMS2 geting and concluded that ARMS2 lacked canon-
ical mitochondrial targeting sequences [80].
The ARMS2 gene, age-related maculopathy sus- Kortvely et al. transfected HEK293 cells with
ceptibility 2, is only present in higher primates plasmid constructs coding for normal or risk vari-
[74]. ARMS2 encodes an 11-kDa protein [75]; ant ARMS2. Immunostaining findings suggested
however the function, localization, and native that ARMS2 is a secreted protein and a compo-
structure of the protein have not been firmly nent of the extracellular matrix (ECM), found
established. An in silico approach to predict the mainly in choroid pillars of human eye [81].
structure of ARMS2 concluded that it has 15 Using a yeast two-hybrid system, followed by
putative phosphorylation sites, four putative gly- coprecipitation, ARMS2 was found to bind sev-
cation sites of ε-amino groups of lysine, two eral ECM proteins [81]. Further, ARMS2 colo-
putative O-linked glycation sites, and three calized with the endoplasmic reticulum (ER) in
potential binding sites [76]. The alanine at posi- transfected ARPE-19 cells expressing ARMS2.
tion 69 is predicted to be a functional residue for Adding a small N-terminal tag to ARMS2 did not
protein binding. have any effect, whereas the same tag at the
Current Views on Chr10q26 Contribution to Age-Related Macular Degeneration 29

C-terminus abolished ER colocalization, indicat- to human apoptotic and necrotic cells and initiate
ing that the C-terminus is integral to proper properdin-mediated complement activation and
­targeting [81]. A follow-up study concluded that C3b surface opsonization for phagocytosis [75].
ARMS2 is secreted via an unconventional, Golgi-­ Two additional variants of ARMS2 have been
independent pathway. Blocking the canonical ER reported: an insertion-deletion (indel) polymor-
to Golgi transport did not affect ARMS2 secre- phism (NM_001099667.1:c.*372_815del443
tion. Instead, ARMS2 was shown to colocalize ins54) in the 3′untranslated region (3′-UTR) and
with calnexin-positive and protein disulfide a coding nonsense polymorphism (R38X) [79].
isomerase-negative vesicle-like structures and, The ARMS2 indel lies within the HTRA1 cis-­
with GRASP65, a marker for unconventional regulatory region, upstream of its transcription
protein secretion. Exon 2 of ARMS2 codes for start site [83, 84], and is suggested to regulate
eight amino acids (-SIIHTAAR) at the HTRA1 expression. In heterozygous retinal and
C-terminus. By generating a set of mutants, the placental samples from human donors, Fritsche
two isoleucines were found to be indispensable et al. showed that this indel mutation destabilizes
for correct targeting. The intracellular distribu- the ARMS2 transcript by removing the polyade-
tion of the A69S risk allele was similar to the nylation signal and inserting a 54-bp element
non-risk allele, with both protein variants secreted known to mediate rapid mRNA turnover [79].
into the culture medium [82]. The group validated their findings by developing
Micklisch et al. found endogenous ARMS2 recombinant ARMS2 isoforms, quantifying heter-
gene expression in human blood-derived mono- ologous ARMS2 expression in vitro and conclud-
cytes and in human-induced pluripotent stem ing that the indel polymorphism in ARMS2 leads
cell-derived microglia (iPSdM) by PCR and laser to reduced mRNA levels [83].
scanning microscopy. Contrary to the study by The ARMS2 variant identified by Fritsche
Kortvely et al. which showed translation of the et al., rs2736911(C>T:R38X), is predicted to
ARMS2 A69S variant, this study reported that result in a premature stop codon (R38X) and a
ARMS2 protein was absent in patients homozy- truncated protein [79]. Yang et al. observed a
gous for the ARMS2 AMD risk variant A69S or 50% decrease in mRNA in patients heterozygous
the non-risk variant R38X [75]. for R38X [85], presumably due to nonsense-­
mediated mRNA decay. They reasoned that the
loss of ARMS2 is insufficient to explain AMD
2.2 
ARMS2 Risk Variants and Their susceptibility as both the indel and R38X variants
Putative Role in AMD result in a decrease in ARMS2, but only the for-
mer confers risk to AMD, while the latter is
The missense variant, rs10490924 (G>T:A69S) mildly protective [85, 86]. This led to a hypothe-
[73, 80, 82], encoding an alanine-to-serine sub- sis of dual causality, in which concomitant down-
stitution at position 69 (A69S) is the most fre- regulation of ARMS2 and upregulation of HTRA1
quently observed ARMS2 risk allele in the human explains the disease [85]. Similar results were
population. Little is known about the effect of obtained in another study that showed reduced
this mutation on ARMS2 protein structure and ARMS2 expression in human postmortem retinal
function. One study reports that the A69S varia- and RPE samples, heterozygous for R38X [83].
tion in ARMS2 creates a new putative phosphory- However, a subsequent investigation failed to
lation site that breaks a predicted α-helix [73]. It detect a rs2736911-dependent alteration in
has been suggested that the A69S change affects ARMS2 expression [87].
ARMS2’s function in either Mt [73, 79] or its Since ARMS2 is not found in mice, to gain a
role in the cytoskeleton in COS7cells [80]. A better understanding of ARMS2 function, two
recent study implicated ARMS2 as a surface com- transgenic mouse models expressing human
plement regulator [75]. Recombinant ARMS2, ARMS2 and ARMS2 A69S were developed. The
expressed in Pichia pastoris, was shown to bind constructs used a ubiquitous cytomegalovirus
30 N. Gogna et al.

(CMV) [88] or chicken actin (CAG) promoter 293, and Y79 (human retinoblastoma) cells trans-
[89] and were either inserted into the ROSA fected with an HTRA1 promoter driving luciferase
locus [88] or randomly in the mouse genome activity that rs11200638 SNP had no significant
[89].While mRNA and protein expression of impact on HTRA1 promoter activity, and HTRA1
ARMS2 and the A69S variant were confirmed in mRNA expression was not significantly different
both models, no pathological changes were between control and AMD retinas [73].
observed in the mouse eye. Once the AMD risk-associated indel polymor-
phism in the 3’-UTR of ARMS2 was discovered,
many studies focused on identifying its impact on
3 
HTRA1 HTRA1 expression. Using a luciferase reporter
assay on cultured human RPE cells and mouse
HTRA1 is a member of the high temperature RPE in vivo, Yang et al. observed a twofold
requirement A family of four serine proteases. increase in expression in constructs that modelled
While the exact role of these HTRAs is not well the disease haplotype, containing both the ARMS2
understood, HTRA1 is known to be a ubiqui- indel and risk SNP rs11200638 in the HTRA1 pro-
tously expressed, secreted protein with nonspe- moter [85]. No increase was observed with either
cific protease activity [90–92]. The 51-kDa variant alone, suggesting that multiple risk vari-
protein is composed of insulin-like growth factor ants within the Chr10q26 risk haplotype may be
binding, Kazal-like protease inhibitor, conserved necessary to induce heightened HTRA1 expression
serine protease, and PDZ domains [91, 92]. It has [85]. Likewise, a subsequent study by Friedrich
been implicated in physiological processes such et al. in the human ARPE-19 and rat Müller rMC-1
as cell invasion and migration [93, 94], osteogen- cell lines failed to detect an effect of the ARMS2
esis [95, 96], protein degradation [97, 98], and risk allele on HTRA1 promoter activity [83]. A
TGF-β signaling [95, 99]. study by Iejima et al. demonstrated a cell type-
specific change in HTRA1 promoter activity [84].
While no differences in activity in ARPE-19 cells
3.1 Reported Tissue and Cellular were observed, a ~threefold and ~twofold increase
Expression of HTRA1/HTRA1 in luciferase activity was detected in the rat retinal
ganglion cell line RGC-5 and the mouse photore-
In 2006, DeWan et al. and Yang et al. identified a ceptor cell line 661W, respectively. Using con-
single-nucleotide polymorphism (SNP), structs with and without ARMS2 indel mutation,
rs11200638, in the promoter region of HTRA1, as the authors showed that the indel alters the sup-
a major genetic risk factor for AMD [53, 62]. pressor and activator cis-elements of HTRA1 and
Using a GWA mapping strategy, DeWan et al. induces an upregulation of the HTRA1 in pluripo-
concluded that the presence of the risk-associated tent stem cells from AMD patients [84]. An in vivo
HTRA1 genotype increased the likelihood for study was done in rhesus monkey (M. mulatta), a
development of wet AMD tenfold compared to primate model that is susceptible to age-related
individuals with a wild-type genotype [53]. Using maculopathy [74], and forms macular drusen
human ARPE19 and HeLaS3 cells transfected deposits [101]. In this model, polymorphisms in
with luciferase reporter plasmids driven by either orthologous ARMS2 and HTRA1 genes were asso-
wild-type or risk-associated HTRA1 promoter, ciated with drusen formation [102]. While one
this study also demonstrated that the risk variant study attributed the increased risk to the HTRA1
caused an upregulation in HTRA1. Analyzing promoter variant [102], another study could not
mRNA expression and protein levels in primary replicate the results in a different rhesus popula-
cultured human RPE cells revealed a two–three- tion [103].
fold increase in HTRA1 expression in the pres- Investigations using human tissue also yielded
ence of the HTRA1 risk haplotype [100]. In variable results. Yang et al. reported an increase in
contrast, Kanda et al. showed in ARPE-19, HEK- expression levels of HTRA1 mRNA and protein in
Current Views on Chr10q26 Contribution to Age-Related Macular Degeneration 31

lymphocytes and RPE cells [62]. Chan et al. also Another function suggested for HTRA1 has
reported HTRA1 upregulation specifically in mac- been as a regulator of TGF-β, which has been
ular regions with lower expression in the periph- associated with AMD due to its role in angiogen-
eral retina [104]. However, contrasting results esis, inflammation, vascular fibrosis, immune
were obtained from independent studies that responses, as well as cross-talk with other signal-
showed no differences in expression between risk- ing pathways [121–123]. Some studies suggest
and non-risk-associated HTRA1 transcripts that HTRA1 can inhibit TGF-β signaling through
obtained from human lymphocytes and retina [87, a number of mechanisms [124–126], while others
105–107]. A recent study by Williams et al. report that HTRA1 activates it [99, 127–129]. A
showed that HTRA1 mRNA is reduced specifically study by Akhtar-Schaefer et al. reported no effect
in RPE and not in neural retina or choroid derived of HTRA1 on enhancing or inhibiting TGF-β sig-
from human donors homozygous for the risk vari- naling [130].
ants that disrupt a cis-regulatory element within
the Chr10q26 locus [108]. Studies involving AMD
patient-derived induced pluripotent stem cells 4 Conclusions
(iPSCs) also report conflicting results, with some
studies reporting higher HTRA1 expression in Both ARMS2 and HTRA1 have been extensively
iPSCs from patients harboring the ARMS2 indel studied for their roles in AMD. However, con-
polymorphism [84] whereas others showing no flicting outcomes from previous investigations
change in HTRA1 expression levels [109]. make it difficult to draw conclusions. Thus far, no
consensus exists about where ARMS2 is
expressed or how it functions. Also, although the
3.2 Contribution of HTRA1 Gene function and localization of HTRA1 are better
Variants to AMD understood, studies of its regulation and associa-
tion with AMD have also generated ambiguous
A large number of functional studies have been results.
undertaken to identify how the HTRA1 protein The use of different tissues, cell types, pri-
might lead to AMD disease phenotypes. HTRA1, mary cells or cell lines, and reagents makes com-
for example, has been found to bind several ECM parisons difficult. Ideally, functional studies and
proteins [90, 110–119], which could potentially developing therapies would be performed in ani-
affect the posterior eye. For example, an altered mal models. Working with primate models can be
elastogenesis of Bruch’s membrane was reported challenging, since they present the same genetic
in transgenic mice overexpressing HTRA1 in the heterogeneity as humans and require large sam-
RPE [115]. Using a differential stable isotope ple sizes. Mouse models, whose genetics and
labeling of amino acids in cell culture strategy, environment can be easily controlled and manip-
RPE proteins selectively cleaved by HTRA1 ulated, offer a potential solution. Care should be
were identified, and its involvement in comple- taken to select and interpret relevant phenotypes
ment regulation and amyloid deposition during because of the lack of a macula per se. In addi-
AMD pathogenesis was suggested [100]. In tion, because AMD is a complex disease, a sus-
another study, two ECM proteins were found to ceptible genetic background has to be identified,
be processed by HTRA1: EFEMP1, an ECM pro- which may have to be specific for the AMD risk
tein, known to be mutated in Doyne honeycomb variant to be studied.
retinal dystrophy [120], a genetic eye disease Nevertheless, intriguing leads have resulted
with similarities to AMD, and thrombospondin 1 from studies conducted thus far, and prospects
(TSP1), an inhibitor of angiogenesis. Further­ are good that we can progress in understanding
more, HTRA1 may play a role in AMD by regu- the functional relevance of these genes and help
lating the ECM and potentially by playing a role to identify novel molecular targets and open new
in neovascularization [118]. therapeutic avenues for the treatment of AMD.
32 N. Gogna et al.

References 16. Handa JT, Cano M, Wang L, Datta S, Liu T. Lipids,


oxidized lipids, oxidation-specific epitopes, and Age-­
related Macular Degeneration. Biochim Biophys
1. VISION 2020 Global Initiative for the Elimination
Acta Mol Cell Biol Lipids. 2017;1862(4):430–40.
of Avoidable Blindness: Action plan 2006–2011.
17. Chen M, Xu H. Parainflammation, chronic inflam-
VISION 2020 Global Initiative for the Elimination
mation, and age-related macular degeneration. J
of Avoidable Blindness: Action plan 2006–2011;
Leukoc Biol. 2015;98(5):713–25.
2007.
18. Knickelbein JE, Chan CC, Sen HN, Ferris FL,
2. de Jong EK, Geerlings MJ, den Hollander
Nussenblatt RB. Inflammatory mechanisms of age-­
AI. Chapter 10 – Age-related macular degeneration.
related macular degeneration. Int Ophthalmol Clin.
In: Genetics and genomics of eye disease. London:
2015;55(3):63–78.
Academic; 2020. p. 155–80.
19. van Lookeren Campagne M, Strauss EC, Yaspan
3. Rein DB, Wittenborn JS, Zhang X, Honeycutt
BL. Age-related macular degeneration: Complement
AA, Lesesne SB, Saaddine J, et al. Forecasting
in action. Immunobiology. 2016;221(6):733–9.
age-related macular degeneration through the year
20. Kauppinen A, Paterno JJ, Blasiak J, Salminen A,
2050: the potential impact of new treatments. Arch
Kaarniranta K. Inflammation and its role in age-­
Ophthalmol. 2009;127(4):533–40.
related macular degeneration. Cell Mol Life Sci.
4. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng
2016;73(9):1765–86.
CY, et al. Global prevalence of age-related macular
21. Lambert NG, ElShelmani H, Singh MK, Mansergh
degeneration and disease burden projection for 2020
FC, Wride MA, Padilla M, et al. Risk factors and
and 2040: a systematic review and meta-analysis.
biomarkers of age-related macular degeneration.
Lancet Glob Health. 2014;2(2):e106–16.
Prog Retin Eye Res. 2016;54:64–102.
5. Jager RD, Mieler WF, Miller JW. Age-related macular
22. Tan PL, Bowes Rickman C, Katsanis N. AMD
degeneration. N Engl J Med. 2008;358(24):2606–17.
and the alternative complement pathway: genet-
6. Bird AC, Bressler NM, Bressler SB, Chisholm IH,
ics and functional implications. Hum Genomics.
Coscas G, Davis MD, et al. An international clas-
2016;10(1):23.
sification and grading system for age-related macu-
23. Gelfand BD, Ambati J. A revised hemodynamic
lopathy and age-related macular degeneration. The
theory of age-related macular degeneration. Trends
International ARM Epidemiological Study Group.
Mol Med. 2016;22(8):656–70.
Surv Ophthalmol. 1995;39(5):367–74.
24. Schwartz SG, Hampton BM, Kovach JL, Brantley
7. Bressler NM, Silva JC, Bressler SB, Fine SL, Green
MA Jr. Genetics and age-related macular degen-
WR. Clinicopathologic correlation of drusen and ret-
eration: a practical review for the clinician. Clin
inal pigment epithelial abnormalities in age-related
Ophthalmol. 2016;10:1229–35.
macular degeneration. Retina. 1994;14(2):130–42.
25. Smith W, Assink J, Klein R, Mitchell P, Klaver CC,
8. Crabb JW. The proteomics of drusen. Cold Spring
Klein BE, et al. Risk factors for age-related macular
Harb Perspect Med. 2014;4(7):a017194.
degeneration: pooled findings from three continents.
9. Ferris FL 3rd, Wilkinson CP, Bird A, Chakravarthy
Ophthalmology. 2001;108(4):697–704.
U, Chew E, Csaky K, et al. Clinical classification of
26. Chakravarthy U, Wong TY, Fletcher A, Piault E,
age-related macular degeneration. Ophthalmology.
Evans C, Zlateva G, et al. Clinical risk factors
2013;120(4):844–51.
for age-related macular degeneration: a system-
10. Klein R, Klein BE, Jensen SC, Meuer SM. The five-­
atic review and meta-analysis. BMC Ophthalmol.
year incidence and progression of age-related macu-
2010;10:31.
lopathy: the Beaver Dam Eye Study. Ophthalmology.
27. Ferris FL 3rd. Senile macular degeneration:
1997;104(1):7–21.
review of epidemiologic features. Am J Epidemiol.
11. Fine SL, Berger JW, Maguire MG, Ho AC. Age-­
1983;118(2):132–51.
related macular degeneration. N Engl J Med.
28. Hyman LG, Lilienfeld AM, Ferris FL 3rd, Fine
2000;342(7):483–92.
SL. Senile macular degeneration: a case-control
12. Pennington KL, DeAngelis MM. Epigenetic mecha-
study. Am J Epidemiol. 1983;118(2):213–27.
nisms of the aging human retina. J Exp Neurosci.
29. Goldberg J, Flowerdew G, Smith E, Brody JA, Tso
2015;9(Suppl 2):51–79.
MO. Factors associated with age-related macu-
13. Armstrong RA, Mousavi M. Overview of risk fac-
lar degeneration. An analysis of data from the first
tors for age-related macular degeneration (AMD). J
National Health and Nutrition Examination Survey.
Stem Cells. 2015;10(3):171–91.
Am J Epidemiol. 1988;128(4):700–10.
14. Gemenetzi M, Lotery AJ. The role of epigenetics
30. Risk factors for neovascular age-related macular
in age-related macular degeneration. Eye (Lond).
degeneration. The Eye Disease Case-Control Study
2014;28(12):1407–17.
Group. Arch Ophthalmol. 1992;110(12):1701–8.
15. Zhang QY, Tie LJ, Wu SS, Lv PL, Huang HW, Wang
31. Mitchell P, Smith W, Attebo K, Wang JJ. Prevalence
WQ, et al. Overweight, obesity, and risk of age-­
of age-related maculopathy in Australia. The
related macular degeneration. Invest Ophthalmol Vis
Blue Mountains Eye Study. Ophthalmology.
Sci. 2016;57(3):1276–83.
1995;102(10):1450–60.
Current Views on Chr10q26 Contribution to Age-Related Macular Degeneration 33

32. Vingerling JR, Dielemans I, Hofman A, Grobbee 47. Seddon JM, Cote J, Page WF, Aggen SH, Neale
DE, Hijmering M, Kramer CF, et al. The prevalence MC. The US twin study of age-related macular degen-
of age-related maculopathy in the Rotterdam Study. eration: relative roles of genetic and environmental
Ophthalmology. 1995;102(2):205–10. influences. Arch Ophthalmol. 2005;123(3):321–7.
33. Cruickshanks KJ, Klein R, Klein BE. Sunlight and 48. Gorin MB, Weeks DE, Baron RV, Conley YP,
age-related macular degeneration. The Beaver Dam Ortube MC, Nusinowitz S. Endophenotypes for age-­
Eye Study. Arch Ophthalmol. 1993;111(4):514–8. related macular degeneration: extending our reach
34. Fletcher AE, Bentham GC, Agnew M, Young IS, into the preclinical stages of disease. J Clin Med.
Augood C, Chakravarthy U, et al. Sunlight exposure, 2014;3(4):1335–56.
antioxidants, and age-related macular degeneration. 49. Fritsche LG, Igl W, Bailey JN, Grassmann F,
Arch Ophthalmol. 2008;126(10):1396–403. Sengupta S, Bragg-Gresham JL, et al. A large
35. Klein R, Klein BE, Linton KL. Prevalence of age-­ genome-wide association study of age-related macu-
related maculopathy. The Beaver Dam Eye Study. lar degeneration highlights contributions of rare and
Ophthalmology. 1992;99(6):933–43. common variants. Nat Genet. 2016;48(2):134–43.
36. Seddon JM, Ajani UA, Mitchell BD. Familial 50. den Hollander AI, de Jong EK. Highly penetrant
aggregation of age-related maculopathy. Am J alleles in age-related macular degeneration. Cold
Ophthalmol. 1997;123(2):199–206. Spring Harb Perspect Med. 2014;5(3):a017202.
37. van Leeuwen R, Klaver CC, Vingerling JR, Hofman 51. Black JR, Clark SJ. Age-related macular degenera-
A, van Duijn CM, Stricker BH, et al. Cholesterol and tion: genome-wide association studies to translation.
age-related macular degeneration: is there a link? Genet Med. 2016;18(4):283–9.
Am J Ophthalmol. 2004;137(4):750–2. 52. Zareparsi S, Branham KE, Li M, Shah S, Klein RJ,
38. Edo A, Pertiwi YD, Hirooka K, Masuda S, Ott J, et al. Strong association of the Y402H variant
Kamaruddin MI, Yanagi M, et al. Association of in complement factor H at 1q32 with susceptibil-
dietary nutrient intake with early age-related macular ity to age-related macular degeneration. Am J Hum
degeneration in Japanese-Americans. Metabolites. Genet. 2005;77(1):149–53.
2021;11(10):673. 53. Dewan A, Liu M, Hartman S, Zhang SS, Liu DT,
39. Newsome DA, Swartz M, Leone NC, Elston RC, Zhao C, et al. HTRA1 promoter polymorphism in
Miller E. Oral zinc in macular degeneration. Arch wet age-related macular degeneration. Science.
Ophthalmol. 1988;106(2):192–8. 2006;314(5801):989–92.
40. VandenLangenberg GM, Mares-Perlman JA, Klein 54. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS,
R, Klein BE, Brady WE, Palta M. Associations Haynes C, et al. Complement factor H polymor-
between antioxidant and zinc intake and the phism in age-related macular degeneration. Science.
5-year incidence of early age-related maculopathy 2005;308(5720):385–9.
in the Beaver Dam Eye Study. Am J Epidemiol. 55. Jakobsdottir J, Conley YP, Weeks DE, Mah TS,
1998;148(2):204–14. Ferrell RE, Gorin MB. Susceptibility genes for age-­
41. Friedman DS, O'Colmain BJ, Munoz B, Tomany related maculopathy on chromosome 10q26. Am J
SC, McCarty C, de Jong PT, et al. Prevalence of age-­ Hum Genet. 2005;77(3):389–407.
related macular degeneration in the United States. 56. Rivera A, Fisher SA, Fritsche LG, Keilhauer
Arch Ophthalmol. 2004;122(4):564–72. CN, Lichtner P, Meitinger T, et al. Hypothetical
42. Sommer A, Tielsch JM, Katz J, Quigley HA, Gottsch LOC387715 is a second major susceptibility gene
JD, Javitt JC, et al. Racial differences in the cause-­ for age-related macular degeneration, contributing
specific prevalence of blindness in east Baltimore. N independently of complement factor H to disease
Engl J Med. 1991;325(20):1412–7. risk. Hum Mol Genet. 2005;14(21):3227–36.
43. Klein R, Rowland ML, Harris MI. Racial/eth- 57. Liu X, Zhao P, Tang S, Lu F, Hu J, Lei C, et al.
nic differences in age-related maculopathy. Third Association study of complement factor H, C2,
National Health and Nutrition Examination Survey. CFB, and C3 and age-related macular degen-
Ophthalmology. 1995;102(3):371–81. eration in a Han Chinese population. Retina.
44. Seddon JM, Willett WC, Speizer FE, Hankinson 2010;30(8):1177–84.
SE. A prospective study of cigarette smoking and 58. Seddon JM, Francis PJ, George S, Schultz
age-related macular degeneration in women. JAMA. DW, Rosner B, Klein ML. Association of CFH
1996;276(14):1141–6. Y402H and LOC387715 A69S with progres-
45. Christen WG, Glynn RJ, Manson JE, Ajani UA, sion of age-related macular degeneration. JAMA.
Buring JE. A prospective study of cigarette smoking 2007;297(16):1793–800.
and risk of age-related macular degeneration in men. 59. Edwards AO, Ritter R 3rd, Abel KJ, Manning A,
JAMA. 1996;276(14):1147–51. Panhuysen C, Farrer LA. Complement factor H
46. Klein R, Klein BE, Moss SE. Relation of smok- polymorphism and age-related macular degenera-
ing to the incidence of age-related maculopathy. tion. Science. 2005;308(5720):421–4.
The Beaver Dam Eye Study. Am J Epidemiol. 60. Hageman GS, Anderson DH, Johnson LV, Hancox
1998;147(2):103–10. LS, Taiber AJ, Hardisty LI, et al. A common hap-
lotype in the complement regulatory gene factor H
34 N. Gogna et al.

(HF1/CFH) predisposes individuals to age-related 74. Francis PJ, Appukuttan B, Simmons E, Landauer
macular degeneration. Proc Natl Acad Sci U S A. N, Stoddard J, Hamon S, et al. Rhesus monkeys
2005;102(20):7227–32. and humans share common susceptibility genes
61. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson for age-related macular disease. Hum Mol Genet.
LM, Gallins P, et al. Complement factor H variant 2008;17(17):2673–80.
increases the risk of age-related macular degenera- 75. Micklisch S, Lin Y, Jacob S, Karlstetter M,
tion. Science. 2005;308(5720):419–21. Dannhausen K, Dasari P, et al. Age-related mac-
62. Yang Z, Camp NJ, Sun H, Tong Z, Gibbs D, Cameron ular degeneration associated polymorphism
DJ, et al. A variant of the HTRA1 gene increases rs10490924 in ARMS2 results in deficiency of
susceptibility to age-related macular degeneration. a complement activator. J Neuroinflammation.
Science. 2006;314(5801):992–3. 2017;14(1):4.
63. Ryu E, Fridley BL, Tosakulwong N, Bailey KR, 76. Jahanfar F, Hamishehkar H. Exploring the associa-
Edwards AO. Genome-wide association analy- tion of rs10490924 polymorphism with age-related
ses of genetic, phenotypic, and environmental macular degeneration: An in silico approach. J Mol
risks in the age-related eye disease study. Mol Vis. Graph Model. 2017;77:280–5.
2010;16:2811–21. 77. Wang G, Spencer KL, Scott WK, Whitehead P,
64. Winkler TW, Grassmann F, Brandl C, Kiel C, Court BL, Ayala-Haedo J, et al. Analysis of the indel
Gunther F, Strunz T, et al. Genome-wide association at the ARMS2 3′UTR in age-related macular degen-
meta-analysis for early age-related macular degener- eration. Hum Genet. 2010;127(5):595–602.
ation highlights novel loci and insights for advanced 78. Bastian FB, Roux J, Niknejad A, Comte A, Fonseca
disease. BMC Med Genet. 2020;13(1):120. Costa SS, de Farias TM, et al. The Bgee suite:
65. Horie-Inoue K, Inoue S. Genomic aspects of age-­ integrated curated expression atlas and compara-
related macular degeneration. Biochem Biophys Res tive transcriptomics in animals. Nucleic Acids Res.
Commun. 2014;452(2):263–75. 2021;49(D1):D831–D47.
66. Cameron DJ, Yang Z, Gibbs D, Chen H, Kaminoh 79. Fritsche LG, Loenhardt T, Janssen A, Fisher SA,
Y, Jorgensen A, et al. HTRA1 variant confers Rivera A, Keilhauer CN, et al. Age-related macular
similar risks to geographic atrophy and neovascu- degeneration is associated with an unstable ARMS2
lar age-related macular degeneration. Cell Cycle. (LOC387715) mRNA. Nat Genet. 2008;40(7):892–6.
2007;6(9):1122–5. 80. Wang G, Spencer KL, Court BL, Olson LM, Scott
67. Landowski M, Kelly U, Klingeborn M, Groelle M, WK, Haines JL, et al. Localization of age-related
Ding JD, Grigsby D, et al. Human complement fac- macular degeneration-associated ARMS2 in cyto-
tor H Y402H polymorphism causes an age-related sol, not mitochondria. Invest Ophthalmol Vis Sci.
macular degeneration phenotype and lipoprotein 2009;50(7):3084–90.
dysregulation in mice. Proc Natl Acad Sci U S A. 81. Kortvely E, Hauck SM, Duetsch G, Gloeckner CJ,
2019;116(9):3703–11. Kremmer E, Alge-Priglinger CS, et al. ARMS2 is
68. Toomey CB, Johnson LV, Bowes Rickman a constituent of the extracellular matrix providing
C. Complement factor H in AMD: bridging genetic a link between familial and sporadic age-related
associations and pathobiology. Prog Retin Eye Res. macular degenerations. Invest Ophthalmol Vis Sci.
2018;62:38–57. 2010;51(1):79–88.
69. Donoso LA, Vrabec T, Kuivaniemi H. The role 82. Kortvely E, Hauck SM, Behler J, Ho N, Ueffing
of complement Factor H in age-related macu- M. The unconventional secretion of ARMS2. Hum
lar degeneration: a review. Surv Ophthalmol. Mol Genet. 2016;25(15):3143–51.
2010;55(3):227–46. 83. Friedrich U, Myers CA, Fritsche LG, Milenkovich
70. McHarg S, Clark SJ, Day AJ, Bishop PN. Age-­ A, Wolf A, Corbo JC, et al. Risk- and non-risk-­
related macular degeneration and the role of the com- associated variants at the 10q26 AMD locus influ-
plement system. Mol Immunol. 2015;67(1):43–50. ence ARMS2 mRNA expression but exclude
71. Wang G. Chromosome 10q26 locus and age-related pathogenic effects due to protein deficiency. Hum
macular degeneration: a progress update. Exp Eye Mol Genet. 2011;20(7):1387–99.
Res. 2014;119:1–7. 84. Iejima D, Itabashi T, Kawamura Y, Noda T, Yuasa S,
72. Leveziel N, Souied EH, Richard F, Barbu V, Zourdani Fukuda K, et al. HTRA1 (high temperature require-
A, Morineau G, et al. PLEKHA1-LOC387715-­ ment A serine peptidase 1) gene is transcription-
HTRA1 polymorphisms and exudative age-related ally regulated by insertion/deletion nucleotides
macular degeneration in the French population. Mol located at the 3′ end of the ARMS2 (age-related
Vis. 2007;13:2153–9. maculopathy susceptibility 2) gene in patients with
73. Kanda A, Chen W, Othman M, Branham KE, Brooks age-related macular degeneration. J Biol Chem.
M, Khanna R, et al. A variant of mitochondrial pro- 2015;290(5):2784–97.
tein LOC387715/ARMS2, not HTRA1, is strongly 85. Yang Z, Tong Z, Chen Y, Zeng J, Lu F, Sun X, et al.
associated with age-related macular degeneration. Genetic and functional dissection of HTRA1 and
Proc Natl Acad Sci U S A. 2007;104(41):16227–32. LOC387715 in age-related macular degeneration.
PLoS Genet. 2010;6(2):e1000836.
Current Views on Chr10q26 Contribution to Age-Related Macular Degeneration 35

86. Teper SJ, Nowinska A, Wylegala E. A69S and R38X degrades tau protein aggregates. J Biol Chem.
ARMS2 and Y402H CFH gene polymorphisms 2012;287(25):20931–41.
as risk factors for neovascular age-related macular 99. Beaufort N, Scharrer E, Kremmer E, Lux V,
degeneration in Poland – a brief report. Med Sci Ehrmann M, Huber R, et al. Cerebral small ves-
Monit. 2012;18(2):PR1-3. sel disease-­ related protease HtrA1 processes
87. Kanda A, Stambolian D, Chen W, Curcio CA, latent TGF-beta binding protein 1 and facilitates
Abecasis GR, Swaroop A. Age-related macu- TGF-beta signaling. Proc Natl Acad Sci U S A.
lar degeneration-associated variants at chromo- 2014;111(46):16496–501.
some 10q26 do not significantly alter ARMS2 and 100. An E, Sen S, Park SK, Gordish-Dressman H, Hathout
HTRA1 transcript levels in the human retina. Mol Y. Identification of novel substrates for the serine
Vis. 2010;16:1317–23. protease HTRA1 in the human RPE secretome.
88. Liu J, Hoh J. Postnatal overexpression of the human Invest Ophthalmol Vis Sci. 2010;51(7):3379–86.
ARMS2 gene does not induce abnormalities in ret- 101. Yiu G, Tieu E, Munevar C, Wong B, Cunefare D,
ina and choroid in transgenic mouse models. Invest Farsiu S, et al. In vivo multimodal imaging of
Ophthalmol Vis Sci. 2015;56(2):1387–8. drusenoid lesions in rhesus macaques. Sci Rep.
89. Nakayama M, Iejima D, Akahori M, Kamei J, Goto 2017;7(1):15013.
A, Iwata T. Overexpression of HtrA1 and exposure to 102. Singh KK, Krawczak M, Dawson WW, Schmidtke
mainstream cigarette smoke leads to choroidal neo- J. Association of HTRA1 and ARMS2 gene varia-
vascularization and subretinal deposits in aged mice. tion with drusen formation in rhesus macaques. Exp
Invest Ophthalmol Vis Sci. 2014;55(10):6514–23. Eye Res. 2009;88(3):479–82.
90. Hu SI, Carozza M, Klein M, Nantermet P, Luk D, 103. Pahl L, Spangenberg A, Schubert S, Schonmann
Crowl RM. Human HtrA, an evolutionarily con- U, Schmidtke J, Stuhrmann M. Characterization of
served serine protease identified as a differentially the 10q26-orthologue in rhesus monkeys corrobo-
expressed gene product in osteoarthritic cartilage. J rates a functional connection between ARMS2 and
Biol Chem. 1998;273(51):34406–12. HTRA1. Exp Eye Res. 2012;98:75–8.
91. De Luca A, De Falco M, Severino A, Campioni 104. Chan CC, Shen D, Zhou M, Ross RJ, Ding X, Zhang
M, Santini D, Baldi F, et al. Distribution of the K, et al. Human HtrA1 in the archived eyes with age-­
serine protease HtrA1 in normal human tissues. J related macular degeneration. Trans Am Ophthalmol
Histochem Cytochem. 2003;51(10):1279–84. Soc. 2007;105:92–7; discussion 7–8.
92. Zumbrunn J, Trueb B. Primary structure of a puta- 105. Chowers I, Meir T, Lederman M, Goldenberg-Cohen
tive serine protease specific for IGF-binding pro- N, Cohen Y, Banin E, et al. Sequence variants in
teins. FEBS Lett. 1996;398(2–3):187–92. HTRA1 and LOC387715/ARMS2 and phenotype
93. Ajayi F, Kongoasa N, Gaffey T, Asmann YW, and response to photodynamic therapy in neovascu-
Watson WJ, Baldi A, et al. Elevated expression of lar age-related macular degeneration in populations
serine protease HtrA1 in preeclampsia and its role in from Israel. Mol Vis. 2008;14:2263–71.
trophoblast cell migration and invasion. Am J Obstet 106. Wang G, Scott WK, Haines JL, Pericak-Vance
Gynecol. 2008;199(5):557e1–10. MA. Genotype at polymorphism rs11200638
94. Chien J, Ota T, Aletti G, Shridhar R, Boccellino M, and HTRA1 expression level. Arch Ophthalmol.
Quagliuolo L, et al. Serine protease HtrA1 associ- 2010;128(11):1491–3.
ates with microtubules and inhibits cell migration. 107. Wang G, Dubovy SR, Kovach JL, Schwartz SG,
Mol Cell Biol. 2009;29(15):4177–87. Agarwal A, Scott WK, et al. Variants at chromosome
95. Graham JR, Chamberland A, Lin Q, Li XJ, Dai D, 10q26 locus and the expression of HTRA1 in the
Zeng W, et al. Serine protease HTRA1 antagonizes retina. Exp Eye Res. 2013;112:102–5.
transforming growth factor-beta signaling by cleav- 108. Williams BL, Seager NA, Gardiner JD, Pappas
ing its receptors and loss of HTRA1 in vivo enhances CM, Cronin MC, Amat di San Filippo C, et al.
bone formation. PLoS One. 2013;8(9):e74094. Chromosome 10q26-driven age-related macular
96. Tiaden AN, Breiden M, Mirsaidi A, Weber FA, degeneration is associated with reduced levels of
Bahrenberg G, Glanz S, et al. Human serine protease HTRA1 in human retinal pigment epithelium. Proc
HTRA1 positively regulates osteogenesis of human Natl Acad Sci U S A. 2021;118(30):20210727.
bone marrow-derived mesenchymal stem cells and 109. Yang J, Li Y, Chan L, Tsai YT, Wu WH, Nguyen
mineralization of differentiating bone-forming cells HV, et al. Validation of genome-wide association
through the modulation of extracellular matrix pro- study (GWAS)-identified disease risk alleles with
tein. Stem Cells. 2012;30(10):2271–82. patient-specific stem cell lines. Hum Mol Genet.
97. Chu Q, Diedrich JK, Vaughan JM, Donaldson 2014;23(13):3445–55.
CJ, Nunn MF, Lee KF, et al. HtrA1 proteolysis of 110. Murwantoko YM, Ueta Y, Murasaki A, Kanda H,
ApoE in vitro is allele selective. J Am Chem Soc. Oka C, et al. Binding of proteins to the PDZ domain
2016;138(30):9473–8. regulates proteolytic activity of HtrA1 serine prote-
98. Tennstaedt A, Popsel S, Truebestein L, Hauske P, ase. Biochem J. 2004;381(Pt 3):895–904.
Brockmann A, Schmidt N, et al. Human high tem- 111. Hou J, Clemmons DR, Smeekens S. Expression
perature requirement serine protease A1 (HTRA1) and characterization of a serine protease that pref-
36 N. Gogna et al.

erentially cleaves insulin-like growth factor binding is pathogenic and causes AMD-like deposits in mice.
protein-­5. J Cell Biochem. 2005;94(3):470–84. Hum Mol Genet. 2007;16(20):2411–22.
112. Grau S, Baldi A, Bussani R, Tian X, Stefanescu R, 121. Wang K, Li H, Sun R, Liu C, Luo Y, Fu S, et al.
Przybylski M, et al. Implications of the serine prote- Emerging roles of transforming growth factor
ase HtrA1 in amyloid precursor protein processing. beta signaling in wet age-related macular degen-
Proc Natl Acad Sci U S A. 2005;102(17):6021–6. eration. Acta Biochim Biophys Sin Shanghai.
113. Hadfield KD, Rock CF, Inkson CA, Dallas SL, Sudre 2019;51(1):1–8.
L, Wallis GA, et al. HtrA1 inhibits mineral deposi- 122. Tosi GM, Orlandini M, Galvagni F. The contro-
tion by osteoblasts: requirement for the protease and versial role of TGF-beta in neovascular age-related
PDZ domains. J Biol Chem. 2008;283(9):5928–38. macular degeneration pathogenesis. Int J Mol Sci.
114. Tsuchiya A, Yano M, Tocharus J, Kojima H, 2018;19(11):E3363.
Fukumoto M, Kawaichi M, et al. Expression of 123. Moses HL, Roberts AB, Derynck R. The discovery
mouse HtrA1 serine protease in normal bone and car- and early days of TGF-beta: a historical perspective.
tilage and its upregulation in joint cartilage damaged Cold Spring Harb Perspect Biol. 2016;8(7) a021865.
by experimental arthritis. Bone. 2005;37(3):323–36. 124. Oka C, Tsujimoto R, Kajikawa M, Koshiba-Takeuchi
115. Vierkotten S, Muether PS, Fauser S. Overexpression K, Ina J, Yano M, et al. HtrA1 serine protease inhib-
of HTRA1 leads to ultrastructural changes in the its signaling mediated by Tgfbeta family proteins.
elastic layer of Bruch’s membrane via cleavage Development. 2004;131(5):1041–53.
of extracellular matrix components. PLoS One. 125. Launay S, Maubert E, Lebeurrier N, Tennstaedt A,
2011;6(8):e22959. Campioni M, Docagne F, et al. HtrA1-dependent
116. Kim GY, Kim HY, Kim HT, Moon JM, Kim CH, proteolysis of TGF-beta controls both neuronal
Kang S, et al. HtrA1 is a novel antagonist controlling maturation and developmental survival. Cell Death
fibroblast growth factor (FGF) signaling via cleav- Differ. 2008;15(9):1408–16.
age of FGF8. Mol Cell Biol. 2012;32(21):4482–92. 126. Shiga A, Nozaki H, Yokoseki A, Nihonmatsu M,
117. Klose R, Adam MG, Weis EM, Moll I, Wustehube-­ Kawata H, Kato T, et al. Cerebral small-vessel dis-
Lausch J, Tetzlaff F, et al. Inactivation of the serine ease protein HTRA1 controls the amount of TGF-­
protease HTRA1 inhibits tumor growth by deregulat- beta1 via cleavage of proTGF-beta1. Hum Mol
ing angiogenesis. Oncogene. 2018;37(31):4260–72. Genet. 2011;20(9):1800–10.
118. Lin MK, Yang J, Hsu CW, Gore A, Bassuk AG, 127. Chang C. Agonists and antagonists of TGF-beta
Brown LM, et al. HTRA1, an age-related macu- family ligands. Cold Spring Harb Perspect Biol.
lar degeneration protease, processes extracellular 2016;8(8) a021923.
matrix proteins EFEMP1 and TSP1. Aging Cell. 128. Derynck R, Budi EH. Specificity, versatility, and
2018;17(4):e12710. control of TGF-beta family signaling. Sci Signal.
119. Chekuri A, Zientara-Rytter K, Soto-Hermida A, 2019;12(570):eaav5183.
Borooah S, Voronchikhina M, Biswas P, et al. Late-­ 129. Robertson IB, Rifkin DB. Regulation of the bioavail-
onset retinal degeneration pathology due to muta- ability of TGF-beta and TGF-beta-related proteins.
tions in CTRP5 is mediated through HTRA1. Aging Cold Spring Harb Perspect Biol. 2016;8(6) a021907.
Cell. 2019;18(6):e13011. 130. Akhtar-Schaefer I, Reuten R, Koch M, Pietsch M,
120. Fu L, Garland D, Yang Z, Shukla D, Rajendran A, Langmann T. AMD-associated HTRA1 variants do
Pearson E, et al. The R345W mutation in EFEMP1 not influence TGF-beta signaling in microglia. Adv
Exp Med Biol. 2019;1185:3–7.
Untargeted Lipidomic Profiling
of Aged Human Retina
With and Without Age-Related
Macular Degeneration (AMD)

Ankita Kotnala, David M. G. Anderson,


Jeffrey D. Messinger, Christine A. Curcio,
and Kevin L. Schey

Abstract and were analyzed by LC-MS/MS and


nanoLC-MS/MS using negative and positive
The molecular characterization of extracellu- ionization modes. Untargeted lipidomics
lar deposits is crucial to understanding the using LC-MS/MS identified 215 lipids from 4
clinical progression of AMD. Liquid lipid classes and 15 subclasses. We observed a
chromatography-­ tandem mass spectrometry 78% increase in lipid identifications using
(LC-MS/MS) analysis is a powerful analytical nanoLC-MS/MS with lipid numbers totaling
discovery tool capable of identifying lipids in 384. The nanoLC-MS/MS method is expected
an untargeted manner. NanoLC-MS/MS is an to provide extensive lipid identifications from
analytical tool capable of identifying lipids small retina samples, e.g., from drusen and
with high sensitivity and minimum sample drusenoid deposits in aged and AMD eyes,
usage. Hence, the purpose of this study was to and could help elucidate how lipids are
compare retina lipid identification from RPE-­ involved in extracellular deposit formation in
choroid samples using high flow LC-MS/MS AMD.
and nanoLC-MS/MS. Manually dissected
paraformaldehyde-fixed human donor tissues Keywords
sections were used for LC-MS/MS and
nanoLC-MS/MS analysis. Lipids were Age-related macular degeneration · Retina ·
extracted with MeOH/MTBE/CHCl3 (MMC) RPE · Fixed · Lipids · LC-MS/MS ·
NanoLC-MS/MS
A. Kotnala
Department of Biochemistry, Vanderbilt University,
Nashville, TN, USA
Department of Ophthalmology, University of 1 Introduction
Alabama at Birmingham, Birmingham, AL, USA
D. M. G. Anderson · K. L. Schey (*) Age-related macular degeneration (AMD) is a
Department of Biochemistry, Vanderbilt University, major cause of blindness in older adults world-
Nashville, TN, USA wide [1]. Extracellular deposits on basal and
e-mail: k.schey@vanderbilt.edu apical sides of the retinal pigment epithelium
J. D. Messinger · C. A. Curcio (RPE) are major risk factors for AMD.
Department of Ophthalmology, University of Degeneration and death of photoreceptors
Alabama at Birmingham, Birmingham, AL, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 37


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_6
38 A. Kotnala et al.

results in vision loss [2, 3]. Ample evidence The RPE-choroid was removed from the glass
supports a role of lipids and associated path- slide with a scalpel blade, under a dissecting
ways in extracellular deposit formation and microscope. Sclera and neural retina were care-
degeneration of vision-critical cells of neural fully removed before removing RPE-choroid tis-
retina [2]. Molecular ­characterization of human sue. The dissected RPE-choroid samples from
RPE and extracellular deposits will further our each tissue section were placed in separate HPLC
understanding of how these deposits form and glass vials, and the lipids were extracted using
confer risk for progression [4]. Liquid chroma- MeOH:MTBE:CHCl3(1.2:1:1) (MMC) extrac-
tography-tandem mass spectrometry LC-MS/ tion mixture. The samples were spiked with
MS analysis is a powerful analytical tool for SPLASH LIPIDOMIX (Avanti Polar Lipids) as
identifying lipids using accurate masses and internal standards, vortexed for 60 s and centri-
fragmentation patterns. LC-MS/MS provides fuged at 3000 rpm for 10 min. The supernatant
molecular identifications of many hundreds of was transferred to a separate HPLC vial and was
lipids in an untargeted fashion [5]. NanoLC-MS/ evaporated to dryness. The sample was then
MS is a highly sensitive method for comprehen- reconstituted with 30 μL methanol, and 10 μL
sive lipidomic coverage from limited samples. was injected for high flow LC-MS/MS analysis.
The nanoLC-MS/MS method has not been For nanoLC-MS/MS analysis, samples were
extensively explored for lipidomic analysis [6, reconstituted with 10 μL BuOH:IPA:H20
7]. This study, for the first time, evaluated its (8:23:69) with 5 mM phosphoric acid, and 1 μL
utility in analyzing human retina. The goal of was subjected for nLC-MS/MS analysis.
this study was to investigate and compare the
lipidome coverage using high flow LC-MS/MS
and nanoLC-­MS/MS. 2.2 High Flow LC-MS/MS
Procedure

2 Material and Methods Chromatographic separation was achieved using


an ACQUITY UPLC BEH C-18 column
2.1 Sample Procurement (2.1 × 150 mm, 1.7 μm particle size) (Waters
and Preparation for LC-MS/MS Corporation, Milford, MA, USA) held at 50 °C
Analysis coupled to a Vanquish binary pump (Thermo
Fisher Scientific, San Jose, CA). A gradient
Donor eyes were obtained from Advancing Sight mobile phase was generated over 55 min with a
Network (formerly Alabama Eye Bank) <6 h flow rate of 250 μL/min and was comprised of
postmortem and immediately prepared for fixa- 10 mM ammonium formate in 40:60 (v/v) water,
tion and/or embedding. The anterior portion of acetonitrile with 0.1% formic acid and 10 mM
the eye was removed before placing in parafor- ammonium formate in 90:10 (v/v) isopropanol,
maldehyde for 48 h at 4 °C. We recently com- and acetonitrile with 0.1% formic acid. Ten μL of
pared lipidomic analysis of fresh-frozen and sample was injected via the Vanquish autosam-
paraformaldehyde fixed human retina tissue sec- pler maintained at 4 °C. HPLC eluate flowed into
tions and observed similar lipidome coverage [5]. a HESI-heated ESI source, and ions were ana-
Fifty μm sections of fixed human donor tissue lyzed using a Q Exactive HF instrument (Thermo
were cut and thaw-mounted on plain glass slides Scientific). Data were acquired at both full (MS1)
for the high flow LC-MS/MS analysis. One bio- and data-dependent MS2 (ddMS2) scan modes
logical replicate of above 80 years of age human using positive and negative modes, separately.
donor retina was used for the LC-MS/MS analy- The full scan mode had a mass resolution of
sis. Twelve-fourteen μm sections from a separate 60,000, mass range of m/z 400–1200 in positive
fixed human donor tissue were cut and subjected polarity and a mass range of m/z 240–1600 in
to nanoLC-MS/MS analysis. negative polarity, and a maximum trap fill time of
Untargeted Lipidomic Profiling of Aged Human Retina With and Without Age-Related Macular… 39

100 ms. ddMS2 data were acquired at 15,000 ion species, accurate m/z values for the precursor
resolution, with a maximum trap fill time of ions and subsequent ddMS2 spectra were
160 ms. The isolation window of selected MS1 acquired from the SPLASH® LIPIDOMIX® mix-
ions was ±1.4 m/z with a normalized collision ture of lipids. Lipid species were identified based
energy (NCE) of 20 and 25. LC-MS/MS data on their fragmentation pattern with a mass toler-
were acquired using Xcalibur version 4.0. ance of 0.2 ppm in negative and positive ioniza-
tion modes. Confirmed lipid identifications were
made by examination of tandem mass spectra for
2.3 NanoLC-MS/MS Procedure head group and fatty acyl chains, mass error mea-
surement, isotopic profile, extracted ion chro-
Custom reverse phase columns (20 cm × 75 μm) matogram, and the retention time for precursor
were packed with 1.9 μm BEH C18 material. ions. LC-MS/MS data were processed to separate
NanoLC-MS/MS was performed using an EASY-­ each lipid identification into its distinct lipid
nLC 1000 (Thermo Scientific) coupled to Q class.
Exactive HF instrument (Thermo Scientific). A
gradient mobile phase comprised of 10 mM
ammonium formate in 40:60 (v/v) water, acetoni- 3 Results
trile with 0.1% formic acid and 10 mM ammo-
nium formate in 90:10 (v/v) isopropanol, and 3.1 Elution Profile of Lipids
acetonitrile with 0.1% formic at 300 nl/min for Identified by High Flow
120 minutes, and the column compartment was LC-MS/MS
heated at 60 °C. One μL of sample was injected.
Data were acquired using both full (MS1) and The high flow LC-MS/MS elution profile of lip-
data-dependent MS2 (ddMS2) scan modes in ids extracted from human RPE is represented in
positive and negative modes, separately. The full Fig. 1. The lysophospholipids including lyso PC,
scan mode had a mass resolution of 60,000, mass lyso PE, and acylcarnitine eluted first from the
range of m/z 400–1200 in positive polarity, and a column with a retention time of 3–10 min.
mass range of m/z 240–1600 in negative polarity. Plasmalogen lipids including plasmanyl PC,
NanoLC-MS/MS data were acquired using plasmenyl PC, plasmanyl PE and plasmenyl PE,
Xcalibur version 4.0. glycerophospholipids (phosphatidylcholines
(PC), phosphatidylethanolamine (PE), phospha-
tidylinositol (PI), phosphatidylserine (PS), phos-
2.4 Data Analysis phatidyl glycerol (PG)), diacylglycerols
(triglyceride and diacylglycerol), and ceramides
Raw LC-MS/MS data files (Thermo .raw files) eluted at 10–30 min. Cardiolipins and cholesteryl
were converted to mgf, mzXML files using the esters eluted at 32–45 min.
MSConvert function of ProteoWizard [8]. Mgf,
mzXML input files were searched using the spec-
trum searcher feature of LipiDex [9] software 3.2 Untargeted Lipidomic
v1.0.2 for lipid identifications using LipiDex_ Analysis of Healthy Human
HCD_Formic and LipiDex_Splash_ISTD_ Donor RPE-Choroid Using
Formic libraries. Precursor mass-to-charge (m/z), High Flow LC-MS/MS
fragmentation spectra, chromatographic reten-
tion time, and identification were integrated using We identified 215 lipids comprised of 4 lipid
LipiDex. Fragmentation patterns of individual classes and 15 lipid subclasses from human donor
lipid standards were manually interpreted and RPE-choroid from paraformaldehyde-fixed tis-
then matched against LipiDex library results. To sue using positive and negative ionization modes
ensure correct annotation of individual molecular of LC-MS/MS as presented in Fig. 2. The 4
40 A. Kotnala et al.

Fig. 1 Base peak chromatogram representative of human donor RPE-choroid samples using LC-MS/MS

Fig. 2 Overall lipidome coverage from high flow LC-MS/MS in negative and positive ionization modes using human
donor RPE-choroid tissue (≥80 years)

classes included 123 glycerophospholipids, 48 prised of 22 sphingomyelin (SM), 11 acylcarnitine


triglycerides, 41 sphingomyelins, and 7 choles- (AC), and 10 ceramides (Cer). A total of 7 cho-
teryl esters. The maximum number of lipids were lesteryl esters (CE) were identified.
identified from glycerophospholipids including
10 subclasses: 37 phosphatidylcholines (PC), 4
phosphatidylethanolamine (PE), 7 phosphati- 3.3 Comparison of Lipid Profiling
dylinositol (PI), 7 phosphatidylserine (PS), 20 from High Flow LC-MS/MS
phosphatidylglycerol (PG), 6 lysophosphatidyl- and nLC-MS/MS
choline (Lyso PC), 6 lysophosphatidylethanol-
amine (Lyso PE), and 9 plasmalogen PC We identified 384 lipids using nanoLC-MS/MS
(plasmanyl PC and plasmenyl PC). Triglycerides analysis and 215 lipids using high flow LC-MS/
(TG) totaled 48 and included 44 TG and 4 diacyl- MS, in separate paraformaldehyde-fixed samples
glycerols (DG). Forty-one sphingomyelins com- of human donor RPE-choroid. The lipid sub-
Untargeted Lipidomic Profiling of Aged Human Retina With and Without Age-Related Macular… 41

Fig. 3 Comparison of high flow LC-MS/MS vs. nanoLC-MS/MS of human RPE-choroid sample analysis in negative
and positive ionization modes

classes were compared from nanoLC-MS/MS However, lipid homeostasis, oxidative stress, and
and high flow LC-MS/MS analysis as shown in chronic inflammation have been associated with
Fig. 3. Lipids from various lipid classes including drusen formation [1] and disease progression.
Cer, DG, lysophospholipids, PC, SP, TG, PE, PI, Although there is a treatment available for the
PS, and HexCer were either absent or were fewer neovascular form of AMD, there is no treatment
in numbers for high flow LC-MS/MS as com- available to prevent disease progression at early
pared to nanoLC-MS/MS. The major subclasses stages [11]. Interestingly, a newer understanding
of lipids identified by nanoLC-MS/MS include: of drusen composition has led to therapeutic tar-
10 AC, 3 CE, 16 Cer, 11 DG, 21 lysoPC, 6 Lyso geting of lipids, a major drusen component [12].
PE, 1 lyso SM, 6 Lyso PG, 4 Lyso PI, 4 Lyso PS, In-depth molecular characterization of retinal
49 PC, 2 PI, 22 SM, 5 SP, 86 TG, 2 Cer, 18 PE, extracellular deposits in aged retina is necessary
11 PE-Nme, 10 PG, 24 PI, 13 PC-O/P, 13 PE-O/P, to understand their mechanism of formation and
16 PS, 9 HexCer, and 22 SM. nanoLC-MS/MS role in AMD progression. Liquid chromatogra-
outperformed high flow LC-MS/MS in the num- phy tandem mass spectrometry (LC-MS/MS)
ber of lipids identified in each class except for AC provides specific molecular identifications of
and PG lipids. Note that roughly 30% of the high hundreds of lipids in an untargeted fashion.
flow LC-MS/MS sample volume was used for Overall, untargeted lipidomic analysis from
nano-LC-MS/MS analysis, yet 78% more lipids paraformaldehyde-­ fixed RPE-choroid tissue
were identified using nano LC-MS/MS as com- identified 215 lipids from 4 lipid classes and 15
pared to high flow LC-MS/MS analysis. lipid subclasses using high flow LC-MS/MS. To
further increase the sensitivity of lipid analysis
and for analysis of limited tissue volumes, we
4 Discussion have developed nanoLC-MS/MS to examine dis-
sected human RPE-choroid samples. We
AMD is a leading cause of blindness among the observed a 78% increase in lipid number while
elderly population [1]. AMD is characterized by going from high flow LC-MS/MS to nanoLC-
the accumulation of extracellular deposits called ­MS/MS with total lipid identifications increasing
drusen located between the basal lamina of the from 215 to 384 with 70% less sample injected.
RPE and the inner collagenous layer of Bruch’s The increase in the number of lipids identified
membrane [10]. The pathophysiology behind was observed for nearly all lipid classes detected.
drusen formation is only partly understood. Further identifications of lipids in aged and AMD
42 A. Kotnala et al.

eyes could help elucidate the sources of lipids 3. Curcio CA, Sloan KR, Kalina RE, Hendrickson
AE. Human photoreceptor topography. J Comp
involved in extracellular deposit formation in Neurol. 1990;292(4):497–523.
AMD. 4. Anderson DMG, Messinger JD, Patterson NH, Rivera
ES, Kotnala A, Spraggins JM, et al. Lipid landscape
of the human retina and supporting tissues revealed by
high-resolution imaging mass spectrometry. J Am Soc
5 Conclusion Mass Spectrom. 2020;31:2426.
5. Kotnala A, Anderson DMG, Patterson NH, Cantrell
Molecular features of lipid signals were identi- LS, Messinger JD, Curcio CA, et al. Tissue fixation
fied and compared using high flow LC-MS/MS effects on human retinal lipid analysis by MALDI
imaging and LC-MS/MS technologies. J Mass
and nanoLC-MS/MS. The increased sensitivity Spectrom. 2021;56(12):e4798.
provided by nanoLC-MS/MS will provide exten- 6. Danne-Rasche N, Coman C, Ahrends R. Nano-LC/
sive lipidome coverage from small retinal depos- NSI MS refines lipidomics by enhancing lipid cov-
its that are involved in AMD formation. erage, measurement sensitivity, and linear dynamic
range. Anal Chem. 2018;90(13):8093–101.
7. Vasilopoulou CG, Sulek K, Brunner A-D, Meitei NS,
Acknowledgments The authors acknowledge the finan- Schweiger-Hufnagel U, Meyer SW, et al. Trapped
cial support from NIH grants R01EY027948 (CAC), P41 ion mobility spectrometry and PASEF enable in-­
GM103391, S10 OD023514 (KLS), Heidelberg depth lipidomics from minimal sample amounts. Nat
Engineering, (UAB institutional support), Research to Commun. 2020;11(1):331.
Prevent Blindness (CAC & KLS), and EyeSight 8. Adusumilli R, Mallick P. Data conversion with
Foundation of Alabama. ProteoWizard msConvert. Methods Mol Biol (Clifton,
NJ). 2017;1550:339–68.
Financial Disclosures (CAC) research funding 9. Hutchins PD, Russell JD, Coon JJ. LipiDex: an inte-
grated software package for high-confidence lipid
from Heidelberg Engineering and Hoffman La identification. Cell Syst. 2018;6(5):621–5.e5.
Roche 10. Chen L, Messinger JD, Kar D, Duncan JL, Curcio
CA. Biometrics, impact, and significance of basal
linear deposit and subretinal drusenoid deposit in age-­
related macular degeneration. Invest Ophthalmol Vis
Sci. 2021;62(1):33.
References 11. Cabral de Guimaraes TA, Daich Varela M, Georgiou
M, Michaelides M. Treatments for dry age-related
1. Fleckenstein M, Keenan TDL, Guymer RH, macular degeneration: therapeutic avenues, clini-
Chakravarthy U, Schmitz-Valckenberg S, Klaver CC, cal trials and future directions. Br J Ophthalmol.
et al. Age-related macular degeneration. Nat Rev Dis 2021:bjophthalmol-2020-318452.
Primers. 2021;7(1):31. 12. Rudolf M, Curcio CA, Schlotzer-Schrehardt U, Sefat
2. Curcio CA. Soft drusen in age-related macu- AMM, Tura A, Aherrahrou Z, et al. Apolipoprotein
lar degeneration: biology and targeting via the A-I mimetic peptide L-4F removes bruch’s membrane
oil spill strategies. Invest Ophthalmol Vis Sci. lipids in aged nonhuman primates. Invest Ophthalmol
2018;59(4):Amd160-amd81. Vis Sci. 2019;60(2):461–72.
Decoding Race and Age-Related
Macular Degeneration: GPR 143
Activity Is the Key

Dorothy Tung and Brian S. McKay

Abstract ments. Together, our data suggests a central


role for GPR143 signaling in RPE-­
Age-related macular degeneration (AMD) is a photoreceptor interaction which is critical to
leading cause of irreversible blindness in the healthy vision.
developed world. Caucasians are eightfold
more likely to develop AMD than any other Keywords
race, indicating a racial bias in AMD inci-
dence which is unexplained. We hypothesize Age-related macular degeneration · Retinal
that pigmentation of the retinal pigment epi- pigment epithelium · GPCR · GPR143 ·
thelium (RPE) and choroid protects from L-dopa · Photoreceptor outer segments · Race
AMD and underlies this peculiar racial bias. · Pigmentation · Melanin
We investigated GPR143, a receptor in the
pigmentation pathway, which is activated by a
melanin synthesis by-product, l-dopa. In this
model, greater pigmentation leads to greater Abbreviations
l-dopa production and, in turn, greater
GPR143 signaling. GPR143 activity upregu- AMD Age-related macular degeneration
lates PEDF and downregulates both VEGF RPE Retinal pigment epithelium
and exosomes; all of which reduce the angio- l-dopa l-3,4-dihydroxyphenylalanine
genic potential in the retina. Moreover, we PEDF Pigment epithelium derived factor
demonstrate that GPR143 signaling enhances VEGF Vascular endothelial growth factor
the digestion of shed photoreceptor outer seg- POS Photoreceptor outer segments

D. Tung
Department of Ophthalmology and Vision Science, 1 Introduction
University of Arizona, Tucson, AZ, USA
e-mail: dorothytung@email.arizona.edu Age-related macular degeneration (AMD) is a
B. S. McKay (*) leading cause of irreversible blindness in devel-
Department of Ophthalmology and Vision Science, oped nations [1–3]. Currently, greater than 10%
University of Arizona, Tucson, AZ, USA of the United States population over the age of
Department of Physiology, University of Arizona, 70 has AMD, and its prevalence will increase as
Tucson, AZ, USA the population ages. Most AMD patients develop
e-mail: bsmckay@eyes.arizona.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 43


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_7
44 D. Tung and B. S. McKay

geographic atrophy, which involves deteriora- mentation results in a characteristic retinal phe-
tion of the macula and gradually impairs vision. notype, including reduced numbers of
The remaining 10–15% of AMD patients photoreceptor and ganglion cells, misrouting of
develop neovascular AMD, which involves the optic chiasm, and foveal hypoplasia [16, 20,
growth of new blood vessels that cause bleed- 21]. Curiously, ocular albinism includes all of the
ing, swelling, and scarring of the macula [4–6]. same retinal defects, despite the fact that the RPE
While neovascular AMD only accounts for a has normal pigmentation [22–24]. Thus, the reti-
minority of AMD cases, it causes 85–90% of nal albinism phenotype is not due to a lack of
vision loss from AMD. AMD incidence exhibits RPE pigmentation but, rather, a lack of GPR143
a striking racial bias, with Caucasians affected signaling. Either there is a loss of melanin and
eight times more often than any other popula- l-dopa production or a loss of the l-dopa receptor,
tion after the age of 80 [7–10]. We hypothesize GPR143, both of which yield the same
that RPE/choroidal pigmentation, is likely dif- phenotype.
ferent among races, and plays a role in this bias
[11, 12]. Despite years of study, the etiology of
AMD is unknown, and we cannot prevent the 2 GPR143
disease [6, 13, 14]. However, we do know that
being Caucasian is an even greater risk factor GPR143 expression is limited to melanin-­
for AMD than age is past 75 years of age containing cells [25]. The expression of GPR143
(Fig. 1). in cells that also produce melanin and l-dopa
To understand the racial bias of AMD, we indicates that GPR143 signals in an autocrine
investigated the melanin synthesis pathway in the manner. Mutations in the gene encoding GPR143
RPE. This pathway includes a little studied cause ocular albinism, a condition where pig-
G-protein-coupled receptor (GPCR), GPR143. mented cells accumulate melanin, but in mishap-
We identified l-dopa as the ligand of GPR143, pen macromelanosomes [25, 26]. This
which is itself a by-product of melanin synthesis observation indicates GPR143 signaling has a
[15]. Defects in GPR143 cause ocular albinism, function in endosomal trafficking [26–28]. The
resulting in low vision similar to all other forms lack of effects on cutaneous or ocular pigmenta-
of albinism which are associated with reduced tion underscores the point that GPR143 signaling
pigment production [16–19]. Loss of RPE pig- does not control melanin production.

Fig. 1 AMD incidence 16%


by race and age. Graph
portrays the percentage 14%
of the identified
population by age and 12%
White
race that have AMD in
2010. (Adapted from the 10% Black
National Eye Institute) Hispanic
8% Other
6%

4%

2%

0%
50-54 55-59 60-64 65-69 70-74 75-79 80+
Decoding Race and Age-Related Macular Degeneration: GPR 143 Activity Is the Key 45

Fig. 2 An overview of
two critical pathways in
the RPE: GPR143
activation and outer
segment degradation.
GPR143 signaling
increases expression of
PEDF, and decreases
both VEGF and
exosome release (blue
dots). GPR143 also
enhances POS digestion

3 GPR143 and Trophic Factors 4 L-Dopa Halts Extracellular


Vesicle Release
GPR143 signaling increases expression of pig-
ment epithelium-derived factor (PEDF), the most Exosomes are nanometer-size extracellular vesi-
potent neurotrophic factor in the eye (Fig. 2) cles secreted by almost all cells [30]. Exosomes
[15]. GPR143 signaling and PEDF make an play a critical role in tissue angiogenesis and are
attractive possibility to account for albinism heavily secreted by most cancerous cells [31, 32].
effects on retinal development and survival [12, Exosomes carry a variety of cargo—including
29]. Recently, Lee et al. demonstrated that sup- proteins, lipids, and nucleic acids—and also play
plemental l-dopa rescues normal retinal develop- a role in inter-tissue communication. GPR143
ment in albino animals, directly showing that activation immediately halts RPE exosome
GPR143 signaling supports proper retinal devel- release [33].
opment [27]. Like the racial bias of AMD, GPR143 effects
GPR143 signaling decreases RPE secretion on RPE exosome release are not subtle. The sheer
of vascular endothelial growth factor (VEGF), magnitude of GPR143’s effect on exosome
a potent angiogenic factor [28]. Reducing ocu- release, compared to the modest effect on trophic
lar VEGF is likely to foster a reduced angio- factors, suggests that exosome release better
genic environment in the RPE. While these matches GPR143 activity to the racial bias of
changes in PEDF and VEGF were significant, AMD [11, 12, 29, 34]. However, we lack any
they were subtle. Thus, the robust eightfold information on the potential racial disparities in
racial bias in AMD incidence seems unlikely to GPR143 expression. There are multiple promoter
be from the subtle alterations in PEDF and and gene polymorphisms in GPR143 that link to
VEGF. race and ethnicity that could affect GPR143
46 D. Tung and B. S. McKay

expression and activity. Unfortunately, while delayed in patients prescribed l-dopa. L-dopa
these changes have the potential to link GPR 143 delayed the age of onset from 71.4 years to
activity to the racial bias of AMD, they have not 79.3 years in patients with geographic atrophy
been studied. and from 75.5 years to 80.8 years in patients with
neovascular AMD.
In a proof of principle prospective clinical
5 GPR 143 Signaling trial, l-dopa significantly increased the best cor-
in Photoreceptor Outer rected visual acuity, decreased subretinal fluid,
Segment Digestion and decreased the mean central retinal thickness
in patients with neovascular AMD [40].
Photoreceptors undergo a daily renewal pro-
cess by shedding the distal 10% of their outer
segments [35]. The RPE plays a critical role in 7 Conclusion
the daily phagocytosis and degradation of the
shed photoreceptor outer segments (POS). Our findings suggest that GPR143 signaling in
Often overlooked, this daily activity makes the response to l-dopa significantly delays or pre-
RPE the most phagocytic tissue in the body vents AMD pathogenesis. Potential mechanisms
[12, 36–38]. This digestive process involves a underlying GPR143 activity include upregulating
series of temporal steps: recognition, binding, PEDF, downregulating VEGF, halting RPE exo-
intracellular signaling, internalization, and some release, and increasing the rate of POS deg-
digestion [36, 37]. Inefficient digestion of POS radation. Since GPR143 is an important
leads to the aggregation of lipids and proteins component of the pigmentation pathway, these
within the RPE, hence playing a role in the observations may underlie the racial bias of the
pathogenesis of various retinopathies includ- disease. Taken as a whole, our results indicate
ing AMD. that GPR143 may be a promising target for future
We tested the effect of GPR143 on the POS research centered on novel drugs and therapies to
uptake and digestion process. We challenged pig- prevent AMD in those at greatest risk.
mented RPE with fluorescently labeled POS with
and without l-dopa to stimulate GPR143 signal-
ing. L-dopa had no effect on the rate of POS References
uptake by the RPE over 2 h. However, when
using a pH-sensitive dye that only fluoresces in 1. Klein R, Klein BEK. The prevalence of age-related
eye diseases and visual impairment in aging: cur-
the acidic lysosomal compartment, l-dopa rent estimates. Investig Ophthalmol Vis Sci. https://
increased the rate of POS degradation, as quanti- doi.org/10.1167/iovs.13-­12789. Epub ahead of print
fied by a more rapid decrease in the area of fluo- 2013.
rescence over time. Our results suggest that 2. Resnikoff S, Pascolini D, Etya’ale D, et al. Global
data on visual impairment in the year 2002. Bull
GPR143 may have an effect on POS digestion, World Health Organ. 2004;82:844–51.
perhaps by increasing the efficiency of endo- 3. Jager RD, Mieler WF, Miller JW. Age-related macular
somal trafficking. degeneration. N Engl J Med. 2008;358:2606–17.
4. Mitchell P, Korobelnik JF, Lanzetta P, et al.
Ranibizumab (Lucentis) in neovascular age-related
macular degeneration: evidence from clinical tri-
6 Clinical Studies with L-Dopa als. Br J Ophthalmol. https://doi.org/10.1136/
and AMD bjo.2009.159160. Epub ahead of print 2010.
5. Kovach JL, Schwartz SG, Flynn HW, et al. Anti-VEGF
treatment strategies for wet AMD. J Ophthalmol.
To explore whether GPR143 signaling affects 2012;2012:786870.
AMD incidence, we conducted a retrospective 6. Group TCR. Ranibizumab and bevacizumab for neo-
analysis of patients prescribed l-dopa [39]. We vascular age-related macular degeneration. N Engl J
found that AMD onset was both reduced and Med. 2011;364:1897–908.
Decoding Race and Age-Related Macular Degeneration: GPR 143 Activity Is the Key 47

7. Frank RN, Puklin JE, Stock C, et al. Race, iris color, 24. White D, Rabago-Smith M. Genotype-phenotype
and age-related macular degeneration. Trans Am associations and human eye color. J Hum Genet.
Ophthalmol Soc. 2000;98:109–17. 2011;56:5–7.
8. Chang MA, Bressler SB, Munoz B, et al. Racial differ- 25. Schiaffino MV, Baschirotto C, Pellegrini G, et al. The
ences and other risk factors for incidence and progres- ocular albinism type 1 gene product is a membrane
sion of age-related macular degeneration: Salisbury glycoprotein localized to melanosomes. Proc Natl
Eye Evaluation (SEE) Project. Invest Ophthalmol Vis Acad Sci U S A. 1996;93:9055–60.
Sci. 2008;49:2395–402. 26. Schiaffino MV, Tacchetti C. The ocular albinism type
9. Milton RC, Clemons TE, Klien R, et al. Risk factors 1 (OA1) protein and the evidence for an intracellular
for the incidence of advanced age-related macular signal transduction system involved in melanosome
degeneration in the Age-Related Eye Disease Study biogenesis. Pigment Cell Res. 2005;18:227–33.
(AREDS): AREDS report no. 19. Ophthalmology. 27. Lee H, Scott J, Griffiths H, et al. Oral levodopa rescues
2005;112:533–9. retinal morphology and visual function in a murine
10. Fisher DE, Klein BEKK, Wong TY, et al. Incidence model of human albinism. Pigment Cell Melanoma
of age-related macular degeneration in a multi-ethnic Res. https://doi.org/10.1111/pcmr.12782. Epub ahead
United States population: the multi-ethnic study of of print 2019.
atherosclerosis. Ophthalmology. 2016;123:1297–308. 28. Falk T, Congrove NR, Zhang S, et al. PEDF and
11. McKay BS, Schwartz SG. Pigmentation and macu- VEGF-A output from human retinal pigment epi-
lar degeneration: is there a role for GPR143? J Ocul thelial cells grown on novel microcarriers. J Biomed
Pharmacol Ther. 2016;32:3–4. Biotechnol. 2012;2012:278932.
12. Figueroa AG, McKay BS. A G-protein coupled recep- 29. Figueroa AG, McKay BS. GPR143 signaling and reti-
tor and macular degeneration. Cell. 2020:1–13. nal degeneration. Adv Exp Med Biol. 2019;1185:15.
13. Chew EY, Clemons TE, Agrón E, et al. Long-term https://doi.org/10.1007/978-­3-­030-­27378-­1_3. Epub
effects of vitamins C and E, β-carotene, and zinc on ahead of print 2019
age-related macular degeneration: AREDS report 30. Ramirez SH, Andrews AM, Paul D, et al. Extracellular
no. 35. Ophthalmology. 120:1604. https://doi. vesicles: mediators and biomarkers of pathology
org/10.1016/j.ophtha.2013.01.021. Epub ahead of along CNS barriers. Fluids Barriers CNS. 2018;15:19.
print 2013. 31. Sahebi R, Langari H, Fathinezhad Z, et al. Exosomes:
14. Rofagha S, Bhisitkul RB, Boyer DS, et al. Seven-­ new insights into cancer mechanisms. J Cell Biochem.
year outcomes in ranibizumab-treated patients in 2020;121:7–16.
ANCHOR, MARINA, and HORIZON: a multi- 32. Todorova D, Simoncini S, Lacroix R, et al.
center cohort study (SEVEN-UP). Ophthalmology. Extracellular vesicles in angiogenesis. Circ Res
2013;120:2292–9. https://doi.org/10.1161/CIRCRESAHA.117.309681.
15. Lopez VM, Decatur CL, Stamer WD, et al. L-DOPA Epub ahead of print 2017.
is an endogenous ligand for OA1. PLoS Biol. 33. Locke CJ, Congrove NR, Dismuke WM, et al.
2008;6:1861–9. Controlled exosome release from the retinal pigment
16. Hoffmann MB, Lorenz B, Morland AB, et al. epithelium insitu. Exp Eye Res. 2014;129:1–4.
Misrouting of the optic nerves in albinism: estima- 34. McKay BS. Pigmentation and vision: is GPR143 in
tion of the extent with visual evoked potentials. Invest control? J Neurosci Res. 97 https://doi.org/10.1002/
Ophthalmol Vis Sci. 2005;46:3892–8. jnr.24246. Epub ahead of print 14 May 2018.
17. d’Addio M, Pizzigoni A, Bassi MT, et al. Defective 35. Guha S, Liu J, Baltazar G, et al. Rescue of compro-
intracellular transport and processing of OA1 is mised lysosomes enhances degradation of photore-
a major cause of ocular albinism type 1. Hum Mol ceptor outer segments and reduces lipofuscin-like
Genet. 2000;9:3011–8. autofluorescence in retinal pigmented epithelial cells.
18. Oetting WS, Summers CG, King RA. Albinism and Adv Exp Med Biol. 2014;801:105–11.
the associated ocular defects. Metab Pediatr Syst 36. Mazzoni R, Safa H, Finnemann SC. Understanding
Ophthalmol. 1994;17:5–9. photoreceptor outer segment phagocytosis: use
19. Summers CG. Albinism: classification, clinical and utility of RPE cells in culture. Exp Eye Res.
characteristics, and recent findings. Optom Vis Sci. 2014;126:51–60.
2009;86:659–62. 37. Kevany BM, Kryzysztof. Phagocytosis of retinal rod
20. King RA, Pietsch J, Fryer JP, et al. Tyrosinase gene and cone photoreceptors. Physiology. 2010;25:8–15.
mutations in oculocutaneous albinism 1 (OCA1): defi- 38. Young RW, Bok D. Participation of the retinal pig-
nition of the phenotype. Hum Genet. 2003;113:502–13. ment epithelium in the rod outer segment renewal pro-
21. Peters S, Kayatz P, Heimann K, et al. Subretinal injec- cess. J Cell Biol. 1969;42:392–403.
tion of rod outer segments leads to an increase in the 39. Brilliant MH, Vaziri K, Connor TB, et al. Mining ret-
number of early-stage melanosomes in retinal pig- rospective data for virtual prospective drug repurpos-
ment epithelial cells. Ophthalmic Res. 2000;32:52–6. ing: L-DOPA and age-related macular degeneration.
22. Lewen RM. Ocular albinism. Arch Ophthalmol. Am J Med. 2016;129:292–8.
1988;106:120–1. 40. Figueroa AG, Boyd BM, Christensen CA, et al.
23. Mártinez-García M, Montoliu LL. Albinism in Levodopa positively affects neovascular age-related
Europe. J Dermatol. 2013;40:319. macular degeneration. Am J Med. 2021;134:122.
Peroxisome Proliferator-Activated
Receptor Gamma
Coactivator-­1Alpha (PGC-1α):
A Transcriptional Regulator
at the Interface of Aging
and Age-Related Macular
Degeneration?

Freya M. Mowat

Abstract Keywords

PGC-1alpha · PPARGC-1alpha · Retina ·


Human age-related macular degeneration
Metabolism · Aging · Age-related macular
(AMD) is a prevalent age-related disease
degeneration
which causes retinal dysfunction and disabil-
ity. Genetic and cell culture studies from AMD
patients have implicated impaired activity of
peroxisome proliferator-activated receptor 1 Introduction
gamma coactivator-1alpha (PGC-1α). PGC-1α
is a transcriptional co-regulator that acts to Peroxisome proliferator-activated receptor
control a plethora of metabolic processes rel- gamma coactivator-1alpha (PGC-1α) is a tran-
evant to AMD pathophysiology including glu- scriptional co-regulator. It mediates the activity
coneogenesis, oxidative phosphorylation, and of a broad range of metabolic pathways including
response to oxidative injury. Perturbation of gluconeogenesis, glucose transport, fatty acid
PGC-1α activity in mice causes AMD-like oxidation, mitochondrial biogenesis, and peroxi-
RPE and retinal pathology. There is potential somal remodeling and is important in the detoxi-
for therapeutic modulation of the PGC-1α fication of reactive oxygen species.
pathway in AMD treatment. The effects of PGC-1α are mediated through
transcription factors including nuclear receptors
such as peroxisome proliferator-activated recep-
tors (PPARs) and nuclear respiratory factors
(NRFs), hormones (thyroid hormone, estrogen),
and glucocorticoids among others. It is highly
F. M. Mowat (*) expressed in tissues with large energy needs such
Department of Ophthalmology and Visual Sciences as neurologic tissues. Ocular tissues, including
(School of Medicine and Public Health), Department retinal photoreceptors and retinal pigmented epi-
of Surgical Sciences (School of Veterinary Medicine), thelium (RPE) cells, express high levels of
Medical Sciences Center, University of Wisconsin-­
Madison, Madison, WI, USA PGC-1α [1, 2]. PGC-1α-related pathways are
e-mail: mowat@wisc.edu strongly implicated in the pathogenesis of human

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 49


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_8
50 F. M. Mowat

age-related macular degeneration (AMD), a activity is regulated at the posttranslational level


prevalent age-related retinal neurodegenerative by the activity of sirtuins [9]. Expression of sirtu-
disorder [3]. ins is greater in the retina than in other tissues in
mice, and most sirtuins are modulated by light-­
dark cycle, with higher expression in the dark,
2 Regulation of Tissue Activity indicating a role for PGC-1α in dark adaptation
of PGC-1α or maintenance of the dark current [9]. Light
damage to the retinal photoreceptors is extensive
Because PGC-1α is pivotal in control of cellular in mice lacking PGC-1α [1]. It seems likely that
metabolic function, its activity is controlled by PGC-1α has an important role in the maintenance
multiple stimuli; many of which change during of photoreceptors and visual function, particu-
the aging process. These stimuli include nutrient larly rod photoreceptors.
availability, temperature, calcium signaling, reac- In the RPE, PGC-1α expression is enhanced
tive oxygen species, circadian rhythm, hormone by binding to photoreceptor outer segments [10].
activity, cytokines, energetic need, and oxygen PGC-1α activity in the RPE facilitates lysosomal
concentration (for a review see [4]). Activity of degradation and lipid metabolism in the process-
PGC-1α is regulated at multiple levels, including ing of outer segments following phagocytosis.
control of expression by alterations in promoter Based on these studies, there may be divergent
activity [5], changes in subcellular localization functions for PGC-1α between the photorecep-
[6], protein stability, and a plethora of posttrans- tors and RPE cells of the retina. Photoreceptor
lational modifications. PGC-1α is more active in the dark-adapted state,
Transcriptional control of PGC-1α expression mediating visual function and dark current,
is an important mechanism to regulate PGC-1α whereas RPE PGC-1α is more active during the
activity in health and disease. Healthy muscle daytime (at the time when rod photoreceptor
and brown fat have transcriptional regulatory sys- outer segments are shed), mediating cellular pro-
tems that fine-tune metabolic activity in response cessing and degradation of the lipid-rich rod
to exercise and temperature [5]. In disease, outer segments.
increased methylation of the PGC-1α promoter
region occurs in obesity, type 2 diabetes, nonal-
coholic fatty liver disease, and Parkinson’s dis- 3 Retinal Effects
ease [5]. Human AMD shares many risk factors of Perturbation of PGC-1α
with these diseases, although epigenetic modifi- Activity
cations affecting PGC-1α expression in human
AMD patients have not been determined. Mice lacking PGC-1α are hyperactive. This
Examples of posttranslational modifications hyperactivity is associated with lesions in the
that increase PGC-1α activity include deacety- striatal region of the brain that controls move-
lation of PGC-1α by the action of sirtuins during ment [11]. PGC-1α null mice are paradoxically
fasting or oxidative stress and phosphorylation of lean and have low fasting blood glucose concen-
PGC-1α by the activity of AMP-activated protein trations, although heterozygous mice are normo-
kinase (AMPK) and glycogen synthase kinase 3 glycemic. Dopaminergic signaling pathways are
beta (GSK3β) during energy deficiency or stress strongly implicated in the brain pathology identi-
[4]. Regulation of PGC-1α activity is highly tis- fied in these mice [12].
sue specific. Most studies disrupting PGC-1α activity in
In the outer retina, PGC-1α expression is reg- the eye have focused on the retinal pigmented
ulated at least in part by melatonin signaling via epithelium, as this is the primary tissue affected
MT1 receptors in the retina [7], which are highly by the dry (atrophic) form of human AMD. In
expressed in photoreceptors [8]. Retinal PGC-1α mice, conditional knockout of RPE PGC-1α and
Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1Alpha (PGC-1α): A Transcriptional… 51

PGC-1β using subretinally delivered viral vec- els of human AMD based on a C57Bl/6 J back-
tors with cre/lox-mediated excision causes disor- ground requires aging to approximately
ganization and degeneration of RPE cells and 22–24 months of age (>80% of anticipated
retinal degeneration [13]. Similarly, global median lifespan). It is also important to note that
double-­knockout of PGC1α and one of two interbreeding of different strains of mice may
PGC-1α nuclear receptor targets, NRF2 [14] or alter the anticipated lifespan and modify the age-­
NFE2L2 [15], also induces RPE disorganization, related phenotype, as has been shown in other
senescence, and degeneration. Mice lacking neurodegenerative mouse models [22].
PGC-1α and NRF2 possess other multisystemic
abnormalities including features of dwarfism,
grey coat color, and microphthalmia [14]. Young 4 Role of PGC-1α in Human
heterozygous PGC-1α null mice fed a high fat AMD
diet develop AMD-like RPE features including
basal laminar deposits, thickening of Bruch’s There is growing evidence that PGC-1α is impor-
membrane, in addition to RPE and photoreceptor tant in the pathophysiology of human AMD. The
degeneration [16]. These changes are associated major risk factor for development of human
with reduced mitochondrial activity, increased AMD is aging, with prevalence of early AMD
reactive oxygen species, decreased autophagy, increasing over the age of 45 years and preva-
and increased inflammation. lence of late AMD increasing over the age of
PGC-1α may also play a role in pathologic 75 years [23]. Other risk factors include cigarette
vascularization, which is a feature of the wet smoking, obesity, serum cholesterol concentra-
(neovascular) form of AMD. PGC-1α-deficient tion [24], and variants in genes related to the
mice have increased expression of vascular endo- complement pathway, lipid transport, angiogen-
thelial growth factor (VEGF) in the inner retina esis, and extracellular matrix remodeling [25].
[17] and outer retina [16]. In one study, PGC-1α Sequence variants in a DNA coding region of
null mice had a blunted neovascular response to PGC-1α and a 3′-UTR region upstream of
hypoxia in oxygen-induced retinopathy [2], PGC-1α are each independently associated with
although this finding was not corroborated in a increased risk of developing the wet (neovascu-
separate study [18]. lar) form of AMD [26]. No studies have exam-
Although retinal structure and function is ined the association between sequence variants in
unaffected in young (13-week-old) mice lacking PGC-1α and risk of developing the more com-
PGC-1α [1], the effect of natural aging of mon dry (atrophic) form of AMD.
PGC-1α knockout mice on RPE or retinal struc- Cultured cells from AMD patients have been
ture and function has not been reported. There is used to examine pathways relevant to disease.
strong supporting evidence for PGC-1α dysfunc- RPE fate-directed induced pluripotent stem cells
tion in the pathogenesis of age-related neurode- (iPSC) derived from AMD patients have reduced
generative diseases [4] [19, 20]. It is important to PGC-1α expression [27]. The expression of a key
note that the PGC-1α knockout mouse line is on activator of PGC-1α (Sirtuin1) is also reduced
a C57Bl/6 J mouse genetic background; the [27]. The effect of this is a detected increase in
median lifespan of this inbred strain is 29 months PGC-1α acetylation in primary RPE cells iso-
[21]. In the USA, median human life expectancy lated from AMD patients [28], which is predicted
is 78.8 years,1 and onset of clinically significant to inhibit PGC-1α activity. Paradoxically, one
dry AMD typically occurs at ages over 65 years, study found increased levels of PGC-1α protein
which is at >80% of anticipated median lifespan. in expanded RPE cultures from AMD patients,
Therefore, natural aging in potential mouse mod- despite a reduction in metabolic function [29],
although neither PGC-1α posttranslational modi-
fication nor activity was reported.
1
https://www.cdc.gov/nchs/fastats/life-expectancy.htm
52 F. M. Mowat

5 Potential Modulation and pathological angiogenesis in the retina. Am J


Pathol. 2013;182(1):255–65.
of the PGC-1α Pathway 3. Blasiak J, Pawlowska E, Sobczuk A, Szczepanska
in AMD Treatment J, Kaarniranta K. The aging stress response and its
implication for AMD pathogenesis. Int J Mol Sci.
Considering the relevance of the PGC-1α path- 2020;21(22):8840.
4. Anderson R, Prolla T. PGC-1alpha in aging and
way to AMD pathophysiology, therapeutic tar- anti-aging interventions. Biochim Biophys Acta.
geting of PGC-1α activity may provide 2009;1790(10):1059–66.
neuroprotection [30]. Upregulation of PGC-1α in 5. Kramer AI, Handschin C. How epigenetic modifica-
primary fetal human RPE cells increases tions drive the expression and mediate the action of
PGC-1alpha in the regulation of metabolism. Int J
­oxidative metabolism [31]. However, the nuances Mol Sci. 2019;20(21):5449.
of differential tissue effects of increased PGC-1α 6. Anderson RM, Barger JL, Edwards MG, Braun KH,
activity must be considered, as overexpression of O’Connor CE, Prolla TA, et al. Dynamic regulation
PGC-1α accelerated cone photoreceptor death in of PGC-1alpha localization and turnover implicates
mitochondrial adaptation in calorie restriction and the
rd1 mutant mice [32], and overexpression in stress response. Aging Cell. 2008;7(1):101–11.
dopaminergic neurons in the brain also causes 7. Kunst S, Wolloscheck T, Kelleher DK, Wolfrum U,
neurodegeneration [33]. Sargsyan SA, Iuvone PM, et al. Pgc-1alpha and Nr4a1
There are numerous candidate therapeutics are target genes of circadian melatonin and dopamine
release in murine retina. Invest Ophthalmol Vis Sci.
that target the PGC-1α pathway. The dyslipid- 2015;56(10):6084–94.
emia drug PU-19 is proposed to upregulate 8. Baba K, Pozdeyev N, Mazzoni F, Contreras-Alcantara
PGC-1α and, when administered in vitro, S, Liu C, Kasamatsu M, et al. Melatonin modulates
increases mitochondrial copy number and mito- visual function and cell viability in the mouse retina
via the MT1 melatonin receptor. Proc Natl Acad Sci U
chondrial function [34]. Similarly, upregulation S A. 2009;106(35):15043–8.
of RPE cell PGC-1α occurs after in vitro admin- 9. Ban N, Ozawa Y, Inaba T, Miyake S, Watanabe M,
istration of a novel compound ZLN005 [35], the Shinmura K, et al. Light-dark condition regulates
mitochondrial-derived peptide SHLP2 [36], or sirtuin mRNA levels in the retina. Exp Gerontol.
2013;48(11):1212–7.
the phytochemical sulforaphane when RPE cells 10. Roggia MF, Ueta T. alphavbeta5 Integrin/FAK/PGC-­
are exposed to toxic blue light [37]. 1alpha pathway confers protective effects on retinal
Although not specifically studied in AMD pigment epithelium. PloS One. 2015;10(8):e0134870.
models, there are several drugs that act on the 11. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J,
Zhang CY, et al. Defects in adaptive energy metabo-
PGC-1α pathway that may be promising as thera- lism with CNS-linked hyperactivity in PGC-1alpha
peutics for AMD. Resveratrol, a natural polyphe- null mice. Cell. 2004;119(1):121–35.
nol compound, activates PGC-1α via Sirtuin1 12. Jiang H, Kang SU, Zhang S, Karuppagounder S,
[38]. 5-Aminoimidazole-4-carboxamide ribonu- Xu J, Lee YK, et al. Adult conditional knockout of
PGC-1α leads to loss of dopamine neurons. eNeuro.
cleoside (AICAR) does so by generating inosine 2016;3(4):ENEURO.0183-16.2016.
monophosphate, which acts as an AMPK agonist 13. Rosales MAB, Shu DY, Iacovelli J, Saint-Geniez
[39]. Pyrroloquinoline quinone [40] and adipo- M. Loss of PGC-1α in RPE induces mesenchymal
nectin receptor agonists [41] also hold promise as transition and promotes retinal degeneration. Life Sci
Alliance. 2019;2(3):e201800212.
other potential modifiers of the PGC-1α 14. Felszeghy S, Viiri J, Paterno JJ, Hyttinen JMT,
pathway. Koskela A, Chen M, et al. Loss of NRF-2 and PGC-­
1alpha genes leads to retinal pigment epithelium dam-
age resembling dry age-related macular degeneration.
Redox Biol. 2019;20:1–12.
References 15. Blasiak J, Koskela A, Pawlowska E, Liukkonen M,
Ruuth J, Toropainen E, et al. Epithelial-mesenchymal
1. Egger A, Samardzija M, Sothilingam V, Tanimoto N, transition and senescence in the retinal pigment epi-
Lange C, Salatino S, et al. PGC-1alpha determines thelium of NFE2L2/PGC-1α double knock-out mice.
light damage susceptibility of the murine retina. PLoS Int J Mol Sci. 2021;22(4):1684.
One. 2012;7(2):e31272. 16. Zhang M, Chu Y, Mowery J, Konkel B, Galli S,
2. Saint-Geniez M, Jiang A, Abend S, Liu L, Sweigard Theos AC, et al. Pgc-1alpha repression and high-­
H, Connor KM, et al. PGC-1alpha regulates normal fat diet induce age-related macular degeneration-­
Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1Alpha (PGC-1α): A Transcriptional… 53

like phenotypes in mice. Dis Model Mech. getics and enhanced resistance to oxidative stress in
2018;11(9):dmm032698. human retinal pigment epithelial cells from donors
17. Guo X, Dason ES, Zanon-Moreno V, Jiang Q, Nahirnyj with age-related macular degeneration. Redox Biol.
A, Chan D, et al. PGC-1alpha signaling coordinates 2017;13:255–65.
susceptibility to metabolic and oxidative injury in the 30. Abrahan C, Ash JD. The potential use of PGC-1α and
inner retina. Am J Pathol. 2014;184(4):1017–29. PGC-1β to protect the retina by stimulating mitochon-
18. Chen J, Michan S, Juan AM, Hurst CG, Hatton CJ, drial repair. Adv Exp Med Biol. 2016;854:403–9.
Pei DT, et al. Neuronal sirtuin1 mediates retinal vas- 31. Iacovelli J, Rowe GC, Khadka A, Diaz-Aguilar D,
cular regeneration in oxygen-induced ischemic reti- Spencer C, Arany Z, et al. PGC-1alpha induces human
nopathy. Angiogenesis. 2013;16(4):985–92. RPE oxidative metabolism and antioxidant capacity.
19. Austin S, St-Pierre J. PGC1α and mitochondrial Invest Ophthalmol Vis Sci. 2016;57(3):1038–51.
metabolism–emerging concepts and relevance in 32. Xiong W, MacColl Garfinkel AE, Li Y, Benowitz LI,
ageing and neurodegenerative disorders. J Cell Sci. Cepko CL. NRF2 promotes neuronal survival in neu-
2012;125(Pt 21):4963–71. rodegeneration and acute nerve damage. J Clin Invest.
20. Róna-Vörös K, Weydt P. The role of PGC-1α in the 2015;125(4):1433–45.
pathogenesis of neurodegenerative disorders. Curr 33. Ciron C, Lengacher S, Dusonchet J, Aebischer
Drug Targets. 2010;11(10):1262–9. P, Schneider BL. Sustained expression of PGC-­
21. Yuan R, Tsaih SW, Petkova SB, Marin de Evsikova 1alpha in the rat nigrostriatal system selectively
C, Xing S, Marion MA, et al. Aging in inbred strains impairs dopaminergic function. Hum Mol Genet.
of mice: study design and interim report on median 2012;21(8):1861–76.
lifespans and circulating IGF1 levels. Aging Cell. 34. Nashine S, Subramaniam SR, Chwa M, Nesburn A,
2009;8(3):277–87. Kuppermann BD, Federoff H, et al. PU-91 drug res-
22. Rodriguez-Gil JL, Watkins-Chow DE, Baxter cues human age-related macular degeneration RPE
LL, Elliot G, Harper UL, Wincovitch SM, et al. cells; implications for AMD therapeutics. Aging
Genetic background modifies phenotypic severity (Albany NY). 2019;11(17):6691–713.
and longevity in a mouse model of Niemann-Pick 35. Zhang LN, Zhou HY, Fu YY, Li YY, Wu F, Gu M,
disease type C1. Disease models & mechanisms. et al. Novel small-molecule PGC-1alpha transcrip-
2020;13(3):dmm042614. tional regulator with beneficial effects on diabetic db/
23. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng db mice. Diabetes. 2013;62(4):1297–307.
CY, et al. Global prevalence of age-related macular 36. Nashine S, Cohen P, Nesburn AB, Kuppermann BD,
degeneration and disease burden projection for 2020 Kenney MC. Characterizing the protective effects of
and 2040: a systematic review and meta-analysis. SHLP2, a mitochondrial-derived peptide, in macular
Lancet Glob Health. 2014;2(2):e106–16. degeneration. Sci Rep. 2018;8(1):15175.
24. Schultz NM, Bhardwaj S, Barclay C, Gaspar L, 37. Yang PM, Cheng KC, Huang JY, Wang SY, Lin YN,
Schwartz J. Global burden of dry age-related macular Tseng YT, et al. Sulforaphane inhibits blue light-­
degeneration: a targeted literature review. Clin Ther. induced inflammation and apoptosis by upregulat-
2021;43(10):1792–818. ing the SIRT1/PGC-1α/Nrf2 pathway and autophagy
25. Singh N, Swaroop A, Ratnapriya R. Making biologi- in retinal pigment epithelial cells. Toxicol Appl
cal sense of genetic studies of age-related macular Pharmacol. 2021;421:115545.
degeneration. Adv Exp Med Biol. 2021;1256:201–19. 38. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane
26. SanGiovanni JP, Chen J, Sapieha P, Aderman CM, H, Lerin C, Daussin F, et al. Resveratrol improves
Stahl A, Clemons TE, et al. DNA sequence variants mitochondrial function and protects against metabolic
in PPARGC1A, a gene encoding a coactivator of the disease by activating SIRT1 and PGC-1alpha. Cell.
omega-3 LCPUFA sensing PPAR-RXR transcription 2006;127(6):1109–22.
complex, are associated with NV AMD and AMD-­ 39. Viscomi C, Bottani E, Civiletto G, Cerutti R, Moggio
associated loci in genes of complement and VEGF M, Fagiolari G, et al. In vivo correction of COX defi-
signaling pathways. PLoS One. 2013;8(1):e53155. ciency by activation of the AMPK/PGC-1alpha axis.
27. Golestaneh N, Chu Y, Cheng SK, Cao H, Poliakov E, Cell Metab. 2011;14(1):80–90.
Berinstein DM. Repressed SIRT1/PGC-1alpha path- 40. Chowanadisai W, Bauerly KA, Tchaparian E, Wong
way and mitochondrial disintegration in iPSC-derived A, Cortopassi GA, Rucker RB. Pyrroloquinoline qui-
RPE disease model of age-related macular degenera- none stimulates mitochondrial biogenesis through
tion. J Transl Med. 2016;14(1):344. cAMP response element-binding protein phosphory-
28. Zhang M, Jiang N, Chu Y, Postnikova O, Varghese lation and increased PGC-1alpha expression. J Biol
R, Horvath A, et al. Dysregulated metabolic path- Chem. 2010;285(1):142–52.
ways in age-related macular degeneration. Sci Rep. 41. Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T,
2020;10(1):2464. Hamagami K, Matsuda K, et al. A small-molecule
29. Ferrington DA, Ebeling MC, Kapphahn RJ, Terluk AdipoR agonist for type 2 diabetes and short life in
MR, Fisher CR, Polanco JR, et al. Altered bioener- obesity. Nature. 2013;503(7477):493–9.
Regulation of ABCA1 by miR-33
and miR-34a in the Aging Eye

Florian Peters and Christian Grimm

Abstract Abbreviations

Many age-related diseases, including age-­ ABCA1 ATP-binding cassette transporter,


related macular degeneration (AMD), go along family A, member 1
with local lipid accumulation and dysregulated AMD Age-related macular degeneration
lipid metabolism. Several genes involved in Apo Apolipoprotein
lipid metabolism, including ATP-­binding cas- ASOs Antisense oligonucleotides
sette transporter A1 (ABCA1), were associated CNV Choroidal neovascularization
with AMD through genome-­wide association HDL High-density lipoprotein
studies. Recent studies have shown that loss of LDL Low-density lipoprotein
ABCA1 in the retinal pigment epithelium (RPE) LXR Liver X receptor
leads to lipid accumulation and RPE atrophy, a miRNA microRNA
hallmark of AMD, and that antagonizing RCT Reverse cholesterol transport
ABCA1-targeting microRNAs (miRNAs) atten- RPE Retinal pigment epithelium
uated pathological changes to the RPE or to
macrophages. Here, we focus on two lipid
metabolism-­modulating miRNAs, miR-33 and 1 Introduction
miR-34a, which show increased expression in
aging RPE cells, and on their potential to regu- Age-related macular degeneration (AMD) is a
late ABCA1 levels, cholesterol efflux, and lipid complex and multifactorial disease and the lead-
accumulation in AMD pathogenesis. ing cause of blindness in the elderly in the Western
countries with an estimated 288 million people
Keywords affected by the year 2040 [38]. Although the exact
causes of AMD remain incompletely understood,
ABCA1 · AMD · Cholesterol · HDL · Lipids ·
emerging genetic, epidemiological, and mecha-
Lipid accumulation · LXR · miRNA · Reverse
nistic evidence suggests that dysregulated lipid
cholesterol transport · RPE
metabolism and associated RPE dysfunction
might play a key role in AMD pathogenesis. A
F. Peters (*) · C. Grimm major function of the RPE is the daily phagocyto-
Laboratory for Retinal Cell Biology, Department of sis of lipid-rich photoreceptor outer segments and
Ophthalmology, University Hospital Zurich, the recycling of unmetabolized lipids to photore-
University of Zurich, Schlieren, Switzerland ceptors or into the choroidal vasculature [1].
e-mail: florian.peters@usz.ch

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 55


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_9
56 F. Peters and C. Grimm

Moreover, cholesterol-rich drusen and accumu- 3 Regulation of ABCA1 by


lated lipoproteins in the sub-­RPE and sub-retinal miR-33
space are hallmarks of AMD [23], which indicates
diminished reverse cholesterol transport (RCT) The most extensively studied ABCA1-regulating
and impaired removal of lipids from the retina for miRNA is miR-33, which is well-known to con-
their degradation in the liver [7]. A key choles- trol the lipid metabolism [8]. The miR-33 family
terol transport protein is ATP-binding cassette was first described in 2010 by several groups and
transporter A1 (ABCA1), which is expressed in in different species as it is highly conserved [24,
the RPE and transfers intracellular cholesterol and 25, 27]. In humans, two isoforms of miR-33 have
other lipids to extracellular lipid-binding proteins been identified, located in different genes on dif-
like ApoA1 or ApoE to generate HDL [26, 36]. ferent chromosomes. While miR-33a localizes to
Polymorphisms in ABCA1 and other genes con- intron 16 of the sterol regulatory element-binding
nected to the lipid metabolism were associated protein (SREBP)-2 gene on chromosome 22,
with an increased risk for AMD development [9], miR-33b is located in intron 17 of SREBP-1.
and loss of Abca1 in mouse RPE led to lipid accu- Only one isoform of miR-33 exists in mouse,
mulation and RPE atrophy with subsequent pho- which corresponds to human miR-33a and is
toreceptor degeneration resembling AMD located in intron 15 of the Srebp-2 gene. SREBPs
pathogenesis [37]. Thus, it is of great interest to are key transcription regulators of genes involved
investigate what factors influence the expression in cholesterol biosynthesis and uptake such as
of ABCA1 and therefore contribute to a dysregu- fatty acid synthase and low-density lipoprotein
lated lipid metabolism. (LDL) receptor [5, 17]. Depletion of cholesterol
results in the upregulation of both SREBPs and
miR-33 and subsequent downregulation of
2 miRNA ABCA1 in a classic feedback loop [25]. To date,
elevated levels of miR-33 have not been described
Expression of genes can be regulated on the RNA in studies of large miRNA screens with AMD
level, e.g., by RNA stability, splicing efficiency, patients. However, there are two publications
and interaction with microRNAs. miRNA are showing elevated levels of miR-33 and reduced
small, ~22 nucleotide-spanning non-coding RNA ABCA1 expression in peritoneal and retinal mac-
molecules that bind to 3′ untranslated regions of rophages [34] and RPE of old mice and nonhu-
target genes, thereby inhibiting translation of man primates [11]. Sene and colleagues described
(large) sets of genes which in further conse- an abnormal polarization of senescent macro-
quence affects entire cellular pathways. On one phages due to high intracellular cholesterol in
hand, modulation of gene expression by miRNAs Abca1-deficient or diet-induced obesity mice
is essential for normal retinal function and tissue [34]. This abnormal polarization resulted in a
integrity as shown for miRNAs 182 and 183 that diminished ability to regulate vascular prolifera-
are necessary for proper cone outer segment tion potentially promoting choroidal neovascu-
maintenance [6]. On the other hand, they are also larization (CNV), a hallmark of wet AMD. In
considered to play a role in the pathology of com- turn, in vivo silencing of miR-33 by anti-miR-33
plex retinal diseases, including AMD [3] and reti- treatment prevented the abnormal macrophage
nitis pigmentosa [21] due to their high gene polarization and inhibited CNV in old mice. A
regulatory potency. Therefore, miRNAs might more recent study investigated the regulation of
serve as potential biomarkers or therapeutic tar- miR-33 and ABCA1 in human RPE cells and RPE
gets [2, 29]. of Western type diet-fed aging mice and nonhu-
Regulation of ABCA1 by miR-33 and miR-34a in the Aging Eye 57

man primates [11]. Subcutaneous delivery of and subsequent cholesterol efflux [20, 32].
anti-miR-33 antisense oligonucleotides (ASOs) Interestingly, elevated levels of miR-34a have
during the high cholesterol diet prevented lipid been found in serum of neovascular AMD
accumulation, immune cell infiltration, and path- patients [30] as well as in aging mouse retina and
ological changes of the RPE. Therefore, the RPE [35]. However, so far, this observation was
authors suggested that clearance of accumulated more connected to DNA damage, cell prolifera-
cholesterol in the RPE by targeting miR-33 might tion, and oxidative stress in RPE cells than to the
attenuate these pathological changes leading to lipid metabolism, as miR-34a is induced by p53
dry AMD. Analogous observations were made in and alters expression of target genes that inhibit
Ldlr–/– mice, a mouse model to study atheroscle- cell proliferation eventually leading to senes-
rosis. Antagonizing miR-33 increased Abca1 cence or apoptosis [13, 15].
expression and circulating HDL, which reduced
atherosclerotic plaque size, lipid content, and
inflammation [28, 31], making it an interesting 5 Concluding Remarks
target for atherosclerosis treatment. However, and Future Therapeutic
long-term deficiency of miR-33 in high-fat diet-­ Perspectives
treated mice causes adverse effects like obesity,
dyslipidemia, and steatosis [12, 14] that might be Age-related diseases often go along with a dys-
prevented by local rather than systemic miR-33 regulated lipid metabolism in cells and accu-
inhibition e.g. by direct delivery of anti-miRs mulated lipid deposits such as drusen in AMD
into the aging eye. or atherosclerotic plaques in atherosclerosis.
The removal of such deposits can be dimin-
ished by a disturbed RCT from peripheral tis-
4 Regulation of ABCA1 by sues. ABCA1, as one of the major lipid export
miR-34a proteins for RCT, and ABCA1-regulatory miR-
NAs gained attention as potential therapeutic
A second miRNA that connects to ABCA1 regu- candidates in atherosclerosis [16, 22]. Since
lation and atherosclerosis is miR-34a, which is expression of ABCA1-targeting miRNAs miR-
encoded by its own transcript and increased dur- 33 and miR-34a increases with age in RPE
ing aging [4, 19] and obesity [10, 18]. A study by cells, they may have a high potency to diminish
Xu et al. identified miR-34a as a regulator of RCT from retinal tissues. This can lead to
ABCA1 and cholesterol efflux in macrophages intracellular lipid accumulation, RPE atrophy,
and achived regression of atherosclerosis by ther- and photoreceptor degeneration [37].
apeutic inhibition of miR-34a in a diet-­induced Therefore, therapeutic targeting those miRNAs
mouse model for atherosclerosis [39]. Of note, that modulate lipid efflux by local application
systemic inhibition or genetic ablation of miR- of ASOs is a promising attempt to attenuate
34a did not result in side effects as dyslipidemia lipid accumulation in the aging eye and AMD
and obesity in Ldlr−/− or Apoe−/− mice as seen pathogenesis [11].
with systemic miR-33 inhibition, making miR-
34a a more promising therapeutic target for ath-
erosclerosis. Besides the direct targeting by References
miR-34a, ABCA1 is additionally regulated by an
upstream pathway including NAD-dependent 1. Anderson DH, Fisher SK, Steinberg RH. Mammalian
cones: disc shedding, phagocytosis, and renewal.
protein deacetylase sirtuin-1 (SIRT1), a key sen-
Invest Ophthalmol Vis Sci. 1978;17:117–33.
sor and regulator of lipid metabolism (exten- 2. Askou AL, Alsing S, Holmgaard A, et al. Dissecting
sively reviewed in [33]). Downregulation of microRNA dysregulation in age-related macular
SIRT1 reduces activation of liver X receptor degeneration: new targets for eye gene therapy. Acta
Ophthalmol. 2018;96:9–23.
(LXR), the major transcription factor for ABCA1,
58 F. Peters and C. Grimm

3. Berber P, Grassmann F, Kiel C, et al. An eye on 19. Li X, Khanna A, Li N, et al. Circulatory miR34a as
age-­related macular degeneration: the role of an RNAbased, noninvasive biomarker for brain aging.
microRNAs in disease pathology. Mol Diagn Ther. Aging (Albany NY). 2011;3:985–1002.
2017;21:31–43. 20. Li X, Zhang S, Blander G, et al. SIRT1 deacety-
4. Boon RA, Iekushi K, Lechner S, et al. MicroRNA-­ lates and positively regulates the nuclear receptor
34a regulates cardiac ageing and function. Nature. LXR. Mol Cell. 2007;28:91–106.
2013;495:107–10. 21. Loscher CJ, Hokamp K, Kenna PF, et al. Altered reti-
5. Brown MS, Goldstein JL. Cholesterol feedback: from nal microRNA expression profile in a mouse model
Schoenheimer’s bottle to Scap’s MELADL. J Lipid of retinitis pigmentosa. Genome Biol. 2007;8:R248.
Res. 2009;50(Suppl):S15–27. 22. Maguire EM, Pearce SWA, Xiao Q. Foam cell for-
6. Busskamp V, Krol J, Nelidova D, et al. miRNAs 182 mation: A new target for fighting atherosclero-
and 183 are necessary to maintain adult cone photo- sis and cardiovascular disease. Vasc Pharmacol.
receptor outer segments and visual function. Neuron. 2019;112:54–71.
2014;83:586–600. 23. Malek G, Li CM, Guidry C, et al. Apolipoprotein B
7. Favari E, Chroni A, Tietge UJ, et al. Cholesterol in cholesterol-containing drusen and basal deposits
efflux and reverse cholesterol transport. Handb Exp of human eyes with age-related maculopathy. Am J
Pharmacol. 2015;224:181–206. Pathol. 2003;162:413–25.
8. Fernandez-Tussy P, Ruz-Maldonado I, Fernandez-­ 24. Marquart TJ, Allen RM, Ory DS, et al. miR-33 links
Hernando C. MicroRNAs and circular RNAs in SREBP-2 induction to repression of sterol transport-
lipoprotein metabolism. Curr Atheroscler Rep. ers. Proc Natl Acad Sci U S A. 2010;107:12228–32.
2021;23:33. 25. Najafi-Shoushtari SH, Kristo F, Li Y, et al.
9. Fritsche LG, Igl W, Bailey JN, et al. A large genome-­ MicroRNA-33 and the SREBP host genes cooper-
wide association study of age-related macular degen- ate to control cholesterol homeostasis. Science.
eration highlights contributions of rare and common 2010;328:1566–9.
variants. Nat Genet. 2016;48:134–43. 26. Quazi F, Molday RS. Lipid transport by mammalian
10. Fu T, Choi SE, Kim DH, et al. Aberrantly ele- ABC proteins. Essays Biochem. 2011;50:265–90.
vated microRNA-34a in obesity attenuates hepatic 27. Rayner KJ, Suarez Y, Davalos A, et al. MiR-33 con-
responses to FGF19 by targeting a membrane core- tributes to the regulation of cholesterol homeostasis.
ceptor beta-Klotho. Proc Natl Acad Sci U S A. Science. 2010;328:1570–3.
2012;109:16137–42. 28. Rayner KJ, Sheedy FJ, Esau CC, et al. Antagonism of
11. Gnanaguru G, Wagschal A, Oh J, et al. Targeting of miR-33 in mice promotes reverse cholesterol trans-
miR-33 ameliorates phenotypes linked to age-related port and regression of atherosclerosis. J Clin Invest.
macular degeneration. Mol Ther. 2021;29:2281. 2011;121:2921–31.
12. Goedeke L, Salerno A, Ramirez CM, et al. Long-term 29. Ren C, Liu Q, Wei Q, et al. Circulating miRNAs as
therapeutic silencing of miR-33 increases circulating potential biomarkers of age-related macular degen-
triglyceride levels and hepatic lipid accumulation in eration. Cell Physiol Biochem. 2017;41:1413–23.
mice. EMBO Mol Med. 2014;6:1133–41. 30. Romano GL, Platania CBM, Drago F, et al. Retinal
13. Hermeking H. The miR-34 family in cancer and apop- and circulating miRNAs in age-related macular
tosis. Cell Death Differ. 2010;17:193–9. degeneration: an in vivo animal and human study.
14. Horie T, Nishino T, Baba O, et al. MicroRNA-33 Front Pharmacol. 2017;8:168.
regulates sterol regulatory element-binding protein 1 31. Rotllan N, Ramirez CM, Aryal B, et al. Therapeutic
expression in mice. Nat Commun. 2013;4:2883. silencing of microRNA-33 inhibits the progres-
15. Hou Q, Tang J, Wang Z, et al. Inhibitory effect of sion of atherosclerosis in Ldlr−/− mice – brief report.
microRNA-34a on retinal pigment epithelial cell pro- Arterioscler Thromb Vasc Biol. 2013;33:1973–7.
liferation and migration. Invest Ophthalmol Vis Sci. 32. Rottiers V, Naar AM. MicroRNAs in metabolism
2013;54:6481–8. and metabolic disorders. Nat Rev Mol Cell Biol.
16. Javadifar A, Rastgoo S, Banach M, et al. Foam cells as 2012;13:239–50.
therapeutic targets in atherosclerosis with a focus on 33. Schug TT, Li X. Sirtuin 1 in lipid metabolism and
the regulatory roles of non-coding RNAs. Int J Mol obesity. Ann Med. 2011;43:198–211.
Sci. 2021;22:2529. 34. Sene A, Khan AA, Cox D, et al. Impaired cholesterol
17. Kawabe Y, Sato R, Matsumoto A, et al. Regulation of efflux in senescent macrophages promotes age-related
fatty acid synthase expression by cholesterol in human macular degeneration. Cell Metab. 2013;17:549–61.
cultured cells. Biochem Biophys Res Commun. 35. Smit-McBride Z, Forward KI, Nguyen AT, et al.
1996;219:515–20. Age-dependent increase in miRNA-34a expression
18. Lee J, Padhye A, Sharma A, et al. A pathway involv- in the posterior pole of the mouse eye. Mol Vis.
ing farnesoid X receptor and small heterodimer 2014;20:1569–78.
partner positively regulates hepatic sirtuin 1 lev- 36. Storti F, Raphael G, Griesser V, et al. Regulated efflux
els via microRNA-34a inhibition. J Biol Chem. of photoreceptor outer segment-derived cholesterol
2010;285:12604–11. by human RPE cells. Exp Eye Res. 2017;165:65–77.
Regulation of ABCA1 by miR-33 and miR-34a in the Aging Eye 59

37. Storti F, Klee K, Todorova V, et al. Impaired ABCA1/ den projection for 2020 and 2040: a systematic
ABCG1-mediated lipid efflux in the mouse retinal review and meta-analysis. Lancet Glob Health.
pigment epithelium (RPE) leads to retinal degenera- 2014;2:e106–16.
tion. elife. 2019;8:e45100. 39. Xu Y, Xu Y, Zhu Y, et al. Macrophage miR-34a is a
38. Wong WL, Su X, Li X, et al. Global prevalence of key regulator of cholesterol efflux and atherosclerosis.
age-related macular degeneration and disease bur- Mol Ther. 2019;28:202.
The Role of Gene Expression
Regulation on Genetic Risk
of Age-­Related Macular
Degeneration

Rinki Ratnapriya

Abstract directions to bridge the gap between genetic


predisposition and biological mechanisms to
Age-related macular degeneration (AMD) is a reap the full benefits of GWAS findings.
major cause of irreversible vision loss in the
elderly. It is a complex multifactorial disease Keywords
that is caused by the cumulative impact of
genetic predisposition, environmental stress, Age-related macular degeneration · GWAS ·
and advanced aging. Knowledge of genetic Transcriptome · Gene expression regulation ·
risk factors underlying AMD susceptibility eQTL · Gene regulatory networks ·
has advanced rapidly in the past decade that Biomarkers
can be largely credited to genome-wide asso-
ciation studies (GWAS) and next-generation
sequencing (NGS) efforts. GWAS have identi-
fied 34 genetic risk loci for AMD; the majority
1 Introduction
of which are in the noncoding genome. Several
Age-related macular degeneration (AMD) is a
lines of evidence suggest that a complex trait-­
common, age-related disease that leads to severe
associated variant is likely to regulate the gene
central visual impairment because of degenera-
expression (acting as expression quantitative
tion of retinal pigment epithelium (RPE) and
trait loci (eQTLs)), and there is a significant
photoreceptors in the central region of the retina
enrichment of GWAS-associated variants
called macula. The burden of AMD on healthcare
within eQTLs. In the last two years, eQTL
is massive, as it afflicts almost 10 million indi-
studies in AMD-relevant tissues have pro-
viduals in the United States and 150 million peo-
vided functional interpretation of several
ple worldwide [17]. It is a chronic, progressive
AMD-GWAS loci. This review highlights the
disease that is caused by dysfunction in photore-
knowledge gained to date and discusses future
ceptors, RPE, Bruch’s membrane (BrM), or cho-
riocapillaris (CC) [11]. Over the last 15 years,
R. Ratnapriya (*) genome-wide association studies (GWAS) have
Department of Ophthalmology, Baylor College of contributed substantially in our understanding of
Medicine, genetic risk factors associated with AMD. The
Houston, TX, USA most recent GWAS analysis of 16,144 AMD
e-mail: rpriya@bcm.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 61


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_10
62 R. Ratnapriya

cases and 17,832 controls identified 52 variants (∼93%) of trait-associated SNPs are located in
across 34 loci that account for over 50% of heri- noncoding regions of the genome [9], and a com-
tability [5]. These findings have implicated com- plex trait-associated variant is more likely to be
plement pathways, lipid transport, and associated with gene expression, i.e., expression
extracellular matrix (ECM) organizations in its quantitative trait loci (eQTL) [2]. These observa-
pathogenesis. Additionally, whole exome and tions suggest a major role of gene expression
genome sequencing have advocated the signifi- regulation in mediating the disease risk. As gene
cant role of rare coding variants in AMD. Rare expression and its regulation often varies across
variants discoveries have helped in identifying cell and tissue types, analysis of disease-relevant
the target genes at several AMD loci and pro- tissues remains central to the functional interpre-
vided insights into the disease mechanism [4, tation of GWAS findings. Genotype-Tissue
13]. Thus, the genetic architecture of AMD Expression (GTEx) project represents the largest
includes common genetic variants with relatively resource that includes 15,201 samples from 838
small effect sizes and rare genetic variants and donors across 49 tissues types [1]. However, the
few common variants with large effect sizes. exclusion of retina, RPE, or BrM from GTEx has
However, for a large number of AMD loci, the been partly responsible for slow progress on
causal variants and genes have remained elusive. post-GWAS functional studies in the field of ocu-
Thus, there is an unmet need to identify genes lar diseases/traits.
and mechanisms underlying AMD as it could A general framework for the functional dis-
lead to possibilities of novel drug targets as well section of a genetic risk locus involves the fol-
as a better prediction for disease onset and lowing key approaches: (1) prioritizing the
progression. variants within the LD through statistical fine-­
mapping, (2) incorporation of functional epig-
enomic data to annotate the variant and genomic
2 Connecting AMD-GWAS region, and (3) testing the function of putative
Discoveries to Biology causal variant and determining the molecular
function of the regulatory variant. Here, we focus
The functional studies of GWAS loci in search of on methods of connecting the noncoding variant
causal variants, genes, and disease mechanisms to gene expression regulation using the eQTL
have not matched the pace with GWAS because approach in the context of AMD.
of several reasons. Firstly, the majority of AMD-­
risk factors reside in nonprotein-coding regions
of the genome whose function is not well studied. 3 Variant to Function by
Secondly, GWAS point to the most significant Studying eQTLs
associated SNP in the region, and the presence of
linkage disequilibrium (LD) between the associ- eQTL analysis is performed to identify the effect
ated variants makes it difficult to point to the of a genetic variant on the expression of one or
causal variant. Within the LD, the associated more genes. It can be divided into those that have
region could harbor multiple genes or no genes at local effects (cis-eQTLs) and those with distant
all. Additionally, a large number of associated effects (trans-eQTLs). A generic pipeline for
variants and their small effect sizes represent sig- eQTL discovery is summarized in Fig. 1. Briefly,
nificant hurdles in translating GWAS discoveries eQTL analysis begins with measuring gene
to disease biology. expression across a cohort using methods such as
Connecting GWAS findings with intermediate RNAseq. RNAseq reads are evaluated for quality
molecular phenotypes such as transcriptome, controls, and normalization and batch corrections
epigenome, and proteome can provide functional are applied. Simultaneously, hundreds of thou-
evidence to map disease associations and trans- sands of SNPs are also genotyped, usually
late them into clinical applications. The majority through genotyping array or genome sequencing.
The Role of Gene Expression Regulation on Genetic Risk of Age-Related Macular Degeneration 63

Fig. 1 A generic pipeline for eQTL discovery

To increase the power of eQTL discovery, geno- get gene. The first reference eQTL map of over
typed SNPs are subjected to haplotype phasing 500 postmortem human donor retinas reported
and genotype imputation. Finally, the association 14,565 genetic variants (eVariants) that regulate
between genotype and gene expression is mea- the expression of 10,474 genes (eGenes) [12].
sured using linear regression. A number of com- Co-localization studies using eCAVIAR identi-
putation tools have been developed to perform fied the target genes at six AMD loci [12]. In
eQTL analysis, such as Matrix eQTL [14] and another study, analysis of the gene expression in
QTLTools [3]. Among these, Matrix eQTL has healthy retinas (n = 311) identified 65 unique
been quite popular because of its fast computing ocular GWAS variants, including AMD, to be
efficiency. regulating gene expression in retinal tissue [15].
eQTL analysis in human RPE/choroid and retina
in bulk tissue samples from the macula and non-
4 eQTL Studies in AMD-­ macular regions in ~120 donors (98 control and
Relevant Human Tissues 23 AMD) identified 15 putative causal genes at
13 known AMD loci [10]. Finally, eQTL analysis
Cis-eQTL analysis is the most commonly used of 23 primary human fetal RPE lines implicated
approach for the identification of candidate sus- four genes [6]. These results are summarized in
ceptibility genes at the risk loci. In the last two Table 1.
years, eQTL studies in AMD-relevant ocular tis- However, interpretations of these results are
sues have been utilized to aid in the interpretation not always straightforward as shown by the asso-
of AMD-GWAS results with moderate success. ciation PILRA/PILRB and RDH5/CD63 loci.
In most analyses, co-localization of eQTL and More than one target gene emerges depending on
GWAS variants was performed to identify the tar- the tissues investigated. For example, the associ-
64 R. Ratnapriya

Table 1 Target genes identified through eQTL studies in human ocular tissues
AMD locus Putative causal gene Target tissues eQTL variant (s) References
B3GALTL B3GLCT Macular and peripheral retina rs9564692, [10, 12]
rs4381465
RDH5/CD63 BLOC1S1 Peripheral retina rs56108400, [10, 12]
rs3138141
RDH5 Peripheral RPE, human fetal RPE rs3138142, [6, 10]
lines rs3138141
AC009779.3 Peripheral retina rs3138141 [15]
ACAD10 SH2B3 Peripheral retina rs61941274 [12]
CFI CFI Macular RPE rs141098594 [10]
PLA2G12A Peripheral retina rs10033900 [12]
PILRB/PILRA PILRA, PILRB Macular and peripheral retina rs6965458, [6, 10, 12]
rs7803454
TMEM97/VTN TMEM199 Macular and peripheral retina rs7212510, [10, 12]
rs11080055
SLC16A8 BAIAP2L2 Nonmacular RPE rs760975 [10]
NPLOC4-­ TSPAN10 Nonmacular RPE rs7405790 [10]
TSPAN10
TRPM1 TRPM1 Nonmacular RPE rs12440880 [10]
MMP9 SLC12A5-AS1 Nonmacular retina rs3761159 [10]
CTRB2-CTRB1 BCAR1 macular retina rs9928736 [10]
COL4A3 COL4A3 Nonmacular retina rs7559693 [10]
TNFRSF10A TNFRSF10A Nonmacular RPE rs11777697 [10]
ARMS2-HTRA1 HTRA1 Macular retina rs11528744 [10]
C2/CFB/SKIV2L HLA-DQB1, Peripheral retina rs204993 [15]
TSBP1-AS1

ated variant, rs3138141, regulates BLOC1S1 in used human donor ocular tissues to estimate
the retina and RDH5 in the RPE. In a different mRNA expression of the ARMS2 and HTRA1
study, low-frequency variants in CFHR2 and genes in human ocular tissues. By comparing
CFHR5 at the CFH locus lead to reduced or age-matched donors with risk and no-risk geno-
absent FHR-2 and FHR-5 concentrations, indi- types, they observed reduced HTRA1 expression
cating the role of these genes at CFH locus [7]. in RPE of donors with risk variants, whereas the
These findings suggest that multiple genes within HTRA1 expression did not change in the neural
a GWAS locus can contribute to the overall dis- retina or choroid [16]. They further show that risk
ease variance. Thus, the “best-fit” candidate gene variant disrupts a cis-regulatory element within
interpretations of GWAS loci might be a simpli- the locus that overlaps with the ARMS2 intron,
fied view of the complex genetic architecture which contains a potential Lhx2 transcription
underlying individual associated loci, and fol- factor binding site.
low-­up functional studies need to investigate all
candidates identified through eQTL studies for
their role in AMD pathology. 5 Future Experimental
Approaches
The HTRA1/ARMS2 Locus 10q26 region, con-
taining two tightly linked genes, age-related mac- AMD has been at the forefront of GWAS studies
ulopathy susceptibility 2 (ARMS2), and and has achieved substantial success. While
high-temperature requirement A serine peptidase genome and exome sequencing-based approaches
1 (HTRA1), has been subjected to intense investi- can continue to identify new variants ge and loci
gation with conflicting results. A recent study associated with AMD, it is also time to focus on
The Role of Gene Expression Regulation on Genetic Risk of Age-Related Macular Degeneration 65

downstream functional dissection of already-­ ing the molecular function of the regulatory vari-
identified AMD-GWAS loci as these efforts can ants and disease mechanisms.
help in providing key insights into the disease
pathophysiology. eQTL studies in the context of Acknowledgment RR is supported by Career
AMD have demonstrated the key role of gene Development Award from Research to Prevent
expression regulation in AMD pathology. Blindness (RPB) and New Investigator Grant from
BrightFocus Foundation. This study was also sup-
However, this area of research is still in its ported by an unrestricted grant from RPB to Baylor
infancy, and the full potential of eQTL studies to College of Medicine.
access and interpret the molecular link between
genetic variant and phenotype has not been real-
ized. Future studies of building bigger reference
data sets for retina, RPE, and choroid will pro- References
vide a valuable framework toward a broad, long-­
1. Consortium GT. The GTEx Consortium atlas of
term goal to bridge the gap between genetic genetic regulatory effects across human tissues.
predispositions and biological mechanisms in Science. 2020;369:1318–30.
AMD. Additionally, most studies have been per- 2. Cookson W, Liang L, Abecasis G, et al. Mapping
formed at the tissues level that cannot define the complex disease traits with global gene expression.
Nat Rev Genet. 2009;10:184–94.
cellular contexts in which disease risk-SNPs 3. Delaneau O, Ongen H, Brown AA, et al. A complete
affect gene expression levels. GTEx consortium tool set for molecular QTL discovery and analysis.
studies have revealed that many eQTLs effects Nat Commun. 2017;8:15452.
are cell-type specific (Consortium, 2020). Thus, 4. den Hollander AI, de Jong EK. Highly penetrant
alleles in age-related macular degeneration. Cold
future studies also necessitate the generation of Spring Harb Perspect Med. 2014;5:a017202.
single-cell eQTL resources to identify the cell 5. Fritsche LG, Igl W, Bailey JN, et al. A large genome-­
type-specific consequences of AMD variants. wide association study of age-related macular degen-
Regulatory variants (eQTLs) are ubiquitous eration highlights contributions of rare and common
variants. Nat Genet. 2016;48:134–43.
across the genome. Thus, relying on co-­ 6. Liu B, Calton MA, Abell NS, et al. Genetic analyses of
localization of eQTL with GWAS alone is not human fetal retinal pigment epithelium gene expres-
sufficient as they can occur as a chance event. sion suggest ocular disease mechanisms. Commun
These signals need to be followed up with inde- Biol. 2019;2:186.
7. Lores-Motta L, van Beek AE, Willems E, et al.
pendent experiments. The establishment and Common haplotypes at the CFH locus and low-­
maintenance of a cell’s transcriptional identity frequency variants in CFHR2 and CFHR5 asso-
are largely driven by the specific activity of cis-­ ciate with systemic FHR concentrations and
regulatory elements (CREs) such as promoter, age-related macular degeneration. Am J Hum Genet.
2021;108:1367–84.
enhancer, and silencer elements [8, 9]. 8. Manolio TA, Collins FS, Cox NJ, et al. Finding the
Additionally, trait and disease-associated vari- missing heritability of complex diseases. Nature.
ants are enriched in the CREs and possibly can 2009;461:747–53.
influence gene transcription via a co-localized 9. Maurano MT, Humbert R, Rynes E, et al. Systematic
localization of common disease-associated variation
genomic regulatory element, such as a transcrip- in regulatory DNA. Science. 2012;337:1190–5.
tion factor binding site within an open chromatin 10. Orozco LD, Chen HH, Cox C, et al. Integration of
region. Thus, characterization and integration of eQTL and a single-cell atlas in the human eye iden-
CREs in ocular tissues is the next step in the pri- tifies causal genes for age-related macular degenera-
tion. Cell Rep. 2020;30:1246–1259 e1246.
oritization of eQTL variants. These findings can 11. Ratnapriya R, Chew EY. Age-related macular
be then validated in cell culture using reporter degeneration-­ clinical review and genetics update.
assay such as massively parallel reporter assays Clin Genet. 2013;84:160–6.
(MPRA) to test the transcriptional activities of 12. Ratnapriya R, Sosina OA, Starostik MR, et al. Retinal
transcriptome and eQTL analyses identify genes asso-
DNA elements. Finally, induced pluripotent stem ciated with age-related macular degeneration. Nat
cell lines or animal models will help in elucidat- Genet. 2019;51:606–10.
66 R. Ratnapriya

13. Ratnapriya R, Acar IE, Geerlings MJ, et al. Family-­ 16. Williams BL, Seager NA, Gardiner JD, et al.
based exome sequencing identifies rare coding vari- Chromosome 10q26-driven age-related macular
ants in age-related macular degeneration. Hum Mol degeneration is associated with reduced levels of
Genet. 2020;29:2022–34. HTRA1 in human retinal pigment epithelium. Proc
14. Shabalin AA. Matrix eQTL: ultra fast eQTL analy- Natl Acad Sci U S A. 2021;118:20210727.
sis via large matrix operations. Bioinformatics. 17. Wong WL, Su X, Li X, et al. Global prevalence of
2012;28:1353–8. age-related macular degeneration and disease bur-
15. Strunz T, Kiel C, Grassmann F, et al. A mega-analysis den projection for 2020 and 2040: a systematic
of expression quantitative trait loci in retinal tissue. review and meta-analysis. Lancet Glob Health.
PLoS Genet. 2020;16:e1008934. 2014;2:e106–16.
Elastin Layer in Bruch’s Membrane
as a Target for Immunization
or Tolerization to Modulate
Pathology in the Mouse Model
of Smoke-Induced Ocular Injury

Bärbel Rohrer, Nathaniel Parsons,


Balasubramaniam Annamalai, Crystal Nicholson,
Elisabeth Obert, Bryan Jones, and Andrew D. Dick

Abstract ates immunogenic elastin neoepitopes that


trigger an increase in α-elastin IgG and IgM
Age-related macular degeneration (AMD) is antibodies, which can then bind to the neoepi-
associated with an overactive complement topes in the target cells or membranes, trigger-
system and an increase in circulating antibod- ing complement activation. Specifically, we
ies. Our search for potential neoantigens that showed that immunization with elastin pep-
can trigger complement activation in disease tide oxidatively modified by cigarette smoke
has led us to investigate elastin. A loss of the (ox-elastin) exacerbated ocular pathology and
elastin layer (EL) of Bruch’s membrane (BrM) vision loss in CSE mice. In contrast, mice
has been reported in aging and AMD together receiving peptide immunotherapy (PIT) with
with an increase of serum elastin-derived pep- ox-elastin did not lose vision over the smok-
tides and α-elastin antibodies. In the mouse ing period and exhibited a more preserved
model of cigarette smoke exposure (CSE), BrM. Immunization and PIT correlated with
damage in BrM, loss of the EL, and vision loss humoral immunity and complement activation
are dependent on complement activation. We and IgG/IgM deposition in the RPE/BrM/cho-
have examined the hypothesis that CSE gener- roid. Finally, PIT modulated immune markers
IFNγ and IL-4. The data further support the
B. Rohrer (*) hypothesis that complement activation, trig-
Department of Ophthalmology, Medical University of gered by immune complex formation in target
South Carolina, Charleston, SC, USA
tissues, plays a role in ocular damage in the
Ralph H. Johnson VA Medical Center, Charleston, CSE model. As PIT with ox-elastin peptides
SC, USA
e-mail: rohrer@musc.edu
reduces damage, we discuss the possibility
that AMD progression might be preventable.
N. Parsons · B. Annamalai · C. Nicholson · E. Obert
Department of Ophthalmology, Medical University of
South Carolina, Charleston, SC, USA Keywords
B. Jones
Department of Ophthalmology, University of Utah, Age-related macular degeneration · Elastin ·
Salt Lake City, UT, USA Immunization · Tolerance · Complement ·
A. D. Dick Smoking
University of Bristol, Bristol, UK

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 67


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_11
68 B. Rohrer et al.

1 Introduction fication status is unknown. Finally, elevating


serum elastin fragments in mouse increased
AMD is a multifactorial, complex disease, which expression and deposition of collagen IV in the
occurs in two forms, wet and dry. Patients experi- RPE/choroid complex [12]. Based on these
ence a loss of central vision, and clinical under- observations, we have previously postulated that
pinnings include drusen and retinal pigment abnormalities in elastin homeostasis together
epithelium (RPE) disturbances, damage to macu- with antibody production may contribute to an
lar photoreceptors, and pathology at the inflammatory feedback loop, ultimately leading
RPE/choroid interface. Among the risk factors to AMD pathology [13].
associated with disease, we wish to highlight fac-
tors relevant for the hypothesis presented here,
which include aging [1], cigarette smoking [2], 2 Cigarette Smoke Exposure
increased susceptibility of elastic fibers to photic (CSE) as a Model for Dry
degenerative stimuli [3], and genetic variants in AMD
genes regulating the complement cascade [4] and
extracellular matrix turnover [5]. We consider CSE, which is a passive inhalation
The complement system is the humoral back- model lasting for 6 months, a model for dry
bone of the innate immune system. Its main role AMD, as it shows structural alteration and com-
is to participate in antimicrobial defense, clear- plement deposition in the RPE/BrM/choriocapil-
ance of immune complexes, and tissue regenera- laris (CC) complex similar to that seen in patients
tion, but excessive complement activation can be [14]. Smoke exposure leads to the formation of
involved in the pathogenesis of disease states, deposits in the RPE/BrM that contain proteins
including AMD [6]. The complement system can also found in human drusen and basal laminar
be activated by three distinct pathways: the clas- deposits such as C3a, C5, MAC, and CFH [15].
sical (CP), lectin (LP), and alternative pathway Also, we have shown that long-term smoke in
(AP), with IgG and natural IgM antibodies bind- C57BL/6J mice reduces scotopic and photopic
ing to ligands generated by injury, immunity, and ERG amplitudes, in addition to contrast sensitiv-
infection participating in CP and LP activation. ity. Structural changes include mitochondrial
This begs the question as to what might be their swelling in the RPE, a loss of CC fenestrations,
ligands. and thickening of BrM due to the deposition of
The potential role of BrM in initiation and material in the outer collagenous layer. These
progression of disease has been investigated structural alterations were not observed in CFB−/−
based on its changes with aging and disease [7], mice, in which the AP is eliminated [14] or can
including lipid buildup and calcifications [8], be repaired by treatment with an AP inhibitor
changes in diffusion characteristics [9], and a [16].
reduction of the cross-linked linear elastin fibers
of the EL [7]. Additional alterations in elastin
metabolism in aging and AMD have been 3 Neoepitopes Trigger Chronic
reported. Blumenkranz et al. published on a gen- Inflammation in Smoke-­
eralized increased susceptibility of elastic fibers Induced Pathology
to photic or other degenerative stimuli and sug-
gested that this might be a new and important risk Based on our premise that breakdown of the EL
factor for choroidal neovascularization [3]. in the presence of aging or oxidative stress gener-
Serum-elastin-derived peptides (s-EDPs) were ates elastin neoepitopes, triggering an immune
found to be significantly higher in AMD patients response that leads to complement activation and
than in controls [10], and serum IgG and IgM pathology, we tested the hypothesis that immuni-
antibodies against elastin are elevated in AMD zation with smoke elastin neoepitopes augments
[11]. However, for both the s-EDPs and the elas- pathology. Mice were immunized with 10 μg of
tin epitopes the antibodies recognize, their modi- mouse lung elastin peptides or 10 μg of cigarette
Elastin Layer in Bruch’s Membrane as a Target for Immunization or Tolerization to Modulate Pathology… 69

smoke-modified elastin peptides mixed with should be decreased by immunotherapy. PIT has
adjuvant followed by a booster shot and exposed been studied for the treatment of various autoim-
to 6 months of smoke. After 6 months, mice were mune diseases, allergy, and cancer (e.g., [17]).
examined for anti-elastin antibody production, Overall, in laboratory animals, peripheral toler-
contrast sensitivity, structural changes in BrM, as ance toward a particular antigen is generated by
well as IgG and IgM deposition and the presence repeated exposure, and while the exact mecha-
of complement activation fragments in the nism of action has not yet been fully defined [18],
RPE/choroid [13]. The amount of antibody gen- it is thought to involve deletion of reactive T
erated was analyzed in ELISA assays, using the cells, the generation of a regulatory T (Treg)-cell
corresponding mouse serum as the antibody response, or an altered macrophage response
source, and demonstrated that ox-elastin (neo-­ [19]. Hence, if the generation of modified s-EDPs
epitope) immunization generated more IgM and and their corresponding antibodies were to trig-
IgG antibodies when compared to control elastin ger pathology in eyes of CSE mice, this vicious
(self-epitope). While room air-raised mice exhib- cycle of inflammation should be preventable by
ited stable contrast sensitivity over the 6-month PIT or tolerization. This hypothesis was tested by
analysis period, smoke-exposed mice immunized providing PIT consisting of 1 or 10 μg of ciga-
with ox-elastin exhibited a greater loss in contrast rette smoke-modified mouse lung elastin pep-
sensitivity than the elastin-immunized mice. This tides weekly during the smoking period. After
loss in vision was correlated with a thicker BrM 6 months, the mice were examined for anti-­elastin
and more damaged RPE mitochondria when antibody production, contrast sensitivity, struc-
compared to nonimmunized mice or those immu- tural changes in BrM, as well as IgG/IgM and
nized with a control elastin peptide. Finally, complement activation in the RPE/choroid. PIT
structural and functional alterations were corre- at the 1 μg dose was efficacious in reducing the
lated with increased levels of IgM, IgG3, and humoral immune response, resulting in the sup-
IgG2b in the RPE/choroid fraction, and the pression of ox-elastin-specific IgG and IgM anti-
increase in antibody binding was correlated with bodies when compared to control smoke-exposed
an increase in complement activation as assayed mice, almost reducing levels to those seen in ani-
by Western blotting, probing for C3 breakdown mals raised in room air. Low dose PIT reduced
products. Based on these results, we would like the amount of IgG and IgM deposited in the
to propose that, in CSE, antibodies, generated RPE/choroid, and this decrease in IgG/IgM depo-
either de novo against ox-elastin (IgG) or natural sition was correlated with reduced complement
antibodies amplified and selected (IgM), bind to activation. Finally, structurally, low dose PIT
ox-elastin generated by smoke in BrM or other resulted in preservation of BrM, and, function-
extracellular matrices containing elastin, to trig- ally, improved contrast could be observed.
ger complement activation. Antibodies might be Finally, we asked whether low dose PIT induces
activating complement via either the CP or LP, tolerance. Tolerance to antigen (neo-antigen or
which is then amplified by the AP [14] leading to against self-antigen) engages both central and
the observed pathology. peripheral mechanisms. Central clonal deletion
providing tolerance to self-antigen occurs fol-
lowing negative selection and deletion during
4 Is Pathology in the CSE thymic development, and peripheral tolerance is
Model Preventable a constant surveillance to prevent immune activa-
with Peptide tion of T cells escaping clonal deletion and to
Immunotherapy? potentially neoantigens. Peripheral tolerance is
mediated largely via natural and inducible Trgs,
Given this data on immunization, albeit with only T helper 3 cells, and T regulatory type 1 cells that
indirect evidence of ox-elastin-induced pathol- orchestrate regulation via immunoregulatory
ogy, we argued that if pathology can be increased cytokines such as TGFβ, IL-10, and IL-4 [20]. As
by increasing antibody production, pathology an induction of tolerance requires, in part,
70 B. Rohrer et al.

decreasing Th1/Th17 activation and increasing mechanisms. Nevertheless, as AMD pathogenesis


Th2 activation or enhancing Treg activity (either has been linked to smoking, complement activa-
antigen or non-antigen specific), we examined tion, and pathogenic T- and B-cell immunity, our
Th1 versus Th2 activation by performing a partial hypothesis that AMD pathology is increased in
representative cytokine analysis. Cytokine analy- the presence of neoantigens and decreased by
sis in the RPE/choroid fraction in response to peptide or antigen immunotherapy to suppress
smoke suggests an induction of tolerance in PIT immunity appears justified. Our results may open
animals, as a decrease IFN-y expression (surro- a novel avenue for immunotherapies in dry AMD.
gate of reduction of non-self-activation) and an
increase in IL-4 expression (promotion of self-­ Acknowledgments Funding was provided in part by the
tolerance) were observed. Overall, these data National Institutes of Health R01EY019320 (BR),
R01EY015128, R01EY028927 and P30EY014800 (BJ),
support the hypothesis by Nussenblatt et al. that the Department of Veterans Affairs RX000444 and
AMD might be suitable for tolerance therapy BX003050 (BR), the South Carolina SmartState
[21]. It would be of great interest to determine the Endowment (BR), and Research to Prevent Blindness
presence of ox-elastin versus control elastin anti- (Department of Ophthalmology, University of Utah).
ADD is supported in part through the National
bodies in AMD, as the modification status of the Institute for Health Research Biomedical Research
s-EDPs and the epitope recognition sites of the Centre at Moorfields Eye Hospital and University
anti-elastin antibodies are unknown. Other neo- College London Institute of Ophthalmology. We would
epitopes that might also be involved in the like to thank Carl Atkinson (MUSC) for the room air
and smoke-exposed FcγRΙΙΙ−/− mice.
immune response identified here might include
malondialdehyde (MDA) or carboxyethylpyrrole
(CEP) adducts. Antibodies against MDA have
been shown to be involved in complement-­ References
dependent mouse choroidal neovascularization 1. Al-Zamil WM, Yassin SA. Recent developments
and to recognize neoepitopes on RPE cells in age-related macular degeneration: a review. Clin
exposed to smoke extract [22], and immunization Interv Aging. 2017;12:1313–30.
of mice with CEP-modified mouse serum albu- 2. Seddon JM, et al. A prospective study of cigarette
smoking and age-related macular degeneration in
min results in complement deposition in BrM, women. JAMA. 1996;276(14):1141–6.
the accumulation of drusen-like deposits with 3. Blumenkranz MS, et al. Risk factors in age-related
aging, and decreased retinal function [23]. maculopathy complicated by choroidal neovascular-
We acknowledge that our studies have a num- ization. Ophthalmology. 1986;93(5):552–8.
4. Degn SE, Jensenius JC, Thiel S. Disease-causing
ber of limitations, which have been addressed in mutations in genes of the complement system. Am J
primary publications [13, 24], and include, among Hum Genet. 2011;88(6):689–705.
others, that the effects of smoke exposure and 5. Nita M, et al. Age-related macular degeneration and
treatment on the eye cannot be distinguished from changes in the extracellular matrix. Med Sci Monit.
2014;20:1003–16.
those of the effects of the two on other organs, as 6. Holers VM. The complement system as a thera-
long-term smoke exposure leads to emphysema peutic target in autoimmunity. Clin Immunol.
and other organ damage in mice [25]. In addition, 2003;107(3):140–51.
we recognize that the gold standard to test the 7. Curcio CA, Johnson M. Structure, function, and
pathology of Bruch’s membrane. London: Elsevier;
hypothesis of the role of T cells, their role in tol- 2013. p. 465–81.
erance, and thereby elucidate pathogenic path- 8. Thompson RB, et al. Identification of hydroxyapatite
ways is the use of adoptive and passive transfer of spherules provides new insight into subretinal pig-
T cells as well as specific ox-elastin antibodies. ment epithelial deposit formation in the aging eye.
Proc Natl Acad Sci U S A. 2015;112(5):1565–70.
However, as our model requires 4–6 months to 9. Hussain AA, et al. Macromolecular diffusion char-
propagate pathology, systemic T-cell responses acteristics of ageing human Bruch’s membrane:
were not established. Also, the sources of the implications for age-related macular degeneration
­
cytokines identified would further illuminate (AMD). Exp Eye Res. 2010;90(6):703–10.
Elastin Layer in Bruch’s Membrane as a Target for Immunization or Tolerization to Modulate Pathology… 71

10. Sivaprasad S, Chong NV, Bailey TA. Serum 18. Sabatos-Peyton CA, Verhagen J, Wraith DC. Antigen-­
elastin-­derived peptides in age-related macu- specific immunotherapy of autoimmune and allergic
lar degeneration. Invest Ophthalmol Vis Sci. diseases. Curr Opin Immunol. 2010;22(5):609–15.
2005;46(9):3046–51. 19. Butcher SK, et al. Toll-like receptors drive specific
11. Morohoshi K, et al. Identification of anti-retinal anti- patterns of tolerance and training on restimulation of
bodies in patients with age-related macular degenera- macrophages. Front Immunol. 2018;9:933.
tion. Exp Mol Pathol. 2012;93:193–9. 20. Li L, Boussiotis V. Control and regulation of periph-
12. Skeie JM, et al. Molecular responses of choroidal eral tolerance in allergic inflammatory disease:
endothelial cells to elastin derived peptides through therapeutic consequences. Chem Immunol Allergy.
the elastin-binding protein (GLB1). Matrix Biol. 2008;94:178–88.
2012;31(2):113–9. 21. Nussenblatt RB, et al. Immune responses in age-­
13. Annamalai B, et al. Immunization against oxidized related macular degeneration and a possible long-term
elastin exacerbates structural and functional dam- therapeutic strategy for prevention. Am J Ophthalmol.
age in mouse model of smoke-induced ocular injury. 2014;158(1):5–11 e2.
Invest Ophthalmol Vis Sci. 2020;61(3):45. 22. Joseph K, et al. Oxidative stress sensitizes RPE cells
14. Woodell A, et al. Alternative complement pathway to complement-mediated injury in a natural antibody-,
deficiency ameliorates chronic smoke-induced func- lectin pathway- and phospholipid epitope-dependent
tional and morphological ocular injury. PLoS ONE. manner. J Biol Chem. 2013;288:12753.
2013;8(6):e67894. 23. Hollyfield JG, Perez VL, Salomon RG. A hapten gen-
15. Wang AL, et al. Autophagy and exosomes in the aged erated from an oxidation fragment of docosahexae-
retinal pigment epithelium: possible relevance to dru- noic acid is sufficient to initiate age-related macular
sen formation and age-related macular degeneration. degeneration. Mol Neurobiol. 2010;41(2–3):290–8.
PLoS ONE. 2009;4(1):e4160. 24. Rohrer B, et al. Peptide-based immunotherapy against
16. Woodell A, et al. A targeted inhibitor of the alterna- oxidized elastin ameliorates pathology in mouse
tive complement pathway accelerates recovery from model of smoke-induced ocular injury. Exp Eye Res.
smoke-induced ocular injury. Invest Ophthalmol Vis 2021;212:108755.
Sci. 2016;57(4):1728–37. 25. Vandivier RW, Ghosh M. Understanding the relevance
17. Larche M, Wraith DC. Peptide-based therapeutic vac- of the mouse cigarette smoke model of COPD: peer-
cines for allergic and autoimmune diseases. Nat Med. ing through the smoke. Am J Respir Cell Mol Biol.
2005;11(4 Suppl):S69–76. 2017;57(1):3–4.
Repurposing Drugs for Treatment
of Age-Related Macular
Degeneration

Sarah G. Francisco and Sheldon Rowan

Abstract Keywords

Age-related macular degeneration · Drug


The need for new drugs to treat dry forms of
repurposing · Medical records database ·
age-related macular degeneration remains
Propensity matching · L-DOPA · Metformin ·
high. A promising approach is repurposing of
Mitochondria · Fluoxetine · Serotonin
FDA-approved medications to treat
receptor agonist · Anti-diabetic drugs
AMD. Databases containing medical and drug
records allow for retroactive identification of
drugs whose use correlates with reduced AMD
diagnosis. This short review summarizes 1 Introduction
progress in several classes of drugs considered
for repurposing: GPR-143 agonists (L-DOPA), Age-related macular degeneration affects
anti-diabetic drugs (metformin, acarbose, approximately 196 million individuals worldwide
empagliflozin, fenofibrate), mitochondrial and is a leading cause of blindness in older adults.
activators (PU-91), and serotonin pathway Advanced forms of the disease come in a dry,
drugs (fluoxetine, flibanserin, xaliproden, bus- atrophic form or wet, neovascular form. Wet
pirone). The promises and caveats of repur- AMD can be treated, in part, through inhibitors
posing are discussed herein. of the VEGF pathway. Dry AMD, which accounts
for 70–90% of reported AMD cases, remains
mostly without effective pharmaceutical
intervention.
Most new drugs for dry AMD target the
S. G. Francisco complement pathway, but, to date, phase 3
JM-USDA Human Nutrition Research Center on
clinical trials for dry AMD treatments are not
Aging, Tufts University, Boston, MA, USA
encouraging [1]. An alternative approach to drug
S. Rowan (*)
discovery is to repurpose FDA-approved drugs
JM-USDA Human Nutrition Research Center on
Aging, Tufts University, Boston, MA, USA with established safety that may have efficacy in
treating AMD due to shared pathoetiological
Department of Ophthalmology, Tufts University
School of Medicine, Boston, MA, USA mechanisms or beneficial off-target effects. It is
now possible to mine medical records for
Friedman School of Nutrition Science and Policy,
Tufts University, Boston, MA, USA hundreds of thousands of individuals and
e-mail: sheldon.rowan@tufts.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 73


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_12
74 S. G. Francisco and S. Rowan

determine whether medication history affects (4.7–4.8 letters compared to sham injection).
AMD incidence. Patients taking L-DOPA were also 50% less
Here, we summarize studies that have likely to initiate anti-VEGF treatments than those
evaluated different classes of medications and with sham injection [4]. These findings suggest
suggest that many FDA-approved, widely used, that L-DOPA may be an attractive option for indi-
and safe drugs potentially reduce AMD diagnosis. viduals who wish to avoid intravitreal injection.

2 L-DOPA 3 Metformin

One of the first documented examples of Metformin is one of the most effective antidiabetic
electronic medical record mining to identify medications. Metformin has several
widely used medications for AMD treatments antihyperglycemic properties, such as decreasing
was an evaluation of L-DOPA. In RPE cells, hepatic glucose production, decreasing intestinal
L-DOPA binds to and activates the GPR143 glucose absorption, and improving insulin sensi-
G-protein-­ coupled receptor, which upregulates tivity and is now being tested as a candidate anti-
production of PEDF and downregulates aging treatment [5]. One function of metformin
production of VEGF-A, thus potentially suggests it may be an effective anti-AMD medi-
mediating homeostatic RPE health [2]. cation: the activation of AMPK signaling and the
These relationships prompted an evaluation of stimulation of glucose metabolism in the retina
whether individuals that took L-DOPA for treat- [6].
ment of movement disorders such as Parkinson’s Five studies have analyzed medical records to
disease had reduced incidence of AMD. McKay determine whether metformin use was associated
and colleagues queried Marshfield Clinic Cohorts with protection against AMD. Brown et al. used
(n = 17,500), the Truven MarketScan database records from the University of Florida in a retro-
(n = 15,215,458), and the Personalized Medicine spective case-control study where controls were
Research Product (n = 20,000) to assess AMD compared to AMD cases using propensity score
incidence in individuals prescribed L-DOPA [3]. matching [7]. From a multivariable logistic
AMD diagnosis was delayed in individuals tak- regression analysis comparing the relative ratios
ing L-DOPA relative to those not taking L-DOPA. of those prescribed with metformin with those
In the massive Truven database, they reported an not (n = 7788), they determined metformin use
odds ratio (OR) for developing AMD when pre- was associated with decreased likelihood of
scribed L-DOPA to be 0.78 (confidence interval developing AMD (OR = 0.58, 95% CI: 0.43–
(CI) of 0.76–0.80) in a logistic regression model 0.79) [7].
controlled for age and sex [3]. One study using a smaller medical record
GPR143 functions as a potential anti-­ database (University of San Francisco, n = 3120)
angiogenesis factor, and L-DOPA prescription indicated a significant protective relationship
was similarly associated with reduced neovascu- (OR = 0.70, 95% CI: 0.55–0.88) [8]. A second
lar AMD (OR = 0.65, 95% CI:0.65–0.69) [3]. In study of the much larger IBM MarketScan
order to prospectively evaluate L-DOPA as treat- Commercial and Medicare Supplemental
ment for neovascular AMD, a small cohort of Databases (n = 624,780; OR = 0.94, 95% CI:
patients newly diagnosed with neovascular AMD 0.92–0.96) revealed a dose relationship where
were randomized to receive a sham intravitreal low-to-moderate doses of metformin had greatest
injection, anti-VEGF treatment (ranibizumab), or potency, but metformin use was a risk factor for
L-DOPA in the form of oral carbidopa-levodopa AMD in patients with coexisting diabetic reti-
combined therapy [4]. The L-DOPA group nopathy [9]. A third study of over a million dia-
showed improvements in best-corrected visual betic enrollees (n = 1,007,213) found decreased
acuity comparable to those taking Ranibizumab risk of AMD among prior users of metformin
Repurposing Drugs for Treatment of Age-Related Macular Degeneration 75

(OR = 0.95, 95% CI:0.92–0.98), but increased notypes. Nevertheless, for drugs that do not have
hazard in those with active metformin use, and a a long history in accessible medical databases,
positive correlation between risk and cumulative preclinical models are necessary for identifying
metformin dose [10]. potential medications to repurpose.
Two Asian cohorts also evaluated AMD
relationships with metformin use. A Taiwanese
cohort (n = 68,205) reproduced findings from 5.1 Mitochondrial Activation
American cohorts (OR = 0.54, 95% CI: 0.50– Drugs
0.58) [11], whereas a smaller South Korean
cohort (n = showed no association between AMD Activation of mitochondrial function is a target
and metformin use (OR = 1.15, 95% CI: 0.91– for drugs to treat AMD since RPE cells from
1.45) [12]. Metformin is now being studied as an AMD patients show clear mitochondrial defects
anti-AMD treatment in nondiabetics in the [14–16]. Furthermore, compounds that improve
Metformin for the Minimization of Geographic mitochondrial function improve the health of
Atrophy Progression in Patients with AMD AMD-derived RPE cells or cells containing
(METforMIN) phase 2 clinical trial AMD-derived mitochondrial DNA [14, 16].
(ClinicalTrials.gov Identifier: NCT02684578). Kenney and colleagues reported a drug-­
repurposing approach using PU-91 to enhance
mitochondrial function in RPE cells containing
4 Fluoxetine mitochondria from AMD patients [17]. PU-91 is
an FDA-approved drug that upregulates PGC-1α,
Fluoxetine, a specific serotonin reuptake a transcriptional activator of mitochondrial bio-
inhibitor, is one of the most prescribed genesis and a key cellular energy integrator. In
medications to treat depression and anxiety. cell-based assays, PU-91 enhanced mitochon-
Gelfand and colleagues determined through drial function, limited apoptotic cell death, and
structural analyses that fluoxetine resembles reduced inflammation and complement activa-
CY-09, a small molecule that binds to NLRP3 tion [17, 18].
and inhibits inflammasome activation, an
important pathway in AMD progression [13]. In
vitro and in vivo experiments demonstrated that 5.2 Serotonin Receptor Agonists
fluoxetine inhibited NLRP3 assembly and
activation in cells in response to Alu RNA A significant body of preclinical research
transfection and inhibited RPE degeneration indicated that serotonin receptor agonists,
in vivo in a dry AMD model. The authors utilized originally used to treat psychiatric conditions,
the Truven MarketScan Commercial Claims were capable of rescuing retinal degeneration
database and PearlDiver Mariner database, which in several rodent disease models [19–21]. At
contains data on over 100 million Americans. least three orally available drugs are currently
The combined multivariable-­ corrected model in preclinical testing. Flibanserin has shown
(n = 196,010) indicated significant protection by neuroprotection against light-induced retinal
fluoxetine use (OR = 0.85, 95% CI: 0.73–0.99). degeneration when delivered intraperitoneally
[22]. Oral administration of xaliproden
improved retinal degeneration and visual func-
5 Drugs Being Evaluated tion decline in Sod2-deficient mice [23]. In a
in Preclinical Models sodium iodate mouse model of RPE oxidative
damage, intraperitoneal delivery of buspirone
Drug testing for AMD is an ongoing challenge activated key antioxidant enzyme pathways
since there are few validated preclinical models and helped preserve retinal tissues from dam-
of AMD, and many require years to assess phe- age [24]. Future studies should assess whether
76 S. G. Francisco and S. Rowan

AMD incidence was altered in patients pre- vary across populations, countries, and health-
scribed these drugs. care systems making it difficult to compare stud-
ies in diverse populations. Statistical
methodologies used in propensity matching
5.3 Antidiabetic Drugs require caution as well. Medical databases are
not randomized, contain residual confounders,
Our work and others have implicated glycemia-­ and may have selection biases. Finally, even if
related stress as a pathoetiologic factor in AMD excellent candidate drugs are identified, they may
in human populations and mouse studies [25]. need to be optimized for safe ocular delivery.
We have developed a preclinical AMD model Drug repurposing is not a replacement for new
where wild-type mice fed high glycemic diets drug discovery but can be added to the physi-
develop AMD-like disease, while those fed low cian’s toolkit to customize medical treatment to a
glycemic diets, or are switched from a high-to-­ patient’s existing conditions, genetics, and
low glycemic diet late in life, maintain retinal physiology.
health [26]. The protective effect of the low
glycemic diet in other AMD-prone mice like Acknowledgments Funding was provided by NIH
­
Nrf2-­deficient mice [27] suggested that pharma- RO1EY028559, NIH RO1EY026979, USDA NIFA 2016-
ceutical agents that mimic properties of the low 08885, USDA 8050-51000-089-01S, Thome Memorial
Foundation, and BrightFocus Foundation. This material is
glycemic diet (like metformin) have potential to based upon work supported by the USDA-Agricultural
treat AMD. Research Service (ARS), under Agreement No.
We are now undergoing experiments using 58-8050-9-004.
high glycemic fed aged wild-type mice to test
three antidiabetic drugs: acarbose, empagliflozin,
and fenofibrate. Acarbose is an alpha-glucosidase References
inhibitor used to treat type 2 diabetes mellitus,
1. Kim J-B, Lad EM. Therapeutic options under
operating to control post-prandial glucose levels development for Nonneovascular age-related macular
[28]. Empagliflozin is an SGLT2 inhibitor that degeneration and geographic atrophy. Drugs Aging.
promotes glucose excretion via the kidney [29]. 2021;38(1):17–27.
Fenofibrate is not used to treat hyperglycemia but 2. Falk T, Congrove NR, Zhang S, McCourt AD,
Sherman SJ, McKay BS. PEDF and VEGF-A
is an effective treatment for diabetic retinopathy output from human retinal pigment epithelial cells
[30]. Fenofibrate works as an agonist of PPARα; grown on novel microcarriers. J Biomed Biotechnol.
it enhances lipid efflux and reduces ocular inflam- 2012;2012:e278932.
mation [31]. None of these drugs are currently 3. Brilliant MH, Vaziri K, Connor TB, Schwartz SG,
Carroll JJ, McCarty CA, et al. Mining retrospective
under consideration as an AMD treatment; thus, data for virtual prospective drug repurposing:
preclinical data is essential. L-DOPA and age-related macular degeneration. Am
J Med. 2016;129(3):292–8.
4. Figueroa AG, Boyd BM, Christensen CA, Javid CG,
McKay BS, Fagan TC, et al. Levodopa positively
6 Conclusions and Outlook affects neovascular age-related macular degeneration.
Am J Med. 2021;134(1):122–128.e3.
There remains potential for repurposed drugs to 5. Barzilai N, Crandall JP, Kritchevsky SB, Espeland
treat AMD, but well-controlled randomized clini- MA. Metformin as a tool to target aging. Cell Metab.
2016;23(6):1060–5.
cal trials are still required. Retrospective analyses 6. Xu L, Kong L, Wang J, Ash JD. Stimulation of AMPK
of medical record databases are fraught with prevents degeneration of photoreceptors and the
challenges as most databases rely on diagnosis retinal pigment epithelium. Proc Natl Acad Sci U S A.
codes for diseases rather than relevant medical 2018;115(41):10475–80.
7. Brown EE, Ball JD, Chen Z, Khurshid GS,
data like fundus imaging. Thus, patients with Prosperi M, Ash JD. The common antidiabetic drug
AMD but without proper diagnosis or coding will metformin reduces odds of developing age-related
resemble controls. In addition, diagnosis codes
Repurposing Drugs for Treatment of Age-Related Macular Degeneration 77

macular degeneration. Invest Ophthalmol Vis Sci. the retina from mitochondrial oxidative stress. Exp
2019;60(5):1470–7. Eye Res. 2015;140:94–105.
8. Stewart JM, Lamy R, Wu F, Keenan JD. Relationship 20. Collier RJ, Patel Y, Martin EA, Dembinska O,
between oral metformin use and age-related Hellberg M, Krueger DS, et al. Agonists at the
macular degeneration. Ophthalmol Retina. serotonin receptor (5-HT(1A)) protect the retina from
2020;4(11):1118–9. severe photo-oxidative stress. Invest Ophthalmol Vis
9. Blitzer AL, Ham SA, Colby KA, Skondra Sci. 2011;52(5):2118–26.
D. Association of metformin use with age-related 21. Tullis BE, Ryals RC, Coyner AS, Gale MJ,
macular degeneration: a case-control study. JAMA Nicholson A, Ku C, et al. Sarpogrelate, a 5-HT2A
Ophthalmol. 2021;139(3):302–9. receptor antagonist, protects the retina from light-
10. Eton EA, Wubben TJ, Besirli CG, Hua P, McGeehan induced retinopathy. Invest Ophthalmol Vis Sci.
B, VanderBeek BL. Association of metformin and 2015;56(8):4560–9.
development of dry age-related macular degeneration 22. Coyner AS, Ryals RC, Ku CA, Fischer CM, Patel
in a U.S. insurance claims database. Eur J Ophthalmol. RC, Datta S, et al. Retinal neuroprotective effects of
2021;32(1):417–23. Flibanserin, an FDA-approved dual serotonin receptor
11. Chen Y-Y, Shen Y-C, Lai Y-J, Wang C-Y, Lin K-H, Feng agonist-antagonist. PLoS One. 2016;11(7):e0159776.
S-C, et al. Association between metformin and a lower 23. Ahmed CM, Biswal MR, Li H, Han P, Ildefonso
risk of age-related macular degeneration in patients with CJ, Lewin AS. Repurposing an orally available drug
type 2 diabetes. J Ophthalmol. 2019;2019:1649156. for the treatment of geographic atrophy. Mol Vis.
12. Lee H, Jeon HL, Park SJ, Shin JY. Effect of 2016;22:294–310.
statins, metformin, angiotensin-converting enzyme 24. Biswal MR, Paulson RJ, Vichare R, Lewin
inhibitors, and angiotensin II receptor blockers on AS. Buspirone is protective against oxidative stress
age-related macular degeneration. Yonsei Med J. induced changes in cell culture and animal models
2019;60(7):679–86. of RPE oxidative injury. Invest Ophthalmol Vis Sci.
13. Ambati M, Apicella I, Wang S, Narendran 2021;62(8):3277.
S, Leung H, Pereira F, et al. Identification of 25. Whitcomb EA, Chiu C-J, Taylor A. Dietary glycemia
fluoxetine as a direct NLRP3 inhibitor to treat as a determinant of health and longevity. Mol Asp
atrophic macular degeneration. Proc Natl Acad Sci. Med. 2015;46:14–20.
2021;118(41):e2102975118. 26. Rowan S, Jiang S, Korem T, Szymanski J, Chang
14. Ebeling MC, Polanco JR, Qu J, Tu C, Montezuma M-L, Szelog J, et al. Involvement of a gut-retina axis
SR, Ferrington DA. Improving retinal mitochondrial in protection against dietary glycemia-induced age-­
function as a treatment for age-related macular related macular degeneration. Proc Natl Acad Sci U S
degeneration. Redox Biol. 2020;34:101552. A. 2017;114(22):E4472–81.
15. Terluk MR, Kapphahn RJ, Soukup LM, Gong H, 27. Rowan S, Jiang S, Chang M-L, Volkin J, Cassalman
Gallardo C, Montezuma SR, et al. Investigating C, Smith KM, et al. A low glycemic diet protects
mitochondria as a target for treating age-related macular disease-prone Nrf2-deficient mice against age-­
degeneration. J Neurosci. 2015;35(18):7304–11. related macular degeneration. Free Radic Biol Med.
16. Nashine S, Cohen P, Chwa M, Lu S, Nesburn 2020;150:75–86.
AB, Kuppermann BD, et al. Humanin G (HNG) 28. Alssema M, Ruijgrok C, Blaak EE, Egli L, Dussort P,
protects age-related macular degeneration (AMD) Vinoy S, et al. Effects of alpha-glucosidase-­inhibiting
transmitochondrial ARPE-19 cybrids from drugs on acute postprandial glucose and insulin
mitochondrial and cellular damage. Cell Death Dis. responses: a systematic review and meta-analysis.
2017;8(7):e2951–1. Nutr Diabetes. 2021;11(1):11.
17. Nashine S, Subramaniam SR, Chwa M, Nesburn 29. Wu Q, Liu M, Fang Z, Li C, Zou F, Hu L, et al.
A, Kuppermann BD, Federoff H, et al. PU-91 drug Efficacy and safety of empagliflozin at different doses
rescues human age-related macular degeneration RPE in patients with type 2 diabetes mellitus: a network
cells; implications for AMD therapeutics. Aging. meta-analysis based on randomized controlled trials.
2019;11(17):6691–713. J Clin Pharm Ther. 2021;47(3):270–86.
18. Bao A, Nashine S, Atilano S, Chwa M, Federoff 30. Keech AC, Mitchell P, Summanen PA, O’Day J,
H, Kenney MC. Differential responses of AMD Davis TME, Moffitt MS, et al. Effect of fenofibrate on
mitochondrial DNA haplogroups to PU-91, a the need for laser treatment for diabetic retinopathy
mitochondria-­ targeting drug. Mitochondrion. (FIELD study): a randomised controlled trial. Lancet
2021;60:189–200. (Lond Engl). 2007;370(9600):1687–97.
19. Biswal MR, Ahmed CM, Ildefonso CJ, Han P, Li H, 31. Tomita Y, Lee D, Tsubota K, Kurihara T. PPARα
Jivanji H, et al. Systemic treatment with a 5HT1a agonist Oral therapy in diabetic retinopathy.
agonist induces anti-oxidant protection and preserves Biomedicine. 2020;8(10):433.
Part II
Extracellular Vesicles
Extracellular Vesicle RNA Contents
as Biomarkers for Ocular Diseases

Heran Getachew and Eric Pierce

Abstract 1 Introduction

Extracellular vesicles (EVs) are small vesicles Extracellular vesicles (EVs) are nanometer-sized,
secreted from cells into extracellular space. lipid membrane-enclosed vesicles secreted by
EVs contain proteins, lipids, and nucleic acids cells. These vesicles contain lipids, proteins, and
of the cells from which they originate. For this various nucleic acid species of the source cell.
reason, EVs are being studied for use as bio- EVs are a heterogeneous population of vesicles
markers as they can be surrogates for the sta- including exosomes, microvesicles, and apop-
tus of the cell from which they are secreted. totic bodies that are categorized based on biogen-
Moreover, EVs are found in numerous bioflu- esis and range in size from less than 50 nm to
ids and can be taken up by other cells, which micrometers. The biogenesis of exosomes has
allows for transfer of functional cargo, like been previously extensively reviewed [1–4]
RNAs, and changes in gene regulation in the (Fig. 1). Briefly, exosomes originate from late
recipient cell. Several potential RNA biomark- endosomes, which are formed by the inward bud-
ers have been identified in many diseases, and ding of endosomal membrane during maturation.
there is great potential in the vision field for Late endosomes are also referred to as multive-
extracellular RNA biomarkers as a diagnostic sicular bodies due to their morphology and invag-
tool as well as a measure for treatment inations in the multivesicular bodies form
efficacy. intraluminal vesicles. The formation of the multi-
vesicular bodies and intraluminal vesicles
Keywords depends on the endosomal sorting complex
required for transport (ESCRT). This complex is
Extracellular vesicles · Biomarkers · RNA · comprised of four complexes (ESCRT 0-III) and
miRNA · Inherited retinal degenerations · associated proteins (VPS4, VTA1, and ALIX)
Ocular diseases that coordinate membrane formation and vesicle
budding and facilitate protein cargo sorting.
Additionally, tetraspanins, CD9, CD63, and
H. Getachew · E. Pierce (*) CD81, play a role in EV development and are
Ocular Genomics Institute, Department of typically used as markers. The biogenesis of
Ophthalmology, Massachusetts Eye and Ear microvesicles differs as these are formed as a
Infirmary, Harvard Medical School, Boston, MA,
USA result of outward budding and pinching off of the
e-mail: eric_pierce@meei.harvard.edu plasma membrane [6]. Cargos are selectively

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 81


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_13
82 H. Getachew and E. Pierce

Fig. 1 Extracellular vesicle biogenesis. Exosomes origi- brane, also relying on ESCRT proteins and GTPases, such
nate from multivesicular bodies and are formed in an as ARF6. Apoptotic bodies are formed in cells undergoing
ESCRT-dependent manner and are secreted through exo- apoptosis, and these vesicles bleb from the plasma mem-
cytosis. Microvesicles bud directly from the plasma mem- brane. (Figure adapted from Dang et al. [5])

recruited into microvesicles by proteins such as techniques and markers for characterizing EVs
the small GTPase, ARF6, and are also regulated [11]. Many of the lipids found within EVs are
by ESCRT proteins [7, 8]. The largest EVs, apop- derived from lipids found in the cellular plasma
totic bodies, are formed as a cell undergoes apop- membrane, such as sphingolipids and phosphati-
tosis, and blebbing from the plasma membrane dylcholine [12]. Single- and double-stranded
occurs. DNA and mitochondrial DNA have been detected
EVs contain proteins, lipids, and nucleic in EVs [13, 14]. Moreover, several RNA species
acids, as well as organelles, histones, and chro- have been identified in EVs, including mRNAs,
matin, which are found in apoptotic bodies [9, miRNAs, piRNAs, and long and short noncoding
10]. The proteins and lipids found in EVs typi- RNA; however, the majority of RNAs thought to
cally arise from their biogenesis mechanisms. be present within EVs are small RNAs [15–18].
For example, exosomes are enriched with ESCRT RNAs are protected by the lipid bilayer of EVs,
proteins and tetraspanins, microvesicles are which provides protection for enzymes and other
enriched with ß-1 integrin and ARF6, and apop- proteins as well [19].
totic bodies are enriched with annexin V; how- The physiological role of EVs was initially
ever, they can also express CD9, CD63, and believed to be a cellular waste removal mecha-
CD81. Therefore, the International Society for nism; however, mounting evidence suggests a
Extracellular Vesicles suggests using multiple role in intercellular communication [20–23]. One
Extracellular Vesicle RNA Contents as Biomarkers for Ocular Diseases 83

mechanism for EV-mediated intercellular com- degeneration, there was increased secretion of
munication is through transfer and uptake of EVs by the retina; however, to date, no RNA bio-
RNA, which can alter gene regulation in the markers have been defined for retinitis pigmen-
recipient cells [24]. Furthermore, EVs can carry tosa or other IRDs [31]. Identification of
gRNA or Cas9 proteins of the CRISPR/Cas sys- biomarkers for IRDs would be of great benefit in
tem expressed in cells following transfection, assessing efficacy of therapies for IRDs as well
thereby transferring the gene editing activity of as additional diagnostic tool in other ocular
this system between donor and recipient cells, disorders.
providing further evidence for EVs-mediated Tear fluid is a noninvasively available biofluid,
gene expression alteration [25]. and while volume may be a limiting factor, a few
Given the information contained within EVs, studies have identified potential biomarkers in
it has been suggested and demonstrated that they tear fluid [32]. There are currently two FDA-­
can provide valuable information about the status approved tests using tear fluid, InflammaDry,
of their parent cells and have been studied as bio- which measures levels of MMP-9, and Advanced
markers in several diseases including cancers, Tear Diagnostics, which measures lactoferrin, as
and similar studies are emerging in the vision diagnostic tools for dry eye disease [32, 33]. EVs
field. from tears contain RNAs, and one study found
that the total miRNA was higher in tears from
Alzheimer disease patients compared to healthy
2 EV RNAs as Biomarkers controls [34]. Moreover, miRNA-200b-5p was
in Ocular Disorders elevated in tears from individuals with
Alzheimer’s disease [34]. Pucker et al. collected
The FDA/NIH Biomarker Working Group defines tear fluid from healthy individuals and using
a biomarker as “a defined characteristic that is RNA-seq were able to identify miRNAs con-
measured as an indicator of normal biological tained in tear EVs which can be used as a refer-
processes, pathogenic processes, or responses to ence in future studies examining the RNA from
an exposure or intervention, including therapeu- tears of individuals with ocular diseases [35].
tic interventions” [26]. Because EVs can be Several small RNAs have been identified in
found within numerous biological specimens, the vitreous including miR-9, miR-9*, miR-­
including plasma, serum, urine, saliva, aqueous 125a-­3p, miR-184, miR-211, miR-214, miR-­
humor, vitreous humor, tears, and cerebrospinal 302c, miR-452, miR-628, and miR-639, which
fluid, they are an excellent candidate to be used are either under expressed or not detectable in
as biomarkers. The cancer and neurodegenera- serum [36]. A study examined the vitreous sam-
tion fields have reported success in identifying ples of patients with primary vitreoretinal lym-
RNA biomarkers from EVs, demonstrating their phoma (PVRL) or chronic vitritis as they are
promising potential in the vision field [27–29]. often difficult to distinguish clinically [37]. The
One potential application in the vision field is for results showed that miR-92, miR-19b, and miR-­
inherited retinal degenerations (IRDs), which are 21 were present at significantly higher levels in
a heterogeneous group of diseases with muta- the vitreous of patients with PVRL than in vitritis
tions in 280 genes that cause vison loss and blind- samples, suggesting that these miRNAs biomark-
ness [30]. While gene therapies, such as the ers can be used in addition to morphological,
FDA-approved therapy for RPE65-associated immunological, and genetic evaluations to make
retinal degenerations, are promising treatments diagnoses and monitor patient progress [37].
for IRDs, the clinical measures to assess improve- Tanaka et al. found 11 significantly upregulated
ment in vison following application of therapies and 18 significantly downregulated miRNAs in
takes time, highlighting the need for additional the aqueous humor of individuals with glaucoma
measures of clinical efficacy. One study found compared to healthy controls [38]. Another study
that, in mice with Pde6b-associated retinal identified a long noncoding RNA
84 H. Getachew and E. Pierce

(ENST00000607393) in aqueous humor as a 3 Concluding Remarks


potential biomarker and therapeutic target for
glaucoma [39]. While more invasive than collect- In order to use EVs as biomarkers for ocular dis-
ing blood, aqueous and vitreous humors provide eases, ocular-specific EV markers and contents
a way to isolate and characterize locally secreted need to be fully defined in health and disease
EVs, from which specific markers and cargo can states [45]. Thus, a potential focus of future stud-
be defined and potentially identified in blood ies is characterization of the contents of EVs, like
circulation. RNAs, secreted from the visual system. Defining
Identifying biomarkers in circulation is the EVs secreted from the various cell types of
appealing as it is an available biofluid and is rich the visual system will allow for evaluation of EV
with EVs [40]. Some circulating RNA biomark- contents as ocular biomarkers in various bioflu-
ers have been identified in circulation, such as ids, which can be assessed for changes during
miR146a which was upregulated in the serum of disease, which will provide a method to monitor
patients with uveal melanoma [41]. Additionally, disease progression, additional endpoints in clin-
miR-126, miR-410, and miR-19a were upregu- ical trials, and therapeutic targets.
lated in the serum of patients with atrophic age-­
related macular degeneration, and another study
identified five other circulating miRNAs as References
potential biomarkers for neovascular age-related
macular degeneration [42, 43]. Since tissues like 1. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: bio-
genesis, biologic function and clinical potential. Cell
the retina and retinal pigment epithelium (RPE) Biosci. 2019;9(1):19.
are not able to be biopsied, it would be of great 2. Patil AA, Rhee WJ. Exosomes: biogenesis, com-
benefit to characterize EV-derived RNAs from position, functions, and their role in pre-metastatic
the retina, RPE, and other cells in the visual sys- niche formation. Biotechnol Bioprocess Eng.
2019;24(5):689–701.
tem in order to: (1) determine if they are detect- 3. Hessvik NP, Llorente A. Current knowledge on exo-
able in circulation and (2) change during disease some biogenesis and release. Cell Mol Life Sci.
progression in order to be used as a surrogate 2018;75(2):193–208.
measure of retinal health status. 4. Colombo M, Raposo G, Théry C. Biogenesis, secre-
tion, and intercellular interactions of exosomes and
In vitro studies can also be helpful in charac- other extracellular vesicles. Annu Rev Cell Dev Biol.
tering ocular EVs, especially given the flexibil- 2014;30(1):255–89.
ity compared to studies using limited patient 5. Dang XTT, Kavishka JM, Zhang DX, Pirisinu M, Le
sample such as tears or aqueous and vitreous MTN. Extracellular vesicles as an efficient and versa-
tile system for drug delivery. Cell. 2020;9(10):2191.
humors. In a 2017 study, Klingeborn et al. iden- 6. D’Souza-Schorey C, Clancy JW. Tumor-derived
tified 631 proteins in EVs secreted from cul- microvesicles: shedding light on novel microenviron-
tured polarized RPE cells [44]. Of these proteins, ment modulators and prospective cancer biomarkers.
299 were uniquely released apically and 94 Genes Dev. 2012;26(12):1287–99.
7. Taylor J, Bebawy M. Proteins regulating microves-
uniquely released basolaterally, indicating that icle biogenesis and multidrug resistance in cancer.
EVs can reflect polarity-specific functions of Proteomics. 2019;19(1–2):e1800165.
RPE cells. This study provides key information 8. Muralidharan-Chari V, Clancy J, Plou C, Romao M,
on the proteome of the RPE and can be used a Chavrier P, Raposo G, et al. ARF6-regulated shedding
of tumor cell-derived plasma membrane microvesi-
reference for future studies aimed at identifying cles. Curr Biol. 2009;19(22):1875–85.
protein biomarkers of the other cell types of the 9. Battistelli M, Falcieri E. Apoptotic bodies: particular
visual system. Future studies defining the RNA extracellular vesicles involved in intercellular com-
contents of EVs secreted from specific cell types munication. Biology (Basel). 2020;9(1):21.
10. Emlen W, Niebur J, Kadera R. Accelerated
of the visual system will allow for the identifica- in vitro apoptosis of lymphocytes from patients
tion of additional RNA biomarkers and thera- with systemic lupus erythematosus. J Immunol.
peutic targets. 1994;152(7):3685–92.
Extracellular Vesicle RNA Contents as Biomarkers for Ocular Diseases 85

11. Thery C, Witwer KW, Aikawa E, Alcaraz MJ, 25. Chen R, Huang H, Liu H, Xi J, Ning J, Zeng W, et al.
Anderson JD, Andriantsitohaina R, et al. Minimal Friend or foe? Evidence indicates endogenous exo-
information for studies of extracellular vesicles somes can deliver functional gRNA and Cas9 protein.
2018 (MISEV2018): a position statement of the Small. 2019;15(38):e1902686.
International Society for Extracellular Vesicles and 26. Cagney DN, Sul J, Huang RY, Ligon KL, Wen
update of the MISEV2014 guidelines. J Extracell PY, Alexander BM. The FDA NIH Biomarkers,
Vesicles. 2018;7(1):1535750. EndpointS, and other Tools (BEST) resource in neuro-­
12. Subra C, Laulagnier K, Perret B, Record M. Exosome oncology. Neuro-Oncology. 2018;20(9):1162–72.
lipidomics unravels lipid sorting at the level of multi- 27. Kim KM, Meng Q, Perez de Acha O, Mustapic
vesicular bodies. Biochimie. 2007;89(2):205–12. M, Cheng A, Eren E, et al. Mitochondrial RNA in
13. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Alzheimer’s disease circulating extracellular vesicles.
Costa-Silva B, et al. Double-stranded DNA in exo- Front Cell Dev Biol. 2020;8:581882.
somes: a novel biomarker in cancer detection. Cell 28. Akin D, Wang SK, Habibzadegah-Tari P, Law B,
Res. 2014;24(6):766–9. Ostrov D, Li M, et al. A novel ATG4B antagonist
14. Guescini M, Genedani S, Stocchi V, Agnati inhibits autophagy and has a negative impact on osteo-
LF. Astrocytes and glioblastoma cells release exo- sarcoma tumors. Autophagy. 2014;10(11):2021–35.
somes carrying mtDNA. J Neural Transm (Vienna). 29. Happel C, Ganguly A, Tagle DA. Extracellular
2010;117(1):1–4. RNAs as potential biomarkers for cancer. J Cancer
15. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard Metastasis Treat. 2020;6:32.
CC, Gibson DF, et al. Argonaute2 complexes carry a 30. RetNet. Available at: https://sph.uth.edu/retnet/home.
population of circulating microRNAs independent of htm
vesicles in human plasma. Proc Natl Acad Sci U S A. 31. Sahaboglu A, Vidal-Gil L, Sancho-Pelluz J. Release
2011;108(12):5003–8. of retinal extracellular vesicles in a model of retinitis
16. Ni H, Capodici J, Cannon G, Communi D, Boeynaems Pigmentosa. Adv Exp Med Biol. 2019;1185:431–6.
JM, Karikó K, et al. Extracellular mRNA induces 32. Tamhane M, Cabrera-Ghayouri S, Abelian G,
dendritic cell activation by stimulating tumor necrosis Viswanath V. Review of biomarkers in ocular matri-
factor-alpha secretion and signaling through a nucleo- ces: challenges and opportunities. Pharm Res.
tide receptor. J Biol Chem. 2002;277(15):12689–96. 2019;36(3):40.
17. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche 33. Roy NS, Wei Y, Kuklinski E, Asbell PA. The grow-
L, Sena-Esteves M, et al. Glioblastoma microvesicles ing need for validated biomarkers and endpoints for
transport RNA and proteins that promote tumour dry eye clinical research. Invest Ophthalmol Vis Sci.
growth and provide diagnostic biomarkers. Nat Cell 2017;58(6):BIO1–BIO19.
Biol. 2008;10(12):1470–6. 34. Kenny A, Jiménez-Mateos EM, Zea-Sevilla MA,
18. Hoy AM, Buck AH. Extracellular small RNAs: what, Rábano A, Gili-Manzanaro P, Prehn JHM, et al.
where, why? Biochem Soc Trans. 2012;40(4):886–90. Proteins and microRNAs are differentially expressed
19. Momen-Heravi F, Getting SJ, Moschos in tear fluid from patients with Alzheimer’s disease.
SA. Extracellular vesicles and their nucleic Sci Rep. 2019;9(1):15437.
acids for biomarker discovery. Pharmacol Ther. 35. Pucker AD, Fortinberry H, Ngo W, Postnikoff CK,
2018;192:170–87. Nichols JJ. Differential expression of extracellular
20. Pitt JM, Kroemer G, Zitvogel L. Extracellular vesicles and microRNAs in normal human tear sam-
vesicles: masters of intercellular communication ples. ARVO Annu Meet. 2020;61:1393.
and potential clinical interventions. J Clin Invest. 36. Ragusa M, Caltabiano R, Russo A, Puzzo L,
2016;126(4):1139–43. Avitabile T, Longo A, et al. MicroRNAs in vitreus
21. Maacha S, Bhat AA, Jimenez L, Raza A, Haris M, humor from patients with ocular diseases. Mol Vis.
Uddin S, et al. Extracellular vesicles-mediated inter- 2013;19:430–40.
cellular communication: roles in the tumor microenvi- 37. Kakkassery V, Schroers R, Coupland SE, Wunderlich
ronment and anti-cancer drug resistance. Mol Cancer. MI, Schargus M, Heinz C, et al. Vitreous microRNA
2019;18(1):55. levels as diagnostic biomarkers for vitreoretinal lym-
22. Berumen Sánchez G, Bunn KE, Pua HH, Rafat phoma. Blood. 2017;129(23):3130–3.
M. Extracellular vesicles: mediators of intercellu- 38. Tanaka Y, Tsuda S, Kunikata H, Sato J, Kokubun T,
lar communication in tissue injury and disease. Cell Yasuda M, et al. Profiles of extracellular miRNAs
Commun Signal. 2021;19(1):104. in the aqueous humor of glaucoma patients assessed
23. Yoon YJ, Kim OY, Gho YS. Extracellular vesicles as with a microarray system. Sci Rep. 2014;4:5089.
emerging intercellular communicasomes. BMB Rep. 39. Xie L, Mao M, Wang C, Zhang L, Pan Z, Shi J, et al.
2014;47(10):531–9. Potential biomarkers for primary open-angle glau-
24. Valadi H, Ekstrom K, Bossios A, Sjostrand M, coma identified by long noncoding RNA profiling in
Lee JJ, Lotvall JO. Exosome-mediated transfer of the aqueous humor. Am J Pathol. 2019;189(4):739–52.
mRNAs and microRNAs is a novel mechanism 40. Palviainen M, Saraswat M, Varga Z, Kitka D,
of genetic exchange between cells. Nat Cell Biol. Neuvonen M, Puhka M, et al. Extracellular ves-
2007;9(6):654–9. icles from human plasma and serum are carriers of
86 H. Getachew and E. Pierce

e­ xtravesicular cargo—implications for biomarker dis- 43. Ertekin S, Yıldırım O, Dinç E, Ayaz L, Fidancı
covery. PLoS One. 2020;15(8):e0236439. SB, Tamer L. Evaluation of circulating miRNAs
41. Ragusa M, Barbagallo C, Statello L, Caltabiano R, in wet age-related macular degeneration. Mol Vis.
Russo A, Puzzo L, et al. miRNA profiling in vitreous 2014;20:1057–66.
humor, vitreal exosomes and serum from uveal mel- 44. Klingeborn M, Dismuke WM, Skiba NP, Kelly U,
anoma patients: pathological and diagnostic implica- Stamer WD, Bowes Rickman C. Directional exo-
tions. Cancer Biol Ther. 2015;16(9):1387–96. some proteomes reflect polarity-specific functions in
42. ElShelmani H, Wride MA, Saad T, Rani S, Kelly DJ, retinal pigmented epithelium monolayers. Sci Rep.
Keegan D. Identification of novel serum MicroRNAs 2017;7(1):4901.
in age-related macular degeneration. Translational 45. Klingeborn M, Dismuke WM, Bowes Rickman C,
vision. Sci Technol. 2020;9(4):28. Stamer WD. Roles of exosomes in the normal and
diseased eye. Prog Retin Eye Res. 2017;59:158–77.
Proteomics of Retinal Extracellular
Vesicles: A Review into
an Unexplored Mechanism
in Retinal Health and AMD
Pathogenesis

Adrian V. Cioanca, Riccardo Natoli,


and Yvette Wooff

Abstract and RPE-derived EV largely reflect the func-


tion of the host cells, while in disease RPE-EV
Extracellular vesicles (EV) are nanosized protein composition becomes altered, favor-
delivery vehicles that participate in cell-to-cell ing inflammatory modulation and potentially
communication through the selective transfer contributing to drusen formation. While these
of molecular materials including RNA, DNA, studies shed light on the potential roles of EV
lipids, and proteins. In the retina, the role of proteins in the neural retina and RPE, it is
EV proteins is largely unclear, in part due to clear that comprehensive proteomic and
the lack of studies and the depth of proteomic molecular studies are required, in particular
analyses of EV cargo. This review summa- using in vivo models of retinal degenerations.
rizes the existing knowledge on retinal EV
proteins and provides a comparative reanaly- Keywords
sis of existing retinal EV proteomic datasets.
Collective findings highlight that in homeosta- Proteomics · Extracellular vesicles · Retinal
sis, the protein components of neural retinal degeneration · Retinal pigmented epithelium ·
Age-related macular degeneration · Exosome

A. V. Cioanca · R. Natoli · Y. Wooff (*)


John Curtin School of Medical Research, The
Abbreviations
Australian National University, Canberra, ACT,
Australia AMD Age-related macular degeneration
The School of Medicine and Psychology, The BRB Blood retinal barrier
Australian National University, Canberra, ACT, EV Extracellular vesicles
Australia miRNA microRNA
e-mail: valerin.cioanca@anu.edu.au;
Riccardo.natoli@anu.edu.au;
RPC Retinal progenitor cells
Yvette.wooff@anu.edu.au RPE Retinal pigmented epithelium

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 87


J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_14
88 A. V. Cioanca et al.

1 Introduction 2 Part I: Retinal EV Proteome


in Normal Health
Extracellular vesicles (EV) are naturally occur-
ring endogenous membrane-enclosed delivery 2.1 Part I.I: Retinal EV Proteins
vehicles [1] that are released by nearly all cell Have Distinct Roles
types and function to transfer essential molecu- in the Neural Retina and RPE
lar cargo, including DNA, mRNA, noncoding to Support Host-Cell Function
RNA (including microRNA (miRNA)), lipids, in Retinal Homeostasis
and proteins. EV selectively encapsulates and
transports this cargo to target cells to induce a There have only been a few studies investigating
range of biological responses, including in the retinal EV proteome; however, collectively,
immune modulation [2–4]. EV comprise vari- these studies show that the EV proteome from the
ous subclasses of vesicle fractions which differ neural retina and RPE are distinct (Fig. 1a), with
in size, cellular origin, composition, and cargo each regulating different pathways involved in
[3, 5]; however, the most widely studied sub- maintaining retinal homeostasis. In a recent study
class of EV are exosomes, which range in size by Mighty et al. [18], the EV proteome of the
from ~30 to 150 nm and are derived from the adult neural retina was explored using retinal
endocytic pathway. explants from C57Bl/6J mice [18]. 957 proteins
Cell-to-cell communication is critical for tis- were found to be shared between the retinal
sue survival, with dysregulation in EV communi- explants and isolated EV; 82 of which were
cation linked to the development of inflammatory uniquely found in the EV [18]. Protein ontology
and neurodegenerative diseases [2]. In the retina, analysis of these 82 EV proteins identified bio-
EV have been shown to function to maintain logical terms such as “response to DNA damage,”
homeostasis and immune modulation [1, 6–8] “phosphorylation,” “mRNA processing,” and
through the selective incorporation and transfer “DNA-dependent transcription regulation,” while
of molecular cargo between cells, including small molecular functions were related to gene regula-
gene regulators called miRNA [1, 6, 9]. However, tion, and phosphorylation were highly significant.
while investigations have focused on the role and Specifically, these pathways involved protein,
transfer of EV miRNA [1, 6, 9] or individual pro- RNA, DNA, nucleotide, and ATP binding, as well
teins [10–13], little has been done in characteriz- as transferase and kinase activity [18]. A limita-
ing or understanding the entire proteomic tion of this study was that no further bioinformatic
contribution of retinal EV in health or analyses were conducted on either the unique EV
degeneration. proteins or the EV/retinal proteins. However, the
This review therefore collates and reanalyzes results available suggest that retinal EV protein
the available literature and data on retinal EV transfer is required for nucleic acid processing,
proteomics, comparing the role of retinal EV surveillance of DNA damage, and transcriptional
proteins in retinal homeostasis and disease, regulation. As this is the only study to date inves-
including in the pathogenesis of age-related tigating the retinal EV proteome, it is apparent
macular degeneration (AMD). With limited that additional studies, in particular in vivo and in
studies available, it is apparent that, in order to diseased models and human tissue, are required to
further unravel the role of the EV proteome in truly understand the biological role that retinal
retinal health, additional standardized studies EV proteins may play in the retina in health and
need to be conducted and the available pro- in degeneration. Further, elucidating the surface
teomic datasets queried in more depth than is membrane protein composition could provide
currently reported. This review therefore addi- insight into the cell-specific origin of EV in the
tionally attempts to further delve into each pro- retina and which cells they are targeted for,
teomic dataset in retinal health and degeneration enabling the mapping of EV-communication
and highlights key areas where additional pathways in the retina. While brief, Mighty et al.
research could be focused in the future. [18] reported that key photoreceptor and neuronal
Proteomics of Retinal Extracellular Vesicles: A Review into an Unexplored Mechanism in Retinal Health… 89

Fig. 1 Reanalysis of retinal EV proteomic databases. (a) chical clustering and alluvial plots were created with
Venn diagram comparing overlapping and unique proteins ggVennDiagram [14], RVenn [15] and ggalluvial R pack-
in each retinal cell type EV. (b) Hierarchical cluster shows ages [16]. EV markers found in drusen as listed on
cell type specific proteomes, with RPE-EV and neural Vesiclepedia [17] shown in red. BL basolateral, EMT epi-
retinal proteins also present in drusen. (c) Pathway analy- thelial mesenchymal transition, IS inner segments, OLM
sis of retinal EV proteins found in drusen. (d) Diagram outer limiting membrane, OS outer segments
showing EV deposition in drusen. Venn diagrams, hierar-

proteins (Rhodopsin and NeuN, respectively) allows the retina to exist in an immune privileged
were present in the EV, suggesting a photorecep- and closed environment, the RPE also plays key
tor origin or potentially from amacrine and gan- roles in regulating retinal health, in particular
glion cells. Previous findings from our lab group maintaining photoreceptor function [20, 21].
support this result, with a depletion in retinal EV Proteomic studies exploring the role of RPE-EV
concentration correlated to decreased photorecep- have largely been conducted in vitro using mono-
tor survivability in vivo [19]. layer cultures of a commonly used non-­
The majority of the available literature has transformed human RPE cell line called aRPE-19.
been focused on understanding the RPE-EV pro- However, the use of aRPE19 monolayer cultures
teome in health (Sect. 2.1) and disease (Sect. 3). poses issues when understanding the polarized
Functioning not only as a physical barrier that roles of the RPE at each of the respective apical
90 A. V. Cioanca et al.

(retinal) and basolateral (systemic) faces. In fact, however is that the cells were grown in culture
Klingeborn et al. [22] showed using immunocap- for 6 weeks which is likely to affect cellular
ture of CD81-expressing EV from the plasma and behavior through responses to their artificial
from EV collected from aRPE-19 monolayer cul- environment, potentially changing EV profiles
ture media that the proteome of aRPE-19 EV [22] and cargo [31]. In fact, in this study, it was dem-
was more similar to the apically derived RPE EV onstrated that significantly different numbers of
population in their earlier study than from the EV were released between the apical and baso-
basolateral EV population [23]. As the polarity of lateral faces dependent on the addition of FBS
secretions from the RPE dictates separate impor- or B27 as culture media supplements [23].
tant interactions with the systemic circulation and Another limitation of this study was that, while
the retina, the proteomic profile of EV from each proteomics were conducted on the RPE cells
side of the RPE may play unique and mutually themselves, there was no comparative analysis
distinct roles in retinal health and disease mani- between the EV populations and the RPE host
festation. Importantly, as basolateral RPE-EV can cell [23] nor in-depth pathway or network anal-
enter the systemic circulation, they represent yses to elucidate potential pathways regulated
potential biomarkers of retinal disease [22]. by these EV.
Earlier work by Klingeborn et al. [23] The results of these studies support a mecha-
explored this polarized nature by isolating and nism by which retinal EV modulate key functions
characterizing the proteomic composition of EV of their host cell and drive homeostatic mainte-
derived from mature polarized primary porcine nance in the retina. However, understanding the
RPE cells. These cells were grown on perme- role of retinal EV in disease models is important
able support scaffolds allowing for EV release to elucidate pathogenic consequences of dysreg-
from the apical as well as basolateral faces of ulated EV communication and the role that EV
the RPE to be collected in the media [23]. Both proteins may play in disease pathogenesis.
exosomes and dense EV fractions from each
compartment were isolated and compared [23,
24]. 631 proteins were identified in total from 3 Part II: Retinal EV Proteome
this study, with 299 unique to the apical and 94 in Disease
to the basolateral fractions, respectively.
In-depth analyses demonstrated that the unique 3.1 Part II.I: RPE EV Proteins
proteins isolated in exosomes from either side Modulate Cell Death,
of the RPE reflected the polar-specific functions Oxidative Stress,
of the RPE, while both apical and basolateral and Inflammatory Pathways
dense-EV were found to contain extracellular in Retinal Degeneration
matrix proteins including COL18A1 which is
essential for RPE function and Bruch’s mem- AMD is the leading cause of vision loss in the
brane structure [23]. Interestingly, proteins such Western World, characterized by the progressive
as complement component C3 and amyloid beta death of the light-sensing photoreceptor cells and
were found uniquely in the basolateral dense- underlying RPE, resulting in irreversible blind-
EV population but not the apical [23]. As these ness [32]. Oxidative stress and inflammation are
proteins are associated with AMD [25–27] and widely accepted as key pathological features
have been isolated in drusen [28, 29], this result implicated in the development and progression of
suggests that basolateral RPE-EV specifically AMD [27, 33]. Given the essential role that the
may contribute to disease pathogenesis, or RPE plays in regulating oxidative stress and the
potentially drusen formation as proposed by associations between RPE dysfunction and AMD
Curcio et al. [22, 30] and briefly discussed in [34], the majority of the literature has been
Sect. 3.2, therefore making them potential diag- focused on understanding the RPE-EV proteome
nostic biomarkers. A limitation of this study to decipher if secretions of EV and their protein
Proteomics of Retinal Extracellular Vesicles: A Review into an Unexplored Mechanism in Retinal Health… 91

cargo may contribute to the onset and pathogenic 4 other proteins were verified and confirmed as
features of AMD and be used as disease putative diagnostic biomarkers using a receiver
biomarkers. operating characteristic curve analysis, with
Using reverse phase protein arrays, Biasutto cytokeratin 8 having an area under the curve
et al. [35] explored the effects of oxidative score of 0.929 [38]. While this study suggests
stress on the RPE-EV proteome to determine that RPE contributes the major source of EV in
how cell-­ free phosphorylated signaling pro- the aqueous humor, they did not analyze the data
teins could arise in the vitreous of patients with from Müller EV except for a comparison of the
AMD [36]. Thirteen phosphoproteins involved number of EV proteins shared between both
in pathways regulating cell survival, prolifera- groups. Further, the contribution of EV from
tion, metabolism, and apoptosis were found to other retinal cell types such as photoreceptors
be differentially expressed in aRPE-19 exo- was not investigated at all.
somes following exposure to oxidative stress
using either 2.5 μM rotenone or 62.5 μM
methyl viologen. Notably, phosphorylated 3.2 Part II.II: RPE-EV Contributes
VEGFR2 and PDGFRβ have also been previ- to Drusen Formation
ously identified in the vitreous of human AMD
patients [37]. The authors concluded from this Drusen formation is a key facet of AMD patho-
work that the phosphoprotein content of RPE genesis [29, 39, 40]. A growing body of evidence
exosomes following oxidative stress may exists for the formation of drusen to be caused, in
reflect a change in signaling pathway activa- part, by an accumulation of EV. EV markers such
tion and suggest that RPE-exosomes phospho- as CD63 and Lamp2 have all been identified in
proteins modulate or alter signaling pathways drusen deposits of AMD patients [38, 41–44] and
in recipient cells during disease. As phosphor- were suggested by Wang et al. [42] to be released
ylated signaling proteins can be detected in the from the RPE [42, 43]. In a currently unpublished
vitreous of AMD patients, this study supports study by Flores-Bellver et al. [41], drusen and
the potential use of EV proteins as a biomarker AMD-associated proteins were reported in EV
for disease state. collected from human-induced pluripotent stem
Further evidence for the use of RPE-EV pro- cell-derived RPE following cigarette-­ smoke
teins as disease biomarkers comes from a study stimulation to induce oxidative stress [41].
by Kang et al. [38], in which proteins identified Enriched EV proteins were further shown to reg-
in the exosomes of aRPE-19 cells subjected to ulate pathways involved in oxidative stress and
400 μM paraquat, an inducer of oxidative stress, inflammation. While the authors suggest that
were also detected in the aqueous humor of wet-­ RPE-EV may contribute to drusen formation
AMD patients [38]. Using liquid chromatography-­ [41], unfortunately, as this dataset has not yet
tandem mass spectrometry, 37 proteins were been made available, limited conclusions can be
identified both in exosomes from aRPE-19 and in drawn. In support of this hypothesis, however,
the aqueous humor of wet-AMD patients along fibulin-3 (FIB3), a secreted glycoprotein
with 18 proteins identified in exosomes from expressed in the retina and associated with dru-
Müller glia (used as a control) that had under- sen formation was detected on the apical surface
gone oxidative stress [38]. Pathway analysis of of RPE, photoreceptor outer segments, and in
the 37 aRPE-19/aqueous humor exosome pro- drusen deposits in human AMD retinas,
teins identified roles in metabolic and immune co-­
­ localizing with ALIX/PDCD6IP, an EV
system processes [38]. Of note cathepsin D was marker [45, 46].
found to be enriched in these groups compared to In an attempt to investigate this hypothesis,
the respective control groups, which the authors the available datasets reviewed in this paper
suggest may be a mechanism to resist oxidative (termed “Healthy retinal EV,” [18] “Stressed
stress. Cathepsin D along with cytokeratin 8 and Müller EV,” [38] “Healthy RPE EV,” [23] and
92 A. V. Cioanca et al.

“Stressed RPE EV” [35, 38]) were reanalyzed EV proteomes in retinal health and degeneration
and compared with comprehensive proteomic and provide needed avenues for understanding
datasets from human drusen [28, 39]. It was and possibly treating retinal degenerations.
found that retinal EV populations from each cell
type contained distinct proteomes with only nine
proteins shared between the datasets (Fig. 1a, b). 4 Conclusion
When compared to drusen, over half of the
reported drusen proteins could be found within This review summarizes the brief knowledge on
the EV databases analyzed, unsurprisingly, with retinal EV proteomics, finding that, while lim-
the majority coming from the RPE (Fig. 1b). ited studies are available, the collective data
Interestingly, a slightly larger proportion was supports a role for retinal EV in maintaining
derived from the healthy RPE EV than in the retinal homeostasis through cell-to-cell signal-
stressed EV; however, it is difficult to directly ing of host-cell proteins and regulation of
compare given the variation induced by study important biological processes such as inflam-
design, including cell line, culture media, and mation. Furthered understanding of the pro-
adherent vs. permeable membrane conditions as teomic signature of retinal EV, both surface
highlighted by Klingeborn et al. [23]. Key EV membrane proteins and internal contents, will
marker proteins taken from the “Top 100 EV expand the current knowledge base and shed
markers; Vesiclepedia” [17] such as Annexin light on homeostatic communication pathways
family members 1, 2 5 and 6 (ANXA) were also within the retina including EV uptake and
identified in the drusen database (Fig. 1b, red release mechanisms. In addition, proteomic
text). It is noteworthy to mention that these EV assessment of EV in retinal disease states may
markers were only found within the proportion of elucidate novel roles for retinal EV in retinal
drusen proteins that were also expressed in one degenerations such as drusen formation and
the retinal EV datasets, i.e., within the dashed AMD pathogenesis or development. Finally,
box in Fig. 1b, strengthening the hypothesis that proteomic characterization of retinal EV may
retinal EV may contribute to drusen formation. provide opportunities for EV manipulation and
Finally, pathway analysis was conducted on the therapeutic targeting as well as for use as diag-
proteins shared between at least one of the nostic biomarkers.
reviewed EV datasets and also identified in dru-
sen, using EnrichR against MSigDB Hallmark
2020 database [47, 48], as previously described References
[49]. Results from this analysis showed that these
proteins had significant associations with roles in 1. Hessvik NP, Llorente A. Current knowledge on exo-
some biogenesis and release. Cell Mol Life Sci.
epithelial-to-mesenchymal transition (EMT), cell 2018;75:193–208.
death, and immune processes, all known to be 2. Jan AT, et al. Perspective insights of exosomes in
highly important pathological processes in AMD neurodegenerative diseases: a critical appraisal. Front
[26, 27, 50] (Fig. 1c, d). Although this analysis is Aging Neurosci. 2017;9:317. https://doi.org/10.3389/
fnagi.2017.00317.
largely superficial, it demonstrates the capacity 3. Théry C, et al. Minimal information for studies of
of largely unused proteomics datasets to be repur- extracellular vesicles 2018 (MISEV2018): a position
posed in a meaningful way and gives further evi- statement of the International Society for Extracellular
dence for retinal/RPE-EV contribution to drusen Vesicles and update of the MISEV2014 guidelines. J
Extracell Vesicles. 2018;7:1535750. https://doi.org/10
formation. A systems biology approach integrat- .1080/20013078.2018.1535750.
ing in vivo animal models of retinal degenera- 4. Veziroglu EM, Mias GI. Characterizing extracel-
tion, high-throughput proteomic characterization, lular vesicles and their diverse RNA contents.
bioinformatic analyses of existing and novel Front Genet. 2020;11:700. https://doi.org/10.3389/
fgene.2020.00700.
datasets, and computational modelling is required 5. Doyle LM, Wang MZ. Overview of extracellular ves-
to elucidate the biological importance of retinal icles, their origin, composition, purpose, and methods
Proteomics of Retinal Extracellular Vesicles: A Review into an Unexplored Mechanism in Retinal Health… 93

for exosome isolation and analysis. Cell. 2019;8:727. degenerative diseases. Cham: Springer International
https://doi.org/10.3390/cells8070727. Publishing; 2020. p. 21–5.
6. Morris DR, et al. Exosomal MiRNA transfer 23. Klingeborn M, et al. Directional exosome proteomes
between retinal microglia and RPE. Int J Mol Sci. reflect polarity-specific functions in retinal pigmented
2020;21:3541. epithelium monolayers. Sci Rep. 2017;7:4901. https://
7. Knickelbein JE, et al. Modulation of immune doi.org/10.1038/s41598-­017-­05102-­9.
responses by extracellular vesicles from retinal 24. Andersen JS, et al. Proteomic characterization of the
pigment epithelium. Invest Ophthalmol Vis Sci. human centrosome by protein correlation profiling.
2016;57:4101–7. Nature. 2003;426:570–4. https://doi.org/10.1038/
8. Mead B, Tomarev S. Extracellular vesicle therapy for nature02166.
retinal diseases. Prog Retin Eye Res. 2020;79:100849. 25. Wang M, Su S, Jiang S, Sun X, Wang J. Role of
9. Ahn JY, et al. Release of extracellular vesicle miR-­ amyloid β-peptide in the pathogenesis of age-related
494-­3p by ARPE-19 cells with impaired mitochondria. macular degeneration. BMJ Open Ophthalmol.
Biochim Biophys Acta Gen Subj. 2020;1865:129598. 2021;6:e000774. https://doi.org/10.1136/
https://doi.org/10.1016/j.bbagen.2020.129598. bmjophth-­2021-­000774.
10. Zhang W, et al. Photo-oxidative blue-light stimulation 26. Bora NS, Matta B, Lyzogubov VV, Bora PS.
in retinal pigment epithelium cells promotes exosome Relationship between the complement system, risk
secretion and increases the activity of the NLRP3 factors and prediction models in age-related macular
Inflammasome. Curr Eye Res. 2019;44:67–75. degeneration. Mol Immunol. 2015;63:176–83.
11. Nicholson C, et al. J or H mtDNA haplogroups in reti- 27. Kauppinen A, Paterno JJ, Blasiak J, Salminen
nal pigment epithelial cells: effects on cell physiology, A, Kaarniranta K. Inflammation and its role in
cargo in extracellular vesicles, and differential uptake age-related macular degeneration. Cell Mol Life
of such vesicles by naïve recipient cells. Biochim Sci. 2016;73:1765–86. https://doi.org/10.1007/
Biophys Acta (BBA)-Gen Subj. 2021;1865:129798. s00018-­016-­2147-­8.
12. Gangalum RK, Atanasov IC, Zhou ZH, Bhat SP. 28. Crabb JW. The proteomics of drusen. Cold Spring
αB-crystallin is found in detergent-resistant mem- Harb Perspect Med. 2014;4:a017194. https://doi.
brane microdomains and is secreted via exosomes org/10.1101/cshperspect.a017194.
from human retinal pigment epithelial cells. J Biol 29. Crabb JW, et al. Drusen proteome analysis: an
Chem. 2011;286:3261–9. approach to the etiology of age-related macular
13. Sreekumar PG, et al. αB crystallin is apically secreted degeneration. Proc Natl Acad Sci. 2002;99:14682–7.
within exosomes by polarized human retinal pigment 30. Curcio CA, Johnson M. Structure, function,
epithelium and provides neuroprotection to adjacent and pathology of Bruch’s membrane. Retina.
cells. PLoS One. 2010;5:e12578. 2013;1:466–81.
14. Gao C-H, Yu G, Cai P. ggVennDiagram: an intuitive, 31. Palviainen M, et al. Metabolic signature of extracel-
easy-to-use, and highly customizable R package to lular vesicles depends on the cell culture conditions. J
generate venn diagram. Front Genet. 2021;12:706907. Extracell Vesicles. 2019;8:1596669. https://doi.org/10
https://doi.org/10.3389/fgene.2021.706907. .1080/20013078.2019.1596669.
15. Team RC. R: a language and environment for statisti- 32. Mitchell, P. (2011). Eyes on the future: a clear out-
cal computing. http://www.R-­project.org/. 2019. look on age-related macular degeneration. Deloitte/
16. Brunson JC. Ggalluvial: layered grammar for alluvial Macular degeneration foundation Australia.
plots. J Open Source Softw. 2020;5:2017. 33. Ambati J, Atkinson JP, Gelfand BD. Immunology of
17. Pathan M, et al. Vesiclepedia 2019: a compendium of age-related macular degeneration. Nat Rev Immunol.
RNA, proteins, lipids and metabolites in extracellu- 2013;13:438.
lar vesicles. Nucleic Acids Res. 2019;47:D516–d519. 34. Beranova-Giorgianni S, Giorgianni F. Proteomics
https://doi.org/10.1093/nar/gky1029. of human retinal pigment epithelium (RPE) cells.
18. Mighty J, et al. Analysis of adult neural retina extra- Proteomes. 2018;6:22.
cellular vesicle release, RNA transport and proteomic 35. Biasutto L, Chiechi A, Couch R, Liotta LA, Espina
cargo. Invest Ophthalmol Vis Sci. 2020;61:30. V. Retinal pigment epithelium (RPE) exosomes con-
19. Wooff Y, et al. Small-medium extracellular vesicles tain signaling phosphoproteins affected by oxidative
and their miRNA cargo in retinal health and degenera- stress. Exp Cell Res. 2013;319:2113–23. https://doi.
tion: mediators of homeostasis, and vehicles for tar- org/10.1016/j.yexcr.2013.05.005.
geted gene therapy. Front Cell Neurosci. 2020;14:160. 36. Boellner S, Becker K-F. Reverse phase protein
https://doi.org/10.3389/fncel.2020.00160. arrays-quantitative assessment of multiple biomarkers
20. Boulton M, Dayhaw-Barker P. The role of the reti- in biopsies for clinical use. Microarrays (Basel).
nal pigment epithelium: topographical variation and 2015;4:98–114. https://doi.org/10.3390/microarrays
ageing changes. Eye. 2001;15:384–9. https://doi. 4020098.
org/10.1038/eye.2001.141. 37. Davuluri G, et al. Activated VEGF receptor shed into
21. Strauss O. The retinal pigment epithelium in visual the vitreous in eyes with wet AMD: a new class of
function. Physiol Rev. 2005;85:845–81. biomarkers in the vitreous with potential for predict-
22. Klingeborn M, Skiba NP, Stamer WD, Bowes ing the treatment timing and monitoring response.
Rickman C. In: Rickman CB, et al., editors. Retinal Arch Ophthalmol. 2009;127:613–21.
94 A. V. Cioanca et al.

38. Kang G-Y, et al. Exosomal proteins in the aqueous 45. Grillo SL, et al. The extracellular vesicle marker,
humor as novel biomarkers in patients with neovascu- Alix, co-localizes with Fibulin-3 puncta located in
lar age-related macular degeneration. J Proteome Res. human drusen deposits. Invest Ophthalmol Vis Sci.
2014;13:581–95. 2020;61:–2265.
39. Bergen AA, et al. On the origin of proteins in 46. Grillo SL, et al. Descriptive analysis of Fibulin-3
human drusen: the meet, greet and stick hypothesis. and the extracellular vesicle marker, Alix, in drusen
Prog Retin Eye Res. 2019;70:55–84. https://doi. from a small cohort of postmortem human eyes. Exp
org/10.1016/j.preteyeres.2018.12.003. Eye Res. 2021;203:108422. https://doi.org/10.1016/j.
40. Buschini E, Piras A, Nuzzi R, Vercelli A. Age related exer.2020.108422.
macular degeneration and drusen: neuroinflammation 47. Liberzon A, et al. Molecular signatures database
in the retina. Prog Neurobiol. 2011;95:14–25. (MSigDB) 3.0. Bioinformatics. 2011;27:1739–40.
41. Flores-Bellver M, et al. Drusen proteins are released https://doi.org/10.1093/bioinformatics/btr260.
in association with exosomes. Invest Ophthalmol Vis 48. Chen EY, et al. Enrichr: interactive and col-
Sci. 2021;62:2225. laborative HTML5 gene list enrichment analy-
42. Wang AL, et al. Autophagy, exosomes and dru- sis tool. BMC Bioinf. 2013;14:128. https://doi.
sen formation in age-related macular degeneration. org/10.1186/1471-­2105-­14-­128.
Autophagy. 2009;5:563–4. https://doi.org/10.4161/ 49. Cioanca AV, et al. The role of melanocytes in the
auto.5.4.8163. human choroidal microenvironment and inflam-
43. Wang AL, et al. Autophagy and exosomes in the aged mation: insights from the transcriptome. Pigment
retinal pigment epithelium: possible relevance to dru- Cell Melanoma Res. 2021;34:928–45. https://doi.
sen formation and age-related macular degeneration. org/10.1111/pcmr.12972.
PLoS One. 2009;4:e4160. 50. Zhou M, et al. Role of epithelial-mesenchymal transi-
44. Zhang Z, et al. Emerging role of exosomes in retinal tion in retinal pigment epithelium dysfunction. Front
diseases. Front Cell Dev Biol. 2021;9:643680. https:// Cell Dev Biol. 2020;8:501. https://doi.org/10.3389/
doi.org/10.3389/fcell.2021.643680. fcell.2020.00501.
Part III
Gene Editing
Prime Editing Strategy to Install
the PRPH2 c.828+1G>A Mutation

Salvatore Marco Caruso, Yi-Ting Tsai,


Bruna Lopes da Costa, Masha Kolesnikova,
Laura A. Jenny, Stephen H. Tsang,
and Peter M. J. Quinn

Abstract or absence of photoreceptor outer segments.


Here, we report on a patient with PRPH2-­
Mutations in peripherin 2 (PRPH2) are asso- linked RP who exhibited widespread RPE
ciated with a spectrum of inherited retinal dis- atrophy with a central area of macular atrophy
eases (IRDs) including retinitis pigmentosa sparing the fovea. In future studies, we plan to
(RP) and macular degeneration. As PRPH2 is model the pathobiology of PRPH2-based RP
localized to cone and rod outer segments, using induced pluripotent stem cell (iPSC)-
mutations in PRPH2 lead the disorganization derived retinal organoids. To effectively model
rare mutations using iPSC-derived retinal
Authors Salvatore Marco Caruso, Yi-Ting Tsai and Bruna organoids, we first require a strategy that can
Lopes da Costa have equally contributed to this chapter. install the desired mutation in healthy wild-­

Jonas Children’s Vision Care, and Bernard


S. M. Caruso · B. L. da Costa & Shirlee Brown Glaucoma Laboratory,
Department of Biomedical Engineering, Columbia Department of Ophthalmology,
University, New York, NY, USA Columbia University,
New York, NY, USA
Edward S. Harkness Eye Institute, Department of
e-mail: pq2138@cumc.columbia.edu
Ophthalmology, Columbia University Irving Medical
Center/New York-Presbyterian Hospital, S. H. Tsang
New York, NY, USA Department of Biomedical Engineering, Columbia
University, New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee
Brown Glaucoma Laboratory, Department of Edward S. Harkness Eye Institute, Department of
Ophthalmology, Columbia University, Ophthalmology, Columbia University Irving Medical
New York, NY, USA Center/New York-Presbyterian Hospital,
New York, NY, USA
Y.-T. Tsai
Department of Biomedical Engineering, Columbia Jonas Children’s Vision Care, and Bernard & Shirlee
University, New York, NY, USA Brown Glaucoma Laboratory, Department of
Ophthalmology, Columbia University,
M. Kolesnikova · L. A. Jenny · P. M. J. Quinn (*)
New York, NY, USA
Edward S. Harkness Eye Institute, Department of
Ophthalmology, Columbia University Irving Medical Columbia Stem Cell Initiative, Columbia University,
Center/New York-Presbyterian Hospital, New York, NY, USA
New York, NY, USA
Department of Pathology & Cell Biology, Columbia
University, New York, NY, USA
Institute of Human Nutrition, Columbia University,
New York, NY, USA

97
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_15
98 S. M. Caruso et al.

type iPSC, which can efficiently generate our future work in modeling the pathobiology of
well-laminated retinal organoids. In this study, PRPH2-based RP using induced pluripotent stem
we developed an efficient prime editing strat- cell (iPSC)-derived retinal organoids.
egy for the installation of the pathogenic
PRPH2 c.828+1 G>A splice-site mutation
underlying our patient’s disease. 2 Materials and Methods

Keywords 2.1 Clinical Evaluation


Prime editing · iPSC · Disease modeling ·
PRPH2 · RDS One patient with a confirmed mutation in the
PRPH2 gene was examined at the Edward
S. Harkness Eye Institute at Columbia University
Medical Center (New York, NY). The study was
1 Introduction conducted under Columbia University
Institutional Review Board approval (protocol
Mutations in peripherin 2 (PRPH2), alternatively IRB AAAF1849), and all procedures were per-
known as RDS (retinal degeneration slow), lead formed according to the tenets of the Declaration
to a several inherited retinal diseases (IRDs) of Helsinki. Informed consent was gathered
including retinitis pigmentosa (RP) and macular according to IRB AAAF1849. The patient’s best
degeneration. PRPH2 is a membrane glycopro- corrected visual acuity was measured before
tein that localizes to cone and rod outer segments. administration of topical tropicamide (1%) and
As such, mutations in PRPH2 lead the disorgani- phenylephrine hydrochloride (2.5%) for dilation.
zation or absence of photoreceptor outer seg- Near-infrared autofluorescence (NIR-AF)
ments [2]. (787 nm excitation, 830 nm emission, 30° × 30°)
Prime editing is a novel double-strand break was performed using a Spectralis HRA2
(DSB)-independent clustered- regularly inter- (Heidelberg Engineering, Heidelberg, Germany),
spaced short palindromic repeats (CRISPR)/ and wide-angle color fundus photography was
CRISPR-associated (Cas) system that can ame- performed using an Optos 200Tx unit (Optos;
liorate both transition and transversion mutations PLC, Dunfermline, United Kingdom). Lidocaine
in addition to small deletions and insertions [1]. was administered to a small region of the skin on
A prime editing guide RNA (pegRNA) is used in the lower back, and a biopsy punch (McKesson,
conjunction with a prime editor consisting of the Virginia) was used to perform the skin biopsy.
H840A Streptococcus pyogenes Cas9 (spCas9)
nickase linked to an optimized Moloney murine
leukemia virus (MMLV) reverse transcriptase 2.2 Cloning and Plasmid
(RT). The pegRNA contains a spacer, primer Constructs
binding sequence (PBS), and a RT template
(RTT) containing the desired edit. Together, the A modified version of the Cas9 nickase-reverse
engineered prime editor and the pegRNA form transcriptase plasmid (pCMV-PE2, Addgene
the prime editing 2 strategy (PE2). In the PE3 #132775) was used, pCMV-PE(V4), as previously
strategy, an additional nicking single guide RNA described [7]. The pegRNA was cloned into the
(nsgRNA) is used to nick the non-edited strand, pU6-pegRNA-GG-Vector (Addgene, # 132777),
directing DNA repair to use the edited strand as a and nicking single guide RNA (nsgRNA) was
template [1, 4]. cloned into the pU6-spacer-­acceptor, both using
In this study, we report the clinical evaluation BsaI Golden Gate assembly (NEB), as described
of a RP patient with a novel PRPH2 mutation previously [1, 6]. Oligos were ordered from
within the splicing donor of intron 2. Furthermore, Integrated DNA Technologies (IDT): pU6-Sp-
we develop an installation strategy for this muta- pegRNA-PRPH2-828+1G>A-­sub. pegRNA
tion using prime editing that paves the way for spacer; top strand oligonucleotide, 5′˗
Prime Editing Strategy to Install the PRPH2 c.828+1G>A Mutation 99

C A C C G C T C C T C AT T T G G C T C T- incubated at 56 °C for 1 h followed by a 30 min


TCGGTTTC˗3′; bottom strand oligonucleotide, incubation at 95 °C to stop the proteinase K
5 ′ ˗ C T C T G A A A C C - digestion. This crude extract of DNA is then
GAAGAGCCAAATGAGGAGC˗3′; Installation, ready for PCR purpose.
3′-extenion; top strand oligonucleotide, 5′˗GTCC
AGGGCCTATCTCGAAGA-GCCAAATG˗3′;
bottom strand oligo-nucleotide, 5′˗AAAACA- 2.5 Analysis of Prime Editing
TTTGGCTCTTCGAGATAGGCCCT˗3′; pU6- Efficiency
n s g R N A - P R P H 2 - 8 2 8 + 1 G > A -­s u b :
nsgRNA spacer, top strand oligonucleotide, 5′˗ For the determination of editing efficiency, the
CACCGCTGCTGT-AGTAGCTCAGCA˗3′; and PRPH2 c.828+1 locus (Forward Primer, 5′-cac-
bottom strand oligonucleotide, 5′˗AAACTGC cagacggaggagctca-­3′; Reverse Primer,
TGAGCT-ACTACAGCAGC˗3′. 5′-gagggaggcatgctctcca-3′) were amplified using
primers with Illumina adapters. The amplicon
was submitted to Genewiz for the Amplicon EZ
2.3 Cell Culture and Transfection service. The analysis of the sequencing data was
determined using CRISPResso2 https://cris-
To test editing efficiency of the prime editing, the presso.pinellolab.partners.org/submission
plasmids were transfected into HEK293 cells. (Fig. 2) or RGEN tools http://www.rgenome.net/
The HEK293 cells were seeded 1 day before at pe-­analyzer/#! (Fig. 3). The reads of the ampli-
50000 cells/well in 24-well plates. On the next cons less than 0.1% of the total frequency were
day, the medium was refreshed with 500 ul com- excluded for analysis.
plete medium. For the transfection of PE3, the
plasmid constitution is 750 ng:250 ng:83 ng of
CMV-PE-V4: U6-pegRNA: U6-ngRNA. For 3 Results and Discussion
PE2, only CMV-PE-V4 and U6-pegRNA were
added. Lipofectamine 2000 (Thermo Fisher) was Mutations in PRPH2 lead to a spectrum of IRDs
mixed at 1:1 mass ratio with plasmid DNA. The [2]. In this study, we identified a 67-year-old RP
cells were then collected after 72 h post transfec- patient with the splice site mutation c.828+1
tion for DNA extraction and analysis. To deter- G>A in PRPH2. Color fundus photographs of the
mine the time course of prime editing, the same right and left eye demonstrated widespread RPE
number of cells was seeded in 6-well plate atrophy with a central area of macular atrophy
instead, and the same amount of plasmid DNA/ sparing the fovea. There is sparse intraretinal pig-
lipofectamine mixture was used to transfect the ment migration OS > OD in the inferotemporal
cells. The cells were collected at 72 h, 168 h, and periphery (Fig. 1a, b). Fundus autofluorescence
240 h post transfection. imaging of the right and left eye, respectively,
reveal scattered hypoautofluorescence atrophic
lesions OS > OD extending throughout the
2.4 DNA Extraction periphery with relative sparing of the superior
retina (Fig. 1c, d).
The cells were detached from the wells by tryp- Induced pluripotent stem cell (iPSC)-derived
sin. The cells from each well were washed with retinal organoids are sensitive, quantitative, and
DPBS (without Ca2+ and Mg2+) and resuspended scalable phenotypic assays and have been used
with 50ul DPBS. The cells were then incubated to model several IRDs [5]. Prime editing, a
at 95 °C for 20 mins. Subsequently, after cooling, novel DSB-independent gene editing method
4ul of 20 mg/ml proteinase K (Promega) was now affords us the flexibility to model rare
added to each sample. The samples were then mutations using iPSC-derived retinal organoids
100 S. M. Caruso et al.

Fig. 1 Photoreceptor and RPE death in a 67-year-old RP sparing the fovea. There is sparse intraretinal pigment
patient with PRPH2, c.828+1 G>A. Color fundus and migration OS > OD in the inferotemporal periphery. (c, d)
autofluorescence imaging of a patient with a single patho- Fundus autofluorescence imaging of the right and left eye,
genic variant in PRPH2. (a, b) Color fundus photographs respectively, reveal scattered hypoautofluorescence atro-
of the right and left eye, respectively, demonstrate wide- phic lesions OS > OD extending throughout the periphery
spread RPE atrophy with a central area of macular atrophy with relative sparing of the superior retina

Fig. 2 Prime editing strategy and next generation results generated from triple plasmid transfection of
sequencing results for PRPH2 c.828+1 G>A installation. HEK293 cells with PegRNA, PE machinery, and nsgRNA
(a) Schematic detailing the native, healthy wild-type plasmids. Reference sequence is the wild-type sequence,
genomic sequence and the prime editing strategy respon- green arrow points to the precise edited sequence; bold
sible for generating the PRPH2 c.828+1 G>A disease lettering indicates substitution, red boxes indicate random
model in HEK293 cells. Directional protospacer insertions. Results indicate successful installation, which
sequences, respective cut sites, and intended mutation are was then selected for to establish isolated disease model.
annotated. Image from: “SnapGene software (from Images from CRISPResso2 analysis [3]
Insightful Science; available at snapgene.com).” (b) NGS
Prime Editing Strategy to Install the PRPH2 c.828+1G>A Mutation 101

Fig. 3 Evaluation of PE2 VS PE3 systems for PRPH2 ery and pegRNA, a PE3 strategy using identical plasmids
c.828+1 G>A installation. To evaluate the long-term edit- to PE2, and an additional nsgRNA plasmid. Time course
ing efficiency for the PRPH2 c.828+1 G>A installation, results are shown below on days 3, 7, and 10 (a). Results
we performed two transfections in parallel on health and calculations are summarized in adjacent table (b)
HEK293 cells: a PE2 strategy using only the PE machin-

for all types of point mutations and has already This study forms the basis of our future work
been used to develop installation and correction to investigate the pathobiology of PRPH2-based
strategies for IRD-related mutations [1, 4, 6, 7]. RP using induced pluripotent stem cell (iPSC)-
We developed a PE3 strategy for the installa- derived retinal organoids and to develop
tion of the c.828+1 G>A in PRPH2 mutation, prime editing-based therapeutics for their
finding efficient editing of 45.96% in HEK293 amelioration.
cells (Fig. 2a, b). Further, we compared a PE2
vs PE3 strategy for the installation of the Acknowledgments SHT and The Jonas Children’s
c.828+1 G>A in PRPH2 mutation over 10 days Vision Care and Bernard & Shirlee Brown Glaucoma
Laboratory are supported by the National Institutes of
(Fig. 3a, b). We hypothesized that a PE2 strat- Health [P30EY019007, R01EY018213, R01EY024698,
egy may produce a comparable editing effi- R01EY026682, R24EY027285, U01EY030580],
ciency to a PE3 strategy if expressed long National Cancer Institute Core [5P30CA013696],
enough. However, over the three time points Foundation Fighting Blindness [TA-NMT-0116-0692-
COLU], the Research to Prevent Blindness (RPB)
analyzed, the PE3 strategy was always more Physician-Scientist Award. BLD is a recipient of the
efficient than PE2.
102 S. M. Caruso et al.

Capes PhD scholarship. PMJQ is the current recipient of a retinal dystrophies caused by mutations in the periph-
Curing Retinal Blindness Foundation (CRBF) grant, a erin/RDS gene. Prog Retin Eye Res. 2008;27:213–35.
Knights Templar Eye Foundation (KTEF) Career Starter 3. Clement K, Rees H, Canver MC, Gehrke JM, Farouni
grant, an Uplifting Athletes Young Investigator grant, and R, Hsu JY, Cole MA, Liu DR, Joung JK, Bauer DE,
a New York Stem Cell Foundation (NYSCF) – Pinello L. CRISPResso2 provides accurate and rapid
Druckenmiller Fellowship. genome editing sequence analysis. Nat Biotechnol.
2019;37:224–6.
Conflict of Interest Stephen H. Tsang receives finan- 4. Costa BLD, Levi SR, Eulau E, Tsai YT, Quinn
cial support from Abeona Therapeutics, Inc. and Emendo. PMJ. Prime editing for inherited retinal diseases.
He is also the founder of Rejuvitas and is on the scientific Front Genome Ed. 2021;3:775330.
and clinical advisory board for Nanoscope Therapeutics. 5. Manafi N, Shokri F, Achberger K, Hirayama M,
Mohammadi MH, Noorizadeh F, Hong J, Liebau S,
Tsuji T, Quinn PMJ, Mashaghi A. Organoids and
organ chips in ophthalmology. Ocul Surf. 2020;19:1–
15. Available at: http://www.ncbi.nlm.nih.gov/
References pubmed/33220469
6. Tsai YT, Costa BLD, Nolan ND, Caruso SM, Tsang
SH, Quinn PMJ. Prime editing for the installation
1. Anzalone AV, Randolph PB, Davis JR, Sousa AA,
and correction of mutations causing inherited reti-
Koblan LW, Levy JM, Chen PJ, Wilson C, Newby
nal disease: a brief methodology. Methods Mol Biol.
GA, Raguram A, Liu DR. Search-and-replace genome
2021;2560:313–31.
editing without double-strand breaks or donor
7. Tsai YT, Costa BLD, Caruso SM, Nolan ND, Levi
DNA. Nature. 2019;576:149–57.
SR, Tsang SH, Quinn PMJ. Generation of an Avian
2. Boon CJF, den Hollander AI, Hoyng CB, Cremers
Myeloblastosis Virus (AMV) reverse transcriptase
FPM, Klevering BJ, Keunen JEE. The spectrum of
prime editor. Adv Exp Med Biol. 2022;
Analysis of CRB1 Pathogenic
Variants Correctable with CRISPR
Base and Prime Editing

Bruna Lopes da Costa, Laura A. Jenny,


Irene H. Maumenee, Stephen H. Tsang,
and Peter M. J. Quinn

Abstract we carried out an analysis of the Leiden Open


Variation Database. Editable variants
The mouse and human retina contain three accounted for 54.5% for base editing and
major Crumbs homologue-1 (CRB1) iso- 99.8% for prime editing of all CRB1 patho-
forms. CRB1-A and CRB1-B have cell-type- genic variants in the Leiden Open Variation
specific expression patterns making the choice Database. The 10 most common editable
of gene augmentation strategy unclear. Gene pathogenic variants for CRB1 accounted for
editing may be a viable alternative for the 34.95% of all pathogenic variants, with the
amelioration of CRB1-associated retinal c.2843G>A, p.(Cys948Tyr) being the most
degenerations. To assess the prevalence and common editable CRB1 variant. These find-
spectrum of CRB1-associated pathogenic ings outline the next step toward developing
variants amenable to base and prime editing, base and prime editing therapeutics as an

B. L. da Costa
Department of Biomedical Engineering, Columbia
University, New York, NY, USA
Edward S. Harkness Eye Institute, Department of S. H. Tsang
Ophthalmology, Columbia University Irving Medical Department of Biomedical Engineering, Columbia
Center/New York-Presbyterian Hospital, New York, University, New York, NY, USA
NY, USA
Edward S. Harkness Eye Institute, Department of
Jonas Children’s Vision Care, and Bernard & Shirlee Ophthalmology, Columbia University Irving Medical
Brown Glaucoma Laboratory, Department of Center/New York-Presbyterian Hospital, New York,
Ophthalmology, Columbia University, New York, NY, NY, USA
USA
Jonas Children’s Vision Care, and Bernard & Shirlee
L. A. Jenny · I. H. Maumenee · P. M. J. Quinn (*) Brown Glaucoma Laboratory, Department of
Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University, New York, NY,
Ophthalmology, Columbia University Irving Medical USA
Center/New York-Presbyterian Hospital, New York,
Columbia Stem Cell Initiative, Columbia University,
NY, USA
New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee
Department of Pathology & Cell Biology, Columbia
Brown Glaucoma Laboratory, Department of
University, New York, NY, USA
Ophthalmology, Columbia University, New York, NY,
USA Institute of Human Nutrition, Columbia University,
e-mail: pq2138@cumc.columbia.edu New York, NY, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 103
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_16
104 B. L. da Costa et al.

alternative to gene augmentation for the ame- the newly identified CRB1-B, which has dis-
lioration of CRB1-associated retinal played disease-relevant functions in mice [14].
degenerations. Canonically, CRB proteins localize adjacent to
adherens junctions and are essential in maintain-
Keywords ing their stability in photoreceptors (PRC) and
Crumbs-homologue-1 (CRB1) · Prime editing Müller glial cells (MGC) [9, 11–13]. While
· Base editing · Retinitis pigmentosa (RP) · CRB1-B and CRB1-C isoforms share significant
Leber congenital amaurosis (LCA) extracellular domain overlap with CRB1-A,
CRB1-B has unique 5′ and 3′ domains, and
CRB1-C has a unique 3′ domain and is predicted
to be a secreted protein. In the mouse, CRB1-A
1 Introduction and CRB1-B predominately operate in different
cell types (MGC and PRC, respectively) [14].
Mutations in the Crumbs homologue-1 (CRB1) Recently, Fry et al. highlighted the applicabil-
gene lead to a diverse spectrum of autosomal ity of base editing for the correction of patho-
recessive retinal diseases including retinitis pig- genic mutations in large genes which cause
mentosa type 12 (RP12) and leber congenital inherited retinal dystrophies (IRDs) that are not
amaurosis type 8 (LCA8), as well as an increas- amenable to a conventional AAV-mediated gene
ing number of juvenile macular dystrophy cases augmentation strategy [6]. In particular, double-
[16, 17]. LCA8 leads to blindness from near strand break-independent clustered-regularly
birth, and RP12 leads to progressive loss of visual interspaced short palindromic repeats (CRISPR)/
field in early childhood [16]. Approximately CRISPR-associated (Cas) systems are a promis-
80,000 CRB1 patients are affected worldwide, ing avenue for the treatment of IRDs [5]. In this
with a prevalence in the United States of 1 in study, due to the evolving complexity of CRB1
86,500 [15, 16]. At present, there is no treatment retinal isoform diversity [14], we sought to
for CRB1-mediated retinal degenerations; how- ascertain the potential therapeutic potential of
ever, recent developments have identified a gene base and prime editing strategies for CRB1
augmentation approach using CRB2 that have patients.
successfully demonstrated proof of concept for
AAV-mediated gene augmentation in mice [10].
Similar studies packaging the CRB1 isoform, 2 Materials and Methods
CRB1-A, for gene augmentation in mice pro-
vides some morphological but no functional ben- CRB1 variants included in the publicly available
efits in addition to a number of adverse effects LOVD database, 3.0, were downloaded on March
potentially due to ectopic or overexpression and 7, 2021. As such, research ethics committee
immunogenic properties of human CRB1 pro- review was not required for this study. The CRB1
teins in Crb mouse models [2, 10]. Additionally, LOVD records were cleaned and coded for patho-
no rescue has been found when delivering either genicity and variational consequence. Variants
CRB1-A or CRB2 AAV-mediated gene therapy that were benign and likely benign were excluded.
to CRB1 mutant rats which have a severe early- For instances in which variant pathogenicity was
onset retinal phenotype [1]. not annotated, was annotated as a variant of
Until recently, the role of CRB1 in retinal unknown significance, or was obviously misla-
development and disease was focused on the beled (silent mutation listed as likely pathogenic),
canonical CRB1 isoform, CRB1-A. However, we reviewed the variant using the ClinVar data-
new research from the Kay lab has identified base, Poly Phen2 web tool, and literature sources,
three abundantly expressed CRB1 isoforms in and we excluded the variant from analysis if its
mouse and human retina: CRB1-A, CRB1-C, and pathogenicity could not be determined to yield a
Analysis of CRB1 Pathogenic Variants Correctable with CRISPR Base and Prime Editing 105

final data set of pathogenic and likely pathogenic prime editing (transitions, transversions, small
variants. Each variant was categorized by insertions (1–40 bp) and deletions (1–80 bp)).
expected variant consequence into termination, Editable transition and transversion mutations
missense, synonymous, intronic, splice, inser- made up 54.5% and 29.6% of alleles with patho-
tion/duplication/deletion/complex, or unknown. genic variants, respectively. The remaining
Termination was defined as a single-nucleotide 15.9% consisted of insertions, deletions, duplica-
variant (SNV) generating a stop codon. Splice tions, and complex mutations comprising dele-
was defined as a variant disrupting a canonical tion/insertions. Of these, prime editing is
splice donor (+1 and +2) or acceptor (−2 and −1) theoretically capable of correcting all but 0.2% of
site. Intronic was defined as a variant occurring pathogenic alleles, which consisted of large dele-
between the intronic +3 to −3 positions. Complex tions or duplications. This allows prime editing to
mutations comprised deletion/insertions. Data correct 99.8% and base editing 54.5% of all
were analyzed using Excel (Microsoft Corp) and pathogenic CRB1 alleles in this study. Editable
Prism, version 8 (GraphPad). Data were reported variants G>A (28.4%), C>T (12.6%), T>C
as descriptive statistics. (11.5%), and deletions (12.2%) were the most
prevalent types (Fig. 1).
The c.2843G>A, p.(Cys948Tyr) was the most
3 Results and Discussion common editable CRB1 variant accounting for
13.7% of all pathogenic alleles (Table 1). This
To determine the prevalence of alleles with edit- concurs with previous studies which also high-
able CRB1 pathogenic variants, we analyzed data lighted c.2843G>A, p.(Cys948Tyr) as the most
from the LOVD database. Of the 1335 alleles, frequent CRB1 variant [3, 7]. The 10 most com-
1259 pathogenic or likely pathogenic alleles mon CRB1 variants accounted for 34.9% of all
remained after excluding those which were pathogenic alleles (Table 1). Missense variants
benign, likely benign or variants of uncertain sig- were the predominant editable variants observed
nificance (Fig. 1). We then determined the edit- in the CRB1 gene (64.8%) followed by termina-
ability of these variants based on the limitations tion (define as SNVs that are associated with in-
of base editing (only transition mutations) and frame codons being altered to stop codons)
(16.5%) and deletion variants (12.2%) (Fig. 2).
Duplication, splice, intronic, insertion, and com-
plex variants made up the remaining 6.5%.
This study highlights that both base and prime
editing are potential alternatives to gene augmen-
tation for the treatment of CRB1-inherited retinal
diseases, particularly to circumvent the evolving
complexity of CRB1 retinal isoform diversity
[14]. Recent developments that further improve
prime editing efficiency by incorporating struc-
tured RNA motifs to the 3′ terminus of pegRNAs,
preventing degradation of the 3′ extension, or by
modifying the 3′ extension to install silent muta-
tions in addition to the target mutation at the tar-
get site that help evade mismatch repair
Fig. 1 Distribution of CRB1 pathogenic alleles from the mechanisms will only further advance progress
CRB1 Leiden open Variation Database. Analysis of 1259 [4, 8]. Lastly, this study shows that the
individual pathogenic or likely pathogenic CRB1 alleles
c.2843G>A, p.(Cys948Tyr) mutation is the most
in the Leiden Open Variation Database. The most preva-
lent mutation types include G>A (28.4%), C>T(12.6%), common editable CRB1 variant and should be the
T>C (11.5%), and deletions (12.2%) focus of future therapy development.
106 B. L. da Costa et al.

Table 1 The 10 most frequent CRB1 pathogenic variants in the Leiden Open Variation Database
cDNA change Protein change Alleles (n) Proportion of pathogenic alleles (%)
LOVD 1 c.2843G>A p.(Cys948Tyr) 172 13.66
2 c.2401A>T p.(Lys801*) 50 3.97
3 c.2234C>T p.(Thr745Met) 47 3.73
4 c.2290C>T p.(Arg764Cys) 39 3.10
5 c.2688T>A p.(Cys896*) 29 2.30
6 c.613_619del p.(Ile205Aspfs*13) 27 2.14
7 c.498_506del p.(Ile167_Gly169del) 22 1.75
8 c.3307G>A p.(Gly1103Arg) 20 1.59
9 c.4121_4130del p.(Ala1374Glufs*20) 17 1.35
10 c.1148G>A p.(Cys383Tyr) 17 1.35
Total 440 34.95

References
1. Boon N, Alves CH, Mulder AA, Andriessen CA,
Buck TM, Quinn PMJ, Vos RM, Koster AJ, Jost
CR, Wijnholds J. Defining phenotype, tropism, and
retinal gene therapy using Adeno-Associated Viral
Vectors (AAVs) in new-born brown Norway rats
with a spontaneous mutation in Crb1. Int J Mol Sci.
2021;22:1–27.
2. Buck TM, Vos RM, Alves CH, Wijnholds J. AAV-
CRB2 protects against vision loss in an inducible
CRB1 retinitis pigmentosa mouse model. Mol Ther –
Methods Clin Dev, vol. 20; 2020. p. 423.
3. Bujakowska K, Audo I, Mohand-Säid S, Lancelot
ME, Antonio A, Germain A, Eveillard TĹ, Ḿelanie
L, Saraiva JP, Lonjou C, Carpentier W, Sahel JA,
Bhattacharya SS, Zeitz C. CRB1 mutations in inher-
Fig. 2 CRB1 editable alleles by variation consequence ited retinal dystrophies. Hum Mutat. 2012;33:306–15.
from the CRB1 Leiden Open Variation Database. 4. Chen PJ, Hussmann JA, Yan J, Knipping F, Ravisankar
Missense, termination, and deletion represent the predom- P, Chen P-F, Chen C, Nelson JW, Newby GA, Sahin
inant editable CRB1 variants M, Osborn MJ, Weissman JS, Adamson B, Liu
DR. Enhanced prime editing systems by manipulat-
Acknowledgments SHT and The Jonas Children’s ing cellular determinants of editing outcomes. Cell.
Vision Care and Bernard & Shirlee Brown Glaucoma 2021;184:1–18.
Laboratory are supported by the National Institutes of 5. Costa BLD, Levi SR, Eulau E, Tsai YT, Quinn
Health [P30EY019007, R01EY018213, R01EY024698, PMJ. Prime editing for inherited retinal diseases.
R01EY026682, R24EY027285, U01EY030580], Front Genome Ed. 2021;3:775330.
National Cancer Institute Core [5P30CA013696], 6. Fry LE, McClements ME, Maclaren RE. Analysis of
Foundation Fighting Blindness [TA-NMT-0116-0692- pathogenic variants correctable with CRISPR Base
COLU], the Research to Prevent Blindness (RPB) editing among patients with recessive inherited retinal
Physician-Scientist Award. BLD is a recipient of the degeneration. JAMA Ophthalmol. 2021;139:319–28.
Capes PhD scholarship. PMJQ is the current recipient of a 7. Motta FL, Salles MV, Costa KA, Filippelli-Silva R,
Curing Retinal Blindness Foundation (CRBF) grant, a Martin RP, Sallum JMF. The correlation between
Knights Templar Eye Foundation (KTEF) Career Starter CRB1 variants and the clinical severity of Brazilian
grant, an Uplifting Athletes Young Investigator grant, and patients with different inherited retinal dystrophy phe-
a New York Stem Cell Foundation (NYSCF) – notypes. Sci Rep. 2017;7:8654.
Druckenmiller Fellowship. 8. Nelson JW, Randolph PB, Shen SP, Everette KA, Chen
PJ, Anzalone AV, An M, Newby GA, Chen JC, Hsu A,
Conflict of Interest Stephen H. Tsang receives financial Liu DR. Engineered pegRNAs improve prime editing
support from Abeona Therapeutics, Inc. and Emendo. He efficiency. Nat Biotechnol. 2021;40(3):402–10.
is also the founder of Rejuvitas and is on the scientific and 9. Pellissier LP, Alves CH, Quinn PM, Vos RM,
clinical advisory board for Nanoscope Therapeutics. Tanimoto N, Lundvig DMS, Dudok JJ, Hooibrink B,
Analysis of CRB1 Pathogenic Variants Correctable with CRISPR Base and Prime Editing 107

Richard F, Beck SC, Huber G, Sothilingam V, Garcia Koster AJ, Jost CR, Wijnholds J. Loss of CRB2 in
Garrido M, Le Bivic A, Seeliger MW, Wijnholds Müller glial cells modifies a CRB1-associated
J. Targeted ablation of CRB1 and CRB2 in retinal retinitis pigmentosa phenotype into a Leber con-
progenitor cells mimics Leber congenital amaurosis. genital amaurosis phenotype. Hum Mol Genet.
PLoS Genet. 2013;9:e1003976. 2019b;28:105–23.
10. Pellissier LP, Quinn PM, Alves CH, Vos RM, 14. Ray TA, et al. Comprehensive identification of mRNA
Klooster J, Flannery JG, Heimel JA, Wijnholds isoforms reveals the diversity of neural cell-surface
J. Gene therapy into photoreceptors and Müller glial molecules with roles in retinal development and dis-
cells restores retinal structure and function in CRB1 ease. Nat Commun. 2020;11:3328.
retinitis pigmentosa mouse models. Hum Mol Genet. 15. Stone EM, Andorf JL, Whitmore SS, DeLuca AP,
2015;24:3104–18. Giacalone JC, Streb LM, Braun TA, Mullins RF,
11. Quinn PM, Alves CH, Klooster J, Wijnholds Scheetz TE, Sheffield VC, Tucker BA. Clinically
J. CRB2 in immature photoreceptors determines the focused molecular investigation of 1000 con-
superior-inferior symmetry of the developing retina secutive families with inherited retinal disease.
to maintain retinal structure and function. Hum Mol Ophthalmology. 2017;124:1314–31.
Genet. 2018;27:3137–53. 16. Talib M, van Schooneveld MJ, van Genderen MM,
12. Quinn PM, Buck TM, Mulder AA, Ohonin C, Alves Wijnholds J, Florijn RJ, ten Brink JB, Schalij-
CH, Vos RM, Bialecka M, van Herwaarden T, van Delfos NE, Dagnelie G, Cremers FPM, Wolterbeek
Dijk EHC, Talib M, Freund C, Mikkers HMM, R, Fiocco M, Thiadens AA, Hoyng CB, Klaver CC,
Hoeben RC, Goumans M-J, Boon CJF, Koster AJ, Bergen AA, Boon CJF. Genotypic and phenotypic
Chuva de Sousa Lopes SM, Jost CR, Wijnholds characteristics of CRB1-associated retinal dystro-
J. Human iPSC-derived retinas recapitulate the fetal phies: a long-term follow-up study. Ophthalmology.
CRB1 CRB2 complex formation and demonstrate that 2017;124:884–95.
photoreceptors and Müller Glia are targets of AAV5. 17. Tsang SH, Burke T, Oll M, Yzer S, Lee W, Xie YA,
Stem Cell Rep. 2019a;12:906–19. Allikmets R. Whole exome sequencing identifies
13. Quinn PM, Mulder AA, Henrique Alves C, CRB1 defect in an unusual maculopathy phenotype.
Desrosiers M, de Vries SI, Klooster J, Dalkara D, Ophthalmology. 2014;121:1773–82.
Generation of an Avian
Myeloblastosis Virus (AMV)
Reverse Transcriptase Prime Editor

Yi-Ting Tsai, Bruna Lopes da Costa,


Salvatore Marco Caruso, Nicolas D. Nolan,
Sarah R. Levi, Stephen H. Tsang,
and Peter M. J. Quinn

Abstract nology that represents an exciting avenue for


the treatment of inherited retinal diseases
Prime editing (PE) is a novel, double-strand (IRDs). Given the extensive and heterogenous
break (DSB)-independent gene editing tech- nature of the 280 genes associated with IRDs,

Authors Bruna Lopes da Costa, Salvatore Caruso and


Nicolas D Nolan have equally contributed to this chapter.

Y.-T. Tsai S. R. Levi · P. M. J. Quinn (*)


Department of Biomedical Engineering, Columbia Edward S. Harkness Eye Institute, Department of
University, New York, NY, USA Ophthalmology, Columbia University Irving Medical
Center/New York-Presbyterian Hospital,
B. L. da Costa
New York, NY, USA
Department of Biomedical Engineering, Columbia
University, New York, NY, USA Jonas Children’s Vision Care, and Bernard & Shirlee
Brown Glaucoma Laboratory, Department of
Edward S. Harkness Eye Institute, Department of
Ophthalmology, Columbia University,
Ophthalmology, Columbia University Irving Medical
New York, NY, USA
Center/New York-Presbyterian Hospital,
e-mail: pq2138@cumc.columbia.edu
New York, NY, USA
S. H. Tsang
Jonas Children’s Vision Care, and Bernard & Shirlee
Department of Biomedical Engineering, Columbia
Brown Glaucoma Laboratory, Department of
University, New York, NY, USA
Ophthalmology, Columbia University,
New York, NY, USA Edward S. Harkness Eye Institute, Department of
Ophthalmology, Columbia University Irving Medical
S. M. Caruso · N. D. Nolan
Center/New York-Presbyterian Hospital,
Department of Biomedical Engineering, Columbia
New York, NY, USA
University, New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee
Edward S. Harkness Eye Institute, Department of
Brown Glaucoma Laboratory, Department of
Ophthalmology, Columbia University Irving Medical
Ophthalmology, Columbia University,
Center/New York-Presbyterian Hospital,
New York, NY, USA
New York, NY, USA
Columbia Stem Cell Initiative, Columbia University,
Jonas Children’s Vision Care, and Bernard & Shirlee
New York, NY, USA
Brown Glaucoma Laboratory, Department of
Ophthalmology, Columbia University, Department of Pathology & Cell Biology, Columbia
New York, NY, USA University, New York, NY, USA
Columbia Stem Cell Initiative, Columbia University, Institute of Human Nutrition, Columbia University,
New York, NY, USA New York, NY, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 109
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_17
110 Y.-T. Tsai et al.

genome editing has presented countless com- genes Cas9 (SpCas9) nickase H840A mutant and
plications. However, recent advances in an optimized Moloney murine leukemia virus
genome editing technologies have identified (MMLV) reverse-transcriptase (RT), in conjuga-
PE to have tremendous potential, with the tion with a PE guide RNA (pegRNA). The
capability to ameliorate small deletions and pegRNA is comprised of a spacer, scaffold,
insertions in addition to all twelve possible primer binding sequence (PBS) and a RT tem-
transition and transversion mutations. The plate (RTT) which includes the chosen edit [1].
current PE system is based on the fusion of the The flexibility, specificity, and efficacy of PE
Streptococcus pyogenes Cas9 (SpCas9) nick- makes it an exciting tool for the treatment of the
ase H840A mutant and an optimized Moloney 280 gene mutations associated with inherited
murine leukemia virus (MMLV) reverse-­ retinal diseases (IRDs) [5, 8].
transcriptase (RT) in conjunction with a PE To date, no other RT has been used to facilitate
guide RNA (pegRNA). In this study, we devel- PE in mammalian cells besides the
oped a prime editor based on the avian myelo- MMLV-RT. The avian myeloblastosis virus
blastosis virus (AMV)-RT and showed its (AMV)-RT is a heterodimer consisting of a
applicability for the installation of the PRPH2 63-kDa α-subunit and 95-kDa β-subunit unlike
c.828+1G>A mutation in HEK293 cells. MMLV-RT, which is a 75 kDa monomer. The α-
subunit and β-subunit both comprise of poly-
Keywords merase and RNase H domains, but the β-subunit
additionally contains a C-terminal integrase
Prime editing · Avian myeloblastosis virus domain. However, MMLV-RT only contains the
(AMV) · PRPH2 · Retinitis pigmentosa (RP) same domains as the α-subunit of the
· AMV-RT prime editors AMV-RT. Additionally, AMV-RT retains more
DNA synthetic activity at elevated temperatures,
possess a higher intrinsic RNase H activity, and is
1 Introduction more processive than MMLV-RT [2, 6, 9].
In this study, we fused the SpCas9 nickase
Prime editing (PE), a double-strand break (DSB)- H840A to either the α- or β-subunit and expressed
independent, “search-and-replace” gene editing the complementary subunit in tandem using a
approach, has the unique capacity to install all P2A linker, forming Cas9(H840A)–AMV-RT(α-­
types of transition and transversion mutations in P2A-­β) or Cas9(H840A)–AMV-RT(β-P2A-α)
addition to amelioration of small insertions and prime editors, respectively (Fig. 1). Similar edit-
deletions (indels) [1]. The process of PE requires ing efficiencies were found in HEK293 cells
a prime editor, a fusion of the Streptococcus pyo- using both editors for the +1 CTT insertion at

Fig. 1 Schematic of AMV-RT prime editors. The linker, and we also expressed the complementary subunit
AMV-RT is a heterodimer consisting of a α- and β- sub- in tandem using a P2A linker. We compare our version 1
units, unlike the MMLV-RT, which is a monomer. In this AMV-RT-based prime editors Cas9(H840A)–AMV-­
study, we fused the SpCas9 nickase H840A to either the RT(α-P2A-β) and Cas9(H840A)–AMV-RT(β-P2A-α) to
α- or β-subunit of the wild-type AMV-RT using a flexible the current state-of-the-art Cas9(H840A)-MMLV-RT
Generation of an Avian Myeloblastosis Virus (AMV) Reverse Transcriptase Prime Editor 111

HEK3 and for the installation the PRPH2 plete medium. For the transfection of PE3, the
c.828+1G>A mutation. Together, our results plasmid constitution is 750ng:250ng:83ng of
highlight the applicability of AMV-RT-based C M V - P E - V 4 : U 6 - p e g R N A : U 6 -­
prime editors, setting a platform for improving ngRNA. Lipofectamine 2000 (Thermo Fisher)
their efficiency by incorporating mutations that was mixed at 1:1 ratio to the plasmid DNA. The
may increase their thermostability, processivity, cells were then collected after 72 h post transfec-
and DNA-RNA substrate affinity. tion for DNA extraction and analysis.

2 Materials and Methods 2.3 DNA Extraction

2.1 Cloning and Plasmid To extract DNA from cells, the cells were
Constructs detached from the wells by trypsin. The cells
from each well were washed with DPBS (without
The original optimized Cas9 nickase-reverse Ca2+ and Mg2+) and resuspended with 50 μl
transcriptase plasmid (pCMV-PE2, Addgene DPBS. The cells were then incubated at 95 °C for
#132775) was modified to create pCMV-PE(V4) 20 mins. Subsequently, after cooling, 4 μl of
using In-Fusion (Takara Bio) cloning by adding 20 mg/ml proteinase K (Promega) was added to
SwaI and SmaI restriction enzyme sites, eGFP each sample. The samples were then incubated at
driven by PGK promoter, and a modified GSSG 56 °C for 1 h followed by a 30-min incubation at
flexible linker sequence between the MMLV-RT 95 °C to stop the proteinase K digestion. This
and the Cas9(H840A) nickase. Subsequently, we crude extract of DNA is then ready for poly-
ordered by gBlock Gene Fragments (Integrated merase chain reaction (PCR) purpose.
DNA Technologies (IDT)) the Alpha and Beta
domains of the AMV-RT and used In-Fusion
cloning (Takara Bio) to replace the MMLV-RT 2.4 Analysis of Prime Editing
from pCMV-PE(V4) to create CMVp-­ Efficiency
Cas9(H840A)–AMV-RT(α-P2A-β) and CMVp-­
Cas9(H840A)–AMV-RT(β-P2A-α) version 1 For the determination of editing efficiency, the
prime editors. We used the previously published PRPH2 c.828+1 locus (Forward Primer, 5′-cac-
pU6-Sp-pegRNA-HEK3_CTT_ins (Addgene, # cagacggaggagctca-­3′; Reverse Primer,
132778) [1], with 5′- ggcccagactgagcacgtga-3′ as 5′-gagggaggcatgctctcca-3′) or +1 CTT insertion
the spacer and 5′-tctgccatcaaagcgtgctcagtctg-3′ at HEK3 locus (Forward Primer, 5′-atgtgggct-
as the 3′-extension sequence) and the pU6- gcctagaaagg-­3′; Reverse Primer, 5′-gcccagc-
Sp-­pegRNA-PRPH2-828+1G>A-sub [3]. caaacttgtcaacc-­3′) were amplified using primers
Corresponding nsgRNA (e.g., for HEK3, with Illumina adaptors. The amplicon was sub-
5′-gtcaaccagtatcccggtgc-3′ as the spacer) were mitted to Genewiz for the Amplicon EZ service.
cloned into the pU6-spacer-acceptor, using BsaI The analysis of the sequencing data was deter-
Golden Gate assembly (NEB), as previously mined using CRISPResso2 https://crispresso.
described [1, 3, 8]. pinellolab.partners.org/submission. The reads of
the amplicons less than 1% of the total frequency
were excluded for visualization.
2.2 Cell Culture and Transfection

To test editing efficiency of the PE system, the 3 Results


plasmids were transfected into HEK293 cells.
The HEK293 cells were seeded 1 day before at In this study, we fused the SpCas9 nickase
50,000 cells/well in 24-well plates. On the next H840A to either the α- or β-subunit of the wild-­
day, the medium was refreshed with 500 μl com- type AMV-RT using a flexible linker and
112 Y.-T. Tsai et al.

expressed the complementary subunit in tandem based prime editors, we again compared them to
using a P2A linker. We first compared our version the MMLV-RT-based prime editor. PE efficiency
1 wild-type AMV-RT-based prime editors was highest using the MMLV-RT-based prime
Cas9(H840A)–AMV-RT(α-P2A-β) and editor, 43.66%, compared to 2.02% and 1.62%
Cas9(H840A)–AMV-RT(β-P2A-α) to the current for the version 1 wild-type AMV-RT(α-P2A-β)
state of the art Cas9(H840A)-MMLV-RT for and AMV-RT(β-P2A-α) prime editors, respec-
incorporating the +1 CTT insertion at the HEK3 tively (Fig. 3). Together, this data demonstrates
locus in HEK293 cells (Fig. 2). All experiments the potential of using an alternative reverse tran-
were done using a PE3 methodology, whereby an scriptase for PE and sets a platform for improv-
additional nicking single-guide RNA (nsgRNA) ing AMV-RT-based prime editors.
is incorporated to nick the non-edited strand,
directing DNA repair to use the edited strand as a
template [1]. PE efficiency was highest using the 4 Discussion
MMLV-RT-based prime editor, 37.15%, com-
pared to 8.03% and 3.66% for the version 1 wild-­ In this study, we showed that both of our version
type AMV-RT(α-P2A-β) and AMV-RT(β-P2A-α) 1 wild-type AMV-RT-based prime editors could
prime editors, respectively. We recently also successfully introduce the specified edits but
designed a PE strategy for the installation of the with limited efficiency compared to the engi-
PRPH2 c.828+1G>A mutation [3]. To further neered MMLV-RT-based prime editor 2 (PE2)
test the applicability of our version 1 AMV-RT-­ [1]. Previously, with PE1, Anzalone et al. used

Fig. 2 Installation of +1 CTT insertion at HEK3 using an editors Cas9(H840A)–AMV-RT(α-P2A-β) (b) and
AMV-RT prime editor. Next-generation sequencing CMVp-Cas9(H840A)–AMV-RT(β-P2A-α) (c). Green
results comparing the +1 CTT insertion at HEK3 locus in arrows point to the precise edited sequence, and the red
bulk HEK293 cells with the optimized Cas9(H840A)- box indicates CTT insertion. (Images from CRISPResso2
MMLV-RT (a) against the version 1 AMV-based prime analysis [4])
Generation of an Avian Myeloblastosis Virus (AMV) Reverse Transcriptase Prime Editor 113

Fig. 3 Installation of PRPH2 c.828+1G>A mutations RT(α-P2A-β) (b)


using an AMV-RT prime editor. Next-generation sequenc- and CMVp-Cas9(H840A)–AMV-RT(β-P2A-α) (c). Green
ing results comparing the installation of the PRPH2 arrows point to the precise edited sequence; bold lettering
c.828+1G>A mutation in bulk HEK293 cells with the indicates substitution. (Images from CRISPResso2 analy-
optimized Cas9(H840A)-MMLV-RT (a) against the ver- sis [4])
sion 1 AMV-based prime editors Cas9(H840A)–AMV-­

the wild-type MMLV-RT but, subsequently, AMV-RT-based prime editors to PE1 may be
after proof-of-concept, engineered the wild- due to the higher RNase H activity exhibited by
type MMLV-RT to make PE2 by introducing AMV-RT than MMLV-RT [2]. Future studies,
mutations that would increase the thermostabil- which directly compare the MMLV-RT-
ity, processivity, and DNA-RNA substrate affin- engineered PE2 alongside an engineered
ity of the RT. This led to substantial AMV-RT based prime editor, are required to see
improvements in prime editing efficiency at the the true potential of AMV-RT-based prime edi-
five genomic sites analyzed. For the +1 CTT tors. Similarly to MMLV-RT, mutations for
insertion at the HEK3 locus, this caused an AMV-RT have been identified to increase its
improvement from approximately 16% with thermostability and to inactivate its RNase H
PE1 to 28% with PE2 in Hek293 cells. All the activity [6, 7]. Further, due to the intrinsic dif-
experiments we carried out in this study were ferences between MMLV-RT and AMV-RT,
done using a prime editing 3 methodology, prime editors, based on the latter, may have dif-
where an additional nsgRNA is incorporated to ferent constraints to be optimized and explored
nick the non-edited strand, directing DNA repair (i.e., the editing window possible for indels may
to use the edited strand as a template [1]. For the be different).
+1 CTT insertion at the HEK3 locus using PE3,
Anzalone et al. could get upwards of approxi- Acknowledgments SHT and the Jonas Children’s Vision
mately 70% editing efficiency; we got a more Care and Bernard & Shirlee Brown Glaucoma Laboratory
are supported by the National Institutes of Health
modest 37.15% under the conditions we used [P30EY019007, R01EY018213, R01EY024698,
with our slightly modified PE2 vector. By con- R01EY026682, R24EY027285, U01EY030580],
trast, we got editing efficiencies of 8.03% and National Cancer Institute Core [5P30CA013696],
3.66% for the version 1 wild-type AMV-RT(α- Foundation Fighting Blindness [TA-NMT-0116-0692-
COLU], the Research to Prevent Blindness (RPB)
P2A-β) and AMV-RT(β-­P2A-­α) prime editors, Physician-Scientist Award. BLD is a recipient of the
respectively. Although no clear and direct com- Capes PhD scholarship. PMJQ is the current recipient of a
parison was carried out, the presumed reduced Curing Retinal Blindness Foundation (CRBF) grant, a
editing efficiency from our version 1 wild-type Knights Templar Eye Foundation (KTEF) Career Starter
114 Y.-T. Tsai et al.

grant, an Uplifting Athletes Young Investigator grant, and genome editing sequence analysis. Nat Biotechnol.
a New York Stem Cell Foundation (NYSCF) – 2019;37:224–6.
Druckenmiller Fellowship. 5. Costa BLD, Levi SR, Eulau E, Tsai YT, Quinn
PMJ. Prime editing for inherited retinal diseases.
Front Genome Ed. 2021;3:775330.
6. Gerard GF, Potter RJ, Smith MD, Rosenthal K,
References Dhariwal G, Lee J, Chatterjee DK. The role of
template-primer in protection of reverse transcrip-
1. Anzalone AV, Randolph PB, Davis JR, Sousa AA, tase from thermal inactivation. Nucleic Acids Res.
Koblan LW, Levy JM, Chen PJ, Wilson C, Newby 2002;30:3118–29.
GA, Raguram A, Liu DR. Search-and-replace genome 7. Konishi A, Yasukawa K, Inouye K. Improving the
editing without double-strand breaks or donor thermal stability of avian myeloblastosis virus reverse
DNA. Nature. 2019;576:149–57. transcriptase α-subunit by site-directed mutagenesis.
2. Bustin SA. Absolute quantification of mrna using real-­ Biotechnol Lett. 2012;34:1209–15.
time reverse transcription polymerase chain reaction 8. Tsai YT, Costa BLD, Nolan ND, Caruso SM, Tsang
assays. J Mol Endocrinol. 2000;25:169–93. SH, Quinn PMJ. Prime editing for the installation
3. Caruso SM, Tsai YT, Costa BLD, Tsang SH, Quinn and correction of mutations causing inherited reti-
PMJ. Prime editing strategy to knock-in the PRPH2 nal disease: a brief methodology. Methods Mol Biol.
c.828+1G>A mutation. Adv Exp Med Biol. 2022; 2022;2560:313–31.
4. Clement K, Rees H, Canver MC, Gehrke JM, Farouni 9. Yasukawa K, Nemoto D, Inouye K. Comparison of the
R, Hsu JY, Cole MA, Liu DR, Joung JK, Bauer DE, thermal stabilities of reverse transcriptases from avian
Pinello L. CRISPResso2 provides accurate and rapid myeloblastosis virus and moloney murine leukaemia
virus. J Biochem. 2008;143:261–8.
Part IV
Gene Therapy
Preexisting Neutralizing
Antibodies against Different
Adeno-Associated Virus Serotypes
in Humans and Large Animal
Models for Gene Therapy

Divya Ail and Deniz Dalkara

Abstract Keywords

Adeno-associated virus · AAV serotypes ·


Gene therapy is a potential cure for several
Neutralizing antibodies · Large animal
inherited retinal dystrophies, and adeno-­
models · Retinal gene therapy · Anti-AAV
associated virus (AAV) has emerged as a
immune response
vector of choice for therapeutic gene deliv-
ery to the retina. However, prior exposure to
AAVs can cause a humoral immune response
resulting in the presence of antibodies in the 1 Introduction
serum, which can subsequently interfere
with the AAV-mediated gene therapy. The Adeno-associated virus (AAV) has emerged as a
antibodies bind specifically to a serotype but vector of choice for gene therapy due to their
often display broad cross-reactivity. A sub- small size, good transduction profiles across dif-
set of these antibodies called neutralizing ferent tissue types, and relatively low immunoge-
antibodies (NABs) can render the AAV inac- nicity [5]. AAV is a small non-enveloped virus
tive, thereby reducing the efficacy of the that contains a single-stranded DNA and depends
therapy. The preexisting NAB levels against on adenovirus for its replication. It is 25 nm in
different serotypes vary by species, and size and has a packaging capacity of approxi-
these variations need to be considered while mately 4.5 kb. Minor differences in the capsid
designing studies. Since large animals often structure determine the cellular receptors it can
serve as preclinical models to test gene ther- bind and determine its cell and tissue specificity.
apies, in this review we compile studies These different AAV variants are considered
reporting preexisting NABs against com- serotypes when distinguished by a common set
monly used AAV serotypes in humans and of antigens [31].
large animal models and discuss strategies to There are several hundred known naturally
deal with NABs. occurring variants, and millions of laboratory
generated AAVs that were screened to perform
specific biological tasks [15]. Of the naturally
occurring and most used serotypes, AAV1–AAV9
D. Ail (*) · D. Dalkara are well characterized. AAV1, AAV2, AAV3,
Sorbonne Université, INSERM, CNRS, Institut de la AAV4, and AAV6 were first isolated as contami-
Vision, Paris, France nants in adenovirus preparations. AAV5 was iso-
e-mail: divya.ail@inserm.fr

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 117
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_18
118 D. Ail and D. Dalkara

Exposure
to AAV Binding Enhanced immune
antibodies response/
BABs Inflammation

Total anti-AAV
antibodies Neutralizing
antibodies
People exposed to AAV NABs Virus neutralization/
develop anti-AAV antibodies Reduction in efficacy

Fig. 1 Schematic representation of the production of serotype-specific anti-AAV binding antibodies (BABs) and neu-
tralizing antibodies (NABs) in human population that is exposed to AAVs

lated from human condylomatous wart. A immune responses, humoral immune response
high-throughput functional screen of 474 nonhu- produces antibodies against surface antigens of
man primates and 259 human tissues resulted in AAVs. Of the total repertoire of antibodies pro-
the identification of the variants AAV7, AAV8, duced in the hosts, all antibodies that specifically
and AAV9 [11]. bind to AAVs are known as binding antibodies
Most research to develop, optimize, and test (BABs), and a subclass of these binding antibod-
AAV-mediated gene therapies is conducted in ies that not only bind but also neutralize AAV
rodents. However, rodent models have certain activity are known as neutralizing antibodies
drawbacks such as the differences in the structure (NABs). Hence, exposure to AAVs results in the
of the retina compared to humans. The rodent development of anti-AAV antibodies (BABs and
vision is rod dominated; they lack a macula and NABs) that are serotype specific (Fig. 1). When
fovea and hence do not recapitulate all the essen- hosts that possess preexisting antibodies due to
tial features of most diseases or give complete prior exposure receive AAV-based gene thera-
information of the efficacy of therapy in the pies, they can have a stronger immune response,
human context. Hence, large animal models are resulting in the elimination of the AAV contain-
preferred for preclinical studies. Among the large ing the therapy, thereby reducing the efficacy of
animals, the nonhuman primate retinas are the the treatment or increasing the risk of developing
closest to human retinas as they rely on high acu- ocular inflammation [5].
ity cone vision and have a macula and fovea. On
the other hand, animals such as pigs, dogs, cats,
sheep, and even horses have been used in some 2 Preexistence of Antibodies
studies [30]. Most of these animals have a cone-­ Against Different Serotypes
rich retina as they are diurnal animals like in Humans
humans. They do not possess a macula and fovea,
but some of these like the dogs and pigs have a Several studies have reported the prevalence of
cone-enriched region in the retina known as the preexisting antibodies to WT AAV [4, 7, 8]. An
visual streak [14]. analysis of the total anti-AAV IgG antibodies
AAVs are preferred for their low immunoge- revealed that prevalence of anti-AAV1 (67% of
nicity and excellent safety profile, especially for the population tested possessed anti-AAV1 anti-
use in the retina as it is believed to be an immune-­ bodies) and anti-AAV2 (72%) was higher than
privileged tissue. However, several studies have those of anti-AAV5 (40%), anti-AAV6 (46%),
shown that AAVs can elicit both innate and adap- anti-AAV8 (38%), and anti-AAV9 (47%) [4].
tive immune responses. As part of adaptive Another study reported the prevalence of neutral-
Preexisting Neutralizing Antibodies against Different Adeno-Associated Virus Serotypes in Humans… 119

izing antibodies in serum samples collected from macaques, 20 marmosets, and 20 chimpanzees.
10 countries and 4 continents. NABs against All marmosets were seropositive for AAV9, and
AAV2 were the highest (30–60% of the popula- 60% of the chimpanzees were seropositive for
tion), whereas anti-AAV7, AAV8, and AAV9 had anti-AAV1 NABs. AAV8 and AAV2 NABs were
lower prevalence (15–30%). Anti-AAV1 NABs most prevalent in the rhesus macaque cohort with
prevalence was lower than that of anti-AAV2 but close to 100% of the samples being seropositive,
higher than AAV7, AAV8, and AAV9 in most whereas the seroprevalence was approximately
regions [7]. There is an interest in studying 45% for AAV1 and AAV5. Prevalence levels of
cohorts of patients with specific diseases to check NABs against AAV1 were similar in the cyno-
the prevalence of anti-AAV antibodies and check molgus macaques (43%), whereas they were
if they can benefit from gene therapies. A study higher for AAV8 (75%) and AAV9 (68%) [28].
involving 1552 patients with heart condition We recently reported the BABs against AAV8
showed close to 60% of the cohort were seroposi- and AAV9 were significantly higher than AAV5
tive for NABs against AAV1 [3]. Interestingly, a BABs in a cohort of 41 cynomolgus macaques
study comparing the preexisting antibodies [2]. A 10-year study on a cohort of 25 chimpan-
against AAV2, AAV5, and AAV6 in patients with zees tested for NABs against AAV8 reported
cystic fibrosis against a control population three distinct groups – a group that remained
revealed that the Nab levels were lower in the naïve for NABs, another one that had chronically
patients for each of the serotype compared to the high levels throughout the study period, and a
control population, with the lowest levels being group that seroconverted from high to low levels
for the NABs against AAV5 [12]. Another study over the span of 10 years [9]. A study involving
involving HIV1-infected patients in China 23 rhesus macaques showed that 70% of the sam-
showed no difference in the seroprevalence com- ples were seropositive for NABs against AAV8
pared to healthy controls, and that prevalence [24]. Overall, in the diverse nonhuman primate
was higher for AAV2 (96.6%) and AAV8 (82.0%) studies, antibodies against AAV8 and AAV9
than for AAV5 (40.2%) [29]. Another study in appear more predominantly, whereas anti-AAV1
Japanese population comparing hemophilia and AAV5 antibodies are the least prevalent.
patients with healthy population revealed a prev- In a study involving 70 dogs, all were positive
alence of NABs in approximately 30–40% of the for NABs against AAV1 and AAV6 with a higher
population for all the serotypes tested (AAV1, prevalence of AAV1, and they were negative for
AAV2, AAV5, AAV8, and AAV9), and the levels all other serotypes tested (AAV2, AAV5, and
did not differ significantly between the patients AAV8), irrespective of the colony or genetic
and healthy controls [20]. All these human stud- background of the animal [6]. Another study also
ies reveal that antibodies against the common confirmed prevalence of high titers of NABs
AAV serotypes are prevalent in the human against AAV6 in their cohort of dog samples,
population, and the geographic location most
­ whereas there were NABs present albeit at low
likely determines the possibility of exposure and titers against AAV1 and AAV2, but no NABs
development of the antibodies. Interestingly, against AAV9 were detected [22]. Contrary to
other disease conditions do not necessarily have this, a study involving 16 dogs reported all of
an impact on prevalence of NABs. them seropositive for AAV9, but these were not
tested for AAV6 [28]. A study in dogs tested 72
naïve dogs for NABs against AAV6, AAV8, and
3 Preexistence of Antibodies AAV9 and found that the NABs against AAV6
Against Different Serotypes were the highest and most prevalent irrespective
in Large Animal Models of the age, gender, and disease status. The other
two serotypes tested were below detection limit
The most comprehensive reporting of NAB in most animals tested, except a few young pups
­prevalence in nonhuman primates was conducted that had low titers of NABs against AAV8 or
on 1632 rhesus macaques, 790 cynomolgus AAV9 [23].
120 D. Ail and D. Dalkara

In pigs, AAV5 was the most prevalent with all they had very low antibodies or were seronega-
the animals tested being seropositive, but unlike tive for all other tested serotypes [6].
other large animals pigs were also seropositive Rodents are often used for gene therapy stud-
for all other tested serotypes (AAV1, AAV2, ies but rarely tested for anti-AAV antibodies. One
AAV6, and AAV8), with the lowest prevalence study revealed that naïve mice acquired from
being against AAV6 [6]. Another study tested commercial vendors harbored anti-AAV antibod-
NABs against AAV1, AAV2, AAV6, and AAV9 ies, with highest prevalence against AAV1 and
and showed the presence of antibodies against all AAV6 and low prevalence against AAV2 and
tested serotypes in the pig samples at high titers AAV9. The rats tested in this study had the lowest
[22]. NABs among all the animal species tested. They
In a study conducted on 230 client-owned and possessed some NABs against AAV6 and very
20 specific pathogen-free (SPF) cats in low NAB titers against AAV2 and were seronega-
Switzerland that were tested for NABs, it was tive for AAV1 and AAV9 [22].
revealed that 53% had NABs against at least one
of the serotypes tested. The lowest prevalence
was against AAV6 (5%), followed by AAV1 and 4 Strategies to Deal
AAV5 (7% each), AAV2 (17%), AAV9 (20%), with the Anti-AAV
and AAV8 (26%), and the highest prevalence and Antibodies
the highest titers were observed against AAV7
(28%) [17]. In another study conducted with 85 Most preclinical and clinical studies use some
cats living in the Northeastern United States (35 form of immunosuppression, but direct evidence
client-owned, 20 feral, and 30 SPF cats), BABs of the efficacy of such treatments is rarely
tested against 11 AAV serotypes (AAV1–AAV11) explored. Also, these treatments vary consider-
showed the presence of antibodies against the ably from one study to another. In fact, a study
different serotypes to varying extents, including points toward the adverse effect of using an
the SPF animals. However, upon testing NABs immunosuppressive regimen [27]. Hence,
against AAV2, AAV6, and AAV9, no significant researchers are actively exploring other methods
neutralizing activity was detected, indicating that to evade the AAV-directed immune responses,
even though the animals had anti-AAV antibod- some of which are as follows:
ies, the levels of NABs were low [1].
A small study with 3 sheep serum samples Designing Novel AAV Variants A strategy using
showed low titers against AAV2 and AAV6, while structural information acquired from cryo-­
the serums were seronegative for AAV1 and electron microscopy of antigen-binding domains
AAV9 [22]. On the other hand, a study involving on AAVs and generating AAV variants by mutat-
a cohort of 41 sheep reported all of them sero- ing these domains resulted in a neutralization
positive for AAV9, which was the only serotype evading variant [26]. One strategy to avoid recog-
tested [28]. Another study involving 6 animals nition and binding by preexisting antibodies is to
showed varying levels of BABs against all sero- generate novel AAV variants. This can be done
types tested (AAV1, AAV2, AAV5, AAV6, AAV8, by using directed evolution of the capsids and
and AAV9), and some of the animals tested screening them by their ability to evade neutral-
showed NABs against AAV2 and at lower titers ization by antibodies. First, a library of sequences
against AAV8 [25]. is generated by making changes to the coding
Horse is not a model commonly used for eye sequence of the AAV capsid using error-prone
diseases but serves as a good preclinical model PCR, and then this is tested and screened for anti-
for osteoarthritis, and it is interesting to note that body resistance. One such approach resulted in
a study testing NABs against different AAV sero- AAV vectors with an approximately 10- and 100-­
types found that all the horses in the study were fold resistance to neutralizing antibodies [19].
seropositive for NABs against AAV5, whereas Capsid shuffling is another technique used to
Preexisting Neutralizing Antibodies against Different Adeno-Associated Virus Serotypes in Humans… 121

generate AAV variant libraries, wherein the cap- showed that this effect was abolished in Tlr9 KO
sid sequences of known AAVs are fragmented mice. Further, they depleted CpG sequences in
and reassembled to generate variant sequences, the viral genome and showed that these modified
which are then tested and screened for antibody AAVs were evading immune responses [10].
resistance [18].

Tlr9 Inhibition Immune cells display a family


AAV Capsid Decoys In this strategy, empty cap- of surface receptors called TLRs, and TLR9 nor-
sids are injected along with the therapeutic gene mally senses DNA from pathogenic viruses that
carrying AAVs so that the empty capsids bind the contain unmethylated CpG motifs. CpG binding
NABs. Further, the empty capsids have their to TLR9 promotes its dimerization and activates
receptor binding sites mutated, so they cannot TLR9 signaling. This results in innate immune
enter the cell and hence only serve to bind anti- responses eventually recruiting other immune
bodies. This approach was shown to successfully cells to the site of infection and primes adaptive
overcome the inhibitory effect of NABs and immune responses [13]. A study showed that
increase gene transfer efficacy in both mice and short TLR9 inhibitory sequences (approximately
nonhuman primates [21]. However, to overcome 12–24 nucleotides) could be incorporated into
high titers of NABs, high doses of empty capsids the viral genome for blocking TLR9 activation.
are required that will significantly increase the The TLRi containing vectors elicited reduced
total viral load used, which is likely to aggravate immune responses and enhanced gene expression
the immune response in an AAV dose-dependent in mouse and pig models [10].
manner [2]. This strategy is thus questionable in
the context of retinal gene therapy. Taking into account all these animal studies, it
is evident that the levels of antibodies against dif-
ferent AAV serotypes show interspecies differ-
Endopeptidase Digestion A recent study dem- ences, which need to be considered before
onstrated that circulating IgGs could be elimi- selecting a suitable serotype for each animal
nated by the enzymatic degradation using an model. Furthermore, the host can be pretested for
endopeptidase called imlifidase (IdeS). In this antibodies against the serotype to be used, and
study, mice were passively immunized with strategies can be put in place to evade immune
serum IgGs, followed by IdeS treatment and responses and ensure efficient gene transfer.
AAV injection. They reported decreased anti-­
AAV antibodies and more efficient gene transfer.
They were able to show that IdeS administration References
was safe and efficient in nonhuman primates.
Further, in vitro testing of human plasma samples 1. Adachi K, Dissen GA, Lomniczi A, Xie Q, Ojeda SR,
collected from gene therapy trial patients resulted Nakai H. Adeno-associated virus-binding antibodies
detected in cats living in the Northeastern United States
in reduced anti-AAV antibody levels [16]. lack neutralizing activity. Sci Rep. 2020;10(1):1–13.
https://doi.org/10.1038/s41598-­020-­66596-­4.
2. Ail D, Ren D, Brazhnikova E, Nouvel-Jaillard C,
Cytosine-Phosphate-Guanine (CpG) Bertin S, Mirashrafi SB, Fisson S, Dalkara D. Systemic
and local immune responses to intraocular AAV vec-
Depletion Many unmethylated CpG motifs tor administration in non-human primates. Mol Ther
present in clusters known as CpG islands are Methods Clin Dev. 2022;24(March):306–16. https://
common in AAV vector sequences. These CpG doi.org/10.1016/j.omtm.2022.01.011.
motifs interact with Tlr9 receptors and contribute 3. Greenberg B, Butler J, Felker GM, Felker GM,
Ponikowsk P, Zsebo KM. Prevalence of AAV1 neu-
toward eliciting an immune response. In a study, tralizing antibodies and consequences for a clinical
the authors used an immunogenic variant of AAV trial of gene transfer for advanced heart failure. Gene
to induce an immune response in WT mice and Ther. 2016;23:313–9.
122 D. Ail and D. Dalkara

4. Boutin S, Monteilhet V, Veron P, Leborgne C, presence of anti-AAV neutralizing antibodies. Nat


Benveniste O, Montus MF, Masurier C. Prevalence Med. 2020;26(7):1096–101. https://doi.org/10.1038/
of serum IgG and neutralizing factors against adeno-­ s41591-­020-­0911-­7.
associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in 17. Li P, Boenzli E, Hofmann-Lehmann R, Katrin Helfer-­
the healthy population: implications for gene therapy Hungerbuehler A. Pre-existing antibodies to candidate
using AAV vectors. Hum Gene Ther. 2010;21(6):704– gene therapy vectors (adeno-associated vector sero-
12. https://doi.org/10.1089/hum.2009.182. types) in domestic cats. PLoS One. 2019;14(3):1–19.
5. Bucher K, Rodríguez-Bocanegra E, Dauletbekov D, https://doi.org/10.1371/journal.pone.0212811.
Dominik Fischer M. Immune responses to retinal 18. Li W, Asokan A, Zhijian W, Van Dyke T, DiPrimio N,
gene therapy using adeno-associated viral vectors – Jarrod JS, Govindaswamy L, et al. Engineering and
implications for treatment success and safety. Prog selection of shuffled AAV genomes: a new strategy
Retin Eye Res. 2020;83(August):100915. https://doi. for producing targeted biological nanoparticles. Mol
org/10.1016/j.preteyeres.2020.100915. Ther. 2008;16(7):1252–60. https://doi.org/10.1038/
6. Calcedo R, Franco J, Qin Q, Richardson DW, mt.2008.100.
Mason JB, Boyd S, Wilson JM. Preexisting neutral- 19. Maheshri N, Koerber JT, Kaspar BK, Schaffer
izing antibodies to adeno-associated virus capsids DV. Directed evolution of adeno-associated
in large animals other than monkeys may confound virus yields enhanced gene delivery vectors. Nat
in vivo gene therapy studies. Hum Gene Ther Biotechnol. 2006;24(2):198–204. https://doi.
Methods. 2015;26(3):103–5. https://doi.org/10.1089/ org/10.1038/nbt1182.
hgtb.2015.082. 20. Mimuro J, Mizukami H, Shima M, Matsushita T, Taki
7. Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson M, Muto S, Higasa S, et al. The prevalence of neutral-
JM. Worldwide epidemiology of neutralizing anti- izing antibodies against adeno-associated virus cap-
bodies to adeno-associated viruses. J Infect Dis. sids is reduced in young Japanese individuals. J Med
2009;199(3):381–90. https://doi.org/10.1086/595830. Virol. 2014;86(11):1990–7. https://doi.org/10.1002/
8. Calcedo R, Wilson JM. Humoral immune response to jmv.23818.
AAV. Front Immunol. 2013;4(October):1–7. https:// 21. Mingozzi F, Hui DJ, Yazicioglu M, Elkouby L,
doi.org/10.3389/fimmu.2013.00341. Hinderer CJ, Basner-tschakarjan E, Wright JF,
9. Calcedo R, Wilson JM. AAV natural infection induces High KA. Overcoming pre-existing humoral
broad cross-neutralizing antibody responses to mul- immunity to AAV using capsid decoys. Mol
tiple AAV serotypes in chimpanzees. Hum Gene Ther Ther. 2013;21(61):S45. https://doi.org/10.1016/
Clin Dev. 2016;27(2):79–82. https://doi.org/10.1089/ s1525-­0016(16)34445-­8.
humc.2016.048. 22. Rapti K, Louis-Jeune V, Kohlbrenner E, Ishikawa
10. Faust SM, Bell P, Cutler BJ, Ashley SN, Zhu Y, K, Ladage D, Zolotukhin S, Hajjar RJ, Weber
Rabinowitz JE, Wilson JM. CpG-depleted adeno-­ T. Neutralizing antibodies against AAV serotypes 1,
associated virus vectors evade immune detection. J 2, 6, and 9 in sera of commonly used animal models.
Clin Investig. 2013;123(7):2994–3001. https://doi. Mol Ther. 2012;20(1):73–83. https://doi.org/10.1038/
org/10.1172/JCI68205. mt.2011.177.
11. Gao G, Vandenberghe L, Wilson J. New 23. Shin JH, Yue Y, Smith B, Duan D. Humoral immunity
recombinant serotypes of AAV vectors. Curr to AAV-6, 8, and 9 in normal and dystrophic dogs.
Gene Ther. 2005;5(3):285–97. https://doi. Hum Gene Ther. 2012;23(3):287–94. https://doi.
org/10.2174/1566523054065057. org/10.1089/hum.2011.125.
12. Halbert CL, Miller D, Mcnamara S, Emerson J, 24. Sun L, Lingli T, Gao G, Sun X, Duan J, You
Ronald L, Ramsey B, Aitken ML. Populations: L. Assessment of a passive immunity mouse model
implications for gene therapy using AAV vectors. to quantitatively analyze the impact of neutralizing
Hum Gene Ther. 2015;17(4):440–7. https://doi. antibodies on adeno-associated virus-mediated gene
org/10.1089/hum.2006.17.440.Prevalence. transfer. J Immunol Methods. 2013;387(1–2):114–20.
13. Hamilton BA, Fraser Wright J. Challenges posed by https://doi.org/10.1016/j.jim.2012.10.003.
immune responses to AAV vectors: addressing root 25. Tellez J, Van Vliet K, Tseng YS, Finn JD, Tschernia N,
causes. Front Immunol. 2021;12(May):1–8. https:// Almeida-Porada G, Arruda VR, Agbandje-­McKenna
doi.org/10.3389/fimmu.2021.675897. M, Porada CD. Characterization of naturally-­
14. Kostic C, Arsenijevic Y. Animal modelling for inher- occurring humoral immunity to AAV in sheep. PLoS
ited central vision loss. J Pathol. 2016;238(2):300–10. One. 2013;8(9):1–11. https://doi.org/10.1371/journal.
https://doi.org/10.1002/path.4641. pone.0075142.
15. Kotterman MA, Schaffer DV. Engineering adeno-­ 26. Tse LV, Klinc KA, Madigan VJ, Castellanos RM,
associated viruses for clinical gene therapy. Nat Rev Rivera LF, Wells LP, Havlik JK, Smith MA-MK,
Genet. 2014;15(7):445–51. https://doi.org/10.1038/ Asokan A. Structure-guided evolution of anti-
nrg3742. genically distinct adeno-associated virus variants
16. Leborgne C, Barbon E, Alexander JM, Hanby H, for immune evasion. Proc Natl Acad Sci U S A.
Delignat S, Cohen DM, Collaud F, et al. IgG-cleaving 2017;114(24):E4812–21. https://doi.org/10.1073/
endopeptidase enables in vivo gene therapy in the pnas.1704766114.
Preexisting Neutralizing Antibodies against Different Adeno-Associated Virus Serotypes in Humans… 123

27. Unzu C, Hervás-Stubbs S, Sampedro A, Mauleón bodies against AAV2, AAV5 and AAV8 in healthy
I, Mancheño U, Alfaro C, de Salamanca RE, et al. and HIV-1-infected subjects in China: implica-
Transient and intensive pharmacological immunosup- tions for gene therapy using AAV vectors. Gene
pression fails to improve AAV-based liver gene transfer Ther. 2014;21(8):732–8. https://doi.org/10.1038/
in non-human primates. J Transl Med. 2012;10:122. gt.2014.47.
https://doi.org/10.1186/1479-­5876-­10-­122. 30. Winkler PA, Occelli LM, Petersen-Jones SM. Large
28. Wang D, Zhong L, Li M, Li J, Tran K, Ren L, He R, animal models of inherited retinal degenerations: a
et al. Adeno-associated virus neutralizing antibodies review. Cell. 2020;9(4):882. https://doi.org/10.3390/
in large animals and their impact on brain intraparen- cells9040882.
chymal gene transfer. Mol Ther Methods Clin Dev. 31. Wu Z, Asokan A, Jude Samulski R. Adeno-associated
2018;11(December):65–72. https://doi.org/10.1016/j. virus serotypes: vector toolkit for human gene
omtm.2018.09.003. therapy. Mol Ther. 2006;14(3):316–27. https://doi.
29. Wang Y, Liu Q, Huang W, Zhang H, Wang Y, Zhao org/10.1016/j.ymthe.2006.05.009.
J, Song A, Xie H, Zhao C, Gao D. Neutralizing anti-
Optimization of Capillary-Based
Western Blotting for MYO7A

Kaitlyn R. Calabro, Sanford L. Boye,


and Shannon E. Boye

Abstract 1 Introduction

Myosin VIIA (MYO7A)-associated Usher syn- Usher syndrome (USH) is a complex disease that
drome type 1B (USH1B) is a severe disorder affects the visual, auditory, and vestibular sys-
that impacts the auditory, vestibular, and tems of patients. Usher syndrome type 1B
visual systems of affected patients. Due to the (USH1B) is the most severe form of the disease
large size (~7.5 kb) of the MYO7A coding as well as one of the most common, accounting
sequence, we have designed a dual adeno-­ for 40–50% of all USH cases [1–3]. Affected
associated virus (AAV) vector-based approach patients are born deaf, with vestibular dysfunc-
for the treatment of USH1B-related vision tion, and begin losing vision within the first
loss. Due to the added complexity of dual-­ decade of life [4–7].
AAV gene therapy, careful attention must be Our lab is developing an adeno-associated
paid to the protein products expressed follow- virus (AAV)-based gene therapy for the treatment
ing vector recombination. In order to improve of USH1B-related retinal degeneration. However,
the sensitivity and quantifiability of our immu- this comes with challenges. USH1B is caused by
noassays, we adapted our traditional western mutations in MYO7A, a relatively large gene of
blot protocol for use with the Jess™ Simple approximately 7.5 kb, whose coding sequence is
Western System. Following several rounds of too large to fit into a single AAV vector (packag-
testing, we optimized our protocol for the ing capacity ~4.8 kb). To overcome this obstacle,
detection of MYO7A in two of our most fre- we developed dual-AAV vector systems that split
quently used sample types, mouse eyes, and MYO7A into two halves that can be co-delivered
infected HEK293 cell lysates. [8]. When the dual vectors enter a target cell,
recombination of the full-length gene is pro-
Keywords moted via the presence of shared, identical
sequences within the vectors [8–11].
MYO7A · Usher syndrome · USH1B · Gene
The added complexity of dual AAV vectors
therapy
relative to traditional single-AAV gene therapies
demands additional scrutiny to ensure safety. For
instance, ensuring proper expression of full-­
K. R. Calabro · S. L. Boye · S. E. Boye (*) length protein and determining whether expres-
Department of Pediatrics, University of Florida, sion of truncated protein emanating from
Gainesville, FL, USA individual vectors is an important safety consid-
e-mail: shaire@ufl.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 125
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_19
126 K. R. Calabro et al.

eration are crucial. We previously found that our 2.2 Use of Animals and Eye
first-generation “hybrid” dual vectors produced a Collections
truncated protein from non-recombined front half
vectors and that this front half vector led to par- All animal procedures were performed according
tial declination of retinal function following to the guidelines of the ARVO statement for the
­subretinal injection in mice [8]. However, the “Use of Animals in Ophthalmic and Vision
expression of this truncated protein was relatively Research” and were approved by the Institutional
low compared to full-length expression, which Animal Care and Use Committees at the
made its characterization and quantification dif- University of Florida. Generation and character-
ficult via traditional western blotting. ization of Myo7a−/− mice has been previously
Our lab previously developed a traditional described [12]. Eyes from C57BL/6J (WT) and
western blotting protocol for the detection of Myo7a−/− (KO) mice were enucleated and flash
MYO7A [8, 12]. This procedure uses 8–12% frozen in protease inhibitors (ThermoFisher,
Tris-Acetate gels to separate denatured protein 87,786). Whole eyes were homogenized in RIPA
samples followed by a transfer to PVDF (polyvi- (radioimmunoprecipitation assay) lysis buffer
nylidene fluoride) membranes. The membranes (ThermoFisher, PI89901) using a Sonic
are incubated overnight with α-MYO7A and Dismembrator (Fisher Scientific, FB50A220).
imaged the following day after secondary anti-
body incubation. The membrane is then incu-
bated with α-VCL (vinculin) as a loading control 2.3 In Vitro Infections and Cell
and reimaged. Developments over recent years Harvesting
have led to advances in immunoblotting technol-
ogy. The Jess™ Simple Western System HEK293 cells were maintained in Dulbecco’s
(ProteinSimple, San Jose, CA, USA) is an auto- Modified Eagle Media (DMEM) containing 10%
mated, capillary-based, size separation, and fetal bovine serum and 50 mg/mL gentamicin
immunoassay system. In addition to the reduc- (complete media) and were incubated at 37 °C in
tion in hands-on time required for a Jess™ assay 5% CO2. Cell morphology and growth rates were
compared to traditional western blot, the auto- closely monitored throughout passages to main-
mated system also allows for reduced variability tain near-identical conditions across infection
and increased sensitivity, leading to more repro- experiments.
ducible results and more accurate quantification. Cells were infected with each virus at a multi-
In this chapter, we will detail the process of opti- plicity of infection (MOI) of 10,000 as previously
mizing our MYO7A immunoassay for use with described [8]. Twenty-four hours post-infection,
Jess™. virus containing media was removed and replaced
with complete media. Cells were harvested 72 h
post-infection. Following collection, RIPA buffer
2 Materials and Methods was added to each sample. Cells were homoge-
nized by vortexing for 30 s.
2.1 AAV Production

All vectors used in this study were packaged in 2.4 Jess™ Simple Western
AAV2. Vector packaging was performed using Optimization
standard plasmid-based transfection methods
in adherent Human Embryonic Kidney (HEK) Prior to use, the total protein concentration of each
293 cells and purified by iodixanol density gra- sample was determined using bicinchoninic assays
dient centrifugation [13]. Following produc- (BCAs, Thermo Scientific, 23,225). To determine
tion, all viruses were titered using PicoGreen the optimal conditions for MYO7A detection in
(ThermoFisher, P7589), as previously mouse eyes and in vitro infection samples, the
described [14]. standard protocol was followed for the 66-440 kDa
Optimization of Capillary-Based Western Blotting for MYO7A 127

Jess™ separation module (ProteinSimple, (WT, C57BL/6 J) mouse eyes and (2) HEK293
SM-W008). To optimize protein concentrations, cells infected with AAV2-smCBA-hybrid (first-­
samples were used at a range from 0.1 to 2.0 μg/ generation) dual vectors (hybrid lysate).
μL. Primary MYO7A antibody dilutions were Negative controls included Myo7a knockout
tested from 1:5 to 1:125 (Santa Cruz, sc-74,516). (KO) mouse eyes and noninfected HEK293 cell
Loading control optimization was performed with lysate (NI lysate). First, we determined the sam-
vinculin (VCL; Cell Signaling Technologies, ple protein concentration and α-MYO7A anti-
13,901) for samples loaded at 2.0 μg/μL with anti- body dilution that would result in saturating
body dilutions ranging from 1:10 to 1:1000. After conditions. Working at the point of saturation is
preliminary runs, it was determined that the ideal because it allows for the most consistent/
RePlex™ module (ProteinSimple, RP-001) was replicable results and the most accurate level of
required to avoid cross-reactivity between anti- quantification.
bodies. All Jess™ data was analyzed using For the first optimization run, WT eyes and
Compass for SW (ProteinSimple, version 5.0.1). hybrid lysate samples were used at 0.1, 0.5, or
2.0 μg/μL with an α-MYO7A dilution of either 1:10
or 1:25 (Fig. 1a). We determined that 2.0 μg/μL
3 Results resulted in prominent ~230 kDa MYO7A signal
and appeared ideal for both the in vivo eye samples
3.1 Optimizing Sample and the in vitro cell lysate. The expected ~120 kDa
and Antibody Concentrations band corresponding to truncated MYO7A (TR
for MYO7A and VCL MYO7A) was also visible in the hybrid lysate.
While the 1:10 antibody dilution looked promising,
The MYO7A Jess™ optimization was per- further testing was conducted to confirm that this
formed using two main samples: (1) Wild-type dilution resulted in saturating conditions.

Fig. 1 Optimizing α-MYO7A and α-VCL for Jess™. (a) ing conditions because halving of antibody concentration
Lane view of α-MYO7A optimization run with sample to 1:10 does not halve signal height. (d) Lane view of
concentrations of 0.1–2.0 μg/μL and antibody dilutions of α-VCL optimization runs with sample concentrations of
1:10 or 1:25. (b) Lane view of α-MYO7A optimization 2.0 μg/μL and antibody dilutions of 1:10, 1:50, 1:250, or
run with sample concentrations of 2.0 μg/μL and antibody 1:100. WT wild-type (C57BL/6J), KO Myo7a−/−, TR
dilutions of 1:5, 1:10, or 1:15. (c) Graph view of WT and MYO7A Truncated MYO7A, S/N signal to noise ratio
KO eyes from (b). α-MYO7A dilution of 1:5 is at saturat-
128 K. R. Calabro et al.

Next, the positive controls were run with a sity data from these samples, we determined that
smaller range of α-MYO7A to confirm saturating the 1:50 antibody dilution was at saturation since
conditions, and negative controls (KO eye and NI the halving of the antibody concentration from
lysate) were used to ensure MYO7A signal speci- 1:50 to 1:100 did not result in a halving of signal
ficity. All samples were used at the previously height/intensity (Fig. 2b). However, when both
identified 2.0 μg/μL concentration, and antibodies were run simultaneously (MYO7A in
α-MYO7A was used at a 1:5, 1:10, or 1:15 dilu- the chemiluminescent channel; VCL in the NIR
tion (Fig. 1b). The positive controls produced channel) in the sample capillary, it was found that
full-length MYO7A signal at all antibody dilu- there was cross-reactivity of VCL with the
tions tested, and hybrid lysate samples also had mouse-HRP (Horseradish Peroxidase) second-
detectable TR MYO7A signal. The graphical ary. This cross-reactivity can clearly be seen in a
analysis of this data (Fig. 1c, hybrid lysate data NI lysate sample (Fig. 2c) in which the VCL sig-
not shown) revealed that the 1:5 α-MYO7A anti- nal is present in both the chemiluminescent and
body dilution resulted in saturating conditions NIR channels.
(halving the antibody concentration from 1:5 to Due to this cross-reactivity, it was determined
1:10 did not result in a halving of signal height/ that the ProteinSimple RePlex™ module would
intensity). This dilution also produced an ade- be necessary to image MYO7A and VCL in the
quate signal-to-noise ratio (S/N, ≥10). In con- same capillary. The RePlex™ reagents are a
trast, no signal was detected in the KO eye or the stripping buffer that removes all reagents (pri-
NI lysate. mary/secondary antibodies, etc.) from the capil-
With the ideal settings for MYO7A detection laries so a second immunoassay can be run
established, we next determined the optimal con- without interference. In other words, once the
ditions for detecting the loading control, vinculin MYO7A immunoassay has been completed, the
(VCL). Because the α-MYO7A is a mouse mono- RePlex™ module removes the antibodies/
clonal antibody, a rabbit monoclonal antibody reagents from the capillaries, and a second,
was selected for α-VCL to avoid interference. sequential, immunoassay with VCL can then be
The optimal MYO7A sample protein concentra- performed.
tion of 2.0 μg/μL was used for the VCL optimiza- For the final run, in which optimized condi-
tion to ensure both antibodies could be used tions were used, HEK293 samples were run at a
simultaneously. Antibody dilutions ranged from concentration of 2.0 μg/μL, and α-MYO7A was
1:10 to 1:1000 (Fig. 1d). Results showed that all used at a 1:5 dilution. Once the MYO7A immu-
antibody concentrations resulted in the ~120 kDa noassay was completed, the RePlex reagents
VCL band. However, due to the similar sizes of were introduced to strip the capillaries. α-VCL
VCL and TR MYO7A, it was determined that was then introduced to the capillaries at 1:50
VCL would need to be run in a separate imaging (Fig. 2d). The results from these immunoassays
channel instead of multiplexed (i.e., MYO7A in provide clear, quantifiable readouts for MYO7A,
chemiluminescent channel; VCL in near infrared, TR MYO7A, and VCL expression without inter-
NIR). ference or cross-reactivity between antibodies.

3.2 Determining Final Run 4 Discussion


Conditions for MYO7A Jess™
Assays Following several rounds of testing, we success-
fully optimized the Jess™ Simple Western proto-
To confirm the optimized antibody dilution for col for the detection of MYO7A in mouse eyes
VCL in the NIR channel, samples were run at and infected HEK293 samples. With these opti-
2.0 μg/μL with either a 1:50 or 1:100 α-VCL mized conditions, we can generate highly sensi-
dilution (Fig. 2a). When viewing the signal inten- tive and quantifiable immunoassay results to
Optimization of Capillary-Based Western Blotting for MYO7A 129

Fig. 2 Determining final run conditions for Jess™. (a) NI Lysate. (d) Lane view of final run conditions.
Lane view of α-VCL optimization run with sample con- α-MYO7A is run in the chemiluminescent channel, and
centrations of 2.0 μg/μL and antibody dilutions of 1:50 or α-VCL is run in the NIR channel after RePlex™ stripping.
1:100. (b) Graph view of hybrid lysate from A. α-VCL (Data from chemiluminescent channel = black; data from
dilution of 1:50 in the near-IR channel (NIR) is at saturat- NIR channel = red). S/N signal to noise ratio, TR MYO7A
ing conditions because halving of antibody concentration truncated MYO7A, CH chemiluminescent channel, NIR
to 1:100 does not halve signal height. (c) Graph view of near-infrared channel
cross-reactivity of α-VCL with mouse-HRP secondary in

3. Stabej PL, Saihan Z, Rangesh N, Steele-Stallard


characterize and improve our dual AAV vector-­ HB, Ambrose J, Coffey A, et al. Comprehensive
based approach for the treatment of USH1B. sequence analysis of nine Usher syndrome genes in
the UK National Collaborative Usher Study. J Med
Acknowledgments We thank Dr. Angel Simpson (UF Genet. 2012;49(1):27–36. https://doi.org/10.1136/
ICBR Monoclonal Antibody Core Facility) and Dr. Greg jmedgenet-­2011-­100468.
Kunkel (ProteinSimple) for their technical assistance and 4. Jacobson SG, Cideciyan AV, Aleman TS, Sumaroka A,
training. Roman AJ, Gardner LM, et al. Usher syndromes due
to MYO7A, PCDH15, USH2A or GPR98 mutations
share retinal disease mechanism. Hum Mol Genet.
2008;17(15):2405–15. https://doi.org/10.1093/hmg/
References ddn140.
5. Jacobson SG, Aleman TS, Sumaroka A, Cideciyan
1. Ouyang XM, Yan D, Du LL, Hejtmancik JF, Jacobson AV, Roman AJ, Windsor EA, et al. Disease boundaries
SG, Nance WE, et al. Characterization of Usher syn- in the retina of patients with usher syndrome caused
drome type I gene mutations in an Usher syndrome by MYO7A gene mutations. Invest Ophthalmol Vis
patient population. Hum Genet. 2005;116(4):292–9. Sci. 2009;50(4):1886–94. https://doi.org/10.1167/
https://doi.org/10.1007/s00439-­004-­1227-­2. iovs.08-­3122.
2. Jaijo T, Aller E, Beneyto M, Najera C, Graziano C, 6. Jacobson SG, Cideciyan AV, Gibbs D, Sumaroka A,
Turchetti D, et al. MYO7A mutation screening in Roman AJ, Aleman TS, et al. Retinal disease course in
Usher syndrome type I patients from diverse origins. J usher syndrome 1B due to MYO7A mutations. Invest
Med Genet. 2007;44(3):e71. https://doi.org/10.1136/ Ophthalmol Vis Sci. 2011;52(11):7924–36. https://
jmg.2006.045377. doi.org/10.1167/iovs.11-­8313.
130 K. R. Calabro et al.

7. Sumaroka A, Matsui R, Cideciyan AV, McGuigan 11. Ghosh A, Yue Y, Duan D. Efficient transgene recon-
DB III, Sheplock R, Schwartz SB, et al. Outer reti- stitution with hybrid dual AAV vectors carrying
nal changes including the ellipsoid zone band in the minimized bridging sequences. Hum Gene
usher syndrome 1B due to MYO7A mutations. Invest Ther. 2011;22(1):77–83. https://doi.org/10.1089/
Ophthalmol Vis Sci. 2016;57(9):OCT253–OCT61. hum.2010.122.
https://doi.org/10.1167/iovs.15-­18860. 12. Calabro KR, Boye SL, Choudhury S, Fajardo D,
8. Dyka FM, Boye SL, Chiodo VA, Hauswirth WW, Peterson JJ, Li W, et al. A novel mouse model of
Boye SE. Dual adeno-associated virus vectors result MYO7A USH1B reveals auditory and visual system
in efficient in vitro and in vivo expression of an Haploinsufficiencies. Front Neurosci. 2019;13 https://
oversized gene, MYO7A. Hum Gene Ther Methods. doi.org/10.3389/fnins.2019.01255.
2014;25(2):166–77. https://doi.org/10.1089/ 13. Zolotukhin S, Potter M, Zolotukhin I, Sakai Y, Loiler
hgtb.2013.212. S, Fraites TJ, et al. Production and purification of
9. Duan DS, Yue YP, Engelhardt JF. Expanding AAV serotype 1, 2, and 5 recombinant adeno-associated
packaging capacity with trans-splicing or over- viral vectors. Methods. 2002;28(2):158–67. https://
lapping vectors: a quantitative comparison. Mol doi.org/10.1016/s1046-­2023(02)00220-­7.
Ther. 2001;4(4):383–91. https://doi.org/10.1006/ 14. Piedra J, Ontiveros M, Miravet S, Penalva C,
mthe.2001.0456. Monfar M, Chillon M. Development of a rapid,
10. Ghosh A, Yue Y, Lai Y, Duan D. A hybrid vector sys- robust, and universal PicoGreen-based method to
tem expands adeno-associated viral vector packaging titer adeno-associated vectors. Hum Gene Ther
capacity in a transgene-independent manner. Mol Methods. 2015;26(1):35–42. https://doi.org/10.1089/
Ther. 2008;16(1):124–30. https://doi.org/10.1038/ hgtb.2014.120.
sj.mt.6300322.
AAV Serotypes and Their
Suitability for Retinal Gene
Therapy

Lynn J. A. Ebner and Christian Grimm

Abstract promotors which play a critical role for cell


targeting.
Throughout the last 25 years, exceptional
progress in retinal gene therapy was achieved. Keywords
The major breakthrough was realized in 2017
Gene therapy · Adeno-associated virus ·
when the FDA approved the adeno-associated
Serotype · Tropism · Photoreceptor transduc-
virus (AAV)-based gene therapy for treatment
tion · Capsid
of the monogenetic disorder Leber congenital
amaurosis type 2 (LCA2). Since then, many
therapies for inherited retinal diseases (IRD)
reached phase I/II clinical trials, targeting dis-
Abbreviations
eases like achromatopsia, choroideremia, reti-
nitis pigmentosa, Stargardt disease, and many
AAP Assembly-activating protein
more (reviewed in (Trapani and Auricchio,
AAV Adeno-associated virus
Trends Mol Med 24:669–681, 2018)).
c-Met Hepatocyte growth factor receptor
Advanced vector and capsid design technolo-
FGFR1 Fibroblast growth factor receptor 1
gies as well as improved gene transfer and
HSPG Heparan sulfate proteoglycan
gene editing methods may lead to refined ther-
IRD Inherited retinal diseases
apies for various eye diseases. Many research
ITR Inverted terminal repeats
departments worldwide focus on optimizing
LCA2 Leber congenital amaurosis type 2
transgene expression by designing novel AAV
rAAV Recombinant AAV
serotypes. Besides serotype tropism, the
RPE Retinal pigment epithelium
method of injection (intravitreal, subretinal, or
suprachoroidal) (Han et al., Hum Gene Ther
31:1288–1299, 2020) defines the efficiency
outcome along with the use of tissue-specific 1 Introduction – Adeno-­
Associated Viral Gene
Therapy
L. J. A. Ebner (*) · C. Grimm
Lab for Retinal Cell Biology, Department of AAVs are single-stranded DNA viruses and
Ophthalmology, University Hospital Zurich, belong to the Parvovirus family (Parvoviridae).
University of Zurich, Schlieren, Switzerland With only 20 nm in diameter, they are small
e-mail: Lynn.Jenny.Alix.Ebner@uzh.ch

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 131
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_20
132 L. J. A. Ebner and C. Grimm

viruses with a genome of approximately 4.8 kb. 2 Serotype Tropism Defines


Inverted terminal repeats (ITRs) of 145 bp flank Cell- Tissue- and
the DNA strand on both sides. They are respon- Host-Infection
sible for efficient multiplication of the genome
[4], integration into the host genome, and Throughout infection, the capsid represents the
encapsidation [21]. Three Cap (Capsid) pro- primary contact to the host cell. This interaction
teins (VP1, VP2, and VP3) encode for the ico- is defined by tissue and cellular tropism and
sahedral capsid structure which protects the needs to be optimized for each experimental
genome and encodes cell binding activity. Four approach. rAAVs enter cells through the interac-
Rep (Replication) proteins (Rep78, Rep68, tion with specific carbohydrates presented on the
Rep52, and Rep40) regulate AAV replication surface of target cells, including sialic acid,
and packaging (reviewed in [15]). In 2010 one galactose, and heparin sulfate. Differences in the
additional protein coding region has been binding preferences, determined by the capsid
described, the aap, which codes for an assem- structure, influence target cell-type specificity
bly-activating protein (AAP) responsible for and transduction efficiency. For example, sero-
proper capsid assembly [12, 16]. However, sub- type 2 cell receptors include heparan sulfate pro-
nuclear localization of AAP varies among dif- teoglycan (HSPG) as a primary receptor [17].
ferent serotypes, and AAP was described as The wide expression of HSPG on the surface of
being nonessential in AAV4, AAV5, and AAV11 many cell types explains the wide tropism of
[16]. AAV2. In the eye, only rAAVs of serotype 2 can
In total 13 AAV serotypes have been described efficiently transduce the retina after intravitreal
so far. While serotypes 2, 3, 5, and 6 were iso- injections, mainly because its binding receptor
lated from humans, serotypes 1, 4, and 7–11 were HSPG is part of the inner limiting membrane
discovered in nonhuman primates [7, 20]. allowing the virus particles to accumulate at the
Serotypes share 65–99% sequence and 95–99% vitreoretinal junction. In contrast, AAV5, which
structural identity (reviewed in [7]). has a sialic acid binding site fails to transduce the
During their lysogenic cycle, 0.1–0.5% of retina upon vitreal injections [5].
wildtype AAVs integrate into the host genome at Post attachment, additional receptors such as
the AAVS1 site of chromosome 19, which con- αVβ5 and α5β1 integrins [2, 18], fibroblast growth
tains a Rep binding element. At a much lower factor receptor 1 (FGFR1) [14], and hepatocyte
frequency, random genome integration is also growth factor receptor (c-Met) [10] mediate cel-
possible (reviewed in [6, 11]). lular entry and function as co-receptors.
Gene therapeutic approaches design recombi-
nant AAVs (rAAV), which only contain ITR
flanked transgenes and no viral DNA to ensure 3 The Eye as a Therapeutic
low immunogenicity and cytotoxicity. To pro- Target
duce virus particles for injection, the required
Rep protein is supplied in trans to exclude The eye and the retina specifically feature a
genome integration at the AAVS1 site in vivo. structural environment suitable for AAV deliv-
Thus, transduced cells usually keep the genome ery. Not only does the accessible, enclosed
of the injected rAAVs as a double-stranded circu- structure make the eye suitable for treatment,
lar episome. However, the ability of the delivered but the high compartmentalization also allows a
DNA to integrate at nonhomologous sites is precise delivery of vectors to the vicinity of tar-
given with a frequency of about 0.1% (reviewed get cells. Systemic side effects are minimized
in [11]). by the blood-retina barrier, and the immune-
AAV Serotypes and Their Suitability for Retinal Gene Therapy 133

privileged site reduces immunogenic clearance. induced-­pluripotent stem cells, and retinal organ-
Defects in photoreceptors represent the majority oid systems are likely to lead to a more precise
of causes for IRDs. Thus, the identification of a design of serotypes. A major drawback of AAV
photoreceptor transducing serotype is highly therapy is the limited size of the expression cas-
requested. Such a serotype has been identified in sette. Thus, lentivirus or nanoparticles display
AAV2 that can mediate expression in photore- alternative options for delivering large expression
ceptors and in the retinal pigment epithelium cassettes. Especially, multifactorial diseases like
(RPE) through binding to HSPG (neural retina), age-­related macular degeneration will depend on
FGFR1 co-receptor (Müller- and RPE cells), gene-independent therapies that target common
and αVβ5 integrin (apical surface of RPE) [1]. disease pathways to reach a broad spectrum of
To further expand the repertoire of AAV vectors patients, independently from underlying
and achieve a more cell-type-specific transduc- mutations.
tion in the retina, hybrid vectors were generated.
They contain the same therapeutic gene packed
between serotype 2 ITRs but are combined with References
a capsid from a different AAV serotype such as
AAV8 (named AAV2/8). These pseudotype 1. Ali RR, Reichel MB, Thrasher AJ, Levinsky RJ,
Kinnon C, Kanuga N, Hunt DM, Bhattacharya
AAVs can circumvent preexisting immunity to SS. Gene transfer into the mouse retina mediated by
one serotype and improve retinal transduction an adeno-associated viral vector. Hum Mol Genet.
kinetics (reviewed in [3]). AAV2/5, for example, 1996;5:591–4.
is more efficient in targeting neurons than 2. Asokan A, Hamra JB, Govindasamy L, Agbandje-­
McKenna M, Samulski RJ. Adeno-associated
AAV2/2 and shows a greater area of transduc- virus type 2 contains an integrin alpha5beta1 bind-
tion. Additional recombinant techniques like ing domain essential for viral cell entry. J Virol.
direct evolution or random peptide library inser- 2006;80:8961–9.
tions are used to create AAVs with unique and 3. Auricchio A. Pseudotyped AAV vectors for constitu-
tive and regulated gene expression in the eye. Vis Res.
suited characteristics. Even exchanges of single 2003;43:913–8.
surface residues such as tyrosine-­ to- 4. Bohenzky RA, Lefebvre RB, Berns KI. Sequence
phenylalanin (Y-F) mutations in AAV2 are used, and symmetry requirements within the internal pal-
for example, to prevent proteasomal degrada- indromic sequences of the adeno-associated virus ter-
minal repeat. Virology. 1988;166:316–27.
tion and increase transduction efficiency in the 5. Dalkara D, Kolstad KD, Caporale N, Visel M,
retina [13]. Klimczak RR, Schaffer DV, Flannery JG. Inner
limiting membrane barriers to AAV-mediated
retinal transduction from the vitreous. Mol Ther.
2009;17:2096–102.
4 Concluding Remarks 6. Deyle DR, Russell DW. Adeno-associated virus vec-
and Future Directions tor integration. Curr Opin Mol Ther. 2009;11:442–7.
7. Drouin LM, Agbandje-McKenna M. Adeno-­
Efficient treatment for IRDs by gene therapy is associated virus structural biology as a tool in vector
development. Futur Virol. 2013;8:1183–99.
highly dependent on efficient transduction of 8. Fitzpatrick Z, Leborgne C, Barbon E, Masat E,
specific retinal cell types. Thus, precise under- Ronzitti G, van Wittenberghe L, Vignaud A, Collaud
standing of capsid tropism is necessary, and engi- F, Charles S, Simon Sola M, Jouen F, Boyer O,
neered capsids are beneficial to reach this goal. Mingozzi F. Influence of pre-existing anti-capsid neu-
tralizing and binding antibodies on AAV vector trans-
Additional techniques are employed to reduce duction. Mol Ther Methods Clin Dev. 2018;9:119–29.
sensitivity to neutralizing antibodies which influ- 9. Han IC, Cheng JL, Burnight ER, Ralston CL, Fick
ence transduction efficiency [8]. Large screen- JL, Thomsen GJ, Tovar EF, Russell SR, Sohn EH,
ings identifying efficient serotypes in nonhuman Mullins RF, Stone EM, Tucker BA, Wiley LA. Retinal
tropism and transduction of adeno-associated virus
primates or human-­derived tissues are necessary varies by serotype and route of delivery (intravitreal,
for a safe and effective translation into the clinics. subretinal, or suprachoroidal) in rats. Hum Gene Ther.
Thus, also improved techniques in human derived 2020;31:1288–99.
134 L. J. A. Ebner and C. Grimm

10. Kashiwakura Y, Tamayose K, Iwabuchi K, Hirai Y, 15. Samulski RJ, Muzyczka N. AAV-mediated gene ther-
Shimada T, Matsumoto K, Nakamura T, Watanabe M, apy for research and therapeutic purposes. Annu Rev
Oshimi K, Daida H. Hepatocyte growth factor recep- Virol. 2014;1:427–51.
tor is a coreceptor for adeno-associated virus type 2 16. Sonntag F, Schmidt K, Kleinschmidt JA. A viral
infection. J Virol. 2005;79:609–14. assembly factor promotes AAV2 capsid formation in
11. McCarty DM, Young SM, Samulski RJ. Integration of the nucleolus. Proc Natl Acad Sci. 2010;107:10220–5.
adeno-associated virus (AAV) and recombinant AAV 17. Summerford C, Samulski RJ. Membrane-associated
vectors. Annu Rev Genet. 2004;38:819–45. heparan sulfate proteoglycan is a receptor for adeno-
12. Naumer M, Sonntag F, Schmidt K, Nieto K, Panke associated virus type 2 virions. J Virol. 1998;72:1438–45.
C, Davey NE, Popa-Wagner R, Kleinschmidt 18. Summerford C, Bartlett JS, Samulski RJ. AlphaVbeta5
JA. Properties of the adeno-associated virus assembly-­ integrin: a co-receptor for adeno-associated virus type
activating protein. J Virol. 2012;86:13038–48. 2 infection. Nat Med. 1999;5:78–82.
13. Petrs-Silva H, Dinculescu A, Li Q, Min S-H, Chiodo 19. Trapani I, Auricchio A. Seeing the light after 25
V, Pang J, Zhong L, Zolotukhin S, Srivastava A, Lewin years of retinal gene therapy. Trends Mol Med.
AS, Hauswirth WW. High-efficiency t­ransduction of 2018;24:669–81.
the mouse retina by tyrosine-mutant AAV serotype 20. Weitzman MD, Linden RM. Adeno-associated virus
vectors. Mol Ther. 2009;17:463–71. biology. In: Snyder RO, Moullier P, editors. Adeno-­
14. Qing K, Mah C, Hansen J, Zhou S, Dwarki V, associated virus. Methods and protocols. New York:
Srivastava A. Human fibroblast growth factor receptor Humana Press; 2011. p. 1–23.
1 is a co-receptor for infection by adeno-associated 21. Zhou X, Muzyczka N. In vitro packaging of adeno-­
virus 2. Nat Med. 1999;5:71–7. associated virus DNA. J Virol. 1998;72:3241–7.
Gene Augmentation for Autosomal
Dominant CRX-Associated
Retinopathies

Chi Sun and Shiming Chen

Abstract Tet-On-hCRX system and adeno-associated


virus (AAV)-mediated gene augmentation. The
outcome of proposed studies will guide future
translational research and suggest guidelines
The cone-rod homeobox (CRX) protein is a for therapy evaluation in terms of treatment
key transcription factor essential for photore- safety and efficacy.
ceptor function and survival. Mutations in
human CRX gene are linked to a wide spec- Keywords
trum of blinding diseases ranging from mild
macular dystrophy to severe Leber congenital CRX mutations · Dominant negative effects ·
amaurosis (LCA), cone-rod dystrophy (CRD), Gene augmentation · Tet-On · AAV gene
and retinitis pigmentosa (RP). These diseases therapy
are still incurable and mostly inherited in an
autosomal dominant form. Dysfunctional
mutant CRX protein interferes with the func- 1 Pathogenic Mechanisms
tion of wild-type CRX protein, demonstrating of CRX Mutations
the dominant negative effect. At present, gene
augmentation is the most promising treatment The human cone-rod homeobox gene CRX
strategy for hereditary diseases. This study (OMIM #602225) is located on chromosome
aims to review the pathogenic mechanisms of 19q13.33. There are four exons in CRX, specifi-
various CRX mutations and propose two ther- cally, the first noncoding and three coding exons.
apeutic strategies to rescue sick photorecep- CRX codes for a 299 amino acid transcription
tors in CRX-associated retinopathies, namely, factor which is predominantly expressed in pho-
toreceptors and the pineal gland, regulating pho-
C. Sun (*) toreceptor development and maintenance [1, 2].
Department of Ophthalmology and Visual Sciences, The protein consists of three major domains: the
Washington University, St. Louis, MO, USA homeodomain at residues 39–99 facilitates the
e-mail: sunchi@wustl.edu DNA binding; the activation domain at residues
S. Chen 113–284, including a WSP motif at residues
Department of Ophthalmology and Visual Sciences, 158–170, contains binding sites for other tran-
Washington University, St. Louis, MO, USA
scription coregulators; a conserved carboxyl ter-
Department of Developmental Biology, Washington
University, St. Louis, MO, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 135
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_21
136 C. Sun and S. Chen

minus motif, also known as the OTX tail, is found Pathogenic mechanisms thus are constituted by
at resides 284–295 [1, 3]. altered DNA binding properties and/or transacti-
To date, 93 disease-causing CRX mutations vation activities of CRX mutant proteins.
have been identified in human patients (www. CRX R90W mutation presents a hypomorphic
hgmd.cf.ac.uk). Pathogenic CRX variants are missense mutation located in the homeodomain
associated with a complex group of inherited reti- [3, 12] and is associated with a dominant late-­
nal diseases (IRDs) such as macular dystrophy onset mild CRD and recessive LCA. The mutant
[4], cone-rod dystrophy (CRD) [1], retinitis pig- protein has reduced DNA binding activity and
mentosa (RP) [5], and Leber congenital amauro- thus malfunctions to transactivate CRX-­
sis (LCA) [6, 7]. Mutations within CRX are downstream target genes such as Rhodopsin [3,
known to occur de novo or to be inherited mostly 12]. The transactivation activity of wild-type
in an autosomal dominant pattern, consisting of (WT) CRX seems normal in the heterozygous
substitution (nonsense and missense mutations) model [3], which proves the limited dominant
and frameshift mutations (deletions and inser- negative effect of the CRX R90W mutation
tions) [8]. Our understanding of these mutations through the process of retinal development. On
has significantly expanded, as a great variety are the other hand, CRX E80A and K88N mutations
being analyzed with the advent of cutting-edge represent distinct antimorphic missense muta-
tools from the sequencing and analytic technolo- tions located in the homeodomain [13–15]. Both
gies. However, a tremendous challenge for the mutations manifest LCA in human patients [1,
pathogenic analysis of CRX-associated retinopa- 14]. Interestingly, some LCA-associated CRX
thies, particularly of the autosomal dominant dis- homeodomain mutations are located at non-­
orders, is to explore the phenotype-genotype conserved residues in human homeodomain par-
correlations between disease severity and CRX alogues [4]. These mutant proteins are predicted
mutations. to bind discrete DNA sequences and show differ-
CRX mutations could cause dominant disor- ent transactivation activities from the WT pro-
ders by two possible mechanisms, namely, the tein, although this has not yet been studied in
haploinsufficiency of the functional CRX and/or mammalian models. Hence, the dominant nega-
various dominant negative or gain-of-function tive effects of the mutant proteins, if any, remain
effects of the mutant CRX. To the extent of data to be proven.
published, haploinsufficiency may not cause The frameshift deletion CRX E168d2 presents
severe phenotypes. The study on Crx+/− mice cor- an antimorphic mutation located in the activation
roborates this claim, as they do not develop any domain [3] and is associated with dominant LCA
detectable functional defects up to 6 months [9]. in human patients [6, 16]. This mutation results
A rare case of human patients with the heterozy- in the early truncation of the activation domain,
gosity of CRX also supports these observations, producing a protein that retains the ability to bind
as only the patients with the nullizygosity of CRX target DNA but fails to transactivate CRX-­
develop LCA [10]. However, in the autosomal downstream target genes [3]. In addition, CRX
dominant disorders, it remains unknown if the E168d2 allele overproduces the mutant protein at
mutant CRX allele could partially abrogate the about 4 times more than the WT protein in het-
production of a functional CRX from the normal erozygous mice, which exacerbates the dominant-­
allele. Further studies are needed to address this negative effect on the binding competition [3]. As
question in detail. Alternatively, dominant nega- a result, the heterozygous mice have impaired
tive activities of various CRX mutant proteins visual functions at 1 month-old (MO) [3] and
have been demonstrated in animal models [9, limited electroretinography (ERG) response at
11]. The reported dominant-negative mutations 3MO (data not shown). Cone degeneration occurs
that arise in the homeodomain are mostly mis- prior to rod degeneration in the heterozygous
sense mutations, and those identified in the acti- mice. As compared to the WT samples, the het-
vation domain are largely frameshifts [7, 9]. erozygous mice have only about 30% survived
Gene Augmentation for Autosomal Dominant CRX-Associated Retinopathies 137

cones at 1 month-old (MO) and no detectable [20]. Five important questions need to be
cones at 3MO (data not shown); mutant rods are answered in order to apply gene augmentation to
functional with shorter outer segments (OS) at treat CRX-associated autosomal dominant disor-
1MO but undergo progressive degeneration till ders. Firstly, how much of augmented CRX is
complete lost at 6MO [3]. Interestingly, the ratio needed to overcome the dominant negative effect
of mutant to WT CRX proteins directly correlates of a given mutation? If a mutant protein competes
with the disease phenotype severity and age of with WT CRX proteins for binding sites, how
onset [3]. In addition, truncation at the last exon much is augmented CRX needed to balance the
by frameshift results in premature terminations ratio of mutant to WT CRX proteins in each pho-
[7, 8], hence the production of shortened but sta- toreceptor? Can altering this ratio rescue the
ble mutant mRNA that can avoid nonsense-­ mutant phenotypes? Secondly, considering CRX
mediated decay [17], which partially contributes is an early photoreceptor transcription factor that
to the dominant negative effect. On the other activates the downstream gene regulatory net-
hand, CrxRip presents the c.763del1 mutation work in the retinal development, could gene aug-
located in the last exon, which results in a skip- mentation impose overexpression toxicity in a
ping of the OTX tail and a non-homologous treated retina? Thirdly, when is augmented CRX
extension of 133 residues [18]. The mutant pro- needed in the treatment? Fourthly, as human
tein does not transactivate CRX-downstream tar- patients with CRX-associated retinopathies are
get genes, suggesting its antimorphic activity often diagnosed at different stages of disease pro-
[18]. More notably, the mutant protein does not gression, what are the windows of opportunities
bind target DNA [18]. Similar to the retinal mani- for gene augmentation to a given disease? Do dis-
festation in patients, CrxRip/+ mice show a LCA-­ eased photoreceptors possess the neuroplasticity
like phenotype [18]. Photoreceptors in CrxRip/+ for gene augmentation outside the development
mice do not form outer segments, due to impaired window? Lastly, can the treated photoreceptors
photoreceptor gene expression and incomplete retain their fated cell identity and functional
differentiation at early development [18]. In addi- capability upon gene augmentation?
tion, the mutant protein is not overexpressed in Answers to these questions could inform the
CrxRip/+ mice. Taken together, the dominant nega- feasibility and strategies to treat CRX-associated
tive effect of CrxRip mutation does not signify a retinopathies. To address these questions at least
competition between the mutant and WT proteins partially, a new transgenic mouse model, Tet-On-­
but likely arises from the disruption of the photo- hCRX, has been developed to allow both quanti-
receptor transcription factor network. tative and temporal control of augmented human
CRX (hCRX) gene expression under a given CRX
mutant background (Fig. 1). Upon binding of
2 Gene Augmentation doxycycline, a conformation change of the
as a Strategy to Treat CRX-­ reverse tetracycline-controlled transcriptional
Associated Retinopathies activator (rtTA) is triggered [21], which allows
the resulted complex bind to tetracycline-­
There are currently limited therapeutic options responsive element (TRE) (Tet-On) [22]. FLAG-­
for CRX-associated retinopathies. Fortunately, tagged human CRX protein will hence be
retinal gene therapy recently shines promise for produced (Fig. 1a). Moreover, the photoreceptor-­
treatment of various inherited blindness [19]. In specific induction can be achieved by the combi-
particular, the gene augmentation approach has nation of two transgenes, namely, a Cre-dependent
been developed to deliver a healthy gene to a dis- pCAG-LSL-rtTA (Fig. 1b) and a Crx promoter-­
eased retina to rescue the defective phenotypes driven Cre, pCrx-Cre [23] (Fig. 1c). Proof-of-­
and visual functions without replacing the mutant concept validation of gene augmentation by
allele. At present, successes of gene augmenta- Tet-On-hCRX system shows that CRX expression
tion are largely restricted to recessive mutations can be induced and detected in Crx−/− mouse reti-
138 C. Sun and S. Chen

Fig. 1 Components of Tet-On-hCRX system. (a) TRE-hCRX (b) pCAG-LSL-rtTA (c) pCrx-Cre

downstream Rho expression. Furthermore, the


comparisons between treated mice with different
starts of the treatments suggests the “early the
better” effect on induced CRX and Rho expres-
sion at P35, as samples with P0–35 treatment dis-
play significantly higher induced CRX and Rho
expression than those with P14–35 or P21–35
treatments (Fig. 2a, b). However, as compared to
the WT samples, treated mice still have signifi-
cantly lower CRX and Rho expression at P35
under current treatment schemes, implying a par-
tial rescue effect. These results suggest that Crx-­
null photoreceptors retain the machinery for
CRX-mediated gene expression and neuroplasti-
city for therapeutic intervention.
As compared to Tet-On-hCRX system, a clini-
Fig. 2 Quantitative PCR (qPCR) analysis of hCrx and
cally applicable approach is adeno-associated
Rho expression in untreated Crx−/−, Crx-BAC-Cre+, Tet-­
On-­hCRX ; doxycycline-treated Crx , Crx-BAC-Cre ,
+ −/− + virus (AAV)-mediated gene augmentation. The
Tet-On-hCRX ; WT mice. (a) Treatments started at P0;
+ retina is surgically accessible, which facilitates
samples were harvested at P5, P14, P21, or P35. (b) intravitreal or subretinal administration of the
Treatments started at P14 or P21; samples were harvested
viral vector [24, 25]. Since the retina is a rela-
at P35. Results are plotted as relative Log2 expression to
untreated samples (n ≥ 4). Asterisks (**, ***, ****) tively immune privileged tissue [24] and photore-
denote p ≤ 0.01, p ≤ 0.001 and p ≤ 0.0001, respectively, ceptors are nondividing and differentiated [26], a
by T-test small amount of the non-integrating AAV viral
vector can be tolerated to generate a therapeutic
nae at all tested ages throughout the entire devel- response [24, 25] with minimal risk of stimulat-
opmental window (postnatal (P) day 5, 14, 21 ing a severe inflammatory immune response [25].
and 35) (Fig. 2), confirming the success of Tet-­ Yet, two immediate challenges emerge during
On-­hCRX system. Doxycycline-treated samples testing AAV-mediated CRX gene augmentation
also display significant upregulation of CRX-­ with animal models, i.e., choice of AAV vectors
Gene Augmentation for Autosomal Dominant CRX-Associated Retinopathies 139

and cell-type specificity. To date, the AAV2/5 vec- [34–37] can also be used to knockout the mutant
tor yields efficient transduction and good tropism allele.
for photoreceptors in P0 mice [27]. Also, the size Since gene augmentation utilizes the tran-
of human or mouse CRX cDNA well fits into the scription machinery of the host cells to express
AAV packaging capacity. Also, photoreceptor-­ the functional protein, cellular conditions of the
specific promoters, including CRX or GRK, can host cells require close monitoring. In a develop-
be employed to achieve desirable sensitivity and ing retina, CRX gene augmentation may not fully
precision. rescue mutations that block early differentiation
However, there are still several safety precau- in photoreceptors, due to an insufficient but early
tions of applying AAV-mediated gene augmenta- window of opportunity. On the contrary, when
tion. Although subretinal administration is the host cells are at the late stage of degeneration
currently the most efficient route for targeting or the degeneration tends to progress too fast,
photoreceptors [28], it causes a transient detach- gene augmentation may not succeed. Prerequisite
ment of the RPE from photoreceptors, which anti-apoptotic or neuroprotective therapies can
might damage photoreceptors at the injection be combined to target the defective photorecep-
site. A possible solution is the use of low-volume tors in order to gain a required time window for
and low-concentration multiple blebs to reduce gene augmentation [38, 39].
the danger of the RPE detachment and systemic Maculopathy has been reported in human
exposure of high concentration by a single bleb, patients of different CRX mutations with large
despite of associated minor risks [29, 30]. In variability in disease progression and symptoms
addition, immunity and tolerance to AAV is [4, 40–42]. In addition, CRX may be involved in
untested in human photoreceptors at early and foveal development, as the haploinsufficiency of
middle childhood [31], and multiplicity of infec- CRX results in subclinical foveal abnormalities in
tion (MOI) of viral vectors per cell may vary at human patients [10]. None of these observations
different stages of the disease, all of which can be validated and studied in non-primate
require further trials on human patients with models.
early-onset diseases. The phenotypic and onset variability between
patients sharing the same CRX mutant allele has
constantly been reported [4, 7, 10, 43, 44]. It
3 Unanswered Questions may be due to, firstly, possible polymorphisms
and Challenges in the CRX promoter region; secondly, the
impacts of possible epigenetic modifications in
One-step gene augmentation may not rescue all coding regions; thirdly, any differential expres-
CRX-associated autosomal dominant disorders. sion of other transcription factors such as NRL
For examples, gain-of-function activities of a and NR2E3; fourthly, variable expression levels
mutant protein with altered DNA binding speci- of the WT and/or mutant alleles; lastly, sexual
ficity, such as CRX K88N, may generate the irre- dimorphisms [42], suggesting CRX may inter-
versible but detrimental effects on the act with specific targets on the sex chromo-
developmental program. In such cases, expres- somes. None of these hypotheses has
sion from the mutant allele needs to be silenced systematically been tested in animal models. In
to prevent the disease progression. Similar addition, the stochastic effects of CRX muta-
approaches of treating RHO-associated autoso- tions and overexpression toxicity on the retinal
mal dominant RP in the mouse and canine mod- degeneration are currently unknown, which
els have shown encouraging outcomes [32, 33]. appends uncertainty on the application of gene
In addition, the CRISPR-Cas9/gRNA technique augmentation.
140 C. Sun and S. Chen

4 Methods Acknowledgments The authors are grateful to Dr. Tom


Glaser for pCrx-Cre mice as a gift, to Dr. Ke Jiang for
critical evaluation of the manuscript. Funding was
4.1 Transgenic Mouse Lines provided by R01 EY012543 and R01 EY032136 (SC),
RPB unrestricted fund (WU-DOVS), EY002687
All male and female mice in this study were on (WU-DOVS), and McDonnell Center for Cellular and
Molecular Neurobiology Fellowship (CS).
the genetic background of C57BL/6J. TRE-hCRX
line was generated for this study. The transgene
had a TRE promoter upstream of full-length
human CRX cDNA sequence, all inserted into the
References
H11 locus. pCAG-LSL-rtTA (Rosa26:pCAG-­ 1. Freund CL, et al. Cone-rod dystrophy due to muta-
LSL-­rtTA) line was obtained from the Jackson tions in a novel photoreceptor-specific Homeobox
Laboratory (Stock No: 029617). pCrx-Cre gene (CRX) essential for maintenance of the photore-
(BAC-Tg Crx-Cre) line was obtained from a pub- ceptor. Cell. 1997;91(4):543–53.
2. Chen S, et al. Crx, a novel Otx-like paired-­
lished colony [23]. Doxycycline diets (200 mg/ homeodomain protein, binds to and Transactivates
kg, Bio-Serv, NJ) were provided to mice (>P14) photoreceptor cell-specific genes. Neuron.
and breeding dams. Intraperitoneal (IP) injec- 1997;19(5):1017–30.
tion of 100 μL of 20 mg/mL doxycycline solu- 3. Tran NM, et al. Mechanistically distinct mouse mod-
els for CRX-associated retinopathy. PLoS Genet.
tion was provided to mice (>P21) once a week 2014;10(2):e1004111.
to maximize the treatment effect. All animal 4. Hull S, et al. The phenotypic variability of retinal
procedures were conducted according to the dystrophies associated with mutations in CRX, with
Guide for the Care and Use of Laboratory report of a novel macular dystrophy phenotype. Invest
Ophthalmol Vis Sci. 2014;55(10):6934–44.
Animals of the National Institute of Health and 5. Sohocki MM, et al. Prevalence of mutations causing
were approved by the Washington University in retinitis pigmentosa and other inherited retinopathies.
St. Louis Institutional Animal Care and Use Hum Mutat. 2001;17(1):42–51.
Committee. 6. Freund CL, et al. De novo mutations in the CRX
homeobox gene associated with Leber congenital
amaurosis. Nat Genet. 1998;18(4):311–2.
7. Rivolta C, et al. Novel frameshift mutations in CRX
4.2 Quantitative PCR (qPCR) associated with Leber congenital amaurosis. Hum
Mutat. 2001;18(6):550–1.
8. Stenson PD, et al. The human gene mutation data-
Each RNA sample was extracted from 2 retinae base: building a comprehensive mutation repository
of a mouse using the NucleoSpin RNA Plus kit for clinical and molecular genetics, diagnostic test-
(Macherey-Nagel, PA). 1 μg of RNA was used to ing and personalized genomic medicine. Hum Genet.
produce cDNA using First Strand cDNA 2014;133(1):1–9.
9. Tran NM, Chen S. Mechanisms of blindness: animal
Synthesis kit (Roche, IN). Technical triplicates models provide insight into distinct CRX-associated
were run for each gene. Primers (5′ to 3′) retinopathies. Dev Dyn. 2014;243(10):1153–66.
used in this study were Crx (F) TGTCCC 10. Ibrahim MT, et al. A complete, homozygous CRX
ATACTCAAGTGCCC, (R) TGCTGTTTCTGC deletion causing nullizygosity is a new genetic
mechanism for Leber congenital amaurosis. Sci Rep.
TGCTGTCG; Rho (F) GCTTCCCTACGCC 2018;8(1):5034.
AGTGTG, (R) CAGTGGATTCTTGCCGCAG; 11. Ruzycki PA, et al. Crx-L253X mutation produces
Ubb (F) CAACATCCAGAAAGAGTCAACC, dominant photoreceptor defects in TVRM65 mice.
(R) ATGTTGTAATCAGAGAGGGTGC. The Invest Ophthalmol Vis Sci. 2017;58(11):4644–53.
12. Swaroop A, et al. Leber congenital Amaurosis caused
reaction master mix consisted of EvaGreen poly- by a homozygous mutation (R90W) in the homeodo-
merase (Bio-Rad Laboratories, CA), 1 μM primer main of the retinal transcription factor CRX: direct
mix, and diluted cDNA samples. Samples were evidence for the involvement of CRX in the devel-
run using a two-step 40-cycle protocol on a opment of photoreceptor function. Hum Mol Genet.
1999;8(2):299–305.
Bio-Rad CFX96 Thermal Cycler (Bio-Rad
­ 13. Chen S, et al. Functional analysis of cone–rod homeo-
Laboratories, CA). The statistical analysis is box (CRX) mutations associated with retinal dystro-
done by Student’s t-test with p < 0.05, CI:95%. phy. Hum Mol Genet. 2002;11(8):873–84.
Gene Augmentation for Autosomal Dominant CRX-Associated Retinopathies 141

14. Nichols LL II, et al. Two novel CRX mutant pro- 29. Peng Y, Tang L, Zhou Y. Subretinal injection: a review
teins causing autosomal dominant Leber congenital on the novel route of therapeutic delivery for vitreo-
amaurosis interact differently with NRL. Hum Mutat. retinal diseases. Ophthalmic Res. 2017;58(4):217–26.
2010;31(6):E1472–83. 30. Xue K, et al. Technique of retinal gene therapy: deliv-
15. Terrell D, et al. OTX2 and CRX rescue overlapping ery of viral vector into the subretinal space. Eye.
and photoreceptor-specific functions in the drosophila 2017;31(9):1308–16.
eye. Dev Dyn. 2012;241(1):215–28. 31. Verdera HC, Kuranda K, Mingozzi F. AAV vector
16. Jacobson SG, et al. Retinal degenerations with immunogenicity in humans: a long journey to suc-
truncation mutations in the cone-rod homeo- cessful gene transfer. Mol Ther. 2020;28(3):723–46.
box (CRX) gene. Invest Ophthalmol Vis Sci. 32. Millington-Ward S, et al. Suppression and replace-
1998;39(12):2417–26. ment gene therapy for autosomal dominant disease in
17. Lejeune F, Maquat LE. Mechanistic links between a murine model of dominant retinitis pigmentosa. Mol
nonsense-mediated mRNA decay and pre-mRNA Ther. 2011;19(4):642–9.
splicing in mammalian cells. Curr Opin Cell Biol. 33. Cideciyan AV, et al. Mutation-independent rho-
2005;17(3):309–15. dopsin gene therapy by knockdown and replace-
18. Roger JE, et al. OTX2 loss causes rod differentiation ment with a single AAV vector. Proc Natl Acad Sci.
defect in CRX-associated congenital blindness. J Clin 2018;115(36):E8547.
Invest. 2014;124(2):631–43. 34. Yu W, Wu Z. In vivo applications of CRISPR-based
19. Campa C, et al. The role of gene therapy in the treat- genome editing in the retina. Front Cell Dev Biol.
ment of retinal diseases: a review. Curr Gene Ther. 2018;6:53.
2017;17(3):194–213. 35. Hung SSC, et al. AAV-mediated CRISPR/Cas gene
20. Vazquez-Dominguez I, Garanto A, Collin editing of retinal cells in vivo. Invest Ophthalmol Vis
RWJ. Molecular therapies for inherited retinal Sci. 2016;57(7):3470–6.
diseases-­current standing. Oppor Chall Genes (Basel). 36. Trapani I, Auricchio A. Seeing the light after 25
2019;10(9):654. years of retinal gene therapy. Trends Mol Med.
21. Das AT, Tenenbaum L, Berkhout B. Tet-on systems 2018;24(8):669–81.
for doxycycline-inducible gene expression. Curr 37. Suzuki K, et al. In vivo genome editing via CRISPR/
Gene Ther. 2016;16(3):156–67. Cas9 mediated homology-independent targeted inte-
22. Gossen M, et al. Transcriptional activation by gration. Nature. 2016;540(7631):144–9.
tetracyclines in mammalian cells. Science. 38. Petit L, Punzo C. Gene therapy approaches for
1995;268(5218):1766–9. the treatment of retinal disorders. Discov Med.
23. Prasov L, Glaser T. Pushing the envelope of retinal 2016;22(121):221–9.
ganglion cell genesis: context dependent function of 39. Chinskey ND, Besirli CG, Zacks DN. Retinal cell
Math5 (Atoh7). Dev Biol. 2012;368(2):214–30. death and current strategies in retinal neuroprotection.
24. Öner A. Recent advancements in gene therapy for Curr Opin Ophthalmol. 2014;25(3):228–33.
hereditary retinal dystrophies. Turkish J Ophthalmol. 40. Ozdek S, et al. Bilateral disc edema and unilateral
2017;47(6):338–43. macular hole in a patient with retinitis pigmentosa.
25. Gupta PR, Huckfeldt RM. Gene therapy for inher- Eur J Ophthalmol. 2006;16(3):487–90.
ited retinal degenerations: initial successes and future 41. Griffith JF, DeBenedictis MJ, Traboulsi EI. A novel
challenges. J Neural Eng. 2017;14(5):051002. dominant CRX mutation causes adult-onset macular
26. Patrício MI, et al. Inclusion of the woodchuck dystrophy. Ophthalmic Genet. 2018;39(1):120–4.
hepatitis virus posttranscriptional regulatory ele- 42. Gonzalez-Gonzalez LA, et al. Novel clinical presen-
ment enhances AAV2-driven transduction of tation of a CRX rod-cone dystrophy. BMJ Case Rep.
mouse and human retina. Mol TherNucleic Acids. 2021;14(4):e233711.
2017;6:198–208. 43. Paunescu K, et al. Genotype–phenotype correlation in
27. Watanabe S, et al. Tropisms of AAV for subretinal a german family with a novel complex CRX mutation
delivery to the neonatal mouse retina and its appli- extending the open reading frame. Ophthalmology.
cation for in vivo rescue of developmental photo- 2007;114(7):1348–57.e1.
receptor disorders. PLoS One. 2013;8(1):e54146. 44. Lu Q-K, et al. A novel CRX mutation by whole-exome
28. Planul A, Dalkara D. Vectors and gene delivery to the sequencing in an autosomal dominant cone-rod dys-
retina. Annual Rev Vision Sci. 2017;3(1):121–40. trophy pedigree. Int J Ophthalmol. 2015;8(6):1112–7.
Txnip Gene Therapy of Retinitis
Pigmentosa Improves Cone Health

Yunlu Xue

Abstract Keywords

Retinitis pigmentosa · Cone photoreceptors ·


Retinitis pigmentosa (RP) is a hereditary reti-
Retina metabolism · Gene therapy · Retinal
nal degenerative disease that can lead to blind-
degeneration
ness. In RP, rod photoreceptors die first,
followed by cone photoreceptors death due to
unknown mechanisms. However, one clue for
cone death concerns their metabolism. Early 1 Introduction
changes suggest that they do not have enough
glucose, which normally fuels their metabo- RP is one of the most prevalent inherited blinding
lism. We sought to design adeno-associated diseases. It affects ~1 in 4000 people worldwide.
virus (AAV)-based gene therapy to address RP also has a high heterogeneity in terms of the
their metabolic challenges and found that types and number of disease genes. Mutation in
overexpressing Txnip is an effective gene any one of ~100 genes creates night blindness,
therapy that extends cone survival and vision which is followed by loss of cone-mediated day-
in three strains of RP mice. The Txnip-­ light, color, and high acuity vision. With the pro-
mediated rescue was found to be dependent gression of the disease, RP patients gradually
upon lactate dehydrogenase b (Ldhb), which lose their peripheral vision, being left with “tun-
is required for lactate catabolism. Txnip also nel vision.” Eventually, some of the patients lose
was found to improve mitochondrial health. the central high acuity vision and become legally
Herein, we propose a model in which Txnip blind. This devastating disease typically origi-
shifts cones from their normal reliance on glu- nates within the photoreceptors.
cose to enhanced utilization of lactate to ben- In RP, rods are typically directly affected by
efit cones in a condition where the glucose the disease genes. At least some of the disease
supply is limiting. genes are only expressed in rods (e.g., PDE6B)
and cones are initially totally functional. When
rods die, the cones gradually get sick and begin to
lose function. The cones start to die only when
the majority of the neighboring rods are gone,
Y. Xue (*)
leading to the severe loss of the daylight color
Department of Genetics, Blavatnik Institute,
Harvard Medical School, Boston, MA, USA vision in the corresponding visual field.
e-mail: yunlu_xue@hms.harvard.edu Regardless of what disease genes patients carry,

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 143
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_22
144 Y. Xue

it seems that the RP cone degeneration pattern is glucose uptake by cones via the binding or
always the same, that is, secondary to the rod RdCVF to its chaperone, Basigin1. This led to
death. Our ultimate goals are (1) to understand the activation of GLUT1, the major glucose
the mechanism of the nonautonomous cone death transporter of cones [1]. With loss of rods in RP,
in RP and (2) to find a generic (i.e., gene-­agnostic) RdCVF secretion dropped, hampering glucose
treatment to preserve cones in RP to save the day- uptake by cones. In a second study, Wang et al.
light color vision. proposed that glucose could be trapped in the
RPE of RP animals using 2-NBDG, a fluorescent
analog of glucose, creating a glucose supply
2 RP Glucose Hypothesis issue for RP cones [11].
However, all of the studies provided only indi-
Several hypotheses of RP cone degeneration have rect evidence in supporting the RP glucose
been proposed. One of them is called the RP glu- hypothesis. It was technically challenging to
cose hypothesis [10]. Punzo et al. worked with measure glucose level in cones directly due to
four RP mice strains to conduct microarray their scarce number in retina. As one of our goals
experiments and identified common changes in was to find a therapy to delay RP cone degenera-
metabolic gene expression. Inspired by this find- tion, we decided to focus on treatment first. It is
ing, Punzo et al. looked into the metabolic path- well known that glucose goes glycolysis to be
ways more carefully and found multiple lines of converted to pyruvate, which has two fates: either
evidence which suggested an imbalance in RP turns into lactate via lactate dehydrogenase and
cone metabolism at an early stage of degenera- exits the cell or turns into acetyl-CoA in mito-
tion. One of the observations concerned mTOR chondria and fuels the TCA cycle. To test if aug-
signaling, which is a major regulator of metabo- menting glycolytic pathway can provide any
lism. Phosphorylated-mTOR (Ser2448) was therapeutic effect for RP cone survival, we over-
found in the inner segments of wild-type cone expressed key glycolytic enzymes (e.g., hexoki-
photoreceptors but was missing at a very early nases, phosphofructokinase, and pyruvate kinase)
stage of degeneration in RP cones. The loss of in RP cones.
mTOR-phosphorylation could be induced by cul-
turing wild-type retinal explants in glucose-free
medium. In addition, evidence of an upregulation 3 Txnip Presents a Robust
in chaperone-mediated autophagy was found, Therapeutic Effect
which is consistent with a nutrient shortage.
Punzo et al. proposed that RP cones suffer from a We used AAV serotype 8 with a cone-specific
glucose shortage, which might subsequently trig- promoter to provide safe and specific gene deliv-
ger the nonautonomous cone death in RP. The ery to cones. We worked with three RP mouse
cause of this hypothesized glucose shortage was strains: rd1, rd10, and Rho−/− that degenerate
suspected to be a disruption in the interactions from fast to slow. Both rd1 and rd10 carry muta-
between the RPE processes and photoreceptor tions in Pde6b, a rod-specific phototransduction
inner and outer segments, which is a major inter- gene, similar to Rho. Subretinal injection was
face for the uptake of nutrients and shedding of performed in RP mice at P0 to achieve a full-­
waste for photoreceptors. retina infection, as well allowing us working with
Two independent studies subsequently pro- the fast-degenerating rd1 retina. We co-injected a
vided evidence for the RP glucose hypothesis. small fraction of AAV-H2BGFP to label the cone
Aït-Ali et al. studied the mechanism for cone sur- nuclei and to facilitate the cell recognition by an
vival provided by rod-derived cone viability fac- automated counting software. We screened 20
tor (RdCVF). They found that it could stimulate genes and some of their combinations using rd1.
Txnip Gene Therapy of Retinitis Pigmentosa Improves Cone Health 145

Among these tested genes, only Txnip, the thio- Therefore, we investigated if Txnip rescued by
redoxin interacting protein, showed enhanced switching RP cone fuel source from glucose to
rd1 cone survival at P50 [13]. Similar rescue lactate. Using shRNAs, we found that Txnip res-
effects were also observed in rd10 and Rho−/− cue was dependent on Ldhb, a critical gene for
strains, suggesting Txnip to be a target for gene-­ lactate to pyruvate conversion, suggesting that
agonistic RP cone treatment. Using optomotor lactate catabolism was critical for Txnip rescue.
behavioral assay, we demonstrated that the loss We also observed that Txnip enhanced the
of cone-mediated vision was also significantly ATP:ADP level in RP cones bathed in lactate-­
delayed by Txnip in rd10 and Rho−/− strains. only culture medium, and this phenotype was
What is Txnip? Txnip is a member of α-arrestin dependent on Ldhb as well. Mitochondria in
family proteins. It is best known for three func- Txnip-treated RP cones appeared to be healthier
tions: (1) modulating thiol redox signaling of than control in terms of size and transmembrane
cells [4, 7], (2) reducing glucose uptake [12], and potential; however, mitochondrial improvement
(3) switching fuel source to non-glucose fuels alone was not sufficient to rescue RP cones. With
[2]. We started to investigate the possible rescue Txnip treatment, the Na+/K+ ATPase pumps func-
mechanism of Txnip along these three tioned better in lactate-only medium. The cone
directions. opsins also appeared to be better retained at pro-
It has been well established that Txnip inter- tein level with Txnip treatment. This observation
acts with thioredoxins via C247 residue to modu- was consistent with an improved energy status by
late the thiol redox signaling [8, 9]. A serine Txnip addition.
mutation of this residue (Txnip.C247S) com- With some additional experimental evi-
pletely abolishes the interaction between Txnip dences, we proposed a working model for
and thioredoxins. If modulating thiol redox sig- Txnip rescue, in which the wild-type photore-
naling is important for Txnip rescue, Txnip. ceptors are genetically programmed to rely on
C247S would hamper the therapeutic effect. To glucose as the major fuel. In wild-type retina,
test this hypothesis, we made a Txnip.C247S glucose from choriocapillaris gets through RPE
vector and tested its efficacy. Surprisingly, we to photoreceptors without much retention. In
observed that Txnip.C247S was more efficacious return, the photoreceptors export lactate for the
than the wild-type allele of Txnip in all three RP RPE to use. In addition, photoreceptors daily
strains. This result suggested that Txnip did not lose 10% of outer segments, which are enriched
work through interacting with thioredoxins, and by amino acids and lipids, to RPE as additional
the well-known Txnip-Thioredoxins interaction fuels. This metabolic relationship between RPE
was indeed deleterious to the rescue. The C247S and photoreceptors has been described as an
mutation might also free up both Txnip and thio- ecosystem in the eye by Hurley and colleagues
redoxin proteins to better benefit RP cone sur- [5]. In RP, due to the loss of rods, the fuel
vival. In addition, we also ruled out that knocking exporting from photoreceptors to RPE is
down GLUT1 alone can enhance RP cone sur- expected to be dramatically reduced. RPE
vival, suggesting the Txnip rescue was unlikely might start to intersect the glucose for its own
to be induced by reducing the glucose uptake use. As a potential consequence of this glucose
alone. shortage, photoreceptors may start to starve and
In a metabolomics study using conditional show symptoms as described by Punzo et al.
knockout mice, Txnip was proposed to function [10]. Adding Txnip could reprogram the fuel
to switch the fuel source from glucose to non-­ selection of RP cones and enable them to use
glucose fuels such as lipids, lactate, amino acids, lactate instead of glucose, which they originally
and ketones [2]. In addition, a recent study rely on. Lactate could be more available to RP
showed that Txnip-null patients presented lactic cones than glucose from the blood circulation
acidosis [6], suggesting that Txnip may function [3], to relieve the starvation and to help them to
to enhance the lactate catabolism of cells. survive.
146 Y. Xue

4 Future Directions 3. Hui S, Ghergurovich JM, Morscher RJ, et al. Glucose


feeds the TCA cycle via circulating lactate. Nature.
2017;551:115–8.
Currently, the molecular mechanism of Txnip 4. Junn E, Han SH, Im JY, et al. Vitamin D3 up-regulated
fuel switching is still not clear. Because Txnip protein 1 mediates oxidative stress via suppressing the
is one type of α-arrestin proteins, it is most thioredoxin function. J Immunol. 2000;164:6287–95.
5. Kanow MA, Giarmarco MM, Jankowski CS, et al.
likely that Txnip works through protein-protein Biochemical adaptations of the retina and retinal pig-
interaction. However, neither reducing glucose ment epithelium support a metabolic ecosystem in the
uptake, via GLUT1 interaction, nor improving vertebrate eye. elife. 2017;6:e28899.
mitochondrial health, possibly via PARP1 6. Katsu-Jiménez Y, Vázquez-Calvo C, Maffezzini C,
et al. Absence of TXNIP in humans leads to lactic aci-
interaction, alone could enhance the RP cone dosis and low serum methionine linked to deficient
survival. Txnip’s interaction with thioredoxins respiration on pyruvate. Diabetes. 2019;68:709–23.
is indeed detrimental to the rescue. Other than 7. Nishiyama A, Matsui M, Iwata S, et al. Identification
GLUT1, PARP1, or thioredoxins, no protein of thioredoxin-binding protein-2/vitamin D(3) up-­
regulated protein 1 as a negative regulator of thio-
partner has been reported to functionally inter- redoxin function and expression. J Biol Chem.
act with Txnip to explain the rescue to our 1999;274:21645–50.
knowledge. In the future, it is important to 8. Patwari P, Chutkow WA, Cummings K, et al.
identify this unknown downstream protein Thioredoxin-independent regulation of metabo-
lism by the α-arrestin proteins. J Biol Chem.
partner(s) of Txnip and to study how they phys- 2009;284:24996–5003.
ically interact with each other. With this miss- 9. Patwari P, Higgins LJ, Chutkow WA, et al. The inter-
ing piece uncovered, we also may be able to action of thioredoxin with Txnip: evidence for forma-
modify Txnip to benefit RP patients even tion of a mixed disulfide by disulfide exchange. J Biol
Chem. 2006;281:21884–91.
further. 10. Punzo C, Kornacker K, Cepko CL. Stimulation of
the insulin/mTOR pathway delays cone death in a
mouse model of retinitis pigmentosa. Nat Neurosci.
References 2009;12:44–52.
11. Wang W, Lee SJ, Scott PA, et al. Two-step reactivation
of dormant cones in retinitis pigmentosa. Cell Rep.
1. Aït-Ali N, Fridlich R, Millet-Puel G, et al. Rod-
2016;15:372–85.
derived cone viability factor promotes cone sur-
12. Wu N, Zheng B, Shaywitz A, et al. AMPK-dependent
vival by stimulating aerobic glycolysis. Cell.
degradation of TXNIP upon energy stress leads to
2015;161:817–32.
enhanced glucose uptake via GLUT1. Mol Cell.
2. DeBalsi KL, Wong KE, Koves TR, et al. Targeted
2013;49:1167–75.
metabolomics connects thioredoxin-interacting pro-
13. Xue Y, Wang SK, Rana P, et al. AAV-Txnip prolongs
tein (TXNIP) to mitochondrial fuel selection and reg-
cone survival and vision in mouse models of retinitis
ulation of specific oxidoreductase enzymes in skeletal
pigmentosa. elife. 2021;10.
muscle. J Biol Chem. 2014;289:8106–20.
Part V
Human Retinal Degeneration
Factors Affecting Readthrough
of Natural Versus Premature
Termination Codons

Avigail Beryozkin, Kerstin Nagel-Wolfum,


Eyal Banin, and Dror Sharon

Abstract and the efficiency of the process during


readthrough following TRIDs treatments.
Nonsense mutations occur within the open-­
reading frame of a gene resulting in a prema- Keywords
ture termination codon (PTC). PTC-containing
mRNAs can either be degeraded or cause pre- Inherited retinal diseases · Nonsense muta-
mature translation termination producing a tions · Stop codons · Translational
truncated protein that can be either nonfunc- readthrough · Translation termination
tional or toxic. Translational readthrough
inducing drugs (TRIDs) are small molecules
that are able to induce readthrough, resulting
in the restoration of full-length protein expres- Abbreviations
sion. The re-expressed proteins usually harbor
a missense change. The effciency of individ- aa Amino acid
ual TRIDs is variable and varies between dif- eRF1 Eukaryote release factor 1
ferent genes and even different nonsense IRDs Inherited retinal diseases
mutations in the same gene. This review sum- NMD Nonsense-mediated mRNA decay
marizes factors, including the sequences PTC Premature termination codon
located upstream and downstream the disease-­ TR Translational readthrough
causing mutation and the type of PTC, affect- TRID Translational readthrough inducing
ing the translational readthrough process by drug
modulating the type of amino acid insertion UTR Untranslated region

1 Introduction
A. Beryozkin · E. Banin · D. Sharon (*)
Department of Ophthalmology, Hadassah Medical
Center, Faculty of Medicine, The Hebrew University Nonsense mutations are responsible for ∼11% of
of Jerusalem, Jerusalem, Israel disease-causing mutations [1] and ∼18% in
e-mail: dror.sharon1@mail.huji.ac.il inherited retinal diseases (IRDs) [2]. Different
K. Nagel-Wolfum translational readthrough (TR)-inducing drugs
Institute of Molecular Physiology & Institute of (TRIDs) are known as a potential treatment strat-
Developmental Biology and Neurobiology, Johannes egy for these mutations. Gentamicin was one of
Gutenberg University of Mainz, Mainz, Germany

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 149
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_23
150 A. Beryozkin et al.

the first reported TRIDs, but its severe side for the tRNA that already delivered its aa. During
effects, including in the retina, do not allow long-­ the initiation of translation, the initiator tRNA is
term treatment [3–5]. Additional compounds located at the P site. When elongation starts, a
with less side effects were reported, including new aminoacyl–tRNA enters the A site. The ribo-
PTC124 (Translarna™) and G418 (Geneticin) [3, some is responsible for the correct selection of
6]. The efficiency of each TRID is different and tRNA. During this proofreading process, tRNAs
varies among genes and termination codons [6]. with incorrect anticodons are rejected and
In this mini review, we summarize factors affect- replaced by a new tRNA. A failure at this proof-
ing the efficiency of the readthrough process. reading step results in a missense change. The
fidelity of the decoding is very high, and mis-
sense errors can be found in a frequency of 10−7
2 Protein Translation to 10−4, depending on the position of the aa in the
protein and the mismatch type [7].
The eukaryote ribosome (consisting of two sub-
units: 40S and 60S, Fig. 1) reads the mRNA
sequence and translates it into a chain of amino 3 The Process of Translation
acids (aas) which build the encoded protein. The Termination
ribosome contains three sites (Fig. 1): A, the
entry site for a new tRNA attached to an aa; P, The termination step occurs when one of the
occupied by peptidyl-­ tRNA which carries the three stop codons (UAA, UAG, or UGA) is posi-
growing polypeptide chain; and E, the exit site tioned at the A site. None of the aminoacyl–

Fig. 1 Factors affecting readthrough during translation. codon and the tRNA anticodon (grey polygon), the aa
The eukaryote ribosome (60S subunit: light yellow, 40S joins the chain of aas of a previously synthesized peptide.
subunit: dark yellow) slides from 5′ toward 3′ on the Different positions on the mRNA are marked by num-
mRNA strand. New tRNA molecule charged with aa bered squares. Nucleotides that show higher preference
enters the A site. If there is a match between the mRNA for readthrough are shown in the upper left panel
Factors Affecting Readthrough of Natural Versus Premature Termination Codons 151

tRNAs fit these codons, and the polypeptide is of nucleotides at positions +5 to +9 varies
released from the ribosome. The basal level of between the three termination codons (Table 1)
stop codon readthrough frequency is usually less and between organisms [16, 23, 24]. A study in
than 10−4 [8], but it can also appear more fre- yeasts identified a consensus sequence [CA(A/G)
quently, depending on different cis and trans fac- N(U/C/G)A] downstream the stop codon, which
tors [9, 10]. The stop codon itself might also promotes more than 5% readthrough efficiency
influence the frequency of the basal readthrough, [9]. A similar study preformed in HEK293T cells
where UGA has the highest basal readthrough revealed a much higher level of TR (7–31%) in
potential [11, 12]. genes in which UGA as a stop codon was fol-
lowed by a CUAG sequence [16].

4 Nucleotide Sequences
Upstream the Stop Codon 6 Nonsense Mutations and TR

Nonrandom distribution of nucleotides located Nonsense mutations, also known as premature


upstream to the stop codon was observed in vari- termination codons (PTCs), result either in the
ous species. In addition, several codons appear activation of the nonsense-mediated mRNA
more frequently before specific termination decay (NMD) process that degrades the aberrant
codons. These sequences are speculated to modu- mRNA molecules or cause premature termina-
late the efficacy of peptide chain termination and tion of protein synthesis, leading to a truncated
stop codon readthrough [13]. Several experi- protein that might be toxic. Natural readthrough
ments performed in yeasts aiming to estimate the of PTCs is higher (0.01–1%) [11, 25] than the
effect of the 5′ sequences revealed that adenine in basal level of TR of termination codons (0.001–
positions −1 and −2 stimulates TR of mainly 0.1%) [10]. This difference stems from several
UAG but also other termination codons [14, 15]. reasons, such as different upstream and down-
Similar experiments on HEK293T cells showed stream sequence signatures and proximity to 3′
that purines in position −1 are associated with UTR poly(A)-binding proteins.
higher TR levels, and uracil is associated with About 18.5% of IRD-causing mutations are
lower levels [16, 17]. nonsense [2], and several genes show even higher
percentage (e.g., >30% of choroideremia cases)
[26]. Therefore, targeting nonsense mutations
5 Nucleotide Sequence with different TRIDs can serve as a potential
Downstream the Stop Codon treatment for a relatively high percentage of
IRDs.
The role of the +4 position in influencing TR The mechanism of TR of natural termination
level and its correspondence with the stop codon codons is different from that of PTCs [27, 28],
itself was studied extensively (Table 1). While it and therefore the impact of the surrounding
is well accepted that the effect of TR depends sequences is different. Interestingly, the efficacy
mostly on the stop codon itself, the findings of TR depends mostly on the sequence of the stop
regarding the effect of the nucleotide in +4 posi- codon itself, with the same ranking order as natu-
tion are controversial. The highest basal levels of ral termination codons but also on the drug and
TR in mammalian cells (up to 3–4%) were the sequence surrounding the PTC [11, 18–22,
observed in UGA followed by cytosine. Other 29, 30]. It is well accepted that cytosine at posi-
stop codons also showed enhanced TR when fol- tion +4 (Fig. 1 and Table 1) is associated with
lowed by cytosine. However, the TR level of higher TR. The effect of TR on other nucleotides
other nucleotides in +4 position varies among in this position is variable. Basal TR level of nat-
studies and therefore is probably influenced by ural termination codons might result in the inser-
their cis and trans factors [11, 18–20]. The effect tion of different aas at different frequencies
152 A. Beryozkin et al.

Table 1 The effect of a stop codon and a nucleotide at +4 position on the TR potential with or without TRID
treatment
Stop codon
UGA > UAG > UAA Treatment Reference
C~U>G>A C > U> > G > A C > U> > G > A Untreated [18]
C> > A > U ~ G U>C>G~A C>G>U~A Untreated [11]
C>A~G~U C>A~G~U C>A~G~U Untreated [19]
C>A>G>U C>A~G~U C>A~G~U Untreated [20]
C>U>A~G C>U~G~A C~U~G~A Gentamicin [21]
C>A=G>U U>C=G>A C>G>A=U Gentamicin [11]
C>A~G~U C>A~G~U C>A~G~U Gentamicin [19]
C> > A = U = G C>A=U=G C>A=U=G Gentamicin [22]
C>A=U=G C>A=U=G C>A=U=G PTC124 [22]
C> > A = U = G C>A=U=G C>A=U=G G418 [22]
C>U>A>G C>U~G~A C>U>G~A G418 [21]
C>A=G>U U>C=G>A C>G>A=U G418 [11]

Table 2 Possible and actual aas that are inserted instead of termination codons
Termination Possible Possible Basal RT of natural Basal RT After ataluren After G418
codon mispairing aa termination codons of PTC treatment treatment
UGA UGU/C Cys [31, 32] [33, 34] [33, 35] [33, 35]
UGG Trp [31, 36, 37] [33, 35] [33, 35] [33, 35]
UAA Ter
UCA Ser
UUA Leu
GGA Gly
C/AGA Arg [31] [33–35] [33, 35] [33, 35]
UAG UAA Ter
UAU/C Tyr [37] [33–35, [33, 35] [33, 35]
38]
UGG Trp [35, 38] [35]
UCG Ser
UUG Leu
GAG Glu [31, 37] [34]
AAG Lys [33, 34, [33] [33, 35]
38]
CAG Gln [33, 35] [33, 35] [33, 35]
UAA UAG Ter
UAU/C Tyr [32, 37] [34, 35] [33, 35] [33, 35]
UCA Ser
UGA Ter
UUA Leu
GAA Glu [31, 37] [34]
AAA Lys [34]
CAA Gln [35] [33, 35] [33, 35]

instead of the termination codon (Table 2). 7 Decoding of Termination


Several studies showed that mainly tryptophan Codons
and arginine are inserted during readthrough of
UGA [31, 36, 37]. Natural readthrough of UAG TR of natural termination codons and PTCs can
and UAA usually results in insertion of a glu- appear in a basal level with the latter showing
tamic acid (Tables 2 and 3) [31, 37]. higher readthrough levels. Studies in various
Factors Affecting Readthrough of Natural Versus Premature Termination Codons 153

Table 3 Amino acids inserted during readthrough


Condition Termination codon Alternatively inserted aas References
Naturally occurring termination codons UGA Arg > Cys > Trp [31]
Cys [32]
Trp [36]
Trp [37]
UAG Glu > Tyr > Gln [37]
Glu [31]
UAA Glu > Tyr > Gln [37]
Glu [31]
Tyr [32]
PTC Natural TR UGA Trp > Cys > Arg [33]
Arg > Trp [35]
Ata Arg > Trp > Cys [33]
Arg > Trp > Cys [35]
G418 Arg > Cys > Trp [33]
Arg > Trp > Cys [35]
Natural TR UAG Gln > Tyr > Lys [33]
Tyr > Gln > Trp [35]
Ata Gln > Tyr > Trp [33]
Gln > Tyr > Trp [35]
G418 Gln > Tyr [33]
Gln > Tyr > Lys [35]
Natural TR UAA Gln > Tyr > Lys [33]
Tyr > Gln [35]
Ata Tyr > Gln [33]
Tyr [35]
G418 Gln > Tyr [33]
Tyr > Gln [35]

organisms showed that TR is not a random pro- To summarize, termination of the translation
cess. When TR occurs on natural termination process is important for functional proteins to be
codons, they might be decoded only as particu- produced. The distribution of nucleotides sur-
lar aas (Table 2). TR of PTCs can result in the rounding the natural termination codon is not ran-
same aas as in natural termination codons, as dom and therefore a PTC that appears within the
well as additional ones (Table 2). Studies in open reading frame will not be flanked by the
HEK293T cells revealed differences in the fre- same elements appearing around natural termina-
quency of the integrated aas after ataluren (argi- tion codons and will therefore show different sen-
nine, 69%; tryptophan, 28%; cysteine, 0.7%) sitivity to TRIDs. Since treatment with TRIDs
and gentamicin (arginine, 48%; cysteine, 27%; does not yield WT proteins, but rather full-length
tryptophan, 24%) treatments [33]. Interestingly, proteins with a single aa change, one needs to ver-
most aas that are inserted after drug treatment ify that the function of the newly produced, slightly
are the same as those inserted due to spontane- different proteins is high enough to correct the
ous TR of PTC but with different frequencies phenotype.
(Tables 2 and 3). An additional interesting
observation is that some of the aas are not Acknowledgments This study was funded by the Israeli
decoded, even though there is only a single Ministry of Health (grant number 3-14995), FAUN
Foundation and German Research Council/DFG,
nucleotide difference between the PTC and SPP2127 – Gene and Cell based therapies to counteract
codon of the potential aa. neuroretinal degeneration (NA 399443882).
154 A. Beryozkin et al.

Declaration of Competing Interest The authors declare 13. Arkov AL, Korolev SV, Kisslev LL. 5′ contexts of
that they have no competing interests. Escherichia coli and human termination codons are
similar. Nucleic Acids Res. 1995;23:4712–6.
14. Bonetti B, Fu L, Moon J, Bedwell DM. The efficiency
References of translation termination is determined by a syner-
gistic interplay between upstream and downstream
sequences in Saccharomyces cerevisiae. J Mol Biol.
1. Mort M, Ivanov D, Cooper DN, Chuzhanova NA. A 1995;251:334–45.
meta-analysis of nonsense mutations causing human 15. Tork S, Hatin I, Rousset JP, Fabret C. The major 5′
genetic disease. Hum Mutat. 2008;29:1037–47. determinant in stop codon read-through involves
2. Schneider N, Sundaresan Y, Gopalakrishnan P, two adjacent adenines. Nucleic Acids Res.
Beryozkin A, Hanany M, Levanon EY, Banin E, 2004;32:415–21.
Ben-Aroya S, Sharon D. Inherited retinal diseases: 16. Loughran G, Chou MY, Ivanov IP, Jungreis I, Kellis
linking genes, disease-causing variants, and relevant M, Kiran AM, Baranov PV, Atkins JF. Evidence of
therapeutic modalities. Prog Retin Eye Res. 2022 efficient stop codon readthrough in four mammalian
Jul;89:101029. genes. Nucleic Acids Res. 2014;42:8928–38.
3. Goldmann T, Overlack N, Moller F, Belakhov V, 17. Cassan M, Rousset JP. UAG readthrough in mam-
van Wyk M, Baasov T, Wolfrum U, Nagel-Wolfrum malian cells: effect of upstream and downstream stop
K. A comparative evaluation of NB30, NB54 and codon contexts reveal different signals. BMC Mol
PTC124 in translational read-through efficacy for Biol. 2001;2:1–8.
treatment of an USH1C nonsense mutation. EMBO 18. Mccaughan KK, Brown CM, Dalphin ME, Berry
Mol Med. 2012;4:1186–99. MJ, Tate WP. Translational termination efficiency in
4. Lopez-Novoa JM, Quiros Y, Vicente L, Morales AI, mammals is influenced by the base following the stop
Lopez-Hernandez FJ. New insights into the mecha- codon. Proc Natl Acad Sci U S A. 1995;92:5431–5.
nism of aminoglycoside nephrotoxicity: an integrative 19. Floquet C, Hatin I, Rousset JP, Bidou L. Statistical
point of view. Kidney Int. 2011;79:33–45. analysis of readthrough levels for nonsense muta-
5. Goldmann T, Rebibo-Sabbah A, Overlack N, tions in mammalian cells reveals a major deter-
Nudelman I, Belakhov V, Baasov T, Ben-Yosef T, minant of response to gentamicin. PLoS Genet.
Wolfrum U, Nagel-Wolfrum K. Beneficial read-­ 2012;8(3):e1002608
through of a USH1C nonsense mutation by designed 20. Beznosková P, Wagner S, Jansen ME, Von Der Haar
aminoglycoside NB30 in the retina. Invest Ophthalmol T, Valášek LS. Translation initiation factor eIF3 pro-
Vis Sci. 2010;51:6671–80. motes programmed stop codon readthrough. Nucleic
6. Lee HLR, Dougherty JP. Pharmaceutical therapies Acids Res. 2015;43:5099–111.
to recode nonsense mutations in inherited diseases. 21. Howard MT, Shirts BH, Petros LM, Flanigan KM,
Pharmacol Ther. 2012;136:227–66. Gesteland RF, Atkins JF. Sequence specificity of
7. Manickam N, Nag N, Abbasi A, Patel K, Farabaugh aminoglycoside-induced stop codon readthrough:
PJ. Studies of translational misreading in vivo show Potential implications for treatment of Duchenne
that the ribosome very efficiently discriminates muscular dystrophy. Ann of Neuro. 2001;48:164–169.
against most potential errors. RNA. 2014;20:9–15. 22. Martorell L, Cortina V, Parra R, Barquinero J, Vidal
8. Schueren F, Thoms S. Functional translational F. Variable readthrough responsiveness of non-
readthrough: a systems biology perspective. PLoS sense mutations in hemophilia A. Haematologica.
Genet. 2016;12(8):e1006196. 2020;105:508–18.
9. Namy O, Hatin I, Rousset JP. Impact of the six nucle- 23. Beier H, Grimm M. Misreading of termination codons
otides downstream of the stop codon on translation in eukaryotes by natural nonsense suppressor tRNAs.
termination. EMBO Rep. 2001;2:787–93. Nucleic Acids Res. 2001;29:4767–82.
10. Tate WP, Poole ES, Horsfield JA, Mannering SA, 24. Urban C, Zerfaß K, Fingerhut C, Beier H. UGA sup-
Brown CM, Moffat JG, Dalphin ME, McCaughan pression by tRNACmCATrp occurs in diverse virus
KK, Major LL, Wilson DN. Translational termination RNAs due to a limited influence of the codon context.
efficiency in both bacteria and mammals is regulated Nucleic Acids Res. 1996;24:3424–30.
by the base following the stop codon. Biochem Cell 25. Keeling KM, Wang D, Conard SE, Bedwell
Biol. 1995;73:1095–103. DM. Suppression of premature termination codons as
11. Manuvakhova M, Keeling K, Bedwell a therapeutic approach. Crit Rev Biochem Mol Biol.
DM. Aminoglycoside antibiotics mediate context-­ 2012;47:444–63.
dependent suppression of termination codons in a 26. Moosajee M, Ramsden SC, Black GC, Seabra MC,
mammalian translation system. RNA. 2000;6:1044–55. Webster AR. Clinical utility gene card for: choroider-
12. Cridge AG, Crowe-Mcauliffe C, Mathew SF, Tate emia. Eur J Hum Genet. 2014;22(4):572.
WP. Eukaryotic translational termination effi- 27. Dabrowski M, Bukowy-Bieryllo Z, Zietkiewicz
ciency is influenced by the 3′ nucleotides within E. Translational readthrough potential of natural ter-
the ribosomal mRNA channel. Nucleic Acids Res. mination codons in eucaryotes-the impact of RNA
2018;46:1927–44. sequence. RNA Biol. 2015;12:950–8.
Factors Affecting Readthrough of Natural Versus Premature Termination Codons 155

28. Rodnina MV, Korniy N, Klimova M, Karki P, Peng Bedwell DM, Welch EM, Jacobson A. Ataluren
BZ, Senyushkina T, Belardinelli R, Maracci C, stimulates ribosomal selection of near-cognate
Wohlgemuth I, Samatova E, Peske F. Translational tRNAs to promote nonsense suppression. PNAS.
recoding: canonical translation mechanisms reinter- 2016;113:12508–13.
preted. Nucleic Acids Res. 2020;48:1056–67. 34. Blanchet S, Rowe M, Von Der Haar T, Fabret C,
29. Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen Demais S, Howard MJ, Namy O. New insights into
WJ, Trifillis P, Paushkin S, Patel M, Trotta CR, stop codon recognition by eRF1. Nucleic Acids Res.
Hwang S, Wilde RG, Karp G, Takasugi J, Chen G, 2015;43:3298–308.
Jones S, Ren H, Moon Y-C, Corson D, Turpoff AA, 35. Dabrowski M, Bukowy-Bieryllo Z, Zietkiewicz
et al. PTC124 targets genetic disorders caused by non- E. Advances in therapeutic use of a drug-stimulated
sense mutations. Nature. 2007;447:87–91. translational readthrough of premature termination
30. Wangen JR, Green R. Stop codon context influ- codons. Mol Med BioMed Central Ltd. 2018;24.
ences genome-wide stimulation of termination 36. Zerfass K, Beier H. The leaky UGA termination codon
codon readthrough by aminoglycosides. elife. of tobacco rattle virus RNA is suppressed by tobacco
2020;9:e52611. chloroplast and cytoplasmic tRNAs(Trp) with CmCA
31. Feng YX, Copeland TD, Oroszlan S, Rein A, Levin anticodon. EMBO J. 1992;11:4167.
JG. Identification of amino acids inserted during sup- 37. Nilsson M, Rydén-Aulin M. Glutamine is incor-
pression of UAA and UGA termination codons at the porated at the nonsense codons UAG and UAA in
gag-pol junction of Moloney murine leukemia virus. a suppressor-­free Escherichia coli strain. Biochim
PNAS USA. 1990;87:8860–3. Biophys Acta. 2003;1627:1–6.
32. Urban C, Beier H. Cysteine tRNAs of plant ori- 38. Fearon K, Mcclendon V, Bonetti B, Bedwell
gin as novel UGA suppressors. Nucleic Acids Res. DM. Premature translation termination mutations
1995;23:4591–7. are efficiently suppressed in a highly conserved
33. Roy B, Friesen WJ, Tomizawa Y, Leszyk JD, region of yeast SteGp, a member of the ATP-binding
Zhuo J, Johnson B, Dakka J, Trotta CR, Xue X, cassette (ABC) transporter family. J Biol Chem.
Mutyam V, Keeling KM, Mobley JA, Rowe SM, 1994;269:17802–8.
Integrating Computational
Approaches to Predict the Effect
of Genetic Variants on Protein
Stability in Retinal Degenerative
Disease

Michelle Grunin, Ellen Palmer, Sarah de Jong,


Bowen Jin, David Rinker, Christopher Moth,
John A. Capra, Jonathan L. Haines,
William S. Bush, and Anneke I. den Hollander

Abstract can be affected by a large change in the ter-


tiary (3D) structure of the protein or due to
Protein function can be impacted by changes free-energy changes caused by single amino
in protein structure stability, but determining acid substitutions. Changes in the DNA
which change has impact is complex. Stability sequence can have minor or major impact on
protein stability, which can lead to disease.
Authors Michelle Grunin, Ellen Palmer, Sarah de Jong, Inherited retinal degenerations are generally
Jonathan L. Haines, William S. Bush, and Anneke I. den caused by single mutations which are mostly
Hollander have equally contributed to this chapter. located in protein-coding regions, while age-­

C. Moth
M. Grunin (*) · E. Palmer · B. Jin · J. L. Haines ·
Center for Structural Biology, Vanderbilt University,
W. S. Bush
Nashville, TN, USA
Department of Population and Quantitative Health
e-mail: chris.moth@vanderbilt.edu
Sciences, Case Western Reserve University,
Cleveland, OH, USA J. A. Capra
Department of Biological Sciences, Vanderbilt
Cleveland Institute for Computational Biology,
University, Nashville, TN, USA
Case Western Reserve University, Cleveland,
OH, USA Center for Structural Biology, Vanderbilt University,
e-mail: mag235@case.edu; elp76@case.edu; Nashville, TN, USA
bxj139@case.edu; jlh213@case.edu; wsb36@case.
Vanderbilt Genetics Institute, Vanderbilt University
edu
School of Medicine, Nashville, TN, USA
S. de Jong (*)
Department of Biomedical Informatics, Vanderbilt
Department of Ophthalmology, Donders
University School of Medicine, Nashville, TN, USA
Institute for Brain, Cognition, and Behavior, Radboud
e-mail: tony.capra@vanderbilt.edu
University Medical Center, Nijmegen, the
Netherlands A. I. den Hollander
e-mail: sarah.dejong@radboudumc.nl Department of Ophthalmology, Donders Institute for
Brain, Cognition, and Behavior, Radboud University
D. Rinker
Medical Center, Nijmegen, the Netherlands
Department of Biological Sciences,
Vanderbilt University, Nashville, Department of Human Genetics, Radboud University
TN, USA Medical Center, Nijmegen, the Netherlands
e-mail: david.rinker@vanderbilt.edu e-mail: Anneke.denHollander@radboudumc.nl

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 157
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_24
158 M. Grunin et al.

related macular degeneration (AMD) is a tions are still unknown, while this may in part be
complex disorder that can be influenced by due to the ambiguous classification of genetic
some genetic variants impacting proteins variants [7]. As sequencing-based genetic studies
involved in the disease, although not all AMD increase the number of identified protein-altering
risk variants lead to amino acid changes. Here, mutations, more work in this area will be critical
we review ways that proteins may be affected, to understand their role in complex diseases[8]
the identification and understanding of these and to discriminate between harmless and more
changes, and how to identify causal changes damaging VUS.
that can be targeted to develop treatments to Missense variants comprise over 60% of all
alleviate retinal degenerative disease. known monogenic disease mutations [9], but they
disrupt protein structure and function in various,
Keywords sometimes unclear, ways [10]. Changes to alpha
Proteins · Age-related macular degeneration · helices or beta sheets are the most clearly under-
Mutations · Variants · Free-energy stood, as they generally impact protein stability
by changing hydrophobicity that affects protein
folding, like mutations in TGFb or Pim1 in can-
cer [9, 11, 12]. Amino acid changes in other loca-
1 Part I. Protein Stability: Role tions may affect the stability of a protein or
and Importance protein complex [6] through changing the tertiary
structure of the protein or through changing the
Multiple efforts are underway to gather informa- free energy needed to fold into a functional form.
tion on clinically meaningful mutations in pro- Depending on the context of the single amino
tein coding genes in databases such as the Human acid change, protein folding (i.e., free-energy
Gene Mutation Database (HGMD) [1, 2] or change) can be impacted enough to prevent the
ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/) formation of structural motifs needed for critical
[3]. In 2003, HGMD contained more than 1000 function [6]. However, directly measuring free-­
mutations that were directly causative of disease; energy changes resulting from an amino acid
the number of reported mutations currently in substitution is a difficult experimental task
HGMD has grown to 323,661 as of Oct. 2021. because current approaches typically are expen-
ClinVar currently contains 1,159,307 unique sive, are time consuming, are performed on a
disease-­contributing variations (Oct. 16, 2021). single mutation at a time, and are hard to scale. In
Many of the mutations found in these databases, lieu of directly measuring experimental changes
but not all, can affect protein stability [4–6]. The in free-energy, computational methods are often
deleterious impact of mutations that clearly affect employed [13]. These methods typically compare
a protein’s sequence, fold, and function is some- wild-type and mutant protein folding using a
times obvious, such as frameshift mutations that solved template protein structure from the pro-
alter much of the protein sequence. More often, tein data bank (PDB, https://www.rcsb.org/) and
missense mutations are identified, such as single-­ then changing those structures to model the
point mutations that cause some familial forms of mutant protein using programs like PoPMusic
Alzheimer’s or Parkinson’s disease. Missense [14] or Phyre [15]. The free energy before and
mutations are generally more difficult to classify after folding is calculated for both the wild-type
and understand, and newly discovered variants and mutant proteins to produce two respective
may be given an ambiguous classification free-energy change measures (ΔG). These mea-
(referred to as “variants of unknown signifi- sures are then compared to estimate a change in
cance”; VUS), as these changes may alter protein ΔG due to the amino acid substitution (ΔΔG).
structure at different, hard to measure levels (e.g., Both experimentally solved and high-resolution
tertiary structure). An excellent example is retini- computational models of protein structures like
tis pigmentosa, as 60% of disease-causing muta- those found in the PDB can be used for accurate
Integrating Computational Approaches to Predict the Effect of Genetic Variants on Protein Stability… 159

folding estimates [16, 17]. PDB contains 183,386 often severe Mendelian diseases. Some
structures in 2021, which were either determined approaches and programs either calculate free-­
experimentally or by homology modeling based energy changes and take the free-energy changes
on similar protein structures [18]. As of 2020, into account when evaluating protein stability,
14,028 solved and proven experimentally struc- like I-Mutant2.0 (http://gpcr2.biocomp.unibo.
tures from the 183,386 were included in the it/%7Eemidio/I-­Mutatnt/I-­Mutant.htm) [30],
PDB. The sequence based or ab initio approaches SDM (http://www-­cryst.bioc.cam.ac.uk/~sdm/
for identifying impactful variants are faster but sdm.php) [31], CUPSAT (http://cupsat.tu-­bs.de/)
depend on machine learning training sets and do [32], FoldX (http://fold-­x.embl-­heidelberg.de)
not focus on the ultimate impact on the full post-­ [33], ROSETTA (https://www.rosettacommons.
secondary protein structure, utilizing protein org/) [34], or AUTO-MUTE (http://proteins.gmu.
modeling, while the structure-based approaches edu/automute) [35]. However, only few programs
use multiple scoring methods as well as intensive take into account both information from tertiary
calculations, like the free-energy perturbation structures and the impact of free-energy change
(FEP) and thermodynamic integration (TI) meth- on the protein structure. Only PolyPhen,
ods [19, 20]. I-Mutant2.0, and HOPE (https://www3.cmbi.
Computationally, protein homology models umcn.nl/hope/about/) [36] consider both
can be constructed using methods like the Swiss-­ sequence and protein structure changes but
Model in the ExPASy webserver [21]. Proteins include these among many other features in a
can be graphed using Mol [22]or other programs machine learning prediction based on highly pen-
to visualize exactly where the structure changes etrant missense variants[6]. A comparison
and where the variant impacts the protein struc- between the different methods used to estimate
ture. Variants that impact the free-energy change the effect of different mutations on protein stabil-
in binding sites or core regions may be the most ity has been described in work of de Groot [37],
damaging, even if the change is not obviously Thiltgen and Goldstein [38], and Kroncke [39].
damaging to the tertiary structure of the protein.
Mutations that destabilize proteins are described
for von Willebrand diseases [23], prion [24], and 3 Part III. New Methodologies:
retinal degenerative diseases that impact rhodop- PathProx and POKEMON
sin [25]. Depending on protein function, some
residue substitutions cause disease by increasing PathProx [16, 17] employs an alternative
protein stability, such as the CLIC2 protein in approach: it specifically focuses on the spatial
some mental disorders [26]. context and stability of the full three-dimensional
protein. PathProx performs two types of analy-
ses: (1) PathProx evaluates the spatial proximity
2 Part II. Common of input variants to known pathogenic variants
Computational Methods (from resources like ClinVar) and to presumed
for Determining Variant neutral variants within the protein; (2) PathProx
Impact calculates differences in the free-energy and sta-
bility of the protein utilizing ROSETTA and
Machine learning algorithms can assess the examines those variants in relationship to other
impact of missense variants using a variety of known pathogenic and presumably neutral vari-
information including protein sequence and in ants that may be present in the protein structure.
some cases, structure and folding. Programs like The spatial proximity portion of the algorithm
SIFT (http://sift-­dna.org) [27], MutPred (http:// uses Euclidean distance to determine if the input
mutpred.putdb.org) [28], or Polyphen2 (http:// variants are nearer to pathogenic risk variants (or
genetics.bwh.harvard.ed/pphw) [29] are trained any variant of interest), as compared to “normal”
using a set of known deleterious mutations for or neutral variants, such as those found in data-
160 M. Grunin et al.

bases like GnomAD, which draws on over 4 Part IV. Retinal Diseases,
141,456 samples from the controls of dozens of AMD, and Protein Stability
disease studies (https://gnomad.broadinstitute.
org/) [40]. Variants that are found closer to known Multiple retinal degenerative diseases are caused
pathogenic variants and that are enriched in cases by mutations that affect protein stability [42].
(when case-control data is used) could indicate One of the best known are rhodopsin mutations,
that a particular protein region plays a significant the most common cause of autosomal dominant
role in disease pathogenesis, whereas variants retinitis pigmentosa [43].The first identified
that are found near neutral variants and are more mutations inhibit protein stability that either
often found in controls or are evenly distributed reduces its export out of the endoplasmic reticu-
between cases and controls are likely neutral or lum (ER) (Class II mutations) [44] or increases
perhaps “protective” variants. The ROSETTA-­ accumulation inside the cell (Class III mutations)
based portion of the method uses free-energy cal- [45]. The majority of these mutations lead to
culations to identify variants that are predicted to folding defects of the protein [45]. Class II muta-
impact the structure of a protein associated with a tions also impact tertiary folding stability [46].
given disease. Together, these two approaches These mutations are common among the G
within PathProx create a candidate variant list for protein-­coupled receptors (GPCRs), but how
further case enrichment, gene-based testing, and their thermodynamics are impacted by folding
functional testing at the bench. Paired together, stability is still not fully understood. Therefore,
these approaches allow for the identification of computational methods to understand the effect
two different patterns of protein-altering of these mutations is the avenue that has been
relationships. most explored [43].
POKEMON [41] adapts gene-based testing Investigation of VUS has been a major focus
methods employing kernel functions (such as in genetic research, and the combination of both
SKAT) to include information about missense computational and functional biology approaches
variant proximity within the 3D structure of a has been useful in identifying the impact of
protein. Using this kernel, a statistical test can be VUSs. If computational approaches can differen-
conducted to determine if the spatial proximity of tiate variants of significance from among the
variants within the protein is related to case sta- VUSs, specifically those that have functional
tus. For example, a pocket of missense variants effect, this would significantly improve variant
found more frequently in cases within one par- interpretation in diagnostic testing. For rhodop-
ticular area of a protein may point to a segment of sin, variant interpretation has been performed
the protein that plays a role in disease. That area using a combination of computational and exper-
may be important as a binding site, or impact pro- imental approaches: gain of function mutations
tein stability at the weakest point, contributing to have systemically been analyzed using a compu-
disease pathogenesis. Results from POKEMON tational approach, in addition to a full-scale
and the spatial-clustering analyses of PathProx experimental screen to evaluate rhodopsin
have been found to be concordant in a study of expression in cells [47]. Two-stage approaches
Alzheimer’s disease and provide orthogonal using computational methods before moving to
methods of identifying disease-associated pro- functional testing have been applied, for exam-
tein regions (unpublished data, 2021). We ple, in Best disease, to determine destabilizing
recently utilized these two methods to identify mutations using I-mutant and then performing
variants that can have a functional impact on pro- functional testing on all variants [48]. Many tests
tein stability and expression of complement fac- have shown that reliable first-stage computa-
tors in AMD (Grunin, Palmer, de Jong et al., tional testing of missense mutations can be done
unpublished). through I-Mutant, Dmutant, and FoldX [49].
Integrating Computational Approaches to Predict the Effect of Genetic Variants on Protein Stability… 161

However, a two-stage approach can be used to protein stability and may provide new avenues
first predict the effect of a variant computation- for identifying treatment-amenable variants with-
ally and then focus experimental work on vari- out the need to bench test every single mutation.
ants that are predicted to affect protein stability In addition, free-energy changes can be utilized
or function, which would narrow the testing pool, to predict the consequences that these variants
time, and costs [50]. In AMD, one of the most might have on the protein of interest. Reliable
studied proteins is complement factor H (CFH). predictive testing allows the identification of
Rare protein-coding AMD risk variants have variants of interest in disease more rapidly and
been mapped on the protein with corresponding allows for targeted functional testing.
issues of protein unfolding or incorrect folding,
with 70% of mutations showing a destabilizing
effect. Recently, 105 variants in CFH were classi- References
fied according to their pathogenicity and effect
on function utilizing functional assays [51]. 1. Stenson PD, Ball E V., Mort M, et al. Human Gene
Mutation Database (HGMD®): 2003 updateHum
Several other proteins of the complement Mutat;21. https://doi.org/10.1002/humu.10212. Epub
pathway are associated with AMD including C3, ahead of print 2003.
C9, CFB, and CFI [52]. Rare protein-coding vari- 2. Stenson PD, Mort M, Ball E V., et al. The Human Gene
ants in these complement genes have been asso- Mutation Database: towards a comprehensive reposi-
tory of inherited mutation data for medical research,
ciated with differences in protein concentration genetic diagnosis and next-generation sequencing
in patients and controls [53–57]. However, in studies. Hum Genet;136. https://doi.org/10.1007/
AMD, a combination of common variants with s00439-­017-­1779-­6. Epub ahead of print 2017.
modest effect and rare variants with large effect 3. Landrum MJ, Lee JM, Benson M, et al. ClinVar:
improving access to variant interpretations and sup-
collectively contribute to the disease, and how porting evidence. Nucleic Acids Res;46. https://doi.
these variants contribute to AMD and whether org/10.1093/nar/gkx1153. Epub ahead of print 2018.
they have synergistic effects are currently 4. Wang Z, Moult J. SNPs, protein structure, and disease.
unknown. Analysis of variants in RPE65 and rho- Hum Mutat;17. https://doi.org/10.1002/humu.22.
Epub ahead of print 2001.
dopsin have shown similar results in destabiliza- 5. Casadio R, Vassura M, Tiwari S, et al. Correlating
tion of the protein, utilizing an unfolding mutation disease-­related mutations to their effect on protein sta-
screen (UMS) [50]. However, these results were bility: a large-scale analysis of the human proteome.
not followed up by actual functional testing on Hum Mutat;32. https://doi.org/10.1002/humu.21555.
Epub ahead of print 2011.
those mutations, because the mutations were 6. Stefl S, Nishi H, Petukh M, et al. Molecular mecha-
already known, and thus the method did not show nisms of disease-causing missense mutations. J Mol
predictive power. We recently applied computa- Biol;425. https://doi.org/10.1016/j.jmb.2013.07.014.
tional methodology utilizing PathProx and Epub ahead of print 2013.
7. González-Del Pozo M, Fernández-Suárez E, Martín-­
POKEMON to patients with AMD and identified Sánchez M, et al. Unmasking Retinitis Pigmentosa
variants that impact proteins in the complement complex cases by a whole genome sequencing
system. These variants were located in unique algorithm based on open-access tools: hidden reces-
spatial locations in the protein and lead to distinct sive inheritance and potential oligogenic variants. J
Transl Med;18. https://doi.org/10.1186/s12967-­020-­
free-energy changes. We determined through 02258-­3. Epub ahead of print 2020.
computational mining which variants cause an 8. Capriotti E, Calabrese R, Casadio R. Predicting
in vitro change in complement protein expres- the insurgence of human genetic diseases associ-
sion. Therefore, the application of computational ated to single point protein mutations with sup-
port vector machines and evolutionary information.
filtering allows us to identify variants that have a Bioinformatics;22. https://doi.org/10.1093/bioinfor-
foreseeable functional impact (Grunin, Palmer, matics/btl423. Epub ahead of print 2006.
deJong, unpublished data, 2021) 9. Yue P, Li Z, Moult J. Loss of protein structure sta-
In conclusion, utilizing novel methods like bility as a major causative factor in monogenic dis-
ease. J Mol Biol;353. https://doi.org/10.1016/j.
PathProx and POKEMON will enable under- jmb.2005.08.020. Epub ahead of print 2005.
standing of which variants are likely to affect
162 M. Grunin et al.

10. Calamini B, Lo DC, Kaltenbach LS. Experimental 23. Xu AJ, Springer TA. Mechanisms by which von wil-
models for identifying modifiers of polyglutamine-­ lebrand disease mutations destabilize the A2 domain.
induced aggregation and neurodegeneration. J Biol Chem;288. https://doi.org/10.1074/jbc.
Neurotherapeutics;10. https://doi.org/10.1007/ M112.422618. Epub ahead of print 2013.
s13311-­013-­0195-­4. Epub ahead of print 2013. 24. Jetha NN, Semenchenko V, Wishart DS, et al.
11. Kucukkal TG, Alexov E. Structural, dynamical, and Nanopore analysis of wild-type and mutant prion pro-
energetical consequences of RETT syndrome muta- tein (PrPC): single molecule discrimination and PrPC
tion R133c in MeCP2. Comput Math Methods Med. kinetics. PLoS One;8. https://doi.org/10.1371/journal.
2015. https://doi.org/10.1155/2015/746157. Epub pone.0054982. Epub ahead of print 2013.
ahead of print 2015. 25. Rakoczy EP, Kiel C, McKeone R, et al. Analysis of
12. Petukh M, Dai L, Alexov E. SAAMBE: webserver to disease-linked rhodopsin mutations based on struc-
predict the charge of binding free energy caused by ture, function, and protein stability calculations. J Mol
amino acids mutations. Int J Mol Sci;17. https://doi. Biol;405. https://doi.org/10.1016/j.jmb.2010.11.003.
org/10.3390/ijms17040547. Epub ahead of print 2016. Epub ahead of print 2011.
13. Magliery TJ. Protein stability: computation, 26. Witham S, Takano K, Schwartz C, et al. A missense
sequence statistics, and new experimental methods. mutation in CLIC2 associated with intellectual dis-
Curr Opin Struct Biol;33. https://doi.org/10.1016/j. ability is predicted by in silico modeling to affect
sbi.2015.09.002. Epub ahead of print 2015. protein stability and dynamics. Proteins Struct Funct
14. Dehouck Y, Kwasigroch JM, Gilis D, et al. Bioinf;79. https://doi.org/10.1002/prot.23065. Epub
PoPMuSiC 2.1: a web server for the estimation ahead of print 2011.
of protein stability changes upon mutation and 27. Sim NL, Kumar P, Hu J, et al. SIFT web server: pre-
sequence optimality. BMC Bioinf;12. https://doi. dicting effects of amino acid substitutions on proteins.
org/10.1186/1471-­2105-­12-­151. Epub ahead of Nucleic Acids Res;40 https://doi.org/10.1093/nar/
print 2011. gks539. . Epub ahead of print 2012.
15. Kelley LA, Mezulis S, Yates CM, et al. The Phyre2 28. Pejaver V, Urresti J, Lugo-Martinez J, et al. Inferring
web portal for protein modeling, prediction and anal- the molecular and phenotypic impact of amino acid
ysis. Nat Protoc. 2015;10:845–58. variants with MutPred2. Nat Commun;11. https://
16. Sivley RM, Sheehan JH, Kropski JA, et al. Three-­ doi.org/10.1038/s41467-­020-­19669-­x. Epub ahead of
dimensional spatial analysis of missense variants in print 2020.
RTEL1 identifies pathogenic variants in patients with 29. Adzhubei IA, Schmidt S, Peshkin L, et al. A method
Familial Interstitial Pneumonia. BMC Bioinf;19. and server for predicting damaging missense muta-
https://doi.org/10.1186/s12859-­018-­2010-­z. Epub tions. Nat Methods;7. https://doi.org/10.1038/
ahead of print 2018. nmeth0410-­248. Epub ahead of print 2010.
17. Sivley RM, Dou X, Meiler J, et al. Comprehensive 30. Capriotti E, Fariselli P, Casadio R. I-Mutant2.0: pre-
analysis of constraint on the spatial distribution dicting stability changes upon mutation from the
of missense variants in human protein structures. protein sequence or structure. Nucleic Acids Res;33.
Am J Hum Genet;102. https://doi.org/10.1016/j. https://doi.org/10.1093/nar/gki375. Epub ahead of
ajhg.2018.01.017. Epub ahead of print 2018. print 2005.
18. Berman HM, Battistuz T, Bhat TN, et al. The protein 31. Pandurangan AP, Ochoa-Montaño B, Ascher DB,
data bank Acta Crystallogr Sect D Biol Crystallogr;58. et al. SDM: a server for predicting effects of mutations
https://doi.org/10.1107/S0907444902003451. Epub on protein stability. Nucleic Acids Res;45. https://doi.
ahead of print 2002. org/10.1093/nar/gkx439. Epub ahead of print 2017.
19. Petukh M, Kucukkal TG, Alexov E. On human 32. Parthiban V, Gromiha MM, Schomburg D. CUPSAT:
disease-­causing amino acid variants: Statistical study prediction of protein stability upon point mutations.
of sequence and structural patterns. Hum Mutat;36. Nucleic Acids Res;34. https://doi.org/10.1093/nar/
https://doi.org/10.1002/humu.22770. Epub ahead of gkl190. Epub ahead of print 2006.
print 2015. 33. Schymkowitz J, Borg J, Stricher F, et al. The FoldX
20. Michel J, Foloppe N, Essex JW. Rigorous free web server: an online force field. Nucleic Acids
energy calculations in structure-based drug design Res;33. https://doi.org/10.1093/nar/gki387. Epub
Mol Inf;29. https://doi.org/10.1002/minf.201000051. ahead of print 2005.
Epub ahead of print 2010. 34. Alford RF, Leaver-Fay A, Jeliazkov JR, et al. The
21. Waterhouse A, Bertoni M, Bienert S, et al. SWISS-­ Rosetta all-atom energy function for macromolecu-
MODEL: homology modelling of protein structures lar modeling and design. J Chem Theory Comput;13.
and complexes. Nucleic Acids Res;46. https://doi. https://doi.org/10.1021/acs.jctc.7b00125. Epub ahead
org/10.1093/nar/gky427. Epub ahead of print 2018. of print 2017.
22. Sehnal D, Bittrich S, Deshpande M, et al. Mol∗Viewer: 35. Masso M, Vaisman II. AUTO-MUTE 2.0: a portable
modern web app for 3D visualization and analysis of framework with enhanced capabilities for predicting
large biomolecular structures. Nucleic Acids Res;49. protein functional consequences upon mutation. Adv
https://doi.org/10.1093/nar/gkab314. Epub ahead of Bioinf. 2014. https://doi.org/10.1155/2014/278385.
print 2021. Epub ahead of print 2014.
Integrating Computational Approaches to Predict the Effect of Genetic Variants on Protein Stability… 163

36. Venselaar H, te Beek TAH, Kuipers RKP, et al. 47. Wan A, Place E, Pierce EA, et al. Characterizing vari-
Protein structure analysis of mutations causing inher- ants of unknown significance in rhodopsin: A func-
itable diseases. An e-Science approach with life sci- tional genomics approach. Hum Mutat;40. https://doi.
entist friendly interfaces. BMC Bioinf;11. https://doi. org/10.1002/humu.23762. Epub ahead of print 2019.
org/10.1186/1471-­2105-­11-­548. Epub ahead of print 48. Milenkovic A, Milenkovic VM, Wetzel CH, et al.
2010. BEST1 protein stability and degradation pathways
37. De Groot BL, Van Aalten DMF, Scheek RM, et al. differ between autosomal dominant Best disease and
Prediction of protein conformational freedom autosomal recessive bestrophinopathy accounting for
from distance constraints. Proteins Struct Funct the distinct retinal phenotypes. Hum Mol Genet;27.
Genet;29. https://doi.org/10.1002/(SICI)1097-­ https://doi.org/10.1093/hmg/ddy070. Epub ahead of
0134(199710)29:2<240::AID-­PROT11>3.0.CO;2-­O. print 2018.
Epub ahead of print 1997. 49. Khan S, Vihinen M. Performance of protein stability
38. Thiltgen G, Goldstein RA. Assessing predictors of predictors. Hum Mutat;31. https://doi.org/10.1002/
changes in protein stability upon mutation using self-­ humu.21242. Epub ahead of print 2010.
consistency. PLoS One;7. https://doi.org/10.1371/ 50. McCafferty CL, Sergeev Y V. In silico mapping of
journal.pone.0046084. Epub ahead of print 2012. protein unfolding mutations for inherited disease.
39. Kroncke BM, Duran AM, Mendenhall JL, et al. Sci Rep;6. https://doi.org/10.1038/srep37298. Epub
Documentation of an imperative to improve ahead of print 2016.
methods for predicting membrane protein stabil- 51. Martin Merinero H, Zhang Y, Arjona E, et al.
ity. Biochemistry;55. https://doi.org/10.1021/acs. Functional characterization of 105 Factor H vari-
biochem.6b00537. Epub ahead of print 2016. ants associated with atypical HUS: lessons for vari-
40. Karczewski KJ, Francioli LC, Tiao G, et al. The muta- ant classification. Blood. https://doi.org/10.1182/
tional constraint spectrum quantified from variation in blood.2021012037. Epub ahead of print 2021.
141,456 humans. Nature;581. https://doi.org/10.1038/ 52. Fritsche LG, Igl W, Bailey JNC, et al. A large genome-­
s41586-­020-­2308-­7. Epub ahead of print 2020. wide association study of age-related macular degen-
41. Jin B, Capra JA, Benchek P, et al. An association eration highlights contributions of rare and common
test of the spatial distribution of rare missense vari- variants. Nat Genet;48. https://doi.org/10.1038/
ants within protein structures improves statistical ng.3448. Epub ahead of print 2016.
power of sequencing studies. bioRxiv. https://doi. 53. Hallam TM, Marchbank KJ, Harris CL, et al. Rare
org/10.1101/2021.08.09.455695 genetic variants in complement factor I lead to low
42. Tzekov R, Stein L, Kausha S. Protein misfolding FI plasma levels resulting in increased risk of age-­
and retinal degeneration Cold Spring Harb Perspect related macular degeneration. Investig Ophthalmol
Biol;3. https://doi.org/10.1101/cshperspect.a007492. Vis Sci;61. https://doi.org/10.1167/IOVS.61.6.18.
Epub ahead of print 2011. Epub ahead of print 2020.
43. Marinko JT, Huang H, Penn WD, et al. Folding and 54. Kremlitzka M, Geerlings MJ, De Jong S, et al.
misfolding of human membrane proteins in health Functional analyses of rare genetic variants in com-
and disease: from single molecules to cellular pro- plement component C9 identified in patients with age-­
teostasis. Chem Rev;119. https://doi.org/10.1021/acs. related macular degeneration. Hum Mol Genet;27.
chemrev.8b00532. Epub ahead of print 2019. https://doi.org/10.1093/hmg/ddy178. Epub ahead of
44. Sung CH, Davenport CM, Hennessey JC, et al. print 2018.
Rhodopsin mutations in autosomal dominant retinitis 55. Geerlings MJ, Kersten E, Groenewoud JMM, et al.
pigmentosa. Proc Natl Acad Sci U S A;88. https://doi. Geographic distribution of rare variants associated
org/10.1073/pnas.88.15.6481. Epub ahead of print with age- related macular degeneration. Mol Vis.
1991. 2018;9:75–82.
45. Kaushal S, Khorana HG. Structure and function in 56. de Jong S, Gagliardi G, Garanto A, et al. Implications
rhodopsin. 7. Point mutations associated with autoso- of genetic variation in the complement system in age-­
mal dominant retinitis pigmentosa. Biochemistry;33. related macular degeneration. Prog Retin Eye Res;84.
https://doi.org/10.1021/bi00186a011. Epub ahead of https://doi.org/10.1016/j.preteyeres.2021.100952.
print 1994. Epub ahead of print 2021.
46. Rader AJ, Anderson G, Isin B, et al. Identification 57. de Jong S, Volokhina EB, de Breuk A, et al. Effect
of core amino acids stabilizing rhodopsin. Proc of rare coding variants in the CFI gene on factor I
Natl Acad Sci U S A;101. https://doi.org/10.1073/ expression levels. Hum Mol Genet;29. https://doi.
pnas.0401429101. Epub ahead of print 2004. org/10.1093/hmg/ddaa114. Epub ahead of print 2020.
Network Biology and Medicine
to Rescue: Applications for Retinal
Disease Mechanisms and Therapy

Anupam K. Mondal and Anand Swaroop

Abstract developments, especially gene agnostic treat-


ment opportunities.
Inherited retinal degenerations (IRDs) are
clinically and genetically heterogenous blind- Keywords
ing diseases that manifest through dysfunction
of target cells, photoreceptors, and retinal pig- Inherited retinal degeneration · Network
ment epithelium (RPE) in the retina. Despite medicine · Systems biology · Diseasome ·
knowledge of numerous underlying genetic Neurodegeneration
defects, current therapeutic approaches,
including gene centric applications, have had
limited success, thereby asserting the need of 1 Introduction
new directions for basic and translational
research. Human diseases have commonalities Inherited retinal degenerations (IRDs) manifest
that can be represented in a network form, through untimely dysfunction and degeneration
called diseasome, which captures relation- of the retina causing permanent vision loss.
ships among disease genes, proteins, metabo- Genetic etiology of retinal neurodegeneration is
lites, and patient meta-data. Clinical and remarkably diverse. IRDs arise from Mendelian
genetic information of IRDs suggest shared inheritance of pathogenic mutations in one of at
relationships among pathobiological factors, least 300 genes [1], though involvement of more
making these a model case for network medi- than one gene and modifier genes have been
cine. Characterization of the diseasome would implicated in producing a clinical phenotype [2].
considerably improve our understanding of In contrast, age-related macular degeneration
retinal pathologies and permit better design of (AMD), glaucoma, and diabetic retinopathy
targeted therapies for disrupted regions within (DR) are complex multifactorial pathologies,
the integrated disease network. Network med- where a multitude of factors including genetics,
icine in synergy with the ongoing artificial environment, diet, and other health conditions
intelligence revolution can boost therapeutic contribute to disease risk [3]. IRDs present as a
spectrum of phenotypes, and appropriate clinical
A. K. Mondal (*) · A. Swaroop management depends on accurate prognosis or
Neurobiology, Neurodegeneration & Repair diagnosis, pathology forecast, drug development,
Laboratory, National Eye Institute, National Institutes and treatment. However, IRDs remain generally
of Health, Bethesda, MD, USA untreatable despite identification of many disease
e-mail: mondalak@nih.gov

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 165
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_25
166 A. K. Mondal and A. Swaroop

genes. Treatments with gene replacement therapy ogy or network medicine, as this specialty is now
would be ideal for recessive IRD-like diseases broadly referred to, is an exciting new way of
exhibiting loss of function phenotypes, but in unraveling disease complexity through network
practice challenges are manifold. At this stage, science, multi-omic data integration, and machine
only one regimen has FDA approval, while few intelligence [15, 16]. Network medicine has been
other candidates are at various stages of develop- particularly insightful through creation of the
ment and clinical trials [4, 5]. Overall, gene ther- human disease network, the “diseasome,” which
apy has proven cumbersome and expensive, has led to realizations that nearly two-third of all
notwithstanding the complexity in designing pathologies might share comparable genetic
gene therapies for the huge diversity of genetic inceptions, while a majority of disease genes co-­
mutations and disease genes associated with occur in pathologies [17]. Network medicine has
IRDs. This has gravitated researchers to gene-­ guided improved understanding of neurological
agnostic methods that have shown promising pre- diseases [18, 19] as well as notable advances in
liminary outcomes. Recently, a disease COVID-19 research among others [20–24].
gene-independent optogenetics-based vision Artificial intelligence is undergoing revolu-
augmentation technology was found to provide tionary changes and is projected to have consid-
visual feedback in a patient with severe blindness erable impact on the biomedical research
[6]. Stem cell-derived photoreceptor and RPE community [25, 26]. Ophthalmological scientists
replacement provide potentially interesting have been at the forefront of using AI; however,
approaches [7]; however, much remains to be its use has remained confined to medical image
done especially with respect to cellular and func- analyses of specific diseases [27]. AI has tremen-
tional integration of grafted cells into host retina dous potential in retinal disease research, particu-
with extensive degeneration. The applicability of larly for biomarker detection and drug discovery
gene agnostic therapies to more than one pathol- [28, 29]. In this book chapter, we discuss the cur-
ogy makes these an arguably better treatment rent advances and future prospect of network
alternative for IRDs [8, 9]. Nevertheless, success medicine and artificial intelligence as applicable
of translational research depends on knowledge to retinal disease, especially IRD, research.
of molecular etiology, and retinal degenerative
disease mechanisms remain poorly understood.
Systems biology approaches in the post-­ 2 Molecular Genetics of IRDs
genomic era have guided unprecedented insights
into biomolecular interactions underlying human IRDs constitute a broad family of retinal dys-
health and diseases. Advent of high-throughput functions and dystrophies, with estimated prev-
large data analytics and their subsequent adop- alence of ~1 in 3000 individuals and affecting
tion in biology and medicine have launched novel more than two million people globally [30, 31].
methodological advances that have defied tradi- Inheritance is generally monogenic; however,
tional “one-gene one-disease” research and genotype-phenotype correlations have been
instead banked on integrating molecular land- extremely difficult to establish [32]. In some
scapes to offer a “bird’s eye view” of cellular sys- instances, di- or multi-genic inheritance pat-
tems [10]. These strategies have been terns have been noted [2], but modifier genes
transformative for studying protein-protein inter- have also been implicated for generating spe-
actions, metabolic simulations, cellular signal- cific phenotypic variability [33]. To date, more
ing, and gene regulation, among others [11–14]. than 10,000 mutations in nearly 300 genes have
A notable feature in this movement has been the been attributed to IRDs, following autosomal-
use of graph theory to capture biomolecular recessive, autosomal-dominant, X-linked, or
interactions in network graphs that can be ana- mitochondrial patterns of inheritance [1].
lyzed using innovative algorithms. Network biol- Recent large-scale genomic studies have esti-
Network Biology and Medicine to Rescue: Applications for Retinal Disease Mechanisms and Therapy 167

mated that pathogenic mutations related with 3 Network Medicine


IRDs are prevalent at a rate of 1 in 3 individuals and Healthcare
[30, 32]. This presents an interesting notion that
retinal disease mutations are more common The genomics era has brought revolutionary
than previously predicted. Although IRD muta- changes in biomedical research. In this age of data
tions affect a few retinal cell classes, photore- intense research, an important distinction is a
ceptors and particularly rods are the most departure from the reductionist approach in favor
frequent targets [34, 35]. However, estimates of a top-down scheme to study comprehensive
suggest nearly 30–50% clinical cases remain molecular interactions regulating health and dis-
unresolved, indicating that the genetic basis of ease. This has been made possible by technologi-
IRD is yet to be comprehensively characterized cal and conceptual innovations that are now a
[32]. Clinically, retinal degeneration can begin mainstay in contemporary research. A pivotal
at various stages of human lifetime. Leber con- application in this movement has been the use of
genital amaurosis (LCA) is when retinal degen- networks to integrate biomolecular interactions at
eration initiates at birth or during early systems resolution [11]. Network graphs are natu-
childhood. Most adult-onset pathologies rally suited to represent biological relationships,
marked by early rod dysfunction and night and assessment of biological networks using graph
blindness are called retinitis pigmentosa (RP). theory tools have uncovered valuable knowledge
In many cases, photoreceptors of the human of the complex biology of health and disease [36].
central retina (i.e., macula) undergo dystrophy
and thus contribute to macular degeneration Network Medicine A fundamental assumption
(MD). An example is Stargardt disease, a form of network medicine is that pathology is estab-
of early onset MD, where the ABCA4 gene is lished in the finite space of interactome, and
the site of mutations that cause central vision therefore diseases can be predicted to have shared
loss [34]. Juvenile onset MD is distinct from mechanistic components [15, 36]. The human
age-related macular degeneration (AMD), a diseasome and the human symptoms disease net-
multifactorial disease prevalent in older indi- work studies uncovered notable molecular and
viduals. Genetic variants in a number of IRD phenotypic commonalities among diseases [17,
genes can also contribute to AMD, such as in 37]. Cellular homeostasis is preserved by thou-
case of ABCA4 and TIMP3 [1]. Additionally, sands of spatio-temporally regulated meaningful
depending on the disease gene involved, IRD biomolecular interactions. Pathogenic mutations
mutations can have extraocular phenotypes, cause alterations in the expression or activity of
i.e., a syndromic pathology [34]. However, proteins coded by the disease gene; however,
IRDs have interesting genetic etiologies. these abnormalities propagate in the interactome
Depending on the mutation, disease genes can through pathways and functional neighborhoods
cause different pathologies. Genes like ABCA4, of the target protein. In this context, disease onset
PROM1, and PRPH2 are implicated in distinct can be imagined as a result of increasing aberra-
clinical pathologies, while many others are tions in homeostatic molecular events. Network
linked to a single disease. On the other hand, analysis has shown that disease factors have dis-
similar IRD phenotypes can arise from muta- tinct placement in the molecular interactome and
tions in different genes. These associations are can be found to cluster into disease modules [17].
suggestive of shared molecular events deter- Network-based approaches aim to capture these
mining disease onset (Fig. 1a). Therefore, elu- trends in graph structures to create browsable in
cidating the retinal diseasome, i.e., the silico models and have created opportunities
interaction framework of retinal pathologies, where mapping and characterization of disease
disease genes, pathways, and other molecular modules can identify additional operators of
events, would be a step toward holistic under- pathology, which in turn could inspire transla-
standing of IRDs. tional outcomes [38].
168 A. K. Mondal and A. Swaroop

Fig. 1 Integrative biology for retinal disease research. (a) affect the host gene (genes X and Y) and alter immediate
Shared genetic origin of IRDs. Nodes represent IRD interactions (early impact); however, disease onset hap-
pathologies (blue) or disease genes (grey) listed in RetNet pens when functional dysregulation propagates (late
[1], while edges connect disease genes to associated impact). In this scheme, influence from different genes
pathologies. Distinct IRD pathologies can occur from can be imagined to converge at common nodes, poten-
mutations in overlapping genes, whereas a particular IRD tially accounting for overlapping molecular phenotype.
phenotype can be an outcome of mutations in several dif- (c) An integrative framework to model the retinal disea-
ferent genes. This network suggests commonalities in some. Characterizing the retinal diseasome will improve
molecular phenotype underlying IRD pathogenesis. (b) our understanding of pathology mechanisms and promote
Illustration of IRD disease genes and temporally impacted new therapeutic developments
pathways in the cellular interactome. Individual mutations

The Artificial Intelligence (AI) Revolution and driven hypotheses for laboratory investigations
a New Paradigm for Medicine Availability of [43]. Furthermore, network medicine in synergy
vast amounts of diverse biological and clinical with AI can remarkably accelerate scientific dis-
information is alluring data scientists to biomedi- coveries, as exemplified in the ongoing
cal research [26, 39]. AI involves designing com- COVID-­19 pandemic, where significant advances
puter programs to learn, interpret, and predict in understanding pathology mechanisms, drug
real-world classifications mimicking human candidate identification, clinical diagnosis, and
intellect. For instance, machine learning models outbreak forecasts were achieved in record time
can now diagnose retinal pathologies and cardio- [21, 22, 44, 45]. Simply put, network medicine
vascular events with accuracy comparable to that and AI have the potential to enhance clinical
of trained clinicians [40–42]. Interestingly, these and preclinical research through multifaceted
models can be reverse engineered to reveal novel innovations and are projected to transform
patterns in biomedical data and stimulate data-­ biomedicine.
Network Biology and Medicine to Rescue: Applications for Retinal Disease Mechanisms and Therapy 169

4 Retinal Degeneration a constant cell death risk over the course of dis-
Mechanisms and Therapy ease. In the convergent pathway hypothesis,
mutation signals are anticipated to arrive at one
Retinal neurodegenerative diseases have limited of select “pre-apoptotic” switches that then deter-
treatment options and most available therapies mine the disease course [46, 49]. Interestingly, in
attempt to control progression into severe pathol- the majority of these models, pre-pathology inte-
ogy. Mitigation strategies employed so far gration can be predicted from a central regulator
include gene therapy, cell replacement therapies, such as mitochondria, for its crucial role in sig-
and prosthesis, among others [8, 9]. A major con- naling, apoptosis, and bioenergetics. Along these
straint in retinal therapy development is the lack lines, we have observed metabolic adaptations
of understanding of underlying disease mecha- and mitochondrial dysfunction as prominent fea-
nisms, despite availability of numerous animal ture of very early stages preceding disease onset
models and the retina being a well-characterized in a widely studied IRD model (rd1 mouse) [50].
tissue [34]. Furthermore, tremendous genetic Nonetheless, these models are yet to be tested
diversity of mutations and genes involved in exhaustively, and the precise mode of IRD onset
IRDs warrant treatments that can target multiple and progression remains poorly understood.
pathologies. To this end, gene agnostic methods Mapping pathway changes and interactions pre-
backed by network medicine are a natural choice, ceding disease onset onto interactome networks
where innovative approach can lead to better would provide an integrated map of assessing
understanding of disease mechanisms, drug dis- IRD pathology (Fig. 1b).
covery through repurposing/repositioning, and
improved clinical diagnosis and pathology
forecasts. Network Medicine for Retinal Disease
Mechanisms and Therapies Managing retinal
IRD Mechanisms and Models Offer a Case for degenerations requires addressing challenges on
Temporal Multi-omics Study Known retinal all preclinical and clinical fronts. Network
disease genes perform various molecular tasks medicine-­inspired study design and innovative
and participate in a wide range of pathways in AI algorithms can provide definitive steps toward
distinct retinal cells [46]. Across the genes and elucidation of the retinal diseasome and a new
thousands of mutations, it is still unclear how a direction for ocular disease research.
convergent fate of neuronal degeneration devel- Identification of genetic factors and disease genes
ops, albeit with different onset and progression in the clinic remains a challenge [30, 32]; how-
patterns. Over the years, some theoretical models ever, diagnosis can benefit from genomic screen-
have been proposed to describe plausible integra- ing and machine learning algorithms [51]. For
tive mechanisms underlying IRD. In the cumula- delineating disease mechanisms, layers of retinal
tive damage model, accumulation of abnormal molecular landscapes, such as genome, transcrip-
biochemical changes is imagined to trigger cell tome, proteome, metabolome, and cellular sig-
death [47]. The one-hit model proposes a mutant naling events, need to be integrated, ideally at
steady state in target retinal cells that eventually various time points, before disease onset to pin-
succumb upon a random cause [48]. Similarly, point the originators of cellular dysregulation and
the stochastic model emphasizes a variable cell molecular pathology (Fig. 1c). Following similar
death probability among mutant retinal cells, approaches, recent multi-omics studies of the
where disease onset happens from apoptosis in a retina have uncovered interesting insights [50,
fraction of target cells, and others survive rela- 52]. Network medicine can speed up biomarker
tively longer resulting in a progressive gradient discovery in ophthalmology and illustrate genetic
[46]. Notably, the latter two models subscribe to and molecular etiology of ocular diseases.
170 A. K. Mondal and A. Swaroop

5 Conclusion 13. Orth JD, Thiele I, Palsson BO. What is flux balance
analysis? Nat Biotechnol. 2010;28(3):245–8.
14. Milo R, Shen-Orr S, Itzkovitz S, Kashtan N, Chklovskii
Fundamental insights and breakthroughs are nec- D, Alon U. Network motifs: simple building blocks of
essary to advance therapeutic developments for complex networks. Science. 2002;298(5594):824–7.
clinical management and vision restoration 15. Barabasi AL, Gulbahce N, Loscalzo J. Network medi-
cine: a network-based approach to human disease.
efforts. Multi-omic network medicine investiga- Nat Rev Genet. 2011;12(1):56–68.
tions can distinguish early pathological changes 16. Maron BA, Altucci L, Balligand JL, Baumbach
in the retinal interactome and lead to biomarkers J, Ferdinandy P, Filetti S, et al. A global net-
and drug targets. Similar approach when extended work for network medicine. NPJ Syst Biol Appl.
2020;6(1):29.
to complex retinal phenotypes like aging and 17. Goh KI, Cusick ME, Valle D, Childs B, Vidal M,
age-related pathologies should elucidate the reti- Barabasi AL. The human disease network. Proc Natl
nal diseasome and lead to strategies for healthy Acad Sci U S A. 2007;104(21):8685–90.
vision. 18. Corominas R, Yang X, Lin GN, Kang S, Shen Y,
Ghamsari L, et al. Protein interaction network of
alternatively spliced isoforms from brain links genetic
risk factors for autism. Nat Commun. 2014;5:3650.
References 19. Soler-Lopez M, Zanzoni A, Lluis R, Stelzl U, Aloy
P. Interactome mapping suggests new mechanistic
1. RetNet. RetNet, the Retinal Information Network. details underlying Alzheimer’s disease. Genome Res.
Available from: https://sph.uth.edu/retnet/ 2011;21(3):364–76.
2. Rivolta C, Sharon D, DeAngelis MM, Dryja 20. Dudley JT, Sirota M, Shenoy M, Pai RK, Roedder S,
TP. Retinitis pigmentosa and allied diseases: numer- Chiang AP, et al. Computational repositioning of the
ous diseases, genes, and inheritance patterns. Hum anticonvulsant topiramate for inflammatory bowel
Mol Genet. 2002;11(10):1219–27. disease. Sci Transl Med. 2011;3(96):96ra76.
3. Swaroop A, Chew EY, Rickman CB, Abecasis 21. Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier
GR. Unraveling a multifactorial late-onset disease: K, White KM, et al. A SARS-CoV-2 protein interac-
from genetic susceptibility to disease mechanisms tion map reveals targets for drug repurposing. Nature.
for age-related macular degeneration. Annu Rev 2020;583(7816):459–68.
Genomics Hum Genet. 2009;10:19–43. 22. Gysi DM, Do Valle I, Zitnik M, Ameli A, Gan X, Varol
4. Maguire AM, Simonelli F, Pierce EA, Pugh EN Jr, O, et al. Network medicine framework for identifying
Mingozzi F, Bennicelli J, et al. Safety and efficacy drug repurposing opportunities for COVID-­19. ArXiv
of gene transfer for Leber's congenital amaurosis. N 2020.
Engl J Med. 2008;358(21):2240–8. 23. Luo Y, Zhao X, Zhou J, Yang J, Zhang Y, Kuang
5. Levi SR, Ryu J, Liu PK, Tsang SH. Precision medi- W, et al. A network integration approach for drug-­
cine trials in retinal degenerations. Annu Rev Vis Sci. target interaction prediction and computational drug
2021;7:851–65. repositioning from heterogeneous information. Nat
6. Sahel JA, Boulanger-Scemama E, Pagot C, Arleo A, Commun. 2017;8(1):573.
Galluppi F, Martel JN, et al. Partial recovery of visual 24. Sirota M, Dudley JT, Kim J, Chiang AP, Morgan
function in a blind patient after optogenetic therapy. AA, Sweet-Cordero A, et al. Discovery and preclini-
Nat Med. 2021;27(7):1223–9. cal validation of drug indications using compendia
7. West EL, Ribeiro J, Ali RR. Development of stem cell of public gene expression data. Sci Transl Med.
therapies for retinal degeneration. Cold Spring Harb 2011;3(96):96ra77.
Perspect Biol. 2020;12(8) 1. 25. Goecks J, Jalili V, Heiser LM, Gray JW. How
8. Roska B, Sahel JA. Restoring vision. Nature. machine learning will transform biomedicine. Cell.
2018;557(7705):359–67. 2020;181(1):92–101.
9. Scholl HP, Strauss RW, Singh MS, Dalkara D, 26. Wainberg M, Merico D, Delong A, Frey BJ. Deep
Roska B, Picaud S, et al. Emerging therapies for learning in biomedicine. Nat Biotechnol.
inherited retinal degeneration. Sci Transl Med. 2018;36(9):829–38.
2016;8(368):368rv6. 27. Li JO, Liu H, Ting DSJ, Jeon S, Chan RVP, Kim JE,
10. Hood L, Heath JR, Phelps ME, Lin B. Systems biol- et al. Digital technology, tele-medicine and artificial
ogy and new technologies enable predictive and pre- intelligence in ophthalmology: a global perspective.
ventative medicine. Science. 2004;306(5696):640–3. Prog Retin Eye Res. 2021;82:100900.
11. Barabasi AL, Oltvai ZN. Network biology: under- 28. Campbell JP, Mathenge C, Cherwek H, Balaskas K,
standing the cell's functional organization. Nat Rev Pasquale LR, Keane PA, et al. Artificial intelligence
Genet. 2004;5(2):101–13. to reduce ocular health disparities: moving from
12. Kitano H. Systems biology: a brief overview. Science. concept to implementation. Transl Vis Sci Technol.
2002;295(5560):1662–4. 2021;10(3):19.
Network Biology and Medicine to Rescue: Applications for Retinal Disease Mechanisms and Therapy 171

29. Becker SM, Tumminia SJ, Chiang MF. The NEI event prediction by machine learning: the
audacious goals initiative: advancing the frontier multi-ethnic study of atherosclerosis. Circ Res.
of regenerative medicine. Transl Vis Sci Technol. 2017;121(9):1092–101.
2021;10(10):2. 43. Eraslan G, Avsec Z, Gagneur J, Theis FJ. Deep learn-
30. Hanany M, Rivolta C, Sharon D. Worldwide carrier ing: new computational modelling techniques for
frequency and genetic prevalence of autosomal reces- genomics. Nat Rev Genet. 2019;20(7):389–403.
sive inherited retinal diseases. Proc Natl Acad Sci U S 44. Arik SO, Shor J, Sinha R, Yoon J, Ledsam JR, Le
A. 2020;117(5):2710–6. LT, et al. A prospective evaluation of AI-augmented
31. Berger W, Kloeckener-Gruissem B, Neidhardt J. The epidemiology to forecast COVID-19 in the USA and
molecular basis of human retinal and vitreoretinal dis- Japan. NPJ Digit Med. 2021;4(1):146.
eases. Prog Retin Eye Res. 2010;29(5):335–75. 45. Zhou Y, Hou Y, Shen J, Mehra R, Kallianpur
32. Carss KJ, Arno G, Erwood M, Stephens J, Sanchis-­ A, Culver DA, et al. A network medicine
Juan A, Hull S, et al. Comprehensive rare variant approach to investigation and population-
analysis via whole-genome sequencing to determine based validation of disease manifestations
the molecular pathology of inherited retinal disease. and drug repurposing for COVID-19. PLoS
Am J Hum Genet. 2017;100(1):75–90. Biol. 2020;18(11):e3000970.
33. Kousi M, Katsanis N. Genetic modifiers and oligo- 46. Wright AF, Chakarova CF, Abd El-Aziz MM,
genic inheritance. Cold Spring Harb Perspect Med. Bhattacharya SS. Photoreceptor degeneration: genetic
2015;5(6):a017145. and mechanistic dissection of a complex trait. Nat
34. Veleri S, Lazar CH, Chang B, Sieving PA, Banin E, Rev Genet. 2010;11(4):273–84.
Swaroop A. Biology and therapy of inherited retinal 47. Travis GH. Mechanisms of cell death in the inher-
degenerative disease: insights from mouse models. ited retinal degenerations. Am J Hum Genet.
Dis Model Mech. 2015;8(2):109–29. 1998;62(3):503–8.
35. Hartong DT, Berson EL, Dryja TP. Retinitis pigmen- 48. Clarke G, Collins RA, Leavitt BR, Andrews DF,
tosa. Lancet. 2006;368(9549):1795–809. Hayden MR, Lumsden CJ, et al. A one-hit model of
36. Furlong LI. Human diseases through the lens of net- cell death in inherited neuronal degenerations. Nature.
work biology. Trends Genet. 2013;29(3):150–9. 2000;406(6792):195–9.
37. Zhou X, Menche J, Barabasi AL, Sharma A. Human 49. Swaroop A, Kim D, Forrest D. Transcriptional regu-
symptoms-disease network. Nat Commun. lation of photoreceptor development and homeo-
2014;5:4212. stasis in the mammalian retina. Nat Rev Neurosci.
38. Sonawane AR, Weiss ST, Glass K, Sharma A. Network 2010;11(8):563–76.
medicine in the age of biomedical big data. Front 50. Jiang K, Mondal AK, Adlakha Y, Gumerson J,
Genet. 2019;10:294. Aponte AM, Gieser L, et al. Multiomics analyses
39. Baldi P. Deep learning in biomedical data science. reveal early metabolic imbalance and mitochondrial
Annu Rev Biomed Data Sci. 2018;1(1):181–205. stress in neonatal photoreceptors leading to cell
40. Scruggs BA, Chan RVP, Kalpathy-Cramer J, Chiang death in Pde6brd1/rd1 mouse model of retinal degen-
MF, Campbell JP. Artificial intelligence in retinopa- eration. Hum Mol Genet. 2022;31(13):2137–54.
thy of prematurity diagnosis. Transl Vis Sci Technol. 51. Dockery A, Whelan L, Humphries P, Farrar GJ. Next-­
2020;9(2):5. generation sequencing applications for inherited reti-
41. Ting DSW, Pasquale LR, Peng L, Campbell JP, nal diseases. Int J Mol Sci. 2021;22(11):5684.
Lee AY, Raman R, et al. Artificial intelligence and 52. Cherry TJ, Yang MG, Harmin DA, Tao P, Timms
deep learning in ophthalmology. Br J Ophthalmol. AE, Bauwens M, et al. Mapping the cis-regulatory
2019;103(2):167–75. architecture of the human retina reveals noncoding
42. Ambale-Venkatesh B, Yang X, Wu CO, Liu K, genetic variation in disease. Proc Natl Acad Sci U S
Hundley WG, McClelland R, et al. Cardiovascular A. 2020;117(16):9001–12.
Non-syndromic Retinal
Degeneration Caused
by Pathogenic Variants in Joubert
Syndrome Genes

Riccardo Sangermano, Egle Galdikaité-Braziené,


and Kinga M. Bujakowska

Abstract Keyword

Inherited retinal degenerations (IRDs) are a Non-syndromic IRD · Joubert syndrome ·


group of genetic disorders characterized by Genotype-phenotype correlation · CEP290 ·
progressive dysfunction and loss of photore- OFD1 · ARL3 · AHI1 · INPP5E
ceptors. IRDs are classified as non-syndromic
or syndromic, depending on whether retinal
degeneration manifests alone or in combina- 1 Introduction
tion with other associated symptoms. Joubert
syndrome (JBTS) is a genetically and clini- Inherited retinal degenerations (IRDs) are a group
cally heterogeneous disorder affecting the of genetically and clinically heterogeneous disor-
central nervous system and other organs and ders characterized by progressive photoreceptor
tissues, including the neuroretina. To date, 39 loss due to genetic defects in ~270 genes, inherited
genes have been associated with JBTS, a in all Mendelian modes [1]. Non-­syndromic IRDs
majority of which encode structural or func- manifest as an isolated retinal phenotype, while
tional components of the primary cilium, a syndromic IRDs affect other organs. A prominent
specialized sensory organelle present in most group of syndromic IRDs involves genes that code
post-mitotic cells, including photoreceptors. for proteins that are necessary for the generation
The use of whole exome and IRD panel next-­ and maintenance of the primary cilium, a special-
generation sequencing in routine diagnostics ized organelle present in most post-mitotic cells
of non-syndromic IRD cases led to the discov- [2]. These diseases are known as ciliopathies. As
ery of pathogenic variants in JBTS genes that the photoreceptor outer segment can be regarded
cause photoreceptor loss without other syn- as a specialized primary cilium, many ciliopathies
dromic features. Here, we recapitulate these often involve the retina [3].
findings, describing the JBTS gene defects Joubert syndrome (JBTS, OMIM #PS213300)
leading to non-syndromic IRDs. is an example of ciliopathy with retinal involve-
ment. JBTS is an autosomal or X-linked reces-
sive disorder, caused by defects in 39 different
R. Sangermano · E. Galdikaité-Braziené ·
K. M. Bujakowska (*) genes (Fig. 1). Over the years, the increasing
Ocular Genomics Institute, Massachusetts Eye and mutational screening studies in non-syndromic
Ear, Department of Ophthalmology, Harvard Medical IRD patients led to the identification of several
School, Boston, MA, USA cases carrying variants in genes initially
e-mail: kinga_bujakowska@meei.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 173
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_26
174 R. Sangermano et al.

Fig. 1 Overlap of clinical and genetic causation for congenital amaurosis, MKS Meckel syndrome, MORM
Joubert syndrome, other syndromic conditions, and non-­ mental retardation, truncal obesity, retinal dystrophy, and
syndromic IRD. Gene names highlighted in red indicate micropenis, OFDS oral-facial-digital syndrome, SGBS
that the relationship between the phenotype and the gene Simpson–Golabi–Behmel syndrome, SLS Senior–Løken
is provisional. BBS Bardet–Biedl syndrome, LCA Leber syndrome

a­ ssociated with JBTS. In this chapter, we pro- However, hypomorphic CEP290 mutations may
vide an overview of these novel associations and also cause non-syndromic IRDs, in particular
describe pathogenic variants detected in IRD Leber Congenital Amaurosis (LCA), rod-cone
cases. (RCD) and cone-rod dystrophies (CRD)
(Table 1) [7, 8].
The hypomorphic deep-intronic variant
2 Joubert Syndrome Genes c.2991 + 1655A > G, resulting in a mixture of
Mutated in Non-syndromic truncating (p.Cys998*) and wild-type proteins
Retinal Degeneration [7], is by far the most frequent variant identified
in non-syndromic IRD patients harboring
2.1 
CEP290 CEP290 mutations (https://databases.lovd.nl/
shared/genes/CEP290) [9]. In fact, ~77% of
CEP290 encodes a 2479 residue Centrosomal CEP290-IRD patients carry this variant in a com-
Protein 290 (kDa), which localizes to the centro- pound heterozygous state, while another ~21% is
meres of dividing cells and to the transition zone homozygous [10].
of the cilia, sometimes referred to as ‘connecting Other two well-studied hypomorphic vari-
cilium’ in the photoreceptors [4]. CEP290 plays a ants are c.451C > T, p.(Arg151*) and
pivotal role in regulating cilia assembly and c.4723A > T, p.(Lys1575*), both seemingly
development [5]. A recent study suggests that it is resulting in truncating mutations. The first vari-
essential for the initiation of transition zone ant (c.451C > T), found in family members with
assembly [6]. variable retinal phenotypes, leads to a mixture
CEP290 is the most frequently mutated gene of splicing products, one carrying the predicted
in recessive ciliopathies. Pathogenic variants in stop variant (p.(Arg151*)) and two resulting in
this gene have been associated with Meckel syn- in-frame deletions (p.(Leu148_Glu165del) and
drome (MKS; OMIM #611134), Senior–Løken p.(Leu148_Lys172del)) [11]. The second vari-
syndrome (SLS; OMIM #610189), Joubert syn- ant (c.4723A > T) gives rise to a truncated pro-
drome (JBTS; OMIM #610188), and Bardet– tein (p.(Lys1575*)) and an altered splicing
Biedl syndrome (BBS; OMIM #615991). product resulting in an in-frame deletion (­p.
Non-syndromic Retinal Degeneration Caused by Pathogenic Variants in Joubert Syndrome Genes 175

Table 1 CEP290 variants leading to non-syndromic IRD


Allele_1 (hg19) Allele_2 (hg19) Phenotype Probands References
c.2991 + 1655A > G, c.2991 + 1655A > G, LCA 38 Coppieters et al. (2010), den
p.(Cys998*) p.(Cys998*) Hollander et al. (2006),
Feldhaus et al. (2021),
Perrault et al. (2007),
Sallum et al. (2020), Testa et al.
(2021)
c.2991 + 1655A > G, Termination, frameshift and LCA or 103 Coppieters et al. (2010), den
p.(Cys998*) essential splice site variants RD Hollander et al. (2006),
Feldhaus et al. (2021),
Littink et al. (2010),
Perrault et al. (2007),
Sallum et al. (2020), Simonelli
et al. (2007), Skorczyk-­
Werner et al. (2020),
Testa et al. (2021), and
Tracewska et al. (2021)
c.2991 + 1655A > G, c.185A > G, p.(Lys62Arg) LCA 1 Tracewska et al. (2021)
p.(Cys998*)
c.2991 + 1655A > G, c.6012-12T > A, p.(?) LCA 1 Sallum et al. (2020)
p.(Cys998*)
c.2991 + 1655A > G, c.6271-8T > G, p.(?) LCA 2 Sallum et al. (2020)
p.(Cys998*)
c.164_167delCTCA, c.4723A > T, p.(Lys1575*) LCA 1 Sallum et al. (2020)
p.(Thr55Serfs*3)
c.180 + 1G > T, p.(?) c.7031_7034del, LCA or 1 Testa et al. (2021)
p.(Leu2344Hisfs*2) RD
c.250 + 2T > C, p.(?) c.7027del, RCD 2 Rafalska et al.
p.(Val2343Phefs*4) (2020), Tracewska et al. (2021)
c.353_354insGCAATTG, c.508A > T, p.(Lys170*) LCA 2 Sallum et al. (2020)
p.(Cys118Trpfs*6)
c.384_387delTAGA, c.4723A > T, p.(Lys1575*) LCA 1 Sallum et al. (2020)
p.(Asp128Glufs*34)
c.881C > G, p.(Ser294*) c.1522 + 1G > C, p.(?) LCA 1 Sallum et al. (2020)
c.1187_1188del, c.7341_7344dupACTT, LCA or 1 Testa et al. (2021)
p.(Lys396Argfs*25) p.(Ser2449Thrfs*7) RD
c.1511_1514 del GAGA, c.1511_1514 del GAGA, LCA 1 Aboussair et al. (2010)
p.(Arg504Serfs*10) p.(Arg504Serfs*10)
c.1666delA, c.2052 + 1_2052 + 2delGT, LCA 1 Sallum et al. (2020)
p.(Ile556Phefs*17) p.(?)
c.1666dupA, c.3310–1_3313delGCTTA, LCA 1 Chen et al. (2021)
p.(Ile556Asnfs*20) p.(Leu1104fs*6)
c.1711 + 5G > A, p.(?) c.5587-1G > C, p.(?) LCA 1 Perrault et al. (2007)
c.1936C > T, p.(Gln646*) c.6604delA, LCA 1 Perrault et al. (2007)
p.(Ile2203Leufs*23)
c.3012delA, c.4732G > T, p.(Glu1578*) LCA or 1 Testa et al. (2021)
p.(Glu1005Asnfs*6) RD
c.3640dupG, c.6604delA, LCA 1 Einsenberger et al. (2013)
p.(Glu1214Glyfs*7) p.(Ile2202Leufs*24)
c.4029 + 1G > A, p.(?) c.7048C > T, p.(Gln2350*) LCA 1 Feldhaus et al. (2021)
c.4040G > A, p. c.3104-2delA, p.(?) RCD 1 Shen et al. (2014)
(Trp1347*)
c.4705-1G > T, p.(?) c.3811C > T, p.(Arg1271*) LCA or 1 Testa et al. (2021)
RD
c.4711C > T, (p.Gln1597*) c.4711C > T, p.(Gln1597*) LCA 1 Beryzokin et al. (2014)
(continued)
176 R. Sangermano et al.

Table 1 (continued)
Allele_1 (hg19) Allele_2 (hg19) Phenotype Probands References
c.4723A > T, p.(Lys1575*) c.4723A > T, p.(Lys1575*) LCA 3 Perrault et al.
(2007), Coppieters et al. (2010)
c.4723A > T, p.(Lys1575*) c.1709C > G, p.(Ser570*) LCA 1 Perrault et al. (2007)
c.4882C > T, p.(Gln1628*) c.4882C > T, p.(Gln1628*) RCD 1 Tracewska et al. (2021)
c.4962_4963del, c.6604del, LCA or 1 Testa et al. (2021)
p.(Glu1656Asnfs*3) p.(Ile2202Leufs*24) RD
c.5493delA, c.5587-1G > C, p.(?) LCA 1 Feldhaus et al. (2021)
p.(Ala1832Profs*19)
c.5709 + 2T > G, p.(?) c.384_385del, LCA or 1 Testa et al. (2021)
p.(Asp128Glufs*17) RD
c.5813_5817del, c.6916A > T, p.(Arg2306*) LCA or 1 Testa et al. (2021)
p.(Thr1938Asnfs*15) RD
c.5850delT, c.5587-1G > C, p.(?) LCA 1 Perrault et al. (2007)
p.(Phe1950Leufs*14)
c.6012-2A > G, p.(?) c.6870delT, LCA 1 Einsenberger et al. (2013)
p.(Gln2291Lysfs*10)
c.6604del, c.6836 T > A, p.(Leu2279*) LCA or 1 Testa et al. (2021)
p.(Ile2202Leufs*24) RD
c.6916A > T, p.(Arg2306*) c.1987A > T, p.(Lys663*) LCA or 1 Testa et al. (2021)
RD
c.7341_7344dupACTT, c.1189 + 1G > A, p.(?) LCA or 1 Testa et al. (2021)
p.(Ser2449Thrfs*7) RD
c.289G > T, p.(Glu97*) c.5237G > A, p.(Arg1746Gln) CD 1 Feldhaus et al. (2021)
c.1219_1220del, c.4661_4663del, LCA or 1 Testa et al. (2021)
p.(Met407Glufs*13) p.(Glu1554del) RD
c.1451delA, c.5477T > C, p.(Leu1826Pro) LCA 1 Sallum et al. (2020)
p.(Lys484Argfs*8)
c.1593C > A, p.(Tyr531*) c.2T > A, p.(Met1Lys) LCA 1 Perrault et al. (2007)
c.1666del, c.6401T > C, p.(Ile2134Thr) LCA or 1 Testa et al. (2021)
p.(Ile556Phefs*17) RD
c.1666del, c.1466T > C, p.(Leu489Pro) LCA or 1 Testa et al. (2021)
p.(Ile556Phefs*17) RD
c.1984C > T, p.(Gln662*) c.223A > G, p.(Lys75Glu) RCD 1 Tracewska et al. (2021)
c.2578G > T, p.(Glu860*) c.3758G > A, p.(Arg1253His) LCA 1 Einsenberger et al. (2013)
c.2695C > T, p.(Gln899*) c.5777G > C, p.(Arg1926Pro) LCA 1 Sallum et al. (2020)
c.4723A > T, p.(Lys1575*) c.4696G > C, p.(Ala1566Pro) LCA 2 Perrault et al. (2007),
Coppieters et al. (2010)
c.4962_4963delAA, c.14T > C, p.(Ile5Thr) RD 1 Sallum et al. (2020)
p.(Glu1656Asnfs*3)
c.5493del, c.2723G > A, p.(Arg908Gln) LCA or 1 Testa et al. (2021)
p.(Ala1832Profs*19) RD
c.5850delT, c.4661_4663delAAG, LCA 1 Perrault et al. (2007)
p.(Phe1950Leufs*14) p.(Glu1554del)
c.7394_7395del, c.1664A > T, p.(Lys555Ile) LCA or 1 Testa et al. (2021)
p.(Glu2465Valfs*2) RD
CD cone degeneration, LCA Leber congenital amaurosis, RCD rod-cone degeneration, RD retinal degeneration

(Glu1569_Trp1604del) [11]. Additional The high frequency of the c.2991 + 1655A > G
CEP290 truncating alleles leading to non-­ variant and its intronic location make it an optimal
syndromic retinal degeneration have been dis- target for therapeutic intervention, with gene
covered (Table 1). It is likely that they also lead editing [12] and antisense oligonucleotide (AON)
to an altered splicing pattern and represent [13] protocols undergoing clinical trials
hypomorphic alleles. (NCT03140969, NCT04855045, NCT03396042).
Non-syndromic Retinal Degeneration Caused by Pathogenic Variants in Joubert Syndrome Genes 177

2.2 
OFD1 2.3 
ARL3

The Oral-Facial-Digital 1 (OFD1) on X chromo- ADP-ribosylation factor-like 3 (ARL3) encodes a


some encodes a multitask ciliary protein local- small GTPase involved in ciliogenesis and cyto-
ized at centrosome and basal body of the primary kinesis [24]. ARL3 is predominantly localized to
cilium. OFD1 regulates primary cilia formation the primary cilium transition zone and other
[14] and is part of the microtubule organizing microtubule structures [25]. ARL3 is a key mol-
center (MTOC), which is essential for cell cycle ecule in late-stage photoreceptor ciliogenesis. It
progression [15]. In addition, OFD1 was shown is a cargo displacement factor in photoreceptor
to localize in the nuclei where it may be involved cilia [26], where it directs lipidated proteins into
in chromatin remodeling [16]. the cilium in a GTP-dependent manner [27].
OFD1 is crucial for the embryonic develop- Transgenic mice expressing a dominant active
ment. Complete loss-of-function mutations in form of ARL3-Gln71Leu were shown to have
OFD1 were initially described to cause the photoreceptor nuclei migration defects at post-­
X-linked dominant oral-facial-digital type 1 syn- natal day 6, revealing a novel role of this protein
drome (OMIM 311200), which is lethal in males [28]. The activity of ARL3 is regulated by a gua-
[17]. However, truncating mutations in the nine nucleotide exchange factor, ARL13b and a
C-terminal part of the protein are thought to be GTPase activating protein, RP2, associated with
hypomorphic, and they cause X-linked recessive JBTS and RCD respectively [29, 30].
ciliopathies such as Simpson–Golabi–Behmel Mutations in the ARL3 gene are known to
syndrome (SGBS) [18], JBTS [19], and primary cause autosomal recessive JBTS and dominant or
ciliary dyskinesia (PCD) [20, 21]. Other hypo- recessive non-syndromic IRD (Table 2) [31–35].
morphic alleles in OFD1 were associated with a Clinical data suggest that rods and cones may be
severe non-syndromic X-linked RCD in two fam- differently affected, with maculopathy phenotype
ilies (Table 2) [22, 23]. manifesting at earlier stages [33]. To date, only
In a fifth-generation RCD family, a deep-­ missense ARL3 variants have been detected in
intronic variant c.935 + 706A > G in OFD1 both JBTS and non-syndromic IRD (https://data-
was discovered in affected males [22]. RT-PCR bases.lovd.nl/shared/genes/ARL3). A heterozy-
and direct sequencing of RNA extracted from gous variant c.269A > G, p.(Tyr90Cys) was
patients’ blood revealed insertion of a 62 bp found in two unrelated dominant IRD families
pseudoexon between exons 9 and 10, resulting with early-onset decreased central vision and
in a frameshift and premature termination central retinal thinning [34, 35]. Recently, another
(p.Asn313fs*330). The cryptic exon inclusion heterozygous variant c.200A > T, p.(Asp67Val)
was present in 70% of transcripts, thus acting was identified in a multi-generation Ashkenazi-­
as a hypomorphic allele. In the second family, Jewish family with a dominant form of IRD,
a hemizygous rare OFD1 missense variant characterized by a widespread severe progressive
c.358A > G, p.(Thr120Ala) was detected in retinal degeneration with maculopathy [33]. Two
one patient with an early onset severe form of additional studies reported recessive non-­
RCD [23]. However, it is unclear if this variant syndromic IRD caused by a homozygous
is truly pathogenic as it occurs in a weakly c.296G > T, p.(Arg99Ile) [32] and compound
conserved amino acid position, which is not heterozygous c.91A > G, p.(Thr31Ala) and
part of any functional protein domain. The c.353G > T, p.(Cys118Phe) mutations [31]. Four
amino acid change is also predicted to be additional non-syndromic cases were discovered
benign by multiple algorithms [23]. We cannot in a large mutation screening study (Table 2);
exclude, however, a hypomorphic nature of however, their mode of inheritance was not speci-
this variant that leads to an isolated retinal fied [36]. It is unclear why some variants lead to
phenotype. a syndromic disease and others to an isolated reti-
Table 2 OFD1, ARL3, AHI1, and INPP5E variants leading to non-syndromic IRD
178

Inheritance-­
Gene Allele_1 (hg19) Allele_2 (hg19) phenotype Proband Reference
OFD1 c.358A > G, p.(Thr120Ala) n.a. XL-CRD 1 Chen et al. (2018)
OFD1 c.935 + 706A > G, p.(Asn313fs*330) n.a. XL-CRD 1 Webb et al. (2012)
ARL3 c.91A > G, p.(Thr31Ala) c.353G > T, p.(Cys118Phe) AR-CRD 1 Fu et al. (2021)
ARL3 c.296G > T, p.(Arg99Ile) c.296G > T, p.(Arg99Ile) AR-CRD 2 Sheikh et al. (2019)
ARL3 c.200A > T, p.(Asp67Val) n.a. AD-RD 1 Ratnapriya et al. (2021)

ARL3 c.269A > G, p.(Tyr90Cys) n.a. AD-RCD 1 Holtan et al. (2019),


Strom et al. (2016)
ARL3 c.155A > C, p.(Asn52Thr) n.a. ?_ RCD 1 Koyanagi et al. (2019)
ARL3 c.308C > T, p.(Thr103Met) n.a. ?_ RCD 2 Koyanagi et al. (2019)
ARL3 c.353G > T, p.(Cys118Phe) n.a. ?_ RCD 3 Koyanagi et al. (2019)
ARL3 c.404C > T, p.(Pro135Leu) n.a. ?_ RCD 6 Koyanagi et al. (2019)
AHI1 c.660del, p.(Ser221Glnfs*10) c.2090C > T, p.(Pro697Leu) AR-RCD 1 Nguyen et al. (2017)
AHI1 c.653A > G, p.(Tyr218Cys) c.3257A > G, p.(Glu1086Gly) AR-RCD 1 Huang et al. (2015)

AHI1 c.1328T > A, p.(Val443Asp) c.3032C > G, p.(Ser1011*) AR-RCD 1 Repo et al. (2021)
AHI1 c.2087A > G, p.(His696Arg) c.2429C > T, p.(Pro810Leu) AR-RCD 1 Nguyen et al. (2017)
AHI1 c.[2174G > A; 2488C > T], p.[Trp725*; Arg830Trp] c.2258A > T, p.(Asp753Val) AR-RCD 1 Nguyen et al. (2017)

INPP5E c.473dup, p.(Asn159*) c.[746C > T; 1787G > C], p.[(Ser249Phe); RCD 1 Sangermano et al. (2021)
(Arg596Thr)]
INPP5E c.[746C > T; 1787G > C], p.[(Ser249Phe); c.[746C > T; 1787G > C], p.[(Ser249Phe); RCD 1 Sangermano et al. (2021)
(Arg596Thr)] (Arg596Thr)]

INPP5E c.844G > A, p.(Gly282Arg) c.1629C > A, p.(Tyr543*) PD 1 Birtel et al. (2018)
INPP5E c.874C > G, p.(Arg292Gly) c.1753C > T, p.(Arg585Cys) RCD 1 Stone et al. (2017)
R. Sangermano et al.
INPP5E c.914C > T, p.(Thr305Ile) c.1456C > T, p.(Arg486Cys) RCD 1 Sangermano et al. (2021)

INPP5E c.1073C > T, p.(Pro358Leu) c.1669C > T, p.(Arg557Cys) RCD 1 Xu et al. (2015)
INPP5E c.1094C > T, p.(Ser365Leu) c.1800C > G, p.(Asp600Glu) LCA 1 Sangermano et al. (2021)

INPP5E c.1393G > A, p.(Val465Ile) c.1393G > A, p.(Val465Ile) RCD 1 Sangermano et al. (2021)

INPP5E c.1402C > T, p.(Arg468Cys) c.1861C > T, p.(Arg621Trp) RCD 1 Sangermano et al. (2021)

INPP5E c.1456C > T, p.(Arg486Cys) c.1577C > T, p.(Pro526Leu) RCD 1 Sangermano et al. (2021)

INPP5E c.1543C > T, p.(Arg515Trp) c.1862G > A, p.(Arg621Gln) CRD 1 Stone et al. (2017)
INPP5E c.1670G > A, p.(Arg557His) c.1754G > A, p.(Arg585His) RCD 1 Sangermano et al. (2021)

INPP5E c.1754G > A, p.(Arg585His) c.1760del, p.(Val587Glyfs*7) RCD# 1 Sangermano et al. (2021)

INPP5E c.1861C > T, p.(Arg621Trp) c.1861C > T, p.(Arg621Trp) LCA 1 Wang et al. (2013)
INPP5E c.1862G > A, p.(Arg621Gln) c.1862G > A, p.(Arg621Gln) LCA/LCA# 3 Sangermano et al. (2021)

AD autosomal dominant, AR autosomal recessive, CRD cone-rod degeneration, LCA Leber congenital amaurosis, n.a. not applicable, PD pattern dystrophy, RCD rod-cone
degeneration, RD retinal degeneration, XL X-linked, # mild syndromic features present, ?_ inheritance not reported
Non-syndromic Retinal Degeneration Caused by Pathogenic Variants in Joubert Syndrome Genes
179
180 R. Sangermano et al.

nal phenotype. The disease mechanism of the truncal obesity, retinal dystrophy, and micropenis
dominant versus recessive variants is also syndrome, OMIM #610156) (https://databases.
unknown, and future functional studies would lovd.nl/shared/genes/INPP5E).
need to be undertaken to understand dominant To date, 17 families with severe early-onset
and recessive nature of the ARL3-associated IRD (mainly RCD and LCA) carrying INPP5E
disease. variants have been described [43–47]. Five of
them were identified in the past years, follow-
ing large mutational screening studies.
2.4 
AHI1 However, since these studies lacked a detailed
phenotypic description of patients, a clear asso-
The Abelson helper integration site 1 (AHI1) was ciation between INPP5E and non-syndromic
the first gene associated with JBTS [37, 38]. It IRD was established recently [47]. Non-
encodes Jouberin, a 1196-amino acid protein syndromic RCD was caused predominantly by
containing a coiled-coil region, seven WD40 missense variants (Table 2) [43–47]. A similar
repeats, and one Sarcoma homology 3 (SH3) trend was observed in the JBTS cases, and the
domain [38]. Its role in cerebellar and cortical combined impact of INPP5E variants in syn-
development is unknown, but the domain struc- dromic and non-­syndromic IRD patients does
ture suggests involvement in intracellular signal- not reveal a clear genotype–phenotype correla-
ing pathways [39]. tion, suggesting the involvement of genetic
The majority of reported AHI1 variants in modifiers [47].
JBTS cases are loss of function or homozygous
missense changes located within the WD40
repeats (https://databases.lovd.nl/shared/genes/ 3 Conclusions
AHI1). To date, only five non-syndromic RCD
families carrying AHI1 mutations have been We described five JBTS genes (CEP290, OFD1,
described (Table 2) [40–42]. Affected individuals ARL3, AHI1, and INPP5E) in which specific
from two families were compound heterozygous alleles lead to isolate retinal phenotypes rather
for missense variants [40, 41], and the affected than severe syndromic conditions. Hypomorphic
members of the remaining three families carried alleles due to the activation of cryptic splice
at least one truncating allele. Four of six missense sites and the position of the mutations in
variants leading to non-syndromic RCD are also CEP290 and OFD1 are one of the explanations
located in the WD40 domain; therefore, location for the broad phenotypic spectrum associated
of the mutations does not explain the severity of with these genes. Hypomorphic variants in
the phenotype. AHI1 mutations leading to non-­ AHI1, INPP5E, and recessive ARL3 disease are
syndromic IRD likely preserve some Jouberin also the most likely explanation; however, this
activity, which is sufficient for ciliary function in has yet to be proven. Similar tissue-dependent
all relevant tissues, except for the retinal photore- activity–effect model was also suggested for
ceptor cells. other syndromic and non-­syndromic retinal dis-
ease genes, CLN3, HGSNAT, and MFSD8 [48–
50]. Alternative splicing in the retina as proposed
2.5 
INPP5E for a ciliopathy gene TTC8 or epistatic effects of
other alleles as proposed for the CEP290-
The Inositol polyphosphate-5-phosphatase E associated disease can also be the reason for the
gene (INPP5E) encodes a phosphatase that plays broad range of disease expressions [51, 52].
a critical role in controlling ciliary growth and Understanding the functional consequences of
stability via the phosphoinositide 3-kinase sig- variants on gene function is at the forefront of
naling pathway. Pathogenic INPP5E variants current functional genetics studies and will
have been reported in patients with ciliopathies bring explanation of the pleiotropic effects of
such as JBTS or MORM (mental retardation, many genes.
Non-syndromic Retinal Degeneration Caused by Pathogenic Variants in Joubert Syndrome Genes 181

References 17. Ferrante MI, Giorgio G, Feather SA, et al.


Identification of the gene for oral-facial-digital type 1
syndrome. Am J Hum Genet. 2001;68:569–76.
1. Retinal Information Network (RetNet) [Internet].
18. Budny B, Chen W, Omran H, et al. A novel X-linked
[cited 2020 Apr 1]. Available from: https://sph.uth.
recessive mental retardation syndrome compris-
edu/retnet/home.htm
ing macrocephaly and ciliary dysfunction is allelic
2. Hildebrandt F, Benzing T, Katsanis N. Ciliopathies. N
to oral–facial–digital type I syndrome. Hum Genet.
Engl J Med. 2011;364:1533–43.
2006;120:171–8.
3. Bujakowska KM, Liu Q, Pierce EA. Photoreceptor
19. Coene KL, Roepman R, Doherty D, et al. OFD1 is
cilia and retinal ciliopathies. Cold Spring Harb
mutated in X-linked Joubert syndrome and interacts
Perspect Biol. 2017;9:a028274.
with LCA5-encoded lebercilin. Am J Hum Genet.
4. Sayer JA, Otto EA, O’Toole JF, et al. The centrosomal
2009;85:465–81.
protein nephrocystin-6 is mutated in Joubert syn-
20. Knowles MR, Daniels LA, Davis SD, et al. Primary
drome and activates transcription factor ATF4. Nat
ciliary dyskinesia. Recent advances in diagnostics,
Genet. 2006;38:674–81.
genetics, and characterization of clinical disease. Am
5. Stowe TR, Wilkinson CJ, Iqbal A, et al. The centrio-
J Respir Crit Care Med. 2013;188:913–22.
lar satellite proteins Cep72 and Cep290 interact and
21. Hannah WB, DeBrosse S, Kinghorn BA, et al. The
are required for recruitment of BBS proteins to the
expanding phenotype of OFD1-related disorders:
cilium. Mol Biol Cell. 2012;23:3322–35.
Hemizygous loss-of-function variants in three patients
6. Wu Z, Pang N, Zhang Y, et al. CEP290 is essential
with primary ciliary dyskinesia. Mol Genet Genomic
for the initiation of ciliary transition zone assembly.
Med. 2019;7:1–10.
PLoS Biol. 2020;18:e3001034.
22. Webb TR, Parfitt DA, Gardner JC, et al. Deep intronic
7. Den Hollander AI, Koenekoop RK, Yzer S, et al.
mutation in OFD1, identified by targeted genomic
Mutations in the CEP290 (NPHP6) gene are a fre-
next-generation sequencing, causes a severe form
quent cause of leber congenital amaurosis. Am J Hum
of X-linked retinitis pigmentosa (RP23). Hum Mol
Genet. 2006;79:556–61.
Genet. 2012;21:3647–54.
8. Feldhaus B, Weisschuh N, Nasser F, et al. CEP290
23. Chen X, Sheng X, Liu Y, et al. Distinct mutations with
mutation spectrum and delineation of the associated
different inheritance mode caused similar retinal dys-
phenotype in a large German cohort: a monocentric
trophies in one family: a demonstration of the impor-
study. Am J Ophthalmol. 2020;211:142–50.
tance of genetic annotations in complicated pedigrees.
9. Fokkema IFAC, Taschner PEM, Schaafsma GCP,
J Transl Med. 2018;16:1–12.
et al. LOVD v.2.0: the next generation in gene variant
24. Zhou C, Cunningham L, Marcus AI, et al. Arl2 and
databases. Hum Mutat. 2011;32:557–63.
Arl3 regulate different microtubule-dependent pro-
10. McAnany JJ, Genead MA, Walia S, et al. Visual acu-
cesses. Mol Biol Cell. 2006;17:2476–87.
ity changes in patients with Leber congenital amau-
25. Grayson C, Bartolini F, Chapple JP, et al. Localization
rosis and mutations in CEP290. JAMA Ophthalmol.
in the human retina of the X-linked retinitis pig-
2013;131:178–82.
mentosa protein RP2, its homologue cofactor C and
11. Littink KW, Pott JWR, Collin RWJ, et al. A novel
the RP2 interacting protein Arl3. Hum Mol Genet.
nonsense mutation in CEP290 induces exon skip-
2002;11:3065–74.
ping and leads to a relatively mild retinal phenotype.
26. Hanke-Gogokhia C, Zhang H, Frederick JM, et al. The
Investig Ophthalmol Vis Sci. 2010;51:3646–52.
function of Arf-like proteins ARL2 and ARL3 in pho-
12. Maeder ML, Stefanidakis M, Wilson CJ, et al.
toreceptors. Adv Exp Med Biol. 2016;854:655–61.
Development of a gene-editing approach to restore
27. Cavenagh MM, Breiner M, Schurmann A, et al.
vision loss in Leber congenital amaurosis type 10. Nat
ADP-ribosylation factor (ARF)-like 3, a new mem-
Med. 2019;25:229–33.
ber of the ARF family of GTP-binding proteins
13. Cideciyan AV, Jacobson SG, Drack AV, et al. Effect
cloned from human and rat tissues. J Biol Chem.
of an intravitreal antisense oligonucleotide on vision
1994;269:18937–42.
in Leber congenital amaurosis due to a photoreceptor
28. Wright ZC, Singh RK, Alpino R, et al. ARL3 regu-
cilium defect. Nat Med. 2019;25:225–8.
lates trafficking of prenylated phototransduction
14. Wang J, Chen X, Wang F, et al. OFD1, as a ciliary pro-
proteins to the rod outer segment. Hum Mol Genet.
tein, exhibits neuroprotective function in photorecep-
2016;25:2031–44.
tor degeneration models. PLoS One. 2016;11:1–18.
29. Cantagrel V, Silhavy JL, Bielas SL, et al. Mutations
15. Alfieri M, Iaconis D, Tammaro R, et al. The centro-
in the cilia gene ARL13B lead to the classical form of
somal/basal body protein OFD1 is required for micro-
Joubert syndrome. Am J Hum Genet. 2008;83:170–9.
tubule organization and cell cycle progression. Tissue
30. Veltel S, Gasper R, Eisenacher E, et al. The reti-
Cell. 2020;64:101369.
nitis pigmentosa 2 gene product is a GTPase-­
16. Giorgio G, Alfieri M, Prattichizzo C, et al. Functional
activating protein for Arf-like 3. Nat Struct Mol Biol.
characterization of the OFD1 protein reveals a nuclear
2008;15:373–80.
localization and physical interaction with subunits
31. Fu L, Li Y, Yao S, et al. Autosomal recessive rod-­
of a chromatin remodeling complex. Mol Biol Cell.
cone dystrophy associated with compound hetero-
2007;18:4397–404.
182 R. Sangermano et al.

zygous variants in ARL3 gene. Front Cell Dev Biol. 43. Wang X, Wang H, Sun V, et al. Comprehensive
2021;9:1–12. molecular diagnosis of 179 Leber congenital amau-
32. Sheikh SA, Sisk RA, Schiavon CR, et al. Homozygous rosis and juvenile retinitis pigmentosa patients by
variant in ARL3 causes autosomal recessive targeted next generation sequencing. J Med Genet.
cone rod dystrophy. Investig Ophthalmol Vis Sci. 2013;50:674–88.
2019;60:4811–9. 44. Xu Y, Guan L, Xiao X, et al. Mutation analysis
33. Ratnapriya R, Jacobson SG, Cideciyan AV, et al. A in 129 genes associated with other forms of reti-
novel ARL3 gene mutation associated with autosomal nal dystrophy in 157 families with retinitis pig-
dominant retinal degeneration. Front Cell Dev Biol. mentosa based on exome sequencing. Mol Vis.
2021;9:1–12. 2015;21:477–86.
34. Holtan JP, Teigen K, Aukrust I, et al. Dominant 45. Stone EM, Andorf JL, Whitmore SS, et al. Clinically
ARL3-related retinitis pigmentosa. Ophthalmic focused molecular investigation of 1000 con-
Genet. 2019;40:124–8. secutive families with inherited retinal disease.
35. Strom SP, Clark MJ, Martinez A, et al. De novo occur- Ophthalmology. 2017;124:1314–31.
rence of a variant in ARL3 and apparent autosomal 46. Birtel J, Eisenberger T, Gliem M, et al. Clinical and
dominant transmission of retinitis pigmentosa. PLoS genetic characteristics of 251 consecutive patients
One. 2016;11:1–9. with macular and cone/cone-rod dystrophy. Sci Rep.
36. Koyanagi Y, Akiyama M, Nishiguchi KM, et al. 2018;8:4824.
Genetic characteristics of retinitis pigmentosa in 1204 47. Sangermano R, Deitch I, Peter VG, et al. Broadening
Japanese patients. J Med Genet. 2019;56:662–70. INPP5E phenotypic spectrum: detection of rare
37. Ferland RJ, Eyaid W, Collura RV, et al. Abnormal variants in syndromic and non-syndromic IRD. npj
cerebellar development and axonal decussation due to Genomic Med. 2021;6:53.
mutations in AHI1 in Joubert syndrome. Nat Genet. 48. Kitzmüller C, Haines RL, Codlin S, et al. A func-
2004;36:1008–13. tion retained by the common mutant CLN3 pro-
38. Dixon-Salazar T, Silhavy JL, Marsh SE, et al. tein is responsible for the late onset of juvenile
Mutations in the AHI1 gene, encoding Jouberin, cause neuronal ceroid lipofuscinosis. Hum Mol Genet.
Jeubert syndrome with cortical polymicrogyria. Am J 2008;17:303–12.
Hum Genet. 2004;75:979–87. 49. Haer-Wigman L, Newman H, Leibu R, et al. Non-­
39. Muñoz-Estrada J, Ferland RJ. Ahi1 promotes Arl13b syndromic retinitis pigmentosa due to mutations in
ciliary recruitment, regulates Arl13b stability and the mucopolysaccharidosis type IIIC gene, heparan-­
is required for normal cell migration. J Cell Sci. alpha-­glucosaminide N-acetyltransferase (HGSNAT).
2019;132:jcs230680. Hum Mol Genet. 2015;24:3742–51.
40. Huang XF, Huang F, Wu KC, et al. Genotype-­ 50. Roosing S, Van Den Born LI, Sangermano R, et al.
phenotype correlation and mutation spectrum in a Mutations in MFSD8, encoding a lysosomal mem-
large cohort of patients with inherited retinal dystro- brane protein, are associated with nonsyndromic auto-
phy revealed by next-generation sequencing. Genet somal recessive macular dystrophy. Ophthalmology.
Med. 2015;17:271–8. 2015;122:170–9.
41. Nguyen TMT, Hull S, Roepman R, et al. Missense 51. Riazuddin SA, Iqbal M, Wang Y, et al. A splice-site
mutations in the WD40 domain of AHI1 cause mutation in a retina-specific exon of BBS8 causes
non-syndromic retinitis pigmentosa. J Med Genet. nonsyndromic retinitis pigmentosa. Am J Hum Genet.
2017;54:624–32. 2010;86:805–12.
42. Repo P, Järvinen RS, Sankila EM, et al. Identifying 52. Baala L, Audollent S, Martinovic J, et al.
haplotypes in recessive inherited retinal dystrophies Pleiotropic effects of CEP290 (NPHP6) mutations
using whole-genome linked-read sequencing. Clin extend to Meckel syndrome. Am J Hum Genet.
Genet. 2021;99:193–8. 2007;81:170–9.
Exonic Variants that Affect
Splicing – An Opportunity
for “Hidden” Mutations Causing
Inherited Retinal Diseases

Yogapriya Sundaresan, Eyal Banin,


and Dror Sharon

Abstract Keywords

Inherited retinal diseases (IRDs) are an Inherited retinal diseases · mRNA splicing ·
extremely diverse group of ocular disorders Exonic variants · Splice sites · Exonic
characterized by progressive loss of photore- splicing enhancers
ceptors leading to blindness. So far, patho-
genic variants in over 300 genes are reported
to structurally and functionally affect the ret- 1 Introduction
ina resulting in visual impairment. Around
15% of all IRD mutations are known to affect Pre-mRNA splicing is a molecular process which
an essential regulatory mechanism, pre-­ plays a significant role in regulating gene expres-
mRNA splicing, which contributes to the tran- sion as well as generating functional complexity.
scriptomic diversity. These variants disrupt This fundamental process excises the intervening
potential donor and acceptor splice sites as non-coding intronic sequences and links the cod-
well as other crucial cis-acting elements ing exons, thereby producing a mature mRNA
resulting in aberrant splicing. One group of molecule. Pre-mRNA splicing is regulated by a
these elements, the exonic splicing enhancers class of five small nuclear ribonucleoproteins
(ESEs), are involved in promoting exon defi- (snRNPs), namely U1, U2, U4, U5, and U6 and
nition and are likely to harbor “hidden” muta- around 150 other proteins, which together make
tions since sequence-analyzing pipelines up the macromolecular complex, spliceosome
cannot identify them efficiently. The main [1]. The functioning of the spliceosomal complex
focus of this review is to discuss the molecular depends on a set of cis elements present in the
mechanisms behind various exonic variants pre-mRNA sequence and additional trans factors
affecting splice sites and ESEs that lead to that bind to these elements. The primary splicing
impaired splicing which in turn result in an signal of most of the introns is a short sequence
IRD pathology. (cis element) positioned at the intron-exon junc-
tion that includes the 5′ and 3′ splice site as well
as the branch point [2]. Accurate splicing is
Y. Sundaresan · E. Banin · D. Sharon (*) achieved by gradual assembly of the snRNPs on
Department of Ophthalmology, Hadassah Medical the regulatory cis elements, which in turn results
Center, Faculty of Medicine, The Hebrew University
of Jerusalem, Jerusalem, Israel in removal of introns and two subsequent trans-
e-mail: dror.sharon1@mail.huji.ac.il esterification reactions combining the exonic

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 183
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_27
184 Y. Sundaresan et al.

sequences. Further, the components of the resulted in human genetic diseases such as fron-
­spliceosomal complex are recycled for the fol- totemporal dementia with parkinsonism, spinal
lowing rounds of splicing [3]. muscular atrophy, and Becker muscular dystro-
In addition to the abovementioned pre-mRNA phy [10]. However, only a very few reports are
embedded signals, other cis regulatory elements available on ESEs disrupting mutations in IRD
present in exons and introns are also crucial in genes, leaving the role of ESE-affecting muta-
exon recognition as well as in achieving precise tions in IRDs poorly understood. We propose that
splicing. This includes the exonic splicing either a nonsense-associated premature termina-
enhancers (ESEs), exonic splicing silencers tion codon (PTC) or other types of mutations
(ESSs), intronic splicing enhancers, and intronic including silent or missense variants could pos-
splicing silencers [4]. The molecular interaction sibly affect an ESE or ESS sequence, and their
between the elements and the components of the consequent effect on splicing might also contrib-
splicing machinery enables to recognize an exon ute to IRDs. Hence, besides mutations affecting
prior to the excision of intronic sequences. One splice sites, we hypothesize that ESE-affecting
of these elements, ESEs, plays a significant role variants can cause IRDs, but are likely to be “hid-
in promoting exon definition by facilitating the den” variants that are not yet recognized
recruitment of splicing factors to the upstream through variant analyses.
introns. In addition, ESEs serve to be a binding Hence, the primary focus of this chapter is to
site for the members of Serine/Arginine (SR)- briefly discuss the molecular mechanisms of var-
rich protein family that is also involved in draw- ious exonic variants affecting donor and acceptor
ing the factors of the spliceosomal complex, splice sites as well as ESE sequences, which in
thereby indicating an assisting role in exon defi- turn contribute to a retinal phenotype.
nition [5, 6].
Inherited retinal diseases (IRDs) are geneti-
cally and clinically heterogeneous ocular pheno- 2 Exonic Mutations Affecting
types in which mutations in genes associated the Donor and Acceptor
with retinal development, structure, and function Splice Sites
result in progressive loss of photoreceptors even-
tually compromising vision. Around 13–15% of Genetic alterations found in the boundaries of an
the total IRD-causing mutations affect the splic- exon (first or last nucleotide of exon and penulti-
ing mechanism by disrupting the GU-AG dinu- mate nucleotides, Fig. 1) are called exonic splice
cleotides at the splice donor and acceptor site [7, site variants. These variants can weaken or abol-
8]. The significance of donor and acceptor splice ish a splice site, thereby reducing the ability of
sites for precise splicing is evident by the fact that the spliceosomal complex from recognizing these
15% of the point mutations are located in these sites resulting in impaired splicing. As a conse-
regions resulting in one or a combination of quence, pronounced skipping of the correspond-
splice anomalies such as weakening or deleting a ing exon is evident [11]. Various exonic splice
regular splice site, creation of a novel splice sites site variants are reported in IRD genes, and a few
affecting exon recognition, and intron retention. are explained in this section of the review.
Donor splice mutations are more frequently Rhodopsin (RHO) is the major autosomal
reported compared to mutations in acceptor site dominant gene causing retinitis pigmentosa (RP),
[7–9]. a condition that is characterized by progressive
In addition, disruption of potential ESE loss of rod photoceptors followed by the cones
sequences by mutations might lead to abnormal leading to severe visual impairment [12]. The
splicing by eliminating the exon from the mature gene encodes a G-protein-coupled receptor that
mRNA transcript, which may result in a patho- is abundantly present in rod photoreceptors and is
logical condition. Various mutations have been significantly essential as the primary photorecep-
previously reported in the ESE sequences that tor molecule of vision [13]. So far, more than 100
Exonic Variants that Affect Splicing – An Opportunity for “Hidden” Mutations Causing Inherited Retinal… 185

RHO mutations have been reported as the cause Computational analysis using the tool NetGene2
of RP. A hidden disease-causing mutation in this predicted the loss of donor splice site in the
gene is c.936G>A that does not affect the imme- sequence carrying the variant. Subsequent mRNA
diate amino acid sequence of rhodopsin and can analysis from whole blood of patients carrying
therefore be considered as a silent change. the mutation identified that this variant caused
However, its location as the last nucleotide of skipping of exon 8 where the resulting protein
exon 4 raised the possibility that it might act as a lacks 48 amino acid residues and thus significant
donor site variant [9]. Indeed, c.936G>A was parts of cadherin ectodomains 2 and 3 [14].
shown to generate two misspliced transcripts in
COS7 cells, indicating an interference in exon
definition. About 95% of variants located in the 3 Exonic Variants Affecting
last nucleotide of an exon are predicted to result Exonic Splicing Enhancer
in missplicing, as the U1 snRNA fails to bind to Sequences
the modified exonic splice site. Such pathogenic
variants leading to altered exonic splice sites can ESEs are discrete sequences of 6–8 bps located
be corrected by a potential therapeutic approach within most of the exons that promote constitu-
using mutation-adapted U1 snRNA. The above-­ tive and regulated splicing (see example in
mentioned mutation c.936G>A has been rescued Fig. 1). These purine-rich sequences also serve as
by mutation-adapted U1 snRNA in 90% of binding motifs for various SR proteins and com-
abnormal transcripts in COS7 cells and retinal binedly function to excise the adjacent intron.
explant cultures [9]. Modification in the ESE sequences due to muta-
Another example is an apparently silent tions might greatly affect the binding factors
CDHR1-c.783G>A variant affecting the last leading to missplicing events such as exon skip-
nucleotide of exon 8. The cadherin-related family ping [15]. Identification of variants affecting the
member 1 (CDHR1) gene encodes a ESE sequences is highly demanding as even a
photoreceptor-­specific protein that is responsible silent variant could potentially disrupt an ESE
for the morphogenesis and arrangement of photo- though the amino acid sequence remains
receptor outer segment discs. Mutations in this unchanged. Online prediction tools including
gene are often associated with cone-rod dystro- ESE finder 3.0 are being used to identify putative
phy and central areolar choroidal dystrophy. ESE sequences [16]. However, such tools might

Fig. 1 Putative ESE sequences of BEST1 exon 6 pre- and 5 ESE sequences that had a score above the threshold
dicted by ESE finder 3.0. The illustration represents the and are depicted by different colors based on the corre-
prediction of specific ESE sequences recognized by each sponding SR protein. The image also presents two ESE
SR protein in exon 6 of the BEST 1 gene. The tool pro- affecting variants, c.704T>C and c.707A>G identified in
vides a threshold of 1.956, 2.38, 2.67, and 2.676 for exon 6 where c.704T>C weakened and c.707A>G abol-
SRSF1, SRSF2, SRSF5, and SRSF6, respectively, based ished an ESE sequence causing skipping of exon 6
on the inbuilt algorithm. The program identified 4, 2, 4,
186 Y. Sundaresan et al.

have a high level of false-positive results, and (c.508T), (ii) the mutant (c.508A), and (iii) the
therefore, each suspected ESE sequence needs to transcript skipping exon 8. Whereas the RT-PCR
be verified by RT-PCR analysis of mRNA primers specific for exon 32 produced the follow-
transcripts. ing four transcripts: (i) the canonical transcript
An excellent example of an IRD disease that is (c.4090G), (ii) the mutant (c.4090T), (iii) the
caused by ESE mutations as the major mecha- transcript skipping exon 32, and (iv) the tran-
nism is autosomal dominant vitreoretinocho- script lacking last 106 nucleotides of exon 32
roidopathy (ADVIRC) due to BEST1 mutations [19]. Although both mutations are nonsense and
(Fig. 1). BEST1 encodes Bestrophin-1, a trans- would be easily picked up by NGS pipelines,
membrane protein of basolateral membrane of these results have broader implications, since
the retinal pigment epithelium. Mutations in this only a lower percentage of transcripts include the
gene mainly cause AD Best disease (which is mutation (due to exon skipping), and therefore,
rarely inherited as an AR disease) and can also therapies targeting the mRNA (such as transla-
cause RP and ADVIRC. All ADVIRC-associated tion read-through inducing drugs and RNA edit-
mutations (c.256G>A, p.V86M; c.704T>C, ing) might show limited success.
p.V235A; c.707A>G, p.Y236C; and c.715G>A, Recessive pathogenic variants in USH2A,
p.V239M) were reported to cause skipping of the which encodes the Usherin protein, can cause
corresponding exons and therefore hypothesized either Usher syndrome and non-syndromic RP.
to affect ESE sequences [17, 18]. ESE-dependent One of the most frequent mutations in USH2A is
splice assays and gel-shift assays on two of the the single base pair deletion in exon 13
mutations (c.704T>C, p.V235A; c.707A>G, (c.2299delG). RT-PCR analysis of nasal epithe-
p.Y236C) located in exon 6 (Fig. 1) revealed that lial cells revealed the presence of two different
c.704T>C weakened and c.707A>G abolished an transcripts in patients carrying the deletion com-
ESE sequence causing skipping of exon 6. This pared to healthy individuals [20]. Exon 13 skip-
study also suggests that the mutation might have ping was evident in one group of subjects and
possibly introduced a composite exonic regula- skipping of exon 13 as well as 12 was identified
tory element site affecting precise splicing event in the second group of subjects. Bioinformatic
[18]. tool predicted that the deletion in exon 13 has
In addition, two unrelated patients with muta- disrupted a potential ESE sequence leading to
tions in CEP290 were presented with unusually missplicing. In addition, the study also claims
relatively mild retinal phenotype [19]. One of the that the deletion has not only affected an ESE but
patients (P2) was found to harbor two heterozy- also created an ESS sequence that resulted in
gous nonsense mutations in trans: c.508A>T skipping.
(p.K170*) in exon 8 and c.4090G>T (p.E1364*)
in exon 32. Prediction software analysis of these
two mutations did not show any effect on splice-­ 4 Conclusion
site scores but showed an increase in ESE/ESS
ratio suggesting that these mutations might dis- The human retina displays extraordinary tran-
rupt an ESE sequence leading to the skipping of script diversity, which requires highly regulated
exons 8 and 32 compared to controls. Furthermore, splicing activity that is achieved by specific splic-
the c.4090G>T allele is expected to generate a ing factors. Over 50% of human genes produce
strong donor splice-site 60 bp downstream of the more than one transcript, and therefore, a rela-
start of exon 32. mRNA analysis by RT- PCR and tively high percentage of variants might alter
Sanger sequencing of patient skin-fibroblasts splicing. As described in this review, exonic vari-
confirmed the in silico prediction where RT-PCR ants affecting splice sites and ESE sequences are
primers specific for exon 8 generated the follow- sometimes hidden and present a challenge to be
ing three transcripts: (i) the canonical transcript identified. RNA-based analysis including next-­
Exonic Variants that Affect Splicing – An Opportunity for “Hidden” Mutations Causing Inherited Retinal… 187

generation sequencing might be a suitable exper- therapeutic modalities. Prog Retin Eye Res. 2022;
89:101029.
imental method to determine these variants. 9. Tanner G, Glaus E, Barthelmes D, Ader M,
Additionally, the identification of novel variants Fleischhauer J, Pagani F, Berger W, Neidhardt
and understanding their role in retinal disease J. Therapeutic strategy to rescue mutation-induced
pathogenesis demand retina-specific microenvi- exon skipping in rhodopsin by adaptation of U1
snRNA. Hum Mutat. 2009;30(2):255–63.
ronment. The use of retinal organoids differenti- 10. Blencowe BJ. Exonic splicing enhancers: mechanism
ated from patient-derived iPSCs is a convenient of action, diversity and role in human genetic dis-
disease-on-a-dish model for such functional anal- eases. Trends Biochem Sci. 2000;25(3):106–10.
ysis as they provide more insights. Furthermore, 11. Anna A, Monika G. Splicing mutations in human
genetic disorders: examples, detection, and confirma-
novel mutation-specific therapies such as anti- tion. J Appl Genet. 2018;59(3):253–68.
sense oligonucleotides, small interfering RNA, 12. Berson EL. Retinitis pigmentosa. The Friedenwald lec-
and mutation-adapted U1 snRNA techniques can ture. Invest Ophthalmol Vis Sci. 1993;34(5):1659–76.
be used as a promising therapeutic option to treat 13. Lenahan C, Sanghavi R, Huang L, Zhang
JH. Rhodopsin: a potential biomarker for neurodegen-
missplicing-associated retinal diseases. erative diseases. Front Neurosci. 2020;14:326.
14. Charbel Issa P, Gliem M, Yusuf IH, Birtel J, Müller PL,
Acknowledgments This work was supported by the Mangold E, Downes SM, MacLaren RE, Betz C, Bolz
Israel Science Foundation (grant No. 1778/20). HJ. A specific macula-predominant retinal phenotype
is associated with the CDHR1 variant c.783G>A, a
silent mutation leading to in-frame exon skipping.
Declaration of Competing Interest The Invest Ophthalmol Vis Sci. 2019;60(10):3388–97.
authors declare that they have no competing 15. Taniguchi I, Masuyama K, Ohno M. Role of purine-­
interests. rich exonic splicing enhancers in nuclear reten-
tion of pre-mRNAs. Proc Natl Acad Sci U S A.
2007;104(34):13684–9.
16. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer
AR. ESEfinder: a web resource to identify
References exonic splicing enhancers. Nucleic Acids Res.
2003;31(13):3568–71.
1. Wahl MC, Will CL, Lührmann R. The spliceosome: 17. Yardley J, Leroy BP, Hart-Holden N, Lafaut BA,
design principles of a dynamic RNP machine. Cell. Loeys B, Messiaen LM, Perveen R, Reddy MA,
2009;136(4):701–18. Bhattacharya SS, Traboulsi E, Baralle D, De Laey JJ,
2. Wang Z, Burge CB. Splicing regulation: from a parts Puech B, Kestelyn P, Moore AT, Manson FD, Black
list of regulatory elements to an integrated splicing GC. Mutations of VMD2 splicing regulators cause
code. RNA. 2008;14(5):802–13. nanophthalmos and autosomal dominant vitreoreti-
3. Gehring NH, Roignant JY. Anything but ordinary – nochoroidopathy (ADVIRC). Invest Ophthalmol Vis
emerging splicing mechanisms in eukaryotic gene Sci. 2004;45(10):3683–9.
regulation. Trends Genet. 2021;37(4):355–72. 18. Burgess R, MacLaren RE, Davidson AE, Urquhart JE,
4. Bacchi N, Casarosa S, Denti MA. Splicing-correcting Holder GE, Robson AG, Moore AT, O’Keefe R, Black
therapeutic approaches for retinal dystrophies: where GCM, Manson FDC. ADVIRC is caused by distinct
endogenous gene regulation and specificity matter. mutations in BEST1 that alter pre-mRNA splicing. J
Investig Ophthalmol Vis Sci. 2014;55(5):3285–94. Med Genet. 2009;46(9):620–5.
5. Lam BJ, Hertel KJ. A general role for splicing enhanc- 19. Barny I, Perrault I, Michel C, Soussan M, Goudin N,
ers in exon definition. RNA. 2002;8(10):1233–41. Rio M, Thomas S, Atti-Bitach T, Hamel C, Dollfus
6. Cáceres EF, Hurst LD. The evolution, impact and H, Kaplan J, Rozet JM, Gerard X. Basal exon skip-
properties of exonic splice enhancers. Genome Biol. ping and nonsense-associated altered splicing allows
2013;14(12):R143. bypassing complete CEP290 loss-of-function in indi-
7. Liu MM, Zack DJ. Alternative splicing and retinal viduals with unusually mild retinal disease. Hum Mol
degeneration. Clin Genet. 2013;84(2):142–9. Genet. 2018;27(15):2689–702.
8. Schneider N, Sundaresan Y, Gopalakrishnan P, 20. Lenassi E, Saihan Z, Bitner-Glindzicz M, Webster
Beryozkin A, Hanany M, Levanon EY, Banin E, AR. The effect of the common c.2299delG muta-
Ben-Aroya, Sharon D. Inherited Retinal Diseases: tion in USH2A on RNA splicing. Exp Eye Res.
linking genes, disease- causing variants, and relevant 2014;122:9–12.
Enhanced S-cone Syndrome,
a Mini-review

Yiyi Wang, Jessica Wong, Jacque L. Duncan,


Austin Roorda, and William S. Tuten

Abstract and also shed light on the role of NR2E3


expression in photoreceptor survival.
Enhanced S-cone Syndrome (ESCS) is an
autosomal recessive inherited retinal disease Keywords
mostly associated with disease-causing vari-
ants in the NR2E3 gene. During retinal devel- Enhanced S-cone Syndrome (ESCS) ·
opment in ESCS, rod photoreceptor precursors Inherited retinal degeneration · Visual
are misdirected to form photoreceptors similar function · Retina · Photoreceptors · NR2E3 ·
to short-wavelength cones, or S-cones. S-cones
Compared to a normal human retina, patients
with ESCS have no rods and significantly
increased numbers of S-cones. Night blind- 1 Introduction
ness is the main visual symptom, and visual
acuity and color vision can be normal at early Enhanced S-cone Syndrome (ESCS) is a rare
disease stages. Histology of donor eyes and inherited autosomal recessive retinal degenera-
adaptive optics imaging revealed increased tion, associated with disease-causing variants in
S-cone density outside of the fovea compared the nuclear receptor subfamily 2, group E, mem-
to normal. Visual function testing reveals ber 3 (NR2E3) gene, first described in 1989 with
absent rod function and abnormally enhanced characteristic electroretinogram (ERG) findings
sensitivity to short-wavelength light. Unlike [20, 25, 27]. Patients with ESCS have night
most retinal degenerative diseases, ESCS blindness as the main visual symptom and can be
results in a gain in S-cone photoreceptor func- misdiagnosed as atypical retinitis pigmentosa or
tion. Research involving ESCS could improve congenital stationary night blindness [48].
understanding of this rare retinal condition Clinical appearance can be variable as the early
stage of the disease shows normal visual acuity,
visual field, color vision, and fundus appearance
Y. Wang (*) · A. Roorda · W. S. Tuten in some patients. More severe clinical findings
Herbert Wertheim School of Optometry and Vision are reduced visual acuity, impaired color vision,
Science, University of California, Berkeley, CA, USA
e-mail: yiyiwang@berkeley.edu cystoid maculopathy, macular hole, pigmentary
degeneration along the arcades, and advanced
J. Wong · J. L. Duncan
Department of Ophthalmology, University of macular atrophy [15]. ESCS is a retinal degener-
California, San Francisco, CA, USA ative disease with progressive slow and irrevers-

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 189
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_28
190 Y. Wang et al.

ible vision loss, especially when foveomacular NR2E3 encodes a transcription factor that
schisis is present that later develops into retinal directs retinal photoreceptor precursors toward a
atrophy [15]. Prior to the onset of severe retinal rod fate [19]. Together with CRX, NR2E3 pro-
degeneration and visual impairment, ESCS motes rod differentiation and suppresses the pro-
patients exhibit enhanced sensitivity to short-­ duction of cone photoreceptors, as shown in the
wavelength light, revealed by ERG and perimetry rd7 mutant mouse, a model for human ESCS [7,
[35]. Additionally, some mutations in human 9]. Since S-cones are produced earlier than the
NR2E3 cause not only ESCS but also other phe- other two cone subtypes, they are considered as
notypes of inherited retinal degeneration includ- the default primordial cone cells [43]. S-opsins
ing Goldmann–Favre syndrome (GFS), Clumped are expressed first before a portion of precursor
Pigmentary Retinal Degeneration (CPRD), and cells changes to M-cones in rodent retinas [44].
up to 1% of all autosomal dominant retinitis pig- Additionally, human fetal retina is shown to have
mentosa (adRP) [16, 39]. about 90% S-cones in early development, and
this ratio changes significantly later in photore-
ceptor development, suggesting an identity
2 
NR2E3 Gene Affects switch from the S-opsin to L- or M-opsin expres-
Photoreceptor sion by the precursor cells [43]. Disease-causing
Differentiation variants in NR2E3 disrupt photoreceptor fate
determination and further impact the cell ratio
The retina in a normal human eye has about 120 and topographic distributions of rods and cone
million rod photoreceptors. They are absent at the subtypes in the retina [47].
fovea and have the highest density at about 10–20 Nr2e3 gene variation can affect animal mod-
degrees eccentricity [10]. Rods are very sensitive els differently. The mutant retina of the rd7
to dim light and are responsible for night vision. mouse has shown slow and progressive retinal
Normal human eyes have around six million cone degeneration and abnormal lamination with
photoreceptors, which are used for fine spatial rosette formation, but has normal ERG responses,
resolution and color vision under photopic condi- unlike those seen in human ESCS [9].
tions. There are three types of cone photorecep- Additionally, the rd7 retina shows that most cells
tor, sensitive to long, medium, and short express both rod-specific and cone-specific
wavelengths of light, and are called L-, M-, and genes, suggesting that not only is there an
S-cones, respectively. S-cones comprise 5–10% increase of the S-opsin-expressing cones, but a
of all the cones and a mere ~0.02% of all photo- hybrid photoreceptor cell type is also formed
receptors in normal eyes [36]. from rod precursors that express rod- and cone-­
In ESCS retinal development, photoreceptor specific genes [9].
precursors that would normally develop into
rods are misdirected to form S-cone-like photo-
receptors, most commonly due to disease-caus- 3 Retinal Structure of ESCS
ing variants in a gene called NR2E3 [30]. The
NR2E3 gene product guides photoreceptor dif- Retinal structure information in ESCS has pri-
ferentiation and retinal progenitor competency marily been restricted to postmortem observa-
[18]. A recent study found that NR2E3- tions, which have confirmed an absence of rod
administered therapy can improve photorecep- photoreceptors, a twofold increased cone density
tor structure and function in multiple mouse with 92% being S-cones, and an overall disrupted
models of retinitis pigmentosa [23]. Conversely, retinal structure [30]. This histology study pro-
another group has pointed out that knockout of vides evidence of retinal degeneration in the
nr2e3 in adult mice could de-repress cone genes R311Q NR2E3 phenotype where the retina
in inherited retinal degeneration to improve degenerates with loss of cone photoreceptors,
photoreceptor survival [31]. migration of the RPE cells and gliosis of the
Enhanced S-cone Syndrome, a Mini-review 191

Müller cells [30]. Other histological findings [40]. However, the NR2E3 gene was not shown to
from patients with Goldmann–Favre syndrome, be essential for RS1 gene expression [22].
which is one phenotype of ESCS [21, 39], show Nevertheless, a link between NR2E3 and Müller
absent rods, an abnormal distribution of L/M- cell function is suggested because disease-­
and S-cone opsins [6], diffuse degenerative causing variants in NR2E3 could disrupt retinal
changes, vascular occlusive abnormalities, and integrity by compromising Müller cell function,
pre-retinal glial proliferation [33]. making the ESCS retina more vulnerable to schi-
In living eyes, spectral domain optical coher- sis formation [41].
ence tomography (SD-OCT) studies reveal that In addition to the retinal degeneration result-
ESCS patients have thickening of the outer ing from schisis in the macular region, photore-
nuclear layer and abnormal retinal layer organi- ceptor degeneration of the all-cone retina in
zation [2, 37, 42]. In addition, OCT demonstrates ESCS could also be attributed to the lack of rods
macular abnormalities including foveomacular and rod-derived cone viability factor (RdCVF),
schisis and macular holes [15, 24]. While OCT as RdCVF is secreted by rods to promote cone
provides valuable retinal layer thickness and survival in retinitis pigmentosa [1].
integrity information as a commercially available
technology to study retinal structure in eye dis-
eases, it is unable to resolve or distinguish 4 Visual Function of ESCS
between individual cone or rod photoreceptors,
thus direct observation of cone photoreceptor 4.1 Visual Sensitivity
topography in ESCS is not possible with
SD-OCT. In vivo retinal imaging using adaptive The conventional diagnostic test for ESCS is
optics scanning laser ophthalmoscopy (AOSLO) ERG, which measures full-field electrical
to examine photoreceptor structures in early responses of rods and cones with different light
ESCS patients shows that cone densities outside stimulations and conditions [28]. In a dark-­
the macula are higher than normal [2, 37]. adapted condition, ESCS patients have absent
The progression of ESCS retinal degenera- rod responses, but when the stimulus becomes
tion was described to have three clinical phases brighter, eliciting responses from both rods and
[41]. In the first phase, the mid-peripheral retina cones, a simplified and delayed response is
that is typically densely packed with rods is observed. In the light-adapted condition, the cone
filled with rod-cone hybrid cells that are prone response is large but slow. When short-­wavelength
to cell death at early stages [9], whereas the cen- stimuli are used, ESCS patients show enhanced
tral macular region is preserved due to normally responses to blue light [20, 24, 25]. The ERG fea-
developed L- and M-cones near the fovea [41]. tures suggest that S-cones may partially replace
In the second phase, macular schisis develops L- and M- cones and completely replace rod pho-
during young adulthood at the macular region, toreceptors [26]. Multifocal ERG results show
affecting visual acuity centrally and reducing normal amplitudes and latency in the central ret-
visual field when inner retinal structure is com- ina, but profoundly delayed responses in the
promised mechanically, as is observed similarly periphery, which suggests that central L- and
in X-linked retinoschisis [41]. In the third phase, M-cone development is relatively normal, but
as the retinoschisis resolves, the retinal thick- S-cones are predominantly distributed in the
ness reduces to normal or abnormally thin due peripheral retina and have slower responses [26].
to collapse of schisis cavities, but the retinal S-cone perimetry further supports this hypothe-
sensitivity maintains reduced [41]. sis: short-wavelength automated perimetry
The origin of retinoschisis in ESCS is (SWAP) revealed increased sensitivity to blue
unknown, but it has a similar phenotype to light across the central and the midperipheral
X-linked retinoschisis, caused by mutations in retina compared to normal eyes in early disease
the RS1 gene with reduced levels of retinoschisin stages [2]. As the disease progresses, the super-­
192 Y. Wang et al.

normal sensitivity in the ESCS retina becomes limit of midget ganglion cells as they collect sig-
normal in mild-to-moderate disease stages and nals from multiple cones [3, 4, 38, 45].
eventually becomes abnormal with an S-cone S-cones are generally considered not to con-
scotoma in the peripheral retina beyond 40 tribute to the pathways that support fine spatial
degrees and photoreceptor degeneration over vision for several reasons. First, S-cones are not
time [35]. present in the foveal center [11, 46]. Second, they
are quite sparse elsewhere in the retina compris-
ing only 10% of the cones at most. Third, S-cone
4.2 Color Vision excitatory signals are transmitted by a different
class of retinal ganglion cells, the small bistrati-
Color vision, measured with Ishihara plates and fied retinal ganglion cells (sbRGCs) [14]. In a
Farnsworth D-15 screening tests, was reported to normal eye, S-cone-mediated spatial resolution is
be mostly normal in ESCS patients [32]. very poor and closely correlates with the sam-
pling limit of the small bistratified cells [5, 13].
The density range of the human small bistratified
4.3 Temporal Vision retinal ganglion cells is estimated to range from
400 cells/mm2 centrally to 20 cells/mm2 periph-
Temporal vision in ESCS has been studied to erally [13]. Given a maximal density of around
understand the post-receptoral mechanism of 400 cells/mm2 centrally, the predicted maximal
S-cone signaling in ESCS [34]. S-cone critical S-cone acuity guided by sbRGCs is around three
flicker fusion and S-cone temporal contrast sensi- cycles/deg. The S-cone-mediated acuity reported
tivity in medium-to-high luminance levels are in several psychophysical studies is generally
improved in ESCS compared to normal subjects, below eight cycles/degree in the central and
which implies a faster than normal S-cone path- peripheral retina [5, 8, 29].
way in ESCS [34]. The faster S-cone signals So what, if any, benefits in spatial vision might
observed in the study are suggested to either orig- be conferred by the larger number of cones in an
inate from increased numbers of S-cone path- ESCS retina?
ways or increased S-cone access to the normal The impact of supernormal S-cone density
faster luminance pathways [34]. Spatial acuity on spatial vision is the least well-established of
measurement would be essential to test these the functional metrics. The foveal achromatic
theories. acuity of ESCS patients was reported to be
about 20/40 on average among 56 patients,
ranging from 20/20 to 20/1000 [15]. One study
4.4 Spatial Vision reported that the S-cone-mediated grating acu-
ity six degrees superior to the fovea was
The neural sampling limit for human vision is 8.16 cycles/degree (20/70 Snellen equivalent)
governed by the retinal ganglion cells (RGCs) on average in ESCS, compared to 3.7 cycles/
that convey signals from the retina to the brain degree (20/160 Snellen equivalent) in normal
[12]. At the fovea, there is minimal convergence subjects [17]. The spatial vision in this study
from cones to retinal ganglion cells, as one cone was measured at a single retinal location and
photoreceptor connects to at least one was not compared directly to the topography of
ON-ganglion cell and one OFF-ganglion cell. the photoreceptor mosaic. Thus, it is still unclear
This preserves visual information such that cen- whether spatial vision in ESCS is determined by
tral foveal vision is limited by optical aberrations the sampling density of S-cones or by the post-
and the sampling of the cone photoreceptors [12]. receptoral neural circuitry.
Just outside of the fovea, spatial vision becomes ESCS provides a unique phenotype to study
poorer than predicted by the sampling limit of the how the human visual system adapts to changes
photoreceptors and better matches the sampling in photoreceptor development and altered visual
Enhanced S-cone Syndrome, a Mini-review 193

inputs from abnormally distributed cone photore- 7. Chen J. The rod photoreceptor-specific nuclear recep-
tor Nr2e3 represses transcription of multiple cone-­
ceptors. In ESCS, S-cones are overexpressed at specific genes. J Neurosci. 2005;25:118–29.
the expense of other photoreceptor types [30]. If 8. Coates DR, Chung STL. Crowding in the S-cone
the increased number of S-cones in ESCS are pathway. Vis Res. 2016;122:81–92.
pooled into the normal blue-yellow small bistra- 9. Corbo JC, Cepko CL. A hybrid photoreceptor express-
ing both rod and cone genes in a mouse model of
tified ganglion cell pathway, the extra S-cones enhanced S-cone syndrome. PLoS Genet. 2005;1:e11.
may not contribute to improving spatial resolu- 10. Curcio CA, Sloan KR, Kalina RE, et al. Human
tion that is normally guided by L- and M-cones. photoreceptor topography. J Comp Neurol.
On the other hand, if S-cones that replaced L- and 1990;292:497–523.
11. Curcio CA, Allen KA, Sloan KR, et al. Distribution
M-cones during retinal development are wired and morphology of human cone photorecep-
into the midget ganglion cell pathway, we would tors stained with anti-blue opsin. J Comp Neurol.
expect an improvement in the spatial resolution 1991;312:610–24.
mediated by S-cones only and a lower-than-­ 12. Dacey D. The mosaic of midget ganglion cells in the
human retina. J Neurosci. 1993a;13:5334–55.
normal acuity mediated by the L- and M-cones 13. Dacey DM. Morphology of a small-field bistratified
exclusively. Studying the cone-type-mediated ganglion cell type in the macaque and human retina.
spatial resolution in ESCS will shed light on how Vis Neurosci. 1993b;10:1081–98.
the photoreceptors connect to the downstream 14. Dacey DM, Lee BB. The ‘blue-on’ opponent pathway
in primate retina originates from a distinct bistratified
neural pathways. ganglion cell type. Nature. 1994;367:731–5.
In summary, ESCS is an inherited retinal dis- 15. de Carvalho ER, Robson AG, Arno G, et al. Enhanced
ease that has the unique feature of showing a gain S-cone syndrome: spectrum of clinical, imaging, elec-
in cone photoreceptor function [19]. An increased trophysiologic, and genetic findings in a retrospec-
tive case series of 56 patients. Ophthalmol Retina.
understanding of this rare retinal disease will not 2021;5:195–214.
only help shed light on the role of NR2E3 during 16. Gire AI, Sullivan LS, Bowne SJ, et al. The Gly56Arg
normal retinal development but also allow us to mutation in NR2E3 accounts for 1–2% of auto-
investigate how the ESCS S-cone pathway is somal dominant retinitis pigmentosa. Mol Vis.
2007;13:1970–5.
involved with spatial resolution. 17. Greenstein VC, Zaidi Q, Hood DC, et al. The enhanced
S cone syndrome: an analysis of receptoral and post-­
receptoral changes. Vis Res. 1996;36:3711–22.
References 18. Haider NB, Demarco P, Nystuen AM, et al. The tran-
scription factor Nr2e3 functions in retinal progeni-
tors to suppress cone cell generation. Vis Neurosci.
1. Aït-Ali N, Fridlich R, Millet-Puel G, et al. Rod-derived
2006;23:917–29.
cone viability factor promotes cone survival by stimu-
19. Haider NB, Jacobson SG, Cideciyan AV, et al.
lating aerobic glycolysis. Cell. 2015;161:817–32.
Mutation of a nuclear receptor gene, NR2E3, causes
2. Ammar MJ, Scavelli KT, Uyhazi KE, et al.
enhanced S cone syndrome, a disorder of retinal cell
Enhanced S-cone syndrome: visual function, cross-­
fate. Nat Genet. 2000;24:127–31.
sectional imaging, and cellular structure with adap-
20. Jacobson SG, Marmor MF, Kemp CM, et al. SWS
tive optics ophthalmoscopy. Retin Cases Brief Rep.
(blue) cone hypersensitivity in a newly identified
2021;15:694–701.
retinal degeneration. Invest Ophthalmol Vis Sci.
3. Anderson RS, Wilkinson MO, Thibos
1990;31:827–38.
LN. Psychophysical localization of the human visual
21. Jacobson SG, Roman AJ, Roman MI, et al. Relatively
streak. Optom Vis Sci. 1992;69:171–4.
enhanced S cone function in the Goldmann-Favre
4. Anderson RS, Evans DW, Thibos LN. Effect of win-
syndrome. Am J Ophthalmol. 1991;111:446–53.
dow size on detection acuity and resolution acuity for
22. Langmann T, Lai CCL, Weigelt K, et al. CRX controls
sinusoidal gratings in central and peripheral vision. J
retinal expression of the X-linked juvenile retinoschi-
Opt Soc Am A. 1996;13:697.
sis (RS1) gene. Nucleic Acids Res. 2008;36:6523–34.
5. Anderson RS, Zlatkova MB, Demirel S. What lim-
23. Li S, Datta S, Brabbit E, et al. Nr2e3 is a genetic mod-
its detection and resolution of short-wavelength
ifier that rescues retinal degeneration and promotes
sinusoidal gratings across the retina? Vis Res.
homeostasis in multiple models of retinitis pigmen-
2002;42:981–90.
tosa. Gene Ther. 2020;28:223–41.
6. Bonilha VL, Fishman GA, Rayborn ME, et al. Retinal
24. Magliyah MS, Alsulaiman SM, Schatz P, et al.
pathology of a patient with Goldmann-Favre syn-
Evolution of macular hole in enhanced S-cone syn-
drome. Ophthalmic Genet. 2009;30:172–80.
drome. Doc Ophthalmol. 2021;142:239–45.
194 Y. Wang et al.

25. Marmor MF. Large rod-like photopic signals in a pos- 36. Roorda A, Williams DR. The arrangement of the
sible new form of congenital night blindness. Doc three cone classes in the living human eye. Nature.
Ophthalmol. 1989;71:265–9. 1999;397:520–2.
26. Marmor MF, Tan F, Sutter EE, et al. Topography 37. Roorda A, Sundquist S, Solovyev A, et al. Adaptive
of cone electrophysiology in the enhanced S optics imaging reveals supernormal cone density in
cone syndrome. Invest Ophthalmol Vis Sci. enhanced S-cone syndrome. Invest Ophthalmol Vis
1999;40:1866–73. Sci. 2010;51:2934.
27. Marmor MF, Jacobson SG, Foerster MH, et al. 38. Rossi EA, Roorda A. The relationship between visual
Diagnostic clinical findings of a new syndrome with resolution and cone spacing in the human fovea. Nat
night blindness, maculopathy, and enhanced S cone Neurosci. 2010;13:156–7.
sensitivity. Am J Ophthalmol. 1990;110:124–34. 39. Sharon D. Shared mutations in NR2E3 in enhanced
28. McCulloch DL, Marmor MF, Brigell MG, et al. S-cone syndrome, Goldmann-Favre syndrome, and
ISCEV standard for full-field clinical electroretinogra- many cases of clumped pigmentary retinal degenera-
phy (2015 update). Doc Ophthalmol. 2015;130:1–12. tion. Arch Ophthalmol. 2003;121:1316.
29. Metha AB, Lennie P. Transmission of spatial 40. Sikkink SK, Biswas S, Parry NRA, et al. X-linked ret-
information in S-cone pathways. Vis Neurosci. inoschisis: an update. J Med Genet. 2007;44:225–32.
2001;18:961–72. 41. Sohn EH, Chen FK, Rubin GS, et al. Macular function
30. Milam AH, Rose L, Cideciyan AV, et al. The nuclear assessed by microperimetry in patients with enhanced
receptor NR2E3 plays a role in human retinal pho- S-cone syndrome. Ophthalmology. 2010;117:1199–
toreceptor differentiation and degeneration. Proc Natl 1206.e1191.
Acad Sci U S A. 2002;99:473–8. 42. Sustar M, Perovšek D, Cima I, et al.
31. Kolesnikov A, Murphy DP, Corbo J, et al. Knockout Electroretinography and optical coherence tomog-
of Nr2e3 protects against photoreceptor degeneration raphy reveal abnormal post-photoreceptoral activ-
in two mouse models of retinitis pigmentosa. Invest ity and altered retinal lamination in patients with
Ophthalmol Vis Sci. 2022; 63:3135. enhanced S-cone syndrome. Doc Ophthalmol.
32. Nishiguchi KM, Friedman JS, Sandberg MA, et al. 2015;130:165–77.
Recessive NRL mutations in patients with clumped 43. Swaroop A, Kim D, Forrest D. Transcriptional regu-
pigmentary retinal degeneration and relative preserva- lation of photoreceptor development and homeo-
tion of blue cone function. Proc Natl Acad Sci U S A. stasis in the mammalian retina. Nat Rev Neurosci.
2004;101:17819–24. 2010;11:563–76.
33. Peyman GA, Fishman GA, Sanders DR, et al. 44. Szél Á, Van Veen T, Röhlich P. Retinal cone differen-
Histopathology of Goldmann-Favre syndrome tiation. Nature. 1994;370:336.
obtained by full-thickness eye-wall biopsy. Ann 45. Thibos LN, Cheney FE, Walsh DJ. Retinal limits to
Ophthalmol. 1977;9:479–84. the detection and resolution of gratings. J Opt Soc Am
34. Ripamonti C, Aboshiha J, Henning GB, et al. Vision A. 1987;4:1524–9.
in observers with enhanced S-cone syndrome: an 46. Williams DR, MacLeod DI, Hayhoe MM. Foveal trit-
excess of S-cones but connected mainly to conven- anopia. Vis Res. 1981;21:1341–56.
tional S-cone pathways. Invest Ophthalmol Vis Sci. 47. Xiao M, Hendrickson A. Spatial and temporal expres-
2014;55:963. sion of short, long/medium, or both opsins in human
35. Roman AJ, Powers CA, Semenov EP, et al. Short-­ fetal cones. J Comp Neurol. 2000;425:545–59.
wavelength sensitive cone (S-cone) testing as an out- 48. Yzer S, Barbazetto I, Allikmets R, et al. Expanded
come measure for NR2E3 clinical treatment trials. Int clinical spectrum of enhanced S-cone syndrome.
J Mol Sci. 2019;20:2497. JAMA Ophthalmol. 2013;131:1324–30.
Part VI
Inflammation
The Role of Microglia in Inherited
Retinal Diseases

Asha Kumari and Shyamanga Borooah

Abstract 1 Introduction

Inherited retinal diseases (IRDs) are a leading Microglia are the primary resident immune cells
cause of irreversible visual loss in the devel- of the retina and perform a major role in immune
oped world. The primary driver of pathology surveillance within the eye [3, 33]. After post-­
in IRDs is pathogenic genetic variant. natal development, in health, microglia normally
However, there is increasing evidence, from maintain a ramified morphology and reside pri-
recent studies, for a role of the immune system marily within the inner plexiform (IPL) and gan-
in disease mechanism, particularly retinal glion cell layers (GCLs) of the retina [34] (Fig. 1).
microglia. Microglia are the primary immune Microglia take part in the repair and maintenance
cells in the retina and actively contribute to of retinal tissue by inducing regeneration of new
disease pathogenesis when activated locally cells and blood vessels. Their ramified processes
by phagocytosing photoreceptors, inducing remain in close proximity to neuronal synapses,
inflammation and signaling infiltration of cir- and microglial activity is important for normal
culating monocytes. In this article, we discuss retinal visual signal transmission [40]. There is
the evidence for the contribution of retinal heterogeneity even among retinal microglia, and
microglia to IRD pathogenesis reported so far recent studies have identified different subclasses
using mice model. of microglia in the different retinal layers consis-
tent with distinct functional roles, and as a result,
Keywords microglia are likely to play a dynamic role
in location-based homeostasis within the retina
Microglia · Inflammation · Inherited retinal [18, 26]. In disease, microglia can rapidly induce
dystrophy · Animal model · Retinal retinal damage driven by pro-inflammatory cyto-
degeneration kines, neurotoxic molecules, complement path-
way proteins, and oxidative stress [32].
Additionally, activated microglia can recruit
A. Kumari · S. Borooah (*) other immune cells, including macrophages,
Jacobs Retina Center, Shiley Eye Institute, University from the systemic blood circulation, iris, or cili-
of California San Diego,
La Jolla, CA, USA ary body to the retina which further intensifies
e-mail: sborooah@health.ucsd.edu damage [18, 25]. In retinal degeneration,

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 197
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_29
198 A. Kumari and S. Borooah

Fig. 1 Retinal cryosection immunofluresence staining of IPL-Inner plexiform layer, INL-Inner nuclear layer, OPL-
wild-type mouse, Panel I-Iba-1 (pan microglial marker), Outer plexiform layer, ONL-Outer nuclear layer, IS/OS-
Panel II-CD68 (microglial activation marker), Panel III- Inner and Outer segments, RPE-Retinal pigment
DAPI, Panel IV-Brightfield. Most of the microglia are epithelium)
present in the IPL and OPL (GCL-Ganglion cell layer,

­ icroglia proliferate and accumulate near areas


m 2 Phosphodiesterase 6B
of retinal damage, where they can phagocytose (Pde6b)
living photoreceptors and contribute to cell death
[17, 43]. In addition, there is a transition from The rd1 mouse model results from homozygous
inactive ramified to more active ameboid forms nonsense mutations in exon 7 of the Pde6b gene,
with subsequent secretion of pro-inflammatory which encodes the beta subunit of cGMP-­Specific
cytokines such as TNF-α, IL-1β, ROS, NO, 3′,5′-Cyclic Phosphodiesterase [8, 31]. This
CCL2, and CCL3, which may accelerate damage mutation results in defects in the rod photorecep-
[4, 42]. The duration of this active state and cyto- tor signaling cascade. High cGMP levels facili-
kine profile is thought to be determined by the tate prolonged opening of cGMP-channels,
local tissue environment, location, as well as the resulting in the apoptosis of rod photoreceptors
timing and duration of stimuli [12]. A change in and degeneration [7]. Degeneration occurs
the microglial niche can trigger transcriptional extremely early in this model, with rod dysfunc-
reprogramming, which can also contribute to a tion occurring after P7. By 1 month of age, rd1
heterogenic microglial population associated mice demonstrate retinal thinning on SD-OCT
with pathogenicity [27]. This review focuses on imaging and responses are lost on full-field elec-
the evidence that microglia propagate inherited troretinogram (ffERG). Microglial activation has
retinal disease (IRDs), with particular focus on been reported in rd1 mice as early as P8. At this
the evidence from mouse models based on stage, microglia migrate from the IPL to the
the major affected IRD associated genes (Fig. 2). ONL. This microglial migration peaks at P14.
The Role of Microglia in Inherited Retinal Diseases 199

Fig. 2 Retinal cryosection immunofluresence staining of region. There is a marked increase in microglia compared
Mfrp loss of function mouse model. Panel I—Iba-1 (pan to wild-type mouse retina (GCL ganglion cell layer, IPL
microglial marker), panel II—CD68 (microglial activa- inner plexiform layer, INL inner nuclear layer, OPL outer
tion marker), panel III—DAPI, panel IV—brightfield. plexiform layer, ONL outer nuclear layer, IS/OS inner and
Most of the microglia are present in IPL, OPL, IS/OS outer segments, RPE retinal pigment epithelium)

Microglia in rd1 mice have classically activated secrete pro-inflammatory cytokines, including
microglial expression phenotypes with high IL-1β, leading to further degeneration [43].
CD86+CD206−, CD16/32, CD40, and pro-­ Administration of the IL-1β antagonist anakinra,
inflammatory cytokines (TNF-α, IL-6, and intraocularly, led to greater ONL preservation
CCL2). The number of CD206+CD86− cells was compared to control-treated eyes. This study pro-
low [44]. The rd10 mouse model results from vides strong evidence of retinal microglial activa-
another mutation in Pde6b. In this model, mis- tion and that microglial phagocytosis and
sense mutations are present in exon 13 of the beta cytokine signaling play a crucial role in retinal
subunit of Pde6b. This leads to decreased protein degeneration in the rd10 mouse model of
stability, transport, and phosphodiesterase activ- IRD. Peng et al. also used intraperitoneal admin-
ity. Increased cGMP levels lead to the increased istration of minocycline to inhibit microglial acti-
opening of the cGMP-gated channels and cal- vation in rd10 mice model. Their group reported
cium influx. This is thought to be the underlying improved retinal structure, function, and visual
cause of cell death of cones as well as rods in this behavior in these mice due to minocycline-­
model [39]. Zhao et al. reported that microglia induced inhibition of photoreceptor apoptosis
actively phagocytize dying rods, which express a and inflammation [30]. These studies suggest that
phosphatidylserine ‘eat me’ signal. Accelerating targeting of activated microglia could be a benefi-
the degeneration process further, microglia cial approach for IRD treatment.
200 A. Kumari and S. Borooah

3 Membrane Frizzled Related mutations have activated subretinal microglia


Protein (Mfrp) However, the role of microglia in this model is
yet to be studied in detail (Fig. 3).
The rd6 mouse model results from a spontaneous
mutation mapped to the mouse membrane friz-
zled receptor protein (Mfrp) gene on chromo- 4 Crumbs Cell Polarity
some 9. The gene has a human homolog on Complex Component 1 Gene
Chr11q23. The rd6 mouse mutation was found to (Crb1)
result from a four-base pair deletion in Mfrp in a
splice donor site, which resulted in skipping exon Mutations in the gene encoding human Crumbs
4 and a predicted truncation of MFRP protein cell polarity complex component 1 gene (CRB1)
[20]. Phenotypically, there is abnormal develop- have been associated with several retinal dystro-
ment of photoreceptor outer segments with phies including Leber congenital amaurosis and
defects including disorganized and reduced api- RP. The Crumbs protein is linked with several
cal RPE microvilli, confirming the importance of biological processes in the mammalian eye,
MFRP for normal functioning and maintenance including retinal patterning, ciliogenesis, and
of photoreceptors [15]. There are several other vesicular transport [16]. Rd8 mice have muta-
models associated with mutations in Mfrp. A tions in the Crb1 gene resulting in white retinal
deletion in exon 3 had been reported in the pro- spots from retinal folding known as neuroretinal
tein truncation represented as 174delG. Mice rosettes [8]. There is marked phenotypic variabil-
develop whitish autofluorescent spots across ity in mice harboring this mutation, and evidence
their fundus. The white dot appearance in the suggests that variation in the genetic background
subretinal cells in mice was identified as inflam- may further affect microglial responses [23]. It
matory monocyte-derived cells with F4/80, has been reported that there is a clear correlation
CD11b, Iba1, and CD68 expression [13]. To between these fundus white spots and Iba-1+
investigate the origin of the inflammatory cells, microglia number in mice with homozygous
the authors generated bone marrow chimeras mutations of Crb1. With increasing age, there
with labeled monocytes in mice carrying muta- was a greater correlation between spots and Iba-­
tions in Mfrp. The labeled monocytes were 1+ microglia in 14–21-month-old mice compared
observed approximately 8 weeks later. However, to 1–9-month-old mice [22]. Evidence for
these mice were lethally irradiated as weanlings, microglial activation was seen in a comparative
which may have affected the BRB and retinal study using (C57BL/6N) carrying the rd8 muta-
microglia, and so this limits the conclusions, tion and (C57BL/6J) without the rd8 mutation,
which can be drawn using this model [14]. which demonstrated increased expression of
Additionally, there was no investigation into CCL2, CFB, C3, NF-kβ, CD200R, and TNF-α in
whether the inflammatory cells modified disease the rd8 mice [23, 24]. C57BL/6N mice were
progression in this model. Another Mfrp model, more prone to subretinal accumulation of microg-
resulting from homozygous insertion of a cyto- lia with a pro-inflammatory shift characterized
sine in exon 5, a frame shift generating a prema- by upregulation of cytokine positive regulatory
ture termination of the Mfrp transcript. This genes and genes involved in oxidative stress [5].
mouse model carrying homozygous Microglial accumulation in the sub-retinal space,
c.498_499insC mutations in Mfrp (MfrpKI/KI) has in addition to upregulation of pro-inflammatory
been recently described [10]. The lack of func- genes and complement pathway proteins, occurs
tional MFRP protein resulted in hyperopia, auto- with age and increasing degeneration [5].
fluorescent spots, and retinal degeneration, which Although markers of activation were noted in the
were detectable by scanning laser ophthalmos- rd8 model, studies to deplete or inhibit active
copy and optical coherence tomography as early microglia, to see if they contribute to retinal
as 1 month of age. Mice carrying c.498_499insC degeneration, have so far not been performed.
The Role of Microglia in Inherited Retinal Diseases 201

Fig. 3 Illustration of the role of microglia in inherited tor 1, Mertk Mer proto-oncogene tyrosine kinase, Rpe65
retinal diseases (IRDs). Pde6b phosphodiesterase 6B, retinoid isomerohydrolase, Nr2e3 nuclear receptor sub-
MFRP membrane frizzled related protein, Crb-1 crumbs family 2 group E member 3, P3h1 prolyl 3-hydroxylase 1
cell polarity complex-1, Cx3cr1 CX3C chemokine recep-

5 CX3C Chemokine Receptor 1 reversed when CX3CL1-CX3CR1 signaling was


(Cx3cr1) improved, using exogenous recombinant
CX3CR1, resulting in lowered microglial infil-
Chemokines are a group of small secretory mol- tration, phagocytosis, activation, and higher pho-
ecules, which bind to their G-protein-coupled toreceptor survival. Current studies suggest that
receptor and play an important role in the regula- CX3CL1-CX3CR1 signaling is an important
tion of the immune system in several diseases molecular mechanism capable of modulating
including retinal degeneration [6]. CX3CR1 (the microglial-mediated degeneration [41].
fractalkine receptor) is the only CX3C family Microglia interact closely with photoreceptors
receptor, but it is nonetheless important for pro- during early retinal development, modulating
liferation and the recruitment of monocytes. The photoreceptor development and maturation.
role of microglia in retinal degeneration was CX3CR1 ablation in Cx3cr1GFP/GFP mice resulted
evaluated using CX3CR1 deficient rd10 mice, in a significant decrease in photoreceptor length
resulting in the augmentation of inflammatory and aberrant photoreceptor maturation during
cytokines, microglial activation markers, and postnatal development when compared with
increased phagosomes. These effects were wild-type mice [19].
202 A. Kumari and S. Borooah

6 MER Proto-Oncogene, nal rosette at P60 [38]. The autofluorescent spots


Tyrosine Kinase (Mertk) were like those present in human cases with
Nr2e3 mutation and autofluorescent spots in the
The Royal College of Surgeons (RCS) rat is one macula. The contribution of BM-derived cells in
of the most commonly used animal models for retinal degeneration was studied by crossing
IRD studies. The model carries mutations in the mice carrying the macrophage Fas-induced apop-
receptor tyrosine kinase gene (Mertk) resulting in tosis transgene (Mafia) with rd7 mice carrying
defective phagocytosis of photoreceptor outer the Nr2e3 mutation. A synthetic dimerizer,
segments. They represent relevant models for AP20187, was used to deplete only BM-derived
studying retinal degeneration and pathologies [2, microglia, resulting in decreased photoreceptor
36]. In an attempt to identify the fate of microglia count and an increase in pro-inflammatory mark-
in mouse models of MERTK deficiency, several ers including IL-1β, IL-6, and TNF-α [37].
transgenic mice were generated. Kohno et al. Additionally, the number of white spots increased
used Mertk −/− Cx3cr1 GFP/+ Ccr2 RFP/+ mice with after the depletion of BM derived cells. This was
defective RPE phagocytosis. As CX3CR1 and driven by a proliferation of resident retinal
CCR2 are relatively specific markers of microg- microglia. This model again provides evidence
lia and activated macrophages, respectively, fluo- for a divergent role for BM-derived and resident
rescent protein sequences were inserted to enable retinal microglia in IRD.
the tagging and investigation of those cells in this
model. CCR2+-activated macrophages and
double-­positive macrophages, which had become 8 Retinoid Isomerohydrolase
microglia-like, were reported in the subretinal (Rpe65)
space at week 8. Initially (4 weeks), only
CX3CR1+ (GFP+) cells were observed, which The rd12 mouse model arose from a spontaneous
represent resident microglia [21]. This suggests homozygous nonsense mutation near the Rpe65
strong evidence of an extra-ocular origin for reti- gene located on mouse chromosome 3. This
nal microglia in the Mertk mouse model. model is useful in the study of slow-onset retinal
degeneration as it has an even slower progression
than rd10 models [28]. Although ultrastructural
7 Nuclear Receptor Subfamily changes were noted as early as 3 weeks of age in
2 Group E Member 3 (Nr2e3) this model, the main pathology occurred at
approximately 7 months of age with only six to
The rd7 mouse model results from a spontaneous eight layers of the ONL remaining. However, like
380 nucleotide deletion in Nr2e3 on mouse chro- human disease, the scotopic electroretinography
mosome 9. Nr2e3 is normally expressed by pho- results were profoundly diminished early, even
toreceptors. The deletion results in a frame shift by P21 [29]. Sasahara et al. investigated the role
and produces a premature stop codon [1]. Rd7 of BM-derived microglia in retinal degeneration
mice develop multiple autofluorescent retinal in rd12 alongside the rd1 and rd10 mouse mod-
white spots across the retina at an early age (P27). els. They transplanted BM-derived cells from
The white dot appearance has been linked to his- CAG-EGFP mice into rd12 mice at P30 and into
topathological changes in the retina resulting in rd10 mice at P15. Transplanted (BM-derived)
retinal whorls and rosettes [11]. These mice show microglial cells were seen in the retina at P60 in
an early attenuation of both a- and b-waves. rd12 and at P30 in rd10 model, which suggests
However, electroretinography stabilizes in the that macrophagic migration to the retina coin-
first year before deteriorating further [9]. Rd7 cides with degeneration. Again, a limiting factor
mouse retina have abnormal accumulation of of these studies is that the mice were all lethally
outer segments debris containing microglia, irradiated, and the mice were not tested for rd8
which appeared as autofluorescent spots in reti- mutations [35].
The Role of Microglia in Inherited Retinal Diseases 203

9 Future Perspectives eration in the rd7 mouse. Proc Natl Acad Sci U S A.
2000;97:5551–6.
2. Al-Khersan H, Shah KP, Jung SC, Rodriguez A,
There is currently some evidence, from mouse Madduri RK, Grassi MA. A novel MERTK mutation
models of IRD, pointing to a role for microglia in causing retinitis pigmentosa. Graefes Arch Clin Exp
retinal degeneration. However, further work is Ophthalmol. 2017;255:1613–9.
3. Aloisi F. Immune function of microglia. Glia.
required in most models to establish their causal 2001;36:165–79.
role in retinal degeneration. Microglia and mac- 4. Appelbaum T, Santana E, Aguirre GD. Strong
rophages appear to play a role in the clearance of upregulation of inflammatory genes accompanies
dying photoreceptors, but it is still not clear photoreceptor demise in canine models of retinal
degeneration. PLoS One. 2017;12:e0177224.
whether activated microglia have a deleterious 5. Aredo B, Zhang K, Chen X, Wang CX-Z, Li T, Ufret-­
effect on disease progression. There is a need for Vincenty RL. Differences in the distribution, phe-
focused studies, using animal models, on acti- notype and gene expression of subretinal microglia/
vated retinal microglia. The development of macrophages in C57BL/6N (Crb1 rd8/rd8) versus
C57BL6/J (Crb1 wt/wt) mice. J Neuroinflammation.
inducible models is also likely to assist with the 2015a;12:6.
use of depletion studies. In addition, a better 6. Balkwill FR. The chemokine system and cancer. J
understanding of microglial response is required Pathol. 2012;226:148–57.
in humans with IRD. In vivo clinical studies, 7. Bowes C, Li T, Danciger M, Baxter LC, Applebury
ML, Farber DB. Retinal degeneration in the rd mouse
optimized in animal models, using the latest is caused by a defect in the β subunit of rod cGMP-­
imaging and microglial labeling, could assist. phosphodiesterase. Nature. 1990;347:677.
Single-cell transcriptome profiling can help iden- 8. Chang B, Hawes N, Hurd R, Davisson M, Nusinowitz
tify subgroups of cells and generate better tran- S, Heckenlively J. Retinal degeneration mutants in the
mouse. Vis Res. 2002;42:517–25.
scriptomic networks of activation, which will 9. Chang BHJR, Hawes NL, Davisson MT. A new
allow targeting of retinal microglia. Finally, tech- mouse model of retinal dysplasia and degeneration
niques such as spatial sequencing and highly (rd7). Invest Ophthalmol Vis Sci. 1998;39:s880.
multiplexed immunohistochemistry and RNA 10. Chekuri A, Sahu B, Chavali VRM, Voronchikhina M,
Soto-Hermida A, Suk JJ, Alapati AN, Bartsch D-U,
labeling may provide even more clues to microg- Ayala-Ramirez R, Zenteno JC. Long-term effects
lial heterogeneity by highlighting regional differ- of gene therapy in a novel mouse model of human
ences. The repurposing of existing systemic MFRP-associated retinopathy. Hum Gene Ther.
treatments for local use in the context of retinal 2019;30:632–50.
11. Cheng H, Khan NW, Roger JE, Swaroop A. Excess
degeneration is extremely tempting and may pro- cones in the retinal degeneration rd7 mouse, caused
vide an opportunity to slow degeneration either by the loss of function of orphan nuclear receptor
as a primary treatment or in combination with Nr2e3, originate from early-born photoreceptor pre-
other therapies. cursors. Hum Mol Genet. 2011;20:4102–15.
12. Chiu IM, Morimoto ET, Goodarzi H, Liao JT,
O’Keeffe S, Phatnani HP, Muratet M, Carroll MC,
Acknowledgments S.B. is supported by the Foundation Levy S, Tavazoie S, Myers RM, Maniatis T. A
Fighting Blindness Career Development Award. This neurodegeneration-­ specific gene-expression signa-
artwork was prepared using BioRender.com. ture of acutely isolated microglia from an amyo-
trophic lateral sclerosis mouse model. Cell Rep.
Conflict of Interest None. 2013;4:385–401.
13. Fogerty J, Besharse JC. 174delG mutation in mouse
MFRP causes photoreceptor degeneration and RPE
atrophy. Invest Ophthalmol Vis Sci. 2011;52:7256–66.
14. Fogerty J, Besharse JC. Subretinal infiltration of
References monocyte derived cells and complement misregula-
tion in mice with AMD-like pathology. Adv Exp Med
Biol. 2014a;801:355–63.
1. Akhmedov NB, Piriev NI, Chang B, Rapoport AL,
15. Fogerty J, Besharse JC. Subretinal infiltration of
Hawes NL, Nishina PM, Nusinowitz S, Heckenlively
monocyte derived cells and complement misregula-
JR, Roderick TH, Kozak CA, Danciger M, Davisson
tion in mice with AMD-like pathology. In: Retinal
MT, Farber DB. A deletion in a photoreceptor-­
degenerative diseases. Springer, New York; 2014b.
specific nuclear receptor mRNA causes retinal degen-
p. 355–63.
204 A. Kumari and S. Borooah

16. Gosens I, den Hollander AI, Cremers FP, Roepman 28. Pang J, Chang B, Hawes NL, Hurd RE, Davisson
R. Composition and function of the Crumbs protein MT, Li J, Noorwez SM, Malhotra R, McDowell
complex in the mammalian retina. Exp Eye Res. JH, Kaushal S. Retinal degeneration 12 (rd12):
2008;86:713–26. a new, spontaneously arising mouse model for
17. Gupta N, Brown KE, Milam AH. Activated microg- human Leber congenital amaurosis (LCA). Mol Vis.
lia in human retinitis pigmentosa, late-onset retinal 2005a;11:152–62.
degeneration, and age-related macular degeneration. 29. Pang JJ, Chang B, Hawes NL, Hurd RE, Davisson
Exp Eye Res. 2003;76:463–71. MT, Li J, Noorwez SM, Malhotra R, McDowell JH,
18. Huang Y, Xu Z, Xiong S, Qin G, Sun F, Yang J, Yuan Kaushal S, Hauswirth WW, Nusinowitz S, Thompson
T-F, Zhao L, Wang K, Liang Y-X, Fu L, Wu T, So K-F, DA, Heckenlively JR. Retinal degeneration 12 (rd12):
Rao Y, Peng B. Dual extra-retinal origins of microglia a new, spontaneously arising mouse model for
in the model of retinal microglia repopulation. Cell human Leber congenital amaurosis (LCA). Mol Vis.
Discov. 2018;4:9–9. 2005b;11:152–62.
19. Jobling AI, Waugh M, Vessey KA, Phipps JA, Trogrlic 30. Peng B, Xiao J, Wang K, So KF, Tipoe GL, Lin
L, Greferath U, Mills SA, Tan ZL, Ward MM, Fletcher B. Suppression of microglial activation is neuropro-
EL. The role of the microglial Cx3cr1 pathway in tective in a mouse model of human retinitis pigmen-
the postnatal maturation of retinal photoreceptors. J tosa. J Neurosci. 2014;34:8139–50.
Neurosci. 2018;38:4708–23. 31. Pittler SJ, Baehr W. Identification of a nonsense muta-
20. Kameya S, Hawes NL, Chang B, Heckenlively JR, tion in the rod photoreceptor cGMP phosphodiester-
Naggert JK, Nishina PM. Mfrp, a gene encoding a ase beta-subunit gene of the rd mouse. Proc Natl Acad
frizzled related protein, is mutated in the mouse retinal Sci U S A. 1991;88:8322–6.
degeneration 6. Hum Mol Genet. 2002;11:1879–86. 32. Rashid K, Akhtar-Schaefer I, Langmann T. Microglia
21. Kohno H, Koso H, Okano K, Sundermeier TR, Saito in retinal degeneration. Front Immunol. 2019; 10:
S, Watanabe S, Tsuneoka H, Sakai T. Expression pat- 1975.
tern of Ccr2 and Cx3cr1 in inherited retinal degenera- 33. Ronning KE, Karlen SJ, Miller EB, Burns
tion. J Neuroinflammation. 2015;12:188. ME. Molecular profiling of resident and infiltrating
22. Li T, Aredo B, Chen X, Zhang K, Ufret-Vincenty mononuclear phagocytes during rapid adult retinal
R. Change in the distribution and phenotype of sub- degeneration using single-cell RNA sequencing. Sci
retinal microglia in C57BL/6J and RD8 mutant mice Rep. 2019;9:4858.
with aging. Invest Ophthalmol Vis Sci. 2014;55:81. 34. Santos AM, Calvente R, Tassi M, Carrasco MC,
23. Luhmann UF, Carvalho LS, Holthaus SM, Cowing Martin-Oliva D, Marin-Teva JL, Navascues J,
JA, Greenaway S, Chu CJ, Herrmann P, Smith AJ, Cuadros MA. Embryonic and postnatal develop-
Munro PM, Potter P, Bainbridge JW, Ali RR. The ment of microglial cells in the mouse retina. J Comp
severity of retinal pathology in homozygous Crb1rd8/ Neurol. 2008;506:224–39.
rd8 mice is dependent on additional genetic factors. 35. Sasahara M, Otani A, Oishi A, Kojima H, Yodoi Y,
Hum Mol Genet. 2015;24:128–41. Kameda T, Nakamura H, Yoshimura N. Activation of
24. Mattapallil MJ, Wawrousek EF, Chan CC, Zhao H, bone marrow-derived microglia promotes photore-
Roychoudhury J, Ferguson TA, Caspi RR. The Rd8 ceptor survival in inherited retinal degeneration. Am
mutation of the Crb1 gene is present in vendor lines J Pathol. 2008;172:1693–703.
of C57BL/6N mice and embryonic stem cells, and 36. Shivaram AM. Isolation and characterization of reti-
confounds ocular induced mutant phenotypes. Invest nal microglia in Royal College of Surgeons Rat. 2003.
Ophthalmol Vis Sci. 2012;53:2921–7. 37. Wang N-K, Lai C-C, Liu C-H, Yeh L-K, Chou CL,
25. O’Koren EG, Mathew R, Saban DR. Fate mapping Kong J, Nagasaki T, Tsang SH, Chien C-L. Origin
reveals that microglia and recruited monocyte-derived of fundus hyperautofluorescent spots and their
macrophages are definitively distinguishable by phe- role in retinal degeneration in a mouse model of
notype in the retina. Sci Rep. 2016;6:20636. Goldmann-­ Favre syndrome. Dis Model Mech.
26. O’Koren EG, Yu C, Klingeborn M, Wong AYW, 2013;6:1113–22.
Prigge CL, Mathew R, Kalnitsky J, Msallam RA, 38. Wang NK, Fine HF, Chang S, Chou CL, Cella W,
Silvin A, Kay JN, Bowes Rickman C, Arshavsky VY, Tosi J, Lin CS, Nagasaki T, Tsang SH. Cellular ori-
Ginhoux F, Merad M, Saban DR. Microglial func- gin of fundus autofluorescence in patients and mice
tion is distinct in different anatomical locations dur- with a defective NR2E3 gene. Br J Ophthalmol.
ing retinal homeostasis and degeneration. Immunity. 2009;93:1234–40.
2019;50:723–737.e727. 39. Wang T, Reingruber J, Woodruff ML, Majumder A,
27. O’Koren EG, Yu C, Klingeborn M, Wong AY, Prigge Camarena A, Artemyev NO, Fain GL, Chen J. The
CL, Mathew R, Kalnitsky J, Msallam RA, Silvin A, PDE6 mutation in the rd10 retinal degeneration mouse
Kay JN. Microglial function is distinct in different model causes protein mislocalization and instability
anatomical locations during retinal homeostasis and and promotes cell death through increased ion influx.
degeneration. Immunity. 2019;50(723–737):e727. J Biol Chem. 2018;293:15332–46.
Mini Review: The Role of Microglia in Inherited Retinal Diseases 205

40. Wang X, Zhao L, Zhang J, Fariss RN, Ma W, 42. Zeng H, Ding M, Chen X-X, Lu Q. Microglial NADPH
Kretschmer F, Wang M, Qian H, Badea TC, Diamond oxidase activation mediates rod cell death in the retinal
JS, Gan W-B, Roger JE, Wong WT. Requirement degeneration in rd mice. Neuroscience. 2014;275:54–61.
for microglia for the maintenance of synaptic func- 43. Zhao L, Zabel MK, Wang X, Ma W, Shah P, Fariss
tion and integrity in the mature retina. J Neurosci. RN, Qian H, Parkhurst CN, Gan WB, Wong
2016;36:2827–42. WT. Microglial phagocytosis of living photoreceptors
41. Zabel MK, Zhao L, Zhang Y, Gonzalez SR, Ma W, contributes to inherited retinal degeneration. EMBO
Wang X, Fariss RN, Wong WT. Microglial phago- Mol Med. 2015a;7:1179–97.
cytosis and activation underlying photoreceptor 44. Zhou T, Huang Z, Sun X, Zhu X, Zhou L, Li M,
degeneration is regulated by CX3CL1-CX3CR1 sig- Cheng B, Liu X, He C. Microglia polarization with
naling in a mouse model of retinitis pigmentosa. Glia. M1/M2 phenotype changes in rd1 mouse model of
2016;64:1479–91. retinal degeneration. Front Neuroanat. 2017;11:77.
CD68: Potential Contributor
to Inflammation and RPE Cell
Dystrophy

Mayur Choudhary and Goldis Malek

Abstract space and discuss observations on its localiza-


tion in a mouse model that presents with
Age-related macular degeneration (AMD) is AMD-like features.
the leading cause of visual impairment in the
elderly in developed countries. It is a complex, Keywords
multifactorial, progressive disease with
diverse molecular pathways, including inflam- Age-related macular degeneration ·
mation, regulating its pathogenesis. The Inflammation · CD68 · Retinal pigment
myeloid marker CD68 is a protein highly epithelial cells · Sub-retinal immune cells
expressed in circulating and tissue macro-
phages. Recent observations of immune mark-
ers in human AMD tissues have varied with 1 Introduction
some finding ectopic RPE cells in advanced
AMD and others noting negligible numbers of Age-related macular degeneration (AMD) is a
CD68-positive cells. Additionally, animal major cause of irreversible blindness affecting
models of retinal degeneration have shown elderly people worldwide [1]. It is a complex
upregulation of CD68, in a protective popula- neurodegenerative disease regulated by both
tion of retinal microglia. Herein, we review genetic and environmental risk factors [2].
the potential role of CD68 in regulating RPE Despite substantial advancement in our under-
health and inflammation in the sub-retinal standing of the pathogenesis of AMD, there are
multiple aspects related to its development and
progression, which remain to be investigated in
M. Choudhary
Department of Ophthalmology, Albert Eye Research detail. Though it is well known that the retinal
Institute, Duke University School of Medicine, pigment epithelium (RPE) supports cells ino the
Durham, NC, USA overlying neurosensory retina and underlying
G. Malek (*) choroid, and performs several vital functions
Department of Ophthalmology, Albert Eye Research including phagocytosis of photoreceptor outer
Institute, Duke University School of Medicine, segments, processing of visual-cycle metabolites,
Durham, NC, USA
maintenance of outer blood–retina barrier, and
Department of Pathology, Albert Eye Research secretion of growth factors, cytokines, and lipo-
Institute, Duke University School of Medicine,
Durham, NC, USA protein particles [3], the early molecular events
e-mail: gmalek@duke.edu precipitating RPE damage remain to be identified

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 207
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_30
208 M. Choudhary and G. Malek

and are an area of intense investigation. This has cessing and involvement in lysosomal/autophagy
been a challenge given the complexity of the clin- pathways. CD68 has been shown to bind
icopathology of RPE cells in the early and oxidized-­low density lipoproteins (oxLDL) in
intermediate stages of AMD including altered
­ cultured monocyte THP-1 cells, suggesting that it
RPE morphology, mediated by a number of fac- may function as a receptor for oxLDL, though
tors, including but not limited to accumulation of this function has not been confirmed in vivo [14–
lipid- and protein-rich deposits within and under- 16]. CD68 is also a well-known myeloid-specific
neath the RPE, modulation of cytokines in the surface marker, expressed abundantly by macro-
sub-retinal space, presence of sub-retinal immune phages [13]. In the posterior retina, CD68 expres-
cells, and epithelial-to-mesenchymal transition sion has been reported in primary human RPE
of RPE cells [2, 4–7]. These changes may eventu- cultures as well as freshly isolated RPE cells [17]
ally lead to RPE atrophy and fatally impact the in a predominantly granular perinuclear staining
overlying photoreceptor cells, as seen in late dry pattern similar to that observed in mononuclear
AMD, also referred to as geographic atrophy [2]. phagocytes [17]. Additionally, CD68 expression
Thus, it is imperative to understand the molecular has been studied in the retina of senescence-­
mechanisms that regulate AMD pathogenic accelerated OXYS rats, with CD68-positive cells
changes along the course of the disease, in order detected throughout much of the inner retina
to develop potential therapies to either decelerate including the retinal ganglion cell layer, inner
or prevent loss of RPE function. nuclear layer, inner plexiform layer, and outer
Hints to pathways that may trigger RPE dam- plexiform layer, but not the photoreceptor layer
age include studies demonstrating loss of retinoid [18]. Finally, CD68-positive cells are often
markers such as RPE65 (retinoid isomerohydro- immunopositive for the microglial marker Iba1,
lase) and CRALBP (cellular retinaldehyde-­ suggesting a potential immune cell origin for
binding protein) [8, 9] and altered expression of CD68-positive cells. These observations imply
immune markers such as CD68 and CD163, in that CD68 is expressed in immune cells as well
abnormal RPE cells as observed in human late-­ as RPE cells in the eye [18].
dry AMD retinal cross sections [10]. These RPE
changes have been attributed to transdifferentia-
tion. The focus of this mini review is on the role 3 CD68 Expression Is
of CD68 in regulating RPE cell homeostasis as Associated with RPE
well as the immune environment of the sub-­ Dystrophy and Inflammation
retinal space.
Infiltration of immune cells into the sub-retinal
space has been widely observed in human neo-
2 CD68 Expression in the Eye vascular AMD specimens as well in the experi-
mental choroidal neovascularization mouse
CD68, also known as macrosialin in mice, is a model, developed by a laser-induced breach of
heavily glycosylated type I transmembrane gly- Bruch’s membrane [19–24]. The presence of sub-­
coprotein, mainly associated with endosomal/ retinal immune cells has been recognized as an
lysosomal organelles [11, 12]. It is a member of important factor in the pathogenesis of CNV for-
the LAMP (lysosomal-associated membrane pro- mation [25], potentially sourced from the circula-
teins) family of glycoproteins and is also called tion, and the resting microglia residing in the
LAMP-4. Structurally, CD68 contains a single inner retina [26]. Notably, the presence of sub-­
LAMP-like domain, with two conserved disul- retinal immune cells has been associated with
fide bridges and a mucin-like domain [13]. both intermediate AMD and geographic atrophy
Interestingly, the function of CD68 is still not [25], as revealed by immunolocalization of
well defined, but its localization within late endo- immune markers including CD163+ and comple-
somes suggests a role in peptide transport/pro- ment factor-3+ cells throughout AMD donor tis-
CD68: Potential Contributor to Inflammation and RPE Cell Dystrophy 209

sue [10, 21, 27, 28]. With regard to CD68 as a in vitro experiments were performed in human
marker of immune cells, two studies reported primary RPE cells (93-year-old, female, passages
negligible numbers of CD68+ cells in retinal 4–6), which showed an upregulation of CD68 in
cross sections as a function of age and AMD, response to treatment with AMD-relevant oxi-
suggesting potential heterogeneity in its expres- dants, sodium iodate, and cigarette smoke extract
sion by immune cells in the retina [21, 29]. In (Fig. 3). The RPE cells again displayed a punc-
vivo studies using transgenic mice have provided tate, granular, and perinuclear staining pattern
further evidence for the involvement of immune consistent with the in vivo observations.
cells in disease progression. Aged mice with a Comparable results were observed in a second
genetic deletion of the aryl hydrocarbon receptor mouse model, which also presents with AMD-­
(Ahr), known for its role in toxin metabolism and like pathology, aged Liver-X-Receptor alpha–
clearance, develop human dry AMD-like fea- deficient (Lxra−/−) mice [31]. These mice
tures, including decreased retinal function, accu- (12–14 months) exhibit significant upregulation
mulation of thick sub-RPE basal laminar deposits, of sub-retinal immune cells, increase in proin-
and RPE and choroidal atrophy [24, 30]. These flammatory cytokines, as well as RPE dystrophy.
mice also present with a significantly larger neo- The sub-retinal cells observed in posterior RPE
vascular lesion in response to laser injury [24]. flatmounts stained positive for CD68 and the
Evaluation of the content of the CNV lesions microglial marker Iba1, associated with regions
revealed the presence of a greater number of of RPE dystrophy (data not shown). Collectively,
immune cells, suggesting that dysregulation of these results highlight the interconnection
inflammation plays a role in its etiology. Further between CD68 expression in sub-retinal immune
support for an Ahr-driven inflammatory role cells and RPE, and RPE injury and dystrophy.
comes from in vitro findings, in which knocking
down Ahr resulted in an increase in the produc-
tion of inflammatory cytokines and growth fac- 4 Unresolved Roles of CD68
tors in RPE and choroidal endothelial cells. in AMD Pathogenesis
Importantly, RPE flatmounts prepared from two
cohorts of aged Ahr−/− mice (12–14 months old In light of the discussed observations in murine
and 18–20 months old) exhibited significantly models and clinicopathology studies of human
higher number of sub-retinal immune cells, com- AMD, CD68 appears to be associated with ecto-
pared to age-matched wild-type mice, which pic RPE as well as RPE dystrophy [10]. The pres-
were associated with regions of RPE dystrophy ence of CD68-positive sub-retinal immune cells
(Fig. 1a, b). The flatmounts from both cohorts in the Ahr−/− and Lxra−/− mouse models, which
(12–14 months and 18–22 months) of Ahr−/− present with AMD-like features, adds another
mice also exhibited a significantly larger area layer of complexity in deciphering the function
with RPE dystrophy as compared to wild-type and role of CD68 in AMD pathogenesis indicat-
mice (Fig. 1c, d). When RPE flatmounts from the ing that CD68 should be investigated in the con-
Ahr−/− cohort were probed with an antibody to text of RPE cells, disease-associated sub-retinal
CD68, increased immunoreactivity was detected immune cells, keeping in mind that its role may
in RPE cells, in a granular, punctate perinuclear differ in mice versus humans [21, 29].
staining pattern. Additionally, an upregulation in Immune cells in the retina are derived from
the number of F4/80+ (marker for murine macro- two sources, resident microglia, which constitute
phages) and CD68+ sub-retinal immune cells about 85% of the immune cells in the retina, and
was observed. Interestingly, only 13–21% of the perivascular macrophages [26]. The retinal
F4/80+ immune cells were also CD68+, suggest- microglia are heterogeneous in nature and occupy
ing that there may be distinct populations of two distinct niches in the retina, the outer plexi-
immune cells in the sub-retinal space of these form layer and inner plexiform layer. The microg-
mice (Fig. 2). To identify stimulants for CD68, lia residing in the inner plexiform layer are
210 M. Choudhary and G. Malek

Fig. 1 Aged Ahr−/− mice exhibit a greater number of sub-­ significant). (c) Two representative high-magnification
retinal immune cells concomitant with diffuse RPE dys- images from flatmounts from two Ahr−/− mice showing
trophy. (a) Flatmounts from wild-type (Ahr+/+) and regions of RPE dystrophy. Ahr+/+ mice displayed normal
Ahr−/− mice stained with phalloidin (RPE cytoskeleton), RPE morphology (scale bar = 50 μm). (d) Quantification
F4/80 (murine macrophages), and Hoechst (cell nuclei) of percentage of flatmount area, which displayed RPE
(scale bar = 50 μm). (b) Quantification of F4/80+ cells in dystrophy (n = 4 per group, *p < 0.05, ns not significant)
Ahr+/+ and Ahr−/− mice (n = 4 per group, *p < 0.05, ns not

dependent on interleukin-34 (IL-34), a cytokine microenvironment in the sub-retinal space in


secreted by retinal ganglion cells, for their sur- response to aging as well as disease. In the sub-­
vival, whereas the microglia in the outer retinal space, they adhere to the apical side of the
­plexiform layer are independent of IL-34 signal- RPE cells. This upregulation of microglial cells
ing [32]. Aging and disease can also bring about in the sub-retinal space, associated with retinal
the migration of immune cells into the sub-retinal and RPE degenerative changes, has consistently
space, a known disease-associated niche [26]. been reported in several mouse models, including
Both populations of retinal microglia are the light degeneration model, sodium-iodate-­
central to the modulation of the immune-­ induced RPE injury model, RhoP23H/WT retinitis
CD68: Potential Contributor to Inflammation and RPE Cell Dystrophy 211

Fig. 2 Aged Ahr−/− mice exhibit a greater number of with antibodies to F4/80 (murine macrophages) and CD68
CD68+ and F4/80+ sub-retinal immune cells. (a) (immune cells) (scale bar = 50 μm). (b) Quantification of
Flatmounts from wild-type (Ahr+/+) and Ahr−/− mice CD68+ and F4/80+ cells in Ahr+/+ and Ahr−/− mice (n = 4
stained with phalloidin (RPE cytoskeleton) and probed per group, *p < 0.05)

Fig. 3 RPE cells exhibit an upregulation of CD68 immu- 4–6) treated with (a) DMSO control, (b) sodium iodate
nostaining in response to oxidant injury. Representative (NaIO3), and (c) cigarette smoke extract (CSE) (scale
images of RPE cells from a 93-year-old, female (passages bar = 20 μm)

pigmentosa model, as well as, in the Ahr−/− and have a distinct protective role in the sub-retinal
Lxra−/− knockout mouse models with AMD-like space in mouse models. Additional studies using
pathologies [24, 30–34]. In one study examining human donor tissue are needed to further cor-
retinal degeneration in mice following light-­ roborate this finding.
induced degeneration, a distinct population of CD68 may also be involved in regulating RPE
CD68+ retinal microglia in the sub-retinal space health and function. CD68 has been reported by
was found to express a number of other genes different groups to be upregulated in ectopic RPE
including Lgals3 (Galectin3), Lpl (lipoprotein cells in advanced AMD, downregulated in human
lipase), Fabp5 (fatty acid binding protein 5), AMD samples, as well as expressed in mouse
Apoe (Apolipoprotein e), Lilr4b (leukocyte models exhibiting AMD-like features (Ahr−/− and
immunoglobulin-like receptor B4), Trem2 (trig- Lxra−/−) [10, 21, 23, 24, 29, 31]. Our studies have
gering receptor expressed on myeloid cells 2), brought to light that aged Ahr−/− mice display an
Cstb (cystatin B), and Sqstm1 (sequestosome 1) upregulation of CD68 immunostaining in regions
[32]. Importantly, it was shown that this microg- displaying RPE dystrophy, leading one to con-
lial population protects RPE from disease-­ clude that CD68 may regulate multiple homeo-
associated damage, in light degeneration as well static pathways in RPE cells, which may extend
as the RhoP23H/WT retinitis pigmentosa model [32]. to transdifferentiation, epithelial-to-­mesenchymal
Therefore, it is postulated that microglia may transition, inflammation, autophagy, and lyso-
212 M. Choudhary and G. Malek

some function. It remains to be determined 11. Holness CL, Simmons DL. Molecular cloning of
CD68, a human macrophage marker related to lyso-
whether CD68 expression is upregulated prior to somal glycoproteins. Blood. 1993;81(6):1607–13.
the appearance of RPE cell dystrophy or is a con- 12. Holness CL, da Silva RP, Fawcett J, Gordon S,
sequence of stressed RPE cells attempting to res- Simmons DL. Macrosialin, a mouse macrophage-­
cue its phenotype by pushing autophagy and restricted glycoprotein, is a member of the lamp/lgp
family. J Biol Chem. 1993;268(13):9661–6.
lysosomal pathways into overdrive. Detailed 13. Chistiakov DA, Killingsworth MC, Myasoedova
investigation is warranted to address the role of VA, Orekhov AN, Bobryshev YV. CD68/macrosia-
CD68 in the context of retinal microglia as well lin: not just a histochemical marker. Lab Investig.
as RPE cells. 2017;97(1):4–13.
14. Kurushima H, Ramprasad M, Kondratenko N, Foster
DM, Quehenberger O, Steinberg D. Surface expres-
Acknowledgements This study was supported by NEI sion and rapid internalization of macrosialin (mouse
grants: EY027802 (GM), EY028160 (GM), EY032751 CD68) on elicited mouse peritoneal macrophages. J
(GM), and EY005722 (Duke Eye Center) and Research to Leukoc Biol. 2000;67(1):104–8.
Prevent Blindness core grant (Duke Eye Center). 15. van der Kooij MA, von der Mark EM, Kruijt JK,
van Velzen A, van Berkel TJ, Morand OH. Human
monocyte-derived macrophages express an approxi-
mately 120-kD Ox-LDL binding protein with strong
References identity to CD68. Arterioscler Thromb Vasc Biol.
1997;17(11):3107–16.
1. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng 16. Ramprasad MP, Terpstra V, Kondratenko N,
CY, et al. Global prevalence of age-related macular Quehenberger O, Steinberg D. Cell surface expres-
degeneration and disease burden projection for 2020 sion of mouse macrosialin and human CD68 and
and 2040: a systematic review and meta-analysis. their role as macrophage receptors for oxidized
Lancet Glob Health. 2014;2(2):e106–16. low density lipoprotein. Proc Natl Acad Sci U S A.
2. Ambati J, Fowler BJ. Mechanisms of age-related 1996;93(25):14833–8.
macular degeneration. Neuron. 2012;75(1):26–39. 17. Elner SG, Elner VM, Nielsen JC, Torczynski E,
3. Lakkaraju A, Umapathy A, Tan LX, Daniele L, Philp Yu R, Franklin WA. CD68 antigen expression by
NJ, Boesze-Battaglia K, et al. The cell biology of human retinal pigment epithelial cells. Exp Eye Res.
the retinal pigment epithelium. Prog Retin Eye Res. 1992;55(1):21–8.
2020;78:100846. 18. Telegina DV, Kozhevnikova OS, Bayborodin SI,
4. Ambati J, Atkinson JP, Gelfand BD. Immunology of Kolosova NG. Contributions of age-related altera-
age-related macular degeneration. Nat Rev Immunol. tions of the retinal pigment epithelium and of glia
2013;13(6):438–51. to the AMD-like pathology in OXYS rats. Sci Rep.
5. Wang L, Clark ME, Crossman DK, Kojima K, 2017;7:41533.
Messinger JD, Mobley JA, et al. Abundant lipid 19. Gehrs KM, Heriot WJ, de Juan E Jr. Transmission
and protein components of drusen. PLoS One. electron microscopic study of a subretinal choroidal
2010;5(4):e10329. neovascular membrane due to age-related macular
6. Curcio CA, Johnson M, Rudolf M, Huang JD. The degeneration. Arch Ophthalmol. 1992;110(6):833–7.
oil spill in ageing Bruch membrane. Br J Ophthalmol. 20. Seregard S, Algvere PV, Berglin L. Immuno­
2011;95(12):1638–45. histochemical characterization of surgically removed
7. Curcio CA, Johnson M, Huang JD, Rudolf M. Aging, subfoveal fibrovascular membranes. Graefes Arch
age-related macular degeneration, and the response-­ Clin Exp Ophthalmol. 1994;232(6):325–9.
to-­retention of apolipoprotein B-containing lipopro- 21. Lad EM, Cousins SW, Van Arnam JS, Proia
teins. Prog Retin Eye Res. 2009;28(6):393–422. AD. Abundance of infiltrating CD163+ cells in the
8. Kiser PD. Retinal pigment epithelium 65 kDa pro- retina of postmortem eyes with dry and neovascular
tein (RPE65): an update. Prog Retin Eye Res. age-related macular degeneration. Graefes Arch Clin
2021;88:101013. Exp Ophthalmol. 2015;253(11):1941–5.
9. Xue Y, Shen SQ, Jui J, Rupp AC, Byrne LC, Hattar 22. Sennlaub F, Auvynet C, Calippe B, Lavalette S,
S, et al. CRALBP supports the mammalian reti- Poupel L, Hu SJ, et al. CCR2(+) monocytes infiltrate
nal visual cycle and cone vision. J Clin Invest. atrophic lesions in age-related macular disease and
2015;125(2):727–38. mediate photoreceptor degeneration in experimental
10. Cao D, Leong B, Messinger JD, Kar D, Ach T, subretinal inflammation in Cx3cr1 deficient mice.
Yannuzzi LA, et al. Hyperreflective foci, optical EMBO Mol Med. 2013;5(11):1775–93.
coherence tomography progression indicators in age-­ 23. Choudhary M, Safe S, Malek G. Suppression of aber-
related macular degeneration, include transdifferenti- rant choroidal neovascularization through activation
ated retinal pigment epithelium. Invest Ophthalmol of the aryl hydrocarbon receptor. Biochim Biophys
Vis Sci. 2021;62(10):34. Acta Mol basis Dis. 2018;1864(5 Pt A):1583–95.
CD68: Potential Contributor to Inflammation and RPE Cell Dystrophy 213

24. Choudhary M, Kazmin D, Hu P, Thomas RS, age-related macular degeneration eyes. Clin Exp
McDonnell DP, Malek G. Aryl hydrocarbon recep- Ophthalmol. 2013;41:129.
tor knock-out exacerbates choroidal neovascular- 30. Hu P, Herrmann R, Bednar A, Saloupis P, Dwyer MA,
ization via multiple pathogenic pathways. J Pathol. Yang P, et al. Aryl hydrocarbon receptor deficiency
2015;235(1):101–12. causes dysregulated cellular matrix metabolism and
25. Guillonneau X, Eandi CM, Paques M, Sahel JA, age-related macular degeneration-like pathology.
Sapieha P, Sennlaub F. On phagocytes and macular Proc Natl Acad Sci U S A. 2013;110(43):E4069–78.
degeneration. Prog Retin Eye Res. 2017;61:98–128. 31. Choudhary M, Ismail EN, Yao PL, Tayyari F, Radu
26. Yu C, Roubeix C, Sennlaub F, Saban DR. Microglia RA, Nusinowitz S, et al. LXRs regulate features of
versus monocytes: distinct roles in degen- age-related macular degeneration and may be a poten-
erative diseases of the retina. Trends Neurosci. tial therapeutic target. JCI Insight. 2020;5(1):131928.
2020;43(6):433–49. 32. O’Koren EG, Yu C, Klingeborn M, Wong AYW,
27. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum Prigge CL, Mathew R, et al. Microglial function is
SR, Lambris JD, et al. Drusen complement compo- distinct in different anatomical locations during
nents C3a and C5a promote choroidal neovasculariza- retinal homeostasis and degeneration. Immunity.
tion. Proc Natl Acad Sci U S A. 2006;103(7):2328–33. 2019;50(3):723–37 e7.
28. Gupta N, Brown KE, Milam AH. Activated microg- 33. Reyes NJ, O’Koren EG, Saban DR. New insights into
lia in human retinitis pigmentosa, late-onset retinal mononuclear phagocyte biology from the visual sys-
degeneration, and age-related macular degeneration. tem. Nat Rev Immunol. 2017;17(5):322–32.
Exp Eye Res. 2003;76(4):463–71. 34. O’Koren EG, Mathew R, Saban DR. Fate mapping
29. Madigan MC, van den Berg C, Jager MJ, Provis reveals that microglia and recruited monocyte-derived
JM. Expression of macrophage markers and C3d in macrophages are definitively distinguishable by phe-
central and peripheral choroid of young, aged and notype in the retina. Sci Rep. 2016;6:20636.
Gene Expression of Clusterin,
Tissue Inhibitor
of Metalloproteinase-1, and Their
Receptors in Retinal Pigment
Epithelial Cells and Müller Glial
Cells Is Modulated
by Inflammatory Stresses

Mengmei Zheng, Eun-Jin Lee, Shinwu Jeong,


and Cheryl Mae Craft

Abstract levels of the two genes with quantitative real-­


time PCR (qRT-PCR) with interleukin (IL)-1β
Balanced activities of matrix metalloprotein- treatment, a key pathological component in
ases (MMPs) and their inhibitors are essential retinal degeneration. Endogenous CLU gene
for photoreceptor (PR) cell survival. PR rod expression was significantly downregulated
cell survival in rodent models of inherited reti- by IL-1β in both cell types, whereas TIMP-1
nitis pigmentosa (RP) is prolonged by recom- expression was upregulated in MG cells, sug-
binant tissue inhibitor of metalloproteinase gesting the transcriptional control of CLU is
(TIMP)-1 or clusterin (CLU) proteins. Retinal potentially more sensitive to inflammatory
pigment epithelial cells (RPE) and Müller glia conditions. The expression levels of CLU
(MG) cells support PR cells. In human RPE endocytic receptors revealed that the low-­
and MG cell lines, we measured their mRNA density lipoprotein receptor-related protein 2
(LRP2) was upregulated only in MG cells by
the treatment with no detectable change in
RPE cells. Like LRP2, IL-1β upregulated
TIMP-1 receptor LRP1 expression in MG
M. Zheng · S. Jeong · C. M. Craft (*) cells; however, it was decreased in the expres-
Mary D. Allen Vision Research Laboratory, USC
sion of RPE cells. These data suggest that the
Roski Eye Institute, Department of Ophthalmology,
Keck School of Medicine of the University of gene expression of CLU and TIMP-1 and their
Southern California, Los Angeles, CA, USA receptors may be dynamically modulated in
e-mail: CherylMae.Craft@med.usc.edu inflammatory conditions.
E.-J. Lee
Mary D. Allen Vision Research Laboratory, USC Keywords
Roski Eye Institute, Department of Ophthalmology,
Keck School of Medicine of the University of
Southern California, Los Angeles, CA, USA Retinal degeneration · Cytokine · Oxidative
stress · Clusterin · Tissue inhibitor of metal-
Department of Ophthalmology, Stanford University,
Palo Alto, CA, USA loproteinase 1 · Matrix metalloproteinase

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 215
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_31
216 M. Zheng et al.

1 Introduction grown in six-well plates subconfluently before


treating the cells with interleukin (IL)-1β at
Retinal Müller glial (MG) cells are the most 10 ng/mL for 24 h in FBS-free media. Total RNA
abundant resident radial glial cells and are closely was prepared using TRI Reagent (Sigma Aldrich,
associated with all neuronal cell types in the ret- St. Lois, MO), and High-Capacity RNA-to-­
ina. In contrast, retinal pigment epithelial (RPE) cDNA™ Kit (ThermoFisher Sci., Chino, CA)
cells are differentiated monolayer cells, apically was used for cDNA synthesis. Quantitative
lining photoreceptor (PR) cells. Both MG and Polymerase Chain Reaction (QPCR) was per-
RPE cells are essential for PR’s function and sur- formed on QuantStudio 6 Flex Real-Time PCR
vival [1, 2]. The integrity and function of retinal System (Applied Biosystems, Waltham, MA)
neurons are determined, not only by an internal using gene transcript-specific oligo DNA pri­
balance between survival and death signaling, but mers for the expression of CLU (Fwd
also by a homeostatic maintenance of retinal 5′-ACCAGTACTATCTGCGGGTCA-3′, Rev
architecture composed of multiple neuronal cell 5′-CGTCACAGTGATGGGATCAG-3′), TIMP-1
types and surrounding extracellular matrix (Fwd 5′-CTGCAATTCCGACCTCGTC-3′, Rev
(ECM), which is a target of modulation by pro- 5′-GGAAGTATCCGCAGACACTCTC-3′),
teinases such as matrix metalloproteinases LRP-1 (Fwd 5′-CGAGGCCCCTGAGATTT
(MMPs). GT-3′, Rev 5′-CATCGAGTGTGGGGACAC-3′),
By showing prolonged rod cell survival with LRP-2 (5′-CCTCCCCAAATGCAAGTGT-3′,
the treatments of recombinant clusterin (CLU) Rev 5′-GCCCCTGATCTGAAGGTCA-3′), and
and tissue inhibitor of metalloproteinase β-Actin (Fwd 5′- AGGCACCAGGGCGTG-3′,
(TIMP)-1 proteins, our published work using Rev 5′-CCCGTCACCGGAGTCC-3′) genes.
human-inherited RP models, the rd1 mouse and Quantification of relative amounts of transcripts
S334ter-line-3 transgenic rat, demonstrated that was done using a delta-delta Ct method and
MMP inhibitors have both a positive neuronal β-actin transcription as an internal normalization
cell survival effect and a therapeutic potential to control. Student’s t-test was performed to exam-
slow rod cell death [3–5]. Furthermore, both ine the differences among the control and test
exogenous treatments exert their functions as cell groups, using GraphPad Prism6 (La Jolla, CA,
signaling molecules by binding to their cell sur- USA). The statistical values are presented as
face receptors, including low-density lipoprotein mean ± standard error of the mean (SEM). The
receptor-related protein (LRP) 1 for TIMP-1 and difference between the means of separate treat-
LRP2 for CLU [6–8]. ment groups was considered statistically signifi-
To gain insight into how TIMP-1 and CLU cant at p-value (p) <0.05.
and their receptors may function, we investigated
their transcriptional regulation in vitro in MG and
RPE cells under an inflammatory condition. 3 Results

PR cells are surrounded radially by MG and api-


2 Methods cally by RPE cells. Both cell types play vital
functions to maintain PR function and survival,
2.1 Cytokine Treatments responding to inflammatory stress occurred upon
and Quantitative RT-PCR the onset of retinal degeneration. To gain insight
into how an inflammatory stress affects the gene
Human MG cell line MIO-M1 and RPE cell line expression from these cells, we adopted in vitro
ARPE-19 were maintained in regular Dulbecco’s culture system using MIO-M1 and ARPE-19,
Modified Eagle’s Medium (DMEM) containing which are spontaneously established cell lines
10% fetal bovine serum (FBS). These cells were from in vitro human primary cell cultures of MG
Gene Expression of Clusterin, Tissue Inhibitor of Metalloproteinase-1, and Their Receptors in Retinal… 217

LRP2 Relative Expression Level


b

CLU Relative Expression Level


a 1.5 b 5

4
1.0
3

2
0.5
1

0 0
IL-1b - + - + IL-1b - + - +
MIO-M1 ARPE-19 MIO-M1 ARPE-19
TIMP-1 Relative Expression Level

LRP1 Relative Expression Level


c 3 d 1.5

2 1.0

1 0.5

0 0
IL-1b - + - + IL-1b - + - +
MIO-M1 ARPE-19 MIO-M1 ARPE-19

Fig. 1a-d Sub-confluent MIO-M1 and ARPE-19 cells tive expression amounts of gene transcripts for CLU (a),
were untreated (−) or treated (+), each in triplicate, with LRP2 (b), TIMP-1 (c), and LRP1 (d), the level of β-actin
IL-1β at 10 ng/mL for 1 day before extracting total RNA, transcripts was used as internal control for normalization.
which was then used for cDNA synthesis. QPCR was per- The statistical significance level was calculated using
formed with gene-specific primer sets. To assess the rela- Students’ t-test: *p < 0.05; **p < 0.01; ****p < 0.0001

and RPE cells, respectively. Cells were treated


with IL-1β at 10 ng/mL for 24 h, and gene 4 Discussion
­transcription was measured by quantitative PCR
using gene transcript-specific DNA primers. In Pathological progress of inherited retinal degen-
Fig. 1a, with IL-1β treatment, the level of CLU eration develops with increased levels of disease
transcripts was significantly reduced by 25% and factors such as inflammatory and oxidative
32% in MIO-M1 and ARPE-19, respectively. stresses and leads to an alteration of retinal archi-
However, its receptor LRP2 was increased by tecture [9, 10]. CLU and TIMP-1 share the MMP
223% in MIO-M1 cells, with no change in inhibitory activity with their own distinct proper-
ARPE-­19 (Fig. 1b). In Fig. 1c, TIMP-1 transcrip- ties [6, 11, 12]. CLU has versatile chaperone
tion was highly augmented by 154% in MIO-M1 functions to attenuate extracellular stress by
cells by the treatment. TIMP-1 receptor LRP1 clearing dead cells or macromolecular aggre-
expression level was increased by 28% in MIO- gates; TIMP-1 acts as signaling molecule to pro-
M1 cells but decreased by 20% in ARPE-19 cells mote cell survival. Consistent with these
(Fig. 1d). pro-survival properties of these proteins, we
218 M. Zheng et al.

demonstrated that recombinant CLU and TIMP-1 cell surface, promoting the cells to clear dead PR
proteins extend rod cell survival in RP animal cells and restore the protein homeostasis in the
models [3–5]. However, how they function in the retina. CLU may also work indirectly for PR cell
retina is still unresolved. To gain insight into how survival, by acting as a signaling molecule
these two proteins work in vivo, the regulation of through LRP2 [17], to modulate MG cells to
expression in RPE and MG cells, which are express neuroprotective molecules to neurons. It
essential to supporting PR cell survival, was will be interesting to see if any changes occur in
investigated in vitro. transcriptome or proteome profiles of the MG
CLU has discrete domains, each responsible cells after CLU treatment.
for binding to the receptor LRP-2 and stressed or TIMPs act as inhibitors of MMPs and play a
unstressed proteins [13]. LRP2 expression is cytokine-like role by binding to membrane recep-
detected mainly in RPE and MG cells [14, 15]. tors, including LRP-1 [6, 7]. In contrast to CLU,
We also observed LRP2 expression in Long-­ the TIMP-1 expression was upregulated by the
Evans rat retina at the RPE layer and inner seg- cytokine treatment (Fig. 1c). We recently reported
ment region where MG apical processes are that IL-1β treatment does not change the level of
normally localized (data not shown). MG cells TIMP-1 secretion from MIO-M1 cells but results
are one of the primary responders to the onset of in its intracellular aggregate formation [18]. The
rod cell death in the RP retina, showing reactive current observation of the significant upregulation
gliosis [16]. of TIMP-1 at the transcriptional level may be
In our study, MG cells show highly increased explained by the possibility that the TIMP-1 tran-
expression of the CLU receptor LRP2 with cyto- script is more susceptible to post-transcriptional
kine treatment (Fig. 1b), whereas endogenous degradation under cytokine treatment, and/or that
CLU expression is downregulated in both MIO-­ the increased protein product translated due to the
M1 and ARPE-19 cells under the same treatment increased mRNA levels may be subjected to the
(Fig. 1a). CLU was also down-regulated by oxi- aggregation, attenuating the level of its secretion.
dative stress (H2O2 treatment; data not shown). Since TIMP-1 has pleiotropic cytokine-like sig-
These results suggest that CLU expression is sen- naling activity that plays a critical role in cellular
sitive to pathological factors. Contrary to the cur- activities, its aggregation may affect the response
rent gene regulation in human RPE and MG cells of MG cells to inflammatory stress in the retina.
in vitro, we previously observed an increase in The opposite pattern of LRP1 transcriptional reg-
CLU protein level in rat RP retina [3]. This may ulation shown in MG and RPE cells by the same
reflect the complexity of the CLU gene expres- treatment reflects the complicated in vivo situa-
sion regulation in the retina, spatiotemporally tion that occurs in the RP retina (Fig. 1d).
involving more diverse retinal cell types and fac- In summary, the expressions of CLU and
tors. On the other hand, LRP2 expression is TIMP-1 and their receptors are regulated differ-
increased in MG cells, which could potentially ently by a single cytokine. The current results
recruit more secreted CLU molecules from other suggest that each of the genes may be regulated
cells to MG cells. Consistently, in the RP retina spatiotemporally during RP progression with
of S334-ter-line, increased CLU immunoreactiv- inflammatory or oxidative stress, thus possibly
ity was detected and associated with MG pro- providing different local environmental impacts
cesses, compared with WT control rats [3]. Both in the RP retina of a multi-factorial inflammatory
MG and RPE are important for clearance or nature. From the therapeutic perspective, one
phagocytosis of released PR membrane. In light way to overcome this heterogeneity of RP pathol-
of CLU’s function of clearing dead cells or ogy in treating the diseased retina may be a com-
stressed proteins [11], it is intriguing to propose bined therapy with diverse neuroprotective
that being intravitreally injected recombinant therapeutic candidates; for example, the com-
CLU proteins may not only act against MMPs’ bined treatment, simultaneously or sequentially,
destructive activity but also be recruited to MG of recombinant CLU and TIMP-1 proteins, both
Gene Expression of Clusterin, Tissue Inhibitor of Metalloproteinase-1, and Their Receptors in Retinal… 219

of which are neuroprotective with different 8. Moezzi SM, et al. Apolipoprotein J in Alzheimer’s dis-
ease: shedding light on its role with cell signaling path-
mechanisms. It may be warranted to investigate way perspective and possible therapeutic approaches.
how CLU or TIMP-1 proteins interact with MG ACS Chem Neurosci. 2020;11(24):4060–72.
and RPE cells to benefit their roles for these cells 9. Jones BW, et al. Retinal remodeling in human retinitis
in normal and diseased retinas. pigmentosa. Exp Eye Res. 2016;150:149–65.
10. McMurtrey JJ, Tso MOM. A review of the immuno-
logic findings observed in retinitis pigmentosa. Surv
Acknowledgments We thank Dr. Hossein Ameri for pro- Ophthalmol. 2018;63(6):769–81.
viding the MOMI cell line. This study was supported, in 11. Humphreys DT, et al. Clusterin has chaperone-like
part, by Mary D. Allen Foundation; Dorie Miller; unre- activity similar to that of small heat shock proteins. J
stricted Grant to the Department of Ophthalmology from Biol Chem. 1999;274(11):6875–81.
Research to Prevent Blindness, New York, NY, and the 12. Jeong S, et al. Interaction of clusterin and matrix
NIH-NEI P30EY029220. metalloproteinase-9 and its implication for epithe-
lial homeostasis and inflammation. Am J Pathol.
2012;180(5):2028–39.
13. Lakins JN, et al. Evidence that clusterin has discrete
References chaperone and ligand binding sites. Biochemistry.
2002;41(1):282–91.
1. de Hoz R, et al. Retinal macroglial responses in health 14. Christensen EI, Birn H. Megalin and cubilin: mul-
and disease. Biomed Res Int. 2016;2016:2954721. tifunctional endocytic receptors. Nat Rev Mol Cell
2. Strauss O. The retinal pigment epithelium in visual Biol. 2002;3(4):256–66.
function. Physiol Rev. 2005;85(3):845–81. 15. Nuutinen T, et al. Clusterin: a forgotten
3. Vargas A, et al. Protective effect of clusterin on rod player in Alzheimer’s disease. Brain Res Rev.
photoreceptor in rat model of retinitis pigmentosa. 2009;61(2):89–104.
PLoS One. 2017;12(8):e0182389. 16. Hippert C, et al. Muller glia activation in response
4. Kim HS, et al. Tissue inhibitor of metalloproteinases to inherited retinal degeneration is highly varied and
1 enhances rod survival in the rd1 mouse retina. PLoS disease-specific. PLoS One. 2015;10(3):e0120415.
One. 2018;13(5):e0197322. 17. Li Y, et al. Differential functions of members of the
5. Shin JA, et al. Inhibition of matrix metalloproteinase 9 low density lipoprotein receptor family suggested
enhances rod survival in the S334ter-line3 retinitis pig- by their distinct endocytosis rates. J Biol Chem.
mentosa model. PLoS One. 2016;11(11):e0167102. 2001;276(21):18000–6.
6. Ries C. Cytokine functions of TIMP-1. Cell Mol Life 18. Lee EJ, et al. Matrix metalloproteinase-9 (MMP-9)
Sci. 2014;71(4):659–72. and tissue inhibitor of metalloproteinases 1 (TIMP-­
7. Grunwald B, Schoeps B, Kruger A. Recognizing 1) are localized in the nucleus of retinal Muller glial
the molecular multifunctionality and interactome of cells and modulated by cytokines and oxidative stress.
TIMP-1. Trends Cell Biol. 2019;29(1):6–19. PLoS One. 2021;16(7):e0253915.
Part VII
Mechanisms of Degeneration
Axonal Transport Defects
in Retinal Ganglion Cell Diseases

Iskalen Cansu Topcu Okan, Fatma Ozdemir,


and Cavit Agca

Abstract Abbreviations

For the survival and maintenance of retinal ALS Amyotrophic lateral sclerosis
ganglion cells (RGCs), axonal transportation ATP Adenine triphosphate
is fundamental. Axonal transportation defects CTB Cholera toxin B
can cause severe neuropathies leading to neu- GC Glucocorticoid
ronal loss. Axonal transport defects usually DEX Dexamethasone-21-acetate
precede axonal degeneration and RGC loss in DOA Dominant optic atrophy
disease models. To date, the main causes of IOP Intraocular pressure
axonal transport defects have not been fully LGN Lateral geniculate nucleus
understood. Therefore, elucidation of the LHON Leber’s hereditary optic neuropathy
mechanisms that lead to transport defects will NMDA N-methyly-D-aspartate
help us to develop novel therapeutic targets OPA 1 Optic atrophy 1
and early diagnostic tools. In this review, we POAG Primary open-angle glaucoma
provide an overview of optic neuropathies and RNFL Retinal nerve fiber layer
axonal degeneration with a focus on axonal RGC Retinal ganglion cell
transport. SC Superior colliculus
TEM Transmission electron microscopy
Keywords TTc Tetanus toxin

Axonal transport · Axonal degeneration ·


Glaucoma · OPA1 · Mitochondria 1 Introduction

Axonal transport, or axoplasmic flow, is the pro-


I. C. T. Okan · F. Ozdemir cess by which motor proteins actively travel
Molecular Biology, Genetics and Bioengineering along microtubules to deliver various cargoes,
Program, Sabanci University, Istanbul, Turkey
such as mitochondria, lipids, synaptic vesicles,
C. Agca (*) and other organelles. In this process, neurons
Molecular Biology, Genetics and Bioengineering
Program, Sabanci University, Istanbul, Turkey shuttle miscellaneous substances in a bidirec-
tional fashion along the axonal microtubule,
Nanotechnology Research and Application Center
(SUNUM), Sabanci University, Istanbul, Turkey which is considered essential for the function and
e-mail: cavit.agca@sabanciuniv.edu the survival of neurons. Depending on the

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 223
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_32
224 I. C. T. Okan et al.

d­ irection of material flow, this physiological pro- eration arises when the nerve fibers are damaged
cess is divided into two categories: anterograde and it takes place at the site of the lesion that
(from the cell body to the axon terminal) and ret- occurs in the neuron towards the distal end of the
rograde (from the axon terminal to the neuronal axon. Retrograde degeneration occurs when the
soma) axonal transport [1]. While anterograde degeneration spreads from the proximal site
transport is driven by the kinesin superfamily, (lesion site) along the axon to the cell soma caus-
retrograde axonal transport is dependent on the ing RGC death [30]. This degeneration can be
cytoplasmic dyneins [2, 3]. detected in retinal neurodegenerative diseases by
Axonal transport in RGCs is a sensitive phe- the changes in the thickness of the retinal nerve
nomenon due to the regulation of homeostasis as fiber layer (RNFL) [10].
a requirement for ATP and oxygen consumption. Despite the major factor of optic pathologies
Since RGCs have a relatively larger cell body and being established as degeneration of RGC axons
a unique cytostructure, there is an uneven energy and their soma, the main mechanism whether the
distribution and consumption in RGCs compared axonal degeneration precedes the RGC loss or
to other neuronal cells [4]. Therefore, RGCs are not has been not fully understood. There is an
essentially considered to be more vulnerable to increasing number of supportive studies showing
degeneration [5, 6]. Any disruption or deficiency that axonal degeneration occurs before cell death
in axonal transport in RGCs possibly results in and the same conclusion is applicable in optic
neurodegenerative pathologies causing irrevers- neuropathies like glaucoma [8]. In primate mod-
ible vision loss [7]. Thus, defects in axonal trans- els [11] and also in human glaucoma studies [12],
port have become an intriguing topic. it was also shown that disruption of axonal trans-
Another critical issue that has to be addressed port precedes RGC cell death in both anterograde
is the assessment of the axonal degeneration and and retrograde transports. In a recent study, simi-
transport defects. It is essential to detect early lar results were obtained using an excitotoxic rat
defects to prevent axonal degeneration and RGC retinopathy model generated by N-methyl-D-­
loss. Thus, early diagnostics of optic neuropa- aspartate (NMDA) delivery. Through the in vivo
thies in RGC and their axons has become an imaging, the loss of axonal transport that occurred
emerging field. Unfortunately, available clinical before the retinal cell death was demonstrated
imaging techniques are insufficient as they tend using tetanus toxin (TTc) [8], a fast axonal trans-
to show morphological changes only when vision port biomarker for neuronal imaging [13].
loss occurs late in the course of the disease [8]. Overall, the importance of understanding the
Yet, there is an unmet diagnostic need to detect interrelationship of transport deficits and axonal
the early stages of optic neuropathies before neu- degeneration has been put forward along with the
ronal and visual loss. need for early diagnostic tools for optic
neuropathies.

2 Axonal Degeneration
and Transport Defects 3 Optic Neuropathies

Mutations in coding genes of the crucial compo- Optic neuropathy is attributed to the degeneration
nents of axonal transport machinery lead to well-­ of the optic nerve and more commonly, it leads to
known neurodegenerative diseases, including characteristic changes in RNFL, optic disc, and
amyotrophic lateral sclerosis (ALS), parkinson- axons of RGC. Subsequently, the degeneration in
ism, and Alzheimer’s disease [9]. Unlike existing the axons causes apoptosis of RGC and thus,
optic neuropathies, they are also well character- vision loss may develop [14, 15]. RGCs are quite
ized by deficiencies in axonal transport. In optic vulnerable to mitochondrial disorders. The two
neuropathies, axonal degeneration can occur bidi- well-known hereditary optic neuropathies,
rectionally [30]. Anterograde (Wallerian) degen- Dominant Optic Atrophy (DOA), and Leber
Axonal Transport Defects in Retinal Ganglion Cell Diseases 225

hereditary optic neuropathy (LHON) are related 4 Animal Models and Axonal
to mitochondrial dysfunction [16]. Dominant Transport Defects
Optic Atrophy (DOA) is an autosomal inherited
optic nerve disorder where RGCs and their axons DBA/2 J is a mouse model which mimics the
forming the optic nerve are severely impaired. human primary open-angle glaucoma (POAG)
About 50–60% of cases are related to optic atro- condition due to elevated intraocular pressure
phy 1 (OPA1) gene [17] and the majority of the (IOP) [34]. Therefore, it is commonly used as an
mutations result in OPA1 haploinsufficiency animal model for glaucoma. A recent study using
[18]. OPA1 is a mitochondrial membrane protein fluorescently labeled Cholera toxin B (CTB) for
involved in mitochondrial cristae structure, and anterograde labeling of RGC or retrograde label-
fusion of the outer membrane, together with sev- ing through intracranial injections to retinorecipi-
eral other mitochondrial functions [16, 19]. ent brain regions showed axonal transport defects
Likewise, Leber’s hereditary optic neuropathy in the DBA/2 J animals [6]. In these studies, IOP
(LHON) is a mitochondrial disease due to mito- is surprisingly reported as an associated factor of
chondrial DNA mutations, which selectively glaucoma but aging turns out to be the main risk
degenerate RGCs [20]. Despite all the neurons factor. This was demonstrated in 3- to 10-month-
having the same mutations and mitochondrial old animals as the gradual increase of IOP levels
dysfunction, the selective loss of RGC in both further contributed to degeneration. At 11 months
hereditary optic neuropathies may be a result of of age, complete failure of axonal transport was
RGC-specific features. Therefore, this peculiar also reported [23]. Furthermore, 9–10 month-old
phenomenon may be attributed to lack of myelin- animals showed 69% and 23% reduction for
ation in lamina cribrosa, which requires excess anterograde and retrograde axonal transports,
energy and thus results in the accumulation of respectively. These findings already confirmed
mitochondria in the region. In simple terms, hav- that axonal transport deficiency was predegener-
ing mitochondrial dysfunction makes RGC more ative and preceded the RGC loss. Moreover, it
prone to selective degeneration. was shown that the RGC axons and presynaptic
Glaucoma is another type of optic neuropathy terminals were intact in Superior Colliculus (SC)
that occurs in retinal cells causing irreversible albeit with severe anterograde transport defects
vision loss by damaging the optic nerves [32]. It [6].
is a worldwide disease and approximately 111.8 As mentioned previously, DOA is an autoso-
million people will suffer from this disease by mal inherited optic nerve disorder where RGCs,
2040, which already reached 64.8 million people and their axons forming the optic nerve are
in 2013 [29]. Although it is a multifactorial dis- severely impaired [24]. In the OPA1 haploinsuf-
ease, the first risk factor has been identified as ficiency animal model, it has been shown that the
increased intraocular pressure (IOP) above the RGCs and their axons are intact at the early
limit [22, 33]. Since retrograde degeneration may stages of the disease [25]. Our unpublished
induce apoptosis in RGCs, it also cumulatively results also showed a delay in axonal transport,
leads to glaucoma [21]. Notably, it has been especially in SC and lateral geniculate nucleus
accepted that aging can be an initiating risk factor (LGN) after anterograde transport of the
for glaucoma and this is also correlated with CTB. This also argued for axonal transport defi-
mitochondrial dysfunction [16]. In the last ciency as an early marker for DOA.
decade, an increasing number of studies have In a laser-induced intraocular pressure model,
argued that one of the early-stage characteriza- axonal transport of mitochondria in the optic nerve
tions of glaucoma includes functional deficits in was followed using multiphoton microscopy. In
axonal transport [6, 23]. this model, both the anterograde and the retro-
226 I. C. T. Okan et al.

grade transport of mitochondria were reduced. which have the potential to affect disease out-
This affected the overall mitochondrial distribu- comes. These findings suggest that axonal trans-
tion in RGC axons leading to several mitochon- port can be used as an early biomarker and
dria-free areas. Axonal transport of mitochondria predictor for developing optic neuropathies. This
was further reduced in aged animals showing fur- will allow us to make interventions to save neu-
ther similarities to the DBA2J model [26]. rons and vision, especially in the elderly popula-
Another mouse model, the Glucocorticoid tion considering the fact that aging is a main risk
(GC)-induced RGC degeneration model, was factor for glaucoma.
generated to mimic human POAG disease by To conclude, the treatment of axonal transport
inducing glaucomatous neurodegeneration via defects and improvement of transport efficiency
periocular injections of dexamethasone-21-­ by targeting several aspects of motor proteins
acetate (Dex). Dex is a glucocorticosteroid that is may also be a valuable therapeutic target for
usually used for edema reduction. Analysis of RGC diseases, especially in glaucoma and DOA.
axonal transport by CTB injection and axonal
loss by transmission electron microscopy (TEM)
showed that axonal transport persists at 5 weeks-­
References
time point where there was only 32% axonal loss.
However, when axonal loss reached 48% at 1. Alberts B, Johnson A, Lewis J, Raff M, Roberts K,
8 weeks, complete blockage of CTB in antero- Walter P. The cytoskeleton. In: Bochicchio A, editor.
grade transport occurred. Taken all together, Molecular biology of the cell. New York: Taylor and
these data also show that Dex induction initiates Francis Group; 2014. p. 939–47.
2. Hirokawa N, Niwa S, Tanaka Y. Molecular motors
axonal degeneration, and more importantly, this in neurons: transport mechanisms and roles in
blockage in the axonal transport precedes the brain function, development, and disease. Neuron.
degeneration and RGC loss [31]. 2010;68(4):610–38.
Further evidence supporting the glaucoma-­ 3. Reck-Peterson SL, Redwine WB, Vale RD, Carter
AP. The cytoplasmic dynein transport machin-
related axonal transport defect in animal models ery and its many cargoes. Nat Rev Mol Cell Biol.
involves accumulation of Tau, a microtubule-­ 2018;19(6):382–98.
binding protein. More interestingly, Tau accumu- 4. Yu D-Y, Cringle SJ, Balaratnasingam C, Morgan
lation in RGCs has been shown to disrupt WH, Paula KY, Su E-N. Retinal ganglion cells: ener-
getics, compartmentation, axonal transport, cyto-
anterograde axonal transport. Therefore, the skeletons and vulnerability. Prog Retin Eye Res.
accumulation of Tau protein in the rat glaucoma 2013;36:217–46.
model [27, 28] suggests that Tau accumulation 5. Morgan JE. Circulation and axonal transport in the
and thus, anterograde axonal transport defect is optic nerve. Eye. 2004;18(11):1089–95.
6. Dengler-Crish CM, Smith MA, Inman DM, Wilson
involved in this glaucoma model. Collectively, GN, Young JW, Crish SD. Anterograde transport
these findings demonstrate that axonal transport blockade precedes deficits in retrograde transport in
deficiency is predegenerative and precedes RGC the visual projection of the DBA/2J mouse model of
loss in several RGC disease models. glaucoma. Front Neurosci. 2014;8:290.
7. Fischer RA, Risner ML, Roux AL, Wareham LK,
Sappington RM. Impairment of membrane repolar-
ization accompanies axon transport deficits in glau-
5 Future Perspectives coma. Front Neurosci. 2019;13:1139.
8. Le Roux LG, Qiu X, Jacobsen MC, Pagel MD,
Gammon ST, Piwnica-Worms DR, et al. Axonal trans-
The axonal transport defects that are seen in both port as an in vivo biomarker for retinal neuropathy.
DBA2J and OPA1 haploinsufficiency models are Cell. 2020;9(5):1298.
an early phenomenon and thus independent of 9. Sleigh JN, Rossor AM, Fellows AD, Tosolini AP,
axonal degeneration. This was further supported Schiavo G. Axonal transport and neurological dis-
ease. Nat Rev Neurol. 2019;15(12):691–703.
by mitochondrial transport defects in the laser 10. Herro AM, Lam BL. Retrograde degeneration of reti-
IOP model. Overall, these defects may contribute nal ganglion cells in homonymous hemianopsia. Clin
to metabolic deficiencies in debilitated RGCs, Ophthalmol. 2015;9:1057.
Axonal Transport Defects in Retinal Ganglion Cell Diseases 227

11. Quigley HA, Addicks EM. Chronic experimen- 24. Carelli V, La Morgia C, Valentino ML, Barboni P,
tal glaucoma in primates. II. Effect of extended Ross-Cisneros FN, Sadun AA. Retinal ganglion cell
­intraocular pressure elevation on optic nerve head and neurodegeneration in mitochondrial inherited disor-
axonal transport. IOVS. 1980;19(2):137–52. ders. BBA-Bioenergetics. 2009;1787(5):518–28.
12. Quigley HA, Addicks EM, Green WR, Maumenee 25. Alavi MV, Bette S, Schimpf S, Schuettauf F,
A. Optic nerve damage in human glaucoma: II. The Schraermeyer U, Wehrl HF, et al. A splice site muta-
site of injury and susceptibility to damage. Arch tion in the murine Opa1 gene features pathology of
Ophthalmol. 1981;99(4):635–49. autosomal dominant optic atrophy. Brain. 2007;130
13. Schellingerhout D, Le Roux LG, Bredow S, Gelovani (Pt 4):1029–42.
JG. Fluorescence imaging of fast retrograde axo- 26. Takihara Y, Inatani M, Eto K, Inoue T, Kreymerman
nal transport in living animals. Mol Imaging. A, Miyake S, et al. In vivo imaging of axonal trans-
2009;8(6):319–29. port of mitochondria in the diseased and aged mam-
14. O’Neill EC, Danesh-Meyer HV, Connell PP, Trounce malian CNS. PNAS. 2015;112(33):10515–20.
IA, Coote MA, Mackey DA, et al. The optic nerve 27. Chiasseu M, Vargas JLC, Destroismaisons L,
head in acquired optic neuropathies. Nat Rev Neurol. Velde CV, Leclerc N, Di Polo A. Tau accumula-
2010;6(4):221–36. tion, altered phosphorylation, and missorting pro-
15. O’Neill EC, Mackey DA, Connell PP, Hewitt AW, mote neurodegeneration in glaucoma. J Neurosci.
Danesh-Meyer HV, Crowston JG. The optic nerve 2016;36(21):5785–98.
head in hereditary optic neuropathies. Nat Rev 28. Chiasseu M, Alarcon-Martinez L, Belforte N,
Neurol. 2009;5(5):277–87. Quintero H, Dotigny F, Destroismaisons L, et al. Tau
16. Ito YA, Di Polo A. Mitochondrial dynamics, transport, accumulation in the retina promotes early neuronal
and quality control: a bottleneck for retinal ganglion dysfunction and precedes brain pathology in a mouse
cell viability in optic neuropathies. Mitochondrion. model of Alzheimer’s disease. Mol Neurodegener.
2017;36:186–92. 2017;12(1):1–20.
17. Yu-Wai-Man P, Chinnery PF. Dominant optic atro- 29. Yih-Chung, Tham Xiang, Li Tien Y., Wong Harry A.,
phy: novel OPA1 mutations and revised preva­lence Quigley Tin, Aung Ching-Yu, Cheng (2014) Global
estimates. Ophthalmology. 2013;120(8):1712–e1. Prevalence of Glaucoma and Projections of Glaucoma
18. Pesch UE, Leo-Kottler B, Mayer S, Jurklies B, Burden through 2040 Ophthalmology 121(11) 2081–­
Kellner U, Apfelstedt-Sylla E, et al. OPA1 mutations 2090. https://doi.org/10.1016/j.ophtha.2014.05.013
in patients with autosomal dominant optic atrophy 30. Retrograde and Wallerian Axonal Degeneration Occur
and evidence for semi-dominant inheritance. Hum Synchronously after Retinal Ganglion Cell Axotomy
Mol Genet. 2001;10(13):1359–68. The American Journal of Pathology (2012);181(1):
19. Lenaers G, Neutzner A, Le Dantec Y, Juschke C, Xiao 62–73. https://doi.org/10.1016/j.ajpath.2012.03.030
T, Decembrini S, et al. Dominant optic atrophy: cul- 31. Prabhavathi, Maddineni Ramesh B., Kasetti Pinkal
prit mitochondria in the optic nerve. Prog Retin Eye D., Patel J. Cameron, Millar Charles, Kiehlbauch
Res. 2020;83:100935. Abbot F., Clark Gulab S., Zode. CNS axonal degen-
20. Hage R, Vignal-Clermont C. Leber hereditary optic eration and transport deficits at the optic nerve head
neuropathy: review of treatment and management. precede structural and functional loss of retinal gan-
Front Neurol. 2021;12:651639. glion cells in a mouse model of glaucoma Abstract
21. Evangelho K, Mogilevskaya M, Losada-Barragan Molecular Neurodegeneration 2020;15(1). https://doi.
M, Vargas-Sanchez JK. Pathophysiology of primary org/10.1186/s13024-020-00400-9
open-angle glaucoma from a neuroinflammatory and 32. Neuronal death in glaucoma Progress in Retinal
neurotoxicity perspective: a review of the literature. and Eye Research 1999;18(1):39–57. https://doi.
Int Ophthalmol. 2019;39(1):259–71. org/10.1016/S1350-9462(98)00014-7
22. Group CN-TGS. Comparison of glaucomatous pro- 33. The effectiveness of intraocular pressure reduction in
gression between untreated patients with normal-­ the treatment of normal-tension glaucoma American
tension glaucoma and patients with therapeutically Journal of Ophthalmology 1998;126(4): 498–505.
reduced intraocular pressures. Am J Ophthalmol. https://doi.org/10.1016/S0002-9394(98)00272-4
1998;126(4):487–97. 34. John, Simon & Smith, R & Savinova, Olga & Hawes,
23. Crish SD, Sappington RM, Inman DM, Horner PJ, N & Chang, Bennny & Turnbull, D & Davisson, M
Calkins DJ. Distal axonopathy with structural per- & Roderick, T & Heckenlively, J. Essential iris atro-
sistence in glaucomatous neurodegeneration. PNAS. phy, pigment dispersion, and glaucoma in DBA/2J
2010;107(11):5196–201. mice. Investigative ophthalmology & visual science.
1998;39:951–62.
Connexins Biology
in the Pathophysiology of Retinal
Diseases

Alejandro Ponce-Mora, Andrea Yuste,


Giuliana Perini-Villanueva, María Miranda,
and Eloy Bejarano

Abstract eration is not well understood, recent findings


present Cx as a potential therapeutic target.
Connexins (Cx) are a family of transmem- Therefore, we will briefly discuss pharmaco-
brane proteins that form gap junction intercel- logical approaches and gene therapies that are
lular channels that connect neighboring cells. being explored to modulate Cx function and
These channels allow the passage of ions and fight sight-threatening eye diseases.
other biomolecules smaller than 1 kDa,
thereby synchronizing the cells both electri- Keywords
cally and metabolically. Cxs are expressed in
Connexins · Retina · Gapjunctions ·
all retinal cell types and the diversity of Cx
Intercellular communication · Retinal
isoforms involved in the assembly of the chan-
degeneration
nels provides a functional syncytium required
for visual transduction. In this chapter, we
summarize the status of current knowledge
regarding Cx biology in retinal tissues and 1 Connexin Biology
discuss how Cx dysfunction is associated with in Different Retinal
retinal disease pathophysiology. Although the Components
contribution of Cx deficiency to retinal degen-
Connexins (Cx) are a large family of membrane
proteins that form clustered intercellular chan-
nels known as gap junctions (GJ). The human Cx
family comprises 21 genes and the expression of
A. Ponce-Mora · A. Yuste · M. Miranda the different members is cell-type dependent.
E. Bejarano (*) This allows heterogeneity in direct intercellular
School of Health Sciences and Veterinary School, interactions in our tissues, and gap junctional
Universidad CEU Cardenal Herrera, CEU intercellular communication (GJIC) is key for
Universities, Valencia, Spain
e-mail: eloy.bejaranofernandez@uchceu.es maintaining organismal homeostasis [1].
The retina is a light-sensitive tissue located in
G. Perini-Villanueva
Laboratory for Nutrition and Vision Research, USDA the innermost part of the eye. Due to its special-
Human Nutrition Research Center on Aging, Tufts ization, it is characterized as a high-level, struc-
University, Boston, MA, USA turally complex, neural tissue representing one of

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 229
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_33
230 A. Ponce-Mora et al.

the most intricate cellular circuits in our body. In rod-rod communication remains unclear, Cx36 is
the retina, neurons, glial cells, retinal pigment expressed by cones and is involved in homolo-
epithelial cells, and vascular cells form different gous cone-cone synapses and heterologous rod-­
layers whose functional synchronization is nec- cone synapses [8–10]. Bipolar cells, specialized
essary to preserve visual signal processing and glutamatergic neurons that transmit rod or cone-­
transduction [2–4]. generated signals to retinal ganglion cells,
All components of the vertebrate retina express Cx36 and Cx45 [8]. Homologous bipolar
express Cx, allowing for rapid, bidirectional cell coupling and bipolar to amacrine cell cou-
communication between retinal cell types, and pling also rely on Cx36 and Cx45 [11]. Retinal
contributing to multiple physiological roles [5, ganglion cells integrate vertebrate retina infor-
6]. The communication is enabled by GJ chan- mation and transmit the visual signal to higher
nels, which are formed by two different, six-­ visual areas. This cell type forms homologous
subunit hemichannels from adjacent cells. Each gap junctional channels using Cx30.2, Cx36, and
hemichannel can be constituted of the same Cx45 subunits, as well as heterologous connec-
(homomeric) or different (heteromeric) type of tions with amacrine cells that show dependence
Cx. The channel itself can be built by two identi- on Cx36 and Cx30.2 subunits [10]. Finally,
cal (homotypic) or non-identical (heterotypic) within the neuronal electrical circuit, horizontal
hemichannels, and cell-to-cell communication cells form homologous coupling based on Cx50
can be established between identical (homolo- and Cx57, Cx that provide high conductance lev-
gous) or different (heterologous) cell types. els [11]. Retinal glial cells are also connected
Given that the biophysical properties of intercel- through Cx-based channels, including the two
lular channels rely on the differential composi- retinal macroglia cell types. Several Cxs have
tion of Cx, the expression of these Cx isoforms been reported in Müller cells, although Cx43
represents a key factor for determining the selec- appears to be the major isoform for homologous
tive intercellular passage of biomolecules in reti- GJIC [10]. Astrocytes form functional syncytia
nal cell types. through homologous intercellular channels
Increasingly, new literature demonstrates that mostly comprised of Cx43 subunits [12]. Other
cell-to-cell communication plays a major role in studies reported the presence of Cx26, Cx30, and
neuronal electrical coupling in the retina, Cx45 in astrocytic gap junction channels [13,
because, unlike neurotransmitters, Cx-based 14]. Compelling literature also shows that the
channels allow for rapid neuronal transmission. establishment and correct functioning of cell-to-­
GJIC not only participates in the transduction of cell communication is key for vascular homeo-
the visual signal from photoreceptors to ganglion stasis. Cx43 is the most abundant subunit in
retinal cells but information can also be spread endothelial cells and pericytes, but Cx30.2, Cx37,
laterally, allowing temporal and spatial control of and Cx40 are also expressed [15]. Even retinal
the visual signal transduction. Cx-mediated elec- pigment epithelium cells require GJIC, and those
trical coupling virtually forms a fine-tuned, func- interactions rely on Cx43.
tional syncytium that is modulated by light In summary, Cx isoforms are expressed in a
adaptation and circadian rhythm [7]. GJIC plays cell-dependent manner in the retina, and
a major role in photoreceptors—retinal cells Cx-mediated cell-to-cell communication plays a
responsible for transforming light stimuli into crucial role in the neuronal network and in vision
electric signals. While Cx interaction involved in function.
Connexins Biology in the Pathophysiology of Retinal Diseases 231

2 Connexin Biology in Retinal in the light-damaged retina of an animal model of


Pathophysiology dry AMD. Additionally, Cx43 mimetic blocking
peptides decreased vascular leakage and
GJ channels are not static cellular structures, increased retinal ganglion cell survival after reti-
instead, they are renewed several times a day. Cx nal ischemia—protecting against retinal pigment
turnover requires a precise balance among differ- epithelial cell barrier breakdown [18, 28]. A non-­
ent molecular steps including transcription, intra- specific gap junction antagonist was also shown
cellular trafficking, membrane assembly, to increase retinal ganglion cell and amacrine cell
docking, and degradation. Although Cxs exhibit a surveillance in glaucoma models [23]. To avoid
short lifespan, the half-life is isoform-dependent off-target effects of pharmacological blockers,
and changes associated with Cx turnover are gene therapy has emerged as a potential promis-
dependent on physiological stimuli [16, 17]. ing alternative. Also, it could be relevant due to
Thus, the Cx turnover rate seems to be a vital limited ability of pharmacological blockers to
component in the regulation of Cx function. cross the BRB. Cx43 antisense oligodeoxynucle-
Altered expression of Cx is reported in both otide treatment demonstrated improved vascular
oxygen-­restricted and high glucose conditions, integrity and decreased astrocyte and microglia
contributing to loss of the BRB and vascular activation in an ischemic optic nerve organotypic
abnormalities [18–21]. Data also indicate that model [29]. In addition, lentivirus-mediated
dysfunctional GJIC plays a significant role in the delivery of siRNA against Cx43 after angioplasty
progression of retinal pathological conditions, injury in rats attenuated vascular restenosis and
presenting Cx as putative targets for the treat- intimal hyperplasia [30].
ment of multiple retinal diseases, including DR,
myopia, glaucoma, and AMD [4, 14, 22–25]. It is
controversial whether disturbed Cx turnover is an 3 Concluding Remarks
etiological agent of disease. However, it is
accepted that GJIC transmits intracellular cas- Cx dysfunction is an emerging molecular target
cades related to pathophysiological processes in the pathological processes of different retinal
involved in secondary cell death [26]. This phe- diseases with promising potential for neuromod-
nomenon, called bystander effect, claims that ulation and retinal regeneration. Despite the sur-
after the first cellular aggression, death signals facing of pharmacological and gene therapy
can be transmitted through the retinal cell net- interventions, a better understanding of Cx biol-
work, leading to secondary cell loss. Therefore, ogy in retinal pathophysiology is necessary to
degenerating retinal cells can spread damage to design therapeutic tools that avoid the unspecific
the surrounding healthy environment [27]. For and off-target effects of traditional approaches.
this reason, modulation of Cx pattern expression Differential Cx expression in the retinal compo-
under pathological conditions might offer a nents should be explored and taken into consider-
potential target to fight retinal diseases (Fig. 1). ation for the development of new tools to combat
Pharmacological blockade of Cx function is retinal and eye-related disorders.
being explored as a therapeutic alternative. For
example, oral administration of a Cx43 hemi- Acknowledgments This manuscript has been supported
channel blocker preserved photoreceptor activity by grants RYC2018-024434-I, MINECO PID2020-
119466RB-I00, FUSP-PPC-19-B53C4C64.
232 A. Ponce-Mora et al.

Fig. 1 A vast amount of


experimental evidence
supports the role of
connexin dysfunction in
several eye-related
disorders including
diabetic retinopathy
(DR), age-related
macular degeneration
(AMD), glaucoma, and
myopia. Different
therapeutic strategies
(highlighted in green)
are being explored as
potential interventional
approaches
(pharmacological
blockers of Cx function,
gene therapy, and
electrical/light
stimulation)

central visual processing and relies on Cx36-­


References containing gap junctions. Exp Eye Res. 2016;150:81–
9. https://doi.org/10.1016/j.exer.2015.05.013.
1. Söhl G, Willecke K. Gap junctions and the connexin 3. Roy K, Kumar S, Bloomfield SA. Gap junctional
protein family. Cardiovasc Res. 2004;62:228–32. coupling between retinal amacrine and ganglion cells
https://doi.org/10.1016/j.cardiores.2003.11.013. underlies coherent activity integral to global object
2. Ivanova E, Yee CW, Baldoni R Jr, Sagdullaev perception. Proc Natl Acad Sci. 2017;114:E10484–
BT. Aberrant activity in retinal degeneration impairs 93. https://doi.org/10.1073/pnas.1708261114.
Connexins Biology in the Pathophysiology of Retinal Diseases 233

4. Banerjee S, Wang Q, Zhao F, Tang G, So C, Tse D, To, Cell. 2012;23:2156–69. https://doi.org/10.1091/mbc.


C.-H, Feng Y, Zhou X, Pan F. Increased Connexin36 E11-­10-­0844.
phosphorylation in AII Amacrine cell coupling of the 17. Solan JL, Lampe PD. Specific Cx43 phosphorylation
mouse myopic retina. Front Cell Neurosci. 2020;14 events regulate gap junction turnover in vivo. FEBS
https://doi.org/10.3389/fncel.2020.00124. Lett. 2014;588:1423–9. https://doi.org/10.1016/j.
5. Güldenagel M, Söhl G, Plum A, Traub O, Teubner febslet.2014.01.049.
B, Weiler R, Willecke K. Expression patterns of 18. Danesh-Meyer HV, Kerr NM, Zhang J, Eady EK,
connexin genes in mouse retina. J Comp Neurol. O’Carroll SJ, Nicholson LF, Johnson CS, Green
2000;425:193–201. CR. Connexin43 mimetic peptide reduces vascular
6. Jin NG, Chuang AZ, Masson PJ, Ribelayga CP. Rod leak and retinal ganglion cell death following retinal
electrical coupling is controlled by a circadian ischaemia. Brain J Neurol. 2012;135:506–20. https://
clock and dopamine in mouse retina. J Physiol. doi.org/10.1093/brain/awr338.
2015;593:1597–631. https://doi.org/10.1113/ 19. Tien T, Muto T, Zhang J, Sohn EH, Mullins RF, Roy
jphysiol.2014.284919. S. Association of reduced Connexin 43 expression
7. Cao J, Ribelayga CP, Mangel SC. A circadian with retinal vascular lesions in human diabetic reti-
clock in the retina regulates rod-cone gap junc- nopathy. Exp Eye Res. 2016;146:103–6. https://doi.
tion coupling and neuronal light responses via acti- org/10.1016/j.exer.2015.12.011.
vation of adenosine A(2A) receptors. Front Cell 20. Ivanova E, Kovacs-Oller T, Sagdullaev BT. Vascular
Neurosci. 2020;14:605067. https://doi.org/10.3389/ pericyte impairment and connexin43 gap junction
fncel.2020.605067. deficit contribute to vasomotor decline in diabetic
8. Kántor O, Varga A, Nitschke R, Naumann A, retinopathy. J Neurosci. 2017;37:7580–94. https://doi.
Énzsöly A, Lukáts Á, Szabó A, Németh J, Völgyi org/10.1523/jneurosci.0187-­17.2017.
B. Bipolar cell gap junctions serve major signal- 21. Kovacs-Oller T, Ivanova E, Bianchimano P,
ing pathways in the human retina. Brain Struct Sagdullaev BT. The pericyte connectome: spatial pre-
Funct. 2017;222:2603–24. https://doi.org/10.1007/ cision of neurovascular coupling is driven by selective
s00429-­016-­1360-­4. connectivity maps of pericytes and endothelial cells
9. Asteriti S, Gargini C, Cangiano L. Connexin 36 and is disrupted in diabetes. Cell Discov. 2020;6:39.
expression is required for electrical coupling between https://doi.org/10.1038/s41421-­020-­0180-­0.
mouse rods and cones. Vis Neurosci. 2017;34:E006. 22. Szarka G, Balogh M, Tengölics ÁJ, Ganczer A,
https://doi.org/10.1017/s0952523817000037. Völgyi B, Kovács-Öller T. The role of gap junc-
10. González-Casanova J, Schmachtenberg O, Martínez tions in cell death and neuromodulation in the retina.
AD, Sanchez HA, Harcha PA, Rojas-Gomez D. An Neural Regen Res. 2021;16:1911–20. https://doi.
update on connexin gap junction and hemichannels org/10.4103/1673-­5374.308069.
in diabetic retinopathy. Int J Mol Sci. 2021;22:3194. 23. Akopian A, Kumar S, Ramakrishnan H, Roy K,
https://doi.org/10.3390/ijms22063194. Viswanathan S, Bloomfield SA. Targeting neuronal
11. Deans MR, Volgyi B, Goodenough DA, Bloomfield gap junctions in mouse retina offers neuroprotec-
SA, Paul DL. Connexin36 is essential for transmis- tion in glaucoma. J Clin Invest. 2017;127:2647–61.
sion of rod-mediated visual signals in the ­mammalian https://doi.org/10.1172/jci91948.
retina. Neuron. 2002;36:703–12. https://doi. 24. Yang GY, Liu FY, Li X, Zhu QR, Chen BJ, Liu
org/10.1016/s0896-­6273(02)01046-­2. LQ. Decreased expression of gap junction delta-2
12. Zahs KR, Newman EA. Asymmetric gap junctional (GJD2) messenger RNA and connexin 36 protein
coupling between glial cells in the rat retina. Glia. in form-deprivation myopia of guinea pigs. Chin
1997;20:10–22. Med J. 2019;132:1700–5. https://doi.org/10.1097/
13. Mansour H, McColm JR, Cole L, Weible M 2nd, cm9.0000000000000319.
Korlimbinis A, Chan-Ling T. Connexin 30 expres- 25. Slavi N, Toychiev AH, Kosmidis S, Ackert J,
sion and frequency of connexin heterogeneity in Bloomfield SA, Wulff H, Viswanathan S, Lampe
astrocyte gap junction plaques increase with age in PD, Srinivas M. Suppression of connexin 43 phos-
the rat retina. PLoS One. 2013;8:e57038. https://doi. phorylation promotes astrocyte survival and vascular
org/10.1371/journal.pone.0057038. regeneration in proliferative retinopathy. Proc Natl
14. Danesh-Meyer HV, Zhang J, Acosta ML, Rupenthal Acad Sci U S A. 2018;115:E5934–e5943. https://doi.
ID, Green CR. Connexin43 in retinal injury and dis- org/10.1073/pnas.1803907115.
ease. Prog Retin Eye Res. 2016;51:41–68. https://doi. 26. Ripps H. Cell death in retinitis pigmentosa: gap
org/10.1016/j.preteyeres.2015.09.004. junctions and the ‘bystander’ effect. Exp Eye
15. Roy S, Kim D, Lim R. Cell-cell communication in Res. 2002;74:327–36. https://doi.org/10.1006/
diabetic retinopathy. Vis Res. 2017;139:115–22. exer.2002.1155.
https://doi.org/10.1016/j.visres.2017.04.014. 27. Ma Y, Han X, de Castro RB, Zhang P, Zhang K, Hu
16. Bejarano E, Girao H, Yuste A, Patel B, Marques C, Z, Qin L. Analysis of the bystander effect in cone
Spray DC, Pereira P, Cuervo AM. Autophagy mod- photoreceptors via a guided neural network platform.
ulates dynamics of connexins at the plasma mem- Sci Adv. 2018;4:eaas9274. https://doi.org/10.1126/
brane in a ubiquitin-dependent manner. Mol Biol sciadv.aas9274.
234 A. Ponce-Mora et al.

28. Kuo C, Green CR, Rupenthal ID, Mugisho model of optic nerve ischaemia. J Clin Neurosci.
OO. Connexin43 hemichannel block protects against 2008;15:1253–63. https://doi.org/10.1016/j.
retinal pigment epithelial cell barrier breakdown. Acta jocn.2008.08.002.
Diabetol. 2020;57:13–22. https://doi.org/10.1007/ 30. Han XJ, Chen M, Hong T, Zhu LY, He D, Feng JG,
s00592-­019-­01352-­3. Jiang LP. Lentivirus-mediated RNAi knockdown of
29. Danesh-Meyer HV, Huang R, Nicholson LF, Green the gap junction protein, Cx43, attenuates the devel-
CR. Connexin43 antisense oligodeoxynucleotide opment of vascular restenosis following balloon
treatment down-regulates the inflammatory response injury. Int J Mol Med. 2015;35:885–92. https://doi.
in an in vitro interphase organotypic culture org/10.3892/ijmm.2015.2078.
Role of Nuclear NAD+ in Retinal
Homeostasis

Emily E. Brown, Michael J. Scandura,


and Eric Pierce

Abstract Keywords

NAD+ · Retinal homeostasis · NMNAT1 ·


The retina is one of the most metabolically
DNA damage · PARP · Genotoxicity · Retinal
active tissues and maintenance of metabolic
metabolism
homeostasis is critical for retinal function.
Nicotinamide adenine dinucleotide (NAD+) is
a cofactor that is required for key processes,
including the electron transport chain, glycol- 1 Introduction
ysis, fatty acid oxidation, and redox reactions.
NAD+ also acts as a co-substrate for enzymes In mammalian cells, NAD+ can be synthesized in
involved in maintaining genomic DNA integ- three different pathways. These include the kyn-
rity and cellular homeostasis, including poly-­ urenine pathway, in which NAD+ is synthesized
ADP ribose polymerases (PARPs) and de novo from tryptophan, the Preiss-Handler
Sirtuins. This review highlights the impor- pathway, in which NAD+ is synthesized from
tance of NAD+ in the retina, including the role NA, and the salvage pathway, which is most of
of enzymes involved in NAD+ production in the NAD+ is recycled from nicotinamide (NAM),
the retina and how NAD+-consuming enzymes the byproduct produced in reactions that con-
may play a role in disease pathology. We also sume NAD+. NAD+-consuming enzymes include
suggest a cell death pathway that may be com- poly (ADP-ribose) polymerases (PARPs) and
mon in multiple models of photoreceptor Sirtuins. NAM can then be converted back into
degeneration and highlight the role that NAD+ the NAD+ precursor, NMN, by nicotinamide
likely plays in this process. Finally, we explore phosphoribosyltransferase (NAMPT) in the sal-
future experimental approaches to enhance vage pathway. The conversion of NAM to NMN
our understanding of the role of NAD+ in the by NAMPT is the rate-limiting step of NAD+
retina. synthesis in this pathway. Nicotinamide mono-
nucleotide adenylyltransferases (NMNATs) then
convert NMN into NAD+.
E. E. Brown · M. J. Scandura · E. Pierce (*) Loss of NAD+-producing enzymes, activation
Ocular Genomics Institute, Massachusetts Eye and of NAD+-consuming enzymes, and reduced lev-
Ear, Boston, MA, USA els of NAD+ are all associated with retinal degen-
Department of Ophthalmology, Harvard Medical eration. These studies, which will be discussed in
School, Boston, MA, USA greater detail in this review, indicate that NAD+ is
e-mail: eric_pierce@meei.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 235
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_34
236 E. E. Brown et al.

essential for proper retinal function, and perhaps temic effects. There are at least 34 mutations in
that the photoreceptors specifically have a unique NMNAT1 that lead to retinal degeneration, and
requirement for NAD+. These findings also sug- many of these mutations result in decreased
gest that NAD+ reduction may be a common enzymatic function of NMNAT1[7]. These find-
pathogenic mechanism in several different mod- ings further support the hypothesis that NAD+ is
els of retinal degeneration. This review will dis- essential for retinal function and that its depletion
cuss these studies in greater detail and will uniquely affects the retina. Knockout of Nmnat1
conclude with a discussion of the potential next in mice is embryonic lethal, suggesting that at
directions to develop a better understanding of least some enzymatic function is required for
the role NAD+ plays in retinal homeostasis and proper development [5]. Mice harboring the p.
disease. V9M mutation in Nmnat1 recapitulate many fea-
tures of NMNAT1-associated disease that occur
in humans, including early onset retinal degen-
2 Enzymes Involved in NAD+ eration, optic atrophy, and RPE cell loss [12].
Synthesis Are Essential Reduced enzymatic activity of NMNAT1 in this
in the Retina model is also associated with reduced levels of
NAD+ in the retina, but not other tissues [11].
The enzymes in the salvage pathway are essential Other studies have shown that conditional knock-
for retinal function, suggesting the importance of out of Nmnat1 in the mouse retina results in rapid
NAD+ in maintaining retinal homeostasis. retinal degeneration [24]. Interestingly, cell type-­
Ablation of NAMPT activity specifically in the specific knockout of Nmnat1 either in the rods or
rod or cone photoreceptors leads to retinal degen- cones is sufficient to cause retinal degeneration
eration in mice [16], suggesting that NAD+ is [24], suggesting a photoreceptor-specific role for
critical for both rod and cone photoreceptor sur- the enzyme or a photoreceptor-specific require-
vival. Small molecule inhibition of NAMPT has ment for nuclear NAD+.
been shown to lead to vision, but not hearing loss,
in a zebrafish model [3], suggesting that the ret-
ina is uniquely susceptible to reduced levels of 3 Role of NAD+ Consumers
NAD+. In the retinal pigment epithelium (RPE) and NAD+ Depletion
of mice, expression of NAMPT and the levels of in Retinal Degeneration
NAD+ were both shown to decline with age,
while pharmacological inhibition of NAMPT in NAD+-consuming enzymes have also been shown
RPE cells resulted in reduced Sirtuin expression to play an important role in disease pathogenesis
and activity and increased levels of inflammation in a variety of models of retinal degeneration. In
[13]. These findings suggest that NAD+ is critical the nucleus, the primary NAD+-consuming
for the maintenance of cellular homeostasis in enzymes are PARPs and Sirtuins. There are 18
the RPE and neural retina. different PARP enzymes, which are encoded by
Several studies have also shown that NMNAT1 different genes. Most of these have poly-ADP
is required for normal retinal function. NMNATs ribosyltransferase activity. Parthanatos, a PARP-­
are ubiquitously expressed, and there are dependent mode of cell death [6], has been pro-
three NMNAT1 isoforms, which are encoded by posed as a mechanism of photoreceptor
different genes. The NMNAT isoforms are distin- degeneration [23]. PARP overactivation has been
guished based on their subcellular localization. shown to lead to photoreceptor cell death in the
NMNAT1 is found in the nucleus, NMNAT2 is rd1 mouse model of inherited retinal degenera-
found in the cytoplasm, and NMNAT3 is found in tion, in the RHO P23H mouse model of retinitis
the mitochondria. Mutations in NMNAT1, the pigmentosa (RP), and in the RHO S334ter rat
nuclear isoform, lead to inherited retinal degen- model of RP [14, 19]. Our group has also sug-
eration [1, 4, 7, 15, 20], with few reports of sys- gested that PARP overactivation and PAR-­
Role of Nuclear NAD+ in Retinal Homeostasis 237

mediated cell death play a role in ity. However, the mechanisms underlying this
NMNAT1-associated disease. Nmnat1V9M/V9M mice phenomenon are still poorly understood. This
exhibit photoreceptor-specific increases in PAR hypothesis also fails to explain why the retina is
expression, which is associated with a retina-­ uniquely affected in NMNAT1-associated dis-
specific decline in the levels of NAD+ [11]. In ease, as NMNAT1-mediated SARM1 inhibition
addition to PARP overactivation contributing to should be affected in other neurons. Although the
disease pathogenesis, studies have also shown precise mechanisms underlying these processes
that inhibition of PARP can prevent photorecep- remain to be fully understood, depletion of NAD+
tor degeneration. Knockout of PARP1 does not appears to be a common feature in many of these
significantly affect retinal structure or function in models with retinal degeneration, suggesting its
wild-type mice [22]. However, knockout of importance in disease pathogenesis.
PARP1 in the Rd1 mouse model significantly Interestingly, depletion of NAD+ occurs early
reduced the rate of retinal degeneration [22] and in the retinal dysfunction process in several
the PARP inhibitor, Olaparib, has been shown to mouse models of retinal degeneration, including
delay cell death in this model [21]. light-induced retinal degeneration, aging-­
Other studies suggest that sterile alpha and associated retinal dysfunction, and in diabetic
TIR motif containing 1 (SARM1), a NADase that retinopathy induced by streptozotocin [16]. Our
depletes NAD+ levels in axons in response to group has also shown that in the Nmnat1V9M/V9M
injury [10], plays a role in retinal degeneration. mouse model of NMNAT1-associated retinal
Activation of SARM1 NADase activity results in degeneration, NAD+ depletion occurs prior to
the loss of NAD+ pools and energy depletion, retinal degeneration [11]. These findings indicate
which ultimately results in cell death [10, 27]. In that reduced levels of NAD+ are a common fea-
the axons of retinal ganglion cells, activation of ture in early disease, suggesting that NAD+ deple-
SARM1 has been shown to promote degenera- tion may be driving retinal pathology in a wide
tion, but not cell death in models of optic crush-­ range of retinal degenerative diseases.
induced injury and toxicity induced by kainic
acid [8, 17]. NMNAT1 has been shown to play an
essential role in blocking SARM1-dependent 4 Conclusions
NAD+ consumption in the axons of injured mouse
dorsal root ganglion cells [26] and overexpres- The studies discussed in this review indicate the
sion of NMNAT1 or expression of a mutant importance of NAD+ for retinal homeostasis.
NMNAT1 fusion protein are protective against Future studies should aim to develop a better
axonal injury in other models [2, 9, 25]. These understanding of the role of NAD+ depletion in
findings suggest that pathology due to loss of the pathogenesis of inherited retinal degenera-
NMNAT1 activity is the result of indirect NAD+ tions, the role of nuclear vs cytosolic or mito-
depletion from lack of inhibition of SARM1, chondrial NAD+ pools in this process, and the
rather than a reduction NAD+ as a direct cause of role NAD+ consuming enzymes play in disease
reduced NMNAT1 enzymatic activity. etiology.
Consistent with this hypothesis, recent work
suggests that the essential role of NMNAT1 in
the retina is to inhibit SARM1. Knockout of References
Sarm1 in addition to Nmnat1 was shown to pre-
vent retinal degeneration [24]. Using the Rho 1. Abad-Morales V, Wert A, Ruiz Gomez MA, Navarro
R, Pomares E. New insights on the genetic basis
knockout model of inherited retinal degeneration,
underlying SHILCA syndrome: characterization
another study showed that knocking out Sarm1 of the NMNAT1 pathological alterations due to
resulted in delayed photoreceptor cell death [18]. compound heterozygous mutations and identifica-
These findings suggest that the key function of tion of a novel alternative isoform. Int J Mol Sci.
2021;22(5):2262.
NMNAT1 in the retina is to inhibit SARM1 activ-
238 E. E. Brown et al.

2. Araki T, Sasaki Y, Milbrandt J. Increased nuclear 13. Jadeja RN, Powell FL, Jones MA, Fuller J, Joseph
NAD biosynthesis and SIRT1 activation prevent axo- E, Thounaojam MC, Bartoli M, Martin PM. Loss of
nal degeneration. Science. 2004;305(5686):1010–3. NAMPT in aging retinal pigment epithelium reduces
3. Cassar S, Dunn C, Olson A, Buck W, Fossey S, NAD(+) availability and promotes cellular senescence.
Ramos MF, Sancheti P, Stolarik D, Britton H, Cole Aging (Albany NY). 2018;10(6):1306–23.
T, Bratcher N, Huang X, Peterson R, Longenecker K, 14. Kaur J, Mencl S, Sahaboglu A, Farinelli P, van Veen
LeRoy B. From the cover: inhibitors of nicotinamide T, Zrenner E, Ekstrom P, Paquet-Durand F, Arango-­
phosphoribosyltransferase cause retinal damage in Gonzalez B. Calpain and PARP activation during pho-
larval zebrafish. Toxicol Sci. 2018;161(2):300–9. toreceptor cell death in P23H and S334ter rhodopsin
4. Chiang PW, Wang J, Chen Y, Fu Q, Zhong J, Chen mutant rats. PLoS One. 2011;6(7):e22181.
Y, Yi X, Wu R, Gan H, Shi Y, Chen Y, Barnett C, 15. Koenekoop RK, Wang H, Majewski J, Wang X, Lopez
Wheaton D, Day M, Sutherland J, Heon E, Weleber I, Ren H, Chen Y, Li Y, Fishman GA, Genead M,
RG, Gabriel LA, Cong P, Chuang K, Ye S, Sallum JM, Schwartzentruber J, Solanki N, Traboulsi EI, Cheng
Qi M. Exome sequencing identifies NMNAT1 muta- J, Logan CV, McKibbin M, Hayward BE, Parry DA,
tions as a cause of Leber congenital amaurosis. Nat Johnson CA, Nageeb M, C. Finding of Rare Disease
Genet. 2012;44(9):972–4. Genes Canada, Poulter JA, Mohamed MD, Jafri H,
5. Conforti L, Janeckova L, Wagner D, Mazzola F, Rashid Y, Taylor GR, Keser V, Mardon G, Xu H,
Cialabrini L, Di Stefano M, Orsomando G, Magni G, Inglehearn CF, Fu Q, Toomes C, Chen R. Mutations
Bendotti C, Smyth N, Coleman M. Reducing expres- in NMNAT1 cause Leber congenital amaurosis and
sion of NAD+ synthesizing enzyme NMNAT1 does identify a new disease pathway for retinal degenera-
not affect the rate of Wallerian degeneration. FEBS J. tion. Nat Genet. 2012;44(9):1035–9.
2011;278(15):2666–79. 16. Lin JB, Kubota S, Ban N, Yoshida M, Santeford A,
6. David KK, Andrabi SA, Dawson TM, Dawson Sene A, Nakamura R, Zapata N, Kubota M, Tsubota
VL. Parthanatos, a messenger of death. Front Biosci K, Yoshino J, Imai SI, Apte RS. NAMPT-mediated
(Landmark Ed). 2009;14:1116–28. NAD(+) biosynthesis is essential for vision in mice.
7. Falk MJ, Zhang Q, Nakamaru-Ogiso E, Kannabiran Cell Rep. 2016;17(1):69–85.
C, Fonseca-Kelly Z, Chakarova C, Audo I, Mackay 17. Massoll C, Mando W, Chintala SK. Excitotoxicity
DS, Zeitz C, Borman AD, Staniszewska M, Shukla upregulates SARM1 protein expression and pro-
R, Palavalli L, Mohand-Said S, Waseem NH, Jalali S, motes Wallerian-like degeneration of retinal ganglion
Perin JC, Place E, Ostrovsky J, Xiao R, Bhattacharya cells and their axons. Invest Ophthalmol Vis Sci.
SS, Consugar M, Webster AR, Sahel JA, Moore AT, 2013;54(4):2771–80.
Berson EL, Liu Q, Gai X, Pierce EA. NMNAT1 muta- 18. Ozaki E, Gibbons L, Neto NG, Kenna P, Carty
tions cause Leber congenital amaurosis. Nat Genet. M, Humphries M, Humphries P, Campbell M,
2012;44(9):1040–5. Monaghan M, Bowie A, Doyle SL. SARM1 defi-
8. Fernandes KA, Mitchell KL, Patel A, Marola OJ, ciency promotes rod and cone photoreceptor cell
Shrager P, Zack DJ, Libby RT, Welsbie DS. Role of survival in a model of retinal degeneration. Life Sci
SARM1 and DR6 in retinal ganglion cell axonal and Alliance. 2020;3:5.
somal degeneration following axonal injury. Exp Eye 19. Paquet-Durand F, Silva J, Talukdar T, Johnson LE,
Res. 2018;171:54–61. Azadi S, van Veen T, Ueffing M, Hauck SM, Ekstrom
9. Fernando FS, Conforti L, Tosi S, Smith AD, Coleman PA. Excessive activation of poly(ADP-ribose)
MP. Human homologue of a gene mutated in the polymerase contributes to inherited photoreceptor
slow Wallerian degeneration (C57BL/Wld(s)) mouse. degeneration in the retinal degeneration 1 mouse. J
Gene. 2002;284(1–2):23–9. Neurosci. 2007;27(38):10311–9.
10. Gerdts J, Brace EJ, Sasaki Y, DiAntonio A, 20. Perrault I, Hanein S, Zanlonghi X, Serre V, Nicouleau
Milbrandt J. SARM1 activation triggers axon degen- M, Defoort-Delhemmes S, Delphin N, Fares-Taie
eration locally via NAD(+) destruction. Science. L, Gerber S, Xerri O, Edelson C, Goldenberg A,
2015;348(6233):453–7. Duncombe A, Le Meur G, Hamel C, Silva E, Nitschke
11. Greenwald SH, Brown EE, Scandura MJ, Hennessey P, Calvas P, Munnich A, Roche O, Dollfus H, Kaplan
E, Farmer R, Du J, Wang Y, Pierce EA. Mutant J, Rozet JM. Mutations in NMNAT1 cause Leber con-
Nmnat1 leads to a retina-specific decrease of NAD+ genital amaurosis with early-onset severe macular and
accompanied by increased poly(ADP-ribose) in a optic atrophy. Nat Genet. 2012;44(9):975–7.
mouse model of NMNAT1-associated retinal degen- 21. Sahaboglu A, Barth M, Secer E, Amo EM, Urtti
eration. Hum Mol Genet. 2021;30(8):644–57. A, Arsenijevic Y, Zrenner E, Paquet-Durand
12. Greenwald SH, Charette JR, Staniszewska M, Shi LY, F. Olaparib significantly delays photoreceptor loss in
Brown SDM, Stone L, Liu Q, Hicks WL, Collin GB, a model for hereditary retinal degeneration. Sci Rep.
Bowl MR, Krebs MP, Nishina PM, Pierce EA. Mouse 2016;6:39537.
models of NMNAT1-Leber Congenital Amaurosis 22. Sahaboglu A, Tanimoto N, Kaur J, Sancho-Pelluz
(LCA9) recapitulate key features of the human dis- J, Huber G, Fahl E, Arango-Gonzalez B, Zrenner
ease. Am J Pathol. 2016;186(7):1925–38. E, Ekstrom P, Lowenheim H, Seeliger M, Paquet-­
Role of Nuclear NAD+ in Retinal Homeostasis 239

Durand F. PARP1 gene knock-out increases resistance 25. Sasaki Y, Milbrandt J. Axonal degeneration is blocked
to retinal degeneration without affecting retinal func- by nicotinamide mononucleotide adenylyltransferase
tion. PLoS One. 2010;5(11):e15495. (Nmnat) protein transduction into transected axons. J
23. Sancho-Pelluz J, Arango-Gonzalez B, Kustermann S, Biol Chem. 2010;285(53):41211–5.
Romero FJ, van Veen T, Zrenner E, Ekstrom P, Paquet-­ 26. Sasaki Y, Nakagawa T, Mao X, DiAntonio A,
Durand F. Photoreceptor cell death mechanisms Milbrandt J. NMNAT1 inhibits axon degeneration
in inherited retinal degeneration. Mol Neurobiol. via blockade of SARM1-mediated NAD(+) depletion.
2008;38(3):253–69. elife. 2016;5:20161001.
24. Sasaki Y, Kakita H, Kubota S, Sene A, Lee TJ, Ban 27. Summers DW, Gibson DA, DiAntonio A, Milbrandt
N, Dong Z, Lin JB, Boye SL, DiAntonio A, Boye J. SARM1-specific motifs in the TIR domain
SE, Apte RS, Milbrandt J. SARM1 depletion rescues enable NAD+ loss and regulate injury-induced
NMNAT1-dependent photoreceptor cell death and SARM1 activation. Proc Natl Acad Sci U S A.
retinal degeneration. elife. 2020;9:20201001. 2016;113(41):E6271–80.
Retinal Pigmented
Epithelium-­Derived Ectopic Norrin
Does Not Promote Intraretinal
Angiogenesis in Transgenic Mice

Andrea E. Dillinger and Ernst R. Tamm

Abstract 1 Introduction

Formation of intraretinal capillaries and inner Development of the intraretinal vasculature starts
blood-retinal barrier during development with the radial outgrowth of vessels from the
requires norrin, a ligand of the canonical optic nerve, followed by the sprouting of endo-
wingless/integrated (Wnt)/β-catenin signaling thelial cells into the retina to form the deep and
pathway. Here we addressed the question intermediate vascular plexuses with multiple
whether retinal pigmented epithelium (RPE)- interconnected vessels [3]. The endothelial cells
derived overexpression of norrin in transgenic form a continuous, non-fenestrated vascular net-
mice rescues the vascular phenotype caused work, with distinct molecular characteristics to
by norrin deficiency. To this end, we generated form the inner blood-retinal barrier (BRB). The
NdpKO/Rpe65-Norrin mice and analyzed the processes prevent the flux of unneeded molecules
activation of β-catenin signaling, the develop- and cells, while providing enough oxygen and
ment of intraretinal capillaries, and the expres- nutrients for the neuronal environment. High and
sion of blood-retinal barrier marker molecules. specialized expression of tight junction proteins,
RPE-derived norrin induced retinal β-catenin like claudins, causes reduced paracellular trans-
signaling but failed to rescue the vascular port, and low rates of transcytosis lead to limited
developmental defects and the breakdown of transcellular transport [4]. Intraretinal angiogen-
the blood-retinal barrier in norrin-deficient esis and formation of the inner BRB critically
mice. Sites of ectopic norrin expression and require the signaling molecule norrin which is
the amounts of secreted transgenic protein are secreted by Müller cells [6, 10, 12, 13]. Norrin
critical factors to enable the angiogenic prop- belongs to the cysteine-knot growth factor super-
erties of norrin. family and is a ligand of the canonical wingless/
integrated (Wnt) signaling pathway. Norrin binds
Keywords as a dimer to the N-terminal extracellular
cysteine-­rich domain of frizzled class receptor 4
Norrin · Retinal vascular development · (FZD4) and to the low-density lipoprotein
Blood-retinal barrier · β-catenin receptor-­related protein 5/6 (LRP5/6) co-receptor
and activates canonical β-catenin signaling.
A. E. Dillinger (*) · E. R. Tamm Norrin-induced β-catenin signaling is enhanced
Institute of Human Anatomy and Embryology, by the stabilization of FZD4 at the cell membrane
University of Regensburg, Regensburg, Germany through the co-activator tetraspanin 12. Upon
e-mail: andrea.dillinger@ur.de

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 241
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_35
242 A. E. Dillinger and E. R. Tamm

Norrin-FZD4 binding, the APC-destruction com- 2.2 Immunohistochemistry


plex which leads to β-catenin phosphorylation and Retinal Flat Mount
and degradation is inactivated and therefore Preparation
β-catenin is stabilized and promotes gene tran-
scription [5, 11, 13]. Mutations in the Ndp gene For blood vessel presentation, mice were anes-
which encodes norrin lead to the rare X-linked thetized with CO2, euthanized by atlanto-­
genetic disorder Norrie disease (OMIM occipital dislocation, and perfused through
#310600). The clinical features in patients suffer- the left ventricle with 1 mL of PBS that con-
ing from Norrie disease, like retinal dysplasia/ tained 50 mg high molecular weight
pseudoglioma, retinal detachment, vitreous hem- (MW = 2,000,000 g/mol) FITC-dextran (TdB
orrhages, and cataract inevitably cause blindness. Consultancy, Uppsala, Sweden). For retinal flat
Mutant Ndp-deficient mice exhibit a lack of intra- mounts, eyes were enucleated and placed in
retinal capillaries, glomeruloid-like vascular 4% paraformaldehyde for 1 h. Retinae were
malformations near the inner retinal surface, and dissected and flat mounted with Mowiol
high vascular leakage due to an impaired BRB [6, mounting medium. For immunohistochemis-
10]. Previous studies showed that a retinal pig- try, eyes with and without FITC-dextran perfu-
mented epithelium (RPE) derived overexpression sion were enucleated and fixed in 10% glacial
of norrin protects photoreceptors after light-­ acetic acid, 60% methanol, and 30% chloro-
induced cell death [2] and accelerates vascular form for 4 h at RT, transferred to 50% and 25%
repair following oxygen-induced damage [9]. In methanol 10 min each, washed in 0.1 M phos-
this study, we investigated whether ectopic RPE-­ phate buffer, equilibrated in 10%, 20%, and
derived norrin secretion to the inner retina pro- 30% sucrose, embedded in Tissue-Tek optimal
motes intraretinal angiogenesis, the formation of cooling temperature compound (Sakura
an intact inner BRB, and overall rescues the reti- Finetek Europe B. V., Zoeterwounde,
nal phenotype of norrin-deficient mutant mice. Netherlands) and stored at −20 °C. Frozen sec-
tions were cut on a cryostat, incubated with 2%
bovine serum albumin, 0.2% cold water fish
2 Material and Methods gelatin, 0.1% Triton X-100 in 0.1 M phosphate
buffer, and stained with specific antibodies as
2.1 Animals follows: rat anti-­PLVAP (1:50; Santa Cruz,
Santa Cruz, USA), rabbit anti-caveolin1
Ndptm2Nat (The Jackson Laboratory Stock No. (1:100; Cell Signaling Technology, Danvers,
012287 [12]) and Rpe65-Norrin [9] mice were USA), rabbit anti-claudin5 (1:100; Invitrogen,
crossed. Resulting Ndpy/−/Rpe65-Norrin and Carlsbad, USA), rabbit anti-­β-­catenin (1:100,
Ndp−/−/Rpe65-Norrin were used as experimental Cell signaling), rabbit anti-­ RPE65 (1:100,
mice, and Ndpy/+/Rpe65-Norrin, Ndpy/−, Ndp−/− Abcam, Cambridge, UK), donkey anti-rabbit
and wild-type mice were referred to as controls. IgG (H+L) Alexa Fluor 647 (1:200, Thermo
Mice were housed under standardized conditions Fisher Scientific, Waltham, USA), Cy™3 goat
of 62% air humidity and 21 °C room temperature anti-rabbit IgG (H+L) (1:2000, Jackson
(RT). Feeding was ad libitum. Animals were kept Immuno Research, Cambridgeshire, UK) and
at a 12 h light/dark cycle (6:00–18:00). All proce- Cy™3 donkey anti-rat IgG (H+L) (1:2000;
dures conformed to the tenets of the National Jackson Immuno Research). To control for
Institutes of Health Guidelines on the Care and unspecific binding of the secondary antibody,
Use of Animals in Research, the EU Directive negative controls were performed in which pri-
2010/63/E and the ARVO Statement for the Use mary antibodies had been omitted. Finally,
of Animals in Ophthalmic and Vision Research, 4,6-diamidino-2-phenylindole (DAPI, Vector
and were approved by local authorities (54-­ Laboratories) was added to counterstain
2532.1-44/12; Regierung Oberpfalz, Germany). nuclear DNA.
Retinal Pigmented Epithelium-Derived Ectopic Norrin Does Not Promote Intraretinal Angiogenesis… 243

2.3 Fluorescence Microscopy suited to detect norrin by immunohistochemis-


try, we used an indirect approach and analyzed
Specimens were analyzed using a Zeiss Axio first the neonatal expression of RPE65 in wild-
Imager microscope (Carl Zeiss AG, Jena, type mice (Fig. 1a). Intense and specific immu-
Germany). noreactivity for RPE65 was detected throughout
the entire RPE at postnatal day (P)14 and
6 weeks of age. In contrast, at P7 no or only
3 Results very weak staining for RPE65 was detectable.
Next, we wanted to know if the RPE65 pro-
For simplicity, we will refer to Ndpy/−and Ndp−/− moter fragment is driving the expression of
mice as NdpKO control mice; however, Ndpy/−/ norrin. To this end, we analyzed the expression
Rpe65-Norrin and Ndp−/−/Rpe65-Norrin mice of β-catenin, a downstream molecule of norrin
are referred to as experimental NdpKO/Rpe65-­ signaling and key mediator of canonical Wnt-
Norrin mice. In addition, data from wild-type and signaling. We performed immunohistochemis-
Ndpy/+/Rpe65-Norrin mice will be presented. try in the retina of 6-week-old control and
experimental mice (Fig. 1b). Wild-type and
Ndpy/+/Rpe65-Norrin mice showed an intense
3.1 Canonical Wnt/β-Catenin signal for β-catenin which preferentially accu-
Signaling Is Activated mulated in the nuclei of neurons in the inner
in NdpKO/Rpe65-Norrin Mice nuclear and ganglion cell layer. In contrast, in
Norrin-­deficient control mice, the β-catenin
We first wanted to learn if the RPE65 minimal signal was dramatically reduced. Remarkably,
promoter [1] that was used to drive norrin in NdpKO/Rpe65-Norrin mice the β-catenin sig-
expression in our mouse model would be active nal intensity was enhanced when compared to
during the norrin-mediated formation of retinal that of NdpKO mice. The finding clearly indi-
capillaries. Because of the lack of an antibody cated that RPE-­ derived transgenic norrin is

Fig. 1 (a) Immunohistochemistry of RPE65 (magenta) in NdpKO/Rpe65-Norrin mice. Nuclei were counterstained
the wildtype retina at P7, P14, and at 6 weeks of age. (b) with Dapi. GCL ganglion cell layer, INL inner plexiform
Immunohistochemistry of β-catenin (red) in the retina of layer, ONL outer plexiform layer, RPE retinal pigmented
6-week-old wildtype, Ndpy/+/Rpe65-Norrin, NdpKO, and epithelium; n = 3
244 A. E. Dillinger and E. R. Tamm

secreted to the sensory retina and activates cue the retinal phenotype of norrin-deficient
there the Wnt/β-catenin pathway. mice. The mice showed a lack of intraretinal cap-
illaries, with glomeruloid-like malformations
extending into the deeper layers of the retina
3.2 RPE-Derived Norrin Does Not (Fig. 2a). The findings were confirmed by FITC-­
Promote Retinal Angiogenesis dextran-­perfused retinal cross sections (Fig. 2b).

Intraretinal capillary development was analyzed


on cross sections and flat mounts of FITC-­ 3.3 Analysis of Blood-Retinal
Dextran-­ perfused retina of 6-week-old mice. Barrier Integrity
Images of retinal flat mounts are presented as
merged picture: the superficial plexus is depicted Next, we analyzed by immunohistochemistry
in magenta, the intermediate plexus in gray and the tight-junction protein claudin5 (CLN5), the
the deep plexus in cyan (Fig. 2a). Wildtype and marker of fenestrated capillaries PLVAP, and the
Ndpy/+/Rpe65-Norrin mice showed normal devel- caveolae protein caveolin1 (CAV1) (Figs. 3, 4
opment of the retinal vascular network. The three and 5). In wildtype and Ndpy/+/Rpe65-Norrin
retinal plexuses were fully developed and the mice staining for PLVAP was detected in the
interconnected capillaries of all three plexuses choriocapillaris (arrowhead) but was completely
were visible (Fig. 2a). In contrast, in NdpKO mice missing in the intraretinal capillaries (Fig. 3). In
retinal vessels were restricted to the superficial contrast, in NdpKO mice, PLVAP was localized
plexus. The retinae showed lack of capillaries in both to the choriocapillaris (arrowhead) and the
the deep and intermediate plexuses and intraretinal vessels (arrow), indicating the for-
glomeruloid-­like or club-like malformations of mation of fenestrated capillaries and the pres-
the superficial vasculature (Fig. 2a, arrow), both ence of an impaired BRB (Fig. 3). Finally, the
characteristic findings in norrin-deficient animals analysis of NdpKO/Rpe65-Norrin mice showed a
[6, 8, 10]. The analysis of NdpKO/Rpe65-Norrin similar staining pattern for PLVAP as the signal
mice showed that RPE-derived norrin did not res- was detected in the choriocapillaris (arrowhead)

Fig. 2 FITC-dextran perfused retinae of 6-week-old (magenta), IP intermediate plexus (gray), DP deep plexus
wild-type, Ndpy/+/Rpe65-Norrin, NdpKO, and NdpKO/ (cyan). Arrow: glomerular-like malformations (b) Retinal
Rpe65-Norrin mice. (a) The three vascular plexuses of the cross sections. GCL ganglion cell layer, INL inner nuclear
retina are overlaid in one image. SP superficial plexus layer, ONL outer nuclear layer. n ≥ 3
Retinal Pigmented Epithelium-Derived Ectopic Norrin Does Not Promote Intraretinal Angiogenesis… 245

Fig. 3 Immunohistochemistry of PLVAP (red) in the ret- retinal capillaries. GCL ganglion cell layer, INL inner
ina of 6-week-old wildtype, Ndpy/+/Rpe65-Norrin, NdpKO, nuclear layer, ONL outer nuclear layer, CC choriocapil-
and NdpKO/Rpe65-Norrin mice. Arrowhead: PLVAP sig- laris; nuclei were counterstained with Dapi. n = 3
nal in the choriocapillaris. Arrow: PLVAP signal in intra-

and intraretinal capillaries (arrow). CAV1 is a highly enriched (Fig. 4, arrow). Interestingly, in
marker for vesicle trafficking, and thereby its NdpKO/Rpe65-Norrin mice the intensity of the
expression in vascular endothelial cells indi- CAV1 signal was slightly reduced when com-
cates transcytosis. In wildtype and Ndpy/+/ pared to NdpKO mice (Fig. 4, arrowhead).
Rpe65-Norrin mice, CAV1 showed a moderate Finally, we analyzed the tight-junction protein
immunohistochemical signal in the retina that CLN5 (Fig. 5). In the two control groups, wild-
was mostly located in endothelial cells. In con- type and Ndpy/+/Rpe65-Norrin mice CLN5 was
trast, in NdpKO mice the signal for CAV1 in located predominantly in endothelial cells
endothelial cells of the superficial plexus was (Fig. 5, arrow). CLN5 intensity was dramati-
246 A. E. Dillinger and E. R. Tamm

Fig. 4 Immunohistochemistry of caveolin1 (magenta) in the NdpKO mice. Arrowhead: moderate CAV1 signal in
the retina of 6-week-old wildtype, Ndpy/+/Rpe65-Norrin, NdpKO/Rpe65-Norrin mice. GCL ganglion cell layer, INL
NdpKO, and NdpKO/Rpe65-Norrin mice. Arrow: intense inner nuclear layer, ONL outer nuclear layer, CC chorio-
signal of CAV1 in capillaries of the superficial plexus of capillaris; nuclei were counterstained with Dapi. n = 3

cally reduced in NdpKO mice, which additionally 4 Discussion


showed the features of an impaired BRB.
Remarkably, in the NdpKO/Rpe65-Norrin ani- The results of this study show that ectopic, RPE-­
mals a slight fluorescence signal of CLN5 in derived transgenic norrin is not sufficient to pro-
endothelial cells of the superficial plexus was mote the formation of retinal capillaries in
detectable (Fig. 5). All in all, the activation of norrin-deficient mice. Possible explanations are
the Wnt/β-­catenin signaling by the RPE-derived that (1) norrin was not secreted in high-enough
Norrin lead to a slight, but not sufficient amounts, (2) did not reach the newly formed ves-
improvement of the BRB, at least for CLN5 and sels of the inner retina, or (3) was not available at
CAV1. a time when the vessels are competent to react to
Retinal Pigmented Epithelium-Derived Ectopic Norrin Does Not Promote Intraretinal Angiogenesis… 247

Fig. 5 Immunohistochemistry of claudin5 (magenta) in of CLN5 in retinal endothelial cells. GCL ganglion cell
the retina of 6-week-old wildtype, Ndpy/+/Rpe65-Norrin, layer, INL inner nuclear layer, ONL outer nuclear layer,
NdpKO, and NdpKO/Rpe65-Norrin mice. Arrow: localiza- CC choriocapillaris; Nuclei were counterstained with
tion of CLN5 in retinal endothelial cells. Asterisk: absence Dapi. n = 3

norrin. We regard explanation (2) as unlikely, followed by an increase from P1-P15 and a sta-
since ectopic norrin broadly induced β-catenin in bile expression in adulthood [7]. We therefore
cells of the inner retina in norrin-deficient ani- conclude that transgenic norrin was not present in
mals, an observation that indicates the presence high enough amounts in the inner retina (expla-
of transgenic norrin. We also rule out explanation nation 1). This finding is surprising as we
(3) as we observed high expression of Rpe65 at a observed in a previous study that Rpe65-Norrin
time when intraretinal capillaries are formed. Our mice have substantially higher amounts of reti-
findings in the mouse eye correlate with those nal norrin than their wildtype littermates.
observed in rat eyes in which mRNA expression Furthermore, transgenic norrin in these
of Rpe65 was first detected at embryonic day 18, animals promotes vascular repair following
248 A. E. Dillinger and E. R. Tamm

­oxygen-­induced retinopathy [9] and protects but not Wnt-induced FZD4/beta-catenin signaling.
Cell. 2009;139:299–311.
photoreceptors against light-induced damage [2]. 6. Luhmann UFO, Lin J, Acar N, Lammel S, Feil
Apparently very high amounts of ectopic norrin S, Grimm C, Seeliger MW, Hammes H-P, Berger
are required in the inner retina to augment capil- W. Role of the Norrie disease pseudoglioma gene
larization [8]. This observation should be taken in sprouting angiogenesis during development of
the retinal vasculature. Invest Ophthalmol Vis Sci.
into account in scenarios in which recombinant 2005;46:3372–82.
Norrin is planned to be used as a therapeutic 7. Manès G, Leducq R, Kucharczak J, Pagès A, Schmitt-­
agent to treat retinal vascular abnormalities. Bernard CF, Hamel CP. Rat messenger RNA for the
retinal pigment epithelium-specific protein RPE65
gradually accumulates in two weeks from late embry-
Acknowledgments We thank Eva Wirkert for expert
onic days 1. FEBS Lett. 1998;423:133–7.
technical assistance.
8. Ohlmann A, Scholz M, Goldwich A, Chauhan BK,
Hudl K, Ohlmann AV, Zrenner E, Berger W, Cvekl
A, Seeliger MW, Tamm ER. Ectopic norrin induces
References growth of ocular capillaries and restores normal reti-
nal angiogenesis in Norrie disease mutant mice. J
Neurosci. 2005;25:1701–10.
1. Boulanger A, Liu S, Henningsgaard AA, Yu S,
9. Ohlmann A, Seitz R, Braunger B, Seitz D, Bösl MR,
Redmond TM. The upstream region of the Rpe65
Tamm ER. Norrin promotes vascular regrowth after
gene confers retinal pigment epithelium-specific
oxygen-induced retinal vessel loss and suppresses
expression in vivo and in vitro and contains criti-
retinopathy in mice. J Neurosci. 2010;30:183–93.
cal octamer and E-box binding sites. J Biol Chem.
10. Richter M, Gottanka J, May CA, Welge-Lüssen U,
2000;275:31274–82.
Berger W, Lütjen-Drecoll E. Retinal vasculature
2. Braunger BM, Ohlmann A, Koch M, Tanimoto N,
changes in Norrie disease mice. Invest Ophthalmol
Volz C, Yang Y, Bösl MR, Cvekl A, Jägle H, Seeliger
Vis Sci. 1998;39(12):2450–7.
MW, Tamm ER. Constitutive overexpression of
11. Xu Q, Wang Y, Dabdoub A, Smallwood PM, Williams
Norrin activates Wnt/β-catenin and endothelin-2 sig-
J, Woods C, Kelley MW, Jiang L, Tasman W, Zhang
naling to protect photoreceptors from light damage.
K, Nathans J. Vascular development in the retina and
Neurobiol Dis. 2013;50:1–12.
inner ear. Cell. 2004;116:883–95.
3. Connolly SE, Hores TA, Smith L, D’Amore
12. Ye X, Wang Y, Cahill H, Yu M, Badea TC, Smallwood
PA. Characterization of vascular development in the
PM, Peachey NS, Nathans J. Norrin, frizzled-4,
mouse retina. Microvasc Res. 1988;36:275–90.
and Lrp5 signaling in endothelial cells controls a
4. Díaz-Coránguez M, Ramos C, Antonetti DA. The
genetic program for retinal vascularization. Cell.
inner blood-retinal barrier: cellular basis and develop-
2009;139:285–98.
ment. Vis Res. 2017;139:123–37.
13. Ye X, Smallwood P, Nathans J. Expression of the
5. Junge HJ, Yang S, Burton JB, Paes K, Shu X, French
Norrie disease gene (Ndp) in developing and adult
DM, Costa M, Rice DS, Ye W. TSPAN12 regulates
mouse eye, ear, and brain. Gene Expr Patterns.
retinal vascular development by promoting Norrin-
2011;11:151–5.
Caveolin-1 in Müller Glia Exists
as Heat-Resistant, High Molecular
Weight Complexes

Eric N. Enyong, Jami Gurley, Virginie Sjoelung,


and Michael H. Elliott

Abstract revealed an interactome network of intermedi-


ate filament, desmosomes, and actin-, and
Caveolin-1 (Cav1), the core structural and microtubule-based cytoskeleton. These results
scaffolding protein of caveolae membrane suggest Cav1 domains in Müller glia act as a
domains, is highly expressed in many retinal scaffolding nexus for the cytoskeleton.
cells and is associated with ocular diseases.
Cav1 regulates innate immune responses and Keywords
is implicated in neuroinflammatory and neu-
Caveolin-1 · Caveolae · Müller glia ·
roprotective signaling in the retina. We have
Immunoprecipitation · Mass spectrometry ·
shown that Cav1 expression in Müller glia
Cav1 complexes · Cavin1/PTRF
accounts for over 70% of all retinal Cav1
expression. However, the proteins interacting
with Cav1 in Müller glia are not established.
Here, we show that immortalized MIO-M1 1 Introduction
Müller glia, like endogenous Müller glia,
highly express Cav1. Surprisingly, we found Caveolin-1 (Cav1), the signature structural pro-
that Cav1 in MIO-M1 cells exists as heat-­ tein of caveolae is known to play a role in cell
resistant, high molecular weight complexes signaling, lipid metabolism, endocytosis, and
that are stable after immunoprecipitation (IP). mechanotransduction [3, 24, 25]. Cav1 has been
Mass spectrometric analysis of high molecu- associated with ocular neuroinflammation, age-­
lar weight Cav1 complexes after Cav1 IP related macular degeneration, diabetic retinopa-
thy, blood-retinal barrier function, and primary
E. N. Enyong · J. Gurley · M. H. Elliott (*) open-angle glaucoma [7, 9–11, 30]. It is expressed
Department of Physiology, Dean A. McGee Eye in a variety of cell types including retinal pig-
Institute, University of Oklahoma Health Sciences ment epithelium (RPE) and choroidal and retinal
Center, Oklahoma City, OK, USA vascular endothelium [8, 11]. Furthermore, Cav1
Department of Ophthalmology, Dean A. McGee Eye is highly expressed in Müller glia [11, 21, 26],
Institute, University of Oklahoma Health Sciences and its expression correlates with Müller glia dif-
Center, Oklahoma City, OK, USA
e-mail: Michael-Elliott@ouhsc.edu ferentiation [8, 23]. We have shown that neuro-
retinal Cav1 expression overwhelmingly accounts
V. Sjoelung
Department of Cell Biology, University of Oklahoma for the majority of Cav1 expression in the retina,
Health Sciences Center, Oklahoma City, OK, USA with most Cav1 being localized to Müller glia

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 249
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_36
250 E. N. Enyong et al.

[11]. While Cav1 is highly expressed in Müller microvascular endothelial cells (RMECs) were
glia, its function in these cells is only beginning cultured in Endothelial Cell Basal Medium-2
to be appreciated. Cav1 regulates cytokine secre- (Lonza Cat#: CC-3156), supplemented with 2%
tion and immune cell influx into the retina, as FBS, human fibroblast growth factor, vascular
global Cav1 knockout (KO) simultaneously sup- endothelial growth factor, insulin-like growth
presses cytokine secretion and increases immune factor-1, ascorbic acid, gentamicin-amphotericin
cell influx into the retina [19]. Neuroretinal dele- B hydroxycortisone, and human endothelial
tion of Cav1 suppresses both proinflammatory growth factor. Cells were maintained in a humidi-
cytokine secretion and immune cell infiltration fied atmosphere of 5% CO2, at 37 °C.
into the retina [11], further confirming a role for
Müller glial Cav1 in innate immune responses.
Cav1 is significantly upregulated in Müller glia 2.2 Western Blotting
in autoimmune uveitis [12]. The role of Cav1 as
an immune modulator is likely cell-context Cells were lysed in buffer containing 120 mM
dependent as it can either promote or suppress octylglucoside, 150 mM NaCl, 10 mM Tris–HCl
the inflammatory response depending on the cell pH 7.4, 0.5 mM EDTA, 0.1% Triton X-100 and
type examined [19, 31, 32]. Müller glia express protease inhibitor cocktail. Lysates were cleared
toll-like receptors (TLRs), whose activities can by centrifugation and protein concentration was
be enhanced or suppressed by interaction with determined using a BCA reagent (ThermoFisher
Cav1 [22, 31]. Further, we and others have shown Scientific). Equal amounts of proteins were sepa-
Cav1 to be an important regulator of blood-­retinal rated by reducing SDS-PAGE and were trans-
barrier (BRB) function [2, 18, 19, 33]. ferred to nitrocellulose membranes. Membranes
While Müller glia abundantly express Cav1, it were blocked for 1 h in 5% BSA and were probed
is unclear what proteins interact with Cav1 in with primary antibodies of choice: rabbit anti-­
these cells. The aim of this study was to identify Cav1 (Cell Signaling Technology, cat. #3267,
the Cav1 interactome in MIO-M1 Müller glia by 1:1000), rabbit anti-PTRF (Abcam, 1:1000) and
immunoprecipitating Cav1 and analyzing mouse anti-β-actin (Abcam, 1:5000). Primary
immune complexes by mass spectrometry. We antibodies were detected using Horseradish per-
show that Cav1 in MIO-M1 Müller glia exists as oxidase (HRP)-conjugated secondary antibodies.
high molecular weight aggregates, which are To visualize protein bands after SDS-PAGE elec-
resistant to heating in reducing SDS-PAGE buf- trophoresis, gels were stained for 1 h with
fer. Mass spectrometric analysis of Cav1 com- SimplyBlue™ Safestain (Thermofisher
plexes revealed a network of cytoskeletal proteins Scientific). Western blot images were captured
that interact with Cav1. using the In Vivo F-Pro imaging system.

2 Materials and Methods 2.3 Immunoprecipitation

2.1 Cell Lines and Culture Immunoprecipitation was performed using the
Conditions Dynabeads™ Protein G Immunoprecipitation Kit
(ThermoFisher Scientific) according to the man-
Immortalized MIO-M1 Müller glia were cultured ufacturer’s instructions. Briefly, Cav1 primary
in DMEM (1X) + GlutaMax™-I (ThermoFisher antibody (Cell Signaling Technology, cat. #3267)
Scientific) supplemented with 10% fetal bovine was conjugated to magnetic beads for 20 min at
serum (FBS), and 1% penicillin-streptomycin. room temperature (RT). Then, the beads-­antibody
Prostate cancer cells (PC3s) were cultured in conjugate was incubated with equal amounts of
F-12K medium (ATCC Cat#: 30-2004). Retinal protein lysates for 20 min at RT. After several
Caveolin-1 in Müller Glia Exists as Heat-Resistant, High Molecular Weight Complexes 251

rounds of washing, beads were resuspended in to the FASP (filter-aided sample preparation)
Laemmli buffer and the immunoprecipitates were protocol [34] and digested overnight with
separated by reducing SDS-PAGE without heat- Sequencing Grade Modified Trypsin (Promega,
ing. The gel was stained for 1 h using V5111) at 37 °C in 40 mM NH4HCO3. Peptides
SimplyBlue™ SafeStain and washed several were desalted, concentrated, and loaded onto
times with water. Visible Cav1 complexes were C18 sequencing columns (AcclaimTM PepMapTM
excised and used for mass spectrometry analysis 100 C18, ThermoFisher). Peptide elution was
(Fig. 1). A portion of the immune complexes was performed using a 90-min acetonitrile gradient
boiled in Laemmli buffer, separated by reducing for label-free quantification. Eluted peptides
SDS-PAGE, and transferred to nitrocellulose were analyzed by LC-MS/MS analysis using a
membranes for Western blotting. Thermo Lumos Fusion tribrid Orbitrap mass
spectrometer, coupled to an Ultimate 3000 RSLC
nano ultra-high-performance liquid chromatog-
2.4 Mass Spectrometry raphy (UHPLC) system. Proteins were identified
by Proteome Discoverer 2.4, with SEQUEST as
Cav1 immunoprecipitates were separated by the search engine. Protein identification required
SDS-PAGE, gel-stained, and visible high molec- the detection of at least two peptides per protein.
ular weight Cav1 complexes excised for mass STRING open-source software was used to iden-
spectrometry (Fig. 2d). Proteins were subjected tify protein networks.

Gel staining and excision


Mass spectrometry
of Cav1 complexes
Protein
extraction Stained gel
80000
Cav1 70000
Complexes
60000
IgG
Protein 50000
40000
mixture 30000
20000
10000
0
1014 1016 1018 1020 1022 1024 1026
Mock

MIO-M1

SDS-PAGE
Cav1 IP electrophoresis
Western blotting
Cav1 blot Data analysis
Cav1
(STRING)
Complexes

IgG

Cav1
monomer
Mock

MIO-M1

Fig. 1 Schematic overview of Cav1 immunoprecipitation without heating. Gels were stained for 1 h and visible
and mass spectrometry workflow. Proteins were extracted Cav1 complexes were excised and analyzed by mass spec-
from untreated MIO-M1 Müller glia and were immuno- trometry. A portion of the immunoprecipitate was trans-
precipitated using Cav1 primary antibodies. Cav1 immu- ferred to nitrocellulose membranes to evaluate interactions
noprecipitates were separated by reducing SDS-PAGE by Western blotting
252 E. N. Enyong et al.

3 Results mass spectrometry. Interestingly, the majority of


these proteins including β-actin (ACTB), myosin,
3.1 Cav1 in MIO-M1 Müller Glia vimentin (VIM), plectin (PLEC), and nestin
Exist as High Molecular (NES) have been shown to play a role in the cyto-
Weight Complexes skeleton and cell-cell junction structure [4].

To determine the protein composition of Cav1


complexes, we first evaluated the expression of 4 Discussion
Cav1 in authenticated MIO-Müller glia and com-
pared the expression to retinal microvascular In this study, we sought to identify the protein
endothelial cells (RMEC), which also abundantly composition of Cav1 complexes in MIO-M1
express Cav1 [29]. Our results show that MIO-­ cells. We show for the first time that Cav1 in
M1 cells, like endogenous Müller glia, abun- MIO-M1 cells exists as heat-resistant, high
dantly express Cav1 (Fig. 2). To our surprise, we molecular weight aggregates, which interact
observed that the majority of Cav1 in MIO-M1 with important cytoskeletal proteins. Cav1 is the
cells exist as high molecular weight complexes major protein component of caveolae [5, 6, 27],
(Fig. 2a) which are resistant to heating in reduc- flask-­
shaped plasma membrane invaginations
ing SDS-PAGE buffer (Fig. 2b). However, upon whose formation requires another protein called
heating at 98 °C for 10 min, Cav1 complexes dis- Cavin1 or PTRF (Polymerase I and Transcript
sociate and migrate as monomers on reducing Release Factor) [14, 20]. It is currently unclear
SDS-PAGE (Fig. 2c). High molecular weight why Cav1 exist in this aggregated form in MIO-
Cav1 complexes in MIO-M1 cells remain rela- M1 cells, as opposed to the monomeric form in
tively stable after Cav1 immunoprecipitation RMEC cells. However, we speculate that the
(Fig. 2d). On the contrary, Cav1 in RMECs absence of Cavin1/PTRF expression in MIO-
migrates predominantly as monomers on SDS-­ M1 cells may provide an explanation for this
PAGE gels without heating. While most Cav1 phenomenon. Cavin1/PTRF stabilizes Cav1 in
complexes in MIO-M1s remain stable after heat- multiple tissues, as Cavin1/PTRF deficiency
ing at 70 °C for 10 min, the small fraction of downregulates Cav1 protein expression [20].
aggregated Cav1 in RMECs dissociates to mono- However, Cav1 is stably expressed in prostate
mers after heating at 70 °C. These data suggest cancer (PC3) cells without Cavin1/PTRF [1, 14,
that different proteins may be interacting with and15] in functional domains described as “Cav1
stabilizing Cav1 complexes in MIO-M1 cells. scaffolds” [16, 17]. Further, multiple studies
have shown that approximately 150 Cav1 mol-
ecules are required per caveola formed [25].
3.2 Proteins Associated with Cav1 However, during caveolae biogenesis, Cav1
Complexes Are Involved form oligomers of 12–16 Cav1 molecules,
with the Cell Cytoskeleton which associate with lipid rafts in the Golgi and
adopt detergent-resistant properties [13, 28],
Next, we were interested in identifying the pro- similar to Cav1 on the plasma membrane. Thus,
teins that interact with Cav1 complexes in MIO-­ it is intriguing to speculate that in the absence of
M1 cells. To identify the protein composition of Cavin1/PTRF in MIO-M1 Müller glial cells,
Cav1 complexes, we immunoprecipitated Cav1 these Cav1 high molecular weight oligomers
from MIO-M1 cells and analyzed the high molec- represent non-caveolar Cav1 scaffolds previ-
ular weight complexes by mass spectrometry. We ously described [16, 17]. Therefore, the expres-
analyzed the same complexes from three repli- sion of Cavin1/PTRF in MIO-M1 cells likely
cate samples (Fig. 2d). Thirty-three proteins were provides a mechanism to biochemically resolve
found to associate with Cav1 complexes after Cav1 scaffolds (Fig. 3).
Caveolin-1 in Müller Glia Exists as Heat-Resistant, High Molecular Weight Complexes 253

A B C

D Stained Gel Cav1 blot


Mock Mock
IP 1 2 3 IP 1 2 3

B1
B2 Cav1
190 B3 complexes
120
85 IgG
60
50
40

25 Cav1
monomer
20
15

10

Fig. 2 Cav1 in MIO-M1 cells exists in heat-resistant, heating at 70 °C for 10 min. (c) Representative Western
high molecular weight complexes. (a) Representative blots showing that Cav1 migrates as a monomer in reduc-
Western blots showing expression of Cav1 in MIO-M1 ing SDS-PAGE buffer only after rigorous heating at 98 °C
and RMEC cells. MIO-M1 cells, like endogenous Müller for 10 min. (d) Stained gel and representative Western blot
glia, abundantly expresses Cav1. Interestingly, Cav1 in showing stable Cav1 complexes after Cav1 immunopre-
these cells exists in high molecular weight complexes. (b) cipitation. Bands from the stained gel (B1, B2, and B3
Cav1 complexes are resistant to heating at 70 °C for indicated by arrows) were excised and analyzed by mass
10 min in reducing SDS-PAGE. On the contrary, Cav1 in spectrometry
RMEC cells was mostly monomeric both at RT and after
254 E. N. Enyong et al.

Fig. 3 Proteins that interact with Cav1 complexes are actions. A total of 33 proteins were found to interact with
associated with the cell cytoskeleton. Cav1 protein-­ Cav1 complexes, most of which play a role in the cell
protein interaction by STRING analysis. Cav1 complexes cytoskeletal architecture and include β-actin (ACTB),
were analyzed by mass spectrometry and STRING open-­ myosin (MYO5A), vimentin (VIM) plectin (PLEC), and
source database was used to identify protein-protein inter- nectin (NEC)

Acknowledgments This work was supported by NIH Virginie Sjoelund was supported in part by the National
Grants R01EY019494 (MHE) and NEI Core Grant Institute of General Medical Sciences of the National
P30EY021725, by Presbyterian Health Foundation Institutes of Health under award number P20GM103447.
Research Scholar Award to Eric Enyong, and by an unre- We thank the Laboratory for Molecular Biology and
stricted grant to the OUHSC Department of Cytometry Research at OUHSC for the use of the Core
Ophthalmology from Research to Prevent Blindness, Inc. Facility, which provided the proteomics services.
Caveolin-1 in Müller Glia Exists as Heat-Resistant, High Molecular Weight Complexes 255

References cholesterol dynamics and the actin cytoskeleton


impacting secretion pathways. Mol Cell Proteom.
2012;11:M111012245.
1. Aung CS, Hill MM, Bastiani M, Parton RG, Parat
16. Khater IM, Aroca-Ouellette ST, Meng F, Nabi IR,
MO. PTRF-cavin-1 expression decreases the migra-
Hamarneh G. Caveolae and scaffold detection from
tion of PC3 prostate cancer cells: role of matrix metal-
single molecule localization microscopy data using
loprotease 9. Eur J Cell Biol. 2011;90:136–42.
deep learning. PLoS One. 2019a;14:e0211659.
2. Chow BW, Gu C. Gradual suppression of transcyto-
17. Khater IM, Liu Q, Chou KC, Hamarneh G, Nabi
sis governs functional blood-retinal barrier formation.
IR. Super-resolution modularity analysis shows poly-
Neuron. 2017;93:1325–1333 e1323.
hedral caveolin-1 oligomers combine to form scaf-
3. Cohen AW, Hnasko R, Schubert W, Lisanti MP. Role
folds and caveolae. Sci Rep. 2019b;9:9888.
of caveolae and caveolins in health and disease.
18. Klaassen I, Hughes JM, Vogels IM, Schalkwijk CG,
Physiol Rev. 2004;84:1341–79.
Van Noorden CJ, Schlingemann RO. Altered expres-
4. Fletcher DA, Mullins RD. Cell mechanics and the
sion of genes related to blood-retina barrier disrup-
cytoskeleton. Nature. 2010;463:485–92.
tion in streptozotocin-induced diabetes. Exp Eye Res.
5. Glenney JR Jr. Tyrosine phosphorylation of a 22-kDa
2009;89:4–15.
protein is correlated with transformation by Rous sar-
19. Li X, Gu X, Boyce TM, Zheng M, Reagan AM, Qi
coma virus. J Biol Chem. 1989;264:20163–6.
H, Mandal N, Cohen AW, Callegan MC, Carr DJ,
6. Glenney JR Jr, Soppet D. Sequence and expression
Elliott MH. Caveolin-1 increases proinflammatory
of caveolin, a protein component of caveolae plasma
chemoattractants and blood-retinal barrier breakdown
membrane domains phosphorylated on tyrosine in
but decreases leukocyte recruitment in inflammation.
Rous sarcoma virus-transformed fibroblasts. Proc
Invest Ophthalmol Vis Sci. 2014;55:6224–34.
Natl Acad Sci U S A. 1992;89:10517–21.
20. Liu L, Brown D, McKee M, Lebrasseur NK, Yang D,
7. Gu X, Fliesler SJ, Zhao YY, Stallcup WB, Cohen AW,
Albrecht KH, Ravid K, Pilch PF. Deletion of Cavin/
Elliott MH. Loss of caveolin-1 causes blood-retinal
PTRF causes global loss of caveolae, dyslipidemia,
barrier breakdown, venous enlargement, and mural
and glucose intolerance. Cell Metab. 2008;8:310–7.
cell alteration. Am J Pathol. 2014a;184:541–55.
21. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar
8. Gu X, Reagan A, Yen A, Bhatti F, Cohen AW, Elliott
K, Goldman M, Tirosh I, Bialas AR, Kamitaki N,
MH. Spatial and temporal localization of caveolin-1
Martersteck EM, Trombetta JJ, Weitz DA, Sanes JR,
protein in the developing retina. Adv Exp Med Biol.
Shalek AK, Regev A, McCarroll SA. Highly parallel
2014b;801:15–21.
genome-wide expression profiling of individual cells
9. Gu X, Reagan AM, McClellan ME, Elliott
using nanoliter droplets. Cell. 2015;161:1202–14.
MH. Caveolins and caveolae in ocular physiol-
22. Mirza MK, Yuan J, Gao XP, Garrean S, Brovkovych
ogy and pathophysiology. Prog Retin Eye Res.
V, Malik AB, Tiruppathi C, Zhao YY. Caveolin-1 defi-
2017;56:84–106.
ciency dampens Toll-like receptor 4 signaling through
10. Gurley JM, Elliott MH. The role of caveolin-
eNOS activation. Am J Pathol. 2010;176:2344–51.
1 in retinal inflammation. Adv Exp Med Biol.
­
23. Nelson BR, Ueki Y, Reardon S, Karl MO, Georgi S,
2019;1185:169–73.
Hartman BH, Lamba DA, Reh TA. Genome-wide
11. Gurley JM, Gmyrek GB, McClellan ME, Hargis
analysis of Muller glial differentiation reveals a
EA, Hauck SM, Dozmorov MG, Wren JD, Carr DJJ,
requirement for Notch signaling in postmitotic cells
Elliott MH. Neuroretinal-derived caveolin-1 promotes
to maintain the glial fate. PLoS One. 2011;6:e22817.
endotoxin-induced inflammation in the murine retina.
24. Parton RG. Caveolae: structure, function, and
Invest Ophthalmol Vis Sci. 2020;61:19.
relationship to disease. Annu Rev Cell Dev Biol.
12. Hauck SM, Dietter J, Kramer RL, Hofmaier F,
2018;34:111–36.
Zipplies JK, Amann B, Feuchtinger A, Deeg CA,
25. Parton RG, del Pozo MA. Caveolae as plasma mem-
Ueffing M. Deciphering membrane-associated molec-
brane sensors, protectors and organizers. Nat Rev Mol
ular processes in target tissue of autoimmune uveitis
Cell Biol. 2013;14:98–112.
by label-free quantitative mass spectrometry. Mol
26. Roesch K, Jadhav AP, Trimarchi JM, Stadler
Cell Proteom. 2010;9:2292–305.
MB, Roska B, Sun BB, Cepko CL. The transcrip-
13. Hayer A, Stoeber M, Bissig C, Helenius A. Biogenesis
tome of retinal Muller glial cells. J Comp Neurol.
of caveolae: stepwise assembly of large caveolin and
2008;509:225–38.
cavin complexes. Traffic. 2010;11:361–82.
27. Rothberg KG, Heuser JE, Donzell WC, Ying YS,
14. Hill MM, Bastiani M, Luetterforst R, Kirkham M,
Glenney JR, Anderson RG. Caveolin, a protein
Kirkham A, Nixon SJ, Walser P, Abankwa D, Oorschot
component of caveolae membrane coats. Cell.
VM, Martin S, Hancock JF, Parton RG. PTRF-Cavin,
1992;68:673–82.
a conserved cytoplasmic protein required for caveola
28. Sargiacomo M, Scherer PE, Tang Z, Kubler E, Song
formation and function. Cell. 2008;132:113–24.
KS, Sanders MC, Lisanti MP. Oligomeric structure of
15. Inder KL, Zheng YZ, Davis MJ, Moon H, Loo D,
caveolin: implications for caveolae membrane organi-
Nguyen H, Clements JA, Parton RG, Foster LJ, Hill
zation. Proc Natl Acad Sci U S A. 1995;92:9407–11.
MM. Expression of PTRF in PC-3 Cells modulates
256 E. N. Enyong et al.

29. Stitt AW, Burke GA, Chen F, McMullen CB, Vlassara 31. Wang XM, Kim HP, Nakahira K, Ryter SW, Choi
H. Advanced glycation end-product receptor interac- AM. The heme oxygenase-1/carbon monoxide
tions on microvascular cells occur within caveolin-­ pathway suppresses TLR4 signaling by regulating
­
rich membrane domains. FASEB J. 2000;14:2390–2. the interaction of TLR4 with caveolin-1. J Immunol.
30. Thorleifsson G, Walters GB, Hewitt AW, Masson G, 2009;182:3809–18.
Helgason A, DeWan A, Sigurdsson A, Jonasdottir 32. Wang XM, Kim HP, Song R, Choi AM. Caveolin-1
A, Gudjonsson SA, Magnusson KP, Stefansson H, confers antiinflammatory effects in murine macro-
Lam DS, Tam PO, Gudmundsdottir GJ, Southgate phages via the MKK3/p38 MAPK pathway. Am J
L, Burdon KP, Gottfredsdottir MS, Aldred MA, Respir Cell Mol Biol. 2006;34:434–42.
Mitchell P, St Clair D, Collier DA, Tang N, Sveinsson 33. Wang Z, Liu CH, Huang S, Fu Z, Tomita Y, Britton
O, Macgregor S, Martin NG, Cree AJ, Gibson J, WR, Cho SS, Chen CT, Sun Y, Ma JX, He X, Chen
Macleod A, Jacob A, Ennis S, Young TL, Chan JC, J. Wnt signaling activates MFSD2A to suppress vas-
Karwatowski WS, Hammond CJ, Thordarson K, cular endothelial transcytosis and maintain blood-­
Zhang M, Wadelius C, Lotery AJ, Trembath RC, Pang retinal barrier. Sci Adv. 2020;6:eaba7457.
CP, Hoh J, Craig JE, Kong A, Mackey DA, Jonasson 34. Wisniewski JR. Filter-aided sample prepara-
F, Thorsteinsdottir U, Stefansson K. Common variants tion for proteome analysis. Methods Mol Biol.
near CAV1 and CAV2 are associated with primary 2018;1841:3–10.
open-angle glaucoma. Nat Genet. 2010;42:906–9.
Role of VLC-PUFAs in Retinal
and Macular Degeneration

Aruna Gorusupudi, Uzoamaka Nwagbo,


and Paul S. Bernstein

Abstract 1 Introduction

Very-long-chain polyunsaturated fatty acids Very-long-chain polyunsaturated fatty acids


(VLC-PUFAs) are a special class of fatty acids (VLC-PUFAs) are a special class of fatty acids
that are present in the retina and a few other present in mammalian retina and a few other tis-
human tissues. They cannot be synthesized de sues. The rod outer segments (ROS) of the retina
novo and are rarely present in dietary sources. contain a stack of closed membranous compart-
Structurally, these lipids are composed of a ments known as disks encased by plasma mem-
proximal end with a typical saturated fatty brane connected to the inner segment by a
acid character and a distal end more character- modified cilium [19]. ROS membranes are
istic of common PUFAs. They have not been enriched in phospholipids with DHA and VLC-­
studied in detail until recently due to their low PUFAs at the sn-2 and sn-1 positions, respec-
abundance in these tissues and technical diffi- tively [2]. Lipids in disk membranes of ROS not
culties in assaying these lipids by conven- only serve as structural support for rhodopsin [3]
tional chromatography. This unique class of and for maintenance of ROS disk morphology,
lipids has chain lengths greater than 24 car- but they also play a critical role in maintaining
bons, with the longest typically 38 carbons the activity of other receptor proteins [12, 14].
long. There is increasing interest in under- In 1987, Aveldano described the presence of
standing their roles in the maintenance of reti- unusually long polyenoic fatty acids in mamma-
nal membrane integrity and the prevention of lian retina [4]. Later studies revealed the structure
macular degeneration and inherited retinal and number of double bonds and the n-3 and n-6
diseases. variations of these VLC-PUFAs in retina [4]. The
synthetic pathway of VLC-PUFAs was first
Keywords investigated by Rotstein and Aveldano in 1987
using C14-acetate [21, 22]. After incubation of
VLC-PUFAs · STGD3 · AMD · ELOVL4 · bovine retinal tissue with labeled 20:4 n-6 (AA),
32:6 n-3 22:5 n-3 (DPA) and 22:6 n-3 (DHA), the authors
observed efficient uptake and esterification into
A. Gorusupudi · U. Nwagbo · P. S. Bernstein (*) phospholipids. They also found that radiolabel-
Department of Ophthalmology and Visual Sciences, ing predominated in VLC-PUFAs in microsomal
John A. Moran Eye Center, University of Utah fractions of photoreceptor membranes, and they
School of Medicine, Salt Lake City, UT, USA observed that the selective absorption was
e-mail: paul.bernstein@hsc.utah.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 257
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_37
258 A. Gorusupudi et al.

Fig. 1 Biosynthetic
pathway of VLC-PUFAs

22:5 > 22:6 > 20:4. In vivo retinal injections of mutant protein mislocalizes, or it co-aggregates
labeled LC-PUFAs in murine models also with wild-type protein and then mislocalizes,
revealed that EPA gets elongated to VLC-PUFAs, thereby leading to disruption of VLC-PUFA bio-
whereas DHA does not [25]. After the discovery synthesis [13, 28]. Mice with homozygous
of the ELOVL4 elongase enzyme, responsible for knockout of Elovl4 [27] or knock-in of human
the biosynthesis of VLC-PUFA in retina (Fig. 1), ELOVL4 mutations [17] exhibit neonatal lethal-
transduction experiments in PC-12 cells con- ity due to disruption of the skin permeability bar-
firmed that EPA and DPA can act as direct pre- rier, while heterozygous mutations in mice show
cursors for the biosynthesis of n-3 VLC-PUFAs, lipofuscin accumulation in the retina [5, 20], loss
while DHA cannot [1]. of VLC-PUFAs [5], and RPE atrophy similar to
human STGD3 and AMD [13], but at a slow
pace. STGD3 disease in humans is characterized
2 ELOVL4 and STGD3 by progressive loss of central vision starting dur-
ing adolescence accompanied by foveal atrophy
Three independent mutations of the ELOVL4 and butterfly pattern fleck-like deposits at the
gene are associated with dominant Stargardt-3 level of the retinal pigment epithelium. The
macular dystrophy (STGD3) in humans [6, 16, severity of human STGD3 disease is inversely
30]. All three mutations lead to premature termi- correlated with the dietary intake of VLC-PUFA
nation of the protein and removal of the signal precursors [11], and since there are no reasonable
sequence for post-translational modification in dietary sources of VLC-PUFAs, we recommend
the endoplasmic reticulum. As a result, the the intake of fish oil VLC-PUFA precursors
Role of VLC-PUFAs in Retinal and Macular Degeneration 259

from diet or supplements for this patient to perform mouse feeding studies [29]. We chose
­population [7]. this particular VLC-PUFA because it is present in
both mouse and human retinas, and it could still
be elongated further by endogenous ELOVL4. In
3 ELOVL4 and AMD our studies, we observed rapid and selective reti-
nal bioavailability of our synthetic VLC-PUFA in
Age-related macular degeneration (AMD) is the mice, and we documented improvement in visual
leading cause of vision loss among the elderly. acuity and photopic and scotopic ERG measure-
Epidemiological studies have shown an inverse ments in both wild-type and Elovl4 conditional
correlation of n-3 PUFA dietary intake from fish knockout mice [9]. Although mouse models of
oil or fish consumption with AMD disease pro- human retinal diseases like STGD3 and AMD are
gression [18, 23, 24], and previous studies from available, mice do not have maculas, and Elovl4
our laboratory have reported significantly lower conditional knockouts do not show as severe a
DHA and VLC-PUFA levels in AMD eyes in retinal degeneration as humans with STGD3.
comparison to age-matched controls [15]. In Additionally, standard laboratory mouse diets are
another study, although we did not observe a sig- surprisingly healthy with regard to n-3 LC-PUFA
nificant change in ELOVL4 expression among levels and n-3/n-6 ratios relative to typical human
the subjects, we found a significant decrease in diets [10], so creating substantial deficiencies of
serum EPA/AA and retinal n3/n6 LC-PUFA and VLC-PUFAs in mouse retinas can be quite chal-
VLC-PUFA ratios in AMD donor punches in lenging. Therefore, our laboratory continues to
comparison to age-matched controls [8]. We also seek out alternatives to current mouse models of
showed that retinal VLC-PUFA and n-3/n-6 Elovl4 dysfunction and VLC-PUFA deficiency.
ratios were correlated with systemic biomarkers
of dietary intake of VLC-PUFA precursors. This
suggests that AMD could be due in part to age-­ 5 Future Directions
related ELOVL4 dysfunction and/or inadequate
intake of VLC-PUFA precursors and that inter- In the past decade, there have been great advances
ventions to increase retinal VLC-PUFA levels in cell culture models of retinal degeneration
might be protective against AMD [8]. Therefore, using patient-derived induced pluripotent stem
we have hypothesized that if STGD3 or AMD cells (iPSCs) and retinal organoids (“disease in a
patients were to consume an adequate supply of dish”), and we think such techniques could have
VLC-PUFAs that could be directly used in the value in ELOVL4 and VLC-PUFA research as
retina, it may be possible to bypass the steps of well. In collaboration with Drs. Martin
elongation mediated by ELOVL4 and delay or Friedlander and Kevin Eade at The Scripps
prevent degeneration; however, until recently, Research Institute, we have knocked out ELOVL4
there have been no adequate natural or commer- in human retinal organoids, and we are currently
cial sources of high purity natural or synthetic studying the developmental and metabolic effects
VLC-PUFAs for laboratory or clinical studies to of complete VLC-PUFA deficiency and potential
test their value for treating or preventing eye rescue by supplementation with exogenous syn-
disease. thetic VLC-PUFAs. We have also successfully
created a germline total knockout of the zebrafish
enzyme responsible for VLC-PUFA synthesis in
4 Synthesis of VLC-PUFAs the retina to study physiological and morphologi-
cal effects of the complete absence of VLC-­
In collaboration with members of the Department PUFAs in the living vertebrate eye. Zebrafish are
of Chemistry at the University of Utah, we have an attractive alternative to mice because they live
synthesized a >98% pure VLC-PUFA (32:6 n-3) in an aqueous environment, and they are not
at nearly a one-gram scale, a sufficient quantity susceptible to integumentary drying seen in
­
260 A. Gorusupudi et al.

t­errestrial animals with a total knockout of 6. Bernstein PS, Tammur J, Singh N, Hutchinson A,
Dixon M, Pappas CM, Zabriskie NA, Zhang K,
ELOVL4. Finally, with our potential ability to Petrukhin K, Leppert M, Allikmets R. Diverse macu-
scale up our organic synthetic scheme to make a lar dystrophy phenotype caused by a novel complex
wide variety of VLC-PUFAs in kilogram quanti- mutation in the ELOVL4 gene. Invest Ophthalmol Vis
ties, it is time to revisit AREDS2’s previous nega- Sci. 2001;42(13):3331–6.
7. Choi R, Gorusupudi A, Bernstein PS. Long-term
tive results for EPA and DHA supplementation follow-up of autosomal dominant Stargardt macular
for AMD prevention [26]. It is possible that fish dystrophy (STGD3) subjects enrolled in a fish oil
oil supplementation with VLC-PUFA precursors supplement interventional trial. Ophthalmic Genet.
in AREDS2 was not sufficient to counteract age-­ 2018;39(3):307–13.
8. Gorusupudi A, Liu A, Hageman GS, Bernstein
related declines in ELOVL4 activity in the human PS. Associations of human retinal very long-chain
retina. Oral administration of pre-formed syn- polyunsaturated fatty acids with dietary lipid bio-
thetic VLC-PUFAs would bypass the need for markers. J Lipid Res. 2016;57(3):499–508.
ELOVL4-mediated elongation and could directly 9. Gorusupudi A, Rallabandi R, Li B, Arunkumar R,
Blount JD, Rognon GT, Chang FY, Wade A, Lucas
raise retinal VLC-PUFA levels just as we did in S, Conboy JC, Rainier JD, Bernstein PS. Retinal bio-
mice, so VLC-PUFAs should be considered as a availability and functional effects of a synthetic very-­
component of a supplement formulation to be long-­chain polyunsaturated fatty acid in mice. Proc
tested in a future AREDS3 clinical trial. Natl Acad Sci U S A. 2021;118(6):e2017739118.
10. Hasegawa Y, Chen S-Y, Sheng L, Jena PK, Kalanetra
KM, Mills DA, Wan Y-JY, Slupsky CM. Long-term
Conflict of Interest AG, PSB, and the University effects of western diet consumption in male and
of Utah have filed for a patent on the synthesis of female mice. Sci Rep. 2020;10(1):14686.
VLC-PUFAs to treat eye disease. 11. Hubbard AF, Askew EW, Singh N, Leppert M,
Bernstein PS. Association of adipose and red blood
cell lipids with severity of dominant Stargardt macu-
lar dystrophy (STGD3) secondary to an ELOVL4
Grant Support Foundation Fighting Blindness; mutation. Arch Ophthalmol. 2006;124(2):257–63.
NIH EY-14800; Research to Prevent Blindness. 12. Jastrzebska B, Tsybovsky Y, Palczewski
K. Complexes between photoactivated rhodopsin
and transducin: progress and questions. Biochem J.
2010;428(1):1–10.
13. Karan G, Lillo C, Yang Z, Cameron DJ, Locke KG,
References Zhao Y, Thirumalaichary S, Li C, Birch DG, Vollmer-­
Snarr HR, Williams DS, Zhang K. Lipofuscin
accumulation, abnormal electrophysiology, and pho-
1. Agbaga M-P, Brush RS, Mandal MNA, Henry K,
toreceptor degeneration in mutant ELOVL4 transgenic
Elliott MH, Anderson RE. Role of Stargardt-3 macu-
mice: a model for macular degeneration. Proc Natl
lar dystrophy protein (ELOVL4) in the biosynthesis
Acad Sci U S A. 2005;102(11):4164–9.
of very long chain fatty acids. Proc Natl Acad Sci.
14. Litman BJ, Niu S-L, Polozova A, Mitchell DC. The
2008;105(35):12843–8.
role of docosahexaenoic acid containing phospholip-
2. Agbaga MP, Mandal MN, Anderson RE. Retinal very
ids in modulating G protein-coupled signaling path-
long-chain PUFAs: new insights from studies on
ways. J Mol Neurosci. 2001;16(2):237–42.
ELOVL4 protein. J Lipid Res. 2010;51(7):1624–42.
15. Liu A, Chang J, Lin Y, Shen Z, Bernstein PS. Long-­
3. Aveldano MI. Phospholipid species containing long
chain and very long-chain polyunsaturated fatty acids
and very long polyenoic fatty acids remain with rho-
in ocular aging and age-related macular degeneration.
dopsin after hexane extraction of photoreceptor mem-
J Lipid Res. 2010;51(11):3217–29.
branes. Biochemistry. 1988;27(4):1229–39.
16. Maugeri A, Meire F, Hoyng CB, Vink C, Van
4. Aveldano MI, Sprecher H. Very long chain (C24 to
Regemorter N, Karan G, Yang Z, Cremers FP, Zhang
C36) polyenoic fatty acids of the n-3 and n-6 series in
K. A novel mutation in the ELOVL4 gene causes
dipolyunsaturated phosphatidylcholines from bovine
autosomal dominant Stargardt-like macular dystro-
retina. J Biol Chem. 1987;262(3):1180–6.
phy. Invest Ophthalmol Vis Sci. 2004;45(12):4263–7.
5. Barabas P, Liu A, Xing W, Chen CK, Tong Z,
17. McMahon A, Butovich IA, Mata NL, Klein M,
Watt CB, Jones BW, Bernstein PS, Križaj D. Role
Ritter R 3rd, Richardson J, Birch DG, Edwards AO,
of ELOVL4 and very long-chain polyunsaturated
Kedzierski W. Retinal pathology and skin barrier
fatty acids in mouse models of Stargardt type 3
defect in mice carrying a Stargardt disease-3 mutation
retinal degeneration. Proc Natl Acad Sci U S A.
in elongase of very long chain fatty acids-4. Mol Vis.
2013;110(13):5181–6.
2007;13:258–72.
Role of VLC-PUFAs in Retinal and Macular Degeneration 261

18. Merle BM, Benlian P, Puche N, Bassols A, Delcourt 25. Suh M, Clandinin MT. 20:5n-3 but not 22:6n-3 is a
C, Souied EH, Nutritional AMDTSG. Circulating preferred substrate for synthesis of n-3 very-long-
omega-3 Fatty acids and neovascular age-related chain fatty acids (C24-C36) in retina. Curr Eye Res.
macular degeneration. Invest Ophthalmol Vis Sci. 2005;30(11):959–68.
2014;55(3):2010–9. 26. The Age-Related Eye Disease Study 2 Research,
19. Nechipurenko IV. The enigmatic role of lipids in cilia G. Lutein + zeaxanthin and Omega-3 fatty acids for
signaling. Front Cell Dev Biol. 2020;8:777. age-related macular degeneration: the age-related eye
20. Raz-Prag D, Ayyagari R, Fariss RN, Mandal disease study 2 (AREDS2) randomized clinical trial.
MN, Vasireddy V, Majchrzak S, Webber AL, JAMA. 2013;309(19):2005–15.
Bush RA, Salem N Jr, Petrukhin K, Sieving 27. Vasireddy V, Uchida Y, Salem N Jr, Kim SY, Mandal
PA. Haploinsufficiency is not the key mechanism MNA, Reddy GB, Bodepudi R, Alderson NL, Brown
of pathogenesis in a heterozygous Elovl4 knockout JC, Hama H, Dlugosz A, Elias PM, Holleran WM,
mouse model of STGD3 disease. Invest Ophthalmol Ayyagari R. Loss of functional ELOVL4 depletes
Vis Sci. 2006;47(8):3603–11. very long-chain fatty acids (≥C28) and the unique
21. Rotstein NP, Aveldano MI. Labeling of lipids of retina ω-O-acylceramides in skin leading to neonatal death.
subcellular fractions by [1-14C]eicosatetraenoate Hum Mol Genet. 2007;16(5):471–82.
(20:4(n-6)) docosapentaenoate (22:5(n-3)) and doco- 28. Vasireddy V, Jablonski MM, Khan NW, Wang XF,
sahexaenoate (22:6(n-3)). Biochim Biophys Acta. Sahu P, Sparrow JR, Ayyagari R. Elovl4 5-bp dele-
1987a;921(2):221–34. tion knock-in mouse model for Stargardt-like macular
22. Rotstein NP, Aveldano MI. Labeling of phosphati- degeneration demonstrates accumulation of ELOVL4
dylcholines of retina subcellular fractions by [1-14C] and lipofuscin. Exp Eye Res. 2009;89(6):905–12.
eicosatetraenoate (20:4(n-6)), docosapentaeno- 29. Wade A, Rallabandi R, Lucas S, Oberg C, Gorusupudi
ate (22:5(n-3)) and docosahexaenoate (22:6(n-3)). A, Bernstein PS, Rainier JD. The synthesis of the very
Biochim Biophys Acta. 1987b;921(2):235–44. long chain polyunsaturated fatty acid (VLC-PUFA)
23. SanGiovanni JP, Chew EY. The role of omega-3 long-­ 32:6 n-3. Org Biomol Chem. 2021;19(25):5563–6.
chain polyunsaturated fatty acids in health and disease 30. Zhang K, Kniazeva M, Han M, Li W, Yu Z, Yang
of the retina. Prog Retin Eye Res. 2005;24(1):87–138. Z, Li Y, Metzker ML, Allikmets R, Zack DJ, Kakuk
24. Souied E, Delcourt C, Querques G, Merle B, Smith LE, Lagali PS, Wong PW, MacDonald IM, Sieving
T, Benlian P, N. S. Group. NAT2 study: OMEGA-3 PA, Figueroa DJ, Austin CP, Gould RJ, Ayyagari R,
levels in red blood cells membranes correlates Petrukhin K. A 5-bp deletion in ELOVL4 is associ-
the preventive effect. Invest Ophthalmol Vis Sci. ated with two related forms of autosomal dominant
2013;54(15):3277–7. macular dystrophy. Nat Genet. 2001;27(1):89–93.
Ocular Amyloid, Condensates,
and Aggregates – Higher-Order
Protein Assemblies Participate
in Both Retinal Degeneration
and Function

Michael H. Hayes, DaNae R. Woodard,


and John D. Hulleman

Abstract form higher-order assemblies under normal


conditions within the retina and what function
The formation of higher-order protein assem- do these structures serve? Herein, we present
blies (commonly called protein aggregates) data suggesting that protein aggregation is
has long been associated with disease states, identifiable in multiple retinal layers of nor-
particularly in neurodegenerative disorders. mal, healthy murine retina, and briefly discuss
Within the eye, protein aggregation has also the potential contributions of aggregated pro-
been implicated in various retinal degenera- teins to normal retinal function, with a focus
tive diseases ranging from retinitis pigmen- on the photoreceptor inner and outer
tosa (RP) to Malattia Leventinese/Doyne segment.
Honeycomb Retinal Dystrophy (ML/DHRD)
to age-related macular degeneration (AMD). Keywords
Yet, many essential cellular processes includ-
ing transcription, translation, and the forma- Higher-order assembly · Protein aggregation ·
tion of non-membrane bound organelles Condensate · Amyloid · Thioflavin T · Retinal
require the formation of functional, non-­ degeneration
pathologic protein aggregates to maintain cel-
lular homeostasis. Thus, functional protein
aggregates, also called condensates, likely
play essential roles in maintaining normal 1 Introduction
retina function. However, currently, there is a
critical gap in our knowledge: What proteins The coalescence of proteins, nucleic acids, and
other biomolecules into a recognizable higher-­
M. H. Hayes · D. R. Woodard order assembly called an aggregate is most com-
Department of Ophthalmology, University of Texas monly associated with disease states [1, 2].
Southwestern Medical Center, Dallas, TX, USA However, similar structures, commonly called
J. D. Hulleman (*) condensates, have been shown to contribute to a
Department of Ophthalmology, University of Texas variety of cellular functions [1, 2]. Experimentally
Southwestern Medical Center, Dallas, TX, USA differentiating between these structures can be
Department of Pharmacology, University of Texas difficult, partially a result of confusing nomen-
Southwestern Medical Center, Dallas, TX, USA clature that has recently been clarified by Alberti
e-mail: John.Hulleman@UTSouthwestern.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 263
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_38
264 M. H. Hayes et al.

and Hyman [2]. For brevity and ease of under- bilized with 0.1% Triton-X100, washed with
standing, throughout this book chapter we will PBS, and stained. Thioflavin T (ThT) staining
use the term “aggregate” to describe functional was performed while the sample was on the
condensates as well as disease-associated aggre- microscope stage (50 μM in PBS, Sigma,
gates. In this context, aggregates form a spectrum T3516). Immunofluorescent labeling was per-
of assemblies ranging from soluble oligomeric formed by blocking permeabilized slides in PBS
structures to phase-separated liquid droplets, gel-­ with 1% goat serum and incubating in blocking
like condensates, and solid-like aggregates [1]. buffer or blocking buffer supplemented with
These physical properties are determined by how primary amyloid oligomer antibody (A11,
ordered the interactions are between components 1:500, EMD Millipore, AB9234) overnight.
and determine where along this spectrum an Sections were washed and incubated with sec-
aggregate lies [3]. Additionally, aggregates can ondary antibody (AlexaFluor 546, 1:1000,
be heterogeneous, forming multiple phases Invitrogen, A-11037) for 3 h, stained with DAPI,
within a single structure [3, 4]. Within an aggre- washed, and mounted with Fluoromount G
gate, some proteins act as scaffolds that stabi- (Invitrogen, 00-4959-52). Images were acquired
lize the aggregate and the environment it with a Zeiss Observer microscope at 10x magni-
creates, whereas other proteins freely diffuse into fication. All experiments were conducted ethi-
and out of the aggregate at will [5]. The scaffold- cally in accordance with the ARVO Statement of
ing proteins likely display a more solid-like phe- the Use of Animals in Ophthalmic and Vision
notype, potentially due to a higher-order structure Research and the UT Southwestern Medical
called amyloid [4]. Many proteins can form the Center Institutional Animal Care and Use
underlying cross-ß structure of amyloid, which Committee guidelines.
can be detected by fluorescent dyes, such as thio-
flavin T, or antibodies against oligomeric or
fibrillar amyloid species [6, 7]. The presence of 3 Results
amyloid (which has typically been associated
only with disease states) within normal retina tis- 3.1 Aggregates Are Identifiable
sue highlights a need to understand how some in Multiple Retinal Layers
aggregates are functional, pathological, or
both depending on the environmental context Functional protein aggregation is implicated in
[1, 3, 4, 8]. essential universal cellular functions – for
example, transcription, translation, centrosome
formation, and control of nucleo-cytoplasmic
2 Methods flux as well as cell-type specific functions [1, 2,
9, 10]. Cell-type specific functions of aggre-
2.1 Thioflavin T gated proteins include participation in synaptic
and Immunofluorescence vesicle and peptide hormone clustering and
Staining storage, post-­synaptic signaling, cell-cell inter-
actions, and maintenance of long-term memory
Wild-type C57BL/6J mice were raised for [1, 2]. Because diverse functions of aggregated
12 months under standard conditions, eutha- proteins are observed in other tissues and cell
nized, and their eyes enucleated. The anterior types, we hypothesized that commercially avail-
chamber, lens, and vitreous were immediately able stains and antibodies used for protein
dissected away and unfixed eye cups were aggregation could identify the presence and
embedded into optimal cutting temperature location of aggregates in the retina. To test this
compound and frozen in liquid nitrogen. 12 μm hypothesis, we isolated retinal tissue from
cryosections were collected and briefly permea- healthy 12-month-old wild-­type mice and per-
Ocular Amyloid, Condensates, and Aggregates – Higher-Order Protein Assemblies Participate in Both… 265

formed cryosectioning of unfixed tissue. We did 4 Discussion


not fix the tissue in order to better preserve
native amyloid and to avoid fixative-induced 4.1 Do Aggregates Contribute
protein aggregation. Robust ThT positive stain- to the Structure and Function
ing was observed in multiple retinal layers of the Basal Body
(Fig. 1c). Of note, increased ThT signal was and Permeability Barrier
observed at the juncture of the inner and outer of the Connecting Cilium?
segment (Fig. 1c, arrowhead), suggesting the
presence of robust amyloid-containing struc- Arguably, the most important structure of the
tures at this location. We further corroborated retina is the photoreceptor (PR) outer segment –
this result by staining similar unfixed sections which is connected to the inner segment by the
with an antibody that recognizes peptides in an connecting cilium, a modified primary cilium.
oligomeric form of amyloid, which also recog- Sitting at its base is the basal body, a structure
nized multiple retinal layers (Fig. 1g). remarkably similar to the centrosome and likely
Differences in the ThT and antibody-based relies upon a similar mechanism for assembly
detection of aggregates are likely due to tissue and maintenance. Centrosomes are microtubule
penetration (small molecule vs. large antibody) organizing centers formed by the coalescence,
and specificity differences. Nonetheless, these phase separation, and aggregation of pericentrio-
results demonstrate protein aggregation within lar matrix around centrioles [11]. Thus, aggrega-
normal murine retina, raising the question: what tion and phase separation likely play a role in the
are the functions of these aggregated proteins assembly and maintenance of the structure and
within retinal tissues? function of the connecting cilium. In addition,

Fig. 1 Thioflavin T (ThT) and anti-oligomeric amyloid (e) anti-rabbit secondary antibody, or (g) A11 anti-oligo-
antibodies react with distinct retinal layers. (a) Phase con- meric amyloid antibody and secondary antibody.
trast image of a unfixed cryosection from a 12-month-old Magnification and exposure are identical between corre-
wild-type C57BL/6J mouse. (b) Lack of fluorescence sig- sponding panels. NFL nerve fiber layer, ILM inner limit-
nal immediately before staining the cryosection with ing membrane, RGC retinal ganglion cell layer, IPL inner
50 μM ThT (c). Staining was performed on the micro- plexiform layer, INL inner nuclear layer, OPL outer plexi-
scope stage with images in (b) and (c) taken seconds form layer, ONL outer nuclear layer, IS inner segment, OS
apart. White arrowhead highlights increased staining of outer segment, RPE retinal pigmented epithelium. Scale
photoreceptor inner segments. Similarly treated unfixed bars = 100 μm
cryosections were stained with DAPI (d, f) to stain nuclei,
266 M. H. Hayes et al.

the membrane and intracellular contents of the RIM proteins lining the disc rim. This organi-
outer segment are unique, the result of a selective zation could be attributed to the formation of
permeability barrier – the “gates” to the outer lipid raft structures. Membrane lipids are
segment. These gates form at the transition zone, known to separate into liquid-ordered (Lo) and
the area just distal to the basal body in the proxi- liquid-­disordered (Ld) phases. However, pro-
mal axoneme [11, 12]. Initiation of transition tein aggregation and phase separation can
zone assembly requires CEP290, a protein that is occur on the surface of membranes, and these
mutated in Leber Congenital Amaurosis and 2-­dimensional protein aggregates can have a
Bardet-Beidel syndrome – both of which are significant influence on membrane shape –
associated with retinal degenerative diseases including membrane curvature [8].
[12]. The prototypical permeability barrier of an Interestingly, the lipid species present within
incompletely membrane-bound organelle is that the membrane can influence the oligomeriza-
of the nucleus. The nuclear membrane is studded tion and aggregation of proteins [14]. Do mem-
with pore complexes that are freely permeable to brane-to-protein aggregate interactions
small molecules but restrict larger molecules in a influence the organization of PR discs?
size- and shape-dependent manner, properties Peripherin is known to form homo- or hetero-­
nearly identical to the permeability barrier pres- oligomers, and peripherin oligomerization
ent between the inner and outer segment [13]. In controls disc enclosure, though it is not
fact, the ciliary permeability barrier contains required for it [15]. Furthermore, aggregation-­
nucleoporins. Nucleoporins contain FG repeats – prone mutant forms of rhodopsin have been
intrinsically disordered regions rich in phenylala- shown to destabilize the PR disc membrane
nine and glycine dipeptides. These proteins have [16]. Thus, it is imperative to identify
been shown to phase separate to form ThT-­ aggregation-­prone proteins that associate with
positive hydrogels suggesting amyloid plays a the disc membrane within the PR. Doing so
role in their function [9]. Interestingly, when we would provide an avenue to further understand
stained cryosections of the unfixed healthy how aggregation-­prone proteins interact with
murine retina with ThT we see increased staining membrane components – such as lipid compo-
at the junction of the inner and outer segment sition and transmembrane proteins – to influ-
(Fig. 1c, arrowhead). These observations may ence membrane structure and function.
suggest that the PR ciliary permeability barrier
also relies on amyloid containing structures for
its barrier function. Identifying which proteins 5 Summary and Future
aggregate within the basal body, transition zone, Directions
and if the PR ciliary permeability barrier is
dependent on amyloid-containing aggregates will Despite a long history of being associated with
provide insight into normal retinal physiology disease, aggregated proteins are known contribu-
and routes for potential novel therapeutic tors to cellular function. Our initial experiments
intervention. with ThT and A11 antibody staining suggest
aggregated proteins are commonplace through-
out the retina. Identifying the proteins that aggre-
4.2 Do Aggregates Participate gate, in both normal and diseased retina, is the
in the Structure first step in understanding the contribution of
and Organization aggregated proteins to normal retinal function,
of the Photoreceptor Disc? the pathophysiology of retinal degenerative dis-
eases, and how functional aggregates relate to
The PR disc membrane is a highly organized their disease-associated brethren. Furthermore,
structure. The light-sensing pigment rhodopsin differentiating between functional and disease-­
is located in the center with peripherin and associated protein aggregation provides an
Ocular Amyloid, Condensates, and Aggregates – Higher-Order Protein Assemblies Participate in Both… 267

opportunity to develop novel therapeutic 6. Biancalana M, Koide S. Molecular mechanism of


Thioflavin-T binding to amyloid fibrils. Biochim
approaches. A plethora of strategies have been Biophys Acta (BBA) – Protein Proteomics.
pursued for the treatment of aggregation-­ 2010;1804(7):1405–12.
associated neurological diseases. This includes 7. Kayed R, Head E, Sarsoza F, Saing T, Cotman CW,
anti-aggregation drugs, therapeutic proteases, Necula M, et al. Fibril specific, conformation depen-
dent antibodies recognize a generic epitope com-
immunomodulatory drugs, and small molecule mon to amyloid fibrils and fibrillar oligomers that is
inhibitors [17]. Dyes such as Congo Red, ThT, absent in prefibrillar oligomers. Mol Neurodegener.
and Nile Red have been shown to have therapeu- 2007;2(1):18.
tic potential in decreasing amyloid content [17]. 8. Nesterov SV, Ilyinsky NS, Uversky VN. Liquid-liquid
phase separation as a common organizing principle
However, unlike the brain, the eye is uniquely of intracellular space and biomembranes providing
accessible. Thus, in addition to the potential ther- dynamic adaptive responses. Biochim Biophys Acta
apeutic benefit, applying these dyes to ocular tis- (BBA) – Mol Cell Res. 1868;2021(11):119102.
sue provides an opportunity to identify, and thus 9. Schmidt HB, Görlich D. Nup98 FG domains from
diverse species spontaneously phase-separate into
potentially treat retinal pathology prior to the particles with nuclear pore-like permselectivity. elife.
onset of devastating symptoms such as irrevers- 2015;4:e04251.
ible vision loss. 10. De Joukov N. The centrosome and the primary cil-
ium: the yin and Yang of a hybrid organelle. Cell.
2019;8(7):701.
Acknowledgments This work was funded by the Roger
11. Raff JW. Phase separation and the centrosome: a fait
and Dorothy Hirl Endowed Research Fund and the NEI
accompli? Trends Cell Biol. 2019;29(8):612–22.
Core Grant for Vision Research (P30-EY030413).
12. Wu Z, Pang N, Zhang Y, Chen H, Peng Y, Fu
J, et al. CEP290 is essential for the initiation
of ciliary transition zone assembly. PLoS Biol.
References 2020;18(12):e3001034.
13. Verhey KJ, Yang W. Permeability barriers for gener-
ating a unique ciliary protein and lipid composition.
1. Fassler JS, Skuodas S, Weeks DL, Phillips BT. Protein
Curr Opin Cell Biol. 2016;41:109–16.
aggregation and disaggregation in cells and develop-
14. Amaro M, Šachl R, Aydogan G, Mikhalyov II, Vácha
ment. J Mol Biol. 2021;433(21):167215.
R, Hof M. GM 1 ganglioside inhibits β-amyloid
2. Alberti S, Hyman AA. Biomolecular condensates
oligomerization induced by sphingomyelin. Angew
at the nexus of cellular stress, protein aggrega-
Chem Int Ed. 2016;55(32):9411–5.
tion disease and ageing. Nat Rev Mol Cell Biol.
15. Lewis TR, Makia MS, Castillo CM, Al-Ubaidi MR,
2021;22(3):196–213.
Naash MI, Arshavsky VY. Photoreceptor disc enclo-
3. Shin Y, Brangwynne CP. Liquid phase conden-
sure is tightly controlled by Peripherin-2 oligomeriza-
sation in cell physiology and disease. Science.
tion. J Neurosci. 2021;41(16):3588–96.
2017;357(6357):eaaf4382.
16. Haeri M, Knox BE. Rhodopsin mutant P23H destabi-
4. Hayes MH, Weeks DL. Amyloids assemble as part of
lizes rod photoreceptor disk membranes. PLoS One.
recognizable structures during oogenesis in Xenopus.
2012;7(1):e30101.
Biol Open. 2016;5(6):801–6.
17. Chen G-F, Xu T-H, Yan Y, Zhou Y-R, Jiang Y,
5. Banani SF, Rice AM, Peeples WB, Lin Y, Jain S,
Melcher K, et al. Amyloid beta: structure, biology
Parker R, et al. Compositional control of phase-­
and structure-­ based therapeutic development. Acta
separated cellular bodies. Cell. 2016;166(3):651–63.
Pharmacol Sin. 2017;38(9):1205–35.
Photoreceptor Ion Channels
in Signaling and Disease

Shivangi M. Inamdar, Colten K. Lankford,


and Sheila A. Baker

Abstract ion channels. Members of four of these families


(CNG, HCN, Kv, and Cav) are expressed in PRs
Photoreceptors (PRs) in the neural retina con- and are key to communicating light detection.
vert photon capture into an electrical signal The structure of rods and cones can be most
that is communicated across a chemical syn- simply divided into three compartments; outer
apse to second-order neurons in the retina and segment (OS), inner segment (IS), and synaptic
on through the rest of the visual pathway. This terminal (ST). Each of the ion channels we dis-
information is decoded in the visual cortex to cuss here has a distinct expression pattern along
create images. The activity of PRs depends on the PRs compartments. The OS houses the photo-
the concerted action of several voltage-gated transduction cascade in the disc membranes and
ion channels that will be discussed in this CNG channels in the OS plasma membrane that
chapter. carry the inward dark current [1]. The IS or
“housekeeping” part of the cell contains the ion
Keywords channels responsible for shaping light responses,
HCN1, and Kv2.1/Kv8.2. HCN1 is found
Photoreceptors · Ion channels · CNG · Cav1.4 throughout the IS plasma membrane, axon, and
· HCN1 · Kv2.1 · Kv8.2 · Retinal synapse. Kv2.1/Kv8.2 is restricted to the apical
degeneration portion of the IS plasma membrane. How that
restriction is accomplished is unknown. The ST
is where neurotransmitter release to communi-
1 Introduction cate the light/dark state to second-order neurons
in the retina occurs and is characterized by a syn-
The human genome encodes a superfamily of aptic ribbon beneath which Cav1.4 channels are
voltage-gated ion channels (VGICs) that conduct clustered [2, 3].
the cations sodium, potassium, or calcium. This
superfamily consists of ten distinct families of
2 CNG Channels Respond
to Light
S. M. Inamdar (*) · C. K. Lankford · S. A. Baker
Department of Biochemistry and Molecular Biology,
University of Iowa, Cyclic nucleotide-gated (CNG) channels are
Iowa City, IA, USA responsible for carrying the inward flowing dark
e-mail: shivangi-inamdar@uiowa.edu; current (IDARK). IDARK is a depolarizing mixed Na+/
lankfordcolten@ufl.edu; sheila-baker@uiowa.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 269
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_39
270 S. M. Inamdar et al.

Ca2+ current that maintains PR depolarization (at are additionally linked to Leber congenital amau-
~ −35 to −40 mV). These channels are not prom- rosis (LCA), and Oligocone trichomacy [24].
inently gated by voltage but are held in the open Achromatopsia is diagnosed early in childhood
state when cGMP is bound [4, 5]. CNG channels with severe cone dysfunction with patients report-
in rods and cones have distinct biophysical prop- ing color vision problems, reduced visual acuity,
erties due to their composition. Rod CNG is a 3:1 and photophobia. Patients show loss of PR in the
assembly of A1 and B1 subunits. Cone CNG is foveal region and thinning of the macula [25].
composed of A3 and B3 subunits at a stoichiom- Studies using mouse models have shown that loss
etry that has been reported to be either 3:1 or 2:2 of CNGA3 disrupts the targeting of the cone opsin
[6–11]. to the OS and cones undergo apoptosis without
The levels of cGMP that gate CNG channels affecting rod function and viability [26, 27]. In
are controlled by the phototransduction pathway mice with loss of CNGB3, there is reduced cone
such that levels are high in the dark but drop pre- function with slow but progressive cone degener-
cipitously in response to light [12]. CNG chan- ation that is attributed to the expression of homo-
nels do not desensitize to cGMP binding allowing meric CNGA3 channels with altered gating
CNG channels to be continuously open in the properties [28]. Recombinant AAV vector-­
dark, a feature necessary for maintaining the dark mediated delivery of CNGA3 in CNGA3 KO
resting potential of PRs [13]. When CNG chan- mice has shown recovery of cone function and
nels close in response to light, the loss of the Na+ delayed cone death [29]. Similarly, AAV-­mediated
influx hyperpolarizes the PR membrane. Loss of delivery of CNGB3 in KO mice showed success-
the Ca2+ influx serves a more important role in ful and long-term restoration of cone function
regulation. Ca2+ entering the OS is extruded by [30]. Excitingly, clinical trials of AAV-­CNGB3
Na+/Ca2+, K+ exchangers that are physically cou- gene therapy for achromatopsia patients have
pled to CNG channels [14–16]. This extrusion completed two phases of testing and are showing
does not stop in the light so Ca2+ levels inside the promising results ([28, 31], clinicaltrials.gov).
OS decrease in the light which is read by calcium-­
binding proteins that regulate several compo-
nents of the phototransduction cascade including 3 Cav1.4 Channels Respond to
CNG channels themselves [17]. CNG-Regulated Membrane
Mutations in rod-specific CNGA1 and Potential
CNGB1 cause autosomal recessive retinitis pig-
mentosa (arRP) [4, 18, 19]. In arRP, rods are dys- Cav1.4 channels in the synapse respond to the
functional, so patients present as night blind. In membrane potential set by the light-regulated
the absence of either of the subunits, functional opening or closing of CNG channels to control
CNG channels fail to be expressed at the plasma communication to the inner retina. Cav1.4 chan-
membrane resulting in photoreceptor death [20– nels consist of three subunits, the pore-forming
22]. As rods die, secondary death of cones takes α1F, an intracellular β2 that provides an indirect
place further reducing the visual field and ulti- link between the channel and the synaptic ribbon
mately leading to complete blindness [18]. Gene complex, and an extracellular α2δ4 that is thought
replacement in CNGB1 knockout (KO) mice to be involved in transsynaptic adhesion [32–37].
using adeno-associated viral (AAV) vectors have The β2 and α2δ4 subunits also participate in traf-
been shown to be effective in restoring rod CNG ficking and stability of the channel and can modu-
and halting degeneration, but this approach has late the biophysical properties of the pore-forming
not yet been translated into clinical trials of a subunit [38]. Additional regulation of Cav1.4 is
therapy for adRP patients [18, 23]. provided by the expression of alternative splicing
Mutations in cone-specific CNGA3 and products or interaction with the calcium-binding
CNGB3 cause recessive achromatopsia or reces- protein, CaBP4 [39, 40]. Cav1.4 channels are
sive cone-rod dystrophy. Mutations in CNGB3 expressed in both rods and cones [3, 41–43].
Photoreceptor Ion Channels in Signaling and Disease 271

Synaptic vesicle fusion with the plasma mem- ST include – HCN1 and Kv2.1/Kv8.2. Of these
brane and release of glutamate is triggered by the channels, HCN1 is the best studied. The local-
binding of Ca2+ to synaptotagmin proteins. The ization of the channel is primarily IS, but it
synaptic ribbon complex ensures that a large num- also labels the soma, axon, and synapse [52–
ber of synaptic vesicles are docked and primed in 58]. HCN1 channels belong to the
close association with Cav1.4 channels which hyperpolarization-­activated cyclic nucleotide-
carry 95% of the current that brings Ca2+ into the gated (HCN) family of ion channels which are
synapse at the active zone (Ica) [3, 44]. Unlike most structurally similar to CNG channels. However,
neurons, PRs do not fire all-or-none-­action poten- HCN1 channels are primarily driven by volt-
tials but respond to light by graded decreases in age-gating which can be regulated by
glutamate release which allows for responses to cAMP. There is one family member insensitive
fine increases or decreases in light intensity [45]. to cAMP regulation and that is HCN1, which is
Two features of Cav1.4 that supports the unique the HCN family member expressed in both
needs of PR neurotransmitter release are that these rods and cones [59]. HCN channels are unique
channels activate at more negative potentials than in that they open in response to hyperpolariza-
other synaptic Cav channels so that they are open tion, [60]. The mechanism that allows these
in dark-adapted PRs. Second, Cav1.4 channels are channels to open at especially negative poten-
resistant to calcium-­dependent inhibition so can tials is unknown.
stay open longer [46]. HCN1 in PRs carries the hyperpolarization-­
Mutations in CACNA1F, encoding the α1F activated current (Ih) which is required to control
subunit of Cav1.4 are most frequently diagnosed response timing. As light-triggered closure of
as incomplete congenital stationary night blind- CNG channels drives the membrane potential
ness (CSNB2). CSNB2 is like complete CSNB, more and more negative HCN1 channels will
which results from mutations in the bipolar den- open and the influx of mixed Na+ and K+ ions
dritic signaling complex, in that patients present depolarize the membrane, thus acting as a reset,
with symptoms that include night blindness, pho- particularly following brighter flashes of light.
tophobia, color vision abnormalities, and Pharmacological experiments have shown the
decreased visual acuity [47]. However, CSNB2 is importance of this function with rod voltage
more variable [47, 48]. The most helpful diag- recovery being up to two-fold slower in the pres-
nostic feature of CSNB is the lack of a b-wave in ence of Ih blockers [52, 61, 62].
the scotopic ERG. In complete CSNB, the b-wave The role of HCN1 has also been described
is completely flat while in CSNB2 there is a small using ERG recording of KO mice. In response to
b-wave that is never large enough to cross the a brief flash, HCN1 knockout mice exhibit a pro-
baseline so is termed electronegative [47]. The longed rod-driven b-wave indicative of prolonged
stationary part of CSNB2 is often true although activation of neurons downstream from rods [57,
some CACNA1F mutations cause degeneration 63–66]. Interestingly, altered rod function in the
in the form of cone-rod dystrophy 3 [49, 50]. HCN1 knockout mice allows rods to continue
Further exemplifying the variability of Cav1.4-­ signaling even under photopic conditions when
associated disease is that mutations in the auxil- rods would normally be shut off. This excessive
iary α2δ4 subunit (CACNA2D4) cause retinal rod signaling ends up preventing the transmission
cone dystrophy 4 [51]. of cone signals [65, 67]. Strangely, we did not
observe any negative consequences to ablating
HCN1 specifically from cones [67].
4 HCN1 Channels Shape Light Understanding why cones express HCN1 is a
Responses question that needs further investigation.
HCN1 is widely expressed throughout the
Two channels residing in the IS that function in brain and plays a well-characterized role in regu-
shaping the visual responses initiated in the OS lating neuronal excitability [68–70]. As such it is
and communicate to the inner retina at the not surprising that people with HCN1 mutations
272 S. M. Inamdar et al.

have epilepsy. Sometimes the disease is severe that activates at more negative potentials and
resulting in childhood mortality, while other inactivates more slowly than Kv2.1 homotetra-
mutations are benign [71, 72]. People with HCN1 meric channels. The biophysical properties of the
mutations have not reported visual problems but hybrid channel match those of IKx recorded from
that does not necessarily undercut the important either amphibian or rodent PRs [61, 76, 77].
role HCN1 plays in the retina. Instead consider Second, there have been several recent knockout
that people with mutations causing very mild mouse studies confirming that loss of either
symptoms may have unrecognized visual dys- Kv2.1 or Kv8.2 causes phenotypes consistent
function and those with mutations that cause with loss of IKx, this will be discussed in more
early mortality would not have had an opportu- detail in the context of disease associated with
nity to notice and report visual abnormalities [71, this channel [79–82].
73, 74]. In animal models, HCN1 loss is insuffi- The primary function of IKx is to set the mem-
cient to drive retinal degeneration; however, brane resting potential [61]. In the dark, the
CNGB KO mice are a model for retinitis pigmen- inward flow of current through outer segment
tosa, and it was found that double KO of CNGB CNG channels is balanced by a standing out-
and HCN1 accelerated rod loss, likely by altered ward current originating from the inner seg-
calcium homeostasis and aberrant calpain activ- ment. Kv2.1/Kv8.2 accounts for 70–80% of this
ity [75]. Therefore, seemingly mild mutations in standing outward dark current [77, 81]. The
HCN1 should be considered as possible genetic constant flow of K+ ions out of the PR in the
modifiers in inherited retinal degeneration. dark is only possible because of the constant
activity of NKA which burns ATP to simultane-
ously transport two K+ ions into the cell and
5 Kv2.1/Kv8.2 Channels three Na+ ions out. Because of this imbalance in
total charge being transported across the mem-
Kv2.1/Kv8.2 is a heterotetrameric channel that is brane, NKA is electrogenic and accounts for the
found in the IS of both rod and cone PRs [76, 77]. remaining portion of the standing outward dark
The Kv2 family of delayed outwardly rectifying current [81]. Note, the abundance and constant
potassium channels consists of two members, activity of NKA in PRs contribute to these cells
Kv2.1 and Kv2.2, that form homomeric channels being often cited as the most metabolically
[78]. Kv2.1 channels are expressed in the rods demanding [83]. Upon light stimulation,
and cones of rodents and primates. Primate cones Kv2.1/Kv8.2 channels slowly inactivate which
additionally express Kv2.2 although the signifi- is important for increasing sensitivity to chang-
cance of that is currently unknown [76]. Kv2.1 ing illumination. As the PR further undergoes
homotetrameric channels activate just outside the hyperpolarization, Kv2.1/8.2 channels close and
normal operational range of PRs but can incorpo- HCN1 channels open to reset the membrane
rate a so-called “electrically silent” or KvS sub- back towards the resting potential [2].
unit that alters the activity of the channel [76, 77]. Mutations in KCNB1 (encoding Kv2.1) are
The KvS protein expressed in PRs is Kv8.2 associated with developmental and epileptic
which co-localizes with Kv2.1 [76]. Both Kv encephalopathy [84, 85] but vision defects have
subunits, and Na+/K+ ATPase (NKA), are present not been reported for these patients. On the other
in the apical portion of the IS plasma membrane, hand, mutations in KCNV2 (encoding Kv8.2) do
which is adjacent to the mitochondria-rich cause an autosomal recessive retinal degenerative
ellipsoid. disease called cone dystrophy with supernormal
There are two lines of evidence that heterotet- rod response (CDSRR) or KCNV2 retinopathy.
rameric Kv2.1/Kv8.2 channels carry IKx – one of Patients are usually diagnosed with CDSRR early
the principal currents that describe basic PR sig- in adolescence with the symptoms common to
naling. The first co-expression of Kv8.2 with cone dystrophies [86]. The characteristic feature
Kv2.1 in Xenopus oocytes can create a channel of CDSRR is the unusual electrical activity of the
Photoreceptor Ion Channels in Signaling and Disease 273

retina which is diagnosed primarily by the ampli- one study reported cone loss, but this was not
tude of the ERG b-wave. The amplitude of the replicated in a subsequent study [79, 82].
rod-driven b-wave under the dimmest illumina- However, rapid cone loss was observed in the lat-
tion is reduced but becomes supernormal as the ter study after crossing the Kv8.2 KO mice to a
light intensity increases, the amplitude of the strain with an all-cone retina [82]. This implies
cone-driven b-wave is consistently reduced. that both loss of Kv8.2 and an environmental
Recent mouse studies have provided models for stress, that is, loss of rods in mice, or perhaps
further study of this disease. highlight exposure in the case of the human mac-
The Kv8.2 KO mice have abnormal ERG ula, are needed to trigger cone death.
responses that phenocopy CDSRR [79, 80, 82]. While there are no current treatment options
Consistent with the loss of IKx and the need to available, the size of KCNV2 is amenable to
rely primarily on NKA to provide the outward packaging in AAV so even though it is a rare dis-
arm of the standing dark current, isolated PR ease, one expects gene replacement therapy is a
responses are reduced by 20%. In Kv2.1 KO viable treatment strategy.
mice a reduction in the extracellular K+ concen-
tration was documented and both Kv2.1 and
Kv8.2 KO mice have greatly reduced ERG 6 Future Directions
c-waves indicating the same is happening in the
Kv8.2 KO retina [79, 81]. A better understanding Here we have presented an overview of our cur-
of how potassium homeostasis is altered in rent state of knowledge regarding four ion chan-
CDSRR will require a closer examination of the nels that are integral to PR signaling. A pressing
expression levels and activity of K+ channels and need in this field that we did not touch on is
exchangers in the RPE and Muller glia since understanding how the activity of these channels
those two cells are essential to so many aspects of is influenced by other ion channels and exchang-
PR biology. ers. Such knowledge may be used to supplement
Loss of Kv8.2 should leave homotetrameric current gene replacement strategies to help
Kv2.1 channels, which normally function outside patients with vision loss due to alteration of ion
of the physiological range of the PRs. That means channel function.
Kv8.2 and Kv2.1 KO phenotypes should be the
same, which is what is mostly observed in ERG
(except for one report of young Kv2.1 KO mice, References
the rod-driven supernormal b-wave is unique to
Kv8.2 KO) and double Kv2.1/Kv8.2 KO mice 1. Pearring JN, Salinas RY, Baker SA, Arshavsky
VY. Protein sorting, targeting and trafficking in pho-
are similar to the single KOs [79–82]. For Kv2.1 toreceptor cells. Prog Retin Eye Res. 2013;36:24–51.
homotetrameric channels to compensate for the 2. Lankford CK, Laird JG, Inamdar SM, Baker
loss of Kv8.2, it is expected that the resting mem- SA. A comparison of the primary sensory neurons
brane potential would have to be more depolar- used in olfaction and vision. Front Cell Neurosci.
2020;14:595523.
ized than normal. This has been documented in 3. Van Hook MJ, Nawy S, Thoreson WB. Voltage- and
the Kv2.1 KO but not directly tested in the Kv8.2 calcium-gated ion channels of neurons in the verte-
KO [77, 81]. brate retina. Prog Retin Eye Res. 2019;72:100760.
The Kv8.2 KO mice do not adequately pheno- 4. Biel M, Michalakis S. Cyclic nucleotide-gated chan-
nels. Handb Exp Pharmacol. 2009;191:111–36.
copy CDSRR when it comes to neurodegenera- 5. Craven KB, Zagotta WN. CNG and HCN chan-
tion. In humans, progressive cell loss is restricted nels: two peas, one pod. Annu Rev Physiol.
to the macula which leads one to expect wide- 2006;68:375–401.
spread cone degeneration in mice even though 6. Zheng J, Trudeau MC, Zagotta WN. Rod cyclic
nucleotide-­gated channels have a stoichiometry of
the human macula contains a high density of rods three CNGA1 subunits and one CNGB1 subunit.
and cones surrounding the cone-pure fovea. Neuron. 2002;36(5):891–6.
Kv8.2 KO mice have a mild rod degeneration and
274 S. M. Inamdar et al.

7. Zhong H, Molday LL, Molday RS, Yau KW. The gated channel subunit CNGB1. J Neurosci.
heteromeric cyclic nucleotide-gated channel adopts a 2005;25(1):130–8.
3A:1B stoichiometry. Nature. 2002;420(6912):193–8. 23. Michalakis S, Koch S, Sothilingam V, Garcia Garrido
8. Weitz D, Ficek N, Kremmer E, Bauer PJ, Kaupp M, Tanimoto N, Schulze E, et al. Gene therapy restores
UB. Subunit stoichiometry of the CNG channel of rod vision and delays degeneration in the CNGB1(−/−)
photoreceptors. Neuron. 2002;36(5):881–9. mouse model of retinitis pigmentosa. Adv Exp Med
9. Peng C, Rich ED, Varnum MD. Subunit configuration Biol. 2014;801:733–9.
of heteromeric cone cyclic nucleotide-gated channels. 24. Sun W, Zhang Q. Diseases associated with muta-
Neuron. 2004;42(3):401–10. tions in CNGA3: genotype-phenotype correlation
10. Matveev AV, Quiambao AB, Browning Fitzgerald and diagnostic guideline. Prog Mol Biol Transl Sci.
J, Ding XQ. Native cone photoreceptor cyclic 2019;161:1–27.
nucleotide-­gated channel is a heterotetrameric com- 25. Remmer MH, Rastogi N, Ranka MP, Ceisler
plex comprising both CNGA3 and CNGB3: a study EJ. Achromatopsia: a review. Curr Opin Ophthalmol.
using the cone-dominant retina of Nrl−/− mice. J 2015;26(5):333–40.
Neurochem. 2008;106(5):2042–55. 26. Michalakis S, Geiger H, Haverkamp S, Hofmann F,
11. Ding XQ, Matveev A, Singh A, Komori N, Matsumoto Gerstner A, Biel M. Impaired opsin targeting and
H. Biochemical characterization of cone cyclic cone photoreceptor migration in the retina of mice
nucleotide-gated (CNG) channel using the infrared lacking the cyclic nucleotide-gated channel CNGA3.
fluorescence detection system. Adv Exp Med Biol. Invest Ophthalmol Vis Sci. 2005;46(4):1516–24.
2012;723:769–75. 27. Biel M, Seeliger M, Pfeifer A, Kohler K, Gerstner A,
12. Burns ME, Arshavsky VY. Beyond counting photons: Ludwig A, et al. Selective loss of cone function in mice
trials and trends in vertebrate visual transduction. lacking the cyclic nucleotide-gated channel CNG3.
Neuron. 2005;48(3):387–401. Proc Natl Acad Sci U S A. 1999;96(13):7553–7.
13. Kaupp UB, Seifert R. Cyclic nucleotide-gated ion 28. Michalakis S, Schon C, Becirovic E, Biel M. Gene
channels. Physiol Rev. 2002;82(3):769–824. therapy for achromatopsia. J Gene Med. 2017;19(3).
14. Bauer PJ, Drechsler M. Association of cyclic GMP-­ 29. Michalakis S, Muhlfriedel R, Tanimoto N,
gated channels and Na(+)-Ca(2+)-K+ exchangers in Krishnamoorthy V, Koch S, Fischer MD, et al.
bovine retinal rod outer segment plasma membranes. Restoration of cone vision in the CNGA3−/− mouse
J Physiol. 1992;451:109–31. model of congenital complete lack of cone photore-
15. Schwarzer A, Schauf H, Bauer PJ. Binding of the ceptor function. Mol Ther. 2010;18(12):2057–63.
cGMP-gated channel to the Na/Ca-K exchanger in rod 30. Carvalho LS, Xu J, Pearson RA, Smith AJ,
photoreceptors. J Biol Chem. 2000;275(18):13448–54. Bainbridge JW, Morris LM, et al. Long-term and
16. Poetsch A, Molday LL, Molday RS. The cGMP-­ age-dependent restoration of visual function in
gated channel and related glutamic acid-rich pro- a mouse model of CNGB3-associated achroma-
teins interact with peripherin-2 at the rim region of topsia following gene therapy. Hum Mol Genet.
rod photoreceptor disc membranes. J Biol Chem. 2011;20(16):3161–75.
2001;276(51):48009–16. 31. Koch M, Scheel C, Ma H, Yang F, Stadlmeier M,
17. Vinberg F, Chen J, Kefalov VJ. Regulation of calcium Gluck AF, et al. The cGMP-dependent protein kinase
homeostasis in the outer segments of rod and cone 2 contributes to cone photoreceptor degeneration in
photoreceptors. Prog Retin Eye Res. 2018;67:87–101. the Cnga3-deficient mouse model of achromatopsia.
18. Koch S, Sothilingam V, Garcia Garrido M, Tanimoto Int J Mol Sci. 2020;22(1).
N, Becirovic E, Koch F, et al. Gene therapy restores 32. Wang Y, Fehlhaber KE, Sarria I, Cao Y, Ingram
vision and delays degeneration in the CNGB1(−/−) NT, Guerrero-Given D, et al. The auxiliary calcium
mouse model of retinitis pigmentosa. Hum Mol channel subunit alpha2delta4 is required for axonal
Genet. 2012;21(20):4486–96. elaboration, synaptic transmission, and wiring of rod
19. Michalakis S, Becirovic E, Biel M. Retinal cyclic photoreceptors. Neuron. 2017;93(6):1359–74.e6.
nucleotide-gated channels: from pathophysiology to 33. Dembla E, Dembla M, Maxeiner S, Schmitz
therapy. Int J Mol Sci. 2018;19(3). F. Synaptic ribbons foster active zone stability and
20. Dryja TP, Finn JT, Peng YW, McGee TL, Berson EL, illumination-dependent active zone enrichment of
Yau KW. Mutations in the gene encoding the alpha RIM2 and Cav1.4 in photoreceptor synapses. Sci Rep.
subunit of the rod cGMP-gated channel in autosomal 2020;10(1):5957.
recessive retinitis pigmentosa. Proc Natl Acad Sci U 34. Hagiwara A, Kitahara Y, Grabner CP, Vogl C, Abe M,
S A. 1995;92(22):10177–81. Kitta R, et al. Cytomatrix proteins CAST and ELKS
21. Liu Y, Wang Y, Xiao Y, Li X, Ruan S, Luo X, et al. regulate retinal photoreceptor development and main-
Retinal degeneration in mice lacking the cyclic tenance. J Cell Biol. 2018;217(11):3993–4006.
nucleotide-gated channel subunit CNGA1. FASEB J. 35. Hibino H, Pironkova R, Onwumere O, Vologodskaia
2021;35(9):e21859. M, Hudspeth AJ, Lesage F. RIM binding pro-
22. Huttl S, Michalakis S, Seeliger M, Luo DG, Acar N, teins (RBPs) couple Rab3-interacting molecules
Geiger H, et al. Impaired channel targeting and retinal (RIMs) to voltage-gated Ca(2+) channels. Neuron.
degeneration in mice lacking the cyclic nucleotide-­ 2002;34(3):411–23.
Photoreceptor Ion Channels in Signaling and Disease 275

36. Lee A, Wang S, Williams B, Hagen J, Scheetz TE, somal recessive cone dystrophy. Am J Hum Genet.
Haeseleer F. Characterization of Cav1.4 complexes 2006;79(5):973–7.
(alpha11.4, beta2, and alpha2delta4) in HEK293T cells 52. Barrow AJ, Wu SM. Low-conductance HCN1
and in the retina. J Biol Chem. 2015;290(3):1505–21. ion channels augment the frequency response
37. Zabouri N, Haverkamp S. Calcium channel-­dependent of rod and cone photoreceptors. J Neurosci.
molecular maturation of photoreceptor synapses. 2009;29(18):5841–53.
PLoS One. 2013;8(5):e63853. 53. Fain GL, Quandt FN, Bastian BL, Gerschenfeld
38. Dolphin AC, Lee A. Presynaptic calcium channels: HM. Contribution of a caesium-sensitive conduc-
specialized control of synaptic neurotransmitter tance increase to the rod photoresponse. Nature.
release. Nat Rev Neurosci. 2020;21(4):213–29. 1978;272(5652):466–9.
39. Haeseleer F, Imanishi Y, Maeda T, Possin DE, Maeda 54. Fain GL, Sampath AP. Light responses of mammalian
A, Lee A, et al. Essential role of Ca2+−binding pro- cones. Pflugers Arch. 2021.
tein 4, a Cav1.4 channel regulator, in photoreceptor 55. Hestrin S. The properties and function of inward recti-
synaptic function. Nat Neurosci. 2004;7(10):1079–87. fication in rod photoreceptors of the tiger salamander.
40. Koschak A, Fernandez-Quintero ML, Heigl T, Ruzza J Physiol. 1987;390:319–33.
M, Seitter H, Zanetti L. Cav1.4 dysfunction and con- 56. Ingram NT, Sampath AP, Fain GL. Membrane con-
genital stationary night blindness type 2. Pflugers ductances of mouse cone photoreceptors. J Gen
Arch. 2021;473(9):1437–54. Physiol. 2020;152(3).
41. Bader CR, Bertrand D, Schwartz EA. Voltage-­ 57. Knop GC, Seeliger MW, Thiel F, Mataruga A,
activated and calcium-activated currents studied in Kaupp UB, Friedburg C, et al. Light responses in
solitary rod inner segments from the salamander ret- the mouse retina are prolonged upon targeted dele-
ina. J Physiol. 1982;331:253–84. tion of the HCN1 channel gene. Eur J Neurosci.
42. Yagi T, Macleish PR. Ionic conductances of mon- 2008;28(11):2221–30.
key solitary cone inner segments. J Neurophysiol. 58. Muller F, Scholten A, Ivanova E, Haverkamp
1994;71(2):656–65. S, Kremmer E, Kaupp UB. HCN channels are
43. Zamponi GW, Striessnig J, Koschak A, Dolphin expressed differentially in retinal bipolar cells and
AC. The physiology, pathology, and pharmacology of concentrated at synaptic terminals. Eur J Neurosci.
voltage-gated calcium channels and their future thera- 2003;17(10):2084–96.
peutic potential. Pharmacol Rev. 2015;67(4):821–70. 59. Zong X, Krause S, Chen CC, Kruger J, Gruner C,
44. Xu JW, Slaughter MM. Large-conductance calcium-­ Cao-­Ehlker X, et al. Regulation of hyperpolarization-­
activated potassium channels facilitate transmit- activated cyclic nucleotide-gated (HCN) channel activ-
ter release in salamander rod synapse. J Neurosci. ity by cCMP. J Biol Chem. 2012;287(32):26506–12.
2005;25(33):7660–8. 60. Lee CH, MacKinnon R. Structures of the human
45. Pangrsic T, Singer JH, Koschak A. Voltage-gated HCN1 hyperpolarization-activated channel. Cell.
calcium channels: key players in sensory cod- 2017;168(1–2):111–20 e11.
ing in the retina and the inner ear. Physiol Rev. 61. Beech DJ, Barnes S. Characterization of a voltage-­
2018;98(4):2063–96. gated K+ channel that accelerates the rod response to
46. Joiner ML, Lee A. Voltage-gated Cav1 channels in dim light. Neuron. 1989;3(5):573–81.
disorders of vision and hearing. Curr Mol Pharmacol. 62. Barnes S. After transduction: response shaping
2015;8(2):143–8. and control of transmission by ion channels of
47. Waldner DM, Bech-Hansen NT, Stell WK. Channeling the photoreceptor inner segments. Neuroscience.
vision: CaV1.4-a critical link in retinal signal trans- 1994;58(3):447–59.
mission. Biomed Res Int. 2018;2018:7272630. 63. Della Santina L, Piano I, Cangiano L, Caputo A,
48. Stone EM, Andorf JL, Whitmore SS, DeLuca AP, Ludwig A, Cervetto L, et al. Processing of retinal sig-
Giacalone JC, Streb LM, et al. Clinically focused nals in normal and HCN deficient mice. PLoS One.
molecular investigation of 1000 consecutive fami- 2012;7(1):e29812.
lies with inherited retinal disease. Ophthalmology. 64. Gargini C, Demontis GC, Bisti S, Cervetto L. Effects
2017;124(9):1314–31. of blocking the hyperpolarization-activated cur-
49. Mantyjarvi M, Nurmenniemi P, Partanen J, Myohanen rent (Ih) on the cat electroretinogram. Vis Res.
T, Peippo M, Alitalo T. Clinical features and a fol- 1999;39(10):1767–74.
low-­up study in a family with X-linked progres- 65. Seeliger MW, Brombas A, Weiler R, Humphries
sive cone-rod dystrophy. Acta Ophthalmol Scand. P, Knop G, Tanimoto N, et al. Modulation of rod
2001;79(4):359–65. photoreceptor output by HCN1 channels is essen-
­
50. Jalkanen R, Mantyjarvi M, Tobias R, Isosomppi J, tial for regular mesopic cone vision. Nat Commun.
Sankila EM, Alitalo T, et al. X linked cone-rod dys- 2011;2:532.
trophy, CORDX3, is caused by a mutation in the 66. Sothilingam V, Michalakis S, Garcia Garrido M,
CACNA1F gene. J Med Genet. 2006;43(8):699–704. Biel M, Tanimoto N, Seeliger MW. HCN1 chan-
51. Wycisk KA, Zeitz C, Feil S, Wittmer M, Forster nels enhance rod system responsivity in the retina
U, Neidhardt J, et al. Mutation in the auxiliary under conditions of light exposure. PLoS One.
calcium-­ channel subunit CACNA2D4 causes auto- 2016;11(1):e0147728.
276 S. M. Inamdar et al.

67. Lankford C, Umino Y, Poria D, Kefalov V, Solessio E, and the retinal subunit kv8.2 in Xenopus oocytes. J
Baker S. Cone-driven retinal responses are shaped by Neurophysiol. 2007;98(3):1213–22.
rod but not cone HCN1. bioRxiv; 2021. 78. Bocksteins E, Snyders DJ. Electrically silent Kv sub-
68. Moosmang S, Stieber J, Zong X, Biel M, Hofmann F, units: their molecular and functional characteristics.
Ludwig A. Cellular expression and functional char- Physiology (Bethesda). 2012;27(2):73–84.
acterization of four hyperpolarization-activated pace- 79. Hart NS, Mountford JK, Voigt V, Fuller-Carter
maker channels in cardiac and neuronal tissues. Eur J P, Barth M, Nerbonne JM, et al. The role of the
Biochem. 2001;268(6):1646–52. voltage-­gated potassium channel proteins Kv8.2 and
69. Notomi T, Shigemoto R. Immunohistochemical local- Kv2.1 in vision and retinal disease: insights from the
ization of Ih channel subunits, HCN1-4, in the rat study of mouse gene knock-out mutations. eNeuro.
brain. J Comp Neurol. 2004;471(3):241–76. 2019;6(1).
70. Shah MM. Cortical HCN channels: function, traffick- 80. Jiang X, Rashwan R, Voigt V, Nerbonne J, Hunt
ing and plasticity. J Physiol. 2014;592(13):2711–9. DM, Carvalho LS. Molecular, cellular and functional
71. Marini C, Porro A, Rastetter A, Dalle C, Rivolta I, changes in the retinas of young adult mice lacking the
Bauer D, et al. HCN1 mutation spectrum: from neo- voltage-gated K(+) channel subunits Kv8.2 and K2.1.
natal epileptic encephalopathy to benign generalized Int J Mol Sci. 2021;22(9).
epilepsy and beyond. Brain. 2018;141(11):3160–78. 81. Fortenbach C, Peinado Allina G, Shores CM, Karlen
72. Nava C, Dalle C, Rastetter A, Striano P, de Kovel CG, SJ, Miller EB, Bishop H, et al. Loss of the K+ chan-
Nabbout R, et al. De novo mutations in HCN1 cause nel Kv2.1 greatly reduces outward dark current and
early infantile epileptic encephalopathy. Nat Genet. causes ionic dysregulation and degeneration in rod
2014;46(6):640–5. photoreceptors. J Gen Physiol. 2021;153(2).
73. Bonzanni M, DiFrancesco JC, Milanesi R, 82. Inamdar SM, Lankford CK, Poria D, Laird JG,
Campostrini G, Castellotti B, Bucchi A, et al. A novel Solessio E, Kefalov VJ, et al. Differential impact of
de novo HCN1 loss-of-function mutation in genetic Kv8.2 loss on rod and cone signaling and degenera-
generalized epilepsy causing increased neuronal tion. Hum Mol Genet. 2021;
excitability. Neurobiol Dis. 2018;118:55–63. 83. Viegas FO, Neuhauss SCF. A metabolic landscape for
74. Tang B, Sander T, Craven KB, Hempelmann A, maintaining retina integrity and function. Front Mol
Escayg A. Mutation analysis of the hyperpolarization-­ Neurosci. 2021;14:656000.
activated cyclic nucleotide-gated channels HCN1 and 84. Shieh CC, Coghlan M, Sullivan JP, Gopalakrishnan
HCN2 in idiopathic generalized epilepsy. Neurobiol M. Potassium channels: molecular defects, dis-
Dis. 2008;29(1):59–70. eases, and therapeutic opportunities. Pharmacol Rev.
75. Schon C, Asteriti S, Koch S, Sothilingam V, Garcia 2000;52(4):557–94.
Garrido M, Tanimoto N, et al. Loss of HCN1 85. Speca DJ, Ogata G, Mandikian D, Bishop HI, Wiler
enhances disease progression in mouse models of SW, Eum K, et al. Deletion of the Kv2.1 delayed
CNG channel-­linked retinitis pigmentosa and achro- rectifier potassium channel leads to neuronal and
matopsia. Hum Mol Genet. 2016;25(6):1165–75. behavioral hyperexcitability. Genes Brain Behav.
76. Gayet-Primo J, Yaeger DB, Khanjian RA, Puthussery 2014;13(4):394–408.
T. Heteromeric KV2/KV8.2 channels mediate delayed 86. Rashwan R, Hunt DM, Carvalho LS. The role of
rectifier potassium currents in primate photorecep- voltage-gated ion channels in visual function and
tors. J Neurosci. 2018;38(14):3414–27. disease in mammalian photoreceptors. Pflugers Arch.
77. Czirjak G, Toth ZE, Enyedi P. Characterization of 2021;473(9):1455–68.
the heteromeric potassium channel formed by kv2.1
The Role of Peripherin-2/ROM1
Complexes in Photoreceptor Outer
Segment Disc Morphogenesis

Tylor R. Lewis, Muayyad R. Al-Ubaidi,


Muna I. Naash, and Vadim Y. Arshavsky

Abstract loop mediates well-established intermolecular


interactions of peripherin-2 molecules among
The light-sensitive outer segment organelle of themselves and a homologous protein ROM1.
photoreceptor cells contains a stack of hun- These interactions lead to the formation of
dreds of flat, disc-shaped membranes called large, highly ordered oligomers. In this chap-
discs. The rims of these discs contain a ter, we discuss the supramolecular organiza-
photoreceptor-­specific tetraspanin protein tion of peripherin-2/ROM1 complexes and
peripherin-2 (also known as rds or PRPH2). their contribution to the process of outer seg-
Mutations in the PRPH2 gene lead to a wide ment disc morphogenesis and enclosure.
variety of inherited retinal degenerations in
humans. The vast majority of these mutations Keywords
occur within a large, intradiscal loop of
peripherin-2, known as the D2 loop. The D2 Peripherin-2 · Photoreceptor · Outer Segment
· Disc · PRPH2 · RDS · ROM1 · Disc Rim ·
Disc Enclosure · Retina
T. R. Lewis (*)
Department of Ophthalmology, Duke University
Medical Center,
Durham, NC, USA 1 Peripherin-2 Is a Tetraspanin
e-mail: tylor.lewis@duke.edu Protein Whose Mutations
M. R. Al-Ubaidi · M. I. Naash Cause Retinal Degeneration
Department of Biomedical Engineering, College of
Engineering, University of Houston, Houston, TX, 1.1 Molecular Structure
USA
of Peripherin-2 and ROM1
College of Optometry, University of Houston,
Houston, TX, USA
e-mail: malubaid@central.uh.edu; Peripherin-2 is a glycosylated photoreceptor-­
mnaash@central.uh.edu specific tetraspanin protein [1, 2]. Both N- and
V. Y. Arshavsky C-termini of peripherin-2 reside on the cytoplas-
Department of Ophthalmology, Duke University mic face of the membrane, while there are two
Medical Center, intradiscal loops (termed D1 and D2 loops) that
Durham, NC, USA face the interior of the outer segment discs [1, 2].
Department of Pharmacology and Cancer Biology, The conserved site for glycosylation is in the
Duke University Medical Center, Durham, NC, USA intradiscal D2 loop [2]. Notably, another highly
e-mail: vadim.arshavsky@duke.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 277
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_40
278 T. R. Lewis et al.

homologous tetraspanin, ROM1, also localizes to lecular interactions of peripherin-2 are critical
outer segment disc rims [3]. ROM1 shares 35% to its biological function.
conservation at the amino acid level with periph-
erin-­2 and has a similar membrane topology [3],
but is not glycosylated [4]. 1.3 Supramolecular Organization
of Peripherin-2/ROM1

1.2 Mutations in Peripherin-2 Are As illustrated in Fig. 1, peripherin-2 can interact


Associated with Retinal non-covalently with other peripherin-2 molecules
Degeneration and with ROM1 to form either homomeric or het-
eromeric tetramers that are considered the “core”
Since the initial identification of PRPH2 muta- units of peripherin-2/ROM1 supramolecular
tions in patients with autosomal dominant reti- organization [3, 17–19]. These non-covalent tet-
nitis pigmentosa in 1991 [5], over 200 PRPH2 ramers can interact covalently among themselves
mutations have been identified that lead to a het- through the formation of disulfide bonds at the
erogenous set of inherited retinal diseases C150 residue of peripherin-2 to form large oligo-
including retinitis pigmentosa, cone-rod dystro- meric complexes [14, 20–22]. In vitro analysis
phy, and macular dystrophies [6]. While most of showed that ROM1 was absent from the large
these PRPH2 mutations lead to disease in an oligomeric complexes of peripherin-2, although
autosomal dominant fashion, autosomal reces- it was found in smaller, intermediate-sized oligo-
sive cases have also been reported [7, 8], as well mers [20]. Whether the latter is also the case
as mutations that combine with mutations in in vivo remains to be determined.
ROM1 to cause digenic retinitis pigmentosa [9,
10]. Yet, it is unclear whether mutations in
ROM1 alone are pathogenic [11–13]. 2 The Function of Peripherin-2
Interestingly, the vast majority of disease-asso-
ciated mutations in PRPH2 exist in the intradis- Peripherin-2 has two critical functions, each per-
cal D2 loop [6], which is involved in the formed by different parts of its molecule. The
well-established interactions of peripherin-2 first, performed by the C-terminus, is the reten-
with either ROM1 or other peripherin-­2 mole- tion of membranes at the photoreceptor cilium to
cules [14–16]. This suggests that the intermo- enable the formation of outer segment discs. The

Fig. 1 Supramolecular organization of peripherin-2 and disulfide bonds can lead to the formation of large, high-­
ROM1. Peripherin-2/ROM1 can form homo- and hetero-­ order oligomers of peripherin-2 that reportedly exclude
tetramers through non-covalent interactions. Covalent ROM1, at least in vitro
The Role of Peripherin-2/ROM1 Complexes in Photoreceptor Outer Segment Disc Morphogenesis 279

second, performed by the tetraspanin core, is the membrane curvature when reconstituted into
formation of the highly curved disc rim. liposomes [32] and leads to the formation of
highly curved intracellular membranes when
expressed in mammalian cells [27, 33, 34]. These
2.1 Discs Are Formed through properties of peripherin-2 are conveyed by its tet-
Peripherin-2-Dependent raspanin core, which is sufficient for generating
Suppression of Ciliary membrane curvature in both living animals and
Ectosome Release cell culture [27]. Other experiments showed that
the replacement of the tetraspanin core of periph-
Loss of peripherin-2 in the rds mouse completely erin-­2 with that of ROM1 in mice affects the cur-
prevents outer segment formation and causes the vature of disc rims [35].
subretinal space to be packed with extracellular These studies eventually led to a model in
vesicles [23, 24] containing rhodopsin [25, 26]. which long oligomeric chains of peripherin-2 and
Recently, it has been shown that these vesicles ROM1 run around the entire circumference of an
are released from the photoreceptor cilium [27] enclosed disc to generate and maintain the shape
and, therefore, are ciliary ectosomes, i.e., extra- of its rim. It was first suggested that there are two
cellular vesicles ubiquitously released by many of these long oligomeric chains surrounding each
types of primary cilia [28]. The function of disc [33], but a recent study utilizing cryo-­
peripherin-2 is to prevent the release of ecto- electron tomography revised this model by dem-
somes from the photoreceptor cilium, thereby onstrating that the disc rim is fortified by three
allowing the retention of these membranes and parallel interconnected oligomeric chains of
their transformation into outer segment discs peripherin-2/ROM1 [36], as illustrated in Fig. 2.
[27]. This function is performed by the C-terminus In support of the need for disulfide bonds
of peripherin-2 [27]. among individual tetramers to form long oligo-
meric chains of peripherin-2/ROM1, the inability
to form these disulfide bonds by the C150S
2.2 The Role of Peripherin-2 peripherin-2 mutant in a knockin mouse yielded
in Disc Rim Formation highly dysmorphic outer segment ultrastructure
and Enclosure [22]. A subsequent study revealed that even
minor defects in the ability of peripherin-2 to
The second function of peripherin-2 is to fortify oligomerize lead to defects in the fidelity of disc
the rims of mature photoreceptor discs separated enclosure [37]. Yet, surprisingly, the C150S
from the outer segment plasma membrane. This knockin mouse still exhibited some degree of
separation occurs during a complex membrane disc rim formation and enclosure, despite the
remodeling process called disc enclosure [29]. grossly aberrant outer segment structure [38].
Given that peripherin-2 redistributes along the Most likely, this is explained by the intrinsic abil-
disc rim during the process of disc enclosure [30, ity of peripherin-2/ROM1 oligomers to non-­
31], it seems likely that peripherin-2 plays an covalently interact, although not nearly as
active role driving the disc enclosure process. efficiently as when reinforced by disulfide
Consistent with this notion, peripherin-2 induces bridges.

Fig. 2 Model of peripherin-2 supramolecular organization at the disc rim. Three interconnected chains of peripherin-2
tetramers run parallel along the circumference of the disc rim
280 T. R. Lewis et al.

3 Concluding Remarks 4. Moritz OL, Molday RS. Molecular cloning, mem-


brane topology, and localization of bovine rom-1 in
and Future Avenues of Study rod and cone photoreceptor cells. Invest Ophthalmol
Vis Sci. 1996;37(2):352–62.
In this short chapter, we briefly highlighted the 5. Kajiwara K, Hahn LB, Mukai S, Travis GH, Berson
involvement of peripherin-2/ROM1 oligomeriza- EL, Dryja TP. Mutations in the human retinal degen-
eration slow gene in autosomal dominant retinitis pig-
tion in outer segment disc morphogenesis (see mentosa. Nature. 1991;354(6353):480–3.
other recent reviews for more details about periph- 6. Peeters M, Khan M, Rooijakkers A, Mulders T, Haer-­
erin-2/ROM1 [39, 40] or disc morphogenesis Wigman L, Boon CJF, et al. PRPH2 mutation update:
[29]). Yet, a lot remains unknown about these pro- in silico assessment of 245 reported and 7 novel vari-
ants in patients with retinal disease. Hum Mutat. 2021.
teins. For example, human mutations in the 7. Wang X, Wang H, Sun V, Tuan HF, Keser V, Wang
PRPH2 gene lead to a wide variety of retinal K, et al. Comprehensive molecular diagnosis of 179
degenerations. What is the basis for such a diver- Leber congenital amaurosis and juvenile retinitis pig-
sity of disease, from retinitis pigmentosa to cone-­ mentosa patients by targeted next generation sequenc-
ing. J Med Genet. 2013;50(10):674–88.
rod dystrophy, as well as several macular 8. Khan AO, Al Rashaed S, Neuhaus C, Bergmann C,
dystrophies? What is the exact contribution of Bolz HJ. Peripherin mutations cause a distinct form of
ROM1 to outer segment development and mainte- recessive Leber congenital amaurosis and dominant
nance? Can mutations in the ROM1 gene alone phenotypes in asymptomatic parents heterozygous for
the mutation. Br J Ophthalmol. 2016;100(2):209–15.
underlie retinal disease or do they simply modu- 9. Kajiwara K, Berson EL, Dryja TP. Digenic reti-
late PRPH2 mutations? Lastly, what is the precise nitis pigmentosa due to mutations at the unlinked
state of peripherin-2/ROM1 supramolecular orga- peripherin/RDS and ROM1 loci. Science.
nization that occurs in vivo in the disc rim and how 1994;264(5165):1604–8.
10. Dryja TP, Hahn LB, Kajiwara K, Berson
exactly does peripherin-2/ROM1 oligomerization EL. Dominant and digenic mutations in the periph-
regulate the dynamics of disc rim assembly and erin/RDS and ROM1 genes in retinitis pigmentosa.
disc enclosure? Further research into each of these Invest Ophthalmol Vis Sci. 1997;38(10):1972–82.
areas is warranted for a complete understanding of 11. Bascom RA, Liu L, Heckenlively JR, Stone EM,
McInnes RR. Mutation analysis of the ROM1
the function of ­peripherin-­2 and ROM1 and their gene in retinitis pigmentosa. Hum Mol Genet.
involvement in human visual disease. 1995;4(10):1895–902.
12. Sakuma H, Inana G, Murakami A, Yajima T, Weleber
Acknowledgments This work was supported by RG, Murphey WH, et al. A heterozygous putative null
National Institutes of Health Grants EY012859, mutation in ROM1 without a mutation in peripherin/
EY030451, and EY005722 to VYA, Grant EY010609 to RDS in a family with retinitis pigmentosa. Genomics.
MIN and MRA-U, and Grant EY033763 to TRL and 1995;27(2):384–6.
Research to Prevent Blindness Inc. Unrestricted Award 13. Reig C, Martinez-Gimeno M, Carballo M. A het-
(Duke University). erozygous novel C253Y mutation in the highly con-
served cysteine residues of ROM1 gene is the cause of
retinitis pigmentosa in a Spanish family? Hum Mutat.
2000;16(3):278.
References 14. Goldberg AF, Loewen CJ, Molday RS. Cysteine resi-
dues of photoreceptor peripherin/rds: role in subunit
1. Connell GJ, Molday RS. Molecular cloning, primary assembly and autosomal dominant retinitis pigmen-
structure, and orientation of the vertebrate photore- tosa. Biochemistry. 1998;37(2):680–5.
ceptor cell protein peripherin in the rod outer segment 15. Kedzierski W, Weng J, Travis GH. Analysis of the
disk membrane. Biochemistry. 1990;29(19):4691–8. rds/peripherin.rom1 complex in transgenic photore-
2. Travis GH, Sutcliffe JG, Bok D. The retinal degen- ceptors that express a chimeric protein. J Biol Chem.
eration slow (rds) gene product is a photoreceptor 1999;274(41):29181–7.
disc membrane-associated glycoprotein. Neuron. 16. Ding XQ, Stricker HM, Naash MI. Role of the second
1991;6(1):61–70. intradiscal loop of peripherin/rds in homo and hetero
3. Bascom RA, Manara S, Collins L, Molday RS, associations. Biochemistry. 2005;44(12):4897–904.
Kalnins VI, McInnes RR. Cloning of the cDNA for a 17. Goldberg AF, Molday RS. Subunit composition of
novel photoreceptor membrane protein (rom-1) iden- the peripherin/rds-rom-1 disk rim complex from
tifies a disk rim protein family implicated in human rod photoreceptors: hydrodynamic evidence for
retinopathies. Neuron. 1992;8(6):1171–84. a tetrameric quaternary structure. Biochemistry.
1996;35(19):6144–9.
The Role of Peripherin-2/ROM1 Complexes in Photoreceptor Outer Segment Disc Morphogenesis 281

18. Goldberg AF, Moritz OL, Molday RS. Heterologous 30. Arikawa K, Molday LL, Molday RS, Williams
expression of photoreceptor peripherin/rds and DS. Localization of peripherin/rds in the disk mem-
Rom-1 in COS-1 cells: assembly, interactions, and branes of cone and rod photoreceptors: relationship to
localization of multisubunit complexes. Biochemistry. disk membrane morphogenesis and retinal degenera-
1995;34(43):14213–9. tion. J Cell Biol. 1992;116(3):659–67.
19. Goldberg AF, Molday RS. Defective subunit assem- 31. Ding JD, Salinas RY, Arshavsky VY. Discs of
bly underlies a digenic form of retinitis pigmentosa mammalian rod photoreceptors form through the
linked to mutations in peripherin/rds and rom-1. Proc membrane evagination mechanism. J Cell Biol.
Natl Acad Sci U S A. 1996;93(24):13726–30. 2015;211(3):495–502.
20. Loewen CJ, Molday RS. Disulfide-mediated oligomer- 32. Kevany BM, Tsybovsky Y, Campuzano ID, Schnier
ization of Peripherin/Rds and Rom-1 in photoreceptor PD, Engel A, Palczewski K. Structural and func-
disk membranes. Implications for p­ hotoreceptor outer tional analysis of the native peripherin-ROM1 com-
segment morphogenesis and degeneration. J Biol plex isolated from photoreceptor cells. J Biol Chem.
Chem. 2000;275(8):5370–8. 2013;288(51):36272–84.
21. Chakraborty D, Ding XQ, Conley SM, Fliesler SJ, 33. Milstein ML, Cavanaugh BL, Roussey NM, Volland S,
Naash MI. Differential requirements for retinal Williams DS, Goldberg AFX. Multistep peripherin-2/
degeneration slow intermolecular disulfide-linked rds self-assembly drives membrane curvature for outer
oligomerization in rods versus cones. Hum Mol segment disk architecture and photoreceptor viability.
Genet. 2009;18(5):797–808. Proc Natl Acad Sci U S A. 2020;117(8):4400–10.
22. Zulliger R, Conley SM, Mwoyosvi ML, Al-Ubaidi 34. Milstein ML, Kimler VA, Ghatak C, Ladokhin AS,
MR, Naash MI. Oligomerization of Prph2 and Rom1 Goldberg AFX. An inducible amphipathic helix
is essential for photoreceptor outer segment forma- within the intrinsically disordered C terminus can par-
tion. Hum Mol Genet. 2018;27(20):3507–18. ticipate in membrane curvature generation by periph-
23. Cohen AI. Some cytological and initial biochemical erin-­2/rds. J Biol Chem. 2017;292(19):7850–65.
observations on photoreceptors in retinas of rds mice. 35. Conley SM, Stuck MW, Watson JN, Zulliger R, Burnett
Invest Ophthalmol Vis Sci. 1983;24(7):832–43. JL, Naash MI. Prph2 initiates outer segment morpho-
24. Jansen HG, Sanyal S. Development and degeneration genesis but maturation requires Prph2/Rom1 oligo-
of retina in rds mutant mice: electron microscopy. J merization. Hum Mol Genet. 2019;28(3):459–75.
Comp Neurol. 1984;224(1):71–84. 36. Pöge M, Mahamid J, Imanishi SS, Plitzko JM,
25. Nir I, Papermaster DS. Immunocytochemical localiza- Palczewski K, Baumeister W. Determinants shaping
tion of opsin in the inner segment and ciliary plasma the nanoscale architecture of the mouse rod outer seg-
membrane of photoreceptors in retinas of rds mutant ment. bioRxiv. 2021:2021.08.18.456753.
mice. Invest Ophthalmol Vis Sci. 1986;27(5):836–40. 37. Lewis TR, Makia MS, Castillo CM, Al-Ubaidi MR,
26. Usukura J, Bok D. Changes in the localization and Naash MI, Arshavsky VY. Photoreceptor disc enclo-
content of opsin during retinal development in the rds sure is tightly controlled by peripherin-2 oligomeriza-
mutant mouse: immunocytochemistry and immunoas- tion. J Neurosci. 2021;41(16):3588–96.
say. Exp Eye Res. 1987;45(4):501–15. 38. Lewis TR, Makia MS, Kakakhel M, Al-Ubaidi MR,
27. Salinas RY, Pearring JN, Ding JD, Spencer WJ, Hao Arshavsky VY, Naash MI. Photoreceptor disc enclo-
Y, Arshavsky VY. Photoreceptor discs form through sure occurs in the absence of normal peripherin-2/rds
peripherin-dependent suppression of ciliary ectosome oligomerization. Front Cell Neurosci. 2020;14:92.
release. J Cell Biol. 2017;216(5):1489–99. 39. Stuck MW, Conley SM, Naash MI. PRPH2/RDS and
28. Wood CR, Huang K, Diener DR, Rosenbaum JL. The ROM-1: historical context, current views and future
cilium secretes bioactive ectosomes. Curr Biol. considerations. Prog Retin Eye Res. 2016;52:47–63.
2013;23(10):906–11. 40. Tebbe L, Kakakhel M, Makia MS, Al-Ubaidi MR,
29. Spencer WJ, Lewis TR, Pearring JN, Arshavsky Naash MI. The interplay between peripherin 2 com-
VY. Photoreceptor discs: built like Ectosomes. Trends plex formation and degenerative retinal diseases. Cell.
Cell Biol. 2020;30(11):904–15. 2020;9(3).
Human Mutations in Arl3, a Small
GTPase Involved in Lipidated
Cargo Delivery to the Cilia, Cause
Retinal Dystrophy

Amanda M. Travis and Jillian N. Pearring

Abstract 1 Arl3 Unloads Lipidated


Cargo onto Ciliary
Photoreceptors are highly polarized sensory Membranes
neurons. Precise localization of signaling mol-
ecules within the ciliary outer segment is criti- Arl3 is a member of the ADP-ribosylation fac-
cal for photoreceptor function and viability. tor (Arf) family of small GTPases that are
The small GTPase Arl3 plays a particularly important for membrane trafficking [3]. Arl3
important role in photoreceptors as it regulates is ubiquitously expressed in ciliated cells
outer segment enrichment of lipidated pro- where it regulates the ciliary enrichment of
teins essential for the visual response: lipidated proteins. The GDI-like chaperones
transducin-α, transducin-γ, PDEα, PDE β, and PDEδ [9] and UNC119A/B [23] are Arl3
Grk1. Recently, mutations in Arl3 have been effectors that bind to prenylated and myris-
identified in human patients with nonsyn- toylated proteins, respectively, by hiding the
dromic autosomal recessive and dominant lipid moiety within an intramolecular cavity
inherited retinal degenerations as well as syn- that solubilizes the lipid cargo and enables its
dromic Joubert syndrome including retinal free diffusion (Fig. 1a). The lipid cargo is
dystrophy. eventually released in the cilium through
interactions with active GTP-bound Arl3 [12,
Keywords 13]. Arl3 is specifically activated in the cilium
by its guanine exchange factor (GEF) Arl13B
Arl3 · Photoreceptors · Arl13B · RP2 · Retinal and is stabilized in a GTP-bound state by its
dystrophy effector, BART [4, 7] (Fig. 1a). Hydrolysis of
GTP to GDP in Arl3 is facilitated by its
GTPase-­ activating protein (GAP) RP2 [20,
A. M. Travis
Department of Ophthalmology and Visual Science, 21]. RP2 is a lipidated protein frequently
University of Michigan, Ann Arbor, MI, USA mutated in X-linked retinitis pigmentosa
e-mail: amtr@umich.edu patients [17] and found enriched at the base of
J. N. Pearring (*) the cilium although also present at other loca-
Department of Ophthalmology and Visual Science, tions in the cell [5, 10]. Inactivation of Arl3
University of Michigan, Ann Arbor, MI, USA causes it to dissociate from its chaperone
Department of Cell and Developmental Biology, effectors, which are freed to start another
University of Michigan, Ann Arbor, MI, USA cycle by binding lipidated cargo.
e-mail: pearring@umich.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 283
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_41
284 A. M. Travis and J. N. Pearring

Fig. 1 Arl3 is important for enrichment of lipidated pro- membranes in photoreceptors). Arl3 is inactivated by the
teins to the ciliary outer segment of rod photoreceptors. GAP, RP2, at inner segment membranes. (b) Cartoon
(a)Model showing Arl3’s role in lipidated protein trans- schematic of mouse photoreceptor with specific compart-
port. GDI-like chaperone proteins sequester the lipid moi- ments labeled to the left. The light-sensitive outer segment
ety of cargo proteins allowing for cytoplasmic diffusion. is a modified primary cilium and site for lipidated protein
In the box, the Arl3 GTPase cycle is highlighted. Arl3 is delivery by Arl3. In vivo electroporation of FLAG-tagged
activated by its guanine nucleotide exchange factor human Arl3 in mouse rod photoreceptors shows Arl3 is
(GEF), Arl13B, in the cilium. Active GTP-bound Arl3 is localized throughout the photoreceptor cell body and
stabilized by BART so it can then bind chaperones, releas- outer segment
ing lipidated cargo on ciliary membranes (outer segment

Arl3 is expressed throughout all layers of the 2 Mouse Models of Arl3


retina [22], but studies have focused on its
­localization and function in photoreceptor cells The importance of Arl3 for ciliary function
as their light-sensitive outer segment compart- in vivo is demonstrated clearly by mouse models
ment is a modified primary cilium. Endogenous of Arl3 knockout or disfunction. Mice with no
Arl3 antibody staining shows that Arl3 is local- Arl3 expression due to a gene trap within the first
ized throughout the photoreceptor layer from the intron of Arl3 are born at non-Mendelian ratios
synapse to the outer segment [22]. This staining and phenocopy the loss of other important ciliary
pattern was confirmed in a transgenic mouse proteins, specifically cysts in the kidney, liver,
expressing HA-tagged wild-type mouse Arl3 and pancreas commonly found in models of the
[22] as well as in our own experiments express- ciliopathy polycystic kidney disease [16]. In the
ing a FLAG-tagged human Arl3 in rod photore- retina, the absence of Arl3 strongly affects rod
ceptors by in vivo electroporation [24] (Fig. 1b). photoreceptors, which are unable to form outer
A previous publication shows EGFP-tagged Arl3 segments. Cone photoreceptors do form outer
localized to the inner segment and cell body of segments, but they are abnormally short at P13.
rods [8]; however, this staining pattern is likely Photoreceptor cell death is observed as early as
due to steric volume exclusion from the P13 in these mice, but the precise course of pho-
membrane-­dense outer segment as a result of the toreceptor cell loss is unknown since the Arl3−/−
addition of the comparatively large fluorescent mice fail to thrive and die by 3 weeks of age.
tag [14].
Human Mutations in Arl3, a Small GTPase Involved in Lipidated Cargo Delivery to the Cilia, Cause Retinal… 285

Arl3’s function in the retina was further stud- segment proteins spilling into the inner segment
ied in two different conditional Arl3 floxed mice: plasma membrane [8], many of these lipidated
(1) an early Arl3 knockout in all retinal cells proteins behave differently in the Arl3-Q71L
driven by Six3-Cre and (2) a late Arl3 knockout transgenic mouse [22]. Transducin-α is normal in
in rod photoreceptors driven by iCre75 [8]. both expression and localization, while GRK1 is
Because Six3-Cre causes recombination embry- downregulated but remains exclusively in the
onically, the early Arl3-KO (called retArl3−/− in outer segments. Two lipidated protein complexes,
the original publication) also showed that Arl3 is PDE6α/β and transducin-β/γ, are found in the
critical for the initial building of the ciliary outer inner segment in both the Arl3-Q71L transgenic
segment. In these mice, no ciliary axoneme and late Arl3-KO mouse; however, their location
formed and photoreceptor degeneration was in the presence of Arl3-Q71L appears to be pri-
completed by 1 month. In contrast, in the late marily in large inner segment puncta rather than
Arl3-KO (called rodArl3−/− in the original publica- the diffuse membrane localization seen for mis-
tion), iCre75 causes recombination postnatally localized lipidated proteins in the late Arl3-KO
(P4-P7), allowing rods to form outer segments mouse. These data imply that although both
before Arl3 is knocked out. Despite forming knockout and constitutively active expression of
outer segments, the late Arl3-KO also shows Arl3 cause rod photoreceptor degeneration, the
rapid rod cell degeneration that was completed underlying mechanism behind the degeneration
by 2 months. In these mice, there is a mislocal- could be different. Interestingly, the Arl3-Q71L
ization of lipidated outer segment-specific pro- transgenic mouse also has a layer of rod nuclei
teins like myristoylated transducin-α, farnesylated mislocalized to the inner nuclear layer [22], a
rhodopsin kinase (GRK1), transducin-γ and phenotype not associated with any of the Arl3
Rab28, and prenylated PDE6α/β to the inner seg- knockout mice. A recent study from our lab con-
ments of rods as early as P15 [8]. As expected, firmed that expression of the constitutively active
outer segment transmembrane proteins were Arl3-Q71L variant in wild type mouse rods
unaffected by the loss of Arl3. By allowing the causes a rod nuclear migration defect [24].
formation of normal outer segments, the authors
were able to confirm a role for Arl3 in trafficking
lipidated proteins to the outer segment [8]. From 3 Human Mutations in Arl3
these mouse studies, it is clear that Arl3 is impor-
tant for both building outer segments and main- In the last few years, human mutations in the
taining outer segment composition. small GTPase Arl3 have been linked to various
The nucleotide cycle of Arl3 as a small forms of retinal degeneration. All the published
GTPase protein is also critical for the health of data on Arl3 human disease show early involve-
photoreceptors. Transgenic expression of Arl3-­ ment of both rod and cone photoreceptors that are
Q71L, a GTP-locked Arl3 mutant, in rod photo- affected across the retina and macula. Therefore,
receptors (using the same rhodopsin promoter we will discuss the known Arl3 variants based on
used to drive Cre expression in late Arl3-KO the broader classification of nonsyndromic domi-
mouse) results in rod photoreceptors that build nant retinal degeneration, nonsyndromic reces-
light-responsive outer segments of normal length, sive retinal degeneration, and syndromic
similar to the late Arl3-KO mice. These outer recessive Joubert syndrome. For a thorough dis-
segments do show subtle defects in the packing cussion of the clinical phenotypes presented for
of the outer segment discs at P20 and rod degen- each Arl3 variant, please refer to Ratnapriya et al.
eration ultimately occurs by 2 months [22]. [15].
Comparing the Arl3-Q71L mouse to the late Arl3 was first associated with inherited reti-
Arl3-KO mouse reveals important differences nal degeneration when a missense Arl3 variant,
despite the similar degeneration timeline. c.269A > G (p.Tyr90Cys), was reported as a
Although Arl3-KO mice show lipidated outer possible cause of nonsyndromic dominant reti-
286 A. M. Travis and J. N. Pearring

nal degeneration in a family of European its effectors, RP2, Arl13B, and UNC119A [15].
Caucasian decent and then in a Norwegian fam- For both of these Arl3 variants, only a single
ily [11, 19]. Tyr90 is an evolutionarily con- copy is required to cause retinal disease, so it is
served residue that resides at the center of the likely that the mutation behaves in dominant-
Arl3 structure where it makes interactions with negative manner. Recently, we published a
a neighboring β-sheet and central α-helix of study that investigated the biochemical and cel-
Arl3 (Fig. 2). The central location of the human lular consequences of dominant mutations in
Tyr90Cys mutation could have a destabilizing Arl3 [24]. We found that the two dominant Arl3
effect on Arl3 structure; however, protein stabil- variants, Arl3-Y90C and Arl3-D67V, pheno-
ity was not assessed. Another missense Arl3 copied the constitutively active Arl3-Q71L by
variant, c.200A > T (p.Asp67Val), was then causing mislocalization of rod nuclei basally
identified in a fourth-­ generation family with within ONL and even displaced into the INL. We
nonsyndromic dominant retinal degeneration went on to show that aberrant activity of Arl3 in
[15]. The Asp67 residue resides near the nucle- rods caused the developmental nuclear migra-
otide-binding pocket and is highly conserved tion defect; however, each dominant muta-
across evolution and the small GTPase family tion had a different GTPase behavior: D67V
(Fig. 2). In silico analysis predicts the Asp67Val behaved as a constitutively active GTPase and
substitution will disrupt Arl3 interactions with Y90C functioned as a fast cycling GTPase.

Fig. 2 Location of human Arl3 variants. (a) Protein structure of mouse Arl3 (yellow; [21] bound to GTP (red)
alignment of multiple Arl3 from different species, human and Mg2+ (gray). The residues T31A, D67V, and Y90C
Arl2 and human Arf1. Known disease causing human linked to nonsyndromic dominant retinal degeneration are
Arl3 variants are highlighted (cyan for dominant, magenta shown in cyan. The residues R99I, C118F, and R149H/C
for recessive) showing these residues are highly con- linked to nonsyndromic recessive retinal degeneration and
served. (b) Secondary linear structure and (c) 3D crystal recessive Joubert Syndrome are shown in magenta
Human Mutations in Arl3, a Small GTPase Involved in Lipidated Cargo Delivery to the Cilia, Cause Retinal… 287

Variants in Arl3 have also been associated c.353G > T (p.Cys118Phe), were identified in
with nonsyndromic recessive retinal degenera- Arl3 [6]. The male proband with both variants
tion. A homozygous c.296G > T (p. Arg99Ile) had early onset autosomal recessive retinal
was identified in two large consanguineous degeneration, while his father only had the
Pakistani families with autosomal recessive c.91A > G (p.Thr31Ala) variant and presented
­retinal degeneration [18]. The Arg99 residue is with late-onset dominant retinal degeneration.
highly conserved in the small GTPase family as it The Thr31 residue is within the nucleotide-­
forms ionic bonds to stabilize the canonical small binding pocket of Arl3 where it forms interac-
GTPase phosphate-binding loop (aka P-loop, tions with the Mg2+ ion necessary for nucleotide
Fig. 2). Bacterial expression of the Arl3-Arg99Ile exchange. Mutating the threonine to asparagine
mutant resulted in an insoluble protein, while results in an inactive GDP-bound Arl3 protein;
exogenous expression in cell culture resulted in a however, the authors suggest that the smaller ala-
Arl3 protein that was smaller in size with multi- nine residue would destabilize nucleotide bind-
ple degradative products; together suggesting ing. Similarly, the Cys118Phe mutation
Arl3-Arg99Ile protein is instable and prone to introduces a large, hydrophobic residue that
proteolytic cleavage [18]. Both inheritance pat- would destabilize Arl3 structure. Cycloheximide
tern and molecular instability are consistent with chase assays performed on recombinant protein
the Arg99Ile variant behaving as a loss of showed that both Thr31Ala and Cys118Phe vari-
function. ants had decreased protein stability compared to
Two missense variants, c.445C > T (p. wild-type Arl3 [6]. The authors also report that
Arg149Cys) and c.446G > A (p.Arg149His), RP2 binding is reduced with the Cys118Phe
were identified in two unrelate families with mutations even though this cystine residue
Joubert syndrome, a genetically heterogeneous resides on the opposite side of the RP2 inter-
neurodevelopmental ciliopathy [1]. Clinical phe- phase, further suggesting overall instability of the
notypes of these patients were typical for Joubert Arl3 protein and a loss of function phenotype.
syndrome including molar tooth sign on brain Arl3 is not a common cause of retinal degen-
MRI and retinal dystrophy. The Arg149 residue is eration in humans; however, the recent flurry of
highly conserved through evolution as well as papers showing multiple variants causing domi-
within the small GTPase Arf family (Fig. 2). This nant, recessive, and syndromic retinal disease
residue is present at the interface between Arl3 show the necessity of Arl3 for photoreceptor
and Arl13B where it is involved in ionic interac- health. These studies also raised the possibly that
tions with a highly conserved Glu88 residue on different biochemical alterations in Arl3, whether
Arl13B [1, 7]. Further experiments revealed that the variant is inactivating, destabilizing, or acting
the Arl3-Arg149His mutant is stable but unable as a dominant negative, could produce divergent
to bind Arl13B, essentially making this Arl3 vari- pathobiological mechanisms. Our study
ant inactive. This was further supported when the showed that dominant mutations of Arl3 result in
ciliary localization of two lipidated proteins, a rod nuclear migration defect not seen with Arl3
INPP5E and NPHP3, was shown to be reduced in loss of function mutations, highlighting that these
patient-derived fibroblast cells compared to con- patients will likely require different approaches
trols [1]. Consistent with these findings, a patho- for treatment [24]. Going forward, it will be
genic variant in Arl13B (c.236G>, p.Arg79Gln) important to understand how dominant human
that causes Joubert syndrome was also shown to Arl3 variants alter photoreceptor composition
have reduced GEF activity toward Arl3 [2, 7]. during development and whether mislocalized
In a Finnish family, two compound heterozy- rod nuclei directly cause photoreceptor dysfunc-
gous variants, c.91A > G (p.Thr31Ala) and tion or degeneration.
288 A. M. Travis and J. N. Pearring

References 13. Linari M, Hanzal-Bayer M, Becker J. The delta sub-


unit of rod specific cyclic GMP phosphodiesterase,
PDE δ, interacts with the Arf-like protein Arl3 in a
1. Alkanderi S, Molinari E, Shaheen R, Elmaghloob Y,
GTP specific manner. FEBS Lett. 1999;458:55–9.
Stephen LA, Sammut V, Ramsbottom SA, Srivastava
14. Najafi M, Maza NA, Calvert PD. Steric volume
S, Cairns G, Edwards N, Rice SJ, Ewida N, Alhashem
exclusion sets soluble protein concentrations in pho-
A, White K, Miles CG, Steel DH, Alkuraya FS, Ismail
toreceptor sensory cilia. Proc Natl Acad Sci U S A.
S, Sayer JA. ARL3 mutations cause Joubert syndrome
2012;109:203–8.
by disrupting ciliary protein composition. Am J Hum
15. Ratnapriya R, Jacobson SG, Cideciyan AV, English
Genet. 2018;103:612–20.
MA, Roman AJ, Sumaroka A, Sheplock R, Swaroop
2. Cantagrel V, et al. Mutations in the cilia gene ARL13B
A. A novel ARL3 gene mutation associated with auto-
lead to the classical form of Joubert syndrome. Am J
somal dominant retinal degeneration. Front Cell Dev
Hum Genet. 2008;83:170–9.
Biol. 2021;9:720782.
3. Cavenagh MM, Breiner M, Schurmann A, Rosenwald
16. Schrick JJ, Vogel P, Abuin A, Hampton B, Rice
AG, Terui T, Zhang C, Randazzo PA, Adams M, Joost
DS. ADP-ribosylation factor-like 3 is involved in
HG, Kahn RA. ADP-ribosylation factor (ARF)-like
kidney and photoreceptor development. Am J Pathol.
3, a new member of the ARF family of GTP-binding
2006;168:1288–98.
proteins cloned from human and rat tissues. J Biol
17. Schwahn U, Lenzner S, Dong J, Feil S, Hinzmann
Chem. 1994;269:18937–42.
B, van Duijnhoven G, Kirschner R, Hemberger M,
4. ElMaghloob Y, Sot B, McIlwraith MJ, Garcia E,
Bergen AA, Rosenberg T, Pinckers AJ, Fundele
Yelland T, Ismail S. ARL3 activation requires the
R, Rosenthal A, Cremers FP, Ropers HH, Berger
co-GEF BART and effector-mediated turnover. elife.
W. Positional cloning of the gene for X-linked retini-
2021;10
tis pigmentosa 2. Nat Genet. 1998;19:327–32.
5. Evans RJ, Schwarz N, Nagel-Wolfrum K, Wolfrum U,
18. Sheikh SA, Sisk RA, Schiavon CR, Waryah YM,
Hardcastle AJ, Cheetham ME. The retinitis pigmen-
Usmani MA, Steel DH, Sayer JA, Narsani AK,
tosa protein RP2 links pericentriolar vesicle transport
Hufnagel RB, Riazuddin S, Kahn RA, Waryah AM,
between the Golgi and the primary cilium. Hum Mol
Ahmed ZM. Homozygous variant in ARL3 causes
Genet. 2010;19:1358–67.
autosomal recessive cone rod dystrophy. Invest
6. Fu L, Li Y, Yao S, Guo Q, You Y, Zhu X, Lei
Ophthalmol Vis Sci. 2019;60:4811–9.
B. Autosomal recessive rod-cone dystrophy associ-
19. Strom SP, Clark MJ, Martinez A, Garcia S, Abelazeem
ated with compound heterozygous variants in. Front
AA, Matynia A, Parikh S, Sullivan LS, Bowne SJ,
Cell Dev Biol. 2021;9:635424.
Daiger SP, Gorin MB. De novo occurrence of a variant
7. Gotthardt K, Lokaj M, Koerner C, Falk N, Gießl A,
in ARL3 and apparent autosomal dominant transmis-
Wittinghofer A. A G-protein activation cascade from
sion of retinitis pigmentosa. PLoS One. 2016;11:1–9.
Arl13B to Arl3 and implications for ciliary targeting
20. Veltel S, Kravchenko A, Ismail S, Wittinghofer
of lipidated proteins. elife. 2015;4:1–16.
A. Specificity of Arl2/Arl3 signaling is mediated by
8. Hanke-Gogokhia C, Wu Z, Gerstner CD, Frederick
a ternary Arl3-effector-GAP complex. FEBS Lett.
JM, Zhang H, Baehr W. Arf-lik8e protein 3 (ARL3)
2008a;582:2501–7.
regulates protein trafficking and Ciliogenesis in mouse
21. Veltel S, Gasper R, Eisenacher E, Wittinghofer A. The
photoreceptors. J Biol Chem. 2016;291:7142–55.
retinitis pigmentosa 2 gene product is a GTPase-­
9. Hanzal-Bayer M, Renault L, Roversi P, Wittinghofer
activating protein for Arf-like 3. Nat Struct Mol Biol.
A, Hillig RC. The complex of Arl2-GTP and
2008b;15:373–80.
PDE delta: from structure to function. EMBO J.
22. Wright ZC, Singh RK, Alpino R, Goldberg AFX,
2002;21:2095–106.
Sokolov M, Ramamurthy V. ARL3 regulates traffick-
10. Holopainen JM, Cheng CL, Molday LL, Johal
ing of prenylated phototransduction proteins to the rod
G, Coleman J, Dyka F, Hii T, Ahn J, Molday
outer segment. Hum Mol Genet. 2016;25:2031–44.
RS. Interaction and localization of the retinitis pig-
23. Zhang H, Constantine R, Vorobiev S, Chen Y,
mentosa protein RP2 and NSF in retinal photorecep-
Seetharaman J, Huang YJ, Xiao R, Montelione GT,
tor cells. Biochemistry. 2010;49:7439–47.
Gerstner CD, Davis MW, Inana G, Whitby FG,
11. Holtan JP, Teigen K, Aukrust I, Bragadóttir R, Houge
Jorgensen EM, Hill CP, Tong L, Baehr W. UNC119 is
G. Dominant ARL3-related retinitis pigmentosa.
required for G protein trafficking in sensory neurons.
Ophthalmic Genet. 2019;40:124–8.
Nat Neurosci. 2011;14:874–80.
12. Ismail SA, Chen Y-X, Miertzschke M, Vetter IR,
24. Travis AM, Manocha S, Willer JR, Wessler TS, Skiba
Koerner C, Wittinghofer A. Structural basis for Arl3-­
NP, Pearring JN. Disrupting the ciliary gradient of
specific release of myristoylated ciliary cargo from
active Arl3 affects rod photoreceptor nuclear migra-
UNC119. EMBO J. 2012;31:4085–94.
tion eLife. 2023;12:80533.
Genotype–Phenotype Association
in ABCA4-Associated Retinopathy

Maximilian Pfau, Wadih M. Zein, Laryssa A. Huryn,


Catherine A. Cukras, Brett G. Jeffrey,
Robert B. Hufnagel, and Brian P. Brooks

Abstract Keywords

Stargardt disease (STGD1) is the most com- Stargardt disease · ABCA4-associated


mon inherited retina degeneration. It is caused retinopathy · Disease Progression · Genotype
by biallelic ABCA4 variants, and no treatment · Genotype–Phenotype correlation
is available to date. STGD1 shows marked
phenotypic variability, especially regarding
the age of onset. The underlying genotype can 1 Introduction
partially explain this variability. Notably, a
subset of ABCA4 variants was previously Stargardt disease (STGD1, or ABCA4-­
associated with an earlier disease onset than retinopathy) is caused by biallelic variants in the
truncating ABCA4 variants, pointing toward ATP Binding Cassette Subfamily A Member 4
pathogenic mechanisms beyond the loss of (ABCA4) gene and represents the most frequent
gene function in these patients. On the other inherited retinal degeneration [1]. Dysfunction of
end of the spectrum, variants such as p. the retinal-specific phospholipid-transporting
Gly1961Glu were associated with markedly ATPase ABCA4 results in lipofuscin accumula-
slower extrafoveal disease progression. Given tion in the retinal pigment epithelium (RPE),
that these drastic differences in phenotype are leading to eventual atrophy of the RPE and neu-
based on genotype (resulting in important rosensory retina [2, 3].
prognostic implications for patients), this The clinical course of Stargardt disease [4] is
chapter reviews previous approaches to geno- characterized by centrifugally progressing RPE
type–phenotype correlation analyses in atrophy [5, 6], centrifugally progressing flecks
STGD1. [7, 8], and peripapillary sparing [9]. The typical
age of onset (in terms of symptoms) is between
the first and the second decade of life, but there is
a wide variation in the age of onset [10]. Initially,
M. Pfau · W. M. Zein · L. A. Huryn · C. A. Cukras ·
B. G. Jeffrey · R. B. Hufnagel · B. P. Brooks (*) best-corrected visual acuity may be preserved
National Eye Institute, National Institutes of Health, due to foveal sparing [11].
Bethesda, MD, USA Accumulation of lipofuscin in the RPE is the
e-mail: maximilian.pfau@iob.ch; zeinw@nei.nih.gov; histopathological hallmark of STGD1 [12] and
laryssa.huryn@nih.gov; cukrasc@nei.nih.gov;
jeffreybg@mail.nih.gov; robert.hufnagel@nih.gov; can be quantified in vivo using autofluorescence
brooksb@nei.nih.gov imaging [13]. Importantly, this increase of

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 289
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_42
290 M. Pfau et al.

a­utofluorescence precedes the loss of function, genotype severity estimate. Early genotype–phe-
implying that vision could be preserved if lipo- notype correlation analyses were based on quali-
fuscin accumulation is slowed [13]. Multiple tative, phenotypic descriptions of disease severity
therapeutic approaches are being evaluated in and reported variant severity in a qualitative man-
clinical trials or preclinical models [2, 3]. ner (cf. Sect. 2.1 below). More recent studies
Therapeutic strategies currently include slowing employed semi-quantitative metrics of disease
vitamin A delivery to the retina (e.g., RBP4- severity, such as the Lois electrophysiologic
antagonist STG-001 [ClinicalTrials.gov (ERG) classification [15] or ultra-widefield fun-
Identifier: NCT04489511]), slowing the visual dus autofluorescence imaging [16]. However,
cycle biochemically (e.g., RPE65-inhibitor these two studies reported the variant severity in
Emixustat [NCT03772665]), or slowing the rate a four- or three-step ordinal scaled manner (cf.
of lipofuscin accumulation (e.g., ALK-001, Sect. 2.2 below). Only one study so far applied a
NCT04239625). Other currently studied thera- quantitative, interval-scaled metric of disease
peutic approaches include the complement factor severity and reported variant severity in an
C5 inhibitor Zimura (NCT03364153) and oral interval-­scaled manner (cf. Sect. 2.3) [14].
metformin (NCT04545736). This review focuses only on clinical geno-
Despite marked phenotypic differences among type–phenotype analyses. Besides these clinical
patients with STGD1, genotypic characteristics approaches, complemental laboratory studies
are not considered within inclusion or exclusion have investigated the effect of ABCA4 variants,
criteria in ongoing trials (beyond the confirma- including biochemical assays of ATPase activity
tion of the diagnosis). Previously, a subset of [17–19], of protein processing [20], in vitro
ABCA4 variants was shown to be associated with splice assays [21], and rodent models [22–24].
an earlier disease onset than truncating ABCA4
variants implying pathogenic mechanisms
beyond the mere loss of function [14]. Potentially, 2.1 Qualitative Genotype–
these variants lead to heterogeneity of treatment Phenotype Correlation
effects (hypothetically, patients with these severe Analyses
variants benefit to a lesser degree from gene-­
replacement therapy). On the other end of the Shortly after the identification of ABCA4 as the
spectrum, patients with mild variants may lead to causal gene for STGD1 [25–28], qualitative geno-
underpowered studies due to slow progression type–phenotype correlations were reported. These
rates, diminishing differences in the treatment were based on either self-reported age of onset or
and control group (in the absence of stratified descriptive characterizations of the fundus.
sampling). For example, Rozet et al. reported in 1998 that
Given the importance of understanding phe- they could identify truncating and missense vari-
notypic variability and its underlying genetic cor- ants in patients with a classical STGD1 phenotype
relates to inform patients of their prognosis and and only missense variants in patients with a late-
clinical trial design, this chapter reviews previous onset STGD1 phenotype (termed FFM in their
approaches to genotype–phenotype correlation publication) [28]. Lewis et al. subsequently pro-
analyses in STGD1. vided the first granular genotype–phenotype anal-
ysis highlighting that variants 5′ to codon 863 tend
to be associated with an earlier onset [29].
2 Genotype–Phenotype
Correlation Analyses 2.1.1 Qualitative Descriptions of Mild
Variants
Published genotype–phenotype analyses In another early study, Fishman et al. 1999 [30]
approaches in ABCA4-associated retinopathy can reported that the typical phenotype associated
be subclassified according to (i) the metric of dis- with ABCA4 p.Gly1961Glu was characterized by
ease severity and (ii) the scale of the resulting central atrophy but preserved retina-wide cone
Genotype–Phenotype Association in ABCA4-Associated Retinopathy 291

and rod function as measured by full-field elec- Fakin et al. reported in 2016 a genotype–phe-
troretinogram (ERG). This association of p. notype correlation analysis based on 82 patients
Gly1961Glu with the absence of severe retina-­ with one of 15 missense variants of interest that
wide cone and rod sensitivity loss was also con- were in trans with an established null variant (i.e.,
firmed by Gerth et al. using dark-adapted compound heterozygous for a null and missense
two-color perimetry [31]. ABCA4 p.Gly1961Glu variant; referred to “hemizygous” by Fakin et al.)
has also been linked to the “optical gap pheno- [38]. Further patients homozygous for a subset of
type,” characterized by disruption of the ellipsoid these missense variants (N = 10) and patients with
zone at the fovea, suggesting that the p. biallelic null variants (N = 10) served as compara-
Gly1961Glu variant selectively affects foveal tors. In a first step, the Lois ERG classification
cones. This variant was also shown to lead to a was applied to the data of the “hemizygous”
lesser degree of lipofuscin accumulation using patients. If a given variant in trans with a null vari-
quantitative autofluorescence imaging [32, 33]. ant was always associated with a Lois ERG group
Further, the ABCA4 variant p.Asn1868Ile, 1 phenotype (pattern ERG abnormality, but nor-
which has a minor allele frequency of ~7% in the mal full-field ERG), it was classified as mild (e.g.,
general population, was linked to a mild pheno- p.Gly1961Glu and p.R2030Q). Variants in trans
type [34]. It is characterized by a late age of onset
with a null variant, which were predominantly
and foveal sparing, as well as an overall lower associated with a Lois ERG group 3 phenotype
degree of lipofuscin accumulation [32–34]. (abnormal pattern ERG, abnormal photopic, and
scotopic full-field ERG), were classified as “null-
2.1.2 Qualitative Descriptions like.” Variants in trans with null variants associ-
of Severe Variants ated with a range of ERG phenotypes were
Fukui et al. further extended the spectrum of classified as intermediate. In a second step, the
STGD1 with the description of two brothers phenotype of the “hemizygous” patients was
homozygous for c.1760 + 2 T > G with severe compared to the linear slope of the dark-adapted
pan-retinal degeneration and bone-spicule-like A-wave amplitude in patients with biallelic null
hyperpigmentary changes [35]. Further variants variants. Specifically, the number of eyes that fell
have been linked to a severe STGD1 manifesta- within or outside of the 95% prediction interval
tion with an early onset, including the complex for patients with biallelic null variants was evalu-
allele p.[Leu541Pro;Ala1038Val] and the intronic ated. Based on this ERG analysis, the intermedi-
variant c.5461-10 T > C [36]. ate variants were stratified as “intermediate +”
More recently, more severe forms of early-­ (variants in trans with null that were mostly asso-
onset STGD1 have also been referred to as ciated with a dark-adapted A-wave amplitude out-
“Generalized Choriocapillaris Dystrophy,” [37] side of the 95% prediction interval for biallelic
and “Rapid-Onset Chorioretinopathy” [36]. null variants) and “intermediate-” (dark-adapted
However, clear-cut genetic or phenotypic charac- A-wave amplitude within of the 95% prediction
teristics that differentiate these severe forms of interval for biallelic null variants) [38]. Last, the
STGD1 from the overall spectrum of STGD1 are authors compared patients with variants that led
lacking to date. in trans with null variants to a severe ERG pheno-
type, to patients that were homozygous for these
missense variants. Patients homozygous for some
2.2 Semi-quantitative Genotype– of these variants (p.Arg212Cys, p.Arg1108Cys,
Phenotype Correlation and p.Pro1380Leu) had significantly better ampli-
Analyses tudes compared to their “hemizygous” counter-
parts. Accordingly, these variants were classified
Few studies have summarized the severity of as “intermediate -” [38]. In a later publication,
ABCA4 variants based on interval-scaled metrics deep-intronic ­mutation c.5196 + 1137G > A vari-
of disease severity but reported the results as an ant was added to the classification as “intermedi-
ordinal-scaled, four-step ranking of severity [38]. ate” based on the same approach [39].
292 M. Pfau et al.

This approach separated previously consid- Cideciyan and coworkers evaluated 66 patients
ered mild variants (e.g., p.Gly1961Glu) from with STGD1 using light-adapted and dark-­
those previously reported as severe (e.g., p. adapted perimetry [14]. According to the authors,
Cys2150Tyr, or p.[Leu541Pro;Ala1038Val]). sensitivity loss could be described by a central,
However, the proposed classification approach centrifugally progressing component and a
has several limitations: (i) the approach always retina-­wide component (mean retina-wide sensi-
necessitates data from patients with a known null tivity loss for loci at ≥30° eccentricity from the
or “null-like” variant in trans, (ii) does not allow fovea). In 36 patients, abnormal extramacular rod
to identify variants that are more severe than null-­ or cone sensitivity was evident at one or more
variants, and (iii) provides only ordinal-scaled visits. It progressed at an average rate of 1.1 log/
estimates of variant severity [38]. decade (rod sensitivity loss) and 0.45 log/decade
Recently, Heath Jeffery and coworkers (cone sensitivity loss). Based on these slope esti-
expanded on this classification based on a large mates, the patient’s age, and the respective retinal
cohort of patients imaged using the total lesion sensitivity, the authors computed for each patient
size (defined by the outer boundary of flecks) the age of retina-wide disease initiation (ADI).
measured from ultra-widefield fundus autofluo- For patients with two truncating variants, the ADI
rescence imaging. Specifically, the authors eval- was 10.6 years. Subsequently, the authors esti-
uated 81 STGD1 patients from 65 families. mated for each variant the severity (in terms of
Patients were either classified based on prior the delay of retina-wide disease initiation) assum-
estimates for variant severity (group A: biallelic ing an additive contribution of each variant.
null variants; group B1: with a mild variant [p. Notably, one-third of the nontruncating variants
Gly1961Glu, p.Asn1868Ile]; group B2: with an were found to cause earlier onset disease than
intermediate variant [p.Leu2027Phe, or the truncating variants. This was considered as an
complex allele p.[Gly863Ala,Gly863del;Asn18 evidence for pathogenic pathways beyond the
68Ile]), or an uncertain severity (group C). mere loss of biallelic function [14]. In a later
Then, patients in group C with a self-reported report by the same group, it was quantitatively
age of onset <14 years were assigned into the shown using this approach that the complex vari-
null/severe variant cohort and patients with a ant p.[Leu541Pro;Ala1038Val] is associated with
total lesion size of <200 mm2 as group B1 and an earlier disease onset than the p.Ala1038Val
with a total lesion size >200 mm2 as group B2. variant [22].
Based on this approach, Heath Jeffery and The disadvantages of their approach were that
coworkers estimated the severity for 32 all analyses were based on the assumption of an
variants. invariant slope of sensitivity loss across patients
Like the previous approach, the analysis did [14]. However, the raw data are suggestive of
not fully account for the age-dependent nature of some between-patient variability in the slope
the applied severity metric (total lesion size). (Figure 3 in Cideciyan et al.) [14]. Further, the
These ordinal-scaled estimates of variant severity model of an additive association of variant sever-
result in a significant loss of information. ity with the ADI is most likely an oversimplifica-
tion of the underlying biology.

2.3 Quantitative Genotype–


Phenotype Correlation 3 Future Directions
Analyses
Severity estimates based on clinical characteris-
Only one previous study employed an interval-­ tics are available for many ABCA4 variants.
scaled metric as the criterion for genotype–pheno- Importantly, these estimates of variant severity
type correlation analysis and reported an are mostly congruent. However, four gaps in the
interval-scaled metric for the variant severity [14]. literature are evident that could be addressed.
Genotype–Phenotype Association in ABCA4-Associated Retinopathy 293

3.1 Interval-Scaled Severity 3.3 Differentiating Age-­


Measures as Input Dependent Severity
from Genuine Phenotypic
Previous studies have summarized continuous Differences
data as an ordinal-scaled measure of severity.
However, dichotomizing (or discretizing) a con- Third, the age-dependent nature of disease sever-
tinuous variable will result in a considerable loss ity metrics has not always been (explicitly)
of power [40]. The one study that applied an accounted for in the genotype–phenotype analy-
interval-scaled measure of disease severity in ses. The framework outlined by Cideciyan and
terms of the ADI (based on the sensitivity loss coworkers, which defined a fixed criterion age for
and patient’s age) did so assuming an invariant each patient based on the (estimated) age of
slope of sensitivity loss across subjects [14]. retina-­wide disease initiation, provides a severity
Using linear mixed model analysis, it would be metric independent of the patient’s age of presen-
possible to derive slope estimates for patients tation [14].
with multiple measurements as well as for
patients with only a single measurement (through
“pooling”) [41]. 3.4 Latent Variable Methods

Last, measures for disease severity for STGD1


3.2 Interval-Scaled Reporting have not been assessed systematically using
of Variant Severity latent variable methods. Outside of “station-
ary” dystrophies (e.g., congenital stationary
Second, for most ABCA4 variants, only ordinal-­ night blindness or achromatopsia), the degree
scaled (four- or three-step) data have been pub- of rod and cone degeneration tends to be cor-
lished, even though interval-scaled measures of related in most IRDs. While the commonly
disease severity constituted the starting point for applied Lois ERG classification for STGD1
these analyses. This drastic reduction in granular- treats photopic and scotopic B-wave ampli-
ity complicates the comparison of estimates of tudes as differential information [15], these
allele severity across studies. It also hinders the measures are partially redundant in STGD1.
comparison to in vitro splice assay data, which is On the other hand, BCVA appears to be reflec-
typically reported in an interval-scaled manner tive of an unrelated latent process in STGD1.
[21]. Moreover, ABCA4 variants more severe than As a result, the common ABCA4 p.Gly1961Glu
truncating and frameshift variants have been variant has been reported to be “mild” (based
described (e.g., the complex variant p. on full-field ERG, peripheral perimetric sensi-
[Leu541Pro;Ala1038Val]) [14, 22]. However, the tivity) [38], but as “moderate” (based on the
above-mentioned ordinal-scaled classification age of onset [i.e., BCVA]) [42]. Thus, the
does not provide a distinct class for more severe application of latent variable methods (e.g.,
variants than “null-like” variants. From a clinical principal component analysis or factor analy-
trial perspective, identifying variants more severe sis) is warranted to pool redundant visual func-
than “null-like” variants is a priority since disease tion metrics (improving the signal-to-noise
mechanisms beyond the mere loss of function ratio) and identify nonredundant (low covari-
could contribute to treatment effect heterogeneity. ance) metrics of disease severity.
294 M. Pfau et al.

4 Summary 7. Cukras CA, Wong WT, Caruso R, Cunningham D,


Zein W, Sieving PA. Centrifugal expansion of fundus
autofluorescence patterns in Stargardt disease over
Previous studies have reported qualitative pheno- time. Arch Ophthalmol. 2012 Feb;130(2):171–9.
type–genotype correlations at the level of indi- 8. Cideciyan AV, Swider M, Schwartz SB, Stone EM,
vidual variants and few in a semi-quantitative or Jacobson SG. Predicting progression of ABCA4-­
associated retinal degenerations based on longitudinal
quantitative manner [14, 16, 38]. Only one previ- measurements of the leading disease front. Investig
ous publication has accounted for the time-­ Ophthalmol Vis Sci. 2015 Sep;56(10):5946–55.
dependent nature of disease severity metrics and 9. Cideciyan AV, Swider M, Aleman TS, Sumaroka A,
reported a genotype–phenotype analysis with Schwartz SB, Roman MI, et al. ABCA4-associated
retinal degenerations spare structure and function of
interval-scaled estimates of variant severity [14]. the human parapapillary retina. Investig Ophthalmol
Besides larger sample sizes, future genotype– Vis Sci. 2005 Dec;46(12):4739–46.
phenotype analyses in STGD1 could be improved 10. Lambertus S, van Huet RAC, Bax NM, Hoefsloot LH,
by employing interval-scaled measures of dis- Cremers FPM, Boon CJF, et al. Early-onset stargardt
disease: phenotypic and genotypic characteristics.
ease severity, reporting interval-scaled measures Ophthalmology. 2015 Feb;122(2):335–44.
of allele severity, using age-independent disease 11. van Huet RAC, Bax NM, Westeneng-Van Haaften
severity metrics (instead of disease severity at SC, Muhamad M, Zonneveld-Vrieling MN, Hoefsloot
patient’s age of presentation) and applying latent LH, et al. Foveal sparing in Stargardt disease. Invest
Ophthalmol Vis Sci. 2014 Oct;55(11):7467–78.
variable methods to separate redundant informa- 12. Bonilha VL, Rayborn ME, Bell BA, Marino MJ,
tion from genuine phenotypic characteristics. Fishman GA, Hollyfield JG. Retinal histopathology
in eyes from a patient with Stargardt disease caused
Conflict of Interest Statement The authors have by compound heterozygous ABCA4 mutations.
declared that no conflict of interest exists. Ophthalmic Genet. 2016 June;37(2):150–60.
13. Cideciyan AV, Aleman TS, Swider M, Schwartz SB,
Steinberg JD, Brucker AJ, et al. Mutations in ABCA4
result in accumulation of lipofuscin before slowing of
References the retinoid cycle: a reappraisal of the human disease
sequence. Hum Mol Genet. 2004 Mar;13(5):525–34.
1. Hanany M, Rivolta C, Sharon D. Worldwide carrier 14. Cideciyan AV, Swider M, Aleman TS, Tsybovsky Y,
frequency and genetic prevalence of autosomal reces- Schwartz SB, Windsor EAMM, et al. ABCA4 disease
sive inherited retinal diseases. Proc Natl Acad Sci U S progression and a proposed strategy for gene therapy.
A. 2020 Feb;117(5):2710–6. Hum Mol Genet. 2009 Mar;18(5):931–41.
2. Huang D, Heath Jeffery RC, Aung-Htut MT, 15. Lois N, Holder GE, Bunce C, Fitzke FW, Bird
McLenachan S, Fletcher S, Wilton SD, et al. Stargardt AC. Phenotypic subtypes of Stargardt macular:
disease and progress in therapeutic strategies. dystrophy-fundus flavimaculatus. Arch Ophthalmol
Ophthalmic Genet. 2021 Aug;1–26. [Internet]. 2001 Mar;119(3):359–369. Available from:
3. Cremers FPM, Lee W, Collin RWJ, Allikmets http://www.ncbi.nlm.nih.gov/pubmed/11231769
R. Clinical spectrum, genetic complexity and thera- 16. Heath Jeffery RC, Thompson JA, Lamey TM,
peutic approaches for retinal disease caused by McLaren TL, McAllister IL, Constable IJ, et al.
ABCA4 mutations. Prog Retin Eye Res. 2020 Classifying ABCA4 mutation severity using age-­
Nov;79:100861. dependent ultra-widefield fundus autofluorescence-­
4. Stargardt K. Über familiäre, progressive Degeneration derived total lesion size. Retina 2021 Jun;
in der Maculagegend des Auges. Albr von Graefes 17. Sun H, Smallwood PM, Nathans J. Biochemical defects
Arch für Ophthalmol [Internet]. 1909;71(3):534–550. in ABCR protein variants associated with human reti-
Available from:https://doi.org/10.1007/BF01961301. nopathies. Nat Genet. 2000 Oct;26(2):242–6.
5. Strauss RW, Kong X, Ho A, Jha A, West S, Ip M, 18. Shroyer NF, Lewis RA, Yatsenko AN, Wensel TG,
et al. Progression of Stargardt disease as determined Lupski JR. Cosegregation and functional analy-
by fundus autofluorescence over a 12-month period: sis of mutant ABCR (ABCA4) alleles in families
ProgStar report no. 11. JAMA Ophthalmol. 2019 that manifest both Stargardt disease and age-related
Aug;137(10):1134–45. macular degeneration. Hum Mol Genet. 2001
6. Müller PL, Pfau M, Treis T, Pascual-Camps I, Birtel Nov;10(23):2671–8.
J, Lindner M, et al. Progression of Abca4-related 19. Biswas-Fiss EE. Functional analysis of genetic muta-
retinopathy—prognostic value of demographic, func- tions in nucleotide binding domain 2 of the human
tional, genetic, and imaging parameters. Retina. 2020 retina specific ABC transporter. Biochemistry. 2003
Jan;1. Sep;42(36):10683–96.
Genotype–Phenotype Association in ABCA4-Associated Retinopathy 295

20. Wiszniewski W, Zaremba CM, Yatsenko AN, Jamrich Stargardt macular dystrophy/fundus flavimacu-
M, Wensel TG, Lewis RA, et al. ABCA4 mutations latus patients with known ABCA4 mutations and
causing mislocalization are found frequently in evaluation of genotype-­ phenotype correlation.
patients with severe retinal dystrophies. Hum Mol Graefe’s Arch Clin Exp Ophthalmol = Albr von
Genet. 2005 Oct;14(19):2769–78. Graefes Arch fur Klin und Exp Ophthalmol. 2002
21. Sangermano R, Khan M, Cornelis SS, Richelle V, Aug;240(8):628–38.
Albert S, Garanto A, et al. ABCA4 midigenes reveal 32. Duncker T, Stein GE, Lee W, Tsang SH, Zernant
the full splice spectrum of all reported noncanonical J, Bearelly S, et al. Quantitative fundus autofluo-
splice site variants in Stargardt disease. Genome Res. rescence and optical coherence tomography in
2018 Jan;28(1):100–10. ABCA4 carriers. Investig Ophthalmol Vis Sci. 2015
22. Zhang N, Tsybovsky Y, Kolesnikov AV, Rozanowska Nov;56(12):7274–85.
M, Swider M, Schwartz SB, et al. Protein misfold- 33. Müller PL, Gliem M, McGuinnes M, Birtel J, Holz
ing and the pathogenesis of ABCA4-associated FG, Charbel IP. Quantitative fundus autofluores-
retinal degenerations. Hum Mol Genet. 2015 cence in ABCA4-related retinopathy -functional rel-
Jun;24(11):3220–37. evance and genotype-phenotype correlation. Am J
23. Weng J, Mata NL, Azarian SM, Tzekov RT, Birch Ophthalmol [Internet]. 2021;222:340–50. Available
DG, Travis GH. Insights into the function of rim pro- from: https://www.sciencedirect.com/science/article/
tein in photoreceptors and etiology of Stargardt’s dis- pii/S0002939420304797
ease from the phenotype in abcr knockout mice. Cell. 34. Zernant J, Lee W, Collison FT, Fishman GA, Sergeev
1999;98(1):13–23. YV, Schuerch K, et al. Frequent hypomorphic alleles
24. Charbel Issa P, Barnard AR, Singh MS, Carter E, account for a significant fraction of ABCA4 disease
Jiang Z, Radu RA, et al. Fundus autofluorescence in and distinguish it from age-related macular degenera-
the Abca4(−/−) mouse model of Stargardt disease— tion. J Med Genet. 2017 Jun;54(6):404–12.
correlation with accumulation of A2E, retinal func- 35. Fukui T, Yamamoto S, Nakano K, Tsujikawa M,
tion, and histology. Invest Ophthalmol Vis Sci. 2013 Morimura H, Nishida K, et al. ABCA4 gene muta-
Aug;54(8):5602–12. tions in Japanese patients with Stargardt disease and
25. Allikmets R, Singh N, Sun H, Shroyer NF, Hutchinson retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2002
A, Chidambaram A, et al. A photoreceptor cell-­ Sep;43(9):2819–24.
specific ATP-binding transporter gene (ABCR) is 36. Tanaka K, Lee W, Zernant J, Schuerch K, Ciccone L,
mutated in recessive Stargardt macular dystrophy. Nat Tsang SH, et al. The rapid-onset Chorioretinopathy
Genet [Internet]. 1997 Mar;15(3):236–246. Available phenotype of ABCA4 disease. Ophthalmology. 2018
from: http://www.ncbi.nlm.nih.gov/pubmed/9054934 Jan;125(1):89–99.
26. Gerber S, Rozet JM, van de Pol TJ, Hoyng CB, 37. Bertelsen M, Zernant J, Larsen M, Duno M, Allikmets
Munnich A, Blankenagel A, et al. Complete exon-­ R, Rosenberg T. Generalized choriocapillaris dystro-
intron structure of the retina-specific ATP binding phy, a distinct phenotype in the spectrum of ABCA4-­
transporter gene (ABCR) allows the identification associated retinopathies. Investig Ophthalmol Vis Sci.
of novel mutations underlying Stargardt disease. 2014 May;55(4):2766–76.
Genomics. 1998 Feb;48(1):139–42. 38. Fakin A, Robson AG, Chiang JPW, Fujinami K,
27. Nasonkin I, Illing M, Koehler MR, Schmid M, Moore AT, Michaelides M, et al. The effect on reti-
Molday RS, Weber BH. Mapping of the rod photo- nal structure and function of 15 specific ABCA4
receptor ABC transporter (ABCR) to 1p21-p22.1 and mutations: a detailed examination of 82 Hemizygous
identification of novel mutations in Stargardt’s dis- patients. Investig Ophthalmol Vis Sci. 2016
ease. Hum Genet. 1998 Jan;102(1):21–6. Nov;57(14):5963–73.
28. Rozet JM, Gerber S, Souied E, Perrault I, Châtelin S, 39. Khan M, Arno G, Fakin A, Parfitt DA, Dhooge PPA,
Ghazi I, et al. Spectrum of ABCR gene mutations in Albert S, et al. Detailed phenotyping and thera-
autosomal recessive macular dystrophies. Eur J Hum peutic strategies for intronic ABCA4 variants in
Genet. 1998;6(3):291–5. Stargardt disease. Mol Ther Nucleic Acids. 2020
29. Lewis RA, Shroyer NF, Singh N, Allikmets R, Sep;21:412–27.
Hutchinson A, Li Y, et al. Genotype/phenotype analy- 40. Royston P, Altman DG, Sauerbrei W. Dichotomizing
sis of a photoreceptor-specific ATP-binding cassette continuous predictors in multiple regression: a bad
transporter gene, ABCR. Stargardt disease Am J Hum idea. Stat Med. 2006 Jan;25(1):127–41.
Genet. 1999 Feb;64(2):422–34. 41. Bates D, Mächler M, Bolker BM, Walker SC. Fitting
30. Fishman GA, Stone EM, Grover S, Derlacki DJ, linear mixed-effects models using lme4. J Stat Softw
Haines HL, Hockey RR. Variation of clinical expres- [Internet]. 2015 Oct 7;67(1). Available from: https://
sion in patients with Stargardt dystrophy and sequence www.jstatsoft.org/v067/i01
variations in the ABCR gene. Arch Ophthalmol. 1999 42. Simonelli F, Testa F, Zernant J, Nesti A, Rossi S,
Apr;117(4):504–10. Allikmets R, et al. Genotype-phenotype correlation
31. Gerth C, Andrassi-Darida M, Bock M, Preising in Italian families with Stargardt disease. Ophthalmic
MN, Weber BHF, Lorenz B. Phenotypes of 16 Res. 2005;37(3):159–67.
Retinal Pathoconnectomics:
A Window into Neurodegeneration

Rebecca L. Pfeiffer and Bryan W. Jones

Abstract 1 Introduction

Over the past decade, the field of retinal con­ The idea of distributed networks underlying ner­
nectomics has made huge strides in describing vous system function dates back to the Roman
the precise topologies underlying retinal physician Galen. As our knowledge of neuroanat­
visual processing. The same techniques that omy has progressed, understanding neuronal and
allowed these advancements are also appli­ glial diversity, along with the molecular contribu­
cable to understanding the progression of tions underlying neural networks has proven
rewiring in retinal remodeling: retinal patho­ invaluable. Yet, a precise understanding of how
connectomics. Pathoconnectomics is unique neurons are connected to one another, and their
in its unbiased approach to understanding the relationships with glia, is an ideal application for
impacts of deafferentation on the remaining connectomics approaches. Detailing neuronal
network components and identifying aberrant class membership and their precise synaptic con­
connectivities leading to visual processing nections with one another is the basis for the field
defects. Pathoconnectomics also paves the of ultrastructural connectomics [1].
way for identifying underlying rules of rewir­ A connectome is an audacious undertaking,
ing that may be recapitulated throughout the requiring the expertise of a team skilled in anat­
nervous system in other neurodegenerative omy, histology, electron microscopy, chemistry,
diseases. and computational resources to capture, assem­
ble, annotate, and analyze the resulting large data
Keywords sets required to encode canonical volumes of
neural tissue [2]. Connectomics approaches
Retinal remodeling · Connectomics ·
reveal the network architecture ground truth that
Pathoconnectome · Ultrastructure · Early
have remained hidden to prior techniques.
retinal degeneration · Neurodegeneration ·
Connectomics initiatives have revealed numer­
Neural networks
ous specific cell classes possessing greater diver­
sity and network complexity than expected, with
retina leading the way in describing precise net­
R. L. Pfeiffer (*) · B. W. Jones
John A. Moran Eye Center, Department of work topologies [3–12].
Ophthalmology, University of Utah, The retina presents a unique opportunity to
Salt Lake City, UT, USA explore neuronal networks of the central ner­
e-mail: r.pfeiffer@utah.edu; bryan.jones@m.cc.utah. vous system (CNS). Retina is compact, highly
edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 297
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_43
298 R. L. Pfeiffer and B. W. Jones

organized, and consists of complex, yet com­ ideal model to explore network and molecular
plete, networks that perform all of the algorith­ components of neural injury.
mic computations associated with visual Negative plasticity has long been known in the
primitives, organized in a matrix of repeating hippocampus and other regions of CNS [15, 16],
motifs allowing for robust exploration and and retina is no different in its response to deaf­
quantification. Within the past 10 years, retinal ferentation, exhibiting remodeling processes in
connectomics has identified novel circuits trauma or diseases that compromise photorecep­
including ON-OFF bipolar cell crossover motifs tors. Remodeling is not restricted to photorecep­
[4], crossover motifs involving rod and cone tors in outer retina that comprise the sensory
vision [5], nested amacrine cell networks [6], retina. Following injury (e.g., retinal detachment,
novel network topologies between amacrine light-induced retinal degeneration) [17–19] or
cells and ganglion cells [7], and numerous gap disease (e.g., retinitis pigmentosa, age-related
junctional coupling motifs below light micros­ macular degeneration) [20–26], the retina under­
copy resolutions [8] that are highly conserved goes a progressive series of revisions including
across the retina. All these network architec­ neuritic sprouting, alterations in glutamate chan­
tures underly components of retinal visual pro­ nel expression, aberrant synaptogenesis, and glial
cessing, illustrating connectomics approaches responses [27–29]. Of particular interest to con­
for understanding how networks takes seem­ nectomics is the neurite sprouting and aberrant
ingly simple input (photons) and transforms it synaptogenesis underlying rewiring.
via neuronal and glial networks into the compo­ Early investigations of retinal remodeling and
nents necessary for interpreting the visual world rewiring were accomplished through immunohis­
around us. tochemical analyses [29]. These studies demon­
strated a robust, reproducible progression
associated with the severity of photoreceptor
2 Remodeling degeneration, regardless of initial insult. Retinal
remodeling is phased revision; in phase 1, rod
Because neural systems are interrelated net­ photoreceptors become stressed and extend neu­
works, damage to one cell type or region does rite sprouts beyond their normal synaptic part­
not occur without wider network consequences. ners (rod-contacting horizontal cells (HCs) and
This phenomenon was coined “diaschisis” by rod bipolar cells (RodBCs) [24, 26, 30].
von Monakow in 1914 [13] by describing Simultaneously, RodBCs and HCs begin den­
affected remote regions connected in some way dritic retraction from the rod photoreceptors with
to the region of injury. Today, noninvasive imag­ a subset aberrantly contacting cone photorecep­
ing methods including diffusion tensor imaging tors [31]. In phase 2, photoreceptors (including
(DTI) and functional magnetic resonance imag­ cones) continue to degenerate and undergo cell
ing (fMRI) demonstrate pathological disease death. As photoreceptors degenerate, bipolar
spread following injury (e.g., stroke, seizures, cells (BCs) and HCs continue to lose synaptic
and concussion) and in neurodegenerative dis­ input, with BCs becoming completely deaffer­
ease (e.g., Alzheimer’s disease, Parkinson’s dis­ ented following the complete loss of photorecep­
ease, and Huntington’s disease) [14]. tors [21, 22]. HCs extend processes deep into
Redundancy in parallel neural networks com­ inner retina, though what contacts they make is
pensates for losses to some degree, but the fun­ unknown [21–23]. Also, during phase 2, ama­
damental question is: How much damage can crine cells (ACs) and ganglion cells (GCs) begin
neural networks take before failing? to sprout anomalous neurites that extend outside
Understanding the underlying components con­ of their normal patterns, and make new synapses,
tributing to disease spread and how the affected though up to this point precise network motifs
networks breakdown is fundamental to develop­ were unknown [20, 25, 28]. Phase 3 remodeling
ing therapies, and we propose that retina is an is characterized by a complete absence of photo­
Retinal Pathoconnectomics: A Window into Neurodegeneration 299

receptors. At this point, all cell classes have initi­ 4 Early Findings
ated neurite sprouting, coalescing into synaptic
groupings termed “microneuromas” [25]. Within Analysis of RPC1 began by examining RodBCs
these microneuromas, ultrastructural anatomy is as they are the source of the first synapse in the
consistent with the presence of functional syn­ visual system and function as the interneuron
apses as is evident by the presence of ribbons, connecting sensory to neural retina, via rod syn­
vesicle clouds, and associated post-synaptic apses. As predicted from earlier studies, RodBCs
densities. extend processes toward cone axon terminals.
Combined, these studies demonstrated the However, in RPC1, we not only confirmed the
clear presence of network altering morphologies presence of synaptic contacts from RodBCs onto
and individual synapses, but are insufficient to cone pedicles but also demonstrated some cone
fully describe the precise ways in which the net­ inputs were not from the conventional cone ped­
works as a whole are altered. icle, but rather from secondary terminals found
on sprouts off of cones not normally observed in
healthy retina. We also found these novel con­
3 Pathoconnectomics nections with cones occur prior to complete
and RPC1 deafferentation of RodBCs from rod photorecep­
tors, meaning that at least for part of retinal
Connectomics has demonstrated that complete degeneration, there is mixed input from both
network diagrams are required to understand rods and cones. Next, we evaluated RodBC con­
neural circuitry. A building literature illustrates nections in inner retina and were surprised to
that we cannot guess at topologies, we have to find that although chemical synapse number and
map them [1]. Because networks are altered in partnerships appear unchanged, gap junctions
disease, connectomes of pathological tissue, or emerged between RodBCs and the Aii amacrine
“pathoconnectomes,” are therefore necessary to cell [32]. Gap junctions are electrical synapses
determine how network topologies are changed, coupling many neuronal classes in retina. Within
and rules that underly rewiring. Just as retinas retina, gap junctions convey polarization state
were ideal for constructing the first connectome, between Aii amacrine cells and ON-type cone
they also make an ideal platform for creating and bipolar cells, allowing RodBCs making chemi­
exploring the first pathoconnectomes, retinal cal synapses onto Aii amacrine cells to inject
pathoconnectome 1 (RPC1) [32]. their signals to GCs. The emergence of gap junc­
RPC1 was constructed from a 10-month-old tional coupling directly between RodBCs and
transgenic rabbit model of autosomal dominant Aii amacrine cells is a network corruption caus­
human retinitis pigmentosa, originally created in ing a feedback loop never found in healthy ret­
the Kondo laboratory with a rhodopsin proline ina, likely impacting the duration and timing of
347 to leucine mutation [33]. We previously glutamate release by RodBCs. More altered cir­
detailed progression of degeneration in this rab­ cuit topologies are currently under investigation
bit [34] and found that it progresses through as understanding the complete network and how
cone-sparing retinal degeneration analogous to it fails in retinal degenerative disease is our goal.
that seen in humans [27, 35, 36]. RPC1 was We have continued exploration of the Aii ama­
selected for a reduced photoreceptor layer thick­ crine cell and discovering alterations in synapse
ness and shortened rod outer segments, but is partners and numbers as degeneration pro­
prior to complete rod outer segment loss. This gresses. In addition, RPC1 contains numerous
early time point of retinal degeneration allows copies of ACs and GCs that are demonstrating
the description of partial deafferentation of rod neurite sprouting, indicating this process initi­
connected HCs, RodBCs, and what effects this ates earlier in retinal degeneration than was pre­
has on the downstream neurons in the network. viously predicted.
300 R. L. Pfeiffer and B. W. Jones

5 Future References
of Pathoconnectomics
1. Marc RE, Jones BW, Watt CB, Anderson JR,
Sigulinsky C, Lauritzen S. Retinal connectomics:
The next steps are clear as we continue this work towards complete, accurate networks. Prog Retin Eye
in later stages of retinal degeneration in the next Res. 2013;37:141–62.
two pathoconnectomes produced from retinas in 2. Marc RE, Jones BW, Lauritzen JS, Watt CB,
later stages of degeneration: RPC2 and RPC3. Anderson JR. Building retinal connectomes. Curr
Opin Neurobiol. 2012;22(4):568–74.
These next pathoconnectomes will reveal how 3. Anderson JR, Jones BW, Watt CB, Shaw MV, Yang
the network is altered at more advanced stages of JH, Demill D, et al. Exploring the retinal connectome.
disease progression. In a more global sense, we Mol Vis. 2011;17:355–79.
believe pathoconnectomics as a field will inform 4. Lauritzen JS, Anderson JR, Jones BW, Watt CB,
Mohammed S, Hoang JV, et al. ON cone bipolar cell
the wider neuroscience community not only on axonal synapses in the OFF inner plexiform layer of the
the progression of specific neurological diseases rabbit retina. J Comp Neurol. 2013;521(5):977–1000.
but will also demonstrate specific stereotyped 5. Lauritzen JS, Sigulinsky CL, Anderson JR, Kalloniatis
rules in the way networks change in disease and M, Nelson NT, Emrich DP, et al. Rod-cone crossover
connectome of mammalian bipolar cells. J Comp
will play a crucial role due to the difficulties in Neurol. 2019;527(1):87–116.
doing whole-brain ultrastructural connectomics. 6. Marc RE, Anderson JR, Jones BW, Sigulinsky CL,
We have detailed in previous sections, the Lauritzen JS. The AII amacrine cell connectome: a
advantages of the size, organization, and repeated dense network hub. Front Neural Circuits. 2014;8:104.
7. Marc RE, Sigulinsky CL, Pfeiffer RL, Emrich D,
motifs of the retina lending it to ultrastructural Anderson JR, Jones BW. Heterocellular coupling
connectomics. In addition, our recent work sug­ between Amacrine cells and ganglion cells. Front
gests the retina recapitulates neurodegeneration Neural Circuits. 2018;12:90.
by entering a stage of progressive neurodegener­ 8. Sigulinsky CL, Anderson JR, Kerzner E, Rapp CN,
Pfeiffer RL, Rodman TM, et al. Network architecture
ation following prolonged remodeling, which of gap junctional coupling among parallel process­
appears to only end once all of the neurons of the ing channels in the mammalian retina. J Neurosci.
retina have degenerated [29]. We further find 2020;40(23):4483–511.
similarities between retinal degeneration and 9. Diamond JS. Inhibitory interneurons in the retina:
types, circuitry, and function. Annu Rev Vis Sci.
some of the classic neurodegenerative diseases 2017;3:1–24.
(Parkinson’s disease and Alzheimer’s disease) 10. Thoreson WB, Dacey DM. Diverse cell types, cir­
through alterations in α-synuclein and phosphor­ cuits, and mechanisms for color vision in the verte­
ylated α-synuclein expression [29]. These fea­ brate retina. Physiol Rev. 2019;99(3):1527–73.
11. Patterson SS, Bordt AS, Girresch RJ, Linehan CM,
tures make the retina ideal to observe how Bauss J, Yeo E, et al. Wide-field amacrine cell inputs
proteinopathies spread from neuron to neuron as to ON parasol ganglion cells in macaque retina. J
a potential driving component of neurodegenera­ Comp Neurol. 2020;528(9):1588–98.
tion. Lastly, the detailed network topographies 12. Dunn FA, Wong RO. Wiring patterns in the mouse
retina: collecting evidence across the connec­
already mapped from retina make it unique in our tome, physiology and light microscopy. J Physiol.
ability to identify and track network changes ini­ 2014;592(22):4809–23.
tiated during rewiring, potentially revealing new, 13. Feeney DM, Baron JC. Diaschisis. Stroke.
fundamental mechanisms of neurodegeneration. 1986;17(5):817–30.
14. Fornito A, Zalesky A, Breakspear M. The con­
nectomics of brain disorders. Nat Rev Neurosci.
Acknowledgments This work was supported by the 2015;16(3):159–72.
National Institutes of Health R01 Grant EY015128(BWJ), 15. de Lanerolle NC, Kim JH, Robbins RJ, Spencer
R01 Grant EY02576(BWJ), T32 Grant EY024234 (RLP), DD. Hippocampal interneuron loss and plastic­
P30 Grant EY014800(Core), the National Science ity in human temporal lobe epilepsy. Brain Res.
Foundation Grant (2014862), and an Unrestricted 1989;495(2):387–95.
Research Grant from Research to Prevent Blindness, 16. Morimoto K, Fahnestock M, Racine RJ. Kindling and
New York, NY to the Department of Ophthalmology & status epilepticus models of epilepsy: rewiring the
Visual Sciences, University of Utah. brain. Prog Neurobiol. 2004;73(1):1–60.
Retinal Pathoconnectomics: A Window into Neurodegeneration 301

17. Fisher SK, Lewis GP, Linberg KA, Verardo 27. Jones BW, Kondo M, Terasaki H, Lin Y, McCall M,
MR. Cellular remodeling in mammalian retina: results Marc RE. Retinal remodeling. Jpn J Ophthalmol.
from studies of experimental retinal detachment. Prog 2012;56(4):289–306.
Retin Eye Res. 2005;24(3):395–431. 28. Marc RE, Jones BW. Retinal remodeling in inher­
18. Lewis GP, Fisher SK. Retinal plasticity and interac­ ited photoreceptor degenerations. Mol Neurobiol.
tive cellular remodeling in retinal detachment and 2003;28(2):139–47.
reattachment. In: Pinaud R, Tremere LA, De Weerd P, 29. Pfeiffer RL, Marc RE, Jones BW. Persistent remodel­
editors. Plasticity in the visual system: from genes to ing and neurodegeneration in late-stage retinal degen­
circuits. New York: Springer; 2005. p. 55–78. eration. Prog Retin Eye Res. 2020;74:100771.
19. Lewis GP, Linberg KA, Fisher SK. Neurite outgrowth 30. Sethi CS, Lewis GP, Fisher SK, Leitner WP, Mann
from bipolar and horizontal cells after experimen­ DL, Luthert PJ, et al. Glial remodeling and neural
tal retinal detachment. Invest Ophthalmol Vis Sci. plasticity in human retinal detachment with prolif­
1998;39(2):424–34. erative vitreoretinopathy. Invest Ophthalmol Vis Sci.
20. Marc RE, Jones BW, Watt CB, Strettoi E. Neural 2005;46(1):329–42.
remodeling in retinal degeneration. Prog Retin Eye 31. Peng YW, Hao Y, Petters RM, Wong F. Ectopic syn­
Res. 2003;22(5):607–55. aptogenesis in the mammalian retina caused by rod
21. Strettoi E, Pignatelli V. Modifications of retinal neu­ photoreceptor-specific mutations. Nat Neurosci.
rons in a mouse model of retinitis pigmentosa. Proc 2000;3(11):1121–7.
Natl Acad Sci U S A. 2000;97(20):11020–5. 32. Pfeiffer RL, Anderson JR, Dahal J, Garcia JC, Yang
22. Strettoi E, Pignatelli V, Rossi C, Porciatti V, Falsini JH, Sigulinsky CL, et al. A pathoconnectome of early
B. Remodeling of second-order neurons in the retina neurodegeneration: network changes in retinal degen­
of rd/rd mutant mice. Vis Res. 2003;43(8):867–77. eration. Exp Eye Res. 2020;199:108196.
23. Strettoi E, Porciatti V, Falsini B, Pignatelli V, Rossi 33. Kondo M, Sakai T, Komeima K, Kurimoto Y, Ueno S,
C. Morphological and functional abnormalities Nishizawa Y, et al. Generation of a transgenic rabbit
in the inner retina of the rd/rd mouse. J Neurosci. model of retinal degeneration. Invest Ophthalmol Vis
2002;22(13):5492–504. Sci. 2009;50(3):1371–7.
24. Fariss RN, Li ZY, Milam AH. Abnormalities in rod 34. Jones BW, Kondo M, Terasaki H, Watt CB, Rapp K,
photoreceptors, amacrine cells, and horizontal cells Anderson J, et al. Retinal remodeling in the Tg P347L
in human retinas with retinitis pigmentosa. Am J rabbit, a large-eye model of retinal degeneration. J
Ophthalmol. 2000;129(2):215–23. Comp Neurol. 2011;519(14):2713–33.
25. Jones BW, Watt CB, Frederick JM, Baehr W, Chen 35. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Marmor
CK, Levine EM, et al. Retinal remodeling triggered M, Marc RE. Retinal remodeling in human retinitis
by photoreceptor degenerations. J Comp Neurol. pigmentosa. Exp Eye Res. 2016;150:149–65.
2003;464(1):1–16. 36. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Tucker
26. Li ZY, Kljavin IJ, Milam AH. Rod photoreceptor J, Marc RE. Retinal remodeling and metabolic
neurite sprouting in retinitis pigmentosa. J Neurosci. alterations in human AMD. Front Cell Neurosci.
1995;15(8):5429–38. 2016;10:103.
The Role of Ceramide in Inherited
Retinal Disease Pathology

Xinye Qian, Tanmay Srinivasan, Jessica He,


and Rui Chen

Abstract underlying molecular basis for clinical pheno-


types of retinal dystrophies and provide us
Ceramide (Cer) plays an essential role in pho- with potential therapeutic targets.
toreceptor cell death in the retina. On the one
hand, Cer accumulation emerges as a common Keywords
feature during retina neurodegeneration, lead-
Inherited retinal diseases · Photoreceptor ·
ing to the death of photoreceptors. On the
Ceramide · Ceramide synthase · Retinal
other hand, Cer deficiency has also recently
degeneration · Sphingolipid metabolism ·
been associated with retinal dysfunction and
Apoptosis
degeneration. Although more and more evi-
dence supports the importance of maintaining
Cer homeostasis in the retina, mechanistic
explanations of the observed phenotypes, 1 Introduction
especially in the context of Cer deficiency, are
still lacking. An enhanced understanding of Apoptosis of photoreceptors leads to retinal dys-
Cer’s role in the retina will help us explore the function in inherited retinal diseases (IRDs), a
complex heterogeneous group of diseases that
can cause severe vision loss or even blindness.
X. Qian (*)
Verna and Marrs McLean Department of Although individually rare, retinal degeneration
Biochemistry and Molecular Biology, Baylor College in IRD patients leads to the irreversible death of
of Medicine, Houston, TX, USA photoreceptors. IRDs are genetically heteroge-
Human Genome Sequencing Center, Baylor College neous, with more than 270 disease genes discov-
of Medicine, Houston, TX, USA ered to date [1]. Currently, a variety of therapeutic
e-mail: xinye.qian@bcm.edu approaches to treat IRDs are under development,
T. Srinivasan · J. He with some success of neurotrophic factor therapy
Rice University, Houston, TX, USA and gene therapies reported from clinical trials
e-mail: ts59@rice.edu; jh134@rice.edu
[4–8, 18, 25, 31, 44].
R. Chen For years, many have searched for common
Human Genome Sequencing Center, Baylor College
of Medicine, Houston, TX, USA intracellular second messengers for these many
forms of cell death that can be targeted for ther-
Department of Molecular and Human Genetics,
Baylor College of Medicine, Houston, TX, USA apy. Despite the exciting progress in IRD therapy,
e-mail: ruichen@bcm.edu the heterogeneity of IRDs hampers the develop-

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 303
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_44
304 X. Qian et al.

ment of a common treatment for a large number led to cell death [14], providing additional sup-
of patients, calling for the need for some com- port of CerS’s role as a death mediator in retinal
mon intracellular second messenger, which could cells.
be targeted for therapy. Understanding of which In addition to the discoveries made in vitro,
mediators participate in the induction of photore- in vivo studies also support Cer involvement in
ceptor cell death is still limited. Ceramide (Cer), photoreceptor cell death. The correlation between
a class of sphingolipid which is abundant in cell Cer and death of retinal neurons was first found
membranes, is a bioactive lipid proposed to be a in a Drosophila model [2, 3]. Cers has also been
cellular second messenger that signals for apop- associated with retinal apoptosis following reti-
tosis [22, 23, 36]. This chapter will address the nal detachment in a rabbit model [37]. Increased
role of Cers in the pathogenic pathways of retinal retinal Cer levels accompanied by photoreceptor
degenerative diseases. death were observed in rd10 mice, while photore-
ceptors were rescued by myriocin, a powerful
SPT inhibitor [43]. Intravitreal injection of C2-
2 Ceramide Metabolism Cer causes significant vision loss and blood-­
retinal barrier breakdown in rats [30]. In both the
Cers, as the central molecule of the highly inter- P23H-1 and light-damaged rat models, the inhi-
connected sphingolipid metabolic pathways, bition of Cer synthesis with FTY720, a CerS
forms the backbone of all complex sphingolipids inhibitor, protected photoreceptors from degen-
and is an essential player in key cellular func- eration [13, 42].
tions. There are three different pathways for the Interestingly, beyond the realm of IRDs, reti-
biological generation of Cers. De novo synthesis nal impairment and vision loss accompanied by
pathway takes place in the endoplasmic reticu- Cer accumulation have also been observed in
lum through reactions catalyzed by different patients with several types of ‘lipid storage dis-
enzymes, including serine palmitoyltransferase eases,’ which are characterized by inherited
(SPT), ceramide synthase (CerS), and dihydroc- sphingolipid metabolism defects, such as
eramide desaturase [10, 28, 32]. The sphingomy- Krabbe’s disease [11], Niemann-Pick disease
elinase pathway is carried out through the [38], Sandhoff disease [39], and Gaucher disease
degradation of sphingomyelin by sphingomye- [41].
linase (SMase)​​ [21, 26, 45]. In the salvage path-
way, complex sphingolipids are broken down by
different hydrolases, leading to the recycling of 3 Ceramide Depletion by CerSs
sphingosines and complex sphingolipids [15, Deletion Also Has
35]. Deleterious Effects
on the Retina
Ceramide Accumulation: A Common
Activator in Photoreceptor Degeneration In mammals, de novo Cer synthesis is catalyzed
During the past decade, more and more evidence by six CerSs: CerS1–6 [34]. Each CerS regulates
supports a role for Cer as a mediator of photore- the formation of a specific set of Cers with vari-
ceptor death. In rat retina neuronal culture, exog- able chain lengths and mediates distinct func-
enous addition of C2-Cer triggered photoreceptor tions. There is redundancy in the Cers synthesized
apoptosis, whereas inhibition of Cer synthesis by by each synthase, but no CerS can completely
CerS or SMase inhibitor protected photorecep- compensate for another. CerS1 specifically syn-
tors from oxidative stress-induced apoptosis [19, thesizes C18 Cers [20, 33], while CerS4 prefers
40], suggesting a role of Cer in oxidative stress C20 acyl-CoAs [16]. CerS2 generates longer
induced-photoreceptor cell death. Additionally, chain, C22–24 Cers [24], whereas CerS3 synthe-
treatment of C8-Cer and C16-Cer in 661 W cells sizes Cers with ultra-long-chain lengths, up to
The Role of Ceramide in Inherited Retinal Disease Pathology 305

C36 [27]. CerS5 and CerS6 synthesize C14 to References


C16 Cers [17, 29]. Retinal functions were previ-
ously examined in CerS1-, CerS2-, and CerS4-­ 1. RetNet. Available at: http://www.sph.uth.tmc.edu/
RetNet/ Accessed 9 July 2018.
deficient mice [12]. Although retinal dysfunction 2. Acharya JK, Dasgupta U, Rawat SS, Yuan C,
was detected by electroretinograms, no signifi- Sanxaridis PD, Yonamine I, Karim P, Nagashima K,
cant morphological defects were detected in Brodsky MH, Tsunoda S. Cell-nonautonomous func-
these three mouse models [12]. In addition to the tion of ceramidase in photoreceptor homeostasis.
Neuron. 2008;57:69–79.
canonical CerSs, it has been reported in an 3. Acharya U, Patel S, Koundakjian E, Nagashima K,
in vitro study that the protein encoded by the Han X, Acharya JK. Modulating sphingolipid biosyn-
TLCD3B gene also has CerS activity [46]. thetic pathway rescues photoreceptor degeneration.
TLCD3B is responsible for the synthesis of C16-, Science. 2003;299:1740–3.
4. Bainbridge JW, Smith AJ, Barker SS, Robbie S,
C18-, and C20-Cers in vitro [46] and was Henderson R, Balaggan K, Viswanathan A, Holder
­confirmed by an in vivo study on Tlcd3b−/− mouse GE, Stockman A, Tyler N, Petersen-Jones S,
model (Qian et al. unpublished). Patients with Bhattacharya SS, Thrasher AJ, Fitzke FW, Carter BJ,
TLCD3B mutations are diagnosed with cone-rod Rubin GS, Moore AT, Ali RR. Effect of gene therapy
on visual function in Leber's congenital amaurosis. N
dystrophy or maculopathy. Their phenotypes are Engl J Med. 2008;358:2231–9.
restricted to the retina [9], while 7-month-old 5. Bainbridge JW, et al. Long-term effect of gene ther-
Tlcd3b−/− mice demonstrate both retinal dysfunc- apy on Leber's congenital amaurosis. N Engl J Med.
tion and degeneration [9]. 2015;372:1887–97.
6. Bennett J, et al. Safety and durability of effect of
TLCD3B has been proven to have a much contralateral-­eye administration of AAV2 gene ther-
higher expression level than the other 6 canonical apy in patients with childhood-onset blindness caused
CerSs in the retina [9], which may explain why by RPE65 mutations: a follow-on phase 1 trial.
so far only TLCD3B mutations have been associ- Lancet. 2016;388:661–72.
7. Berson EL, Rosner B, Sandberg MA, Hayes KC,
ated with retinopathies. Nicholson BW, Weigel-DiFranco C, Willett W. A
randomized trial of vitamin A and vitamin E supple-
mentation for retinitis pigmentosa. Arch Ophthalmol.
4 Conclusion 1993;111:761–72.
8. Berson EL, Rosner B, Sandberg MA, Weigel-­
DiFranco C, Moser A, Brockhurst RJ, Hayes KC,
Cer is known to act as a key player in various Johnson CA, Anderson EJ, Gaudio AR, Willett WC,
forms of photoreceptor cell death, and recent Schaefer EJ. Clinical trial of docosahexaenoic acid in
studies also demonstrated the importance of patients with retinitis pigmentosa receiving vitamin A
treatment. Arch Ophthalmol. 2004;122:1297–305.
maintaining the overall Cer homeostasis in the 9. Bertrand RE, Wang J, Xiong KH, Thangavel C, Qian
retina. The discovery of Cer as a key mediator in X, Ba-Abbad R, Liang Q, Simões RT, Sampaio SAM,
photoreceptor degeneration has a significant Carss KJ, Lucy Raymond F, Robson AG, Webster
impact on the development of IRD therapies. As AR, Arno G, Porto FBO, Chen R. Ceramide synthase
TLCD3B is a novel gene associated with human reces-
a common arbitrator in the photoreceptor apopto- sive retinal dystrophy. Genet Med. 2021;23:488–97.
sis and pathogenicity, Cer is a perfect target for 10. Bose R, Verheij M, Haimovitz-Friedman A, Scotto
the development of therapeutic approaches K, Fuks Z, Kolesnick R. Ceramide synthase medi-
designed for IRDs. Nevertheless, Cer metabo- ates daunorubicin-induced apoptosis: an alterna-
tive mechanism for generating death signals. Cell.
lism is inherently complex, and it is highly 1995;82:405–14.
involved with other sphingolipids like sphingo- 11. Brownstein S, Meagher-Villemure K, Polomeno
sine and dihydroceramide, or bioactive Cer RC, Little JM. Optic nerve in globoid leukodystro-
metabolites, such as S1P and C1P. Therefore, phy (Krabbe's disease): ultrastructural changes. Arch
Ophthalmol. 1978;96:864–70.
careful investigations need to be done on other 12. Brüggen B, Kremser C, Bickert A, Ebel P, vom
players in the sphingolipid metabolic pathways Dorp K, Schultz K, Dörmann P, Willecke K, Dedek
to determine how their interactions affect photo- K. Defective ceramide synthases in mice cause
receptor degeneration. reduced amplitudes in electroretinograms and altered
306 X. Qian et al.

sphingolipid composition in retina and cornea. Eur J 25. Jacobson SG, et al. Gene therapy for leber congeni-
Neurosci. 2016;44:1700–13. tal amaurosis caused by RPE65 mutations: safety and
13. Chen H, Tran J-TA, Eckerd A, Huynh T-P, Elliott MH, efficacy in 15 children and adults followed up to 3
Brush RS, Mandal NA. Inhibition of de novo ceramide years. Arch Ophthalmol. 2012;130:9–24.
biosynthesis by FTY720 protects rat retina from light-­ 26. Jarvis WD, Kolesnick RN, Fornari FA, Traylor RS,
induced degeneration. J Lipid Res. 2013;54:1616–29. Gewirtz DA, Grant S. Induction of apoptotic DNA
14. Chen H, Tran J-TA, Brush RS, Saadi A, Rahman damage and cell death by activation of the sphin-
AK, Yu M, Yasumura D, Matthes MT, Ahern K, gomyelin pathway. Proc Natl Acad Sci U S A.
Yang H, LaVail MM, Mandal MNA. Ceramide sig- 1994;91:73–7.
naling in retinal degeneration. Adv Exp Med Biol. 27. Jennemann R, Rabionet M, Gorgas K, Epstein S,
2012;723:553–8. Dalpke A, Rothermel U, Bayerle A, van der Hoeven F,
15. Dbaibo GS, El-Assaad W, Krikorian A, Liu B, Diab Imgrund S, Kirsch J, Nickel W, Willecke K, Riezman
K, Idriss NZ, El-Sabban M, Driscoll TA, Perry DK, H, Gröne HJ, Sandhoff R. Loss of ceramide synthase
Hannun YA. Ceramide generation by two distinct 3 causes lethal skin barrier disruption. Hum Mol
pathways in tumor necrosis factor alpha-induced cell Genet. 2012;21:586–608.
death. FEBS Lett. 2001;503:7–12. 28. Kalén A, Borchardt RA, Bell RM. Elevated ceramide
16. Ebel P, Imgrund S, Vom Dorp K, Hofmann K, Maier levels in GH4C1 cells treated with retinoic acid.
H, Drake H, Degen J, Dörmann P, Eckhardt M, Franz Biochim Biophys Acta. 1992;1125:90–6.
T, Willecke K. Ceramide synthase 4 deficiency in 29. Levy M, Futerman AH. Mammalian ceramide syn-
mice causes lipid alterations in sebum and results in thases. IUBMB Life. 2010;62:347–56.
alopecia. Biochem J. 2014;461:147–58. 30. Lou H, Kang D, Yang Q, Lian C, Zhang C, Li Z, Tian
17. Ebel P, Vom Dorp K, Petrasch-Parwez E, Zlomuzica A, H, Lu L, Xu GT, Xu G, Zhang J. Erythropoietin pro-
Kinugawa K, Mariani J, Minich D, Ginkel C, Welcker tects retina against ceramide 2-induced damage in rat.
J, Degen J, Eckhardt M, Dere E, Dörmann P, Willecke Curr Mol Med. 2017;17:699–706.
K. Inactivation of ceramide synthase 6 in mice results 31. MacLaren RE, Groppe M, Barnard AR, Cottriall
in an altered sphingolipid metabolism and behavioral CL, Tolmachova T, Seymour L, Clark KR, During
abnormalities. J Biol Chem. 2013;288:21433–47. MJ, Cremers FP, Black GC, Lotery AJ, Downes SM,
18. Edwards TL, Jolly JK, Groppe M, Barnard AR, Webster AR, Seabra MC. Retinal gene therapy in
Cottriall CL, Tolmachova T, Black GC, Webster patients with choroideremia: initial findings from a
AR, Lotery AJ, Holder GE, Xue K, Downes SM, phase 1/2 clinical trial. Lancet. 2014;383:1129–37.
Simunovic MP, Seabra MC, MacLaren RE. Visual 32. Mandon EC, Ehses I, Rother J, van Echten G, Sandhoff
acuity after retinal gene therapy for choroideremia. N K. Subcellular localization and membrane topology
Engl J Med. 2016;374:1996–8. of serine palmitoyltransferase, 3-­dehydrosphinganine
19. German OL, Miranda GE, Abrahan CE, Rotstein reductase, and sphinganine N-acyltransferase in
NP. Ceramide is a mediator of apoptosis in ret- mouse liver. J Biol Chem. 1992;267:11144–8.
ina photoreceptors. Invest Ophthalmol Vis Sci. 33. Mizutani Y, Kihara A, Igarashi Y. Mammalian Lass6
2006;47:1658–68. and its related family members regulate synthesis of
20. Ginkel C, Hartmann D, vom Dorp K, Zlomuzica specific ceramides. Biochem J. 2005;390:263–71.
A, Farwanah H, Eckhardt M, Sandhoff R, Degen 34. Mullen TD, Hannun YA, Obeid LM. Ceramide syn-
J, Rabionet M, Dere E, Dörmann P, Sandhoff K, thases at the centre of sphingolipid metabolism and
Willecke K. Ablation of neuronal ceramide synthase biology. Biochem J. 2012;441:789–802.
1 in mice decreases ganglioside levels and expression 35. Novgorodov SA, Wu BX, Gudz TI, Bielawski J,
of myelin-associated glycoprotein in oligodendro- Ovchinnikova TV, Hannun YA, Obeid LM. Novel
cytes. J Biol Chem. 2012;287:41888–902. pathway of ceramide production in mitochondria:
21. Hannun YA. The sphingomyelin cycle and the sec- thioesterase and neutral ceramidase produce ceramide
ond messenger function of ceramide. J Biol Chem. from sphingosine and acyl-CoA. J Biol Chem.
1994;269:3125–8. 2011;286:25352–62.
22. Hannun YA. Functions of ceramide in coordinating cel- 36. Pettus BJ, Chalfant CE, Hannun YA. Ceramide in
lular responses to stress. Science. 1996;274:1855–9. apoptosis: an overview and current perspectives.
23. Hannun YA, Obeid LM. The ceramide-cen- Biochim Biophys Acta. 2002;1585:114–25.
tric universe of lipid-mediated cell regulation: 37. Ranty M-L, Carpentier S, Cournot M, Rico-Lattes
stress encounters of the lipid kind. J Biol Chem. I, Malecaze F, Levade T, Delisle M-B, Quintyn
2002;277:25847–50. J-C. Ceramide production associated with retinal
24. Imgrund S, Hartmann D, Farwanah H, Eckhardt M, apoptosis after retinal detachment. Graefes Arch Clin
Sandhoff R, Degen J, Gieselmann V, Sandhoff K, Exp Ophthalmol. 2009;247:215.
Willecke K. Adult ceramide synthase 2 (CERS2)- 38. Robb RM, Kuwabara T. The ocular pathology of
deficient mice exhibit myelin sheath defects, cerebel- type A Niemann-Pick disease: a light and elec-
lar degeneration, and hepatocarcinomas. J Biol Chem. tron microscopic study. Invest Ophthalmol Vis Sci.
2009;284:33549–60. 1973;12:366–77.
The Role of Ceramide in Inherited Retinal Disease Pathology 307

39. Sango K, Yamanaka S, Ajiki K, Arai N, Takano 44. Weleber RG, Pennesi ME, Wilson DJ, Kaushal
M. Involvement of retinal neurons and pigment epi- S, Erker LR, Jensen L, McBride MT, Flotte TR,
thelial cells in a murine model of sandhoff disease. Humphries M, Calcedo R, Hauswirth WW, Chulay
Ophthalmic Res. 2008;40:241–8. JD, Stout JT. Results at 2 years after gene therapy
40. Sanvicens N, Cotter TG. Ceramide is the key mediator for RPE65-deficient Leber congenital Amaurosis
of oxidative stress-induced apoptosis in retinal photo- and severe early-childhood-onset retinal dystrophy.
receptor cells. J Neurochem. 2006;98:1432–44. Ophthalmology. 2016;123:1606–20.
41. Seidova S-F, Kotliar K, Foerger F, Klopfer M, Lanzl 45. Wiesner DA, Dawson G. Staurosporine induces
I. Functional retinal changes in Gaucher disease. Doc programmed cell death in embryonic neurons and
Ophthalmol. 2009;118:151–4. activation of the ceramide pathway. J Neurochem.
42. Stiles M, Qi H, Sun E, Tan J, Porter H, Allegood 1996;66:1418–25.
J, Chalfant CE, Yasumura D, Matthes MT, LaVail 46. Yamashita-Sugahara Y, Tokuzawa Y, Nakachi Y,
MM. Sphingolipid profile alters in retinal dystrophic Kanesaki-Yatsuka Y, Matsumoto M, Mizuno Y,
P23H-1 rats and systemic FTY720 can delay retinal Okazaki Y. Fam57b (family with sequence similar-
degeneration [S]. J Lipid Res. 2016;57:818–31. ity 57, member B), a novel peroxisome proliferator-­
43. Strettoi E, Gargini C, Novelli E, Sala G, Piano I, activated receptor γ target gene that regulates
Gasco P, Ghidoni R. Inhibition of ceramide biosyn- adipogenesis through ceramide synthesis. J Biol
thesis preserves photoreceptor structure and function Chem. 2013;288:4522–37.
in a mouse model of retinitis pigmentosa. Proc Natl
Acad Sci. 2010;107:18706–11.
Extracellular Matrix:
The Unexplored Aspects of Retinal
Pathologies and Regeneration

Dmitri Serjanov and David R. Hyde

Abstract Membrane · Bruch’s Membrane ·


Interphotoreceptor Matrix · Collagen ·
Nearly a billion people worldwide are affected Laminin · Integrin
by vision-impairing conditions, with retinal
degenerative diseases being a major cause of
blindness. Unfortunately, such diseases are
often permanent and progressive, resulting in Abbreviations
further degeneration and loss of sight, due to
the human retina possessing little, if any, AMD Acute Macular Degeneration
regenerative capacity. Despite numerous BM Basement Membrane
efforts and great progress being made to BrM Bruch’s Membrane
understand the molecular mechanisms of ECM Extracellular Matrix
these diseases and possible therapies, the ILM Inner-Limiting Membrane
majority of investigations focused on cell-­ IPM Interphotoreceptor Matrix
intrinsic factors. However, the microenviron- IRBP Interphotoreceptor Retinoid-Binding
ment surrounding retinal cells throughout Protein
these processes also plays an important role, SM Synaptic Matrix
though our current understanding of its
involvement remains limited. Here we present
a brief overview of the current state of the field 1 Introduction
of extracellular matrix studies within the ret-
ina and its potential roles in retinal diseases The CNS of most vertebrates, including humans,
and potential therapeutic approaches. has very limited regenerative capacity. The ret-
ina, an accessible part of the CNS, is no excep-
Keywords tion. Injuries or degenerative diseases often lead
to impaired retinal function and reduced visual
Extracellular Matrix · Retinal Degeneration ·
ability of the affected person. Unfortunately, reti-
Retinal Regeneration · Inner-Limiting
nal injuries and degenerative diseases leading to
vision loss are permanent and often progressive,
D. Serjanov · D. R. Hyde (*) resulting in further degeneration and loss of sight.
Department of Biological Sciences, University of However, some organisms, such as zebrafish,
Notre Dame, Notre Dame, IN, USA possess the ability to regenerate any retinal
e-mail: dserjano@nd.edu; dhyde@nd.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 309
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_45
310 D. Serjanov and D. R. Hyde

n­ eurons that are lost and restore visual behaviors 2 ECM in the Retina
through the reprogramming of Muller glia and
their asymmetric cell division to produce neuro- In general, the ECM may be divided into two cat-
nal progenitor cells [1]. Elucidating what con- egories based on its location and composition: a
trols Muller glia reprogramming, neuronal collagen and fibronectin-rich interstitial matrix,
progenitor cell production, and neuronal differ- which surrounds the cells; and highly organized
entiation during regeneration may provide BMs, which separate the epithelial cells from the
insights in how to induce a regeneration response stroma and provide a vast range of molecular
in mammals. cues necessary for proper tissue development and
Despite great advances in our understanding maintenance [4]. There are two different base-
of the processes governing retinal regeneration, ment membranes in the retina – the ILM, separat-
the investigations of these mechanisms were con- ing the neural retina from the vitreous, which is
centrated mainly on intrinsic factors, while our mainly composed of multiple laminins, collagens
understanding of the role of the microenviron- type IV and XVIII, perlecan, agrin, and nidogen
ment remains limited. Indeed, characterizing the [5]; and the BrM, underlying the RPE, which is
extracellular matrix (ECM) and its role in retinal mainly composed of laminins, multiple colla-
degenerative diseases and as a possible target for gens, fibronectin, elastin, and endostatin [3]. It
regeneration strategies has been understudied. should also be noted that in humans and other
The ECMs in the eye are abundant and diverse in species with a vascularized retina, the BMs of the
their location, composition, and function. They blood vessels play important structural and bio-
include the basement membranes (BMs), such as molecular roles [6–9]. Despite its name, the
the inner limiting membrane (ILM) and Bruch’s external limiting membrane is not a BM, but
membrane (BrM). Numerous studies demon- rather a confluence of cellular junctions between
strated the importance of ILM and BrM signaling the Müller glia and photoreceptors.
and composition in retinal development and tis-
sue architecture [2, 3]. Non-BM matrices include
the interphotoreceptor matrix (IPM) surrounding 3 Inner-Limiting Membrane
the photoreceptor outer segments and filling the
subretinal space and the synaptic matrices (SM), The ILM plays a crucial role in retinal develop-
which surround the synapses connecting the reti- ment and homeostasis. The incredible complexity
nal cells. Their composition is highly dynamic of the ILM composition and development is high-
and changes dramatically over the course of ret- lighted by the findings that the lens and the ciliary
inogenesis and disease progression. body produce the main components required for
For many years, ECM was only considered for ILM assembly, such as laminins and type IV col-
its structural role in tissue maintenance, but grad- lagen, and secrete them into the vitreous body.
ually, its role in molecular signaling has been rec- The vitreous body acts, at least in part, as a source
ognized and appreciated. In recent years, the of these proteins for ILM assembly during devel-
biochemical aspects of the ECM and its role in opment [10]. The ILM is rapidly synthesized dur-
organizing and polarizing the cytoskeleton, as ing development with dynamic composition
well as guiding its tension forces, were identified. changes, while during postnatal and throughout
Despite the many advances in our understanding adulthood, it is maintained with a slow turnover of
of ECM biology in other tissues, investigations components. Indeed, in humans, the matrix com-
into its involvement in retinal morphogenesis, ponent synthesis declines as early as the first
pathogenesis, and regeneration are severely lag- month after birth and nearly stops by the second
ging behind. The following review aims to give a year of life. Similar observations were made in
brief overview of these structures and their other animal models, such as chick and mouse [5].
involvement in neurodegenerative diseases of the In contrast, the ILM composition remains dynamic
retina. within the proliferative niches in retinas of the
Extracellular Matrix: The Unexplored Aspects of Retinal Pathologies and Regeneration 311

animals capable of retinal regeneration [10]. This Structurally, it is highly elastic and can expand
correlation between the slowing down of the syn- and contract in response to changes in intraocular
thesis and slow turnover of the composition of the pressure (IOP) and choroidal blood volume [20],
retinal matrix and the loss of proliferative and serving as an attachment and migration platform
regenerative capacity of the retinal cells presents for the RPE cells during development and wound
this ECM as a very attractive target for studies to healing [21–25], which anchor to its basal lamina
reverse progressive neurodegenerative diseases, via integrin receptors [26]. Interestingly, the
especially in the context of cell replacement thera- anchoring properties of the membrane change
pies. Unfortunately, there have been very few with age [27], a phenomenon thought to be asso-
studies to these ends thus far. ciated with age-related thickening and calcifica-
The role of the ILM in degenerative retinal tion. Additionally, not all its layers demonstrate
pathologies is currently poorly understood. Other the same attachment properties, with the basal
than suggestions of the ILM involvement in glau- lamina having the highest anchoring properties
coma progression [11], the role of the ILM in [21]. During mechanical or light-induced dam-
retinal pathologies was mainly a result of the age to the RPE, the RPE cells divide and migrate
ILM peeling technique. The idea that the ILM to repair the wounds. This process is more effi-
was simply a scaffold on which glial cells migrate cient in the presence of the basal lamina rather
and proliferate and to create a contractile force than cases where it is damaged or absent [28].
led to the widespread use of this clinical approach For example, these processes are disrupted in
for the treatment and closure of full-thickness AMD patients, presumably due to impaired
macular holes [12, 13]. Since then, it became RPE–BrM interactions [29].
apparent that the ILM serves much more impor- Since BrM is located between the choroid and
tant functions. It is now recognized that ILM the RPE, it also serves as a bidirectional molecu-
peeling results not only in the loss of the struc- lar transport and filtration instrument, servicing
tural support for the Müller glia but also in the the molecular trafficking between the two com-
removal of their endfeet [14]. Furthermore, ILM partments. Given its acellular nature, these pro-
peeling results in serious ultrastructural damage cesses are passive and depend on the hydrostatic
to the retinal surface, contributing to pathological pressure on either side of the membrane, its
conditions such as swelling of the nerve fiber porosity, and molecular composition. From the
layer [15], dissociation of the optic nerve fiber choroid vasculature, RPE receives nutrients, vita-
layer [16], inner retinal dimpling [17], nerve fiber min A, pigment precursors, oxygen, and other
layer thickening [18], macular thinning [19], and components [30–32]. At the same time, the RPE
ganglion cell complex thinning [17]. This grow- exports CO2, water, metabolic waste, and waste
ing body of evidence suggests that the ILM plays products from the shed photoreceptor outer seg-
a crucial role in blinding retinal pathologies; ments through the BrM into the choroid vascula-
however, in-depth assessment and studies into ture [30–33]. Both processes are necessary for
this topic remain lacking. proper function and survival of the RPE and the
visual cycle, and it stands to reason that age-­
related structural changes within the BrM would
4 Bruch’s Membrane affect these functions. Indeed, the water permea-
bility of the BrM changes due to age-related col-
Perhaps the best-studied ECM within the eye is lagen crosslinking and the buildup of lipid and
Bruch’s membrane. Crucial developmental debris [34]. It was noted that the hydraulic per-
importance aside, BrM serves two main func- meability loss progresses exponentially with age
tions – structural support for the RPE cell adhe- and is most pronounced in the macular rather
sion and migration and regulation of molecular than peripheral regions [35–38]. Since these
transportation between the choroid and the RPE. changes detrimentally affect the functionality of
312 D. Serjanov and D. R. Hyde

the RPE, understanding the molecular processes the degradation of ECM components, is expressed
behind them is of great importance. at a much higher level in AMD patient cultures
[52]. Also, the expression of αβ crystallin, another
soluble IPM component whose increased expres-
5 Interphotoreceptor Matrix sion has been detected in early AMD and which
was linked to wet AMD pathogenesis, was found
The interphotoreceptor matrix serves important to be upregulated in AMD patient cultures [53].
functions in mediating the communication Moreover, degradation of the RPE barrier that is
between the photoreceptors, the Müller glia, the associated with oxidative stress has been linked
RPE, and the underlying choroid vasculature to aggregation of αβ crystallin at the basal side of
[39]. Curiously, the IPM itself is quite complex the RPE, promoting drusen formation [53, 54].
and not homogenous in its composition, contain- Clearly, further studies into the role of the IPM in
ing distinct structural regions. The soluble frac- pathogenesis and possible therapeutic IPM-­
tion is mainly composed of the interphotoreceptor targeted therapies are warranted.
retinoid-binding protein (IRBP) with the addition
of a broad repertoire of other components [40,
41]. Most of the soluble IPM components are 6 ECM Composition
secreted by the RPE [42]. Additionally, there are
structurally distinct entities, secreted by the pho- The ECM in the retina is composed of a wide
toreceptors and the Müller glia, such as the cone array of proteins and complex carbohydrates,
matrix, also referred to as cone sheaths, which secreted by cells and organized into complex
surround the cone outer segments, and the rod-­ structures. The main ECM components include
associated matrix [41, 43–46]. The makeup of proteoglycans, such as heparan and chondroitin
these matrices and the interactions between their sulfates, and polysaccharides, such as hyaluronin,
components and the surrounding cells has been various collagens, fibronectin and laminins,
the subject of some studies, but remains largely growth factors, extracellular proteases, and their
ambiguous. regulators [55]. The complexity of studying the
The IPM was noted to be involved in several ECM and its molecular mechanisms is partly due
degenerative diseases. For example, RBP3, to the protein–protein interactions within the
encoding the IRBP protein, and IMPG2, encod- matrix. ECM components do not necessarily
ing the SPACRCAN protein, are associated with interact with cell-surface receptors, but bind
autosomal-recessive retinitis pigmentosa [47, other ECM components, modifying their molec-
48], IMPG1, encoding the SPACR protein, has ular and biochemical properties. Currently, the
been associated with autosomal dominant and role of the ECM and its components is much bet-
recessive forms of Best’s disease [49], TIMP3 is ter understood in development. However, given
associated with Sorby’s fundus dystrophy [50], the current interest in regenerative approaches as
and EFEMP1, encoding the Fibulin 3 protein, a therapy for degenerative diseases, it is impor-
with Doyne honeycomb retinal dystrophy [51]. tant to understand the role of ECM in both these
Though mutations in these genes were found in aspects. As the process of regeneration in lower,
patients with these diseases, the molecular mech- vertebrates recapitulate the process of develop-
anisms by which they are involved remain unclear ment in several ways [56], and it stands to reason
and poorly studied. Recently, numerous studies that the ECM molecules that play a role in retino-
noted the possible involvement of the IPM in genesis would also be involved in tissue repair.
AMD [52]. It was demonstrated that RPE cul- Though there are over 300 components that make
tures from AMD patients exhibit robust changes up the ECM in various tissues, including the eye,
in the expression of these components compared we will briefly describe some of the main struc-
to healthy donors. For example, matrix metallo- tural building blocks and how they interact with
protease 2 (MMP2), a protease responsible for retinal cells.
Extracellular Matrix: The Unexplored Aspects of Retinal Pathologies and Regeneration 313

7 Collagens 9 ECM Receptors

Collagens are one of the most abundant proteins One of the most studied receptor families is the
present in mammals and are a major ECM com- integrins. Their ligands include a wide array of
ponent. As with other matrix proteins, it was ini- ECM molecules, including laminins, collagens,
tially viewed as only a structural molecule, but is and fibronectin [73]. They function as heterodi-
now recognized for its involvement in molecular mers that are composed of an α and a β subunit.
signaling, including cell adhesion, survival, These α-β combinations dictate the ligand speci-
­differentiation, and secreted signaling molecule ficity. Integrin-mediated signaling was shown to
diffusion [4, 57–62]. In the neural retina, colla- be important for a multitude of cellular functions,
gens are found within the ILM at the vitreal sur- including cell attachment, migration, prolifera-
face, basement membranes of the blood vessels, tion, and normal homeostatic function [74–79].
BrM, and in the synaptic layers of the tissue [63]. Though integrins were the subject of numerous
The role of collagens in retinal pathogenesis is studies, there are other receptors interacting with
also becoming recognized. Recent evidence sug- the surrounding microenvironment that have not
gests that collagen abnormalities within the tra- been as extensively studied, especially in the eye.
becular meshwork lead to elevated IOP [64–66] Such receptors include dystroglycan, syndecans,
and pathogenesis of primary open-angle glau- lectins, CD44, lutheran, discoidin domain recep-
coma [64, 67, 68]. Collagens have also been tors, and others.
implicated in ganglion cell survival following
optic nerve transection and ILM removal [69,
70]. Together these findings clearly demonstrate 10 Dynamic Reciprocity
the crucial importance of collagens, though our
understanding of the molecular mechanisms they An additional confounding aspect of matrix biol-
govern in the retina remains unclear. ogy is known as the concept of dynamic reciproc-
ity, describing a combination of outside-in and
inside-out signaling cascades, which continu-
8 Laminins ously modify each other [80–92]. In short, the
cells create the ECM with which they interact,
Laminins are a major ECM component and a which in turn governs the behavior and gene
critical building block of the basement mem- expression of the cells, which in turn modify the
brane. Indeed, without laminins, no basement composition of the matrix around them. This
membrane can persist. There are 11 mammalian concept was best demonstrated and studied in the
laminin subunits, 5-α, 3-β, and 3-γ, which com- mammary epithelium. Culturing mammary epi-
bine into 16 distinct heterotrimers composed of thelial cells on 3D laminin-rich matrices results
one α, one β, and one γ chains. Each laminin tri- in actin cytoskeleton polarization configuration
mer possesses distinct biological properties based similar to that observed in vivo, as opposed to the
on the composition and molecular interactions stress fiber formation in conventional 2D sys-
between the subunits [71]. The importance of tems. Inhibition of actin polymerization leads to
laminins and their potential therapeutic use was changes in cell shape and histone deacetylation,
clearly demonstrated in organoid cultures. While suggesting that ECM-cytoskeleton signaling reg-
the effort to grow retinal organoid has been ongo- ulates chromosome organization and gene
ing for some time, it was not until it was discov- expression as a result [83]. Additionally, manipu-
ered that the addition of laminins to the culture lating ECM stiffness in vitro changes gene
medium resulted in consistent laminated retina expression and cell fate. Mesenchymal stem cells
formation [72]. This highlights the potential for grown on matrices with stiffness comparable to
the use of laminins in cell replacement therapies that of the brain, muscle, or bone leads to differ-
and tissue engineering. entiation into respective neuronal, myoblastic, or
314 D. Serjanov and D. R. Hyde

osteoblastic cells [84]. Yet another example of 6. Roy S, Kim D. Retinal capillary basement membrane
thickening: role in the pathogenesis of diabetic reti-
dynamic reciprocity was observed in cancer. nopathy. Prog Retin Eye Res. 2021 May;82:100903.
Normally, the ECM alone is sufficient to inhibit 7. Trost A, Bruckner D, Rivera FJ, Reitsamer
tumorigenesis [87]. Conversely, misexpression of HA. Pericytes in the retina. Adv Exp Med Biol.
ECM genes such as collagen IV creates microen- 2019;1122:1–26.
8. Edwards MM, Lefebvre O. Laminins and reti-
vironment that is permissive to tumor metastasis nal vascular development. Cell Adhes Migr. 2013
in the liver [89, 90]. The studies of dynamic reci- Jan;7(1):82–9.
procity in the eye, however, have proven 9. Miller CG, Budoff G, Prenner JL, Schwarzbauer
extremely difficult to do, and our understanding JE. Minireview: fibronectin in retinal disease. Exp
Biol Med (Maywood). 2017 Jan;242(1):1–7.
of these processes in the eye remains essentially 10. Halfter W, Dong S, Dong A, Eller AW, Nischt
nonexistent. R. Origin and turnover of ECM proteins from the
inner limiting membrane and vitreous body. Eye
(Lond). 2008 Oct;22(10):1207–13.
11. Prager AJ, Blumberg D, Zeng Q, Chang S, Tezel
11 Concluding Remarks TH. Internal limiting membrane Peel is associated
and Future Perspectives with increased ganglion cell loss and poor visual
outcome in patients with primary open angle glau-
Here we reviewed only a small fraction of the coma. Invest Ophthalmol Vis Sci. The Association
for Research in Vision and Ophthalmology; 2015 Jun
retinal ECMs, their components, and how they 11;56(7):4571–1.
interact with retinal cells. Despite all the evi- 12. Gass JDM. Idiopathic senile macular hole: its early
dence gathered in other systems, the ECM stages and pathogenesis. Arch Ophthalmol. American
remains largely unexplored in the eye and is Medical Association. 1988 May 1;106(5):629–39.
13. Kelly NE, Wendel RT. Vitreous surgery for idio-
almost entirely overlooked when it comes to pathic macular holes: results of a pilot study. Arch
developing regenerative strategies. It is abun- Ophthalmol. American Medical Association. 1991
dantly clear at this point that understanding ECM May 1;109(5):654–9.
biology in the context of retinal development and 14. Wolf S, Schnurbusch U, Wiedemann P, Grosche J,
Reichenbach A, Wolburg H. Peeling of the basal
pathogenesis is crucial to the development of membrane in the human retina: ultrastructural effects.
therapeutics that would allow us to prevent, halt, Ophthalmology. 2004 Feb;111(2):238–43.
or even reverse degenerative conditions. 15. Clark A, Balducci N, Pichi F, Veronese C, Morara
M, Torrazza C, et al. Swelling of the arcuate nerve
fiber layer after internal limiting membrane peeling.
Retina. 2012 Sep;32(8):1608–13.
References 16. Tadayoni R, Paques M, Massin P, Mouki-Benani S,
Mikol J, Gaudric A. Dissociated optic nerve fiber
1. Nagashima M, Barthel LK, Raymond PA. A self-­ layer appearance of the fundus after idiopathic
renewing division of zebrafish Müller glial cells gen- epiretinal membrane removal. Ophthalmology. 2001
erates neuronal progenitors that require N-cadherin Dec;108(12):2279–83.
to regenerate retinal neurons. Development. 2013 17. Spaide RF. “Dissociated optic nerve fiber layer
Nov;140(22):4510–21. appearance” after internal limiting membrane
2. Zhang KY, Johnson TV. The internal limiting mem- removal is inner retinal dimpling. Retina. 2012
brane: roles in retinal development and implications Oct;32(9):1719–26.
for emerging ocular therapies. Exp Eye Res. 2021 18. Yamashita T, Uemura A, Kita H, Sakamoto
May;206:108545. T. Analysis of the retinal nerve fiber layer after indo-
3. Booij JC, Baas DC, Beisekeeva J, Gorgels TGMF, cyanine green-assisted vitrectomy for idiopathic mac-
Bergen AAB. The dynamic nature of Bruch’s mem- ular holes. Ophthalmology. 2006 Feb;113(2):280–4.
brane. Prog Retin Eye Res. 2010 Jan;29(1):1–18. 19. Treumer F, Wacker N, Junge O, Hedderich J, Roider
4. Yurchenco PD. Basement membranes: cell scaf- J, Hillenkamp J. Foveal structure and thickness of
foldings and signaling platforms. Cold Spring Harb retinal layers long-term after surgical peeling of
Perspect Biol. 2011 Feb 1;3(2):a004911–1. idiopathic epiretinal membrane. Invest Ophthalmol
5. Halfter W, Dong S, Schurer B, Ring C, Cole GJ, Eller Vis Sci. The Association for Research in Vision and
A. Embryonic synthesis of the inner limiting mem- Ophthalmology. 2011 Feb 9;52(2):744–50.
brane and vitreous body. Investigative Ophthalmology 20. Johnson M, Curcio CA. Structure, function and
& Visual Science. The Association for Research in pathology of Bruch’s membrane. In: Retina. Elsevier;
Vision and Ophthalmology. 2005 Jun;46(6):2202–9. 2017. pp. 522–543.
Extracellular Matrix: The Unexplored Aspects of Retinal Pathologies and Regeneration 315

21. Del Priore LV, Tezel TH. Reattachment rate of human 36. Starita C, Hussain AA, Pagliarini S, Marshall
retinal pigment epithelium to layers of human Bruch’s J. Hydrodynamics of ageing Bruch’s membrane:
membrane. Arch Ophthalmol American Medical implications for macular disease. Exp Eye Res. 1996
Association. 1998 Mar;116(3):335–41. May;62(5):565–72.
22. Del Priore LV, Geng L, Tezel TH, Kaplan 37. Starita C, Hussain AA, Patmore A, Marshall
HJ. Extracellular matrix ligands promote RPE J. Localization of the site of major resistance to fluid
attachment to inner Bruch’s membrane. Current Eye transport in Bruch’s membrane. Invest Ophthalmol
Research. Taylor & Francis; 2009 July 2. Vis Sci. 1997 Mar;38(3):762–7.
23. Gong J, Sagiv O, Cai H, Tsang SH, Del Priore 38. Hillenkamp J, Hussain AA, Jackson TL, Cunningham
LV. Effects of extracellular matrix and neighboring JR, Marshall J. The influence of path length and
cells on induction of human embryonic stem cells into matrix components on ageing characteristics of trans-
retinal or retinal pigment epithelial progenitors. Exp port between the choroid and the outer retina. Invest
Eye Res. 2008 Jun;86(6):957–65. Ophthalmol Vis Sci. 2004 May;45(5):1493–8.
24. Tezel TH, Del Priore LV. Repopulation of different 39. Ishikawa M, Sawada Y, Yoshitomi T. Structure and
layers of host human Bruch’s membrane by retinal function of the interphotoreceptor matrix surround-
pigment epithelial cell grafts. Invest Ophthalmol ing retinal photoreceptor cells. Exp Eye Res. 2015
Vis Sci. The Association for Research in Vision and Apr;133:3–18.
Ophthalmology. 1999 Mar 1;40(3):767–74. 40. Bridges CD. The interphotoreceptor matrix--func-
25. Tezel TH, Del Priore LV, Kaplan HJ. Reengineering tions and possible role in hereditary retinal degenera-
of aged Bruch’s membrane to enhance retinal pig- tions. Prog Clin Biol Res. 1985;190:195–212.
ment epithelium repopulation. Invest Ophthalmol 41. Hageman GS, Johnson LV. Chapter 9 Structure, com-
Vis Sci. The Association for Research in Vision and position and function of the retinal interphotoreceptor
Ophthalmology. 2004 Sep 1;45(9):3337–48. matrix. Progress in Retinal Research Pergamon. 1991
26. Fang I-M, Yang C-H, Yang C-M, Chen Jan 1;10:207–49.
M-S. Overexpression of integrin alpha6 and beta4 42. Kay P, Yang YC, Paraoan L. Directional protein secre-
enhances adhesion and proliferation of human retinal tion by the retinal pigment epithelium: roles in retinal
pigment epithelial cells on layers of porcine Bruch’s health and the development of age-related macular
membrane. Exp Eye Res. 2009 Jan;88(1):12–21. degeneration. J Cell Mol Med. 2013 Jul;17(7):833–43.
27. Gullapalli VK, Sugino IK, Van Patten Y, Shah S, 43. Acharya S, Foletta VC, Lee JW, Rayborn ME,
Zarbin MA. Impaired RPE survival on aged submac- Rodriguez IR, Young WS, et al. SPACRCAN, a novel
ular human Bruch’s membrane. Exp Eye Res. 2005 human interphotoreceptor matrix hyaluronan-binding
Feb;80(2):235–48. proteoglycan synthesized by photoreceptors and pine-
28. Wang H, Ninomiya Y, Sugino IK, Zarbin MA. Retinal alocytes. J Biol Chem. 2000 Mar 10;275(10):6945–55.
pigment epithelium wound healing in human Bruch’s 44. Steinberg RH, Wood I, Hogan MJ. Pigment epithelial
membrane explants. Investigative Ophthalmology & ensheathment and phagocytosis of extrafoveal cones
Visual Science The Association for Research in Vision in human retina. Philos Trans R Soc Lond Ser B Biol
and Ophthalmology. 2003 May;44(5):2199–210. Sci. 1977 Mar 28;277(958):459–74.
29. Binder S, Stanzel BV, Krebs I, Glittenberg 45. Anderson DH, Fisher SK. The relationship of primate
C. Transplantation of the RPE in AMD. Prog Retin foveal cones to the pigment epithelium. J Ultrastruct
Eye Res. 2007 Sep;26(5):516–54. Res. 1979 Apr;67(1):23–32.
30. Hewitt AT, Nakazawa K, Newsome DA. Analysis 46. Hollyfield JG, Rayborn ME, Landers RA. A tech-
of newly synthesized Bruch’s membrane proteogly- nique for isolation of the photoreceptor layer from
cans. Invest Ophthalmol Vis Sci. The Association for other neurons in the human retina. Exp Eye Res. 1990
Research in Vision and Ophthalmology. 1989 Mar Mar;50(3):335–8.
1;30(3):478–86. 47. Hollander den AI, McGee TL, Ziviello C, Banfi S,
31. Bok D. The retinal pigment epithelium: a versatile Dryja TP, Gonzalez-Fernandez F, et al. A homozy-
partner in vision. J Cell Sci Suppl. 1993;17:189–95. gous missense mutation in the IRBP gene (RBP3)
32. Strauss O. The retinal pigment epithelium in associated with autosomal recessive retinitis pig-
visual function. Physiological Reviews American mentosa. Invest Ophthalmol Vis Sci. The Association
Physiological Society. 2005 Jul;85(3):845–81. for Research in Vision and Ophthalmology; 2009
33. Wimmers S, Karl MO, Strauss O. Ion channels in the Apr;50(4):1864–72.
RPE. Prog Retin Eye Res. 2007 May;26(3):263–301. 48. Bandah-Rozenfeld D, Collin RWJ, Banin E, van den
34. Huang J-D, Presley JB, Chimento MF, Curcio CA, Born LI, Coene KLM, Siemiatkowska AM, et al.
Johnson M. Age-related changes in human macular Mutations in IMPG2, encoding interphotoreceptor
Bruch’s membrane as seen by quick-freeze/deep-etch. matrix proteoglycan 2, cause autosomal-recessive
Exp Eye Res. 2007 Aug;85(2):202–18. retinitis pigmentosa. Am J Hum Genet. 2010 Aug
35. Moore DJ, Hussain AA, Marshall J. Age-related 13;87(2):199–208.
variation in the hydraulic conductivity of Bruch’s 49. Manes G, Meunier I, Avila-Fernández A, Banfi S,
membrane. Invest Ophthalmol Vis Sci. 1995 Le Meur G, Zanlonghi X, et al. Mutations in IMPG1
Jun;36(7):1290–7.
316 D. Serjanov and D. R. Hyde

cause vitelliform macular dystrophies. Am J Hum 63. Ponsioen TL, van Luyn MJA, van der Worp RJ, van
Genet. 2013 Sep 5;93(3):571–8. Meurs JC, Hooymans JMM, Los LI. Collagen dis-
50. Weber BH, Vogt G, Pruett RC, Stöhr H, Felbor tribution in the human vitreoretinal interface. Invest
U. Mutations in the tissue inhibitor of metalloprotein- Ophthalmol Vis Sci. The Association for Research in
ases-­3 (TIMP3) in patients with Sorsby’s fundus dys- Vision and Ophthalmology. 2008 Sep;49(9):4089–95.
trophy. Nat Genet. 1994 Dec;8(4):352–6. 64. Last JA, Pan T, Ding Y, Reilly CM, Keller K, Acott TS,
51. Marmorstein LY, Munier FL, Arsenijevic Y, Schorderet et al. Elastic modulus determination of normal and
DF, McLaughlin PJ, Chung D, et al. Aberrant accu- glaucomatous human trabecular meshwork. Invest
mulation of EFEMP1 underlies drusen formation in Ophthalmol Vis Sci. The Association for Research in
Malattia Leventinese and age-related macular degen- Vision and Ophthalmology. 2011 Apr;52(5):2147–52.
eration. Proc Natl Acad Sci USA. National Academy 65. Lütjen-Drecoll E. Functional morphology of the tra-
of Sciences; 2002 Oct 1;99(20):13067–72. becular meshwork in primate eyes. Prog Retin Eye
52. An E, Lu X, Flippin J, Devaney JM, Halligan B, Res. 1999 Jan;18(1):91–119.
Hoffman EP, et al. Secreted proteome profiling in 66. Fuchshofer R, Tamm ER. Modulation of extracellular
human RPE cell cultures derived from donors with age matrix turnover in the trabecular meshwork. Exp Eye
related macular degeneration and age matched healthy Res. 2009 Apr;88(4):683–8.
donors. J Proteome Res. 2006 Oct;5(10):2599–610. 67. Aihara M, Lindsey JD, Weinreb RN. Ocular hyper-
53. De S, Rabin DM, Salero E, Lederman PL, Temple tension in mice with a targeted type I collagen
S, Stern JH. Human retinal pigment epithelium cell mutation. Investigative Ophthalmology & Visual
changes and expression of alphaB-crystallin: a bio- Science. The Association for Research in Vision and
marker for retinal pigment epithelium cell change in Ophthalmology. 2003 Apr;44(4):1581–5.
age-related macular degeneration. Arch Ophthalmol. 68. Tektas O-Y, Lütjen-Drecoll E. Structural changes of
2007 May;125(5):641–5. the trabecular meshwork in different kinds of glau-
54. Sreekumar PG, Kannan R, Kitamura M, Spee C, coma. Exp Eye Res. 2009 Apr;88(4):769–75.
Barron E, Ryan SJ, et al. αB crystallin is apically 69. Rosario CM, Fry KR, Madison R. Rabbit retinal gan-
secreted within exosomes by polarized human retinal glion cells survive optic nerve transection and entubu-
pigment epithelium and provides neuroprotection to lation repair with type I collagen nerve guide tubes.
adjacent cells. PLoS One. 2010 Oct 8;5(10):e12578. Restor Neurol Neurosci. 1989 Jan 1;1(1):31–7.
55. Al-Ubaidi MR, Naash MI, Conley SM. A perspective 70. Halfter W, Willem M, Mayer U. Basement
on the role of the extracellular matrix in progressive membrane-­ dependent survival of retinal gan-
retinal degenerative disorders. Invest Ophthalmol Vis glion cells. Investigative Ophthalmology & Visual
Sci. 2013 Dec 17;54(13):8119–24. Science. The Association for Research in Vision and
56. Ahmad I, Teotia P, Erickson H, Xia X. Recapitulating Ophthalmology. 2005 Mar;46(3):1000–9.
developmental mechanisms for retinal regeneration. 71. Domogatskaya A, Rodin S, Tryggvason K. Functional
Prog Retin Eye Res. 2020 May;76:100824. diversity of laminins. Annu Rev Cell Dev Biol.
57. Yurchenco PD, Ruben GC. Basement membrane 2012;28:523–53.
structure in situ: evidence for lateral associations 72. Eiraku M, Takata N, Ishibashi H, Kawada M,
in the type IV collagen network. J Cell Biol. 1987 Sakakura E, Okuda S, et al. Self-organizing optic-cup
Dec;105(6 Pt 1):2559–68. morphogenesis in three-dimensional culture. Nat Publ
58. Lampe AK, Bushby KMD. Collagen VI related mus- Group. 2011 Apr 7;472(7341):51–6.
cle disorders. J Med Genet. 2005 Sep;42(9):673–85. 73. Humphries JD, Byron A, Humphries MJ. Integrin
59. Pöschl E, Schlötzer-Schrehardt U, Brachvogel B, ligands at a glance. J Cell Sci. 2006 Oct 1;119(Pt
Saito K, Ninomiya Y, Mayer U. Collagen IV is essen- 19):3901–3.
tial for basement membrane stability but dispensable 74. Hynes RO. Integrins: bidirectional, allosteric signal-
for initiation of its assembly during early develop- ing machines. Cell. 2002 Sep 20;110(6):673–87.
ment. Development. 2004 Apr;131(7):1619–28. 75. Aszodi A, Hunziker EB, Brakebusch C, Fässler
60. Claudepierre T, Manglapus MK, Marengi N, R. Beta1 integrins regulate chondrocyte rotation, G1
Radner S, Champliaud M-F, Tasanen K, et al. progression, and cytokinesis. Genes & Development.
Collagen XVII and BPAG1 expression in the Cold Spring Harbor Lab. 2003 Oct 1;17(19):2465–79.
retina: evidence for an anchoring complex in the 76. Gardiner NJ. Integrins and the extracellular matrix:
central nervous system. J Comp Neurol. 2005 Jun key mediators of development and regeneration of
27;487(2):190–203. the sensory nervous system. Prokop A, Reichardt LF,
61. Bhattacharya G, Kalluri R, Orten DJ, Kimberling WJ, editors. Dev Neurobiol. 2011 Oct 11;71(11):1054–72.
Cosgrove D. A domain-specific usherin/collagen IV 77. Lee JL, Streuli CH. Integrins and epithelial cell polar-
interaction may be required for stable integration into ity. J Cell Sci. 2014.
the basement membrane superstructure. J Cell Sci. 78. Li M, Sakaguchi DS. Inhibition of integrin-mediated
2004 Jan 15;117(Pt 2):233–42. adhesion and signaling disrupts retinal development.
62. Timpl R, Wiedemann H, van Delden V, Furthmayr H, Dev Biol. 2004 Nov 1;275(1):202–14.
Kühn K. A network model for the organization of type 79. Manninen A. Epithelial polarity--generating and inte-
IV collagen molecules in basement membranes. Eur J grating signals from the ECM with integrins. Exp Cell
Biochem. 1981 Nov;120(2):203–11. Res. 2015 Jun 10;334(2):337–49.
Extracellular Matrix: The Unexplored Aspects of Retinal Pathologies and Regeneration 317

80. Hetzer MW, Walther TC, Mattaj IW. Pushing the (ADAMTS) superfamily: functions and mechanisms.
envelope: structure, function, and dynamics of J Biol Chem. 2009 Nov 13;284(46):31493–7.
the nuclear periphery. Annu Rev Cell Dev Biol. 87. Bissell MJ, Hines WC. Why don’t we get more
2005;21(1):347–80. cancer? A proposed role of the microenvironment
81. Roskelley CD, Bissell MJ. Dynamic reciprocity revis- in restraining cancer progression. Nat Med. 2011
ited: a continuous, bidirectional flow of information Mar;17(3):320–9.
between cells and the extracellular matrix regulates 88. Tian X, Azpurua J, Hine C, Vaidya A, Myakishev-­
mammary epithelial cell function. Biochem Cell Biol. Rempel M, Ablaeva J, et al. High-molecular-­
1995 Jul;73(7–8):391–7. mass hyaluronan mediates the cancer resistance
82. Maniotis AJ, Chen CS, Ingber DE. Demonstration of the naked mole rat. Nat Publ Group. 2013 Jul
of mechanical connections between integrins, cyto- 18;499(7458):346–9.
skeletal filaments, and nucleoplasm that stabilize 89. Burnier JV, Wang N, Michel RP, Hassanain M, Li S,
nuclear structure. Proc Natl Acad Sci U S A. 1997 Feb Lu Y, et al. Type IV collagen-initiated signals provide
4;94(3):849–54. survival and growth cues required for liver metastasis.
83. Le Beyec J, Xu R, Lee S-Y, Nelson CM, Rizki A, Oncogene. 2011 Sep 1;30(35):3766–83.
Alcaraz J, et al. Cell shape regulates global histone 90. Paszek MJ, Zahir N, Johnson KR, Lakins JN,
acetylation in human mammary epithelial cells. Exp Rozenberg GI, Gefen A, et al. Tensional homeosta-
Cell Res. 2007 Aug 15;313(14):3066–75. sis and the malignant phenotype. Cancer Cell. 2005
84. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix Sep;8(3):241–54.
elasticity directs stem cell lineage specification. Cell. 91. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M,
2006 Aug;126(4):677–89. Erler JT, et al. Matrix crosslinking forces tumor pro-
85. Kheradmand F, Werner E, Tremble P, Symons M, gression by enhancing integrin signaling. Cell. 2009
Werb Z. Role of Rac1 and oxygen radicals in colla- Nov 25;139(5):891–906.
genase-­1 expression induced by cell shape change. 92. Bergamaschi A, Tagliabue E, Sørlie T, Naume B,
Science. 1998 May 8;280(5365):898–902. Triulzi T, Orlandi R, et al. Extracellular matrix signa-
86. Apte SS. A disintegrin-like and metalloprotease ture identifies breast cancer subgroups with different
(reprolysin-type) with thrombospondin type 1 motif clinical outcome. J Pathol. 2008 Feb;214(3):357–67.
Role of TFEB in Diseases
Associated with Lysosomal
Dysfunction

Hsuan-Yeh Pan and Mallika Valapala

Abstract Keywords

Transcription factor EB (TFEB) · Lysosomal


Transcription factor EB (TFEB) plays a very
dysfunction · Retinal pigment epithelium
important role in the maintenance of cellular
(RPE) · Age-related macular degeneration
homeostasis. TFEB is a transcription factor
(AMD) · Neurodegeneration · Reactive
that regulates the expression of several genes
oxygen species (ROS)
in the Coordinated Lysosomal Expression and
Regulation (CLEAR) network. The CLEAR
network genes are known to regulate many
processes associated with the autophagy path-
Abbreviations
way and lysosome biogenesis. Lysosomes,
which are degradative organelles in the cell,
AKT Ak strain transforming protein/­
are associated with several cellular mecha-
protein kinase B
nisms, such as autophagy and phagocytosis.
ALS Amyotrophic lateral sclerosis
Recent studies have shown that TFEB dysreg-
AMD Age-related macular degeneration
ulation and lysosomal dysfunction are associ-
CLEAR Coordinated Lysosomal Expression
ated with several degenerative diseases. Thus,
and Regulation
enhancing TFEB activity and accompanied
CTNS Cystinosin
induction of lysosomal function and autoph-
GBA Glucocerebrosidase
agy can have tremendous therapeutic potential
GCase Glucocerebrosidase enzyme
for the treatment of several degenerative dis-
GSK3β Glycogen synthase kinase-3β
eases including age-related macular degenera-
MCOLN1 Mucolipin TRP cation channel 1
tion (AMD). In this chapter, we briefly
mTORC1 Mammalian target of rapamycin
illustrate the expression and regulation of
(mTOR) complex 1
TFEB in response to several cellular stressors
POS shed outer photoreceptor segments
and discuss the effects of TFEB overexpres-
ROS Reactive oxygen species
sion to induce cellular clearance functions.
TFEB Transcription factor EB
TGF-β1 Transforming growth factor β1
H.-Y. Pan · M. Valapala (*)
School of Optometry, Indiana University,
Bloomington, IN, USA
e-mail: hsupan@iu.edu; mvalapal@iu.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 319
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_46
320 H.-Y. Pan and M. Valapala

1 Introduction results in nuclear translocation of TFEB [12–14].


Inhibition of mTORC1 via mTORC1 inhibitor
Transcriptional factor EB (TFEB) is a helix-loop-­ activates Mucolipin-1 (MCOLN1), a calcium
helix leucine zipper transcription factor belong- channel on lysosome, leading to the release of
ing to microphthalmia-associated transcription calcium from the lysosomes [15]. Increase in cel-
factor/transcription factor E (MiTF/TFE) family lular levels of calcium in cytoplasm results in the
[1, 2]. TFEB expression and activity is regulated activation of a calcium-regulated phosphatase,
by phosphorylation and dephosphorylation calcineurin. Calcineurin further dephosphory-
events at multiple sites on the protein [2, 3]. It lates the S142 and S211 sites in TFEB and pro-
was previously known that phosphorylation of motes nuclear localization of TFEB to trigger
TFEB at two residues, serine 142 (S142) and ser- lysosomal biogenesis and autophagy [13].
ine 211 (S211), by mammalian target of rapamy- Subcellular distribution of TFEB is also regu-
cin complex 1 (mTORC1) influences its nuclear lated by the AKT signaling pathway. TFEB at
localization and activity [2]. In addition to these S467 is phosphorylated by AKT, a serine/threo-
sites, there are other sites that regulate TFEB by nine kinase, further inhibiting TFEB nuclear
phosphorylation, AKT phosphorylates TFEB at translocation [16].
serine 467 (S467), and GSK3β phosphorylates
TFEB at serine 134/138 (S134/S138) [2, 3]. The
regulation of TFEB cytosolic-nucleus transloca- 2 Involvement of Lysosomal
tion is key to manipulate the expression of TFEB-­ Dysfunction
regulated genes and associated cellular functions in the Pathogenesis
[4]. TFEB activation is induced by multiple cell of Several Diseases
stress factors such as nutrient and growth factor
deprivation and mitochondrial stress [5–7]. TFEB Lysosomal dysfunction is implicated in the
has been identified as a master regulator of lyso- pathogenesis of several degenerative diseases
somal function and autophagy, which is an inter- including Parkinson’s disease and amyotrophic
cellular process that promotes degradation of lateral sclerosis [3, 17, 18]. Misfolded
dysfunctional organelles and long-lived proteins α-synuclein, a neuronal protein involved in
and recycles these materials into nutrients during Parkinson’s disease is known to cause an impair-
starvation [3]. TFEB remains in cytoplasm, and ment of the autophagy-lysosomal pathway [19].
autophagy operates at a basal level when cells Studies have shown that loss of activity of the
grow in a nutrient-rich environment [8, 9]. lysosomal enzyme, glucocerebrosidase (GCase),
However, when cells undergo stress, TFEB trans- causes accumulation of α-Synuclein [20, 21]. In
locates to nucleus to enhance autophagy and lys- addition, lysosomal dysfunction is also impli-
osome biogenesis [9, 10]. TFEB regulates cated in the pathogenesis of amyotrophic lateral
autophagy via binding to the promoter regions of sclerosis (ALS) [18]. The presence of GGGGCC
genes in the Coordinated Lysosomal Expression (G4C2) hexanucleotide repeat expansion (HRE)
and Regulation (CLEAR) network to promote in chromosome nine open reading frame 72
autophagy initiation, autophagosome formation, (C9orf72) results in a genetic predisposition to
autophagosome and lysosome fusion, and lyso- ALS [22, 23]. It was reported that C9orf72-HRE
some biogenesis [3, 11]. Subcellular localization interferes with TFEB nuclear transport resulting
of TFEB is regulated by multiple intracellular in TFEB cytosolic retention and failure of lyso-
signaling mediators, such as mTOR and AKT somal acidification [18].
signaling pathways [3]. Subcellular distribution Lysosomal and mitochondrial dysfunction is
and activation of TFEB are regulated by also implicated in the pathogenesis of several dis-
mTORC1; activated mTORC1 phosphorylates eases of the kidney, including diabetic nephropa-
TFEB resulting in cytosolic retention of TFEB. thy and cisplatin-induced acute kidney injury
Nutrient stress-mediated inhibition of mTORC1 [24, 25]. Transforming growth factor β1 (TGF-β1)
Role of TFEB in Diseases Associated with Lysosomal Dysfunction 321

has been shown to be an essential regulator of downregulation of cathepsins, lysosomal basifi-


diabetic nephropathy [24]. TGF-β1 induces phos- cation, and impairment of autophagy [39].
phorylation of mothers against decapentaplegic
homolog 3 (SMAD3), which results in lysosome
depletion and inhibition of lysosome biogenesis 3 Lysosomes and Its
in a TFEB-dependent manner [24]. Studies have Importance in the Proper
shown that cisplatin, an anticancer agent, can Functioning of the RPE
cause mitochondrial dysfunction [25]. Induction
of TFEB expression by overexpression of peroxi- The retinal pigment epithelium (RPE) is a signal
some Peroxisome proliferator-activated receptor layer of cells located at the outer layer of the ret-
gamma coactivator 1-alpha (PGC-1α) can protect ina, between the neuroretina and the choroid.
against mitochondrial dysfunction in cisplatin-­ RPE has multiple functions in the eye; one of the
treated HK2 cells [25]. Trehalose treatment also main functions is to maintain photoreceptor
alleviates mitochondrial dysfunction and cispla- hemostasis by phagocytizing the shed photore-
tin-induced acute kidney injury via induction of ceptor outer segments [40]. RPE also plays an
TFEB-mediated autophagy in acute kidney injury important role in transportation of the metabo-
mice [26]. lites such as glucose and fatty acids from the cho-
Cystinosis, a lysosomal storage disorder, roid capillaries to the photoreceptors [41]. RPE is
occurs as a result of mutations in the cystinosin, a also involved in lipid metabolism by producing
lysosomal cystine transporter (CTNS) gene [27]. the components that are required for lipoprotein
Studies have shown reduction of TFEB in CTNS-­ synthesis and secretion [42]. Lysosomes are
depleted cells [27]. Induction of TFEB in these extremely important to RPE, because RPE needs
cells can reduce cystine stores and rescues abnor- to process not only shed outer photoreceptor seg-
mal lysosomes cystinotic [28]. Patients with cys- ments (POS) but also reactive oxygen species
tinosis also show the deposition of cystine (ROS) [43]. The autophagosomes and phago-
crystals in the cornea [29, 30]. Gaucher disease somes fuse with lysosomes and degrade cellular
(GD) is a lysosomal storage disease caused by a material; hence, lysosomal function in RPE plays
loss of lysosomal enzyme glucocerebrosidase a very important role in autophagy and phagocy-
activity, which degrades glucocerebroside to glu- tosis [44]. Dysfunctional RPE can cause lipid
cose [31, 32]. Patients with Gaucher disease type accumulation within RPE and Bruch’s membrane
3 (GD3) shows several ocular effects of the dis- leading to pathologies as manifested in AMD
ease, including vitreous lesions and drusen-like [42, 45].
deposits in the posterior segment of the eye [33].
Glucocerebrosidase deficiency also results in
TFEB downregulation and loss of TFEB stability 4 Lysosomal Dysregulation
due to increase of proteasomal degradation in in the Pathogenesis of AMD
Gaucher disease iPSC-derived neuronal cells
[34]. Age-related macular degeneration (AMD) is a
Several studies have reported that lysosomal disease that causes loss of central vision in peo-
dysfunction is associated with several ocular dis- ple over 60 years [46]. AMD results from pro-
eases including glaucoma and age-related macu- gressive RPE degeneration and cell death, further
lar degeneration (AMD) [35, 36]. Extracellular leading to impaired cone and rod photoreceptors
matrix (ECM) deposition in the trabecular mesh- [47]. Studies have shown that lysosomal dysfunc-
work (TM) is one of the vital factors leading to tion is one of the major features leading to the
glaucoma [35]. Studies have shown that the activ- progression of AMD [36]. Lipofuscin has been
ity of lysosomal enzyme cathepsin B impacts the linked to the pathogenesis of AMD [48]; lipofus-
amount of extracellular matrix [37, 38]. On the cin accumulates in the lysosome resulting in
other hand, oxidative stress in TM cells causes lysosomal dysfunction [49]. Lysosomal dysfunc-
322 H.-Y. Pan and M. Valapala

tion results in impaired autophagy and further Alzheimer’s disease mouse model also showed a
causes accumulation of incompletely digested decrease of membrane-tethered C-terminal frag-
POS-containing phagosomes [50, 51], eventually ment C99, which is a β-amyloid precursor [64].
leading to RPE dysfunction and AMD. Oxidative Other studies have shown that induction of TFEB
stress has been recognized as one of the major activity by trehalose treatment decreases
factors leading to AMD via imbalance between hydroquinone-­induced oxidative damage and cell
generation and elimination of reactive oxygen death in ARPE-19 cells [61]. Induction of TFEB
species (ROS) in RPE [43]. Studies have shown expression by trehalose or AKT inhibitor,
positive correlation between ROS and lipofuscin mk-2206, could restore autophagy function in
accumulation [52, 53]. It was previously shown immunoproteasome-deficient retinal cells, which
that mitochondrial dysfunction produces massive have impaired autophagy due to the deficiency of
ROS leading to lysosomal dysfunction [54, 55]. immunoproteasome, a proteasome subtype [65].
Increase of ROS can cause loss of lysosome acid-
ification and result in lysosomal dysfunction
[56]. Smoking is one of leading causes of AMD; 6 Conclusion
it is known to cause an increase in oxidative dam-
age in the RPE [57]. Hydroquinone, a constituent TFEB is a promising therapeutic target for sev-
of cigarette smoke, results in the reduction of eral diseases including cancer, inflammatory dis-
mitochondrial membrane permeabilization and eases, and degenerative diseases because of its
the induction of oxidative stress in the RPE [58, unique function as a master regulator of autoph-
59]. Studies have shown that hydroquinone can agy and lysosomal function. An increasing num-
cause subretinal deposits, which can lead to ber of studies have shown that induction of TFEB
AMD [57]. Studies have shown that expression can rescue the symptoms of diseases
hydroquinone-­induced oxidative damage induces in various cell and animal models.
lysosomal alkalization, downregulation of
lysosome-­ associated membrane protein 2
(LAMP2) and cathepsin D expression in ARPE-­ References
19 cells [60]. Hydroquinone-induced oxidative
damage in RPE cells can be rescued by induction 1. Steingrímsson E, Copeland NG, Jenkins
NA. Melanocytes and the microphthalmia tran-
of TFEB expression by treatment with trehalose scription factor network. Annu Rev Genet.
[61]. 2004;38(1):365–411.
2. Puertollano R, Ferguson SM, Brugarolas J, Ballabio
A. The complex relationship between TFEB tran-
scription factor phosphorylation and subcellular
5 Enhancing TFEB localization. EMBO J. 2018;37(11):e98804.
as a Strategy to Induce 3. Zhu S-Y, Yao R-Q, Li Y-X, Zhao P-Y, Ren C, Du X-H,
Lysosomal Function et al. The role and regulatory mechanism of transcrip-
tion factor EB in health and diseases. Front Cell Dev
Biol. 2021;9(2278):667750.
Previous studies have suggested that overexpres- 4. Napolitano G, Ballabio A. TFEB at a glance. J Cell
sion of TFEB induces autophagy and lysosomal Sci. 2016;129(13):2475–81.
function and prevents substrate accumulation in 5. Pan H-Y, Alamri AH, Valapala M. Nutrient depriva-
several diseases [4]. In Parkinson’s disease, tion and lysosomal stress induce activation of TFEB
in retinal pigment epithelial cells. Cell Mol Biol Lett.
induction of TFEB-mediated autophagy through 2019;24(1):33.
p38 MAPK-TFEB signaling pathway can protect 6. Fernández-Mosquera L, Diogo CV, Yambire KF,
against dopaminergic neuronal loss [17]. AAV-­ Santos GL, Luna Sánchez M, Bénit P, et al. Acute and
mediated overexpression of TFEB shows a reduc- chronic mitochondrial respiratory chain deficiency
differentially regulate lysosomal biogenesis. Sci Rep.
tion of α-synuclein accumulation and restoration 2017;7(1):45076.
of autophagy-lysosome function [62, 63]. 7. Settembre C, De Cegli R, Mansueto G, Saha PK,
Overexpression of TFEB in cellular and Vetrini F, Visvikis O, et al. TFEB controls cellular
Role of TFEB in Diseases Associated with Lysosomal Dysfunction 323

lipid metabolism through a starvation-induced auto- 22. Dejesus-Hernandez M, Ian RM, Bradley FE, Adam
regulatory loop. Nat Cell Biol. 2013;15(6):647–58. LB, Baker M, Nicola JR, et al. Expanded GGGGCC
8. Casares-Crespo L, Calatayud-Baselga I, García-­ hexanucleotide repeat in noncoding region of
Corzo L, Mira H. On the role of basal autophagy in C9ORF72 causes chromosome 9p-linked FTD and
adult neural stem cells and neurogenesis. Front Cell ALS. Neuron. 2011;72(2):245–56.
Neurosci. 2018;12:339. 23. Alan ER, Majounie E, Waite A, Simón-Sánchez J,
9. Di Malta C, Cinque L, Settembre C. Transcriptional Rollinson SJ, et al. A Hexanucleotide repeat expan-
regulation of autophagy: mechanisms and diseases. sion in C9ORF72 is the cause of chromosome 9p21-­
Front Cell Dev Biol. 2019;7:114. linked ALS-FTD. Neuron. 2011;72(2):257–68.
10. Pan H-Y, Ladd AV, Biswal MR, Valapala M. Role 24. Yang C, Chen X-C, Li Z-H, Wu H-L, Jing K-P, Huang
of nuclear factor of activated T cells (NFAT) path- X-R, et al. SMAD3 promotes autophagy dysregula-
way in regulating autophagy and inflammation tion by triggering lysosome depletion in tubular epi-
in retinal pigment epithelial cells. Int J Mol Sci. thelial cells in diabetic nephropathy. Autophagy.
2021;22(16):8684. 2021;17(9):2325–44.
11. Palmieri M, Impey S, Kang H, Di Ronza A, Pelz C, 25. Yuan L, Yuan Y, Liu F, Li L, Liu J, Chen Y, et al. PGC-1α
Sardiello M, et al. Characterization of the CLEAR net- alleviates mitochondrial dysfunction via TFEB-­
work reveals an integrated control of cellular clearance mediated autophagy in cisplatin-induced acute kidney
pathways. Hum Mol Genet. 2011;20(19):3852–66. injury. Aging (Albany NY). 2021;13(6):8421–39.
12. Settembre C, Zoncu R, Medina DL, Vetrini F, Erdin 26. Zhu L, Yuan Y, Yuan L, Li L, Liu F, Liu J, et al.
S, Erdin S, et al. A lysosome-to-nucleus signalling Activation of TFEB-mediated autophagy by tre-
mechanism senses and regulates the lysosome via halose attenuates mitochondrial dysfunction in
mTOR and TFEB. EMBO J. 2012;31(5):1095–108. cisplatin-­induced acute kidney injury. Theranostics.
13. Medina DL, Di Paola S, Peluso I, Armani A, De 2020;10(13):5829–44.
Stefani D, Venditti R, et al. Lysosomal calcium sig- 27. Zhang W, Li X, Wang S, Chen Y, Liu H. Regulation
nalling regulates autophagy through calcineurin and of TFEB activity and its potential as a therapeutic
TFEB. Nat Cell Biol. 2015;17(3):288–99. target against kidney diseases. Cell Death Discov.
14. Martina JA, Puertollano R. TFEB and TFE3: the art of 2020;6(1):32.
multi-tasking under stress conditions. Transcription. 28. Rega LR, Polishchuk E, Montefusco S, Napolitano G,
2017;8(1):48–54. Tozzi G, Zhang J, et al. Activation of the transcription
15. Wang W, Gao Q, Yang M, Zhang X, Yu L, Lawas M, factor EB rescues lysosomal abnormalities in cys-
et al. Up-regulation of lysosomal TRPML1 channels tinotic kidney cells. Kidney Int. 2016;89(4):862–73.
is essential for lysosomal adaptation to nutrient starva- 29. Alsuhaibani AH, Wagoner MD, Khan AO. Confocal
tion. Proc Natl Acad Sci. 2015;112(11):E1373–E81. microscopy of the cornea in nephropathic cystinosis.
16. Palmieri M, Pal R, Nelvagal HR, Lotfi P, Stinnett Br J Ophthalmol. 2005;89(11):1530–1.
GR, Seymour ML, et al. mTORC1-independent 30. Biswas S, Gaviria M, Malheiro L, Marques JP,
TFEB activation via Akt inhibition promotes cellular Giordano V, Liang H. Latest clinical approaches
clearance in neurodegenerative storage diseases. Nat in the ocular Management of Cystinosis: a review
Commun. 2017;8(1):14338. of current practice and opinion from the oph-
17. Chen J, Mao K, Yu H, She H, Zhang H, Liu L, et al. thalmology Cystinosis forum. Ophthalmol Ther.
p38-TFEB pathways promote microglia activation 2018;7(2):307–22.
through inhibiting CMA-mediated NLRP3 degrada- 31. Hruska KS, Lamarca ME, Scott CR, Sidransky
tion in Parkinson’s disease. J. Neuroinflammation. E. Gaucher disease: mutation and polymorphism
2021;18:295. spectrum in the glucocerebrosidase gene (GBA). Hum
18. Cunningham KM, Maulding K, Ruan K, Senturk M, Mutat. 2008;29(5):567–83.
Grima JC, Sung H, et al. TFEB/Mitf links impaired 32. Berg RD, Levitte S, O’Sullivan MP, O’Leary SM,
nuclear import to autophagolysosomal dysfunction in Cambier CJ, Cameron J, et al. Lysosomal disorders
C9-ALS. elife. 2020;9:e59419. drive susceptibility to tuberculosis by compromising
19. Teixeira M, Sheta R, Idi W, Oueslati A. Alpha-­ macrophage migration. Cell. 2016;165(1):139–52.
synuclein and the endolysosomal system in 33. Hopf S, Pfeiffer N, Liesenfeld M, Mengel K-E,
Parkinson’s disease: guilty by association. Hennermann JB, Schmidtmann I, et al. A compre-
Biomolecules. 2021;11(9):1333. hensive monocentric ophthalmic study with Gaucher
20. Moors T, Paciotti S, Chiasserini D, Calabresi P, disease type 3 patients: vitreoretinal lesions, retinal
Parnetti L, Beccari T, et al. Lysosomal dysfunction atrophy and characterization of abnormal saccades.
and α-Synuclein aggregation in Parkinson’s disease: Orphanet J Rare Dis. 2019;14(1):257.
diagnostic links. Mov Disord. 2016;31(6):791–801. 34. Awad O, Sarkar C, Panicker LM, Miller D, Zeng X,
21. Cosden M, Jinn S, Yao L, Gretzula CA, Kandebo M, Sgambato JA, et al. Altered TFEB-mediated lysosomal
Toolan D, et al. A novel glucosylceramide synthase biogenesis in Gaucher disease iPSC-derived neuronal
inhibitor attenuates alpha synuclein pathology and cells. Hum Mol Genet. 2015;24(20):5775–88.
lysosomal dysfunction in preclinical models of synu- 35. Liton PB, Lin Y, Gonzalez P, Epstein DL. Potential
cleinopathy. Neurobiol Dis. 2021;159:105507. role of lysosomal dysfunction in the pathogen-
324 H.-Y. Pan and M. Valapala

esis of primary open angle glaucoma. Autophagy. 49. Moreno-García A, Kun A, Calero O, Medina M,
2009;5(1):122–4. Calero M. An overview of the role of lipofuscin in
36. Cerniauskas E, Kurzawa-Akanbi M, Xie L, Hallam D, age-related neurodegeneration. Front Neurosci.
Moya-Molina M, White K, et al. Complement modu- 2018;12:464.
lation reverses pathology in Y402H -retinal pigment 50. Valapala M, Wilson C, Hose S, Bhutto IA, Grebe R,
epithelium cell model of age-related macular degen- Dong A, et al. Lysosomal-mediated waste clearance
eration by restoring lysosomal function. Stem Cells in retinal pigment epithelial cells is regulated by
Transl Med. 2020;9(12):1585–603. CRYBA1/βA3/A1-crystallin via V-ATPase-MTORC1
37. Porter K, Lin Y, Liton PB. Cathepsin B is up-regulated signaling. Autophagy. 2014;10(3):480–96.
and mediates extracellular matrix degradation in tra- 51. Escrevente C, Falcão AS, Hall MJ, Lopes-Da-Silva
becular meshwork cells following phagocytic chal- M, Antas P, Mesquita MM, et al. Formation of
lenge. PLoS One. 2013;8(7):e68668. lipofuscin-­like autofluorescent granules in the retinal
38. Nettesheim A, Shim MS, Dixon A, Raychaudhuri pigment epithelium requires lysosome dysfunction.
U, Gong H, Liton PB. Cathepsin B localizes in the Investig Opthalmol Vis Sci. 2021;62(9):39.
caveolae and participates in the proteolytic cascade 52. Bayati S, Yazdanparast R. Antioxidant and free radi-
in trabecular meshwork cells. Potential new drug cal scavenging potential of yakuchinone B deriva-
target for the treatment of glaucoma. J Clin Med. tives in reduction of lipofuscin formation using
2021;10(1):78. H2O2-treated neuroblastoma cells. Iran Biomed J.
39. Porter K, Nallathambi J, Lin Y, Liton PB. Lysosomal 2011;15(4):134–42.
basification and decreased autophagic flux in oxi- 53. Terman A, Dalen H, Eaton JW, Neuzil J, Brunk
datively stressed trabecular meshwork cells: impli- UT. Aging of cardiac myocytes in culture: oxidative
cations for glaucoma pathogenesis. Autophagy. stress, lipofuscin accumulation, and mitochondrial
2013;9(4):581–94. turnover. Ann N Y Acad Sci. 2004;1019(1):70–7.
40. Yang S, Zhou J, Li D. Functions and diseases of 54. Demers-Lamarche J, Guillebaud G, Tlili M, Todkar
the retinal pigment epithelium. Front Pharmacol. K, Bélanger N, Grondin M, et al. Loss of mito-
2021;12(1976):727870. chondrial function impairs lysosomes. J Biol Chem.
41. Simó R, Villarroel M, Corraliza L, Hernández C, 2016;291(19):10263–76.
Garcia-Ramírez M. The retinal pigment epithelium: 55. Yuan Y, Chen Y, Peng T, Li L, Zhu W, Liu F, et al.
something more than a constituent of the blood-retinal Mitochondrial ROS-induced lysosomal dysfunction
barrier–implications for the pathogenesis of diabetic impairs autophagic flux and contributes to M1 mac-
retinopathy. J Biomed Biotechnol. 2010;2010:190724. rophage polarization in a diabetic condition. Clin Sci.
42. Jun S, Datta S, Wang L, Pegany R, Cano M, Handa 2019;133(15):1759–77.
JT. The impact of lipids, lipid oxidation, and inflam- 56. Song SB, Hwang ES. High levels of ROS impair lyso-
mation on AMD, and the potential role of miR- somal acidity and autophagy flux in glucose-deprived
NAs on lipid metabolism in the RPE. Exp Eye Res. fibroblasts by activating ATM and Erk pathways.
2019;181:346–55. Biomol Ther. 2020;10(5):761.
43. Zhang Z-Y, Bao X-L, Cong Y-Y, Fan B, Li 57. Espinosa-Heidmann DG, Suner IJ, Catanuto
G-Y. Autophagy in age-related macular degeneration: P, Hernandez EP, Marin-Castano ME, Cousins
a regulatory mechanism of oxidative stress. Oxidative SW. Cigarette smoke–related oxidants and the devel-
Med Cell Longev. 2020;2020:2896036. opment of Sub-RPE deposits in an experimental ani-
44. Sinha D, Valapala M, Shang P, Hose S, Grebe R, mal model of dry AMD. Investig Opthalmol Vis Sci.
Lutty GA, et al. Lysosomes: regulators of autophagy 2006;47(2):729.
in the retinal pigmented epithelium. Exp Eye Res. 58. Strunnikova N, Zhang C, Teichberg D, Cousins SW,
2016;144:46–53. Baffi J, Becker KG, et al. Survival of retinal pigment
45. Golestaneh N, Chu Y, Xiao Y-Y, Stoleru GL, Theos epithelium after exposure to prolonged oxidative
AC. Dysfunctional autophagy in RPE, a contribut- injury: a detailed gene expression and cellular analy-
ing factor in age-related macular degeneration. Cell sis. Investig Opthalmol Vis Sci. 2004;45(10):3767.
Death Dis. 2018;8(1):e2537. 59. Bertram KM, Baglole CJ, Phipps RP, Libby
46. Gehrs KM, Anderson DH, Johnson LV, Hageman RT. Molecular regulation of cigarette smoke
GS. Age-related macular degeneration—emerging induced-oxidative stress in human retinal pig-
pathogenetic and therapeutic concepts. Ann Med. ment epithelial cells: implications for age-related
2006;38(7):450–71. macular degeneration. Am J Physiol Cell Physiol.
47. Bhutto I, Lutty G. Understanding age-related macu- 2009;297(5):C1200–C10.
lar degeneration (AMD): relationships between the 60. Abokyi S, Shan S-W, Lam CH-I, Catral KP, Pan F,
photoreceptor/retinal pigment epithelium/Bruch’s Chan HH-L, et al. Targeting lysosomes to reverse
membrane/choriocapillaris complex. Mol Asp Med. hydroquinone-induced autophagy defects and oxida-
2012;33(4):295–317. tive damage in human retinal pigment epithelial cells.
48. Blasiak J. Senescence in the pathogenesis of age-­ Int J Mol Sci. 2021;22(16):9042.
related macular degeneration. Cell Mol Life Sci. 61. Abokyi S, Shan SW, To C-H, Chan HH-L, Tse
2020;77(5):789–805. DY-Y. Autophagy upregulation by the TFEB
Role of TFEB in Diseases Associated with Lysosomal Dysfunction 325

inducer Trehalose protects against oxidative dam- prevents Parkinson’s disease-related neurodegenera-
age and cell death associated with NRF2 inhibition tion. Mol Ther. 2018;26(6):1552–67.
in human RPE cells. Oxidative Med Cell Longev. 64. Bécot A, Pardossi-Piquard R, Bourgeois A, Duplan
2020;2020:1–18. E, Xiao Q, Diwan A, et al. The transcription factor
62. Arotcarena M-L, Bourdenx M, Dutheil N, Thiolat EB reduces the intraneuronal accumulation of the
M-L, Doudnikoff E, Dovero S, et al. Transcription beta-secretase-derived APP fragment C99 in cel-
factor EB overexpression prevents neurodegenera- lular and mouse Alzheimer’s disease models. Cells.
tion in experimental synucleinopathies. JCI Insight. 2020;9(5):1204.
2019;4(16):e129719. 65. Karim MR, Fisher CR, Kapphahn RJ, Polanco JR,
63. Torra A, Parent A, Cuadros T, Rodríguez-Galván B, Ferrington DA. Investigating AKT activation and
Ruiz-Bronchal E, Ballabio A, et al. Overexpression autophagy in immunoproteasome-deficient retinal
of TFEB drives a pleiotropic neurotrophic effect and cells. PLoS One. 2020;15(4):e0231212.
Retinoic Acid Receptor-Related
Orphan Receptors (RORs) in Eye
Development and Disease

Felix Yemanyi, Kiran Bora, Alexandra K. Blomfield,


and Jing Chen

Abstract Keywords

The retinoic acid receptor-related orphan Nuclear receptors · Retinoic acid receptor-­
receptors (RORs) are ligand-mediated tran- related orphan receptors (RORs) · RORα ·
scription factors with important biological RORβ · RORγ · Eye development · Eye
roles in regulating circadian rhythms, metabo- diseases · Retinal vascular diseases · Retinal
lism, immunity, angiogenesis, inflammation, degenerative diseases
and development. They belong to the super-
family of nuclear receptors and include three
family members: RORα, RORβ, and RORγ. 1 Introduction
Currently identified ROR ligands include cho-
lesterol and cholesterol derivatives for RORα The retinoic acid receptor-related orphan recep-
and RORγ, and stearic acid and all-trans reti- tors (RORs) belong to a subfamily of the nuclear
noic acid for RORβ. Aberrant signaling of the receptor (NR) superfamily and include three
RORs is involved in the pathogenesis of sev- members: RORα (NR1F1), RORβ (NR1F2), and
eral human diseases including autoimmune RORγ (NR1F3) [1]. Like other members of the
diseases, metabolic disorders, and certain can- NR superfamily, the RORs exhibit a marked
cers. In the eye, RORs regulate normal devel- sequence homology and conserved structure to
opment of the lens and the retina, and also enable their function as ligand-dependent tran-
contribute to potentially blinding eye diseases, scription regulators. RORs harbor a ligand-­
especially retinal vascular diseases. Here, we independent activation function-1 (AF-1) domain
review the role of RORs in eye development in the N-terminus, followed by a DNA-binding
and disease to highlight their potential as domain (DBD), hinge region, and a ligand-­
druggable targets for therapeutic development binding domain in the C-terminus, with the DBD
in retinal vascular and degenerative diseases. being the most conserved region for DNA recog-
nition [2]. Upon ligand-regulated activation,
RORs bind as monomers to DNA at ROR
response elements (RORE) encompassing an
F. Yemanyi · K. Bora · A. K. Blomfield · J. Chen (*) AGGTCA consensus motif preceded by an A/T-­-
Department of Ophthalmology, Boston Children’s rich region [2]. Together with other transcrip-
Hospital, Harvard Medical School, Boston, MA, tional co-regulators, RORs meditate transcription
USA of many target genes including circadian, meta-
e-mail: Jing.Chen@childrens.harvard.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 327
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_47
328 F. Yemanyi et al.

bolic, and inflammatory genes. They hence play for cone photoreceptor development in the mouse
important physiological roles in regulating circa- retina, where it directly regulates a subset of cone
dian rhythm, development, metabolism, angio- photopigment genes, including S-opsin and
genesis, and immunity [1, 3]. M-opsin, as well as cone arrestin [7].
Dysregulation of RORs is implicated in sev-
eral pathologies like cancer, autoimmune, meta-
bolic, and inflammatory diseases [1, 2, 4]. While 2.2 Genetic Variations of RORA
the endogenous physiological ligands for RORs Are Linked with Age-Related
have not been unequivocally confirmed, choles- Macular Degeneration (AMD)
terol and cholesterol derivatives have been dis-
covered as ligands for RORα and RORγ, and The importance of RORα in eye diseases is
stearic acid and all-trans retinoic acid were iden- exemplified by genetic studies linking its genetic
tified as RORβ ligands based on structural bind- variation with risks of developing AMD, a major
ing [3]. Many synthetic ROR agonists and cause of vision impairment in the elderly.
antagonists have also been developed and are Previous human clinical studies have identified
currently being investigated in experimental common variants and haplotypes within intron 1
studies of ROR-related diseases [3]. Therefore, of the RORA gene and its apparent synergistic
RORs represent attractive druggable targets to interaction with the ARMS2/HTRA1 locus as
ameliorate several debilitating human diseases increased risk factors for wet (neovascular) AMD
including ocular pathologies. Accordingly, this [11, 12]. A subsequent study suggested interac-
mini review summarizes the respective roles of tion between RORA and ROBO1 genes for both
the RORs in eye development and diseases. wet and dry (atrophic) AMD risks [13]. Together,
these works highlight an important role of RORα
in AMD pathogenesis. Consequently, an AAV
2 RORα Regulates Eye approach delivering RORA gene (OCU410,
Development and Vascular AAV-RORA) is currently being investigated by
Eye Diseases Ocugen Inc. as a potential gene therapy for AMD,
and clinical trials are planned.
2.1 RORα Regulates Lens
and Cone Photoreceptor
Development 2.3 RORα Modulates Retinal
Inflammation and
RORα is widely expressed in many tissues Neurovascular Interaction
including the brain, liver, skeletal muscle, skin, in a Mouse Model of Oxygen-­
lung, adipose tissue, and eye [2]. The human Induced Retinopathy (OIR)
RORα (RORA) gene is located on chromosome
15q22.2. Humans express four RORα isoforms Additional function of RORα in ocular angiogen-
(RORα1–4) produced by alternative splicing, esis was uncovered by our group using a mouse
whereas two of these isoforms (RORα1 and model of OIR [14] modeling ischemic prolifera-
RORα4) are also expressed in mice [5]. In the tive retinopathy in humans. We found that RORα
mouse eye, expression of RORα is found in the is a novel regulator of pathologic retinal neovas-
lens, retinal ganglion cells, bipolar cells, photore- cularization by directly modulating the transcrip-
ceptors, and microglia/macrophages [6–9]. tion of suppressor of cytokine signaling 3
RORα is important for the development of the (SOCS3), a key inflammatory regulator, in retinal
lens [10], where it is critical for the differentia- microglia and macrophages [8]. RORα-deficient
tion of lens epithelial cells into fiber cells by mice exhibit decreased neovascularization in
directly activating expression of γF-crystallin OIR with dampened inflammatory response,
gene [10]. In addition, RORα is also important whereas treatment with RORα synthetic inverse
Retinoic Acid Receptor-Related Orphan Receptors (RORs) in Eye Development and Disease 329

agonist ameliorated the neovascular lesions in 3 RORβ Regulates Retinal


both OIR mouse retinas and mice deficient in Neuronal Differentiation
very low-density lipoprotein receptor (VLDLR),
modeling angiomatous proliferation and macular Compared with RORα, expression of RORβ is
telangiectasia [8]. In addition, RORα engenders more restricted, found exclusively in the brain,
pathological retinal angiogenesis in mice by dis- bone, and the eye [2]. The RORβ gene (RORB) is
rupting the physiological interactions between mapped to human chromosome 9q21.13 [3].
retinal neurons and the vasculature via negatively Whereas humans seemingly express only one
regulating class 3 semaphorin E (SEMA3E), a RORβ isoform (RORβ1), in mice, two isoforms
neuron-secreted axonal and vascular guidance (RORβ1 and RORβ2) have been reported [3]. In
factor, in retinal ganglion cells [9]. These works the eye, RORβ is expressed in retinal neurons and
suggest an angiogenic role of RORα in modulat- is vital for the development of rod and cone pho-
ing pathological neovascularization in experi- toreceptors [15–17]. During prenatal and postna-
mental retinopathy via regulating both tal development of the mouse retina, RORβ
inflammation and neurovascular interaction transcriptionally modulates Blimp1 to induce the
(Fig. 1). photoreceptor cell fate by suppressing bipolar
Together, in the eye, RORα is not only impor- cell identity in most of the retinal progenitor cells
tant for lens and retinal development but also [15]. In addition, RORβ directly transactivates
implicated in clinical AMD and experimental S-opsin in cones during photoreceptor develop-
retinopathy (Fig. 2), suggesting that RORα is a ment in mice [16]; therefore, loss of function in
probable druggable target for these eye RORβ results in S-cone morphological abnor-
conditions. malities in addition to rod loss [17]. Moreover,

Fig. 1 Schematic diagram illustrating regulation of path- leading to disrupted neurovascular interaction concomi-
ological retinal angiogenesis by RORα. RORα suppresses tant with pathological neovascularization in retinopathy.
transcription of Socs3 in retinal microglia/macrophage, RORα: retinoic acid receptor-related orphan receptor α,
thereby increasing inflammation to promote pathological Socs3: suppressor of cytokine signaling 3, Sema3E: class
neovascularization in retinopathy. In addition, RORα 3 semaphorin E, RGCs: retinal ganglion cells, RORE:
directly suppresses Sema3E gene transcription in RGCs, ROR response elements
330 F. Yemanyi et al.

Fig. 2 Venn diagram


summarizing the
overlapping and distinct
roles of RORs in eye
development and
disease. RORs: retinoic
acid receptor-related
orphan receptors

the RORβ1 isoform is important for differentia- vasculature [2, 20]. RORγt together with RORα
tion of retinal horizontal and amacrine interneu- is a key transcription factor for T helper (Th) 17
rons through mediating early-acting factors Ptf1a cell differentiation and interleukin 17 (IL-17)
and Foxn4 [18]. All these studies strongly sug- production [21, 22]. Whereas enhancement of
gest a transcriptional regulatory role of RORβ in this RORγt-mediated regulation of Th17 cells
mediating retinal neuronal differentiation, par- and IL-17 is a promising immunotherapy for can-
ticularly photoreceptors and interneu- cer [23] and autoimmune diseases [24] and for
rons (Fig. 2). Whereas RORβ has roles in preventing corneal infections [25], excessive pro-
regulating circadian rhythms (just like RORα) duction of Th17 cells and IL17 by RORγt is a
and bone development [2, 3], and loss of function major hallmark of ocular diseases such as dry eye
in RORB is associated with epilepsy in humans [26], Graves orbitopathy (the most common
[19], whether it plays any role in eye diseases is orbital disease causing blindness and disability)
still unknown. [27, 28], and autoimmune uveitis [29]. Therefore,
in these eye conditions, inhibition of RORγt may
hold promise as a new therapeutic intervention.
4 RORγ in Autoimmune Additionally, genetic variations of IL-17 are also
Regulation and Eye Diseases linked with AMD [30], as well as Th17 cells in
AMD-associated inflammation [31], further sug-
4.1 RORγt, a Key Regulator gesting a putative role of RORγt in AMD.
of Th17 Cells, Is Implicated
in Ocular Autoimmunity
4.2 RORγt/IL17 Signaling Axis
The RORγ gene maps to human chromosome in Retinal Vasculature
1q21.3 and exists as two isoforms in both humans
and mice: RORγ (RORγ1) and RORγt (RORγ2). Recent experimental studies suggest an emerging
RORγ differs from RORγt only at the first 100 role of RORγt in regulating retinal vasculature
nucleotides at the N-terminus [2]. RORγ is under pathological conditions. For example, a
expressed in muscle tissue, prostate, pancreas, study reported pathologic role of the RORγt/IL17
heart, liver, and testicles, whereas RORγt is signaling axis in the retinal vasculature of dia-
expressed in lymphatic tissues and in the retinal betic mice modeling diabetic retinopathy (DR), a
Retinoic Acid Receptor-Related Orphan Receptors (RORs) in Eye Development and Disease 331

microvascular complication of diabetes which References


affects the working-age population [20].
Consequently, small-molecule inhibitors of 1. Fan J, et al. Retinoic acid receptor-related orphan
receptors: critical roles in tumorigenesis. Front.
RORγt (including SR1001, the RORα/γ inverse Immunol. 2018;9:1187. https://doi.org/10.3389/
agonist) ameliorated the vessel loss, neovascular fimmu.2018.01187.
phenotype, and vascular leakage observed in dia- 2. Ladurner A, Schwarz PF, Dirsch VM. Natural
betic mice and OIR retinas [20, 29, 32]. Another products as modulators of retinoic acid recep-
tor-related orphan receptors (RORs). Nat. Prod.
study in mice and rats also demonstrated that reti- Rep. 2021;38(4):757–81. https://doi.org/10.1039/
nal immunocompetent cells, microglia, could be d0np00047g.
an additional source of RORγt and IL17A beyond 3. Solt LA, Griffin PR, Burris TP. Ligand regulation
Th17 cells, to subsequently trigger production of of retinoic acid receptor-related orphan receptors:
implications for development of novel therapeutics.
vascular endothelial growth factor (VEGF) and Curr. Opin. Lipidol. 2010;21(3):204–11. https://doi.
mediate pathological retinal neovascularization org/10.1097/mol.0b013e328338ca18.
in experimental retinopathy [32]. 4. Zhang Y, Luo X-Y, Wu D-H, Xu Y. ROR nuclear
To recapitulate, aberrant RORγt expression receptors: structures, related diseases, and drug dis-
covery. Acta Pharmacol. Sin. 2015;36(1):71–87.
and signaling are implicated in several eye dis- https://doi.org/10.1038/aps.2014.120.
eases including dry eye, autoimmune uveitis, as 5. Lee JM, Kim H, Baek SH. Unraveling the physi-
well as experimental retinopathy (Fig. 2). Small-­ ological roles of retinoic acid receptor-related orphan
molecule inhibitors of RORγt may offer promis- receptor α. Exp. Mol. Med. 2021;53(9):1278–86.
https://doi.org/10.1038/s12276-­021-­00679-­8.
ing therapeutic option to curb the visual 6. Steinmayr M, et al. Staggerer phenotype in retinoid-­
impairment associated with these eye and retinal related orphan receptor-deficient mice. Proc.
diseases. Natl. Acad. Sci. 1998;95(7):3960–5. https://doi.
org/10.1073/pnas.95.7.3960.
7. Fujieda H, Bremner R, Mears AJ, Sasaki H. Retinoic
acid receptor-related orphan receptor α regulates a
5 Conclusions subset of cone genes during mouse retinal develop-
ment. J. Neurochem. 2009;108(1):91–101. https://doi.
While both RORα and RORβ play important org/10.1111/j.1471-­4159.2008.05739.x.
8. Sun Y, et al. Nuclear receptor RORα regulates patho-
roles in retinal development and neuronal differ- logic retinal angiogenesis by modulating SOCS3-­
entiation, both RORα and RORγt are implicated dependent inflammation. Proc. Natl. Acad. Sci.
in retinal vascular diseases and ocular immune 2015;112(33):10401–6. https://doi.org/10.1073/
regulation (Fig. 2). Given these roles of RORs in pnas.1504387112.
9. Sun Y, et al. RORα modulates semaphorin 3E tran-
the eye, targeting them in pathological eye condi- scription and neurovascular interaction in pathological
tions is appealing. Over the past two decades, the retinal angiogenesis. FASEB J. 2017;31(10):4492–
discovery of many endogenous and synthetic 502. https://doi.org/10.1096/fj.201700172r.
ligands for RORs has instilled hope and garnered 10. Tini M, Fraser RA, Giguère V. Functional interac-
tions between retinoic acid receptor-related orphan
attention for drug development [2–4]. Harnessing nuclear receptor (RORα) and the retinoic acid recep-
RORs as drug targets could greatly expand cur- tors in the regulation of the γF-crystallin promoter.
rent treatment options for various eye diseases J. Biol. Chem. 1995;270(34):20156–61. https://doi.
including AMD and other retinal vascular and org/10.1074/jbc.270.34.20156.
11. Silveira AC, et al. Convergence of linkage, gene
degenerative diseases [8, 9, 29, 32]. expression and association data demonstrates the
influence of the RAR-related orphan receptor alpha
Acknowledgments: (RORA) gene on neovascular AMD: a systems biol-
Funding: This work was supported by NIH/NEI R01 ogy based approach. Vision Res. 2010;50(7):698–
grants (EY024963, EY028100, and EY031765) (to J.C.). 715. https://doi.org/10.1016/j.visres.2009.09.016.
12. Schaumberg DA. Prospective study of common vari-
ants in the retinoic acid receptor–related orphan recep-
Author contributions: F.Y. and J.C. conceived the mini
tor α gene and risk of neovascular age-related macular
review and wrote the manuscript; K.B. and J.C. prepared
degeneration. Arch. Ophthalmol. 2010;128(11):1462.
the figures. F.Y., K.B., A.K.B. and J.C. edited and
https://doi.org/10.1001/archophthalmol.2010.261.
approved the manuscript.
332 F. Yemanyi et al.

13. Jun G, et al. Influence of ROBO1 and RORA on risk of molecule therapeutics for cancer immunotherapy.
age-related macular degeneration reveals genetically J. Med. Chem. 2018;61(14):5794–804. https://doi.
distinct phenotypes in disease pathophysiology. PloS org/10.1021/acs.jmedchem.7b01314.
One. 2011;6(10):e25775. https://doi.org/10.1371/ 24. Solt LA, et al. Suppression of TH17 differentiation
journal.pone.0025775. and autoimmunity by a synthetic ROR ligand. Nature.
14. Smith L, et al. Oxygen-induced retinopa- 2011;472(7344):491–4. https://doi.org/10.1038/
thy in the mouse. Invest. Ophthalmol. Vis. Sci. nature10075.
1994;35(1):101–11. 25. Taylor PR, et al. JAK/STAT regulation of Aspergillus
15. Mills TS, et al. Combinatorial regulation of a Blimp1 fumigatus corneal infections and IL-6/23-stimulated
(Prdm1) enhancer in the mouse retina. PloS One. neutrophil, IL-17, elastase, and MMP9 activity.
2017;12(8):e0176905. https://doi.org/10.1371/jour- J. Leukoc. Biol. 2016;100(1):213–22. https://doi.
nal.pone.0176905. org/10.1189/jlb.4a1015-­483r.
16. Srinivas M, Ng L, Liu H, Jia L, Forrest D. Activation 26. Zheng X, De Paiva CS, Li D-Q, Farley WJ,
of the blue opsin gene in cone photoreceptor devel- Pflugfelder SC. Desiccating stress promotion of Th17
opment by retinoid-related orphan receptor β. differentiation by ocular surface tissues through a
Mol. Endocrinol. 2006;20(8):1728–41. https://doi. dendritic cell-mediated pathway. Investig Opthalmol
org/10.1210/me.2005-­0505. Vis Sci. 2010;51(6):3083. https://doi.org/10.1167/
17. Jia L, et al. Retinoid-related orphan nuclear recep- iovs.09-­3838.
tor ROR is an early-acting factor in rod photo- 27. Fang S, et al. Evidence for associations between Th1/
receptor development. Proc. Natl. Acad. Sci. Th17 “hybrid” phenotype and altered lipometabolism
2009;106(41):17534–9. https://doi.org/10.1073/ in very severe graves orbitopathy. J. Clin. Endocrinol.
pnas.0902425106. Metabol. 2020;105(6):1851–67.
18. Liu H, et al. An isoform of retinoid-related 28. Fang S, et al. IL-17A exacerbates fibrosis by pro-
orphan receptor β directs differentiation of reti- moting the proinflammatory and profibrotic function
nal amacrine and horizontal interneurons. Nat. of orbital fibroblasts in TAO. J. Clin. Endocrinol.
Commun. 2013;4(1):1813. https://doi.org/10.1038/ Metabol. 2016;101(8):2955–65. https://doi.
ncomms2793. org/10.1210/jc.2016-­1882.
19. Rudolf G, et al. Loss of function of the retinoid-­ 29. Tan J, et al. Small molecules targeting RORγt inhibit
related nuclear receptor (RORB) gene and epilepsy. autoimmune disease by suppressing Th17 cell dif-
Eur. J. Hum. Genet. 2016;24(12):1761–70. https:// ferentiation. Cell Death Dis. 2020;11(8) https://doi.
doi.org/10.1038/ejhg.2016.80. org/10.1038/s41419-­020-­02891-­2.
20. Zapadka TE, et al. RORγt inhibitor-SR1001 halts 30. Zhang S, Liu Y, Lu S, Cai X. Genetic variants of
retinal inflammation, capillary degeneration, and interleukin 17A are functionally associated with
the progression of diabetic retinopathy. Int. J. Mol. increased risk of age-related macular degenera-
Sci. 2020;21(10):3547. https://doi.org/10.3390/ tion. Inflammation. 2015;38(2):658–63. https://doi.
ijms21103547. org/10.1007/s10753-­014-­9973-­3.
21. Ivanov II, et al. The orphan nuclear receptor RORγt 31. Chen J, Wang W, Li Q. Increased Th1/Th17
directs the differentiation program of proinflamma- responses contribute to low-grade inflamma-
tory IL-17+ T helper cells. Cell. 2006;126(6):1121– tion in age-related macular degeneration. Cell.
33. https://doi.org/10.1016/j.cell.2006.07.035. Physiol. Biochem. 2017;44(1):357–67. https://doi.
22. Wang R, et al. Genetic and pharmacological inhibition org/10.1159/000484907.
of the nuclear receptor RORα regulates TH17 driven 32. Talia DM, Deliyanti D, Agrotis A, Wilkinson-Berka
inflammatory disorders. Nat. Commun. 2021;12(1) JL. Inhibition of the nuclear receptor RORγ and
https://doi.org/10.1038/s41467-­020-­20385-­9. interleukin-17A suppresses Neovascular retinopathy.
23. Qiu R, Wang Y. Retinoic acid receptor-related orphan Arterioscler. Thromb. Vasc. Biol. 2016;36(6):1186–
receptor γt (RORγt) agonists as potential small 96. https://doi.org/10.1161/atvbaha.115.307080.
Part VIII
Mechanisms of Degeneration – Animal
Models
A Novel Mouse Model
for Late-­Onset Retinal
Degeneration (L-ORD) Develops
RPE Abnormalities Due to the Loss
of C1qtnf5/Ctrp5

Shyamanga Borooah, Anil Chekuri,


Shikha Pachauri, Bhubananda Sahu,
Marina Vorochikhina, John J. Suk, Dirk-­
Uwe Bartsch, Venkata R. M. Chavali,
Monica M. Jablonski, and Radha Ayyagari

Abstract nal imaging, light microscopy, and


transmission electron microscopy (TEM) at 6,
Late-onset retinal degeneration (L-ORD) is an 11, and 18.5 mo. Expression of Ctrp5 was
autosomal dominant macular dystrophy analyzed using immunostaining and qRT-­
resulting from mutations in the gene PCR. The Ctrp5−/− mice showed lack of both
CTRP5/C1QTNF5. A mouse model (Ctrp5+/−) Ctrp5 transcript and protein. Presence of a
for the most common S163R developed many significantly larger number of autofluorescent
features of human clinical disease. We gener- spots was observed in Ctrp5−/− mice compared
ated a novel homozygous Ctrp5 gene knock-­ to the WT (P < 0.0001) at 19 mo. Increased
out (Ctrp5−/−) mouse model to further study RPE stress with vacuolization and thinning
the mechanism of L-ORD. The retinal mor- was observed as early as 6 mo in Ctrp5−/−
phology of these mice was evaluated by reti- mice. Further, ultrastructural analyses revealed
a progressive accumulation of basal laminar
sub-RPE deposits in Ctrp5−/− mice from 11
Authors Shyamanga Borooah and Anil Chekuri have mo. The Ctrp5−/− mice shared retinal and RPE
equally contributed to this chapter.

S. Borooah · S. Pachauri · B. Sahu · M. Vorochikhina Department of Ophthalmology, Scheie Eye Institute,


· J. J. Suk · D.-U. Bartsch · V. R. M. Chavali · University of Pennsylvania, Philadelphia, PA, USA
R. Ayyagari (*) e-mail: anil_chekuri@meei.harvard.edu
Shiley Eye Institute, University of California San
M. M. Jablonski
Diego, La Jolla, CA, USA
Department of Ophthalmology, Hamilton Eye
e-mail: sborooah@health.ucsd.edu; spachauri@
Institute, University of Tennessee Health Science
health.ucsd.edu; bsahu@ucsd.edu; jjsuk@ucsd.edu;
Center, Memphis, TN, USA
dbartsch@health.ucsd.edu; vchavali@pennmedicine.
e-mail: mjablon1@uthsc.edu
upenn.edu; rayyagari@health.ucsd.edu
A. Chekuri
Shiley Eye Institute, University of California San
Diego, La Jolla, CA, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 335
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_48
336 S. Borooah et al.

pathology that matches with that previously Patients with L-ORD usually develop early-onset
described for Ctrp5+/− mice suggesting that long anterior lens zonules, late-onset dark-­
pathology in these mice results from the loss adaptation abnormalities, pseudodrusen deposits,
of functional CTRP5 and that the presence of geographic atrophy, and visual symptoms begin-
CTRP5 is critical for normal RPE and retinal ning in fifth decade of life, with late-stage choroi-
function. dal neovascular membrane formation in the
macula [2]. Mutations in the gene C1QTNF5/
Keywords CTRP5 have been implicated in L-ORD pathol-
ogy [3, 4]. A mouse model with the heterozygous
Late-onset retinal degeneration (L-ORD) · S163R mutation in Ctrp5 recapitulated the
CTRP5/C1QTNF5 · Sub-RPE deposits · L-ORD phenotype [5, 6]. Here we describe the
BLamD · Retinal and RPE pathology · Gene phenotype of a homozygous Ctrp5 gene knock-­
knock-out mouse model · Fundus autofluo- out (KO) mouse (Ctrp5−/−) model to further
rescent spots understand the mechanism of L-ORD pathology.

1 Introduction 2 Results

Late-onset retinal degeneration (L-ORD) is an 2.1 Exons 2 and 3 of Ctrp5 Are


autosomal dominant phenotype. The presence of Deleted in Ctrp5−/− Mice
deposits between the retinal pigment epithelium
and Bruch’s membrane has been described as the The Ctrp5 knock-out allele was generated by
hallmark of autosomal dominant L-ORD [1]. excision of exons 2 and 3 of Ctrp5 and contains

Fig. 1 Confirmation of Ctrp5 deletion in Ctrp5−/− mice terior eyecup tissue at 2 mo (*** P < 0.001). (c)
(a) The Ctrp5 knock-out allele was generated by excision Immunostaining of CTRP5 (red) in 2-mo WT and Ctrp5−/−
of exons 2 and 3 of Ctrp5 by Cre-recombinase. The Ctrp5 mouse retina. (d) Western blot analysis of CTRP5 expres-
knock-out allele possesses only exon 1. The restriction sion in posterior eye cup lysates from 2-mo-old WT and
sites and location of exon1 (box) are also shown. (b) Ctrp5−/− mice. β-Actin was used for normalization
Expression of Ctrp5 mRNA in WT and Ctrp5−/− mice pos-
A Novel Mouse Model for Late-Onset Retinal Degeneration (L-ORD) Develops RPE Abnormalities… 337

only exon 1 (Fig. 1.a). Amplification and sequenc- mo (Fig. 3), and signs of RPE stress were evident
ing of genomic DNA using primers flanking the in Ctrp5−/− mice, with areas of vacuolization and
deleted region confirmed the absence of exons 2 thinning, allowing photoreceptor outer segments
and 3 of Ctrp5 in Ctrp5−/− mice. to protrude into spaces that would ordinarily be
occupied by the RPE. By 11 mo of age, sub-RPE
deposits and swollen photoreceptor inner seg-
2.2 Loss of Ctrp5 Expression ments were present. By 18.5 mo, RPE vacuoles
in Ctrp5−/− Mice and sub-RPE deposits were more abundant.
Retinal ultrastructure analysis also showed
CTRP5 is expressed predominantly in the RPE similar structural profile of Ctrp5−/− mice
and ciliary body within the eye of normal indi- (Fig. 4). The RPE of Ctrp5−/− mice aged 6 mo
viduals [7]. In the ocular tissue of 2-mo Ctrp5−/− or older were filled with vacuoles of varying
mice, the Ctrp5 transcript and the protein were sizes. At 11 mo, dense sub-RPE deposits were
undetectable by qRT-PCR, Western blot analysis, present in Bruch’s membrane, and these
and immunohistochemistry (Fig. 1b, c, d). increased in size by 18.5 mo. The most promi-
nent feature of the RPE of 18.5-mo Ctrp5−/−
mice was the presence of electron-dense
2.3 Accumulation lipid-based basal laminar deposits. In addition
of Autofluorescent Spots Bruch’s membrane was periodically disrupted.
in Ctrp5−/− Mice In comparison, no aberrant features were noted
in 21-mo WT mice.
Fundus autofluorescent (AF) scanning laser oph-
thalmoscopy was employed for the examination
of the AF spots in the retina of 19-mo WT 3 Materials and Methods
C57BL/6 and Ctrp5−/− mice. Ctrp5−/− mice
showed a significantly higher number of AF 3.1 Generation of Mice
spots, i.e., 38.4 ± 11.1 (n = 4) as compared to the
WT mice 5.5 ± 2.2 (n = 5) (P < 0.0001) (Fig. 2). The generation and characterization of the hetero-
zygous knock-in (Ctrp5 +/−) mice has been described
previously [5]. To generate homozygous Ctrp5
2.4 Early RPE Stress knock-out (Ctrp5−/−) mice, Ctrp5+/+ mice were
and Accumulation of Sub-RPE crossed with global Cre mice on the same
Deposits with Age C57BL/6 J background to remove exons 2 and 3 of
in Ctrp5−/− Mice the Ctrp5 gene [8]. The Ctrp5−/− genotype was
detected using primers flanking the deleted region
Retinal morphology of Ctrp5−/− mice showed (3329-Ctrp5-F, 5′-GTCTGAGGAAGCCATTCA
abnormal retinal and RPE structure as early as 6 AAG-3′, and 3331-Ctrp5-R, 5′-GGT CCTGGG

Fig. 2 Autofluorescence
imaging of WT and
Ctrp5−/− mice
Representative AF-SLO
images from 19-mo-old
WT and Ctrp5−/− mice
338 S. Borooah et al.

Fig. 3 Analysis of retinal histology in Ctrp5−/− mice cated with the boxes labeled (E-H). Areas of RPE vacuol-
Retinal architecture was analyzed by LM in 6-, 11-, 18.5- ization are marked with black arrows. Magnification
mo Ctrp5−/− mice and 21-mo WT (A-D). Magnified bars = 200 microns in top panels and 500 nanometers in
images of panel A-D generated from the regions demar- lower panels

TCTTTGTCAGA-3′). The mice were genotyped Cambridge, UK) was used for IHC. The expres-
to ensure that they were clear of the rd8 mutation. sion of Ctrp5 transcript was normalized against
The maintenance and care of mice were in accor- GAPDH [5].
dance with the ARVO statement for the Use of
Animals in Ophthalmic and Vision Research and
with protocols approved by the UCSD Institutional 3.3 Autofluorescence Imaging
Animal Care and Use Committee.
SPECTRALIS HRA + OCT scanning laser oph-
thalmoscopy (Heidelberg Engineering, Inc.) was
3.2 Evaluation of the Expression used to take autofluorescence images of murine
of CTRP5 and the Transcript retina. The method was a modified protocol from
that described previously [5]. To facilitate count-
Expression of Ctrp5 transcript and protein was ing deep retinal AF spots, the initial focus for
studied in 2-mo Ctrp5−/− and WT mice RPE-­ images was just in front of the nerve fiber layer,
choroid tissue by qRT PCR, Western blot analysis, and then images were taken in 0.5 diopter steps
and immunohistochemistry (IHC) as descried until the posterior vessels emerging from the disc
­earlier [5]. The anti-CTRP5 polyclonal antibody were in focus to ensure that deep AF spots would
which was generated in-house (1:500) and has be at their best focus. The autofluorescence image
previously been used for western blotting [7], composed of 10 frame per fundus was taken with
­
while the anti-CTRP5 (1:100, ab36893, Abcam, a 55° angle lens. AF spots were counted from WT
A Novel Mouse Model for Late-Onset Retinal Degeneration (L-ORD) Develops RPE Abnormalities… 339

Fig. 4 Retinal ultrastructure in Ctrp5−/− mice brane, which increased in size by 18.5 mo (pound signs in
In Ctrp5−/− mice, arrows in panels B, C, D, F, G, and H panels D and H). The asterisks in panels D and H repre-
represent vacuoles in RPE. The pound sign in panels C sent the presence of an electron dense lipid-based material
and G represent dense sub-RPE deposits in Bruch’s mem- in the RPE of 18.5-mo Ctrp5−/− mice

and Ctrp5−/− mice aged 19 mo (n = 3 for each), L-ORD and the previously characterized Ctrp5+/−
from one field of view centered on the optic disc mouse model [1, 2, 5, 10]. The Ctrp5−/− mice
by a masked observer. developed AF spots which were distributed
throughout the retina, similar to Ctrp5+/− mice.
The number of AF spots increased with age.
3.4 Histology and Ultrastructure The major pathological features of human
Analysis of the Retina L-ORD include the presence of thick sub-RPE
deposits, Bruch’s membrane and RPE abnormal-
Retinal morphology and ultrastructure of eyes of ities and neuroretinla atrophy in late-stage dis-
6-, 11-, and 18.5-mo-old Ctrp5−/− and 21-mo WT ease [1, 11, 12]. Similarly, the Ctrp5+/− mouse
control mice were analyzed by LM and TEM as model also developed sub-RPE deposits and
described earlier [5]. For TEM, the sample from Bruch’s membrane abnormalities as early as 6
18.5-mo Ctrp5−/− mice was processed using an mo and progressive accumulation of focal basal
osmium–tannic acid–phenylenediamine (OTAP) laminar deposits (BLamD) and basal linear
method to preserve lipids [9]. All other sections deposits with age [5, 6]. In the present study,
underwent standard processing. Ctrp5−/− mice developed dense sub-RPE deposits
in Bruch’s membrane by 11 mo of age, and by
18.5 mo, Ctrp5−/− mice had significant basal lam-
4 Discussion inar deposits. An interesting feature in the
Ctrp5−/− mouse retina was the development of
A systematic analysis of the ocular phenotype of electron-dense basal laminar deposits (BLamDs).
the homozygous Ctrp5 gene knock-out model The presence of BLamDs is also a common find-
(Ctrp5−/−) revealed RPE pathology consistent ing in early AMD eyes [13]. Formation of these
with the phenotype observed in patients with deposits has been linked to RPE stress both in
340 S. Borooah et al.

mouse models and AMD [14–16]. The RPE in from a CTRP5 gene mutation. Invest Ophthalmol Vis
Sci. 2005;46(9):3363–71.
Ctrp5−/− mice showed signs of stress as early 6 5. Chavali VR, Khan NW, Cukras CA, Bartsch DU,
mo of age with vacuolization with progressive Jablonski MM, Ayyagari R. A CTRP5 gene S163R
RPE abnormalities. These findings in Ctrp5−/− mutation knock-in mouse model for late-onset retinal
mice suggest that the presence of CTRP5/ degeneration. Hum Mol Genet. 2011;20(10):2000–14.
6. Sahu B, Chavali VR, Alapati A, Suk J, Bartsch DU,
C1QTNF5 is critical for maintaining the normal Jablonski MM, et al. Presence of rd8 mutation does
structure and function of RPE, particularly at an not alter the ocular phenotype of late-onset retinal
older age. degeneration mouse model. Mol Vis. 2015;21:273–84.
It is interesting to note that the phenotype 7. Mandal MN, Vasireddy V, Reddy GB, Wang X,
Moroi SE, Pattnaik BR, et al. CTRP5 is a membrane-­
observed in both Ctrp5+/− and Ctrp5−/− mouse associated and secretory protein in the RPE and
models is similar and recapitulates the deposit ciliary body and the S163R mutation of CTRP5
forming phenotype with RPE abnormalities impairs its secretion. Invest Ophthalmol Vis Sci.
observed in L-ORD patients. The findings in 2006;47(12):5505–13.
8. Zou YR, Müller W, Gu H, Rajewsky K. Cre-­
these mouse models suggest that the disease loxP-­mediated gene replacement: a mouse strain
pathology of L-ORD in patients and Ctrp5+/− producing humanized antibodies. Curr Biol.
mice may result from a dominant negative mech- 1994;4(12):1099–103.
anism leading to the loss of functional CTRP5. 9. Curcio CA, Presley JB, Millican CL, Medeiros
NE. Basal deposits and drusen in eyes with age-­
These models may aid in understanding the related maculopathy: evidence for solid lipid par-
mechanism of L-ORD pathology and serve as ticles. Exp Eye Res. 2005;80(6):761–75.
tools for preclinical evaluation of therapeutic 10. Cukras C, Flamendorf J, Wong WT, Ayyagari R,
strategies. Cunningham D, Sieving PA. Longitudinal structural
changes in late-onset retinal degeneration. Retina.
2016;36(12):2348–56.
Acknowledgments R01-EY030591, RO1-EY21237, 11. Duvall J, McKechnie NM, Lee WR, Rothery S,
T32-EY026590, P30-EY22589, R01-EY031663, Marshall J. Extensive subretinal pigment epithelial
Foundation Fighting Blindness, Nixon Vision Foundation, deposit in two brothers suffering from dominant reti-
Viterbi Family Fund, Edward N. and Della L. Thome nitis pigmentosa. A histopathological study. Graefes
Memorial Foundation, a Research to Prevent Blindness Arch Clin Exp Ophthalmol. 1986;224(3):299–309.
Challenge Grant to the Hamilton Eye Institute. 12. Milam AH, Curcio CA, Cideciyan AV, Saxena S,
John SK, Kruth HS, et al. Dominant late-onset
retinal degeneration with regional variation of sub-­
retinal pigment epithelium deposits, retinal function,
References and photoreceptor degeneration. Ophthalmology.
2000;107(12):2256–66.
1. Kuntz CA, Jacobson SG, Cideciyan AV, Li ZY, Stone 13. Curcio CA. Soft drusen in age-related macular degen-
EM, Possin D, et al. Sub-retinal pigment epithelial eration: biology and targeting via the oil spill strategies.
deposits in a dominant late-onset retinal degeneration. Invest Ophthalmol Vis Sci. 2018;59(4):AMD160–81.
Invest Ophthalmol Vis Sci. 1996;37(9):1772–82. 14. Sarks S, Cherepanoff S, Killingsworth M, Sarks
2. Ayyagari R, Griesinger IB, Bingham E, Lark KK, J. Relationship of basal laminar deposit and membra-
Moroi SE, Sieving PA. Autosomal dominant hem- nous debris to the clinical presentation of early age-­
orrhagic macular dystrophy not associated with the related macular degeneration. Invest Ophthalmol Vis
TIMP3 gene. Arch Ophthalmol. 2000;118(1):85–92. Sci. 2007;48(3):968–77.
3. Hayward C, Shu X, Cideciyan AV, Lennon A, Barran 15. Toomey CB, Kelly U, Saban DR, Bowes
P, Zareparsi S, et al. Mutation in a short-chain col- RC. Regulation of age-related macular degeneration-­
lagen gene, CTRP5, results in extracellular deposit like pathology by complement factor H. Proc Natl
formation in late-onset retinal degeneration: a genetic Acad Sci U S A. 2015;112(23):E3040–9.
model for age-related macular degeneration. Hum 16. Sura AA, Chen L, Messinger JD, Swain TA, McGwin
Mol Genet. 2003;12(20):2657–67. G Jr, Freund KB, et al. Measuring the contributions of
4. Ayyagari R, Mandal MN, Karoukis AJ, Chen L, basal laminar deposit and Bruch’s membrane in age-­
McLaren NC, Lichter M, et al. Late-onset macular related macular degeneration. Invest Ophthalmol Vis
degeneration and long anterior lens zonules result Sci. 2020;61(13):19.
Comparison of Mouse Models
of Autosomal Dominant Retinitis
Pigmentosa Due to the P23H
Mutation of Rhodopsin

Shannon R. Barwick and Sylvia B. Smith

Abstract cause of autosomal dominant retinitis pigmen-


tosa. Here, we provide a brief review of the
The need for robust and reliable animal mod- Rho-P23H mouse models currently available
els is a crucial step in studying any disease. for research.
This certainly applies to inherited retinal
degenerative diseases, in which mutations of Keywords
retinal specific genes result in photoreceptor
cell death and subsequent visual loss. Animal Retinitis pigmentosa · Retina · Mouse model ·
models of retinal gene mutations have proven Rho-P23H · Photoreceptor
valuable to our understanding of disease
mechanisms and as tools to evaluate therapeu-
tic intervention strategies. Notable among 1 Introduction
these models are mice with a mutation of the
rhodopsin gene at amino acid 23 in which pro- Many retinal diseases lead to varying degrees of
line is substituted for histidine (Rho-P23H). visual impairment. Mutations in over 330 genes
The RHO-P23H mutation is the most common are currently known to cause some form of reti-
nal disease [15]. One common form of retinal
disease is retinitis pigmentosa (RP), an inherited
S. R. Barwick (*)
Department of Cellular Biology and Anatomy, retinal degenerative disease (IRD) that leads to
Medical College of Georgia at Augusta University, the death of rod photoreceptor cells (PRCs). Its
Augusta, GA, USA prevalence worldwide is approximately 1:4000
The James and Jean Culver Vision Discovery [19]. RP can be inherited as autosomal dominant
Institute, Augusta University, Augusta, GA, USA (adRP), autosomal recessive (arRP), or X-linked.
e-mail: sbarwick@augusta.edu Mutations in proteins, which function or are
S. B. Smith expressed primarily in the retina, can lead to non-­
Department of Cellular Biology and Anatomy, syndromic RP, whereas mutations in genes
Medical College of Georgia at Augusta University,
Augusta, GA, USA encoding proteins found in a diverse number of
cells (and have cellular functions) outside the eye
The James and Jean Culver Vision Discovery
Institute, Augusta University, Augusta, GA, USA can result in systemic disease (syndromic RP).
The majority of RP cases are autosomal recessive
Department of Ophthalmology, Medical College of
Georgia at Augusta University, Augusta, GA, USA (arRP) and are often syndromic. Autosomal dom-
e-mail: sbsmith@augusta.edu inant cases comprise the second largest group of

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 341
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_49
342 S. R. Barwick and S. B. Smith

inheritance with most mutations occurring in the 2 Mouse Models of the Rho-­
gene encoding rhodopsin (RHO (human) Rho P23H Mutation
(mouse)). Rhodopsin, the light-sensitive visual
pigment, is located in rod outer segment disks. It A number of attempts to recapitulate the human
plays a critical role in rod disk morphogenesis RHO-P23H phenotype have been conducted in
and visual transduction [9]. The most commonly mice [11–13, 16]. Table 1 summarizes the gen-
mutated gene leading to autosomal dominant RP eration and phenotypical presentation of these
(adRP) in humans is the proline to histidine sub- various models, and for clarity of writing, we
stitution at amino acid 23 of rhodopsin (RHO-­ refer to the models by the first authors’ last name.
P23H) [4]. These models have proven useful to study mech-
While RP typically leads to the loss of rod anisms of adRP. Common methods for creating
PRCs initially, cone PRC loss follows [10], which these mouse models include transgene and
is particularly debilitating since cones mediate knock-in technologies. The transgene method
best vision. Clinical features of RP manifest most randomly incorporates the gene of interest into
often in early adulthood. Patients present with the host genome. In contrast, knock-in methods
night blindness and decreased peripheral vision. specifically target the gene of interest to a spe-
Late-stage RP can be identified by fundus exam cific locus of the target genome. Each of these
that reveals pigmentary deposits (bone spicules) genetic alteration strategies has advantages and
thought to be formed due to death of PRCs. At disadvantages that are discussed below.
advanced stages of RP, daylight vision becomes A groundbreaking discovery by the Berson
impaired and retinal vessel attenuation is seen lab occurred when the first genetic mutation
[4]. The progression of RP is typically slow; directly linked to adRP, RHO-P23H, was reported
however, due to the heterogeneous nature of the [5]. Following this discovery, many labs sought
disease, this can be difficult to predict [17]. to study this new mutation. One of the first labs
Patients with the same genetic mutation can vary created a humanized RHO-P23H model utilizing
in age of disease onset as well as progression a mutant allele isolated from a patient known to
rates. This makes diagnosing and treating patients have adRP. This model presented with attenuated
with RP challenging. Currently, there is no cure retinal vasculature, normal inner retina, and
for patients with RP creating the need for diverse decreased electrophysiological responses similar
and reliable models to study RP. A plethora of to human but did show irregular pigmentary
models ranging from mouse and rat to rabbit and deposition. Another feature observed was mislo-
pig have been created to study differing causes calization of rhodopsin within the rod PRCs lead-
and treatments of RP. In this review we describe ing to retinal degeneration only when misfolded
several mouse models of the RHO-P23H muta- protein levels were high [12]. Concurrently, Dr.
tion that are relevant to the study of adRP and Muna Naash and colleagues created a mouse
work underway using these models to advance transgene model utilizing oligonucleotide-­
our understanding and treatment of this disease. directed mutagenesis containing three point

Table 1 Summary of Rho-P23H mouse models


Model Type Phenotype Model creation method
Olsson Human Slow degenerative phenotype, variable, Mutant allele from patient cloned,
[12] transgenic transgene expression, irregular pigmentation injected into mouse embryos
Naash Mouse Slow degenerative phenotype, variable, Oligonucleotide-directed mutagenesis
[11] transgenic transgene expression, no pigmentary changes of murine opsin gene
Price Human Slow degenerative phenotype, mislocalized Targeting vector incorporated using
[13] knock-in protein site-directed mutagenesis
Sakami Mouse Slow degenerative phenotype, closely mimics Targeting vector stem cell injected into
[16] knock-in human disease mouse blastocyst
Comparison of Mouse Models of Autosomal Dominant Retinitis Pigmentosa Due to the P23H Mutation… 343

mutations, including Pro-23-His, in exon 1 of the structure showed greater retinal alteration in the
germ line mouse opsin gene. This model pre- inferior retina compared to the superior retina
sented with gradually declining a/b-wave ampli- [16]. Utilizing this new information, Sakami and
tudes of the scotopic electroretinography (ERG) co-workers created a knock-in Rho-P23H mouse
s. It also appeared that the rod PRC death pro- model to improve upon the previous knock-in
ceeded cone PRC death, much like the human model created by Price and colleagues. The
disease [11]. Interestingly, the RPE did not show Sakami mouse model demonstrates a slow pro-
any pigmentary deposition, which is a hallmark gressive retinal degeneration in which half of the
of human RP. This model expressed the normal rods die before cone death is detected. In addi-
and mutant rhodopsin in relatively equal quanti- tion, a gradual shortening and disorganization of
ties, but misfolded mutant rhodopsin was mislo- outer segments precedes rod PRC death in this
calized to the outer plexiform layer (OPL) and model. A notable feature of the Sakami mouse
outer segments [11]. The lack of pigmentary model is that its degeneration pattern shows
deposits and the variable expression of normal regional retinal degenerative differences such
and mutant rhodopsin were considered limita- that the inferior retina degenerates more rapidly
tions of these transgene models. To aid in the than the superior retina, which is similar to the
ability to understand and study the misfolded superior-inferior differences observed in human
P23H rhodopsin, Dr. Brandee Price and col- RP patients mentioned above. Other features of
leagues created a humanized knock-in mouse the Sakami mouse model are that the scotopic
model containing a RHO-P23H-rhodopsin-­ ERG amplitudes decline slowly followed by
GFP. This model not only allowed for tracking of gradual decline in the photopic ERG. WT and
the P23H-rhodopsin but also utilized knock-in mutant rhodopsin protein production are equiva-
techniques. By so doing, the investigators were lent. Interestingly, it does not appear that rhodop-
able to directly target the mutant gene to the loca- sin is mislocalized, which is present in other
tion of interest allowing for the production of a Rho-P23H mouse models [12, 13]. In aggregate,
slow degenerative phenotype without the need the aforementioned similarities of the humanized
for multiple transgene insertions. This decreased mouse model created in the Palczewski labora-
the variability of normal and mutant rhodopsin tory to human adRP patients make this mutant
expression [13]. These three models were mouse model a particularly attractive tool for
groundbreaking for the study of retinal degenera- investigating mechanisms of disease and inter-
tive mechanisms in adRP; however, they still did ventional strategies.
not fully recapitulate human disease.
The advent of optical coherence tomography
(OCT), which allows in situ visualization of reti- 3 Pathogenic Mechanism
nal structure in human patients (as well as animal Studies
models), paved the way for optimizing a mouse
model that more closely reflects human retinal While the long-term goal of many studies of
structure under disease conditions. To better mouse models of retinal disease is to develop
understand how the P23H mutation of rhodopsin successful treatment strategies, the mechanism(s)
presents in human patients, Dr. Sanae Sakami underlying loss of PRCs must be understood if
and colleagues from Dr. Krystoff Palczewski’s those strategies are to move from the bench to the
laboratory analyzed a cohort of 19 human adRP clinic. Regarding the RHO-P23H mutation, sev-
patients with the RHO-P23H mutation. The eral mechanisms have been investigated as under-
patients were subjected to scotopic and photopic lying the degeneration of PRCs. It is known that
ERG analysis and retinal cross-sectional imaging if misfolded proteins are not properly degraded,
using OCT. Rod ERGs were abnormally reduced they can be retained within the endoplasmic
in all patients, whereas cone responses varied. reticulum (ER) which can lead to ER stress and
Importantly, the in situ assessment of retinal activation of the unfolded protein response
344 S. R. Barwick and S. B. Smith

(UPR). The Palczewski group has reported could pose a promising therapeutic target. Other
incomplete glycosylation of rhodopsin in the labs are investigating therapeutic intervention by
Sakami model, which led to retention of protein genetically modulating the immune system [1] in
within the ER. Further investigation, however, addition to direct modification of the Rho-P23H
indicated that reduced glycosylation was detected mutation [8]. These studies show promising
only in small quantities, suggesting protein is improvements in retinal structure and function
rapidly targeted for degradation differing from and are another encouraging avenue of research
previous data suggesting misfolded protein is for treatment of IRDs.
retained within the ER and induces ER stress Here we have discussed various models of the
[16]. The Sakami mouse model was later crossed Rho-P23H mutation and existing research utiliz-
with an ER stress-activated indicator, and robust ing them. The current ongoing research is prom-
signal was observed within rod PRCs [2]. ising for the future of patients with the RHO-P23H
Subsequent research performed in various Rho-­ mutation as well as other retinal degenerations,
P23H mouse models suggests ER stress and UPR but much work is still needed to translate current
activation may play a role in activation of cell findings to the clinic.
death pathways, but this may not be the whole
story. Comitato and co-workers found that cal- Acknowledgments We gratefully acknowledge NIH
pain activation (characteristic of the apoptotic (RO1EY028103, P30EY031631) and Foundation
Fighting Blindness (TA-NMT-0617-0721-AUG) for their
pathway of cell death) may be contributing more support.
to induction of cell death than ER stress-induced
UPR, more so in the Olsson model than the
Sakami model, suggesting activation of ER stress References
alone may not be sufficient to induce cell death
[3]. Due to the key role ER stress and proper pro- 1. Aguilà M, Bellingham J, Athanasiou D, Bevilacqua
tein folding play in the progression of retinal D, Duran Y, Maswood R, Parfitt DA, Iwawaki T,
degeneration, several labs have investigated Spyrou G, Smith AJ, Ali RR. AAV-mediated ERdj5
overexpression protects against P23H rhodopsin tox-
inducing misfolded protein clearance (or protein icity. Hum. Mol. Genet. 2020;29(8):1310–8.
folding assistance mechanisms) to attenuate reti- 2. Chiang WC, Kroeger H, Sakami S, Messah C,
nal degeneration. The Liu laboratory treated Yasumura D, Matthes MT, Coppinger JA, Palczewski
Sakami mice with methotrexate to pharmacologi- K, LaVail MM, Lin JH. Robust endoplasmic reticulum-­
associated degradation of rhodopsin precedes retinal
cally clear misfolded rhodopsin and observed degeneration. Mol. Neurobiol. 2015;52(1):679–95.
improved protein degradation. Chemical chaper- 3. Comitato A, Schiroli D, La Marca C, Marigo
ones such as vitamin A, 4-phenylbutyrate, and V. Differential contribution of calcium-activated
curcumin have also been utilized to improve rho- proteases and ER-stress in three mouse models
of retinitis pigmentosa expressing P23H mutant
dopsin folding and transport. Importantly, func- RHO. Retin. Degener. Dis. Cham: Springer; 2019.
tional improvement was detected by ERG in p. 311–6.
some of these therapeutic strategies [6]. 4. Dias MF, Joo K, Kemp JA, Fialho SL, da Silva Cunha
Autophagy and altered proteasome activity have Jr A, Woo SJ, Kwon YJ. Molecular genetics and
emerging therapies for retinitis pigmentosa: basic
been proposed as endogenous mechanisms that research and clinical perspectives. Prog. Retin. Eye
clear misfolded proteins and reduce PRC death. Res. 2018;63:107–31.
The Sakami model is known to have increased 5. Dryja TP, McGee TL, Reichel E, Hahn LB, Cowley
autophagy [18]. This led several labs to investi- GS, Yandell DW, Sandberg MA, Berson EL. A point
mutation of the rhodopsin gene in one form of retinitis
gate whether decreasing autophagy improves pigmentosa. Nature. 1990;343(6256):364–6.
proteosomal activity and restores their balance 6. Liu X, Feng B, Vats A, Tang H, Seibel W, Swaroop
within the cell, thereby improving retinal struc- M, Tawa G, Zheng W, Byrne L, Schurdak M, Chen
ture and function [7, 14]. This research suggests Y. Pharmacological clearance of misfolded rhodopsin
for the treatment of RHO-associated retinitis pigmen-
misfolding and degradation of proteins may play tosa. FASEB J. 2020;34(8):10146–67.
a major role in retinal disease progression and
Comparison of Mouse Models of Autosomal Dominant Retinitis Pigmentosa Due to the P23H Mutation… 345

7. Lobanova ES, Finkelstein S, Li J, Travis AM, Hao degradation of human P23H-rhodopsin-GFP in a


Y, Klingeborn M, Skiba NP, Deshaies RJ, Arshavsky knockin mouse model of retinitis pigmentosa. Invest.
VY. Increased proteasomal activity supports Ophthalmol. Vis. Sci. 2011;52(13):9728–36.
­photoreceptor survival in inherited retinal degenera- 14. Qiu Y, Yao J, Jia L, Thompson DA, Zacks
tion. Nat. Commun. 2018;9(1):1–1. DN. Shifting the balance of autophagy and pro-
8. Mao H, James T Jr, Schwein A, Shabashvili AE, teasome activation reduces proteotoxic cell death: a
Hauswirth WW, Gorbatyuk MS, Lewin AS. AAV novel therapeutic approach for restoring photorecep-
delivery of wild-type rhodopsin preserves retinal func- tor homeostasis. Cell Death Dis. 2019;10(8):1–4.
tion in a mouse model of autosomal dominant retinitis 15. RetNet [Internet]. Houston (TX):The University of
pigmentosa. Hum. Gene Ther. 2011;22(5):567–75. Texas Health Science Center; c1996–2021. Available
9. Mendes HF, Van Der Spuy J, Chapple JP, Cheetham online at https://sph.uth.edu/retnet/
ME. Mechanisms of cell death in rhodopsin retinitis 16. Sakami S, Maeda T, Bereta G, Okano K, Golczak
pigmentosa: implications for therapy. Trends Mol. M, Sumaroka A, Roman AJ, Cideciyan AV, Jacobson
Med. 2005;11(4):177–85. SG, Palczewski K. Probing mechanisms of photo-
10. Hamel C. Retinitis pigmentosa. Orphanet J. Rare Dis. receptor degeneration in a new mouse model of the
2006;1(1):1–2. common form of autosomal dominant retinitis pig-
11. Naash MI, Hollyfield JG, Al-Ubaidi MR, Baehr mentosa due to P23H opsin mutations. J. Biol. Chem.
W. Simulation of human autosomal dominant reti- 2011;286(12):10551–67.
nitis pigmentosa in transgenic mice expressing a 17. Van Soest S, Westerveld A, De Jong PT, Bleeker-­
mutated murine opsin gene. Proc. Natl. Acad. Sci. Wagemakers EM, Bergen AA. Retinitis pigmen-
1993;90(12):5499–503. tosa: defined from a molecular point of view. Surv.
12. Olsson JE, Gordon JW, Pawlyk BS, Roof D, Hayes Ophthalmol. 1999;43(4):321–34.
A, Molday RS, Mukai S, Cowley GS, Berson EL, 18. Yao J, Qiu Y, Frontera E, Jia L, Khan NW, Klionsky DJ,
Dryja TP. Transgenic mice with a rhodopsin mutation Ferguson TA, Thompson DA, Zacks DN. Inhibiting
(Pro23His): a mouse model of autosomal dominant autophagy reduces retinal degeneration caused by
retinitis pigmentosa. Neuron. 1992;9(5):815–30. protein misfolding. Autophagy. 2018;14(7):1226–38.
13. Price BA, Sandoval IM, Chan F, Simons DL, Wu 19. Zhang Q. Retinitis pigmentosa: progress and perspec-
SM, Wensel TG, Wilson JH. Mislocalization and tive. Asia-Pac J Ophthalmol. 2016;5(4):265–71.
Compensatory Cone-Mediated
Mechanisms in Inherited Retinal
Degeneration Mouse Models:
A Functional and Gene Expression
Analysis

Alicia A. Brunet, David M. Hunt, Carla Mellough,


Alan R. Harvey, and Livia S. Carvalho

Abstract as an increase in cone-specific genes in the


Rd1 at the peak of rod death. We show that
The retina undergoes compensatory changes Cnga3−/− and Pde6ccpfl1 mice maintained phot-
in response to progressive photoreceptor loss/ opic visual acuity via optomotor responses,
dysfunction; however, studies of inherited despite no recordable cone electroretinogram
retinal diseases (IRDs) often lack a temporal (ERG), while functional measures and photo-
connection between gene expression and receptors loss were correlated in Rd1 mice.
visual function. Here, we used three mouse There were also significant changes to oscilla-
models of IRD – Cnga3−/−, Pde6ccpfl1, and tory potentials (OPs) in Cnga3−/− and
Rd1 – to investigate over time the effect of Pde6ccpfl1, implying an effect on inner retinal
photoreceptor degeneration, particularly cells as a result of cone degeneration. These
cones, on visual function and gene expression. results indicate a potentially malleable retinal
Changes to gene expression include increases environment following cone degeneration;
in cell survival and cell death genes in however, further investigation is needed to
Pde6ccpfl1 before significant cell loss, as well elucidate how these changes compensate for
the loss of cone function.
A. A. Brunet (*) · D. M. Hunt · C. Mellough ·
L. S. Carvalho Keywords
Centre for Ophthalmology and Visual Sciences, The
University of Western Australia, Crawley, WA,
Australia Vision · Inherited retinal disease ·
Achromatopsia · Retinitis pigmentosa · cpfl1 ·
Lions Eye Institute Ltd., Nedlands, WA, Australia
e-mail: alicia.brunet@research.uwa.edu.au; david. rd1 · cnga3 · Mechanisms · Cell death
hunt@uwa.edu.au; carlamellough@lei.org.au;
liviacarvalho@lei.org.au
A. R. Harvey
School of Human Sciences, The University of 1 Introduction
Western Australia,
Crawley, WA, Australia Cone degeneration varies between inherited reti-
Perron Institute for Neurological and Translational nal disease (IRD) mutations in severity and time
Science, course. Previous studies in IRDs have mainly
Nedlands, WA, Australia
focused on morphological and physiological
e-mail: alan.harvey@uwa.edu.au

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 347
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_50
348 A. A. Brunet et al.

changes in the retina at peak photoreceptor zine) using the Celeris ERG (Diagnosys, USA)
degeneration; however, there is a lack of research as previously described [4]. Mice were dark-­
on temporal changes in IRD mice during degen- adapted overnight prior to ERG recordings.
eration. Furthermore, several studies have shown Scotopic responses and oscillatory potentials
a high level of retinal plasticity, leading to poten- (OP1 and OP2) were recorded, followed by light
tial compensatory mechanisms, in response to adaptation for 10 min at 30 cd.s.m.−2 before phot-
photoreceptor degeneration. Synaptic plasticity opic recordings. Responses to a series of increas-
is prevalent in the Cnga3−/− model of achroma- ing light flashes were recorded, but only data at
topsia (ACHM) and in the Rho−/− model of retini- 25 cd.s.m.−2 are presented. Data were analyzed
tis pigmentosa (RP), where remaining using the Diagnosys Espion software and
photoreceptors form ectopic synapses between Microsoft Excel.
cones and rod-bipolar cells, and rods and cone-­
bipolar cells, respectively [1]. Synaptic remodel-
ing is also observed in the Rd1 and Rd10 RP 2.3 Optomotor Response
models, including loss of dendrites, cell body
migration of horizontal cells, and ectopic synapse Optomotor responses were assessed using the
formation with rod bipolar cells [2, 3]. However, automated OptoDrum (Striatech) to measure the
how these mechanisms preserve or contribute to visual reflex to rotating stimuli. The OptoDrum is
vision in IRD models remains unclear. To evalu- a closed box containing four screens that simulate
ate how the course of cone degeneration affects a rotating cylinder of alternating white and black
the visual response, this study combines gene stripes. Mice were placed on a raised platform at
expression and physiological and behavioral the center of the box, with a camera directly
approaches in two cone-dystrophy (ACHM) and above. Movement was tracked using computer-
one rod-cone dystrophy (RP) mouse model. controlled software. Mice were dark-­ adapted
overnight before scotopic recordings, followed by
light adaptation for 2 h and photopic recordings.
2 Materials and Methods Contrast sensitivity and visual acuity (cycles/
degree) were recorded under scotopic (2 mLux)
2.1 Animals and photopic (70 Lux) conditions, respectively.

Mouse (C57BL/6 J background) experiments


were approved by the Harry Perkins Institute 2.4 Flow Cytometry
Animal Ethics Committee, bred and housed at
the Perkins Bioresources Facility, and conducted Whole retinae were dissociated as previously
in accordance with the ARVO Statement for the described [4], and flow cytometry was performed
Use of Animals in Ophthalmic and Vision with a BD FACSMelody Cell Sorter to quantify
Research. All IRD lines used in this study GFP+ cell number.
(Cnga3−/−, Pde6ccpfl1,and Rd1) express green flu-
orescent protein (GFP) in cone photoreceptors, as
previously described [4], and are referred to as 2.5 qRT-PCR Analysis
(IRD line).GFP. The Chrnb4.GFP model [5] was
used as the wild type, also expressing GFP cones Total RNA was extracted from whole retinae using
(referred to as WT). TriReagent (Sigma-Aldrich), and reverse transcrip-
tion was performed using the QuantiTect Reverse
Transcription Kit (Qiagen) as per manufacturer’s
2.2 Electroretinography (ERG) instructions. qRT-PCR was done on a Bio-Rad CFX
Connect Real-Time System using Taqman Fast
ERGs were performed on mice under general Advanced mastermix (ThermoFisher). Relative
anesthesia (40 mg/kg ketamine, 5 mg/kg xyla- expression was expressed as fold change (2-ΔΔct).
Compensatory Cone-Mediated Mechanisms in Inherited Retinal Degeneration Mouse Models… 349

2.6 Statistical Analysis phosphodiesterase 6C, Pde6c) were decreased


overtime in Cnga3.GFP and Pde6c.GFP com-
Data was analyzed using GraphPad PRISM by pared to WT (Fig. 1). However, in Rd1.GFP there
Student’s t-test or two-way ANOVA (post hoc was increased expression of cone genes, particu-
Tukey’s test). Results were expressed as larly Pde6c at P24 (Fig. 1). Rod-specific genes
mean ± standard error of the mean (SEM). (phosphodiesterase 6B, Pde6b; rhodopsin, Rho)
in Cnga3.GFP and Pde6c.GFP were comparable
to WT, except an increase in Pde6b at P16 in
3 Results Pde6c.GFP, and were significantly decreased in
Rd1.GFP. In Pde6c.GFP, cell survival (B-cell
3.1 Compensatory Gene lymphoma, Bcl2; Bcl2 like 1, Bcl2l1; rod-derived
Expression Changes cone viability factor; RdCVF) and cell death
in Response to Cell Death genes (apoptotic peptidase activating factor 1,
Apaf1, Bcl2-associated x protein, Bax, caspase-8,
To examine how photoreceptor degeneration Casp8) were increased at P32 compared to WT,
contributes to changes in gene expression, we but decreased by P60 (Fig. 1).
first assessed cone numbers over time. Cone
numbers showed no significant difference
between Cnga3.GFP and WT, but were signifi- 3.2 ERG and Behavioral Visual
cantly decreased in Pde6c.GFP and Rd1.GFP Responses Persist
(Fig. 1). Significantly fewer cones were observed in the Absence of Cone
at P60 for Pde6c.GFP and P24 for Rd1.GFP. Function
Expression of photoreceptor, cell-survival,
and cell-death genes during the course of degen- Assessment of photoreceptor function via ERG
eration were analyzed using qRT-PCR. Cone-­ showed no photopic responses in Cnga3.GFP
specific genes (cone arrestin, Arr3; cyclic and Pde6c.GFP (Fig. 2a, b). At P16, Rd1.GFP
nucleotide-gated channel subunit alpha 3, Cnga3; had a decreased photopic ERG b-wave (Fig. 2b),
G protein subunit alpha transducing 3; Gnat2; although photopic a-wave levels were compara-

Fig. 1 Changes to gene expression in response to photo- (P < 0.05), which was lost at P60. Cone- and rod-specific
receptor cell death. Cone counts using flow cytometry gene expressions are shown. Relative expression was nor-
showed a significant reduction in average cone numbers in malized to Gapdh and shown as a fold change compared
Pde6c.GFP and Rd1.GFP (P < 0.05) but not in Cnga3. to age-matched WT samples using the ΔΔCt method
GFP (n ≥ 4). Pde6c.GFP had a significant increase in both (n = 3). *P < 0.05, **P < 0.005, ***P < 0.0005,
cell survival and cell death genes compared to WT at P32 ****P < 0.0001
350 A. A. Brunet et al.

Fig. 2 Photopic spatial frequency is retained in the a-wave response (P > 0.05). Scotopic b-wave responses
absence of cone ERG in cone dystrophy models. (a, b) were lower in Cnga3.GFP and Pde6c.GFP models when
Quantification of photopic ERG a-wave (a) and b-wave compared to WT after P16, but was only significant at
(b) responses in Cnga3.GFP, Pde6c.GFP, and Rd1.GFP P24 in Pde6c.GFP (P < 0.001) and P60 in Cnga3.GFP
models compared to WT. Photopic responses were (P < 0.05). (j) Scotopic contrast sensitivity was unchanged
unmeasurable in Cnga3.GFP and Pde6c.GFP, and Rd1. in Cnga3.GFP and Pde6c.GFP compared to age-matched
GFP past P16. (i) Cnga3.GFP and Pde6c.GFP photopic WT mice (P > 0.05) and were unmeasurable in Rd1.
optomotor had no significant difference compared to WT GFP. OPs were analyzed for amplitude (OP1, e, OP2, f)
(P > 0.05). Rd1.GFP had reduced P16 (P < 0.05) and P24 and latency (OP1, g, OP2, h). There was a significant
(P < 0.0001) photopic response before being unmeasur- decrease in OP1 and OP2 response in Cnga3.GFP and
able at P32. Scotopic a-wave (c) and b-wave (d) responses Pde6c.GFP compared to WT at various timepoints
were significantly reduced in Rd1.GFP line (P < 0.0001). (P < 0.05). There was no difference in latency except at
There was no significant difference between Cnga3.GFP P16 in OP2 of Pde6c.GFP (P < 0.0005). n ≥ 4 per age per
and Pde6c.GFP models compared to WT in the scotopic line, *P < 0.05, ***P < 0.0005, ****P < 0.0001

ble to WT (Fig. 2a), but both were lost by P24. but no optomotor scotopic response was detect-
Cnga3.GFP and Pde6c.GFP had a sustained able in Rd1.GFP at all ages (Fig. 2j). OP1 and
optomotor response under photopic conditions OP2 were isolated from the scotopic response of
across time, lower than WT past P16, but not sta- Cnga3.GFP and Pde6c.GFP, and analyzed for
tistically significant (Fig. 2i). Photopic optomo- amplitude (Fig. 2e, f) and latency (Fig. 2g, h). No
tor response was significantly lower in the Rd1. OPs were measurable in Rd1.GFP. OP1 (Fig. 2e)
GFP at P16, and decreased further until being and OP2 (Fig. 2f) amplitudes were significantly
lost by P32. decreased in both Cnga3.GFP and Pde6c.GFP at
In scotopic ERGs, P16 Rd1.GFP possessed a various ages after P16. OP latency was mostly
reduced response which was lost by P24 (Fig. 2c, consistent over time (Fig. 2g, h).
d). Scotopic responses in Cnga3.GFP and Pde6c.
GFP showed no differences in a-wave (Fig. 2c),
but a decreased b-wave compared to WT, signifi- 4 Discussion
cant at P24 in the Pde6c.GFP and P60 in the
Cnga3.GFP (Fig. 2d). Optomotor scotopic con- Our Rd1.GFP data replicates previous findings,
trast sensitivity showed no difference between with a peak in cone death around P24 [7].
Cnga3.GFP and Pde6c.GFP compared to WT, However, Pde6c.GFP has previously been
Compensatory Cone-Mediated Mechanisms in Inherited Retinal Degeneration Mouse Models… 351

reported to also have a peak in cone death at P24 The surprisingly WT-level photopic acuity
[8, 9], much earlier than described here (P60). response seen in Cnga3.GFP and Pde6c.GFP
Previous studies used terminal deoxynucleotidyl may suggest a takeover of the cone visual path-
transferase dUTP nick end labeling (TUNEL) to way, most likely by rods, with an equivalent
estimate dying cone numbers, while our study type of compensation not seen in Rd1.GFP
estimated the amount of viable cones, which may mice. Furthermore, significant changes to the
explain this disparity. Furthermore, distinct dif- scotopic OPs of the Cnga3.GFP and Pde6c.GFP
ferences in cell survival and cell death genes indicate changes to inner retinal structures. The
were found between P32 and P60 in Pde6c.GFP, origin of OPs is still not clear; however, it is
correlating with a marked decrease in cone num- thought that they originate from the inner retinal
bers at P60. Differential expression of cell sur- layer, occurring in the ascending limb of the
vival and cell death genes was not as distinct in scotopic b-wave [10]. Particularly, OP1 and
Cnga3.GFP, supporting the lack of cone loss that OP2 arise from bipolar and amacrine cells [11].
we report in this mouse line at the investigated The overall decrease in amplitude of OP1 and
timepoints. OP2 in Cnga3.GFP and Pde6c.GFP compared
In our cone dystrophy models, there was a to WT may therefore be a result of decreased
decrease in cone-specific gene expression. amacrine and bipolar cell function, perhaps con-
Interestingly, there was a general increase in nected to a “takeover” of the cone system by
cone-specific genes in Rd1.GFP, particularly of surviving rods.
Pde6c, where expression was almost sevenfold To conclude, we found significant changes in
higher at P24 compared to WT. Rd1.GFP has a gene expression at ages of significant cone loss.
defective Pde6b gene, and compensatory mecha- Retention of photopic optomotor responses
nisms may be the cause of the increase in the despite the lack of cone ERG in Cnga3.GFP and
expression of the paralogous Pde6c cone gene in Pde6c.GFP suggests changes to the retinal archi-
an attempt to facilitate phosphodiesterase func- tecture to compensate for cone loss. How exactly
tion in rods during peak cone degeneration. A these changes occur requires further study.
previous study has shown that supplementation Additionally, the effect on inner retinal function
of Pde6c in the Rd1 mouse partially recovered shown through changes to scotopic b-wave ERGs
vision [6]. The opposite is also seen in Pde6c. and decreased OP amplitudes suggests cone-­
GFP, as Pde6b was more than threefold higher dystrophies can have a negative impact on
than WT at P16, although this preceded the drop remaining viable retinal cells.
in Pde6c.GFP cone numbers.
Cnga3.GFP and Pde6c.GFP are cone-specific Acknowledgments This research was supported by a
IRDs with supposedly no detrimental effect on grant from the Lindsay and Heather Payne Medical
Research Charitable Foundation (IPAP2020/0504, LSC).
the remaining retinal cells. For the first time,
using optomotor visual responses, we show that
photopic-mediated vision is retained in these
mice in the absence of a photopic ERG. Such References
responses could potentially be mediated via mor- 1. Haverkamp S, et al. Synaptic plasticity in CNGA3−/−
phological changes previously reported to mice: cone bipolar cells react on the missing
second-­order neurons [1, 2] that may act to main- cone input and form ectopic synapses with rods.
tain visual function to some extent. This is further J. Neurosci. 2006;26(19):5248–55.
2. Phillips MJ, Otteson DC, Sherry DM. Progression of
justified by data from the Rd1.GFP model, with neuronal and synaptic remodeling in the rd10 mouse
significantly less photopic acuity at P16 com- model of retinitis pigmentosa. J. Comp. Neurol.
pared to WT.GFP, then a further decrease at P24 2010;518(11):2071–89.
before being undetectable at P32 and P60, mir- 3. Haq W, et al. Synaptic remodeling generates syn-
chronous oscillations in the degenerated outer mouse
roring the temporal progression of cone death in retina. Front Neural Circuit. 2014;8:108.
Rd1 mice [7].
352 A. A. Brunet et al.

4. Brunet AA, et al. Validating fluorescent chrnb4. 8. Arango-Gonzalez B, et al. Identification of a common
EGFP mouse models for the study of cone photo- non-apoptotic cell death mechanism in hereditary
receptor degeneration. Transl. Vis. Sci. Technol. retinal degeneration. PloS One. 2014;9(11):e112142.
2020;9(9):28–9. 9. Trifunović D, et al. cGMP-dependent cone pho-
5. Siegert S, et al. Genetic address book for retinal cell toreceptor degeneration in the cpfl1 mouse retina.
types. Nat. Neurosci. 2009;12(9):1197–204. J. Comp. Neurol. 2010;518(17):3604–17.
6. Majumder A, et al. Exchange of cone for rod phos- 10. Speros P, Price J. Oscillatory potentials. History,
phodiesterase 6 catalytic subunits in rod photore- techniques and potential use in the evaluation of dis-
ceptors mimics in part features of light adaptation. turbances of retinal circulation. Surv. Ophthalmol.
J. Neurosci. 2015;35(24):9225–35. 1981;25(4):237–52.
7. Narayan DS, et al. Spatio-temporal characteriza- 11. Wachtmeister L, Dowling JE. The oscillatory poten-
tion of S-and M/L-cone degeneration in the Rd1 tials of the mudpuppy retina. Invest. Ophthalmol. Vis.
mouse model of retinitis pigmentosa. BMC Neurosci. Sci. 1978;17(12):1176–88.
2019;20(1):1–17.
Inhibition of Ryanodine Receptor
1 Reduces Endoplasmic Reticulum
(ER) Stress and Promotes ER
Protein Degradation in Cyclic
Nucleotide-Gated Channel
Deficiency

Fan Yang, Hongwei Ma, Rekha Garg, Alfred Lewin,


and Xi-Qin Ding

Abstract Ryr1, manifested as increased expression


levels of cone proteins M-opsin, S-opsin, and
The cone photoreceptor cyclic nucleotide-­ cone arrestin. Knockdown of Ryr1 also led to
gated (CNG) channel plays a pivotal role in reduced ER stress and increased expression
cone phototransduction. Mutations in genes levels of the ER-associated degradation pro-
encoding the channel subunits CNGA3 and teins. This work demonstrates a role of Ryr1 in
CNGB3 account for about 80% of all cases of ER stress and cone degeneration in CNG
achromatopsia and are associated with pro- channel deficiency, and supports strategies tar-
gressive cone dystrophies. CNG channel defi- geting ER calcium regulation for cone
ciency leads to cellular/endoplasmic reticulum preservation.
(ER) calcium dysregulation and ER stress-­
associated cone apoptosis. This work investi- Keywords
gated the role of the ER calcium channel
Cyclic nucleotide-gated channel · Cone ·
ryanodine receptor 1 (Ryr1) in ER stress and
Retina · Ryanodine receptor · Endoplasmic
cone degeneration in CNG channel deficiency.
reticulum calcium homeostasis · Endoplasmic
The AAV-mediated CRISPR/SaCas9 genome
reticulum-associated degradation ·
editing was used to knock down Ryr1 specifi-
Endoplasmic reticulum stress
cally in cones. CNG channel-deficient mice
displayed improved cone survival after sub-
retinal injection of AAV2-SaCas9/gRNA-­
1 Introduction
F. Yang · H. Ma · X.-Q. Ding (*)
Departments of Cell Biology, University of The cone photoreceptor cyclic nucleotide-gated
Oklahoma Health Sciences Center,
Oklahoma City, OK, USA (CNG) channel is essential for phototransduc-
e-mail: Hongwei-ma@ouhsc.edu; xi-qin-ding@ tion. Upon binding of cyclic guanosine mono-
ouhsc.edu phosphate (cGMP) under dark conditions, CNG
R. Garg · A. Lewin channels open and permit the influx of the cal-
Department of Molecular Genetics and Microbiology, cium and sodium ions necessary to maintain the
University of Florida, Gainesville, FL, USA dark current and cellular calcium homeostasis.
e-mail: Garg@UFL.EDU; lewin@UFL.EDU

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 353
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_51
354 F. Yang et al.

Mutations in the CNGA3 and CNGB3 genes that conformed to the guidelines on the care and use
encode the channel subunits account for about of animals adopted by the Society for
80% of all cases of achromatopsia and are associ- Neuroscience and the Association for Research
ated with progressive cone dystrophies [2, 5]. in Vision and Ophthalmology (Rockville, MD).
These diseases are characterized by severely
impaired daylight vision and lack of color
­discrimination. Cone photoreceptors degenerate 2.2 Construction of AAV2/5-­
over time in patients and in mouse models of SaCas9/gRNA-Ryr1 Vector
CNG channel deficiency. With the disease mouse
models, we established that cone death involves The knockdown of Ryr1 in cones was achieved
endoplasmic reticulum (ER) stress-associated using an AAV-mediated CRISPR/SaCas9 genome
apoptosis and cytosolic/ER calcium dysregula- editing technology. The plasmid that expresses an
tion [3, 6]. ER calcium homeostasis is regulated epitope-tagged version of the Cas9 gene from
by two ER calcium channels, the inositol Staphylococcus aureus and a cassette for the
1,4,5-trisphosphate receptor (IP3R) and ryano- expression of guide RNAs containing the U6 pro-
dine receptors (RyR) for calcium efflux out of the moter was provided by Dr. Feng Zhang
ER into the cytosol, and the sarco−/endoplasmic (Massachusetts Institute of Technology), and
reticulum Ca2+-ATPase for calcium influx into modified for directing the expression specially in
the ER. There are three isoforms of RyR, Ryr1, cones. The modified plasmid permits packaging in
Ryr2, and Ryr3, and photoreceptors express all an AAV capsid and contains a hybrid promoter of
these three isoforms [1]. We previously showed the enhancer from the interphotoreceptor retinoid
that CNG channel deficiency leads to reduced binding protein and human transducin alpha-sub-
cytoplasmic calcium level and elevated expres- unit proximal promoter for expression of SaCas9
sion/activity of the ER calcium channels, includ- specifically in cones. The guide RNAs (gRNAs)
ing Ryr1 and Ryr2, and that suppression of ER for each of the first five exons of the coding region
calcium channels by chemical inhibitors or of Ryr1 were identified using the algorithm for
genetic deletion of IP3R1 or Ryr2 preserves cones gRNA design from the Broad Institute (https://
[1, 4, 7]. This work investigated the effects of portals.broadinstitute.org/gpp/public/analysis-­
knockdown of Ryr1, and the findings support the tools/sgrna-­design). We tested five guides in cell
view that Ryr1 contributes to ER stress and cone culture (mouse Neuro 2A cells), based on qRT-
degeneration in CNG channel deficiency. PCR detection for knockdown of exons 1–5 of
Ryr1. A sgRNA that led to significant reduction of
Ryr1 mRNA relative to a non-targeting control
2 Materials and Methods gRNA in the cells was cloned in an AAV2 vector,
and expressed using the U6 RNA pol III promoter.
2.1 Experimental Animals The gRNA sequence is
CCATCATGGGTGATGGAGGAG. The vector
The Nrl−/− mouse was provided by Dr. Anand DNAs were packaged in AAV5 capsids for high
Swaroop (National Eye Institute), the Cnga3−/− efficiency of photoreceptor transduction in mice
mouse was provided by Dr. Martin Biel and were produced using the two-plasmid co-­
(University of Munich), and Cnga3−/−/Nrl−/− transfection method, followed by subsequent puri-
mouse was generated by cross-breeding. The fication and titration, as described previously [8].
double knockout mouse was used as a channel-­
deficient model on a cone-dominant background
to facilitate biochemical evaluations in cones. All 2.3 Subretinal Injection
experiments and animal maintenance were
approved by the local Institutional Animal Care Subretinal injections were performed under a
and Use Committee (Oklahoma City, OK) and Carl Zeiss OPMI VISU 140 surgical operating
Inhibition of Ryanodine Receptor 1 Reduces Endoplasmic Reticulum (ER) Stress and Promotes ER Protein… 355

Table 1 Primary antibody information


Catalog Dilutions used in immunoblotting (IB) or
Antibody Provider number immunofluorescence labeling (IF)
Ryr1 Cell signaling 8153 1:500 (IB)
M-opsin EMD Millipore AB5405 1:500 (IB)
S-opsin Dr. Muna Naash (University 1:500 (IB)
of Houston)
Cone arrestin EMD Millipore AB15282 1:500 (IB), 1:200 (IF)
Phospho-­ Cell signaling 3398 1:500 (IB)
eIF2α
Phospho-­ Abcam Ab-48,187 1:500 (IB)
IRE1α
Syvn1 Proteintech 13,473–1-AP 1:500 (IB)
Derlin-1 Abcam ab176732 1:500 (IB)
β-actin Abcam ab-6276 1:2000 (IB)

microscope. Briefly, after anesthesia on ice for Li-Cor Odyssey Fc Imager and Image Studio Fc
2–3 min and complete dilation with 1% cyclo- software (Li-Cor Biosciences) were used for
pentolate hydrochloride (Akorn), a drop of 2.5% detection and densitometric analysis.
methylcellulose was added to the corneal surface Mouse eye collection, retinal cross-sectional
to visualize the fundus. A 28-gauge beveled preparation, and immunofluorescence labeling
hypodermic needle (BD Biosciences) was used were performed as described previously [4].
to puncture a hole immediately below the limbus. Table 1 shows the dilution information of cone
One microliter of AAV2/5-SaCas9/gRNA-Ryr1 arrestin antibody used in the immunofluores-
vector (4.4 × 109 vg) was injected subretinally cence labeling. The labeling was imaged using an
into one eye of each mouse using a NanoFil Olympus FV1000 confocal laser-scanning micro-
microsyringe injector system with a 33-gauge scope and FluoView imaging software
blunt needle (Hamilton Co.). The contralateral (Olympus).
eye was injected with 1 μl of vehicle (balanced
salt solution, Alcon).
3 Results

2.4 Western Blotting 3.1 Treatment with AAV2/5-­


and Immunofluorescence SaCas9/gRNA-Ryr1 Reduced
Labeling Expression Level of Ryr1
in Cnga3−/−/Nrl−/− Retinas
Retinal protein preparation, SDS-PAGE, and and Increased Expression
Western blotting were performed as described Levels of Cone-Specific
previously [1]. Following gel separation, trans- Proteins
ferring, and blocking, blots were incubated with
primary antibody overnight at 4 °C (see Table 1 Previous studies have shown that the expression
for antibody dilution information) and with level of Ryr1 mRNA was increased in CNG
HRP-­ conjugated secondary antibodies channel-deficient retina [1]. In this work, we
(1:20,000) for 1 h at room temperature. first examined the expression level of Ryr1 pro-
SuperSignal West Dura Extended Duration che- tein in the diseased retina and the effects of Ryr1
miluminescent substrate (Thermo Fisher knockdown. Postnatal day 5 (P5),
Scientific) was used to detect binding of the pri- Cnga3 /Nrl mice received AAV2/5-SaCas9/
−/− −/−

mary antibodies to their cognate antigens. A gRNA-Ryr1 via subretinal injection, and retinas
356 F. Yang et al.

Fig. 1 Knockdown of Ryr1 increased expression levels cone-specific protein detections and corresponding quan-
of cone proteins in Cnga3−/−/Nrl−/−retinas. The expression titative analysis (b). (c) Shown are representative confocal
levels of Ryr1, M-opsin, S-opsin, and cone arrestin (CAR) images of CAR labeling on retinal cross sections. ONL
in Cnga3−/−/Nrl−/− mice treated with AAV2/5-SaCas9/ outer nuclear layer, INL inner nuclear layer. Data are pre-
gRNA-Ryr1 or vehicle were analyzed by western blot sented as mean ± SEM of three independent assays using
analysis and immunofluorescence labeling. (a, b) Shown retinas from 6 to 8 mice (*p < 0.05)
are representative western blot images of Ryr1 (a) and

were collected at P40 for analysis of protein and cone arrestin (CAR) were significantly
expression by western blotting. The evaluation increased in the retinas of Cnga3−/−/Nrl−/− mice
showed that the expression level of Ryr1 in that have been treated with AAV2/5-SaCas9/
Cnga3−/−/Nrl−/− retinas was increased, com- gRNA-Ryr1, compared with mice treated with
pared with that in the Nrl−/− controls, and treat- vehicle (Fig. 1b). The expression level of CAR
ment with the viral vectors nearly completely was increased by about 50% in Cnga3−/−/Nrl−/−
abolished this elevation (Fig. 1a). mice treated with AAV2/5-SaCas9/gRNA-Ryr1,
We next examined the effects of AAV2/5-­ compared with that in the age-matched vehicle-­
SaCas9/gRNA-Ryr1 on the expression of the treated controls (Fig. 1b). Increased expression
cone-specific proteins. The evaluation showed level of CAR was also shown by immunofluores-
that the expression levels of M-opsin, S-opsin, cence labeling (Fig. 1c).
Inhibition of Ryanodine Receptor 1 Reduces Endoplasmic Reticulum (ER) Stress and Promotes ER Protein… 357

3.2 Treatment with AAV2/5-­ However, treatment with AAV2/5-SaCas9/


SaCas9/gRNA-Ryr1 Reduced gRNA-Ryr1 significantly increased expression
ER Stress Responses level of these proteins (Fig. 2b).
in Cnga3−/−/Nrl−/− Retinas
and Increased Expression
Levels of ERAD Proteins 4 Discussion

We then examined the effects of Ryr1 knock- Cones in CNG channel-deficient mice display
down on the ER stress responses. The evaluation characteristics of ER stress-associated apoptosis.
showed that knockdown of Ryr1 significantly Retinas of these mice show elevation of all three
reduced ER stress in CNG channel-deficient reti- arms of the ER stress pathways, i.e., elevated lev-
nas. The elevated levels of phospho-IRE1α and els of phospho-eIF2α and phospho-IRE1α and
phospho-eIF2α were reversed in retinas of increased cleavage of ATF6 [1, 3, 6], as well as
Cnga3−/−/Nrl−/− mice treated with increased nuclear localization of CCAAT/−
­AAV2/5-­SaCas9/gRNA-Ryr1, compared with enhancer-binding protein homologous protein [3,
vehicle-­treated controls (Fig. 2a). 6]. CNG channel-deficient retinas also show
ER retrotranslocation and ER-associated pro- impaired cytosolic and ER calcium homeostasis
tein degradation (ERAD) involve multiple [1, 3, 4, 6]. This work shows that knockdown of
machinery proteins, including Syvn1 (E3 Ryr1 reduces ER stress, promotes ERAD, and
ubiquitin-­protein ligase synoviolin 1) and improves cone survival in CNG channel
Derlin-1 (degradation in ER protein 1), and these deficiency.
proteins have been shown to be upregulated by The cone preservation after knockdown of
deletion of Ryr2 [7]. In this work, we examine the Ryr1 is partial. This observation is similar to that
effects of Ryr1 knockdown. The expression lev- in mice with deletion of IP3R1 or Ryr2 [1, 4, 7],
els of Syvn1 and Derlin-1 were not different suggesting an incomplete correction of ER cal-
between Cnga3−/−/Nrl−/− and Nrl−/− retinas. cium deficiency, likely resulting from compensa-

Fig. 2 Knockdown of Ryr1 reduced ER stress in gRNA-Ryr1 or vehicle were analyzed by western blot
Cnga3−/−/Nrl−/− retinas and increased expression levels of analysis. Shown are representative western blot images of
ERAD proteins. The expression levels of ER stress marker these detections and corresponding quantitative analysis.
proteins phospho-IRE1α and phospho-eIF2α (a) and the Data are presented as mean ± SEM of three independent
expression levels of ERAD proteins Syvn1 and Derlin-1 assays using retinas from 6 to 8 mice (*p < 0.05; **p <
(b) in Cnga3−/−/Nrl−/− mice treated with AAV2/5-SaCas9/ 0.01; n.s., not significant)
358 F. Yang et al.

tory mechanisms. These could include References


compensatory regulation from other isoforms of
the ER calcium channels, from other ER 1. Butler MR, Ma H, Yang F, Belcher J, Le YZ,
Mikoshiba K, Biel M, Michalakis S, Iuso A, Krizaj
calcium/protein folding regulators/chaperones, D, Ding XQ. Endoplasmic reticulum (ER) Ca(2+)-
and from other calcium channels in the inner seg- channel activity contributes to ER stress and cone
ment membranes. Partial protection of cones death in cyclic nucleotide-gated channel deficiency.
might also reflect incomplete transduction of J. Biol. Chem. 2017;292:11189–205.
2. Kohl S, Marx T, Giddings I, Jagle H, Jacobson SG,
photoreceptors following subretinal injection Apfelstedt-Sylla E, Zrenner E, Sharpe LT, Wissinger
with the viral vectors. B. Total colourblindness is caused by mutations in
Similar to deletion of Ryr2, knockdown of the gene encoding the alpha-subunit of the cone pho-
Ryr1 is sufficient to reduce ER stress/cone death toreceptor cGMP-gated cation channel. Nat. Genet.
1998;19:257–9.
and improve ERAD. These findings suggest that 3. Ma H, Butler MR, Thapa A, Belcher J, Yang F,
although the potential compensatory mechanisms Baehr W, Biel M, Michalakis S, Ding XQ. cGMP/
among the different channel isoforms cannot be protein kinase G signaling suppresses inositol
excluded when one isoform is deleted/knocked 1,4,5-­trisphosphate receptor phosphorylation and pro-
motes endoplasmic reticulum stress in photoreceptors
down, these two channel isoforms are indispens- of cyclic nucleotide-Gated Channel-deficient mice.
able and both isoforms contribute to ER calcium J. Biol. Chem. 2015;290:20880–92.
dysregulation in CNG channel deficiency. 4. Ma H, Yang F, Butler MR, Rapp J, Le YZ, Ding
In summary, this work demonstrates that Ryr1 XQ. Ryanodine receptor 2 contributes to impaired
protein localization in cyclic nucleotide-gated
contributes to ER stress/cone degeneration in CNG channel deficiency. eNeuro. 2019;6(3):1–12.
channel deficiency. Together with previous findings ENEURO.0119-19.2019
showing that deletion of IP3R1 or Ryr2 suppresses 5. Nishiguchi KM, Sandberg MA, Gorji N, Berson EL,
ER stress and preserves cones, our findings estab- Dryja TP. Cone cGMP-gated channel mutations and
clinical findings in patients with achromatopsia, mac-
lish a role of ER calcium dysregulation in the ER ular degeneration, and other hereditary cone diseases.
stress/cone degeneration in CNG channel defi- Hum. Mutat. 2005;25:248–58.
ciency and support strategies targeting ER calcium 6. Thapa A, Morris L, Xu J, Ma H, Michalakis S,
regulation to slow down cone degeneration. Biel M, Ding XQ. Endoplasmic reticulum stress-­
associated cone photoreceptor degeneration in cyclic
nucleotide-gated channel deficiency. J. Biol. Chem.
Acknowledgments We thank Drs. Martin Biel and 2012;287:18018–29.
Anand Swaroop for providing Cnga3-/- and Nrl-/- mouse 7. Yang F, Ma H, Butler MR, Ding XQ. Preservation of
lines, and Dr. Feng Zhang for the SaCas9 plasmid. We endoplasmic reticulum (ER) Ca(2+) stores by deletion
thank the Imaging Core Facility and the Histology Core of inositol-1,4,5-trisphosphate receptor type 1 pro-
Facility of the Department of Cell Biology at OUHSC for motes ER retrotranslocation, proteostasis, and protein
technical assistance. This work was supported by grants outer segment localization in cyclic nucleotide-gated
from the National Eye Institute (R01EY027754 and channel-deficient cone photoreceptors. FASEB J.
P30EY021725), the Oklahoma Center for the 2021;35:e21579.
Advancement of Science and Technology, the Presbyterian 8. Young BM, Jones K, Massengill MT, Walsh E, Li
Health Foundation, and support to Dr. Lewin from the H, Lewin AS, Ildefonso CJ. Expression of a CARD
Shaler Richardson Professorship endowment and an unre- slows the retinal degeneration of a geographic atro-
stricted grant to the University of Florida from Research phy mouse model. Mol Ther Methods Clin Dev.
to Prevent Blindness. 2019;14:113–25.
Mouse Choroid Proteome
Revisited: Focus on Aging

Donita Garland, James Harnly,


and Radha Ayyagari

Abstract 1 Introduction

Age is a major risk factor for age-related mac- All of the major risk factors for age-related mac-
ular degeneration (AMD), and age has a role ular dystrophy (AMD) will, to varying degrees,
in the disease phenotypes of heritable macular impact biological aging, i.e., the combination of
dystrophies. The proteomes of C57Bl6/J molecular changes leading to decreased biologi-
mouse choroids at 2 ages were analyzed to cal function [6, 15, 19, 25]. The interplay between
identify biochemical processes affected by these molecular changes is complex and unique
aging. Proteins of interest were identified as for individuals. In addition to the impact of
those contributing most to the variance in genetic variants on aging, it is important to under-
principal component analysis and those show- stand how biological aging influences the impact
ing the largest significant differences between of genetic variants or mutations on macular
ages. These proteins implicated altered ECM disease.
composition, immune system function, and The age-related changes in the functions of
lipid metabolism. the retinal pigment epithelia (RPE) and Bruch’s
membrane (BrM) are key to the development of
Keywords macular dystrophies whether age-related or due
to mutation. Mice have been widely used to
Choroid · Mouse · Aging · Proteome · Bruch’s model the pathology of AMD and heritable forms
membrane · Extracellular matrix · of macular dystrophies [7, 20, 21, 27, 28, 33]. We
Complement · Lipid · Immune system chose to assess the effects of chronological age
on the mouse choroid. The choroid consists of
vessels, the choriocapillaris, and BrM which
D. Garland (*) forms the inner edge of the choroid. The choroid
Harnly LLC, Bethesda, MD, USA
contains a number of cell types, among them
J. Harnly immune cells, melanocytes, and endothelia cells.
Human Nutrition Center, US Department of
Agriculture, Beltsville, MD, USA Each component of the choroid undergoes
e-mail: james.harnly@usda.gov changes with age that are thought to contribute to
R. Ayyagari the development of macular dystrophies. These
Departments of Ophthalmology and Pathology, include the thinning of the choroid, thickening of
Shiley Eye Institute, University of California, San BrM, decrease of the choriocapillaris, and the
Diego, CA, USA activation of immune cells [1, 14, 17, 26]. In
e-mail: rayyagari@health.ucsd.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 359
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_52
360 D. Garland et al.

addition, the choroid is a source of certain com- Proteomics and Metabolomics Facility, Wistar
plement components and regulatory proteins of Institute, Philadelphia, PA. MaxQuant 1.6.0.16
the classical and alternative pathways [3]. was used for database searching, peptide and pro-
Our initial proteome analysis of the mouse cho- tein identification, and label-free quantification
roid with age showed altered biological functions (LFQ) [12, 13].
included immune function, signaling, ECM com- Principal component analysis was done using
position, and intracellular transport processes [21]. SOLO, Eigen Vector.
Recent technical advances and improved software
and databases prompted us to revisit quantitative
proteome analysis of the mouse choroid. This 3 Results
analysis yielded identification of 3700 proteins,
over 4.5 times the number of proteins in the origi- Across all samples 39,000 peptides were identi-
nal study. This allowed identification of lower fied. These were assigned to 3700 protein groups.
abundance proteins and provided a more in-depth The first step was to determine if there were any
picture of the age-­related changes. patterns in the data. This was done using princi-
The aim of this study was to identify the molec- pal component analysis (PCA), a multivariate
ular processes most affected by age in the choroid. analysis method that reduces the dimensionality
This provides a baseline for studying the pathobiol- of large datasets to increase interpretability. As
ogy in mouse models of heritable macular dystro- seen in Fig. 1, the data formed two clusters with
phies and for resolving mutant-­induced pathology respect to age, separated along the x-axis, which
from age-related changes. We outline here our accounted for 68% of the variance in the data.
approach and an overview of the results. The separation along the y-axis accounted for
only 10% of the variance. This separation was
due to biological variation among samples. At
2 Methods 4.4 months the variation among samples is likely
due to gender. For two samples three of the four
The C57Bl6 mice were generated at UC San choroids were from female, and for two samples
Diego. The use of the mice was approved by the three of the four choroids were from male mice.
UCSD Animal Use and Care Committees. The The variation was slightly greater in 19.2-mo-old
cornea, lens, and neural retina were removed and samples and was likely due to the slightly larger
the eyes were held at −80° until used. Room tem- spread in mouse age. The remaining 22% of the
perature PBS/EDTA was added to the eyecups to variance was due to technical variation.
slowly thaw and loosen the RPE [21]. Radial cuts Two approaches were primary in identifying
were made to flatten the tissue. Any remaining proteins of potential significance in aging of the
RPE was washed from the surface of Bruch’s mouse choroid. (1) The PCA “loadings” identify
membrane with repeated streams of PBS/ the contribution of each protein according to their
EDTA. The edge of Bruch’s membrane was held relative contribution to the separation of the clus-
with tweezers and Bruch’s membrane/choroid was ters in PCA. In this case, the loadings associated
peeled out. To address mouse biological variation, with PC1/age were of interest. (2) The student t
samples were composites of choroids, one test was used to identify all proteins that were
eye/mouse/sample. Four biological replicates significantly altered between the two ages. About
were done for each age. The mice were 4.4 mo ± one third (1265) of the proteins were significantly
0.25 mo (16 mice, 8F/8M) and 19.2 mo ± 0.6 mo altered (p < 0.05). Slightly more than half
(13 mice, 5F/8M). These ages correspond to 20–30 ofvthese were decreased; the rest were increased.
and 56–69 human years of age, respectively [18]. Seventy-eight proteins were increased threefold
The proteins in samples were solubilized and or more, and 124 were decreased threefold or
run 2 cm into SDS gels. In-gel trypsin digestion greater. Biological significance was further eval-
of proteins and LC-MS/MS was performed at the uated by comparison of the top ranked loadings
Mouse Choroid Proteome Revisited: Focus on Aging 361

0.1

Scores on PC2 (10.44%) 0.05

–0.05

–0.1

–0.15
–0.25 –0.2 –0.15 –0.1 –0.05 0 0.05 0.1 0.15 0.2 0.25
Scores on PC1 (67.60%)

Fig. 1 Scores plot for PCA analysis. Separation on X-axis (PC1) correlates with age. Separation on Y-axis correlates
with biological variation within samples

with those proteins identified by student t test as lipid-associated proteins were significantly
being significantly altered. The biological func- altered with age. In this case, however, two thirds
tions of those proteins identified as being altered of those were increased.
with age were assessed by using multiple data- Of particular interest was the effect of age on
bases for protein class, gene ontology, pathway the composition of Bruch’s membrane (BrM).
analysis, and molecular interactions. Forty-five known components of BrM were iden-
The presence of endothelial cells, mast cells, tified and most were altered with age. Mutant
macrophages, and melanocytes was verified by forms of the BrM proteins, CTRP5, EFEMP1,
the identification and quantification of specific and TIMP3, lead to heritable forms of macular
markers. For each cell type, the altered levels of dystrophies [2, 4, 9, 31]. Altered levels of another
specific markers suggested critical biological BrM component, HTRA1, confer risk for AMD
functions were impacted by age. [24, 34]. As seen in Table 1, CTRP5 and HTRA1
Components of three classes of proteins were were both increased with age. EFEMP1 was
highly represented in the dataset: extracellular decreased and TIMP3 was not altered.
matrix (ECM), immune system, and lipid metab- Thirteen complement components and nine
olism. Three hundred proteins were identified as complement system regulators were identified
ECM proteins by comparison with the Matrisome in the choroid. The changes observed with
database; 250 proteins were identified as immune increased age for 6 of these are listed in Table 1.
system proteins by comparison with the C4b and Serping1 which increased 2.7 and 1.9
InnateDB; and 319 proteins were associated with fold, respectively (p = 0.0001 and 0.002), are
lipids (LipidMaps) [5, 11, 29]. Nearly 50% of the components of the classical complement path-
proteins of the innate immune system were way. Cfh and Cfhr2, regulators of the alterna-
altered with age (p < 0.05) of which 55% were tive complement pathway, were also increased
increased. About 25% of the ECM proteins were but to a lesser extent. Factors B, P, D, and I of
significantly altered with age ranging from a five- the alternative complement pathway were not
fold increase to a fivefold decrease. Only 32% of detected.
362 D. Garland et al.

Table 1 Ratios of listed protein levels (LFQ intensities) The absence of complement factors B, D, P,
between 19.2 mo and 4.4 mo mice
and I of the alternative complement pathway at
19.2mo/4.4mo p value both ages was unexpected. Gene expression of
CFH 1.4 0.001
each was observed in human choroid, and CFP is
CFHR2 1.8 0.0003
reported to be expressed by proinflammatory cells,
SERPING1 1.9 0.002
C1S 0.9 0.727 cells which were present in the mouse choroid [3,
C3 0.9 0.474 10]. The main sources of ocular CFB are, however,
C4B 2.7 0.0001 RPE and plasma [35]. Other approaches are being
CTRP5 1.4 0.03 used to verify the presence or absence of these
HTRA1 2.7 0.001 alternative complement pathway components.
EFEMP1 0.5 0.0002 The limitations to this approach are primarily
TIMP3 1.0 0.656
technical. Very low abundance proteins may be
below the detection limit, and some proteins are
4 Discussion not amenable to detection by mass spectrome-
try. The amino acid sequences may yield few
Significant changes with age were observed in the tryptic peptides, or the tryptic peptides are too
ECM, immune system, and lipid-associated pro- short, are too long, or have low ionization effi-
teins. Components of each confer risk of develop- ciency. Many ECM proteins are long-lived and
ing macular dystrophies and each is involved in become heavily posttranslationally modified
their pathobiology, and they are functionally with age. Elastin is such an example; it becomes
interdependent. Engin et al. [16] stated “Altered cross-linked yielding few detectable peptides.
expression of ECM affects all biological process Only one elastin peptide was detected in all of
including inflammation” [30, 32]. In addition to the samples analyzed. Proteins which were not
its role in immune homeostasis, ECM is a reser- detected but predicted to be in potentially inter-
voir for signaling molecules and thus plays a criti- esting pathways have to be validated by other
cal role in signaling. Since the composition of techniques. A major advantage of the proteome
ECM dictates its structure and function, changes approach is, however, the generation of testable
of up to fivefold in the levels of some ECM com- hypotheses.
ponents will have a significant impact on the
structure and function of the ECM [22, 23]. It is
interesting that CTRP5 and HTRA1 were both References
increased with age. We recently demonstrated
these were both increased in mice expressing 1. Ambati J, Atkinson JP, Gelfand BD. Immunology of
age-related macular degeneration. Nat. Rev. Immunol.
mutant CTRP5 and are binding partners [8]. 2013;13:438–51.
There is a large literature regarding the role of 2. Anand-Apte B, Chao JR, Singh R, Stöhr H. Sorsby
complement in macular dystrophies, and comple- fundus dystrophy: insights from the past and looking
ment is critical for early stage pathology in to the future. J. Neurosci. Res. 2019;97:88–97.
3. Anderson DH, Radeke MJ, Gallo NB, Chapin EA,
EFEMP1 mutant mice [21]. Choroidal immune Johnson PT, Curletti CR, et al. The pivotal role of the
cells are a source of a number of complement complement system in aging and age-related macular
components and regulators, whereas the RPE and degeneration: hypothesis re-visited. Prog. Retin. Eye
liver are primary sources of others. The signifi- Res. 2010;29:95–112.
4. Ayyagari R, Mandal MNA, Karoukis AJ, Chen L,
cance of the age-related increases in the regula- McLaren NC, Lichter M, Sieving PA. Late-onset mac-
tors (CFH, CFHR2, SERPING1) and C4B is not ular degeneration and long anterior lens zonules result
clear (Table 1). Does it reflect increased comple- from a CTRP5 gene mutation. Invest. Ophthalmol.
ment activity or increased numbers of immune Vis. Sci. 2005;46:3363–71.
5. Breuer K, Foroushani AK, Laird MR, Chen C,
cells expressing complement? In earlier studies Sribnaia A, Lo R. InnateDB: systems biology of innate
on human AMD choroid, increased transcription immunity and beyond – recent updates and continuing
of complement components was not observed [3]. curation. Nucleic Acids Res. 2013;41:D1228–33.
Mouse Choroid Proteome Revisited: Focus on Aging 363

6. Chakravarthy U, Wong TY, Fletcher A, Piault E, 20. Fu L, Garland D, Yang Z, Shukla D, Rajendran A,
Evans C, Zlateva G. Clinical risk factors for age-­ Pearson E, et al. The R345W mutation in EFEMP1
related macular degeneration: a systematic review and is pathogenic and causes AMD-like deposits in mice.
meta-analysis. BMC Ophthalmol. 2010;10:31. https:// Hum. Mol. Genet. 2007;16:2411–22.
doi.org/10.1186/1471-­2415-­10-­31. 21. Garland DL, Fernandez-Godino R, Kaur I, Speicher
7. Chavali VR, Khan NW, Cukras CA, Bartsch KD, Harnly JM, Lambris JD, et al. Mouse genet-
DU, Jablonski MM, Ayyagari R. A CTRP5 gene ics and proteomic analyses demonstrate a critical
S163R mutation knock-in mouse model for late- role for complement in a model of DHRD/ML, an
onset retinal degeneration. Hum. Mol. Genet. inherited macular degeneration. Hum. Mol. Genet.
2011;20:2000–14. 2014;23:52–68.
8. Chekuri A, Zientara-Rytter K, Soto-Hermida A, 22. Hynes RO. Extracellular matrix: not just pretty fibrils.
Borooah S, Voronchikhina M, Biswas P, et al. Late-­ Science. 2009;326:1216–9.
onset retinal degeneration pathology due to mutations 23. Karamanos NK, Theocharis AD, Piperigkou Z,
in CTRP5 is mediated through HTRA1. Aging Cell. Manou D, Passi A, Skandalis SS, et al. A guide to the
2019;18:e13011. composition and functions of the extracellular matrix.
9. Christensen D, Brown F, Cree A, Ratnayaka AJ, Lotery FEBS J. 2021;288:6850. https://doi.org/10.1111/
A. Sorsby fundus dystrophy – a review of pathology febs.15776.
and disease mechanisms. Exp. Eye Res. 2017;165:35. 24. Liu Y, Jin H, Wei D, Li W. HTRA1 rs11200638
https://doi.org/10.1016/j.exer.2017.08.014. variant and AMD risk from a comprehensive anal-
10. Cortes C, Ohtola JA, Saggu G, Ferreira VP. Local ysis about 15,316 subjects. BMC Med. Genet.
release of properdin in the cellular microenviron- 2020;21:107.
ment: role in pattern recognition and amplifica- 25. López-Otín C, Blasco MA, Partridge L, Serrano
tion of the alternative pathway of complement M, Kroemer G. The hallmarks of aging. Cell.
front. Immunology. 2013;3 https://doi.org/10.3389/ 2013;153(6):1194–217.
fimmu.2012.00412. 26. McLeod DS, Grebe R, Bhutto I, Merges C, Baba T,
11. Cotter D, Maer A, Guda C, Saunders B, Subramaniam Lutty GA. Relationship between RPE and choriocap-
S. LMPD: LIPID MAPS proteome database. illaris in age-related macular degeneration. Invest.
Nucleic Acids Res. 2006;34:D507–10. https://doi. Ophthalmol. Vis. Sci. 2009;50:4982–91.
org/10.1093/nar/gkj122. 27. Pandi SPS, Arjuna Ratnayaka JA, Lotery AJ,
12. Cox J, Mann M. MaxQuant enables high peptide Teeling JL. Progress in developing rodent mod-
identification rates, individualized p.p.b.-range mass els of age-related macular degeneration (AMD).
accuracies and proteome-wide protein quantification. 2020;203:108404. https://doi.org/10.1016/j.
Nat. Biotechnol. 2008;26:1367–72. exer.2020.108404.
13. Cox J, Hein MY, Luber CA, Paron I, Nagaraj N, Mann 28. Pennesi ME, Neuringer M, Courtney RJ. Animal
M. Accurate proteome-wide label-free quantification models of age related macular degeneration. Mol.
by delayed normalization and maximal peptide ratio Aspects Med. 2012;33:487–509.
extraction. Termed MaxLFQ Mol Cell Proteomics. 29. Shao X, Taha IN, Clauser KR, Gao U, Naba
2014;13:2513–26. A. MatrisomeDB: the ECM-protein knowledge data-
14. Curcio CA, Johnson M. Structure, function, and base. Nucleic Acids Res. 2020;48:D1136–44.
pathology of Bruch’s membrane. Anat Physiol. 30. Sorokin L. The impact of the extracellular
2013;20:465–81. matrix on inflammation. Nat. Rev. Immunol.
15. Ebrahimi KB, Handa JT. Lipids, lipoproteins, 2010;10:712–23.
and age-related macular degeneration. J Lipids. 31. Stone EM, Lotery AJ, Munier FL, Héon E, Piguet
2011;2011:802059. B, Guymer RH, et al. A single EFEMP1 muta-
16. Engin AB, Nikitovic D, Neagu M, Henrich-Noack tion associated with both Malattia Leventinese and
P, Docea AO, Shtilman MI, Golokhvast K, Tsatsakis Doyne honeycomb retinal dystrophy. Nat. Genet.
AM. Mechanistic understanding of nanoparticles’ 1999;22:199–202.
interactions with extracellular matrix: the cell and 32. Theocharis AD, Manou D, Karamanos NK. The
immune system. Part. Fibre Toxicol. 2017;14:22. extracellular matrix as a multitasking player in dis-
17. Farazdaghi MK, Ebrahimi KB. Role of the choroid ease. FEBS J. 2019;286:2830–69.
in age-related macular degeneration: a current review. 33. Weber BHF, Lin B, White K, Kohler K, Soboleva
J. Ophthalmic Vis. Res. 2019;14:78–87. G, Herterich S, et al. A mouse model for Sorsby
18. Flurkey K, Currer JM, Harrison DE. The mouse in fundus dystrophy. Invest. Ophthalmol. Vis. Sci.
aging research. In: Fox JG, et al., editors. The mouse 2002;43:2732–40.
in biomedical research. 2nd ed. Burlington: American 34. Weber BHF, Lin B, White K, Kohler K, Soboleva
College Laboratory Animal Medicine (Elsevier); G, Herterich S, et al. A mouse model for Sorsby
2007. p. 637–72. fundus dystrophy. Invest. Ophthalmol. Vis. Sci.
19. Fritsche LG, Fariss RN, Stambolian D, Abecasis 2002;43:2732–40.
GR, Curcio CA, Swaroop A. Age-related macu- 35. 35. Chen M, Muckersie E, Robertson M, Forrester
lar degeneration: genetics and biology coming JV, Xu H. Up-regulation of complement factor B in
together. Ann Rev Genomics and Human Genetics. retinal pigment epithelial cells is accompanied by
2014;15:151–71. complement activation in the aged retina. Exp. Eye
Res. 2008;543–550.
Morphological and Functional
Comparison of Mice Models
for Retinitis Pigmentosa

Prakadeeswari Gopalakrishnan, Avigail Beryozkin,


Eyal Banin, and Dror Sharon

Abstract Keywords

Retinitis pigmentosa (RP) is the predominant Retinitis pigmentosa · Fam161a · Mouse


form of inherited retinal degenerations (IRDs) model · Knockout · Retina
caused by abnormalities and loss of photore-
ceptor cells ensuing diminishment of vision.
RP is a heterogenous genetic disorder associ-
ated with mutations in over 80 genes, showing Abbreviations
various inheritance patterns. Laboratory
mouse models are important for our under- IRDs Inherited retinal degenerations
standing of disease mechanisms, modifier PR Photoreceptor
effects, and development of therapeutic RP Retinitis pigmentosa
modalities. In this review, we have summa-
rized a comprehensive comparison of our pre-
viously reported Fam161a knockout (KO) 1 Introduction
mouse model with other well-studied RP
mouse models, Fam161aGT/GT, Pde6brd1, Retinitis pigmentosa (RP; MIM 268000) is the
Nr2e3rd7, Rpgrrd9, and Pde6brd10 using struc- most prevalent inherited retinal degeneration
tural and functional analysis of the retina. (IRD) with a prevalence of 1:2000–1:5000
Fam161atm1b/tm1b mouse models are important individuals and is one of the major causes of
for developing novel therapies and mainly progressive loss of vision in young individuals
AAV-based gene therapy and translational [1, 2]. RP is a heterogeneous disease and can be
read-through-inducing drugs. inherited in various patterns (autosomal
dominant, autosomal recessive, X-linked, and
mitochondrial) [3]. RP is ensued due to the
abnormalities and loss of photoreceptor cells
P. Gopalakrishnan · A. Beryozkin · E. Banin · which are associated with mutations in over 80
D. Sharon (*) genes (https://sph.uth.edu/retnet/, retrieved on
Department of Ophthalmology, Hadassah Medical
Center, Faculty of Medicine, The Hebrew University 27th November 2021). The primary clinical
of Jerusalem, Jerusalem, Israel features of RP are apoptosis of rod photoreceptor
e-mail: prakadee.gopalakrish@mail.huji.ac.il; avigail. cells and subsequent cone photoreceptor cell
beryozkin@mail.huji.ac.il; banine@mail.huji.ac.il; degeneration, resulting in partial or even complete
dror.sharon1@mail.huji.ac.il

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 365
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_53
366 P. Gopalakrishnan et al.

vision impairment. Not only restricted to the eye, (iii) Protein expression by evaluating either
RP can also transpire along with syndromes absence of the appropriate full-length func-
including Usher syndrome and Bardet-Biedl tional protein or presence of truncated non-
syndrome [4]. functional protein by Western blot or
A decade ago, two back-to-back publications immunohistochemistry (IHC) analysis.
[5, 6] first reported that null mutations in the
FAM161A (OMIM 613596) gene cause RP, fol-
lowed by other reports from various populations. 2 Morphological Alterations
Mutations in FAM161A are now known to be the of Retinal Structure
major cause of RP in the Israeli population due to
two founder mutations [7] and additional though Since photoreceptor degeneration is one of the
less common mutations have been identified in hallmarks of RP, one can measure ONL thickness
RP patients from Europe, the USA, India, and in histological analysis and OCT scans as a mea-
China [8–12]. Moreover, we have recently sure of disease progression. The different models
reported on a comprehensive clinical analysis of that are analyzed here show variable progression
over 100 patients with biallelic FAM161A muta- rates and onset. The Fam161aGT/GT model showed
tions [7], a dataset that can serve as a basis for disorganized outer segment of photoreceptors
future gene therapy treatment. It was therefore already at 1 month, and a 50% reduction in reti-
important to understand the underlying patho- nal thickness at 2 months of age (Table 1). At the
genic mechanism of FAM161A mutations associ- age of 6 months, 90% of the ONL entirely disap-
ated with RP. Experimental animal models were peared although the inner retina remained to be
extremely useful so far in understanding disease mostly unaffected [15]. A KO model of the same
mechanism of various RP types, including knock- gene, Fam161atm1b/tm1b, showed a similar rate of
out (KO) and knock-in (KI) models in various degeneration with gradual thinning of ONL from
species [13], and this would be obliging for 23% loss at 1 month to 94% loss at 8 months.
designing therapeutic regimens. This prompted OCT did not reveal any detectable ONL at the
us to generate and characterize a new Fam161a age of 6 months [14]. On the other hand, Pde6brd1
knockout (KO) mouse model, Fam161atm1b/tm1b mice showed a more severe form of retinal degen-
which is lacking the major exon 3 [14]. In this eration with 73% substantial reduction of outer
mini review, we intend to assess and summarize retinal thickness at P13 [27]. Interestingly, the
the common and variable characteristics of vari- inner nuclear layer (INL) seems to be well-­
ous mouse models of RP, including the preserved until P34 [27, 28]. Another long-term
Fam161atm1b/tm1b [14], Fam161aGT/GT [15], characterization study [29] on this model at P14
Pde6brd1 [16–19], Nr2e3rd7 [20, 21], Rpgrrd9 [22, revealed thinned retina as well as loss of inner
23], and Pde6brd10 [24–26]. To this end, we per- segment and outer segment (IS/OS) junction and
formed a comparative analysis of these models external limiting membrane (ELM). The rd10
based on the following parameters: (Pde6brd10) model was reported to have a progres-
sive ONL degeneration starting in the central
(i) Progression of retinal degeneration with retina at P16, spreading to the peripheral retina
subsequent retinal structural differences that later at P20, while no ONL was left at P60 [25].
appear in the outer nuclear layer (ONL) Compared to rd1, the rd10 model exhibited a pro-
along with photoreceptor cell loss as deter- gressive but relatively slow decline of total retinal
mined by optical coherence tomography thickness and only at P103 it reached similar reti-
(OCT) scans and histological analysis nal thickness as rd1 [29]. The retina of the rd7
(ii) Retinal function as measured by model, Nr2e3rd7, illustrated ONL waves, whorls,
electroretinography (ERG) and visual acuity and rosettes at 1 month of age; then, flattened
(VA) waves were observed in 5 months, which disap-
peared at 16-month-old retinas. The ONL thick-
Morphological and Functional Comparison of Mice Models for Retinitis Pigmentosa 367

Table 1 Summary of the comparative analyses of well-studied RP mouse models


Rate of Time
Gene Mutation Strain degeneration point
Pde6brd1 Exon 7; p.Tyr347X: B.C3-Pde6brd1; ABJ/Le; BDP/J; 73% retinal P13
Nonsense point mutation & BUB/BnJ; C3H sublines: CBA/J, thickness
an intronic insertion of a FVB/NJ, PL/J, SB, SJL/J, and reduction
leukemia virus (Xmv-28) SWR/J
Pde6brd10 Exon 13; p.Arg560Cys: B6.CXB1-Pde6brd10/J Diminished P16
Missense mutation ONL P60
No ONL
Nr2e3rd7 Deletion of last 36 B6.Cg-Nr2e3rd7/J 4–5 rows of 16 months
nucleotides of exon 4, intron nuclei
4, & single nucleotide
changes in exon 5
Rpgrrd9 32 bp duplication in exon C57BL/6 J-RpgrRd9/Boc 23% ONL 12 months
ORF15 reduction 18 months
34% ONL
reduction
Fam161aGT/GT Gene trap within exon 3 after 129X1/SvJ x 129S1/Sv F1-Kitl+ 50% ONL 2 months
amino acid 362 reduction 6 months
90% ONL
reduction
Fam161atm1b/tm1b Deletion of exon 3 C57BL/6J-Atm1Brd 63% ONL 3 months
reduction 6 months
79% ONL
reduction

ness is relatively preserved with only 19% loss at amplitude decrease compared to dark-adapted
the age of 1 year and 46% at 18 months [20]. ERGs [15]. Similarly, the other Fam161a model,
Rpgrrd9, a mutant model of X-linked RP, has Fam161atm1b/tm1b, showed both photopic and sco-
revealed a fairly diminished ONL at 12 months of topic ERG response declines from 1 month until
age (23% ONL loss) progressing to 34% at the 6 months of age, where no ERG signals were
age of 18 months (Table 1) [22, 30]. The degree detected [14]. On the other hand, scotopic ERG
of progression of retinal degeneration based on response was not obtainable in Pde6brd1 mice at
the reduction of total retinal thickness and ONL any of the age time points, as they completely
rows at specific time points among the above lack of functional PDE6B and dark-rearing can-
compared groups follows this pattern (see Fig. 1): not delay the aggressive Pde6brd1 retinal degen-
eration [31]. The rd10 model, Pde6brd10, has
Pde6b rd 1 > Pde6b rd 10 > Fam161a GT / GT >
reduced light-adapted and dark-adapted
Fam161a tm1b / tm1b > Rpgr rd 9 > Nr 2e3rd 7. responses to 50% and 30% of wild-type response,
respectively, at P18, deteriorating to 30% and
10% at P30 and unrecordable by P50 [25]. ERG
3 Comparison of Retinal response in the rd7 model, Nr2e3rd7, was reported
Functional Response to be normal until the age of 5 months which sub-
sequently showed progressive reduction of both
Functional analysis of retinal function as cone and rod signals. In the presence of rosettes
measured by ERG is largely compatible with that extensively disrupt retinal integrity at
ONL thickness in retinal degenerative diseases. 1 month of age, a- and b-amplitudes at maximum
The Fam161aGT/GT model has been reported with intensities were slightly reduced compared to
abnormal scotopic response even at the age of 5 months of age when the waves have disap-
1 month that became undetectable at 4 months. peared. ERG amplitudes were reduced by 50% at
The light-adapted ERGs had a lesser amount of 16 months of age [20]. In 1-year-old Rpgrrd9
368 P. Gopalakrishnan et al.

Fig. 1 Comparative illustration of the ONL thickness the following RP models: Pde6brd1 0.5 M (73%), 1 M
reduction associated with rate of degeneration in RP (100%) [27]; Pde6brd10, 2 M (90%), 6 M (100%) [26];
mouse models. The ONL thickness reduction in percent- Fam161aGT/GT, 1 M (17%), 2 M (50%), 3 M (50%), 4 M
age at specific time points showing the rate of progressive (79%), 5 M (84%), 6 M (90%) [15]; and Fam161atm1b/tm1b,
retinal degeneration based on the histological measure- 1 M (23%), 3 M (63%), 6 M (79%), 8 M (94%) [14]
ments of retinal sections from the appropriate studies for

mice, functions analysis revealed a decreased analysis by immunohistochemistry (IHC)


retinal light response [22, 30]. The degree of pro- showed decreased levels of expression in all
gression of retinal degeneration based on the reti- retinal layers. This finding suggests that
nal physiology at specific time point of age, Fam161aGT/GT mice have a weak “leaky”
among the above-compared group, follows up expression of a truncated Fam161a protein that
this pattern Pde6brd1 > Pde6brd10 > Fam161aGT/ might have some residual function [15]. In the
GT
> Fam161atm1b/tm1b > Rpgrrd9 > Nr2e3rd7, which rd10 model, low level of β-PDE protein
is in parallel to the morphological finding. expression was detected by immunoblotting in
P9–P11 retinal extracts. IHC labeling for Pde6b
antibody in cryosections of P22 detected
4 Comparison of Protein immunoreactivity in the inner and outer seg-
Expression ments of photoreceptors. A similar analysis of
the rd1 model did not reveal any expression by
It is highly important to understand the Western blotting or immunostaining [25]. In
mechanism of retinal degeneration in animal Nr2e3rd7 mice retina, IHC staining of the cone
models. One important aspect in this regard is cells evidences their enhanced number which
whether functional protein is being produced in might be responsible for retinal dysplasia [21].
each model and to which extent. Fam161a Western blot analysis of Rpgrrd9, Rpgr-KO, and
protein expression by immunoblotting was wild-type retinal protein extracts by using anti-
absent in 1-month-old Fam161aGT/GT mice bodies with shared Rpgr epitopes for the two
retina; however, longitudinal retinal section major transcripts showed expression of the Rpgr
Morphological and Functional Comparison of Mice Models for Retinitis Pigmentosa 369

Ex1–19 isoform only. No specific reactivity was 4. Ferrari S, Di Iorio E, Barbaro V, Ponzin D,
Sorrentino FS, Parmeggiani F. Retinitis pigmentosa:
reported in Rpgr-KO mice with both antibodies genes and disease mechanisms. Curr. Genomics.
which clearly indicates the absence of both Rpgr 2011;12(4):238–49.
isoforms [22]. 5. Bandah-Rozenfeld D, Mizrahi-Meissonnier L,
Farhy C, Obolensky A, Chowers I, Pe’er J, Merin
S, Ben-Yosef T, Ashery-Padan R, Banin E, Sharon
D. Homozygosity mapping reveals null mutations in
5 Conclusion FAM161A as a cause of autosomal-recessive retinitis
pigmentosa. Am. J. Hum. Genet. 2010;87(3):382–91.
The different RP mouse models presented here 6. Langmann T, Di Gioia SA, Rau I, Stohr H,
Maksimovic NS, Corbo JC, Renner AB, Zrenner E,
show high variability in retinal structure and Kumaramanickavel G, Karlstetter M, Arsenijevic Y,
function. While rd1 and rd10 show an early-onset Weber BH, Gal A, Rivolta C. Nonsense mutations in
relatively severe phenotype, rd9 and rd7 models FAM161A cause RP28-associated recessive retinitis
show a relatively slow process. The two Fam161a pigmentosa. Am. J. Hum. Genet. 2010;87(3):376–81.
7. Beryozkin A, Khateb S, Idrobo-Robalino C, Khan
models show a relatively similar disease progres- M, Cremers F, Obolensky A, Hanany M, Mezer E,
sion which is moderate in severity. This makes Chowers I, Newman H, Ben-Yosef T, Sharon D, Banin
the Fam161atm1b/tm1b KO mouse model promising E. Unique combination of clinical features in a large
for long-term therapeutic modalities such as cohort of 100 patients with retinitis pigmentosa caused
by FAM161A mutations. Sci. Rep. 2020;10(1):15156.
AAV-based gene therapy and translational read-­ 8. Hu Y-S, Song H, Li Y, Xiao Z-Y, Li T. Whole-
through-­inducing drugs (TRIDs) for which KI exome sequencing identifies novel mutations
models will need to be developed. in genes responsible for retinitis pigmentosa
Based on the comparative analysis with three in 2 nonconsanguineous Chinese families. Int.
J. Ophthalmol. 2019;12(6):915–23.
criteria, retinal morphology, retinal physiology, 9. Zobor D, Balousha G, Baumann B, Wissinger
and expression of protein which we performed B. Homozygosity mapping reveals new nonsense
among the selected RP mouse models in this mutation in the FAM161A gene causing autosomal
review, the progression of retinal degeneration recessive retinitis pigmentosa in a Palestinian family.
Mol. Vis. 2014;20:178–82.
seems to be in the following range: 10. Van Schil K, Klevering BJ, Leroy BP, Pott JWR,
Bandah-Rozenfeld D, Zonneveld-Vrieling MN,
Pde6b rd 1  Pde6b rd 10  Fam161a tm1b / tm1b
GT / GT rd 9 rd 7
Sharon D, den Hollander AI, Cremers FPM, De Baere
 Fam161a  Rpgr  Nr 2e3 . E, Collin RWJ, van den Born LI. A nonsense mutation
in FAM161A is a recurrent founder allele in Dutch
and Belgian individuals with autosomal recessive
Acknowledgments This study was funded by the Israel retinitis pigmentosa. Invest. Ophthalmol. Vis. Sci.
Science Foundation (grant number 1147/18). 2015;56(12):7418–26.
11. Duncan JL, Biswas P, Kozak I, Navani M, Syed
Conflict of Interest Statement The authors declare no R, Soudry S, Menghini M, Caruso RC, Jeffrey
financial or proprietary and conflict of interest. BG, Heckenlively JR, Reddy GB, Lee P, Roorda
A, Ayyagari R. Ocular phenotype of a family
with FAM161A-associated retinal degeneration.
Ophthalmic Genet. 2014:1–9.
References 12. Zhou Y, Saikia BB, Jiang Z, Zhu X, Liu Y, Huang
L, Kim R, Yang Y, Qu C, Hao F, Gong B, Tai Z,
1. Cremers FPM, Boon CJF, Bujakowska K, Zeitz Niu L, Yang Z, Sundaresan P, Zhu X. Whole-exome
C. Special Issue Introduction: Inherited Retinal sequencing reveals a novel frameshift mutation in the
Disease: Novel Candidate Genes, Genotype-­ FAM161A gene causing autosomal recessive retinitis
Phenotype Correlations, and Inheritance Models. pigmentosa in the Indian population. J. Hum. Genet.
Genes (Basel). 2018;9(4):215. 2015;60(10):625–30.
2. Hanany M, Rivolta C, Sharon D. Worldwide carrier 13. Rivas MA, Vecino E. Animal models and different
frequency and genetic prevalence of autosomal therapies for treatment of retinitis pigmentosa. Histol.
recessive inherited retinal diseases. Proc. Natl. Acad. Histopathol. 2009;24(10):1295–322.
Sci. 2020;117(5):2710–6. 14. Beryozkin A, Matsevich C, Obolensky A, Kostic C,
3. Hartong DT, Berson EL, Dryja TP. Retinitis Arsenijevic Y, Wolfrum U, Banin E, Sharon D. A new
pigmentosa. Lancet. 2006;368(9549):1795–809.
370 P. Gopalakrishnan et al.

mouse model for retinal degeneration due to Fam161a mouse, a model of X-linked RP. Front. Neurosci.
deficiency. Sci. Rep. 2021;11(1):2030. 2019;13:991.
15. Karlstetter M, Sorusch N, Caramoy A, Dannhausen 24. Chang B, Hawes NL, Hurd RE, Davisson MT,
K, Aslanidis A, Fauser S, Boesl MR, Nagel-Wolfrum Nusinowitz S, Heckenlively JR. Retinal degeneration
K, Tamm ER, Jagle H, Stoehr H, Wolfrum U, mutants in the mouse. Vision Res. 2002;42(4):517–25.
Langmann T. Disruption of the retinitis pigmentosa 25. Chang B, Hawes NL, Pardue MT, German AM,
28 gene Fam161a in mice affects photoreceptor ciliary Hurd RE, Davisson MT, Nusinowitz S, Rengarajan
structure and leads to progressive retinal degeneration. K, Boyd AP, Sidney SS, Phillips MJ, Stewart RE,
Hum. Mol. Genet. 2014;23(19):5197–210. Chaudhury R, Nickerson JM, Heckenlively JR,
16. Keeler C. Retinal degeneration in the mouse is rodless Boatright JH. Two mouse retinal degenerations
retina. J. Hered. 57(2):47–50. caused by missense mutations in the beta-subunit
17. Pittler SJ, Keeler CE, Sidman RL, Baehr W. PCR of rod cGMP phosphodiesterase gene. Vision Res.
analysis of DNA from 70-year-old sections of rodless 2007;47(5):624–33.
retina demonstrates identity with the mouse rd defect. 26. Rösch S, Johnen S, Müller F, Pfarrer C, Walter
Proc. Natl. Acad. Sci. U. S. A. 1993;90(20):9616–9. P. Correlations between ERG, OCT, and anatomical
18. Pittler SJ, Baehr W. Identification of a nonsense findings in the rd10 mouse. J. Ophthalmol.
mutation in the rod photoreceptor cGMP 2014;2014:874751.
phosphodiesterase beta-subunit gene of the rd mouse. 27. Kim KH, Puoris’haag M, Maguluri GN, Umino Y,
Proc. Natl. Acad. Sci. U. S. A. 1991;88(19):8322–6. Cusato K, Barlow RB, de Boer JF. Monitoring mouse
19. Wu W-H, Tsai Y-T, Justus S, Lee T-T, Zhang L, Lin C-S, retinal degeneration with high-resolution spectral-­
Bassuk AG, Mahajan VB, Tsang SH. CRISPR repair domain optical coherence tomography. J. Vis.
reveals causative mutation in a preclinical model of 2008;8(1):17–111.
retinitis Pigmentosa. Mol. Ther. 2016;24(8):1388–94. 28. Nakazawa M, Hara A, Ishiguro S-I. Optical coherence
20. Akhmedov NB, Piriev NI, Chang B, Rapoport AL, tomography of animal models of retinitis Pigmentosa:
Hawes NL, Nishina PM, Nusinowitz S, Heckenlively from animal studies to clinical applications. Biomed.
JR, Roderick TH, Kozak CA, Danciger M, Davisson Res. Int. 2019;2019:8276140.
MT, Farber DB. A deletion in a photoreceptor-­specific 29. Pennesi ME, Michaels KV, Magee SS, Maricle A,
nuclear receptor mRNA causes retinal degeneration Davin SP, Garg AK, Gale MJ, Tu DC, Wen Y, Erker
in the rd7 mouse. Proc. Natl. Acad. Sci. U. S. A. LR, Francis PJ. Long-term characterization of retinal
2000;97(10):5551–6. degeneration in rd1 and rd10 mice using spectral
21. Haider NB, Naggert JK, Nishina PM. Excess cone domain optical coherence tomography. Invest.
cell proliferation due to lack of a functional NR2E3 Ophthalmol. Vis. Sci. 2012;53(8):4644–56.
causes retinal dysplasia and degeneration in rd7/rd7 30. Collin GB, Gogna N, Chang B, Damkham N, Pinkney
mice. Hum. Mol. Genet. 2001;10(16):1619–26. J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs
22. Thompson DA, Khan NW, Othman MI, Chang B, Jia MP. Mouse Models of Inherited Retinal Degeneration
L, Grahek G, Wu Z, Hiriyanna S, Nellissery J, Li T, with Photoreceptor Cell Loss. Cell. 2020;9(4):4.
Khanna H, Colosi P, Swaroop A, Heckenlively JR. Rd9 31. Han J, Dinculescu A, Dai X, Du W, Smith WC, Pang
is a naturally occurring mouse model of a common J. Review: the history and role of naturally occurring
form of retinitis pigmentosa caused by mutations in mouse models with Pde6b mutations. Mol. Vis.
RPGR-ORF15. PloS One. 2012;7(5):e35865. 2013;19:2579–89.
23. Falasconi A, Biagioni M, Novelli E, Piano I, Gargini
C, Strettoi E. Retinal phenotype in the rd9 mutant
Current Advancements in Mouse
Models of Retinal Disease

T. J. Hollingsworth, Xiangdi Wang,


Raven N. Simpson, William A. White,
Robert W. Williams, and Monica M. Jablonski

Abstract multifactorial, and/or polygenic diseases is a


need that has yet to be fulfilled. While models
The field of retinal degenerative (RDs) disease for AMD and RP do exist, they often require
study has been in a state of exponential growth aging the animals for a year or more, feeding
from discovering the underlying genetic com- special diets, or introduction of external mod-
ponents of such diseases as age-related macu- ulators such as exposure to cigarette smoke.
lar degeneration (AMD) and retinitis Currently, work is being done to uncover
pigmentosa (RP) to the first gene therapy high-fidelity naturally occurring models of
developed and approved for human Leber these retinal diseases with the hope and intent
congenital amaurosis. However, a source for of providing the vision community the tools it
high-fidelity animal models of these complex, needs to better understand, treat, and, one day,
cure the patients suffering from these devas-
tating afflictions.
T. J. Hollingsworth (*) · X. Wang · R. N. Simpson ·
W. A. White · M. M. Jablonski Keywords
Department of Ophthalmology, University of
Tennessee Health Sciences Center,
Age-related macular degeneration · Retinitis
Memphis, TN, USA pigmentosa · Retinal dystrophies · Mouse
Department of Medicine, University of Tennessee
models · Retinal degenerations · Preclinical
Health Sciences Center, Memphis, TN, USA models
Hamilton Eye Institute, University of Tennessee
Health Sciences Center, Memphis, TN, USA
e-mail: thollin1@uthsc.edu; xwang17@uthsc.edu;
rdavi122@uthsc.edu; wilawhit@uthsc.edu; 1 Introduction
mjablonski@uthsc.edu
R. W. Williams Retinal neurodegenerative diseases come in mul-
Department of Medicine, University of Tennessee tiple shapes and forms. These can range from dis-
Health Sciences Center, Memphis, TN, USA eases like age-related macular degeneration
Hamilton Eye Institute, University of Tennessee (AMD) and glaucoma which both lead to irre-
Health Sciences Center, Memphis, TN, USA versible blindness but almost exclusively or typi-
Department of Genetics, Genomics, and Informatics, cally affect patients in middle to old age to
University of Tennessee Health Sciences Center, retinitis pigmentosa (RP) and Leber congenital
Memphis, TN, USA amaurosis (LCA) which affect patients from birth
e-mail: rwilliams@uthsc.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 371
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_54
372 T. J. Hollingsworth et al.

and often cause complete blindness by adult- have polygenic origins with a specific example of
hood. These diseases all have different etiologies RDS/ROM1 compound mutations or triallelic
with a strong commonality … each has a genetic cases of BBS2 and BBS6 [7].
component playing a role in its pathogenesis. In
order to study these diseases, high-fidelity animal
models mimicking the human condition are 3 Current Mouse Models of RP
essential. The purpose of this review is to discuss
the current state of mouse models in the study of Mouse models of RP have been fairly simple to
the retinal diseases AMD and RP concluding make. Often, genetically ablating the protein of
with the advancements being made currently in interest has provided models of significant value
enhancing the repertoire of relevant, spontaneous to the field. Countless examples of these knock-
mouse models of the diseases. out (KO) mice exist including those for Rho,
Gnat1, Pde6b, and others [8–12] as well as for
RPE-specific genes such as Rpe65, Lrat, and
2 Inherited Retinal Mertk [4, 7, 13–16]. While these models do
Dystrophies (IRDs): exhibit RP-like phenotypes observed in humans,
A “Monogenic” Retinal they are only “true” models of humans bearing
Degeneration nonsense null mutations and a resultant recessive
RP. To better mimic human RP, researchers have
Many IRDs are classified as RP. RP is typically moved beyond simple genetic ablation into the
initiated by mutations in genes affecting the generation of mouse models in which the normal
health or function of photoreceptors (PRs) or gene is completely replaced with a mutated RP
retinal pigment epithelium (RPE) and has a prev- gene (i.e., X349E, Q344X, and P23H Rho knock-
alence on roughly 1 in 4000–5000 births [1]. RP in mice) or mutating the normal mouse gene
­
inheritance is diverse, with mutations in over 80 using Crispr/Cas9 [1, 6, 17–19].
genes known to cause some form of the disease
[2], and these mutations are heritable by all forms
of inheritance (autosomal dominant and autoso- 4 AMD: A Perfect Storm
mal recessive being the most common forms) [3, of Genetic Complexity
4]. Many genes are culpable in RP, with RHO and Environmental Stressors
being the first gene identified [5]. ~10% of all RP
cases are associated with RHO mutations, the AMD is the leading cause of irreparable blindness
majority of which are dominant [6]. Other genes in elderly populations and globally affects between
involved in phototransduction also cause RP 150 and 200 million individuals with that number
when mutated including GNAT1, PDE6A, increasing dramatically with time to nearly 300
CNGA1, and others [3]. million by 2040 [20, 21]. The primary risk factor
Many severe RP cases often tamper with the for AMD is age itself; however, multiple other risk
rod cell morphogenesis, specifically the outer factors, both modifiable and non-­ modifiable,
segment/connecting cilium [3]. These can include include race, sex, diet/weight, smoking, and genet-
mutations in genes such as CEP290 and RPGR ics [21–23]. As diet, smoking, and weight are
that are essential for the ciliary formation and modifiable risk factors, work on the genetics
genes involved in ciliary cargo transport like underlying the AMD pathophysiology has gar-
KIF3A and BBS genes [3]. More severe RPs are nered much attention. Genes with known risk-
instigated by mutations in genes affecting the associated alleles for AMD include those
visual cycle such as IRBP, LRAT, or RPE65, as associated with inflammation, oxidative stress,
well as genes responsible for the outer segment lipid metabolism, neovascularization, as well as
growth and renewal like MERTK [4]. Some RPs other physiological pathways, especially those
thought to be monogenic have been shown to associated with retinal homeostasis [21–27].
Current Advancements in Mouse Models of Retinal Disease 373

5 Current Mouse Models persion glaucoma [37]; thus, one of the first
of AMD ocular diseases modeled with the BXD family
was glaucoma [38–40]. Very recently, work has
Mouse models of AMD have come a long way begun to use systems genetics; much like has
but without much advancement. Many of the first been performed in glaucoma studies with the
models developed followed the course of aging in BXD mice, to elucidate higher fidelity models
conjunction with monogenic manipulation, per- of human AMD. Over 25 of the known genes
turbation using environmental stressors, or both that have been correlated to AMD pathogenesis/
[23, 25, 28–32]. Due to the strong correlations progression were analyzed in the over 150 BXD
between the complement system, oxidative strains. Of these, 11 genes were observed that fit
stress, and obesity/Western diet, many of the cur- the selection criteria chosen for the study: (1)
rently available models are those having effectors the gene must have an association to human
in these areas. Among the complement models AMD; (2) the gene must possess cis-expression
are the complement factor H Y402H transgenic quantitative trait loci (eQTLs) with a logarithm
mouse which, when aged to 24 months and fed a of the odds (LOD) score above 10 (10 = genome-
high fat, develops a thickening of Bruch’s mem- wide significance); (3) the gene must show dif-
brane, small drusen-like deposits observable by ferential expression between the founder B6 and
histology, and upregulation of apolipoproteins D2 strains; and (4) the genes must bear single
B48 and A1 [25], as well as transgenic comple- nucleotide polymorphisms (SNPs) in regulatory
ment factor 3 overexpressing mice and the cluster or coding regions predicted to have an impact
of differentiation 46 knockout mice [30, 33]. on the gene product’s structure, function, and/or
Some oxidative stress models include superoxide expression. Thus far, two strains of mice have
dismutase (SOD) 1 KO and SOD2 knockdown been observed to show significant phenotypical
mice along with mice exposed to cigarette smoke correlations to human AMD. BXD34 and
toxins. These exhibit AMD phenotypes such as BXD38 thus far appear to be possible models of
Bruch’s membrane thickening, RPE stress, and dry to wet transversion AMD and dry AMD,
loss of PR function [28, 29, 34]. Models pertain- respectively. Surprisingly, a strain with rapid
ing to obesity/Western diet include mice lacking degeneration, BXD32, exhibits an RP-like phe-
functional apolipoprotein E as well as cluster of notype. All three strains present with typical
differentiation 36 and receptors for low-density anatomical and functional deficits associated
lipoproteins which all exhibit Bruch’s membrane with AMD or RP by optical coherence tomogra-
thickening and drusen-like deposits [31, 32, 35]. phy (OCT), funduscopy, fluorescein angiogra-
phy (FA), electroretinography, and optokinetic
nystagmography (Fig. 1).
6 Systems Genetics
and the BXD Family of Mice:
The Future of Retinal Disease 7 Conclusions
Modeling?
Modeling AMD and RP in mice is necessary for
While most RD models originate through studying and developing treatments. While no
genetic manipulation like KOs and KIs, another single model generated to date is 100% ideal at
family of inbred mice has emerged as a unique recapitulating the human disease, many do pro-
and better model system. The BXD mouse fam- vide solid foundations for RP and AMD studies.
ily, initiated in 1971 by breeding a C57B/6 J to The development of the BXD mouse family has
a DBA/2 J, has proven its worth as a source of provided the ability to use varying SNP profiles
models of numerous human diseases including allowing for higher fidelity models of the human
cancer and neurodegeneration [36]. The condition with the ultimate goal of treating and
DBA/2 J mouse is a model for pigmentary dis- curing these blinding diseases.
Fig. 1 The BXD mouse family can model AMD and RP. RP including pigment appearing through the neural retina
(*) and attenuated retinal vessels (
Most mouse models of require in-depth molecular bio-
). BXD34 displays
logical cloning expertise (RP/AMD) and/or special condi-
tions to display a phenotype (i.e., aging to >12 months of hyper-reflective foci ( ) and vascular leakage (#) by
age, feeding HFD, etc. for AMD). By mining the BXD 9 months of age, while BXD38 exhibits no vascular
family of mice, we have found a strain of mouse exhibit- anomalies by 9 months of age while appearing to show
ing a possible polygenic RP phenotype spontaneously
occurring (BXD32) and two possible models of AMD, possible lipid deposits ( ). (c) By ERG, both BXD34
both dry (BXD38) and dry/wet (BXD34) forms. (a) OCT and 38 vary little from B6, while BXD32 shows essen-
analysis shows BXD32 exhibits marked retinal thinning tially a complete loss of retinal function. (d) OKN exams
by 6 months of age while BXD38 and 34 both exhibit revealed little difference between the contrast sensitivities
fairly normal retinal structure but display small (BXD34) of BXD34 and 38 compared to B6; however, BXD34
and remarkably large (BXD38) drusen-like deposits ( showed a loss of visual acuity with age. BXD32 lacked
). (b) By funduscopy and FA, hallmark characteristics of both CS and VA compared to all strains in agreement with
the ERG results
Current Advancements in Mouse Models of Retinal Disease 375

References of 11-cis-vitamin A in the retinal visual cycle. Nat.


Genet. 1998;20(4):344–51.
15. Duncan JL, Yang H, Vollrath D, Yasumura D, Matthes
1. Hollingsworth TJ, Hubbard MG, Levi HJ, White W,
MT, Trautmann N, et al. Inherited retinal dystro-
Wang X, Simpson R, et al. Proinflammatory pathways
phy in Mer knockout mice. Adv. Exp. Med. Biol.
are activated in the human Q344X rhodopsin knock-in
2003;533:165–72.
mouse model of retinitis pigmentosa. Biomolecules.
16. Batten ML, Imanishi Y, Maeda T, Tu DC, Moise
2021;11(8)
AR, Bronson D, et al. Lecithin-retinol acyltransfer-
2. Yi Z, Ouyang J, Sun W, Li S, Xiao X, Zhang
ase is essential for accumulation of all-trans-retinyl
Q. Comparative exome sequencing reveals novel can-
esters in the eye and in the liver. J. Biol. Chem.
didate genes for retinitis pigmentosa. EBioMedicine.
2004;279(11):10422–32.
2020;56:102792.
17. Bales KL, Ianov L, Kennedy AJ, Sweatt JD, Gross
3. Hollingsworth TJ, Gross AK. Defective trafficking
AK. Autosomal dominant retinitis pigmentosa rho-
of rhodopsin and its role in retinal degenerations. Int.
dopsin mutant Q344X drives specific alterations in
Rev. Cell Mol. Biol. 2012;293:1–44.
chromatin complex gene transcription. Mol. Vis.
4. Ferrari S, Di Iorio E, Barbaro V, Ponzin D,
2018;24:153–64.
Sorrentino FS, Parmeggiani F. Retinitis pigmentosa:
18. Hollingsworth TJ, Gross AK. Innate and autoimmu-
genes and disease mechanisms. Curr. Genomics.
nity in the pathogenesis of inherited retinal dystrophy.
2011;12(4):238–49.
Cell. 2020;9(3):3.
5. Dryja TP, McGee TL, Hahn LB, Cowley GS, Olsson
19. Orlans HO, Barnard AR, Patricio MI, McClements
JE, Reichel E, et al. Mutations within the rhodopsin
ME, MacLaren RE. Effect of AAV-mediated rho-
gene in patients with autosomal dominant retinitis
dopsin gene augmentation on retinal degeneration
pigmentosa. N. Engl. J. Med. 1990;323(19):1302–7.
caused by the dominant P23H rhodopsin mutation
6. Hollingsworth TJ, Gross AK. The severe autosomal
in a knock-in murine model. Hum. Gene Ther.
dominant retinitis pigmentosa rhodopsin mutant
2020;31(13–14):730–42.
Ter349Glu mislocalizes and induces rapid rod cell
20. Li H, Chintalapudi SR, Jablonski MM. Current drug
death. J. Biol. Chem. 2013;288(40):29047–55.
and molecular therapies for the treatment of atrophic
7. Rivolta C, Sharon D, DeAngelis MM, Dryja
age-related macular degeneration: phase I to phase III
TP. Retinitis pigmentosa and allied diseases: numer-
clinical development. Expert Opin. Investig. Drugs.
ous diseases, genes, and inheritance patterns. Hum.
2017;26(10):1103–14.
Mol. Genet. 2002;11(10):1219–27.
21. Fleckenstein M, Keenan TDL, Guymer RH,
8. Calvert PD, Krasnoperova NV, Lyubarsky AL, Isayama
Chakravarthy U, Schmitz-Valckenberg S, Klaver CC,
T, Nicolo M, Kosaras B, et al. Phototransduction
et al. Age-related macular degeneration. Nat. Rev.
in transgenic mice after targeted deletion of the rod
Dis. Primers. 2021;7(1):31.
transducin alpha -subunit. Proc. Natl. Acad. Sci. U. S.
22. Rho J, Percelay P, Pilkinton S, Hollingsworth TJ,
A. 2000;97(25):13913–8.
Kornblau I, Jablonski MM. An overview of age-­
9. Choi S, Hao W, Chen CK, Simon MI. Gene expres-
related macular degeneration: clinical, pre-clinical
sion profiles of light-induced apoptosis in arrestin/
animal models and bidirectional translation. animal
rhodopsin kinase-deficient mouse retinas. Proc. Natl.
models in medicine and biology. InTechOpen. 2021;1
Acad. Sci. U. S. A. 2001;98(23):13096–101.
23. Al-Zamil WM, Yassin SA. Recent developments in
10. Humphries MM, Rancourt D, Farrar GJ, Kenna P,
age-related macular degeneration: a review. Clin.
Hazel M, Bush RA, et al. Retinopathy induced in
Interv. Aging. 2017;12:1313–30.
mice by targeted disruption of the rhodopsin gene.
24. Gorin MB, da Silva MJ. Predictive genetics for AMD:
Nat. Genet. 1997;15(2):216–9.
hype and hopes for genetics-based strategies for treat-
11. Liang Y, Fotiadis D, Maeda T, Maeda A,
ment and prevention. Exp. Eye Res. 2020;191:107894.
Modzelewska A, Filipek S, et al. Rhodopsin
25. Landowski M, Kelly U, Klingeborn M, Groelle M,
signaling and organization in heterozygote
Ding JD, Grigsby D, et al. Human complement fac-
rhodopsin knockout mice. J. Biol. Chem.
tor H Y402H polymorphism causes an age-related
2004;279(46):48189–96.
macular degeneration phenotype and lipoprotein
12. Huttl S, Michalakis S, Seeliger M, Luo DG, Acar N,
dysregulation in mice. Proc. Natl. Acad. Sci. U. S. A.
Geiger H, et al. Impaired channel targeting and retinal
2019;116(9):3703–11.
degeneration in mice lacking the cyclic nucleotide-­
26. Lederman M, Weiss A, Chowers I. Association
gated channel subunit CNGB1. J. Neurosci.
of neovascular age-related macular degeneration
2005;25(1):130–8.
with specific gene expression patterns in periph-
13. Saari JC, Nawrot M, Kennedy BN, Garwin GG,
eral white blood cells. Invest. Ophthalmol. Vis. Sci.
Hurley JB, Huang J, et al. Visual cycle impairment
2010;51(1):53–8.
in cellular retinaldehyde binding protein (CRALBP)
27. Mansoor N, Wahid F, Azam M, Shah K, den
knockout mice results in delayed dark adaptation.
Hollander AI, Qamar R, et al. Molecular mechanisms
Neuron. 2001;29(3):739–48.
of ­complement system proteins and matrix metallo-
14. Redmond TM, Yu S, Lee E, Bok D, Hamasaki D,
proteinases in the pathogenesis of age-related macular
Chen N, et al. Rpe65 is necessary for production
degeneration. Curr. Mol. Med. 2019;19(10):705–18.
376 T. J. Hollingsworth et al.

28. Espinosa-Heidmann DG, Suner IJ, Catanuto neovascularization, and retinal pigment epithelium
P, Hernandez EP, Marin-Castano ME, Cousins dysfunction in SOD1-deficient mice: a model of age-­
SW. Cigarette smoke-related oxidants and the devel- related macular degeneration. Proc. Natl. Acad. Sci.
opment of sub-RPE deposits in an experimental ani- U. S. A. 2006;103(30):11282–7.
mal model of dry AMD. Invest. Ophthalmol. Vis. Sci. 35. Dithmar S, Curcio CA, Le NA, Brown S, Grossniklaus
2006;47(2):729–37. HE. Ultrastructural changes in Bruch’s membrane of
29. Justilien V, Pang JJ, Renganathan K, Zhan X, apolipoprotein E-deficient mice. Invest. Ophthalmol.
Crabb JW, Kim SR, et al. SOD2 knockdown mouse Vis. Sci. 2000;41(8):2035–42.
model of early AMD. Invest. Ophthalmol. Vis. Sci. 36. Ashbrook DG, Arends D, Prins P, Mulligan MK, Roy
2007;48(10):4407–20. S, Williams EG, et al. A platform for experimental
30. Lyzogubov VV, Bora PS, Wu X, Horn LE, de Roque precision medicine: the extended BXD mouse family.
R, Rudolf XV, et al. The complement regulatory pro- Cell Syst. 2021;12(3):235–47. e9
tein CD46 deficient mouse spontaneously develops 37. Anderson MG, Smith RS, Hawes NL, Zabaleta
dry-type age-related macular degeneration-like phe- A, Chang B, Wiggs JL, et al. Mutations in genes
notype. Am. J. Pathol. 2016;186(8):2088–104. encoding melanosomal proteins cause pigmen-
31. Picard E, Houssier M, Bujold K, Sapieha P, Lubell W, tary glaucoma in DBA/2J mice. Nat. Genet.
Dorfman A, et al. CD36 plays an important role in the 2002;30(1):81–5.
clearance of oxLDL and associated age-­dependent sub- 38. King R, Li Y, Wang J, Struebing FL, Geisert
retinal deposits. Aging (Albany NY). 2010;2(12):981–9. EE. Genomic locus modulating IOP in the BXD RI
32. Rudolf M, Ivandic B, Winkler J, Schmidt-Erfurth Mouse Strains. G3 (Bethesda). 2018;8(5):1571–8.
U. Accumulation of lipid particles in Bruch’s mem- 39. King R, Struebing FL, Li Y, Wang J, Koch AA, Cooke
brane of LDL receptor knockout mice as a model of Bailey JN, et al. Genomic locus modulating cor-
age-related macular degeneration. Ophthalmologe. neal thickness in the mouse identifies POU6F2 as a
2004;101(7):715–9. potential risk of developing glaucoma. PLoS Genet.
33. Cashman SM, Desai A, Ramo K, Kumar-Singh 2018;14(1):e1007145.
R. Expression of complement component 3 (C3) from 40. Swaminathan S, Lu H, Williams RW, Lu L, Jablonski
an adenovirus leads to pathology in the murine retina. MM. Genetic modulation of the iris transillumina-
Invest. Ophthalmol. Vis. Sci. 2011;52(6):3436–45. tion defect: a systems genetics analysis using the
34. Imamura Y, Noda S, Hashizume K, Shinoda K, expanded family of BXD glaucoma strains. Pigment
Yamaguchi M, Uchiyama S, et al. Drusen, choroidal Cell Melanoma Res. 2013;26(4):487–98.
Single-Cell Transcriptomic
Profiling of Müller Glia in the rd10
Retina

Duygu Sigurdsson and Christian Grimm

Abstract might arise from a novel immune function of


Müller glia in RP. We also describe a possible
Müller glia are the principal macroglia of the decrease in glial lipid biogenesis, which might
retina and support retinal neurons both in affect degenerating photoreceptors.
health and disease. In retinitis pigmentosa
(RP), a highly heterogeneous inherited retinal Keywords
disorder, the most common form of pathology
Single-cell RNA sequencing ·
involves primary rod degeneration, followed
Transcriptomics · Müller glia · Retinal
by secondary cone death. To investigate
degeneration · Retinitis pigmentosa · rd10
Müller glia responses to rod degeneration, we
mouse model
performed droplet-based single-cell RNA
sequencing in the rd10 mouse model of RP
during primary rod degeneration. We con-
firmed known MG behavior on gliosis, meta- 1 Introduction
bolic, and immune functions. Pde6brd10 Müller
glia also exhibited an increased expression of Retinitis pigmentosa (RP) is a highly heteroge-
histocompatibility complex members, which neous inherited retinal degenerative disorder that
is a leading cause of blindness in children and
D. Sigurdsson (*) working-age adults. RP is characterized by pri-
Lab for Retinal Cell Biology, Department of mary rod degeneration, commonly due to a rod-­
Ophthalmology, University Hospital Zurich, specific causative mutation, followed by the
University of Zurich, Zurich, Switzerland secondary death of cones [7]. The genetic hetero-
Center for Integrative Human Physiology, University geneity of RP complicates the correction of caus-
of Zurich, Zurich, Switzerland ative mutations for treatment, e.g., by gene
e-mail: duygu.sigurdsson@iob.ch
therapy. In contrast, the common disease patho-
C. Grimm physiology provides an avenue for mutation-­
Lab for Retinal Cell Biology, Department of
Ophthalmology, University Hospital Zurich, independent approaches that support
University of Zurich, Zurich, Switzerland neuroprotection [15].
Center for Integrative Human Physiology, University Müller glia (MG) are the principal macroglia
of Zurich, Zurich, Switzerland of the retina and provide various kinds of support
Neuroscience Center Zurich, University of Zurich, to surrounding photoreceptors, especially cones,
Zurich, Switzerland including recycling the cone visual pigment [2,

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 377
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_55
378 D. Sigurdsson and C. Grimm

25]. MG also release neurotrophic factors for Bioinformatic Analysis Sequencing data were
photoreceptors upon injury [12, 23]. In addition, mapped to the GRCm38.p5 genome assembly
MG enter gliosis upon damage [5], partake in and were converted to a cell-gene matrix with the
retinal remodeling [18], and contribute to the Cell Ranger pipeline. Downstream analysis was
microglial response [22]. However, the heteroge- performed with Seurat v.3.1 [3]. Only cells with
neity of the retina has so far hindered high-­ 200–6000 unique genes and 1000–20,000 tran-
throughput studies of changes that occur scripts were retained. Data was log-normalized
specifically in MG during rod degeneration, with a scale factor of 10,000 and scaled. 5,000
which may provide clues as to how MG affect most highly variable genes with 0.1–8 average
photoreceptor survival in RP. expression and >1 dispersion were selected for
analysis. The first 50 principal components were
used for the k-nearest neighbor graph, and clus-
2 Materials and Methods tering was performed with modularity optimiza-
tion on a shared nearest neighbor graph with
Animal Experimentation Animal maintenance resolution = 0.8. Cell cluster identity was
and experiments were performed according to assigned using known marker gene expression. A
the standards of the ARVO Statement for the Use previously published algorithm was used for dou-
of Animals in Ophthalmic and Vision Research blet filtering [9]. A cluster expressing markers for
and the regulations of the Veterinary Office of both MG (e.g., Rlbp1, Glul) and rods (e.g., Sag,
Canton Zürich, Switzerland, under the license Rho) was considered to be rod-contaminated MG
ZH141/2016. rd10 (Pde6brd10/rd10) and C57/BL6 and excluded from the analysis. Differential gene
mice were purchased from Jackson Laboratories expression was analyzed with Seurat, using the
(Bar Harbor, USA) and maintained in the Wilcoxon rank-sum test and Bonferroni correc-
Laboratory Animal Services Center facilities of tion. Due to the small size of the dataset, genes
the University of Zürich under a 14 h:10 h light to with an adjusted p-value of 0.1 or lower were
dark cycle with food and water ad libitum. considered significant. The dataset containing all
Euthanasia was performed with CO2 exposure cell types is available in the Gene Expression
and cervical dislocation. Omnibus repository (GSE183206).

Retina Dissociation and scRNAseq Retinas 3 Results


were isolated from mice at postnatal day (P) 21
and dissociated with the Worthington Papain To study single MG transcriptomes during primary
Dissociation System (Worthington, Lakewood, rod degeneration, we performed single-cell RNA
NJ, USA). Cell viability was assessed with sequencing (scRNAseq) on the retinas of two rd10
2 μM calcein-AM (Sigma-Aldrich, St. Louis, animals and two wild-type controls, one male and
MO, USA) and 3 μM propidium iodide (Sigma-­ one female each, at P21. After quality control, 269
Aldrich). Five million cells were fixed with MG identified by marker gene expression (e.g.,
methanol according to the 10X Genomics pro- Glul) were retained for analysis (Fig. 1a).
tocol (10X Genomics, Pleasanton, CA, USA) More MG were isolated from rd10 (206 cells)
for scRNAseq. 10,000 cells from each sample than from wild-type retinas (63 cells), with a
were loaded onto an individual lane of a 10X comparable ratio of cells originating from males
Genomics Chromium Single-Cell microfluid- vs. females (22.8% female for rd10, 23.8% for
ics chip using the V3 chemistry (10X wild types). Therefore, we pooled cells from
Genomics). Libraries were sequenced on an male and female samples together for further
Illumina NovaSeq 6000 platform (Illumina, analyses. rd10 MG expressed more transcripts
San Diego, CA, USA) at the Functional and more unique genes than wild-type MG, with
Genomics Center of the University of Zürich/ a higher percentage of mitochondrial and ribo-
ETH Zürich. somal transcripts (Fig. 1b). The increase in these
Fig. 1 Transcriptomic properties of Müller glia in rd10 (percent.ribo) genes in each cell. (c) Volcano plot for dif-
and wild-type retinas analyzed in this study. (a) Expression ferentially expressed genes in rd10 MG compared to
of the MG marker gene Glul at the single-cell level. (b) wild-type MG. Colored dots refer to genes with an
Violin plots for the main quality control metrics for cells adjusted p-value <0.1. Labeled genes are expressed with
Fig. 1 (continued) from each genotype, including the log2 fold change >0.85. Genes upregulated in rd10 cells
number of transcripts, unique genes expressed, and per- have a positive log2 fold change. UMAP: uniform mani-
centage of mitochondrial (percent.mt) and ribosomal fold approximation and projection. wt wild type
380 D. Sigurdsson and C. Grimm

transcripts may stem from the activation of an rd10 MG. In addition, the levels of two gene
MG response in the rd10 retina that involves expression regulators, Auts2 and Pax6, were sig-
increased levels of mRNA translation and nificantly lower in the rd10 MG captured in our
increased energy metabolism. A possible techni- dataset (Fig. 1c). Auts2 activates neuronal gene
cal artifact of high mtDNA and ribosomal con- expression in the developing brain, where it does
tent due to low cell quality in rd10 MG is not co-localize with glia [4]. Pax6 is a transcrip-
countered by their higher transcript count, as low tion factor expressed in MG, horizontal cells,
cell quality is associated with lower transcript amacrine cells, and retinal ganglion cells [20].
counts [11].
To identify the genes that were differentially
regulated in rd10 MG due to primary rod degen- 4 Discussion
eration, we carried out differential gene expres-
sion analysis between rd10 and wild-type glia in Using scRNAseq on the P21 rd10 retina to study
our dataset (Fig. 1c). Unsurprisingly, gliosis-­ MG, we were able to confirm certain patterns of
associated Gfap was the most highly upregulated MG response to rod degeneration found in other
gene in rd10 glia [5]. The upregulation of extra- models, including gliosis, immune modulation
cellular matrix protein Cd44, seen during second- by release of complement factors, and changes to
ary cone degeneration in other models of RP iron metabolism [21]. Furthermore, we generated
[21], may also contribute to retinal remodeling. some novel insight that might be relevant in
While the increases in leukemia inhibitory factor, understanding glial responses to rod injury in RP.
glycoprotein 130, and ciliary neurotrophic factor Multiple MHC-associated transcripts were
receptor were not statistically significant, the lev- increased in rd10 MG (Fig. 1c). In RP models,
els of cytokine signaling mediator Stat3 and studies on antigen-presenting capabilities of glia
tumor necrosis factor receptor superfamily mem- focused on microglia [16]. However, there has
ber Tnfrsf1a were significantly higher in rd10 been recent evidence both in vitro after lipopoly-
MG. Similar to the rd1 model of RP, mediators of saccharide stimulation [14] and in vivo in an
the complement system were increased in rd10 autoimmune uveoretinitis model [8] that MG
MG (C4b, Serping1) [21]. In addition, we found express components of the MHC and may act as
that several major histocompatibility complex antigen-presenting cells. According to our find-
(MHC) components and regulators (H2-D1, H2-­ ings, in the rd10 retina, where the cause of degen-
K1, B2m, A2m) were also higher in rd10 MG, eration is not directly associated with immunity,
potentially indicating an antigen-presenting role MG might also be performing a similar function.
of these cells in the rd10 retina. As in other RP Not much is known about AC149090.1, the
models [21], rd10 MG upregulated various genes most downregulated transcript in rd10 MG. If
involved in metal homeostasis and anti-­oxidation, AC149090.1 encodes a PSD-like protein, its
including Mt1, Mt2, Trf (Fig. 1c), Selenow, and decrease in the rd10 MG might lead to decreased
Msrb2 (not shown). Another support role MG production of PE, a major lipid moiety in the
may play in degeneration is via increased retinol-­ retina. Since MG can process and provide PE to
binding protein 1 (Rbp1), which is crucial for the photoreceptors [19] and photoreceptor lipid com-
visual cycle [10] that is also increased in other position changes in other models of RP [6],
forms of retinal injury [17]. Müller glial changes in PE biogenesis might also
The most strongly decreased gene in rd10 MG affect photoreceptors. As PE is a docosahexae-
was AC149090.1. This gene is an ortholog of noic acid (DHA) carrier, it is tempting to specu-
phosphatidylserine decarboxylase (PSD), which late that these changes might contribute to
is essential for the biogenesis of phosphatidyl- lowered DHA levels observed in RP [1].
ethanolamine (PE) [24]. Various genes involved PAX6-positive MG are present in rd10 retinas,
in RNA processing (Arglu1, Son, Snrnp70, with whole-retinal Pax6 expression comparable
Prpf4b, Ddx5, Cirbp) were downregulated in to wild types at P21. With further degeneration,
Single-Cell Transcriptomic Profiling of Müller Glia in the rd10 Retina 381

Pax6 levels increased and some PAX6+ MG 9. Hoang T, Wang J, Boyd P, et al. Gene regulatory
networks controlling vertebrate retinal regenera-
nuclei migrated toward the outer nuclear layer tion. Science. 2020; https://doi.org/10.1126/science.
[13]. Since Pax6 is expressed by various inner abb8598.
retinal cells, a partial or transient decrease might 10. Huang J, Possin DE, Saari JC. Localizations of visual
not be detectable in whole-retinal samples but cycle components in retinal pigment epithelium. Mol
Vis. 2009;15:223–34.
still affect the MG transcriptome. Alternatively, 11. Ilicic T, Kim JK, Kolodziejczyk AA, et al.
migrating PAX6+ MG might differentiate from Classification of low quality cells from single-cell
non-migrating MG enough to “escape” the main RNA-seq data. Genome Biol. 2016;17:29.
MG cluster, while Pax6 levels decrease in the 12. Joly S, Lange C, Thiersch M, et al. Leukemia inhibi-
tory factor extends the lifespan of injured photorecep-
non-migrating MG. Together with Auts2, which tors in vivo. J Neurosci. 2008;28:13765–74.
was not previously described in the retina, these 13. Joly S, Pernet V, Samardzija M, Grimm C. Pax6-­
results are of interest for validation, as they can positive Müller glia cells express cell cycle markers
contribute to our understanding of stem cell-like but do not proliferate after photoreceptor injury in the
mouse retina. Glia. 2011;59:1033–46.
properties of mouse MG. 14. Lorenz L, Hirmer S, Schmalen A, et al. Cell surface
profiling of retinal Müller glial cells reveals associa-
Acknowledgments The authors would like to thank the tion to immune pathways after LPS stimulation. Cell.
Functional Genomics Center Zürich for their excellent 2021;10 https://doi.org/10.3390/cells10030711.
guidance and assistance with scRNAseq. This study was 15. Newton F, Megaw R. Mechanisms of photoreceptor
supported by the Swiss National Science Foundation death in retinitis pigmentosa. Genes. 2020;11 https://
(grant nr. 31003A_173008). doi.org/10.3390/genes11101120.
16. Noailles A, Maneu V, Campello L, et al. Persistent
inflammatory state after photoreceptor loss in an
animal model of retinal degeneration. Sci Rep.
References 2016;6:33356.
17. Otake H, Yamamoto T, Deguchi S, et al. Retinal pro-
1. Berson EL, Rosner B, Sandberg MA, et al. Further teomic evaluation of rats following streptozotocin-­
evaluation of docosahexaenoic acid in patients injection using shotgun proteomics. Mol Med Rep.
with retinitis pigmentosa receiving vitamin A 2020;21:379–86.
treatment: subgroup analyses. Arch Ophthalmol. 18. Phillips MJ, Otteson DC, Sherry DM. Progression
2004;122:1306–14. of neuronal and synaptic remodeling in the rd10
2. Bringmann A, Pannicke T, Grosche J, et al. Müller mouse model of retinitis pigmentosa. J Comp Neurol.
cells in the healthy and diseased retina. Prog Retin 2010;518:2071–89.
Eye Res. 2006;25:397–424. 19. Politi L, Rotstein N, Carri N. Effects of docosahexae-
3. Butler A, Hoffman P, Smibert P, et al. Integrating noic acid on retinal development: cellular and molec-
single-cell transcriptomic data across different con- ular aspects. Lipids. 2001;36:927–35.
ditions, technologies, and species. Nat Biotechnol. 20. Roesch K, Jadhav AP, Trimarchi JM, et al. The tran-
2018;36:411–20. scriptome of retinal Müller glial cells. J Comp Neurol.
4. Gao Z, Lee P, Stafford JM, et al. An AUTS2-­ 2008;509:225–38.
Polycomb complex activates gene expression in the 21. Roesch K, Stadler MB, Cepko CL. Gene expression
CNS. Nature. 2014;516:349–54. changes within Müller glial cells in retinitis pigmen-
5. Gargini C, Terzibasi E, Mazzoni F, Strettoi E. Retinal tosa. Mol Vis. 2012;18:1197–214.
organization in the retinal degeneration 10 (rd10) 22. Todd L, Palazzo I, Suarez L, et al. Reactive microg-
mutant mouse: a morphological and ERG study. J lia and IL1β/IL-1R1-signaling mediate neuropro-
Comp Neurol. 2007;500:222–38. tection in excitotoxin-damaged mouse retina. J
6. Hamano F, Kuribayashi H, Iwagawa T, et al. Mapping Neuroinflammation. 2019;16:118.
membrane lipids in the developing and adult mouse 23. Ueki Y, Le Y-Z, Chollangi S, et al. Preconditioning-­
retina under physiological and pathological con- induced protection of photoreceptors requires acti-
ditions using mass spectrometry. J Biol Chem. vation of the signal-transducing receptor gp130 in
2021;296:100303. photoreceptors. Proc Natl Acad Sci U S A.
7. Hartong DT, Berson EL, Dryja TP. Retinitis pigmen- 2009;106:21389–94.
tosa. Lancet. 2006;368:1795–809. 24. Vance JE, Tasseva G. Formation and function of
8. Heng JS, Hackett SF, Stein-O’Brien GL, et al. phosphatidylserine and ­ phosphatidylethanolamine
Comprehensive analysis of a mouse model in mammalian cells. Biochim Biophys Acta.
of spontaneous uveoretinitis using single-cell 2013;1831:543–54.
RNA sequencing. Proc Natl Acad Sci U S A. 25. Wang J-S, Kefalov VJ. The cone-specific visual cycle.
2019;116:26734–44. Prog Retin Eye Res. 2011;30:115–28.
Methods for In Vivo
Characterization of Proteostasis
in the Mouse Retina

Yixiao Wang and Ekaterina S. Lobanova

Abstract 1 Introduction

An increasing number of studies connect The cellular proteome is regulated at multiple


inherited and age-related retinal degenerations levels including protein translation, folding, and
with changes in the regulation of proteostasis. degradation. A number of studies suggest that
Here, we describe technical aspects of exist- alterations in the cellular ability to maintain a
ing assays allowing to assess the status of the healthy proteome contribute to vision loss [1–7].
ubiquitin-proteasome system (UPS), changes Here, we review and show representative exam-
in autophagy, and protein translation in mouse ples for methods allowing assessment of protein
retina in vivo. These methods are helpful for translation and degradation in vivo that were suc-
the development and testing approaches to cessfully used in mouse retina in several recent
modulate proteostasis and delay vision loss. studies [1–7].

Keywords
2 Approaches to Assess
Ubiquitin-proteasome system · Autophagy · Proteostasis in Mouse Retina
Protein translation · Proteostasis · Retinal In Vivo
degeneration
2.1 Assessments of UPS
with Genetic Reporters

The efficiency of protein degradation by


ubiquitin-­proteasome system (UPS) can be eval-
uated in vivo using genetic UPS reporters [8–11].
Y. Wang In our previous studies, we took advantage of
Department of Ophthalmology, University of Florida, transgenic mice expressing the ubiquitin-­
Gainesville, FL, USA proteasome reporter UbG76V-GFP (Fig. 1a) [5–7].
E. S. Lobanova (*) This reporter is barely detectable in healthy pho-
Department of Ophthalmology, University of Florida, toreceptor cells of wild-type mice: it is quickly
Gainesville, FL, USA ubiquitinated and targeted to proteasomes for
Department of Pharmacology and Therapeutics, degradation. However, it is accumulated, and its
University of Florida, Gainesville, FL, USA clearance is impaired in photoreceptors of several
e-mail: elobanova@ufl.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 383
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_56
384 Y. Wang and E. S. Lobanova

Fig. 1 Using UbG76V-GFP reporter for UPS assessments detection. The ECL Prime Blocking Reagent (RPN418,
in photoreceptors. (a) Scheme for degradation of dam- GE Healthcare Life Sciences) was used to block mem-
aged proteins and UbG76V-GFP reporter by UPS. Part of branes. The extracts obtained from littermates negative for
the scheme is created with BioRender.com. (b) UbG76V-­ UbG76V-GFP transgene were used to control for antibody
GFP reporter fluorescence (green) in 20 μm frozen retinal specificity. A nonspecific band detected with anti-GFP
cross sections of WT and heterozygote RhoP23H/WT knock- antibody is marked as “ns band.” Extracts from retinas of
­in mice. Rod outer segments were stained with wheat reporter expressing rod transducin gamma knockout (Gγ1
germ agglutinin (WGA) conjugated to Alexa Fluor 555 KO [5]) mouse were run on the same gel as an additional
(red). The scale bar is 25 μm. (c) Western blot detection of control. (d) The quantification plot for density bands of
the UbG76V-GFP sensor in whole retina lysates (30 μg per the reporter is shown as a percentage of the average signal
lane) from mice of indicated genotypes with the anti-GFP in WT/UbG76V-GFP mice. Rabbit antibodies against Hsc70
antibodies (A11122, Invitrogen; 1:10,000) using ECL protein (ADI-SPA-819) were from Enzo Life Sciences

mouse models of retinal degeneration [6, 7, 12, the degradation of ubiquitinated proteins to delay
13]. Impaired clearance of UbG76V-GFP was vision loss.
observed in rods of transgenic mice expressing The UbG76V-GFP reporter can be detected with
human P23H rhodopsin mutant, heterozygote confocal microscopy in frozen retinal sections or
P23H rhodopsin knock-in mice, two strains of agarose cross sections prepared using standard
rod transducin gamma knockout mice, and rods protocols [6, 7, 14]. In a typical experiment
of peripherin and rhodopsin knockout mice [5– shown in Fig. 1b, the samples from evaluated
7]. Other groups reported changes in the levels of (heterozygote P23H knock-in mouse [15]) and
this reporter in retinas of BBS5 and Lrat knock- wild-type mice expressing UbG76V-GFP reporter
out mice [12, 13]. were processed together. The sections prepared
The level of reporter UbG76V-GFP is set by the from wild-type mice were used to optimize imag-
balance between its synthesis and degradation. ing settings (e.g., gain, laser power) allowing
The rate-limiting steps for processing ubiquiti- reliable detection of fluorescent signal. The same
nated proteins in the diverse mouse models men- settings (laser intensity, gain, z-stack values)
tioned above are currently unknown. It is were then used to collect images from heterozy-
unknown which specific changes in degenerating gote P23H knock-in mouse. Retinal sections pre-
rods could delay clearing of UbG76V-GFP reporter pared from mice that did not express reporter
and whether they are the same in these mice. could be used to control for autofluorescence.
However, these observations point to some kind The imaging studies were supplemented with
of UPS insufficiency as a potential stress factor Western blotting analysis to confirm and quantify
present in the degenerating photoreceptors and reporter changes. Retinal extracts from mice that
encourage the development of tools to stimulate do not express reporter were used to control for
Methods for In Vivo Characterization of Proteostasis in the Mouse Retina 385

antibody specificity. For example, the set of pri- in the levels of LC3-II in comparison with unin-
mary/secondary antibodies used in the experi- jected mice (Fig. 2b, c), confirming that it exerts
ment shown in Fig. 1c detected two nonspecific sufficient inhibitory effects as expected. The
bands (marked as “ns”). The blots of retinal injection and Western blotting procedures should
lysates prepared from mice expressing reporter be optimized in the case of inefficient inhibition
displayed two additional bands. One of the bands, or high variability between samples.
migrating under 37 kDa, corresponded to UbG76V-­ Western blot quantification (Fig. 2c) showed
GFP reporter. The second band, representing the that the CQ-induced LC3-II increase (reflecting
non-proteolyzed nonfluorescent xGFP fragment, the autophagy flux and abbreviated ΔLC3-II) in
migrated around 25 kDa [9]. The quantification control and experimental groups are indistin-
of density plots for UbG76V-GFPreporter (Fig. 1d) guishable from one another and the basal level of
complements the fluorescence imaging findings autophagy (continuous formation of phago-
for heterozygote P23H knock-in mice (Fig. 1b) somes) is not affected. Other commonly used
and together shows impaired clearance of the autophagy markers and assays could be used to
reporter. gain additional insights and are discussed else-
where [16].
The autophagy flux measurements could be
2.2 Autophagy Flux performed using immunodetection of LC3 in
Measurements retinal cross sections or detection of the fluores-
cently tagged LC3 protein in LC3-GFP trans-
The basal activity of the autophagy-lysosomal genic mice [3, 17]. Similarly, the fluorescent
pathway is responsible for the continuous degra- approach to measure autophagy flux relies on
dation of cytoplasmic material. One of the key analyzing the change in the number of phago-
steps of phagosome maturation is coordinated by somes caused by treatment with autophagy inhib-
conjugation of LC3 protein (microtubule-­ itors. It requires accurate controls for consistency
associated protein 1A/1B-light chain 3) to phos- of image collection, data analysis, and sample
phatidylethanolamine. The lipidated form of LC3 processing.
protein is abbreviated as LC3-II and non-­lipidated
as LC3-I (Fig. 2a). The changes in the basal abil-
ity of cells to form phagosomes (autophagic flux) 2.3 Assessments of Protein
could be assessed by analyzing the accumulation Translation
of LC3-II in the presence of an autophagy inhibi-
tor [16]. In the retina, this approach was pio- The adaptational changes in protein translation
neered and validated by Dr. David Zacks [3, 4]. can be efficiently assessed using SUnSET (sur-
In one of the ongoing projects shown in face sensing of translation) method [18]. This
Fig. 2b, we noticed slightly elevated levels of method is based on the ability of puromycin to
LC3-I/LC3-II markers in the sets of experimental incorporate into growing polypeptide chains
(Exp) animals compared to controls (Cntr). (Fig. 3a). Incorporated puromycin is detected by
Therefore, to seek differences in the autophagy-­ Western blotting with anti-puromycin antibodies.
lysosomal system between the groups, we per- Changes in the intensity of puromycin staining
formed autophagy flux measurements. To this are shown to be indicative of the efficiency of
end, animals from both groups were given an protein translation, and overall, this method is
intraperitoneal injection of autophagy inhibitor, shown to be as sensitive in translation studies as a
chloroquine (CQ). An accumulation of LC3-II conventional radioactive method. The use of this
was studied several hours postinjection and com- method in the retina was pioneered by Dr. Marina
pared by Western blotting with uninjected mice. Gorbatyuk [1, 2].
Efficient injection of autophagy inhibitor In a typical experiment, mice are intraperito-
should result in a statistically significant increase neally injected with puromycin solution, their
386 Y. Wang and E. S. Lobanova

Fig. 2 Autophagy flux assessments. (a) Scheme of Data are normalized on the average density of LC3-II in
phagosome formation and fusion with lysosomes demon- untreated control group. Rabbit antibodies against LC3
strating autophagy flux assessments. (b, c) Western blots protein (1:10,000) were from Cell Signaling Technology
and quantification of comparative changes in LC3 mark- (4108) and anti-beta actin antibodies (MA5-15739-D800)
ers in retinal lysates (30 μg per lane) caused by chloro- were from Invitrogen
quine (CQ) injections 4 h prior to the retina collection.

Fig. 3 Protein translation assessments. (a) Scheme dem- were blocked in TBST (20 mM Tris-HCl, pH 8.0, 0.1%
onstrating a puromycin-based approach for protein trans- Tween-20) solution containing 5% of ECL Prime
lation assessments. Part of the scheme is created with Blocking Reagent (RPN418, GE Healthcare Life
BioRender.com. (b, c) Puromycin staining of Western blot Sciences). Puromycin-modified proteins were detected
membranes and quantification of density profiles of reti- using primary mouse anti-puromycin (MABE343, EMD
nal lysates (40 μg of protein per lane) prepared from mice Millipore,1:10,000) and secondary HRP-conjugated anti-­
injected 30 min prior to the analysis with 50–100 μl of mouse IgG2a (115-035-206, Jackson Immuno Research
puromycin (0.04 μmol/g body weight) dissolved in Laboratories) chosen based on the previous studies [2,
PBS. Retinal lysates from uninjected mice were used to 18]. Enhanced chemiluminescent reactions were devel-
control for nonspecific properties of the anti-puromycin oped using ProSignal Pico ECL reagent (20–300, Genesee
antibody. (d) Western blot membranes for the same sam- Scientific). ECL signal for puromycin blots required pro-
ples as in panel (b) were stained with secondary antibod- longed ~10-min exposure. Protein bands were visualized
ies only and imaged using the same settings. Samples with the ChemiDoc Imager system (Bio-Rad Laboratories)

retinas are collected 30 min after injections, and nized in experiments performed with secondary
following solubilization, the lysates are immedi- antibodies only (Fig. 3c). The retinas of
ately analyzed by Western blotting. Samples pre- puromycin-­treated mice show additional staining
pared from uninjected mice are run on the same which was not present in uninjected mice and
gel to identify puromycin-labeled polypeptides. most likely correspond to puromycin-modified
In a typical experiment, we detected four peptide chains (Fig. 3b).
prominent nonspecific bands in the 35–75 kDa The proteins that correspond to four middle-­
region (Fig. 3b). The same bands were recog- range nonspecific bands are unknown; it is
Methods for In Vivo Characterization of Proteostasis in the Mouse Retina 387

unclear why they become darker in the toreceptors using two complementary in vivo report-
ers of proteasomal activity. eNeuro. 2020;7(1):ENE
puromycin-­injected mice (Fig. 3b). Therefore, to URO.0428-19.2019.
present data we plotted density profiles of lanes 6. Lobanova ES, Finkelstein S, Li J, Travis AM, Hao Y,
and avoided quantification of the total lane den- Klingeborn M, et al. Increased proteasomal activity
sity (Fig. 3c). supports photoreceptor survival in inherited retinal
degeneration. Nat Commun. 2018;9(1):1738.
Reagents and conditions giving the best results 7. Lobanova ES, Finkelstein S, Skiba NP, Arshavsky
in our hands are listed in the figure legend, but we VY. Proteasome overload is a common stress factor in
cannot exclude the possibility that other block- multiple forms of inherited retinal degeneration. Proc
ing/developing reagents might perform better Natl Acad Sci U S A. 2013;110(24):9986–91.
8. Salomons FA, Verhoef LG, Dantuma NP. Fluorescent
and increase signal or decrease nonspecific back- reporters for the ubiquitin-proteasome system. Essays
ground. It is worth noting that the ECL signal Biochem. 2005;41:113–28.
from the puromycin-labeled proteins is weak and 9. Dantuma NP, Lindsten K, Glas R, Jellne M, Masucci
requires prolonged exposure (10–15 min). MG. Short-lived green fluorescent proteins for quan-
tifying ubiquitin/proteasome-dependent proteolysis in
To conclude, we hope that detailed technical living cells. Nat Biotechnol. 2000;18(5):538–43.
aspects of execution and data analysis of in vivo 10. Dantuma NP, Bott LC. The ubiquitin-proteasome
proteostasis assays will aid the readers with the system in neurodegenerative diseases: precipitating
incorporation of these methods into their own factor, yet part of the solution. Front Mol Neurosci.
2014;7:70.
studies. Due to the limited space, we were not 11. Lindsten K, Menendez-Benito V, Masucci MG,
able to cite all relevant literature. Dantuma NP. A transgenic mouse model of the
ubiquitin/proteasome system. Nat Biotechnol.
Acknowledgments This work was supported by the 2003;21(8):897–902.
National Institutes of Health grants EY030043 (E.S.L.) 12. Liu YP, Tsai IC, Morleo M, Oh EC, Leitch CC, Massa
and S10D028476, the University of Florida startup funds F, et al. Ciliopathy proteins regulate paracrine signal-
(E.S.L.), and an unrestricted grant from Research to ing by modulating proteasomal degradation of media-
Prevent Blindness Foundation to the Department of tors. J Clin Invest. 2014;124(5):2059–70.
Ophthalmology of the University of Florida. 13. Xu H, Enemchukwu N, Zhong X, Zhang O, Fu
Y. Deletion of M-opsin prevents M cone degeneration
in a mouse model of Leber congenital amaurosis. Am
J Pathol. 2020;190(5):1059–67.
References 14. Lobanova ES, Finkelstein S, Song H, Tsang SH,
Chen CK, Sokolov M, et al. Transducin transloca-
1. Saltykova IV, Elahi A, Pitale PM, Gorbatyuk OS, Athar tion in rods is triggered by saturation of the GTPase-­
M, Gorbatyuk MS. Tribbles homolog 3-­ mediated activating complex. J Neurosci. 2007;27(5):1151–60.
targeting the AKT/mTOR axis in mice with retinal 15. Sakami S, Maeda T, Bereta G, Okano K, Golczak M,
degeneration. Cell Death Dis. 2021;12(7):664. Sumaroka A, et al. Probing mechanisms of photo-
2. Starr CR, Pitale PM, Gorbatyuk M. Translational receptor degeneration in a new mouse model of the
attenuation and retinal degeneration in mice with common form of autosomal dominant retinitis pig-
an active integrated stress response. Cell Death Dis. mentosa due to P23H opsin mutations. J Biol Chem.
2018;9(5):484. 2011;286(12):10551–67.
3. Qiu Y, Yao J, Jia L, Thompson DA, Zacks 16. Klionsky DJ, Abdelmohsen K, Abe A, Abedin
DN. Shifting the balance of autophagy and pro- MJ, Abeliovich H, Acevedo Arozena A, et al.
teasome activation reduces proteotoxic cell Guidelines for the use and interpretation of assays
death: a novel therapeutic approach for restor- for monitoring autophagy (3rd edition). Autophagy.
ing photoreceptor homeostasis. Cell Death Dis. 2016;12(1):1–222.
2019;10(8):547. 17. Mizushima N. Methods for monitoring autophagy
4. Yao J, Qiu Y, Frontera E, Jia L, Khan NW, Klionsky using GFP-LC3 transgenic mice. Methods Enzymol.
DJ, et al. Inhibiting autophagy reduces retinal degen- 2009;452:13–23.
eration caused by protein misfolding. Autophagy. 18. Goodman CA, Hornberger TA. Measuring pro-
2018;14(7):1226–38. tein synthesis with SUnSET: a valid alternative
5. Dexter PM, Lobanova ES, Finkelstein S, Arshavsky to traditional techniques? Exerc Sport Sci Rev.
VY. Probing proteostatic stress in degenerating pho- 2013;41(2):107–15.
Absence of PRCD Leads
to Dysregulation in Lipid
Homeostasis Resulting
in Disorganization
of Photoreceptor Outer Segment
Structure

Sree I. Motipally and Saravanan Kolandaivelu

Abstract in the maintenance of photoreceptor OS struc-


tural and functional integrity.
The outer segments of photoreceptors are spe-
cialized sensory cilia crucial for light detec- Keywords
tion. Any disruption that alters outer segment
morphology can impair photoreceptor func- PRCD · Photoreceptor · Palmitoylation · Disc
tion and therefore vision. Progressive rod-­ membrane · ApoA1 · Lipid homeostasis ·
cone degeneration (PRCD) is an integral Phospholipids · Rhodopsin densities ·
membrane protein exclusively present in the Retinitis pigmentosa
photoreceptor OS with an unknown function.
Multiple mutations in PRCD are linked with
retinitis pigmentosa. The most common 1 Introduction
PRCD mutation observed in both human and
multiple dog breeds, PRCD-C2Y, lacks the Retinal photoreceptor (PR) neurons possess a
lipid modification “palmitoylation,” which is unique outer segment (OS), which is a modified
crucial for protein stability and trafficking to cilium containing over 1000 flattened and closely
the OS. Previous studies including ours show apposed double membranous structures called
impaired disc morphogenesis and rhodopsin discs. These discs contain many phototransduc-
distributions in the absence of PRCD, but the tion proteins including opsins crucial for convert-
precise role of PRCD in maintaining OS struc- ing light into electrical signals. On a daily basis,
ture and function remains unclear. In this one-tenth of these discs are phagocytosed at the
chapter, we discuss the potential role of PRCD apex by retinal pigment epithelium and perfectly
replaced by addition of new discs at the OS base
S. I. Motipally [23]. Disturbances to either processes can lead to
Departments of Neuroscience and Biochemistry,
Robert C. Byrd Health Sciences Centre, WVU Eye impaired visual function and PR degeneration [8,
Institute, Morgantown, WV, USA 10]. The maintenance of OS including disc for-
S. Kolandaivelu (*) mation and maturation are carried out by con-
Departments of Ophthalmology, Visual Sciences and certed actions of multiple proteins and lipids. A
Biochemistry, Robert C. Byrd Health Sciences proteomic study demonstrated that there are 11
Centre, WVU Eye Institute, Morgantown, WV, USA proteins that are found to be strongly associated
e-mail: kolandaivelus@hsc.wvu.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 389
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_57
390 S. I. Motipally and S. Kolandaivelu

with the disc membrane, including progressive affect the structural and functional properties of
rod-cone degeneration (PRCD) linked with the OS. Previous studies show that complete
retinitis pigmentosa (RP) [17]. PRCD is a
­ absence or defect in OS resident proteins can dis-
54-amino acid long protein that is highly concen- rupt OS integrity and ultimately lead to retinal
trated at the PR OS base and plays a critical role degeneration [11]. Early studies on canine
in OS disc morphogenesis [3, 16, 18]. Six muta- PRCD-C2Y models showed abnormal OS
tions in PRCD, including C2Y, R17C, R18X, renewal rates, lamellar disorientation, vesicular
R22X, P25T, and V30M, are associated with RP profiles, and reduced electroretinogram (ERG)
in humans. Our sequence-based prediction analy- responses [1, 2]. A recent study reported normal
sis suggested the presence of several structural visual responses in the absence of PRCD, sug-
domains that include transmembrane helices (aa gesting no changes in rhodopsin packaging den-
3–15), polybasic region (aa 16–18), and 2 poten- sities [18]. In contrast, we showed that
tial phosphorylation sites (aa 37 and 45 in PRCD-deficient retina show ~30% attenuation in
humans; 38 and 46 in mice). We have previously visual responses under bright light conditions.
shown that PRCD undergoes palmitoylation on More importantly, our atomic force microscopy
the sole cysteine residue (Cys2) and demon- studies demonstrate altered rhodopsin packaging
strated that this lipid modification is crucial for densities and distribution in the OS disc mem-
protein stability and OS trafficking [15]. The branes of PRCD-KO retina. Interestingly, previ-
non-palmitoylated PRCD-C2Y mutant protein is ous studies on rhodopsin overexpression and
highly unstable and mistargeted to the PR inner heterozygous rhodopsin mutants show normal
segment [15]. Previous studies in canine PRCD-­ rhodopsin packaging densities [13, 22].
C2Y models show ~35–45% reduction in disc Additionally, although the rhodopsin mRNA lev-
renewal rates; however, studies in PRCD-KO els did not differ between wild-type and PRCD
mice suggest attenuated rates of both disc renewal knockout animals at P30 and P120, we observed
and phagocytosis [1, 3]. Animal models with a 20% decrease in total rhodopsin protein levels
complete absence of PRCD in the OS and models at P120 [16]. Expression levels of rhodopsin are
expressing PRCD-C2Y mutant both show disor- previously shown to have a direct effect on OS
ganized OS and vesicular profiles [1, 16, 18]. length, disk diameters, and incisure depth [13,
Furthermore, previous studies also show that 14]. Taken together, these data suggest a potential
PRCD interacts with rhodopsin and this interac- role for PRCD in regulating opsin densities
tion is crucial for its stability as evidenced by its within the discs which thereby contributes to nor-
complete absence in rhodopsin knockout murine mal OS structure and function. Disorganization
models [19]. However, the precise function of of OS disc membranes and progressive loss of
PRCD’s interaction with rhodopsin and its role in retinal function in PRCD-deficient retinas dem-
the maintenance of PR OS structural and func- onstrate that PRCD is essential for maintaining
tional integrity remain obscure. In this chapter, normal OS structure and function. However, the
we demonstrate that absence of PRCD affects OS exact role of PRCD in OS structural maintenance
disc membrane lipid homeostasis, which likely and why PRs degenerate in the absence of PRCD
underlies the structural defects observed in OS. remains unknown.

2 PRCD Is Essential 3 Phenotypic Variations


for Photoreceptor Outer in Different PRCD Mutant
Segment Maintenance Animal Models

Structural integrity of PR OS is of prime impor- Complete PRCD knockouts from three indepen-
tance for efficient phototransduction that under- dent studies show disorganization of PR disc
lies normal vision. Most inherited retinal membranes and slow progressive degeneration
degenerations arise from molecular defects that [3, 16, 18]. However, some morphological and
Absence of PRCD Leads to Dysregulation in Lipid Homeostasis Resulting in Disorganization… 391

functional variations were observed across these previously shown to be critical for intraretinal
models, such as presence or absence of extracel- reverse transport of lipids and cholesterol homeo-
lular vesicles (EVs) in the interphotoreceptor stasis [20]. Additionally, we also observed accu-
matrix (IPM), attenuated or normal visual mulation of neutral lipids in the inner segments
responses with age, and different rates of PR of PRCD-C2Y retina at P200 which also sug-
degeneration suggesting a potential genetic het- gests dysregulated lipid metabolism (data not
erogeneity. In contrast to PRCD-KO mice, PRCD-­ shown). Based on this information, we hypothe-
C2Y knock-in mutant models do not exhibit sized that PRCD is crucial for maintaining PR
attenuated visual responses until after 6 months disc membrane lipid homeostasis and likely an
of age (data not shown). In addition, while the altered membrane lipid composition underlies
ultrastructure of PRCD-C2Y mutant retina shows the structural defects observed in PRCD mutant
presence of vesicles under PR OS base, however, animal models. To test this hypothesis, we inves-
in contrast to the observations made in PRCD KO tigated complete lipid profile in our PRCD-KO
mice models, the presence of EVs in the IPM was and PRCD-C2Y mice along with age-matched
sparse. Nevertheless, our unpublished data wild-type retina by proteomic analysis using
from murine PRCD-C2Y retina displayed lamel- mass spectrometry (LC-MS/MS). Various studies
lar disorientation and vesicular profiles, in con- suggest that dysregulation of lipid and choles-
sistence with canine C2Y models [1]. Although terol homeostasis is one of the mechanisms driv-
several studies in canine PRCD-C2Y and murine ing PR dysfunction in retinal degenerative
PRCD-KO models demonstrate the critical role diseases including RP [12, 21]. Lipidomic analy-
of PRCD in maintaining PR health, the precise sis of PRCD-C2Y retinas at P30 revealed
role of PRCD in supporting PR structure and decreased levels of phosphatidylethanolamine
function remains elusive. (PE), phosphatidylserine, phosphatidyl glycerol,
and phosphatidic acid (PA) when compared to
WT controls (Fig. 2a). Interestingly, only PE and
4 Retina Lacking PRCD Exhibit PA levels were reduced in PRCD-KO retinas.
Altered Membrane Lipid Although no significant changes in the levels of
Composition various phospholipids were observed at P120,
both PRCD-C2Y and PRCD-KO retinas show
Membrane phospholipid and cholesterol levels higher levels of docosahexaenoic acid (DHA)
are tightly regulated in the neural retina. The OS (Fig. 2b) (Motipally and Kolandaivelu in prep).
disc membranes are composed of phospholipids DHA is the most abundant polyunsaturated fatty
(90–95%), sphingolipids (1%), and cholesterol acid present in the OS that is crucial for visual
(4–6%) in certain proportions that provide the function and PR survival [4]. Elevated DHA in
structural and functional integrity needed for effi- PRCD-KO and C2Y mice at P120 is intriguing
cient phototransduction [9]. The accumulation of and warrants further investigation as previous
EVs and the presence of vesicular profiles in the studies in canine PRCD-C2Y models report
IPM of PRCD-KO and canine PRCD-C2Y reti- lower levels of plasma DHA when compared to
nas, respectively, suggest impaired membrane wild-type controls [5]. Furthermore, studies on
lipid homeostasis that can lead to disrupted OS rds/peripherin and P23H rhodopsin mutant mice
disc membrane integrity [1, 18] (Motipally and also show low DHA levels in rod OS [6, 7].
Kolandaivelu in prep). Interestingly, our co-­ Overall, our observations along with previous
immunoprecipitation followed by mass spec- findings suggest that PRCD deficiency alters the
trometry (Fig. 1a, Table 1) and cell culture studies OS disc lipid homeostasis, which likely underlies
using transiently expressed PRCD and apolipo- the observed defects in rhodopsin packaging den-
protein A1 (ApoA1) plasmids (Fig. 1b) shows sities and impaired OS morphology in PRCD-­
that PRCD binds with ApoA1. ApoA1 has been deficient retinas.
392 S. I. Motipally and S. Kolandaivelu

PRCD-IP IgG-IP

nd

nd
u

ou
bo

te

te
Un l
ta

b
kDa X X X

Elu
u

Un
To

El
15
PRCD

25 ApoA1
100 PDE6

50 β tubulin

WT-PRCD + WT-PRCD +
WT-PRCD Apo-A1 NMNAT1
T U E T U E T U E
29 kD
FLAG-IB
26kD

HA-IB 15 kD

Fig. 1 PRCD interacts with Apolipoprotein A1. (a) further analyzed in HEK-293 T cells. Transiently
Co-immunoprecipitation of PRCD and ApoA1 from P30 expressed PRCD (HA tagged) and ApoA1 (FLAG tagged)
wild-type retinal lysate is performed using affinity-­ were co-immunoprecipitated using HA beads and ana-
purified PRCD antibody coupled with protein A/G beads. lyzed by immunoblotting using antibodies as indicated.
Immunoblots were probed with antibodies as indicated We used lysates from HEK-293T cells co-transfected with
(lanes #1, 2, 4). Similarly, control IP is done using rabbit PRCD-WT alone and flag-tagged NMNAT1 (~29kD)
IgG as indicated (lanes #6, 8). As controls, we used PDE6 with PRCD-WT plasmid (n = 3) as controls. (T total, UB
and β-tubulin (n = 3). (b) PRCD-ApoA1 interaction was unbound, E elute)

Table 1 PRCD interacts with ApoA1 as identified by PRCD Co-IP followed by LC-MS/MS from retinal extracts
Protein ID Coverage Peptide PSM MW
Apolipoprotein-A1 25% 6 11 ~26 kD

5 Conclusions and Future lipid composition may contribute to the rhodop-


Directions sin packaging defects as well as EV accumula-
tion observed, ultimately resulting in disorganized
Several studies on PRCD-C2Y- and PRCD-­ PR OS structure and function. While significant
deficient models demonstrate impaired disc mor- progress has been made in our understanding
phogenesis and visual responses [1, 3, 16, 18]. about the importance of PRCD, there are many
We previously showed that PRCD plays a critical questions that remain to be addressed. Our future
role in regulating rhodopsin packaging densities focus includes the following: (1) investigating
within OS discs which is crucial for achieving why PRCD is exclusively present in the PR disc
normal OS structure and function [16]. The pres- membranes, (2) studying the importance of sev-
ent study shows that absence of PRCD in the PR eral predicted structural domains and their roles,
OS alters membrane lipid composition in both particularly mutations affecting palmitoyl lipid
PRCD-KO and PRCD-C2Y mice models. We modification and mutations in polybasic region
believe the observed alterations in membrane that are linked with RP, and (3) investigating how
Absence of PRCD Leads to Dysregulation in Lipid Homeostasis Resulting in Disorganization… 393

Fig. 2 Absence of PRCD in the photoreceptor OS alters ina. (b) No changes were observed in DHA levels at P30
membrane lipid composition. Retina samples from WT, (Bi), but significant increase in DHA levels is seen in
PRCD-KO, and PRCD-C2Y from postnatal ages 30 and PRCD mutant retinas at P120 (Bii). Groups were com-
120 were used to analyze complete lipid profile by LC-­ pared using a two-way ANOVA and a post hoc t-test. Data
MS/MS. (a) Relative levels of different phospholipids are presented as mean ± SD (n = 4; *p < 0.05, **p < 0.02;
(PS, PG, PE, and PA) are indicated in picomoles per ret- ***p < 0.001)

2. Aguirre G, O’Brien P. Morphological and biochemi-


do some PRs eventually achieve normal OS cal studies of canine progressive rod-cone degenera-
structure in the absence of PRCD. tion. 3H-Fucose autoradiography. Invest Ophthalmol
Vis Sci. 1986;27:635–55.
3. Allon G, Mann I, Remez L, et al. PRCD is concen-
trated at the base of photoreceptor outer segments and
References is involved in outer segment disc formation. Hum Mol
Genet. 2019;28:4078–88.
1. Aguirre G, Alligood J, O’Brien P, et al. Pathogenesis 4. Alvarez RA, Aguirre GD, Acland GM, et al.
of progressive rod-cone degeneration in miniature Docosapentaenoic acid is converted to docosahexae-
poodles. Invest Ophthalmol Vis Sci. 1982;23:610–30. noic acid in the retinas of normal and prcd-affected
394 S. I. Motipally and S. Kolandaivelu

miniature poodle dogs. Invest Ophthalmol Vis Sci. 15. Murphy J, Kolandaivelu S. Palmitoylation of pro-
1994;35:402–8. gressive rod-cone degeneration (PRCD) regulates
5. Anderson E, Aude M. Plasma lipid abnormalities protein stability and localization. J Biol Chem.
in the miniature poodle with progressive rod-cone 2016;291:23036–46.
degeneration. Exp Eye Res. 1991;52:349–55. 16. Sechrest ER, Murphy J, Senapati S, et al. Loss of
6. Anderson RE, Maude MB, Bok D. Low docosahexae- PRCD alters number and packaging density of rho-
noic acid levels in rod outer segment membranes of dopsin in rod photoreceptor disc membranes. Sci Rep.
mice with rds/peripherin and P216L peripherin muta- 2020;10:1–12.
tions. Invest Ophthalmol Vis Sci. 2001;42:1715–20. 17. Skiba NP, Spencer WJ, Salinas RY, et al. Proteomic
7. Anderson RE, Maude MB, McClellan M, et al. Low identification of unique photoreceptor disc com-
docosahexaenoic acid levels in rod outer segments of ponents reveals the presence of PRCD, a protein
rats with P23H and S334ter rhodopsin mutations. Mol linked to retinal degeneration. J Proteome Res.
Vis. 2002;8:351–8. 2013;12:3010–8.
8. Buyukmihci N, Aguirre G, Marshall J. Retinal degen- 18. Spencer WJ, et al. PRCD is essential for high-fidelity
erations in the dog II. Development of the retina in photoreceptor disc formation. Proc Natl Acad Sci.
rod-cone dysplasia. Exp Eye Res. 1980;30:575–91. 2019;96(116):13087–96.
9. Giusto NM, Pasquaré SJ, Salvador GA, et al. Lipid 19. Spencer WJ, Pearring JN, Salinas RY, et al.
second messengers and related enzymes in vertebrate Progressive rod-cone degeneration (PRCD) protein
rod outer segments. J Lipid Res. 2010;51:685–700. requires N-terminal S-acylation and rhodopsin bind-
10. Goldman AI, O’Brien PJ. Phagocytosis in the reti- ing for photoreceptor outer segment localization and
nal pigment epithelium of the RCS rat. Science. maintaining intracellular stability. HHS Public Access
1978;201:1023–5. Biochem. 2016;55:5028–37.
11. Haeri M, Knox BE. Rhodopsin mutant P23H destabi- 20. Tserentsoodol N, Gordiyenko NV, Pascual I, et al.
lizes rod photoreceptor disk membranes. PLoS One. Intraretinal lipid transport is dependent on high den-
2012;7(1):e30101. sity lipoprotein-like particles and class B scavenger
12. Hamano F, Kuribayashi H, Iwagawa T, et al. Mapping receptors. Mol Vis. 2006;12:1319–52.
membrane lipids in the developing and adult mouse 21. Van der Veen JN, Kennelly JP, Wan S, et al. The
retina under physiological and pathological con- critical role of phosphatidylcholine and phos-
ditions using mass spectrometry. J Biol Chem. phatidylethanolamine metabolism in health
2021;296:100303. and disease. Biochim Biophys Acta Biomembr.
13. Liang Y, Fotiadis D, Maeda T, et al. Rhodopsin sig- 2017;1859:1558–72.
naling and organization in heterozygote rhodopsin 22. Wen XH, Shen L, Brush RS, et al. Overexpression of
knockout mice. J Biol Chem. 2004;279:48189–96. rhodopsin alters the structure and photoresponse of
14. Makino CL, Wen XH, Michaud NA, et al. Rhodopsin rod photoreceptors. Biophys J. 2009;96:939–50.
expression level affects rod outer segment mor- 23. Young RW. The renewal of photoreceptor cell outer
phology and photoresponse kinetics. PLoS One. segments. J Cell Biol. 1967;33:61–72.
2012;7:e37832.
Expansion Microscopy of Mouse
Photoreceptor Cilia

Abigail R. Moyel, Michael A. Robichaux,


and Theodore Wensel

Abstract ing of ciliary markers, including tubulin,


CEP290, centrin, and CEP164. The results are
The small size of ciliary structures that under- consistent with those from other super-­
lies photoreceptor function and inherited cil- resolution fluorescence techniques and reveal
iopathies requires imaging techniques adapted new insights into their arrangements with
to visualizing them at the highest possible respect to the subcompartments of photore-
resolution. In addition to powerful super-­ ceptor cilia. This technique is complimentary
resolution imaging modalities, emerging to other imaging modalities used in retinal
approaches to sample preparation, including imaging, and can be carried out in virtually
expansion microscopy (ExM), can provide a any laboratory, without the need for expensive
robust route to imaging specific molecules at specialized equipment.
the nanoscale level in the retina. We describe a
protocol for applying ExM to whole retinas in Keywords
order to achieve nanoscale fluorescence imag-
Expansion microscopy · Connecting cilium ·
CEP290 · Photoreceptor · Super-resolution
A. R. Moyel
Verna and Marrs McLean Department of
Biochemistry and Molecular Biology, Baylor College
of Medicine, Houston, TX, USA
Department of Genetics and Ophthalmology, Institute Abbreviations
of Molecular and Clinical Ophthalmology Basel,
Basel, Switzerland BBS Bardet-Biedl syndrome
M. A. Robichaux BSA bovine serum albumin
Verna and Marrs McLean Department of CC connecting cilium
Biochemistry and Molecular Biology, Baylor College
of Medicine, Houston, TX, USA DAPI 4′,6-diamidino-2-phenylindole
ExM expansion microscopy
Department of Ophthalmology & Visual Sciences and
Department of Biochemistry & Molecular Medicine, IFT intraflagellar transport
West Virginia University, Morgantown, WV, USA NGS normal goat serum
T. Wensel (*) OCT optimal cutting temperature
Verna and Marrs McLean Department of PBS phosphate-buffered saline
Biochemistry and Molecular Biology, Baylor College PFA paraformaldehyde
of Medicine, Houston, TX, USA RT room temperature
e-mail: twensel@bcm.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 395
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_58
396 A. R. Moyel et al.

SDS sodium dodecyl sulfate adapted and optimized by many different labs
SIM structured illumination microscopy over the years, and used for the imaging of a
STORM stochastic optical reconstruction number of ciliary and centriolar structures in cel-
microscopy lular cultures [6–13], and very recently for the
TEMED tetramethylethylenediamine cilia in retinal tissue [14]. This technique is
adaptable to whichever tissue is of interest, and
the extent of expansion can be scaled [15]. The
1 Introduction materials and instruments used for this technique
are relatively accessible as compared to other
In recent years the structure of the mouse rod super-resolution methods.
photoreceptor cilium has been defined at an ever-­ Here we describe an expansion method opti-
increasing level of detail using electron micro- mized for visualizing photoreceptor cilia in
scopic and super-resolution fluorescence mouse retina. It can be used with many micros-
techniques. The tightly packed submicron struc- copy modalities. As an example, we have used
tures of rod photoreceptor sensory cilia require this modified ExM protocol to improve local-
optimized staining procedures and super-­ ization of CEP290 along the microtubules of
resolution for accurate localization analyses. 2D the connecting cilium (CC) axoneme, and have
stochastic optical reconstruction microscopy confirmed and improved localization detail for
(STORM) has been used to localize centrin to the ciliary proteins CEP164 and centrin (Figs. 1
lumen of the CC, surrounded by microtubules and 2).
and mobile components of the CC, like intrafla-
gellar transport (IFT) and BBSome proteins [1].
Comparative studies using super-resolution fluo- 2 Materials and Methods
rescence localization have demonstrated differ-
ences in subcellular localization of ciliary 2.1 Solutions Needed
proteins in retinal ciliopathy mutants, like
Spata7−/− and BBS protein subunit knockout Ames’ Medium [16] (Sigma A1420)
mouse mutants (Bbs2−/−, Bbs4−/−, and Bbs7−/−) Ames’ Block Buffer: 15% (v/v) normal goat serum
[1, 2]. A combination of both structured illumina- (NGS) (Fitzgerald 88R-NG001) + 5% (w/v)
tion microscopy (SIM) and STORM was recently Bovine serum albumin (BSA) (Sigma
used to localize CEP290 along the length of the A9647) + 0.5% (w/v) BSA-c (Aurion
CC, in comparison with other ciliary proteins like 25557) + 5% (w/v) Fish skin gelatin (Sigma
NPHP8 [3]. G7765) + 1x protease inhibitor cocktail
Expansion microscopy (ExM) [4, 5] is a tech- (GenDepot P3100) + 0.2% (w/v) saponin
nique that allows for fluorescence-based imag- (Calbiochem 558255); remaining volume is
ing, on standard microscopes, at nanoscale 1x Ames’ Medium
resolution. It can be combined with SIM, Ames’ Wash Buffer: 2% (v/v) NGS in 1x Ames’
STORM, and other nanoscopic techniques to Ames’ Secondary Buffer: 2% (v/v) NGS in 1x
potentially improve the spatial resolution achiev- Ames’ + 1x protease inhibitor cocktail + 0.2%
able beyond the capabilities of each technique (w/v) saponin
alone. ExM is based on the principle of embed- Phosphate buffered saline (PBS), 1x: 144 mg/L
ding cells and tissues in highly hygroscopic gels KH2PO4, 9000 mg/L NaCl, 421.62 mg/L
polymerized from mixtures of acrylamide and Na2HPO4, pH = 7.4
acrylate, which absorb water, allowing the gel to PBS Block Buffer: 15% NGS + 5% BSA + 0.5%
expand [5]. Proteins are cross-linked to the gel, BSA-c + 5% Fish skin gelatin + 0.2% sapo-
and thus structures containing proteins expand nin + 0.01% (w/v) Na-Azide in 1xPBS
with the gel (Fig. 1). The technique has been PBS Wash Buffer: 2% NGS in 1xPBS
Expansion Microscopy of Mouse Photoreceptor Cilia 397

Fig. 1 Sample preparation during expansion protocol. inches wide, ~0.17 inches long; immediately preceding
(a–d) Display examples of the retina in different steps of freezing, retinas ~0.13–0.17 inches wide, and ~0.23–0.27
the protocol, in order: (a) retina immediately after gela- inches long (~0.08 inch changes in each direction). (e)
tion; (b) retina post-disruption, trimmed and slightly SIM z-projection images displaying cilia, compared in
expanded; (c) retina after overnight incubation with pri- 512 × 512 pixel images, between an expanded section, a
mary antibodies (opacity subsides once they warm to RT); non-expanded control section, and a non-gelled control
and (d) orientation of the retinas for embedding in section stained for the antibodies indicated. Scale bar
OCT. Arrows point to retinas within the gels, and colored 5 μm. (f, g) Dot plots with averages and standard devia-
lines display length and width measurements to show that tions of the lengths and widths of staining in connecting
slight expansion of the retinas during disruption and sub- cilia pre- and post-expansion
sequent steps is, by eye, isotropic. Gelled retinas ~0.08

2.2 Gelation Solution 2.3 Method

2 M acrylamide (Fisher, BP1402-1), 300 ppm Unless otherwise noted, all incubation steps at
(ug/ml) N,N′-methylene-bis (acrylamide) RT and 4 °C were performed with the samples
(Sigma, 146072), 1.1 M Na-Acrylate (Santa rocking and covered. Note that there are three
Cruz Biotech. sc-236893), 1.5 ppt (mg/ml) sequential cycles of incubation with primary
ammonium persulfate and tetramethylethyl- antibodies, followed by secondary antibodies,
enediamine (TEMED) each, in 1xPBS before and after fixation, and before and after
Disruption Buffer: 50 mM Tris-Cl pH 7.5, 5% expansion, a procedure we have found to enhance
sodium dodecyl sulfate (SDS), 200 mM NaCl sensitivity of immunostaining.
398 A. R. Moyel et al.

Fig. 2 Localization of ciliary markers in connecting cil- tubulin, and CEP290 or (b) tubulin, centrin, and CEP164
ium following expansion in whole retina. (a, b) SIM with their corresponding controls. SIM z-projections with
z-projection images of single, straightened cilia stained 3D deconvolution are shown to the right of each panel. On
for either (a) acetylated alpha-tubulin, glutamylated-­ the axes, numbers less than 10 represent μm

Day 1 C-3302-1) filled with 1 mL of Ames Block,


blocked for 2 h. The tubes were positioned so as
We dissected the retinas into ice-cold Ames’ to allow the retinas to float around in the tube
media in 35 mm petri dishes, on ice, and then with gentle agitation. We added primary antibod-
transferred them via a wide-bore pipette tip into ies (5–10 μg each) directly to the block buffer and
hydrophobic microcentrifuge tubes (GeneMate, left the retinas to rock overnight.
Expansion Microscopy of Mouse Photoreceptor Cilia 399

Day 2 tured cells, but we found a full day of incuba-


tion to be best for overall incorporation of
We washed the retinas, gently, with Ames acryloyl-X into the entire retinal tissue.
Wash Buffer six times, 10 min each, on ice. After
the last wash, we added Secondary Buffer and
2°ary antibodies and left the retinas to incubate Day 7
for 2 h. We then washed the retinas with Ames
Wash six times, 5 min each, on ice. After rinsing Just before use, we prepared gelation solution
with 1xPBS to remove serum from the wash, we (1.5 mL per retina) and added 1 mL per retina to
fixed the retinas with 4% (w/v) PFA in 1xPBS, a cryomold (Ted Pella, 27147-2), on ice. After
for 20 min, and then quenched with 100 mM gly- rinsing retinas three times in PBS, we placed
cine HCl in PBS, for 30 min at RT. After one them in the center of the cryomold to incubate,
rinse in 1xPBS, we then blocked the retinas for shaking gently on ice, at 4 °C. We then added the
2 h in PBS Block Buffer at RT. Finally, we added remaining gelation solution to the cryomold
fresh primary antibodies directly to the tubes and before placing a glass coverslip on top (avoiding
stained for 2 full days. bubbles) and incubating for 2 h in a humidified
37 °C incubator to induce gelation. After cool-
Day 5 ing, we gently popped the gels out of the cryo-
molds using a pipette tip and trimmed excess gel
We washed the retinas in PBS Wash six times, from the retinas (Fig. 1a). We placed each gel
10 min each, on ice. We added a fresh 1 mL of PBS into a 15 mL conical tube with 10 mL of disrup-
Wash Buffer to each tube, with the corresponding tion buffer (covered with foil), and incubated in
2°ary antibodies, and incubated overnight. an 80 °C chamber for 3 h. We washed the gels in
water once and PBS several times to remove
Day 6 SDS (Fig. 1b), then transferred them to wells of
a new 24-well plate containing PBS + 0.1%
We washed the retinas in PBS Wash six times, Tween 20 (PBST). We washed the retinas in
5 min each, on ice. We then postfixed the retinas PBST two times, 30 min each, at RT, then added
with 3% PFA in PBS for 1 h at RT, followed by fresh primary antibodies, and incubated
quenching with 100 mM glycine HCl in PBS for overnight:
30 min at RT. After rinsing in PBS, we carefully
cut a small piece of each retina to serve as a non-­ –– Although previous expansion methods call for
gelled control. We quickly transferred these 3 h of gelation time, we found 2 h to be
pieces to tubes filled with 30% sucrose (made in sufficient.
1xPBS), added 1:1 30% sucrose-optimal cutting –– We found less isotropic expansion and
temperature (OCT, Tissue-Tek), and flash froze decreased antibody labeling when we used
them in N2(l). We transferred the rest of the retina digestion rather than denaturation.
to a 0.5 dram glass vial containing 1 mL of
0.1 mg/ml acryloyl-X SE (6-((acryloyl)amino)
hexanoic acid, succinimidyl ester, Thermo Day 8
A20770; for cross-linking proteins to hydrogel),
diluted in PBS, and left them to incubate on a We washed the retinas in PBST three times,
roller or rocker for 24 h at RT: 20 min each, at RT. We added fresh 1 mL of
PBST and 2°ary antibodies to the retinas, and
–– If there is no access to a roller, a rocker can be stained them for three and a half hours at
used as before. RT. DAPI was added at a 1:500 dilution for at
–– Regular eyecup cryosections can also be used least 1 h. After washing the retinas in PBST six
for the non-gelled control. times, 15 min each, at RT, we washed the retinas
–– Incubating for less time in Acryloyl-X SE has in 30% (w/v) sucrose (prepared in 1xPBS) three
been used in other protocols expanding cul- times, 15 min each. We then transferred the reti-
400 A. R. Moyel et al.

nas to 15 ml conical tubes filled with 10 mL of water. We used a fluorescent microscope to


30% sucrose. Once the retinas sank to the bottom choose the best sections for imaging. After care-
of the tube, we transferred them to a petri dish fully sealing the coverslip edges with scotch tape,
filled with OCT and swirled them to remove we marked the positions of the sections with a
excess sucrose from the gel block, and trimmed sharpie. To reduce drying we kept them at 4 °C,
the gels if possible. We then immediately trans- protected from light, until and imaged within the
ferred the retinas to a cryomold filled with fresh day, except for controls which we imaged later
OCT and froze them by placing the cryomolds on that week:
a flat surface in a −80 °C freezer:
–– The rest of the block can be stored in −80 °C,
–– If the retinas are left in 30% sucrose overnight, but fluorescence signal may decrease over
the structure of the gel changes slightly. time.
Fluorescence stays intact, but better to not –– We tested leaving the sections in water longer
leave it overnight. than 10 min, but the expansion factor did not
–– During mounting, the gel was pushed to the increase.
bottom of the mold for longitudinal sectioning –– Placing the section directly in the water is the
and the mold marked to indicate the gel best way to get isotropic expansion of the sec-
position. tion. Drying it on a coverslip first and then
expanding that in water did not produce reli-
able, consistent expansion.
2.4 Sectioning

Within 3 days after freezing, we cut sections. We 2.5 Animals


placed the cryomolds in a microtome cryocham-
ber set to −27–35 °C and left them to warm for Wild-type mice used for this study were C57BL/6
~30 min. In advance we filled multiple 50 mL aged 2–4 months. All procedures were approved
beakers with Milli-Q water, prepared coverslips by the Baylor College of Medicine Institutional
and slides by placing a ~50 ul drop of water on Animal Care and Use Committee and adhered to
each, and left a thin paintbrush out at RT. We then the Association of Research in Vision and
collected sections onto a Superfrost Plus slide Ophthalmology (ARVO) guidelines for the
(Fisher, 12-550-15) to check for fluorescence, humane treatment and ethical use of animals for
mounted in ProLong Glass mounting medium vision research.
(Thermo Fisher P36980), and used those as non-­
expanded controls. Once the area of interest was
reached, we cut 10 μm (or less) sections. We used 2.6 Antibodies
the RT brush to transfer the section to the glass
beaker by dipping the brush in water, lightly The following commercial antibodies were used:
touching it to the section, and transferring the CEP290 (C-terminus), Bethyl Laboratory, A301-­
section directly into the beaker filled with water. 659A; acetylated α-tubulin, Santa Cruz,
We left the sections to expand for 10 min, cov- sc-23950; Centrin, pan-centrin antibody from
ered. We used a loop tool (EMS, 70922-03, or a EMD Millipore, 04-1624, raised against
bigger loop can be made using a wooden stick Chlamydomonas centrin and reported to recog-
and silver wire) to transfer the expanded sections nize all mouse centrin isoforms 1-4 (41, 84);
from the beaker onto the drop of water on a #1.5 CEP164, Proteintech, 22227-1-AP; CEP164,
glass coverslip. After wicking away excess water Santa Cruz, sc-515403; MKS3 (cultured cells)
with a Kimwipe (Kimberly-Clark), we mounted Proteintech, 13975-1-AP; Atto488-Tuba1,
the coverslip on the drop of water on a glass slide. Antibodies-­online.com, ABIN1169085.
We again used a Kimwipe to wick away excess Antibodies were used at a concentration of 10 μg/
Expansion Microscopy of Mouse Photoreceptor Cilia 401

mL, with the exception of Atto488-Tuba1 which ROI for each row of pixels along the length of the
was used at a concentration of 7.5 μg/ ROI. Once converted to nm for accurate scaling,
mL. Secondary antibodies used were F(ab’)2-­ these profiles were used to identify the edges of
goat anti-rabbit IgG Alexa 488, F(ab’)2-goat antibody labeling (33% of the maximum inten-
anti-mouse IgG Alexa 555, and F(ab’)2-goat sity value). All measurements were made in a
anti-mouse IgG Alexa 647 (Thermo Fisher, 8 μg 1.5-μm-wide ROI. We achieved a ~ four-fold
each). expansion factor for length and width (Fig. 1f, g).
GraphPad Prism 7 was used to create the graphs.
The staining patterns for CEP290, centrin, and
3 Results CEP164 are all comparable to previously pub-
lished, non-expanded connecting cilia staining
We combined our whole retina immunostaining [3, 19, 20], but more detailed analysis is in
technique developed for STORM [1], with two progress.
expansion protocols: whole tissue ExM protocol
developed for expanding Drosophila synaptone-
mal complex during meiosis [17, 18] and further 4 Discussion
optimized with insights from an ExM method
termed TREx (tenfold robust expansion micros- We demonstrate in this report that ExM is a use-
copy) that was optimized for both cells and whole ful super-resolution fluorescence tool for localiz-
tissue [15]. We found that by eye the whole retina ing proteins in the rod photoreceptor cilium. In
expanded isotropically in the gels (Fig. 1). the future, the technique could be applied to
Although cryosections were fragile, the integrity localize other critical components of the cilium.
of the retinal tissues was maintained sufficiently Although ExM is useful for cilia cytoskeleton
to identify correctly localized fluorescently labeling with reasonably isotropic expansion,
labeled cilia (in reference to DAPI and non-­ there are some potential drawbacks. Some 2°ary
ciliary tubulin staining) for SIM. Though samples antibodies do not work well; as seen in Fig. 2,
were relatively lightly fixed (3% or 4% PFA for a CEP164 staining in the expanded cilia is less
total of 80 min), fixation could affect the sensitiv- well-maintained than in non-expanded retina, or
ity of some antibodies, which should be taken when compared to the staining under non-gelling
into account while optimizing this protocol for protocols [3], possibly due to problems with the
other antibodies/cells. Alexa 647-labeled secondary [21]. It remains to
With SIM, we collected z-stacks that revealed be determined which structures expand isotropi-
rod photoreceptor cilia immuno-labeled for post- cally. Membranes and protein structures must be
translationally modified tubulin (acetylated and disrupted in order to allow for complete gel
glutamylated), CEP290, CEP164, centrin, and embedding and subsequent expansion. The gel
tubulin that were significantly larger and more composition and protein disruption techniques
expanded than control, unexpanded cilia, as well used by different labs vary widely, depending on
as control non-gelled retina, imaged with the the structure and/or proteins of interest. Bioactive
same settings (Fig. 2a). Reconstructions were lipid membranes, such as the inner segment and
performed in Softworx 7 software, and 3D decon- CC plasma membrane, post-Golgi secretory ves-
volution of SIM reconstructed z-stacks was per- icles, and OS membrane discs, may be adversely
formed using Nikon NIS-Elements software. We affected by the high temperatures used in the pro-
performed the majority of the analysis thereafter tocol. It seems likely there is some anisotropy in
in FIJI/ImageJ. We calculated the expansion fac- the expansion, with some structures expanding
tor at the single cilium level by taking ROIs of more in one direction than in another, but further
individual cilia, digitally straightening them, and analysis and possibly further protocol modifica-
then taking the row-average intensities, which tions will be required. Improvement in further
plots the average intensity across the width of the spatial resolution will require either increasing
402 A. R. Moyel et al.

the expansion factor or by imaging with a more for fluorescence signal amplification. Mol Biol Cell.
2020;31(20):2195–206.
powerful super-resolution microscope like STED 9. Kong D, Loncarek J. Analyzing centrioles and
or STORM. Such changes will also require addi- cilia by expansion microscopy. Methods Mol Biol.
tional adaptations to the protocol. Nonetheless, 2021;2329:249–63.
even in its current state, ExM represents a power- 10. Sahabandu N, Kong D, Magidson V, Nanjundappa
R, Sullenberger C, Mahjoub MR, et al. Expansion
ful tool for localizing proteins within photorecep- microscopy for the analysis of centrioles and cilia. J
tor cilia. Microsc. 2019;276(3):145–59.
11. Gambarotto D, Hamel V, Guichard P. Ultrastructure
Acknowledgments This work was supported by NIH expansion microscopy (U-ExM). Methods Cell Biol.
grant R01-EY26545 (TGW), F32-EY027171 (MAR), 2021;161:57–81.
T32-EY007102 (ARM), and F32-EY031574 (ARM) and 12. Hamel V, Guichard P. Improving the resolution of flu-
a grant from the Knights Templar Eye Foundation (MAR). orescence nanoscopy using post-expansion labeling
3D deconvolution analysis was performed in the West microscopy. Methods Cell Biol. 2021;161:297–315.
Virginia University Microscope Imaging Facility, which 13. Zwettler FU, Reinhard S, Gambarotto D, Bell TDM,
has been supported by the WVU Cancer Institute and NIH Hamel V, Guichard P, et al. Molecular resolution
grants P20RR016440, P30GM103488, and imaging by post-labeling expansion single-molecule
U54GM104942. localization microscopy (Ex-SMLM). Nat Commun.
2020;11(1):3388.
Conflict of Interest The authors have declared that no 14. Mercey O, Kostic C, Bertiaux E, Giroud A, Sadian Y,
conflicts of interest exist. Chang N, et al. The connecting cilium inner scaffold
provides a structural foundation to maintain photore-
ceptor integrity. Plos Bio. 2022;20(6): e3001649.
15. Damstra HGJ, Mohar B, Eddison M, Akhmanova
References A, Kapitein LC, Tillberg PW. Visualizing cel-
lular and tissue ultrastructure using Ten-fold
1. Robichaux MA, Potter VL, Zhang Z, He F, Liu J, Robust Expansion Microscopy (TREx). bioRxiv.
Schmid MF, et al. Defining the layers of a sensory cil- 2021:2021.02.03.428837.
ium with STORM and cryoelectron nanoscopy. Proc 16. Ames A 3rd, Nesbett FB. In vitro retina as an
Natl Acad Sci U S A. 2019;116(47):23562–72. experimental model of the central nervous system. J
2. Dharmat R, Eblimit A, Robichaux MA, Zhang Z, Neurochem. 1981;37(4):867–77.
Nguyen TT, Jung SY, et al. SPATA7 maintains a novel 17. Cahoon CK, Yu Z, Wang Y, Guo F, Unruh JR,
photoreceptor-specific zone in the distal connecting Slaughter BD, et al. Superresolution expansion
cilium. J Cell Biol. 2018;217(8):2851–65. microscopy reveals the three-dimensional organiza-
3. Potter VL, Moye AR, Robichaux MA, Wensel tion of the Drosophila synaptonemal complex. Proc
TG. Super-resolution microscopy reveals Natl Acad Sci U S A. 2017;114(33):E6857–E66.
photoreceptor-­specific subciliary location and func- 18. Wang Y, Yu Z, Cahoon CK, Parmely T, Thomas
tion of ciliopathy-associated protein CEP290. JCI N, Unruh JR, et al. Combined expansion micros-
Insight. 2021;6(20):e145256. copy with structured illumination microscopy
4. Gallagher BR, Zhao Y. Expansion microscopy: a for analyzing protein complexes. Nat Protoc.
powerful nanoscale imaging tool for neuroscientists. 2018;13(8):1869–95.
Neurobiol Dis. 2021;154:105362. 19. Yang TT, Chong WM, Wang WJ, Mazo G, Tanos
5. Chen F, Tillberg PW, Boyden ES. Optical imaging. B, Chen Z, et al. Super-resolution architecture of
Expansion microscopy. Science. 2015;347(6221):543–8. mammalian centriole distal appendages reveals dis-
6. Gambarotto D, Zwettler FU, Le Guennec M, Schmidt-­ tinct blade and matrix functional components. Nat
Cernohorska M, Fortun D, Borgers S, et al. Imaging Commun. 2018;9(1):2023.
cellular ultrastructures using expansion microscopy 20. Shi X, Garcia G 3rd, Van De Weghe JC, McGorty
(U-ExM). Nat Methods. 2019;16(1):71–4. R, Pazour GJ, Doherty D, et al. Erratum: super-­
7. Le Guennec M, Klena N, Gambarotto D, Laporte resolution microscopy reveals that disruption of
MH, Tassin AM, van den Hoek H, et al. A helical ciliary transition-­zone architecture causes Joubert
inner scaffold provides a structural basis for centriole syndrome. Nat Cell Biol. 2017;19(11):1379.
cohesion. Sci Adv. 2020;6(7):eaaz4137. 21. Tillberg PW, Chen F, Piatkevich KD, Zhao Y, Yu
8. Katoh Y, Chiba S, Nakayama K. Practical method for CC, English BP, et al. Protein-retention expansion
superresolution imaging of primary cilia and centri- microscopy of cells and tissues labeled using standard
oles by expansion microscopy using an amplibody fluorescent proteins and antibodies. Nat Biotechnol.
2016;34(9):987–92.
Rod Photoreceptor-Specific
Ablation of Metformin Target,
AMPK, in a Preclinical Model
of Autosomal Recessive Retinitis
Pigmentosa

Nicholas D. Nolan, Laura A. Jenny,


Stephen H. Tsang, and Xuan Cui

Abstract tein kinase (AMPK)-associated metabolic


pathway reprogramming using a gene-­
Retinal gene therapies have shown tremendous independent model of degeneration and
progress in the past decade, but the sheer rescue. We show that recue of photoreceptor
number of disease-causing mutations makes structure and function is not achieved through
their applicability challenging. In this study our model of metabolic reprogramming.
we test our hypothesis that retinitis These results suggest that RP may not be
pigmentosa-­ associated retinal degeneration treatable through AMPK pathway modulation-­
can be prevented through AMP-activated pro- based therapies.

Ophthalmology, Columbia University,


N. D. Nolan
New York, NY, USA
Department of Biomedical Engineering, Columbia
e-mail: xc2342@cumc.columbia.edu
University, New York, NY, USA
S. H. Tsang
Edward S. Harkness Eye Institute, Department of
Department of Biomedical Engineering, Columbia
Ophthalmology, Columbia University Irving Medical
University, New York, NY, USA
Center/New York-Presbyterian Hospital,
New York, NY, USA Edward S. Harkness Eye Institute, Department of
Ophthalmology, Columbia University Irving Medical
Jonas Children’s Vision Care, and Bernard & Shirlee
Center/New York-Presbyterian Hospital,
Brown Glaucoma Laboratory, Department of
New York, NY, USA
Ophthalmology, Columbia University,
New York, NY, USA Jonas Children’s Vision Care, and Bernard & Shirlee
Brown Glaucoma Laboratory, Department of
Columbia Stem Cell Initiative, Columbia University,
Ophthalmology, Columbia University,
New York, NY, USA
New York, NY, USA
L. A. Jenny · X. Cui (*)
Columbia Stem Cell Initiative, Columbia University,
Edward S. Harkness Eye Institute, Department of
New York, NY, USA
Ophthalmology, Columbia University Irving Medical
Center/New York-Presbyterian Hospital, Department of Pathology & Cell Biology, Columbia
New York, NY, USA University, New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee Institute of Human Nutrition, Columbia University,
Brown Glaucoma Laboratory, Department of New York, NY, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 403
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_59
404 N. D. Nolan et al.

Keywords ise in recent years, and is capable of treating


patients on the magnitude necessary to signifi-
Retinitis pigmentosa · Retinal degenerations · cantly reduce disease burden [9–11]. In addition,
Gene therapy · Metabolomics · Late-stage there is a growing body of evidence that supports
treatment the notion of metabolic ties between the retinal
pigment epithelium and photoreceptors, the
breakdown of which occurs in many retinal dys-
1 Introduction trophies [12]. Targeting this breakdown of cou-
pling is a gene-independent strategy and remains
Retinitis pigmentosa (RP) is the third most to be explored herein.
common inheritable cause of vision loss behind In this study, we used a mutant
age-­related macular degeneration and glaucoma, phosphodiesterase 6b (PDE6B) mouse model
and reports have estimated its prevalence from with rod-­ specific excision of AMP-activated
between 1 in 750 and 1 in 9000 in various geo- protein kinase (AMPK). Cellular energy
graphic locations [1–4]. In addition to RP, there homeostasis is a dynamic complex process.
are many related inherited retinal dystrophies, During cell states of low energy, AMPK plays a
caused by a variety of underlying mutations that crucial role phosphorylating specific enzymes
have similar clinical presentations and perturbed which leads to an increased production of ATP
metabolic pathways [5]. Retinitis pigmentosa through downstream targets of fatty acid oxidation
typically begins as a rod photoreceptor degenera- and protein synthesis, and is typically activated
tion, with patients experiencing mild to more following elevated mitochondrial stress [13, 14].
severe night blindness as a result. If left untreated, Other bodies of work have shown a delicate
this leads to tunnel vision in mid-stage RP and interplay between the AMPK and mTOR
eventually progresses to cone loss in the macula, pathways that regulate cell survival and metabolic
or loss of photopic vision altogether [6]. pathways such as mitochondrial homeostasis,
In recent years, gene therapy has emerged as a protein synthesis, and autophagy [15]. Previous
promising therapeutic strategy applied to oph- researchers have attempted to modulate this
thalmology. By directly editing the causative pathway, using the drug metformin to activate
underlying mutation, medical researchers aim to AMPK as a treatment for type 2 diabetes [16].
correct the root of pathology. Over three decades Contrarily, we hypothesize that AMPK ablation
ago, researchers identified the first mutation asso- could shift some of these metabolic pathways and
ciated with autosomal dominant RP, and as of subsequently improve photoreceptor health and
now more than 80 genes have been identified to function. Using this gene-independent model of
be associated with non-syndromic RP [7, 8]. The retinal degeneration rescue, we investigated
large number of identified causative mutations in whether photoreceptor degeneration could be
RP combined with the amount of time and money halted independently of underlying, disease-
it costs to design a therapy for each mutation in causing mutations. Our results reveal that in this
each gene independently makes gene therapies specific mouse model of RP, AMPK ablation does
specific to each mutation challenging. In addi- not significantly mediate gene-independent
tion, most patients are not usually diagnosed until therapeutic restoration of photoreceptor health
far after the onset of disease and degeneration. and function.
Given this, a therapeutic aimed at treating RP
must remain efficacious even in mid- to late-­
stage disease. Strategies targeting specific steps
in metabolic pathways which cause disease have
been explored. Reprogramming of the mamma-
lian target of rapamycin (mTOR)- and sirtuin 6
(SIRT6)-related pathways has shown great prom-
Rod Photoreceptor-Specific Ablation of Metformin Target, AMPK, in a Preclinical Model of Autosomal… 405

2 Materials and Methods analysis of the photoreceptor outer nuclear layer


(ONL) thickness was performed on sections con-
2.1 Animals taining the thickest cross section of the optic
nerve, corresponding to the most central plane of
Our experimental procedures were approved by the eye (Fig. 1a). Distance from the optic nerve
the Institutional Animal Care and Use Committee for all points can be seen in Fig. 1b.
(IACUC) at Columbia University in New York.
All rodents were used in accordance to the
Statement for the Use of Animals in Ophthalmic 2.4 Electroretinography (ERG)
and Vision Research of the Association for
Research in Vision and Ophthalmology and the ERG testing was performed at 6 weeks of age, at
Policy for the Use of Animals in Neuroscience the representative mid-stage of disease progres-
Research of the Society for Neuroscience. sion in this particular mouse model according to
Pde6bH620Q mice were derived via oviduct trans- procedures described [22].
fer as described previously [17, 18]. AMPKfl/fl
mice were ordered from the Jackson Laboratory
#014142. These mice have loxP sites flanking 3 Results
exon 2 of the protein kinase, AMP-activated,
alpha 2 catalytic subunit (Prkaa2) gene. This 3.1 Lack of Functional
exon is deleted following tamoxifen-induced and Structural Rescue
Cre-recombination, crippling the function of in Pde6bH620Q; Pde6gCreERT2;
AMPK in a cell type-specific manner. Pde6gCreERT2 and AMPKfl/fl Mice
were generated as previously described [19–21].
All animals were maintained in the Columbia In order to investigate whether or not AMPK-­
University Eye Institute Annex Animal Care mediated metabolic reprogramming would treat
Services Facility in a standard 12-h light-dark RP independently of the underlying disease-­
cycle. causing mutation, we delivered treatment to our
specific RP mouse model Pde6bH620Q/H620Q;
Pde6gCreERT2/+; and AMPKfl/fl mice. After intraper-
2.2 Tamoxifen Injection itoneal injection of tamoxifen, Pde6gCreERT2/+
becomes activated and halts the AMPK gene
Tamoxifen powder (Sigma-Aldrich T5648) was expression and downstream protein production
diluted in 100% ethanol to a concentration of through a frame shift-mediated premature stop
100 mg/ml at 42 °C. This mixture was further codon introduction. Our group administered
diluted with sunflower oil to a final concentration of tamoxifen treatment to P9-P11 Pde6bH620Q/H620Q;
10 mg/ml and injected at 100 μg/g body weight on Pde6gCreERT2/+; and AMPKfl/fl mice. At 6 weeks of
three consecutive days, starting on postnatal day 9. age, retinal health has severely declined in this
mouse model (Fig. 1). ONL thickness magni-
tudes are seen in Fig. 1b. Photoreceptor ONL
2.3 Immunohistochemistry thickness is reduced dramatically from WT mice
and Morphometry [20]. To assess potential structural rescue from
our treatment, sectioned mouse eyes were subject
Mice were euthanized according to IACUC to morphometry and ONL thickness character-
guidelines. Mouse eyes were enucleated, fixed in ization as described in the methods section of the
paraformaldehyde, and sectioned across the sag- chapter (Fig. 1a). Based on our data, AMPK abla-
ittal plane as described [21]. Sectioned eyes were tion treatment did not significantly reduce ONL
stained with H&E to visualize the photoreceptor degeneration rates (Fig. 1b). In both treated and
nuclear layer. Quantification and subsequent untreated Pde6bH620Q/H620Q; Pde6gCreERT2/+; and
406 N. D. Nolan et al.

Fig. 1 Histological sections and visualization of the outer thickness averages mapped against the distance from
nuclear layer (ONL) in Pde6bH620Q/H620Q; Pde6gCreERT2/+; optic nerve (all measurements in micrometers). nAMPKfl/
AMPKfl/fl eyes. (a) Representative sections stained with fl
= 4, nAMPK−/− = 6
H&E for ease of ONL visualization. (b) Quantified ONL

AMPKfl/fl retinas, ONL thickness was markedly 4 Discussion


decreased from WT.
In addition to the structural characterization, In this study we used a Pde6bH620Q/H620Q;
treated and untreated mice were subject to func- Pde6gCreERT2/+; AMPKfl/fl mouse model which
tional assays aimed at assessing rod and cone allowed the deletion of AMPK uniformly from
function. The standard in ophthalmology for rods in the retina, circumventing common prob-
assessing overall photoreceptor function of ERG lems associated with subretinal injections, and
was implemented to test 6-week-old mice in this simplifying analysis. Administration of tamoxifen
study. We utilized dark-adapted conditions to at P9 allows us to attempt to prevent AMPK path-
stimulate a purely scotopic response, dark-­ way perturbation from an underlying mutation
adapted bright light conditions to stimulate a before the late-stage degeneration occurs. This
mixed rod and cone response, and light-adapted therapy was given early on in degeneration pro-
conditions to stimulate a purely photopic gression in line with many other animal gene
response. Both treated and untreated mice therapy studies. The data generated within this
­exhibited marked degenerations in both A- and study indicates the lack of significant rescue. This
B-wave responses and show little rescue com- could be due to a compensatory adjustment from
pared to the degenerative control (DC) mice AMPK-related pathways which are also dysregu-
(Fig. 2a). When quantified, magnitudes of A- and lated in many underlying RP mutations. Several
B-wave responses showed no statistically signifi- recent gene therapy clinical trials have demon-
cant difference (Fig. 2b). Treatment at P9 failed strated the ability to improve retinal function and
to rescue photoreceptor-associated electrophysi- structural health but lack the ability to prevent fur-
ological functionality. ther photoreceptor degeneration [9–11, 23–25]. A
therapy aimed at correcting downstream pathway
modulation and degeneration should be investi-
gated in future studies. Our study indicates that
other dysregulated metabolic pathways and their
modulation should be studied in the context of
Rod Photoreceptor-Specific Ablation of Metformin Target, AMPK, in a Preclinical Model of Autosomal… 407

B Scotopic Rod b-wave Scotopic Max b-wave Photopic Cone a-wave Photopic Cone b-wave
50 300 200

40
150
200
Amplitude(mV)

Amplitude(mV)

Amplitude(mV)

Amplitude(mV)
30

100
–10
20
100
50
10

0 0 –20 0
DC Ampkfl/fl Ampk-/- DC Ampkfl/fl Ampk-/- DC Ampkfl/fl Ampk-/- DC Ampkfl/fl Ampk-/-

Fig. 2 Electroretinogram (ERG) photoreceptor functional plotted for comparison. Results indicate no statistically
assay. (a) Average trace recordings of rod (left), rod and significant difference between groups. nDC = 6, nAMPKfl/
cone mixed (middle), and cone (right) photoreceptors in fl
= 2, nAMPK−/− = 5. Error bar is standard error of the mean
Pde6bH620Q/H620Q; Pde6gCreERT2/+; AMPKfl/fl; and Pde6bH620Q/
H620Q
(DC) mice. (b) Quantified A- and B-wave magnitudes

New York Regional Research Center Grant [TA-NMT-


established RP models to assess and develop 0116-0692-COLU], Lynette & Richard Jaffe, the Jaffe
potential therapeutics able to overcome current Family Foundation, Nancy and Kobi Karp, the Crowley
hurdles in the field. Family Funds, the Rosenbaum Family Foundation, Alcon
Research Institute, the Gebroe Family Foundation, the
Research to Prevent Blindness (RPB) Physician-Scientist
Acknowledgments We thank Sarah R. Levi and Jonas Award, unrestricted funds from RPB, New York,
Children’s Vision Care (JCVC) for sharing ideas, and for NY, USA.
critically reading the manuscript; JCVC is supported by
the National Institute of Health 5P30CA013696, Conflict of Interest Statement Stephen H. Tsang has
U01EY030580, U01EY034590, U54OD020351, received support from Emendo. He is also the founder of
R24EY028758, R24EY027285, 5P30EY019007, Rejuvitas and is on the scientific and clinical advisory
R01EY018213, R01EY024698, R01EY026682, board for Nanoscope Therapeutics and Medical
R01EY033770, R21AG050437, the Schneeweiss Stem Excellence Capital.
Cell Fund, New York State [SDHDOH01-
C32590GG-3450000], the Foundation Fighting Blindness
408 N. D. Nolan et al.

14. Xu L, Ash JD. The role of AMPK pathway


References in neuroprotection. Adv Exp Med Biol.
2016;854:425–30.
1. Haim M. Epidemiology of retinitis pigmentosa 15. Garza-Lombó C, Schroder A, Reyes-Reyes EM,
in Denmark. Acta Ophthalmol Scand Suppl. Franco R. mTOR/AMPK signaling in the brain: cell
2002;233:1–34. metabolism, proteostasis and survival. Curr Opin
2. Pagon RA. Retinitis pigmentosa. Surv Ophthalmol. Toxicol. 2018;8:102–10.
1988;33(3):137–77. 16. Hawley SA, Gadalla AE, Olsen GS, Hardie DG. The
3. Na KH, Kim HJ, Kim KH, Han S, Kim P, Hann HJ, antidiabetic drug metformin activates the AMP-­
et al. Prevalence, age at diagnosis, mortality, and activated protein kinase cascade via an adenine
cause of death in retinitis pigmentosa in Korea – A nucleotide-independent mechanism. Diabetes.
nationwide population-based study. Am J Ophthalmol. 2002;51(8):2420–5.
2017;176:157–65. 17. Hart AW, McKie L, Morgan JE, Gautier P, West K,
4. Nangia V, Jonas JB, Khare A, Sinha A. Prevalence of Jackson IJ, et al. Genotype–phenotype correlation of
retinitis pigmentosa in India: the Central India eye and mouse Pde6b mutations. Invest Ophthalmol Vis Sci.
medical study. Acta Ophthalmol. 2012;90(8):e649–50. 2005;46(9):3443–50.
5. Verbakel SK, van Huet RAC, Boon CJF, den 18. Davis RJ, Tosi J, Janisch KM, Kasanuki JM,
Hollander AI, Collin RWJ, Klaver CCW, et al. Non-­ Wang N-K, Kong J, et al. Functional rescue of
syndromic retinitis pigmentosa. Prog Retin Eye Res. degenerating photoreceptors in mice homozygous
2018;66:157–86. for a hypomorphic cGMP phosphodiesterase 6 b
6. Sahel J, Bonnel S, Mrejen S, Paques M. Retinitis allele (Pde6b H620Q). Invest Ophthalmol Vis Sci.
pigmentosa and other dystrophies. Dev Ophthalmol. 2008;49(11):5067–76.
2010;47:160–7. 19. Davis RJ, Hsu C-W, Tsai Y-T, Wert KJ, Sancho-Pelluz
7. Dryja TP, McGee TL, Reichel E, Hahn LB, Cowley J, Lin C-S, et al. Therapeutic margins in a novel
GS, Yandell DW, et al. A point mutation of the preclinical model of retinitis pigmentosa. J Neurosci.
rhodopsin gene in one form of retinitis pigmentosa. 2013;33(33):13475–83.
Nature. 1990;343(6256):364–6. 20. Koch SF, Tsang SH. Success of gene therapy in late-­
8. Daiger SP, Sullivan LS, Bowne SJ, Rossiter stage treatment. Adv Exp Med Biol. 2018;1074:101–7.
BJF. RetNet: retinal information network. Na+ Ca2. 21. Koch SF, Duong JK, Hsu C-W, Tsai Y-T, Lin C-S,
Houston: The University of Texas Health Science Wahl-Schott CA, et al. Genetic rescue models refute
Center; 2013. p. 5. nonautonomous rod cell death in retinitis pigmentosa.
9. Venkatesh A, Ma S, Le YZ, Hall MN, Rüegg MA, Proc Natl Acad Sci. 2017;114(20):5259–64.
Punzo C. Activated mTORC1 promotes long-term 22. Koch SF, Tsai Y-T, Duong JK, Wu W-H, Hsu C-W,
cone survival in retinitis pigmentosa mice. J Clin Wu W-P, et al. Halting progressive neurodegeneration
Invest. 2015;125(4):1446–58. in advanced retinitis pigmentosa. J Clin Invest.
10. Zhang L, Du J, Justus S, Hsu C-W, Bonet-Ponce 2015;125(9):3704–13.
L, Wu W-H, et al. Reprogramming metabolism by 23. Cideciyan AV, Jacobson SG, Beltran WA, Sumaroka
targeting sirtuin 6 attenuates retinal degeneration. J A, Swider M, Iwabe S, et al. Human retinal
Clin Invest. 2016;126(12):4659–73. gene therapy for Leber congenital amaurosis
11. Zhang L, Justus S, Xu Y, Pluchenik T, Hsu C-W, Yang shows advancing retinal degeneration despite
J, et al. Reprogramming towards anabolism impedes enduring visual improvement. Proc Natl Acad Sci.
degeneration in a preclinical model of retinitis 2013;110(6):E517–E25.
pigmentosa. Hum Mol Genet. 2016;25(19):4244–55. 24. Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal
12. Brown EE, DeWeerd AJ, Ildefonso CJ, Lewin AS, S, Schwartz SB, Boye SL, et al. Human RPE65 gene
Ash JD. Mitochondrial oxidative stress in the retinal therapy for Leber congenital amaurosis: persistence
pigment epithelium (RPE) led to metabolic dysfunction of early visual improvements and safety at 1 year.
in both the RPE and retinal photoreceptors. Redox Hum Gene Ther. 2009;20(9):999–1004.
Biol. 2019;24:101201. 25. Bainbridge JWB, Smith AJ, Barker SS, Robbie S,
13. Herzig S, Shaw RJ. AMPK: guardian of metabolism Henderson R, Balaggan K, et al. Effect of gene therapy
and mitochondrial homeostasis. Nat Rev Mol Cell on visual function in Leber’s congenital amaurosis. N
Biol. 2018;19(2):121–35. Engl J Med. 2008;358(21):2231–9.
TLR2 Is Highly Overexpressed
in Retinal Myeloid Cells in the rd10
Mouse Model of Retinitis
Pigmentosa

Alonso Sánchez-Cruz, Enrique J. de la Rosa,


and Catalina Hernández-Sánchez

Abstract expression, which was restricted to myeloid


cells, was increased in rd10 retinal microglia.
Retinitis pigmentosa (RP) is a genetically het- These observations, together with our previ-
erogeneous disease and the predominant cause ous finding of delayed RP progression follow-
of hereditary blindness. Irrespective of the ing Tlr2 deletion, point to TLR2 as a potential
causative mutation, traits common to all forms therapeutic target for RP.
of RP include photoreceptor dysfunction and
death, activation of the retinal glial compo- Keywords
nent, and retinal inflammation. Activation of
Retinitis pigmentosa · TLR · TLR2 ·
Toll-like receptors (TLRs) in response to tis-
Microglia · Inflammation · Innate immunity ·
sue damage is associated with inflammatory
Myeloid cells · Retina · Neurodegeneration
processes that contribute to neurodegenera-
tion. We show that retinal expression of the
genes Tlr1 to Tlr9 is increased in the rd10
mouse model of RP, with Tlr2 showing the 1 Introduction
greatest increase (36-fold). Flow cytometry
analysis of the retinal myeloid population Retinitis pigmentosa (RP) is a group of retinal
revealed significant increases in numbers of genetic dystrophies responsible for the most
microglia and infiltrating monocytes and mac- prevalent form of hereditary blindness, and
rophages in rd10 retinas. Furthermore, TLR2 affects around 2 million patients worldwide. RP
is caused by more than 3000 different mutations
in over 60 genes. Despite its genetic heterogene-
A. Sánchez-Cruz · E. J. de la Rosa ity, traits common to all forms of RP include pri-
Department of Molecular Biomedicine, Centro de mary dysfunction and death of photoreceptors,
Investigaciones Biológicas Margarita Salas (CSIC),
Madrid, Spain which triggers a rapid response from retinal
microglial and macroglial cells. The persistent
C. Hernández-Sánchez (*)
Department of Molecular Biomedicine, Centro de presence of the insult caused by the mutation
Investigaciones Biológicas Margarita Salas (CSIC), leads to non-resolving gliosis and sterile inflam-
Madrid, Spain mation that further contributes to disease pro-
Centro de Investigación Biomédica en Red de gression [1]. It has been proposed that targeting
Diabetes y Enfermedades Metabólicas Asociadas of common alterations, such as the altered glial
(CIBERDEM), ISCIII, Madrid, Spain response, may constitute a feasible strategy to
e-mail: chernandez@cib.csic.es

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 409
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_60
410 A. Sánchez-Cruz et al.

treat the wide variety of RP types, regardless of 2.2 RNA Isolation


the causative mutation [2]. and Quantitative PCR
Toll-like receptors (TLRs) are initiators of
the innate immune response that recognize RNA isolation and qPCR were carried out as pre-
both pathogen-associated molecular patterns viously reported [5]. TaqMan assays
and damage-associated molecular patterns (ThermoFisher) are included in Table 1.
(PAMPs and DAMPs, respectively). DAMPs
are cellular molecules that are released or
increased in response to tissue damage. The 2.3 Flow Cytometry
activation of TLRs by DAMPs has prompted
investigation of the role of the former in dis- Myeloid cell numbers and TLR2 levels in WT and
eases with an inflammatory component, includ- rd10 retinas were assessed as previously described
ing neurodegenerative conditions. A growing [4]. Staining was carried out with the following
body of evidence implicates TLRs in brain antibodies: CD45-eFluor450 (30-F11, BioLegend);
neurodegenerative disease [3]. However, little CD11b-PerCPCy5.5 (M170, BioLegend); F4/80-
is known about the role of TLRs in retinal dys- APC (BM8, eBioscience), and TLR2-FITC (6C2,
trophies, particularly RP. We recently demon- eBioscience). Samples without the TLR2-FITC
strated that genetic deletion of Tlr2 preserves antibody were used as a negative control.
photoreceptors and alleviates vision loss in two
mouse models of RP [4], pointing to TLR2 as a
candidate therapeutic target for RP. Here, we 2.4 Statistical Analysis
contextualize the increased expression of Tlr2
in the innate immune response to RP and char- Statistical analyses were performed with
acterize the retinal cellular component involved GraphPad Prism 8.0. For normally distributed
in this increase. data, an unpaired t-test was applied. Analyses of
more than two data sets were performed using a
one-way ANOVA with a Tukey’s post-test.
2 Materials and Methods Comparisons of two variables were performed
using a two-way ANOVA, followed by Sidak’s
2.1 Animals multiple comparison test. Statistical significance
was established at p < 0.05.
The rd10 mouse model of RP carries a homozy-
gous phosphodiesterase 6b mutation (Pde6brd10/
rd10
) on a C57BL/6 J background. rd10 and wild-­ 3 Results
type (WT) mice were obtained from the Jackson
Laboratory. All animals were housed and handled 3.1 Expression of Tlr Genes in WT
at the CIB Margarita Salas [4] in accordance with and rd10 Retinas
the ARVO statements and the guidelines of the
European Union and the local ethics committees First, in both WT and rd10 mouse retinas, we
of the CSIC and Madrid (PROEX 152/16, 287/19 examined the expression of Tlr genes that are
and 266.1/21). conserved between humans and mice (Tlr1 to

Table 1 TaqMan assays for qPCR


Gene TaqMan assay Gene TaqMan assay Gene TaqMan assay
Il1b Mm00434228_m1 Tlr3 Mm01207404_m1 Tlr7 Mm00446590_m1
Tbp Mm00446971_m1 Tlr4 Mm00445273_m1 Tlr8 Mm04209873_m1
Tlr1 Mm00446095_m1 Tlr5 Mm00546288_s1 Tlr9 Mm00446193_m1
Tlr2 Mm00442346_m1 Tlr6 Mm02529782_s1 Tnf Mm00443258_m1
TLR2 Is Highly Overexpressed in Retinal Myeloid Cells in the rd10 Mouse Model of Retinitis… 411

Tlr9). The analysis covered the period from post- tions in rd10 retinas: similar levels were observed
natal day (P)14, before the onset of degeneration, in microglia and monocyte populations, while
to P21, which corresponds to early-stage RP [5]. those quantified in macrophages were three times
By P21 all Tlr genes analyzed showed increased higher (Fig. 2d). In rd10 retinas, TLR2 expres-
expression in rd10 retinas (Fig. 1a), although the sion in activated microglia was four times higher
magnitude of this increase differed between than that observed in WT microglia (Fig. 2e).
genes: expression levels of Tlr1, Tlr2, and Tlr9
were markedly increased (30-, 36-, and 16-fold,
respectively), while those of Tlr3, Tlr4, Tlr6, and 4 Discussion
Tlr7 were increased by 5-, 9-, 6.5-, and 8.5-fold,
respectively, and Tlr5 levels were only modestly Here, we show that expression of all members of
increased (twofold). Tlr8 expression was below the TLR family is increased in the rd10 mouse
the limit of detection in WT animals but was retina in early stages of RP. Moreover, we
detected in rd10 retinas at P21 (not shown). observed a parallel increase in the expression of
Interestingly, the elevated expression of the dif- the proinflammatory cytokine genes Tnf and Il-­
ferent Tlr genes coincided with a dramatic 1β. The greatest increase was observed for Tlr2
increase in expression levels of the proinflamma- (36-fold increase with respect to WT), suggesting
tory cytokine genes Tnf and Il1β (over 90- and that TLR2 may play a prominent role in the
60-fold, respectively) at P21 (Fig. 1b). innate immune response associated with RP. In
fact, we previously demonstrated that deletion of
Tlr2 has a disease-modifying effect in two RP
3.2 TLR2 Levels in Retinal Cells models: elimination of Tlr2 in both rd10 and
from WT and rd10 Retinas P23H/+ mice, which carry different mutations
and have distinct hereditary patterns, attenuates
Flow cytometry analysis was used to characterize the progression of RP [4]. These findings suggest
the distribution of the TLR2 retinal cell popula- that the beneficial effects of Tlr2 deletion reflect
tion. WT and rd10 retinas at P25 were subjected a mutation-independent phenotype, supporting a
to flow cytometry following the gating strategy role of the innate immune response in the course
described by O’Koren [4, 6]. The myeloid popu- of RP.
lation was identified based on CD11b expression. Flow cytometry analysis revealed that TLR2
Within the population of CD11b+ myeloid cells, expression was restricted to the myeloid retinal
macrophages were defined as those with high population, in accordance with previous findings
levels of F4/80 expression. Cells with intermedi- [7], and confirmed the increase in Tlr2 expres-
ate/low F4/80 expression were classified as sion in rd10 retinas revealed by qPCR. The net
microglia (CD45 low) or monocytes (CD45 increase in Tlr2 expression was primarily due to
high). In the nonmyeloid (CD11b-) cell popula- an increased number of microglial cells, in which
tion in both WT and rd10 retinas, levels of TLR2 TLR2 expression was dramatically increased, as
(median of fluorescence values) were similar to well as monocyte and macrophage infiltration
those of the negative controls (Fig. 2a). However, from the periphery. We previously showed that
significantly higher TLR2 levels were observed Tlr2 deletion in rd10 mice results in a decrease in
in the myeloid cell population (CD11b+) microglial cell number [4], suggesting that mod-
(Fig. 2c), indicating selective enrichment of ulation of microgliosis could attenuate RP pro-
TLR2 in the myeloid population. gression. Although microglial cells are necessary
Cell numbers in the three myeloid cell sub- for homeostasis, overreactive microglia engulf
populations were increased in rd10 versus WT living photoreceptors, thus contributing to RP
retinas (Fig. 2b), in agreement with the microg- progression [8]. Together, our findings support
lial activation and monocyte and macrophage the view that TLR2 may constitute a therapeutic
infiltration associated with RP [4]. TLR2 expres- target for RP treatment, irrespective of the caus-
sion differed between the three myeloid popula- ative mutation.
412 A. Sánchez-Cruz et al.

Fig. 1 Expression of Tlr genes in WT and rd10 retinas. mean per group (+SEM). n = 3–4 animals per group.
Expression of Tlr1 to Tlr9 (a) and of the proinflammatory Transcript levels were normalized to Tbp RNA and
cytokine genes Tnf and Il1β (b) was analyzed by qPCR in expressed relative to WT counterparts (= 1) for each time
WT and rd10 retinas at the indicated postnatal ages. Dots point analyzed. ****p < 0.0001; ***p < 0.001 (two-way
represent individual animals, while bars represent the ANOVA followed by Sidak’s multiple comparison test)
TLR2 Is Highly Overexpressed in Retinal Myeloid Cells in the rd10 Mouse Model of Retinitis… 413

Fig. 2 Analysis of TLR2 levels in WT and rd10 retinas. tion. (c) Representative histograms showing TLR2 levels
P25 retinas were subjected to flow cytometry analysis by of the myeloid cell populations (CD11b+). (d, e) Analysis
surface staining with antibodies against CD11b, F4/80, of TLR2 levels in the indicated myeloid cell subpopula-
and CD45, and subpopulations were defined as described tion. Dots represent individual animals, while bars repre-
in the Results section. (a) Representative histograms sent the mean (+SEM) of each group. n = 4 mice.
depicting TLR2 levels (median fluorescence intensity) in ****p < 0.0001; **p < 0.01. (Unpaired t-test in B and E;
nonmyeloid retinal populations (CD11b-). (b) Graphs one-way ANOVA with Tukey’s post-test in D). MFI
show the number of cells in each myeloid cell subpopula- median fluorescence intensity

Acknowledgments We thank Cayetana Murillo and the deletion delays retinal degeneration in two genetically
staff of the CIB Margarita Salas animal facility for techni- distinct mouse models of retinitis pigmentosa. Int J
cal support. This work was supported by a grant from the Mol Sci. 2021;22(15):7815.
Spanish MICINN (PID2019-109506RB-100 to E.J.d.l.R. 5. Sanchez-Cruz A, Villarejo-Zori B, Marchena M,
and C.H.-S.). Zaldivar-Diez J, Palomo V, Gil C, et al. Modulation of
GSK-3 provides cellular and functional neuroprotec-
tion in the rd10 mouse model of retinitis pigmentosa.
Mol Neurodegener. 2018;13(1):19.
References 6. O’Koren EG, Mathew R, Saban DR. Fate map-
ping reveals that microglia and recruited
1. Silverman SM, Wong WT. Microglia in the retina: monocyte-derived macrophages are definitively
­
roles in development, maturity, and disease. Annu Rev distinguishable by phenotype in the retina. Sci Rep.
Vis Sci. 2018;4:45–77. 2016;6(1):20636.
2. Murakami Y, Ishikawa K, Nakao S, Sonoda 7. Hooper MJ, Wang J, Browning R, Ash JD. Damage-­
K-H. Innate immune response in retinal homeosta- associated molecular pattern recognition is
sis and inflammatory disorders. Prog Retin Eye Res. required for induction of retinal neuroprotective
2020;74:100778. pathways in a sex-dependent manner. Sci Rep.
3. Heneka MT, Kummer MP, Latz E. Innate immune 2018;8(1):9115.
activation in neurodegenerative disease. Nat Rev 8. Zhao L, Zabel MK, Wang X, Ma W, Shah P, Fariss
Immunol. 2014;14(7):463–77. RN, et al. Microglial phagocytosis of living photore-
4. Sánchez-Cruz A, Méndez AC, Lizasoain I, de la Villa ceptors contributes to inherited retinal degeneration.
P, de la Rosa EJ, Hernández-Sánchez C. Tlr2 gene EMBO Mol Med. 2015;7(9):1179–97.
Environmental Light Has
an Essential Effect on the Disease
Expression in a Dominant RPE65
Mutation

Wenjing Wu, Yusuke Takahashi, Xiang Ma,


Gennadiy Moiseyev, and Jian-Xing Ma

Abstract reduced electroretinography (ERG) ampli-


tude, recapitulating that observed in human
The retina pigmented epithelium 65 kDa pro- patients. Our findings indicated the impor-
tein (RPE65) is an essential enzyme in the tance of the light environment in the mecha-
visual cycle that regenerates the 11-cis-retinal nism of RPE65 D477G pathogenicity.
chromophore obligatory for vision. Mutations
in RPE65 are associated with blinding dis- Keywords
eases. D477G (C.1430G > A) is the only
RPE65 · Luminance · Retinitis pigmentosa ·
known RPE65 variant to cause autosomal
D477G · Blindness · Inherited retinopathy ·
dominant retinitis pigmentosa (adRP).
Visual cycle
Previously, we reported that the heterozygous
D477G knock-in (WT/KI) mice exposed to
dim light intensity demonstrated delayed
chromophore regeneration rates and slowed 1 Introduction
recovery of photoreceptor sensitivity follow-
ing photobleaching. However, visual function Inherited retinopathies (IR) are a major cause of
and retinal architecture were indistinguishable childhood blindness affecting 1 in 2000 people
from the wild-type (WT) mice. In this study, worldwide [4]. Mutations in over 300 genes
when maintained under the physiological day-­ expressed in both the photoreceptor (PR) and the
light intensity (2 K lux), the WT/KI heterozy- retinal pigment epithelium (RPE) cause irrepara-
gous mice displayed retina degeneration and ble blindness in humans. In the United States,
mutations in the RPE65 retinol isomerase account
for no less than 13% of all IR [1]. These mutations
W. Wu · X. Ma · G. Moiseyev (*) cause Leber congenital amaurosis (LCA), early-
Department of Physiology, University of Oklahoma
Health Sciences Center, Oklahoma City, OK, USA onset severe retinal dystrophy (EOSRD), and reti-
e-mail: Gennadiy-Moiseyev@ouhsc.edu nitis pigmentosa (RP) [18]. Collectively, these
Y. Takahashi · J.-X. Ma (*) disorders cause progressive vision loss, eventually
Department of Physiology, University of Oklahoma leading to blindness [3]. As the causes of these
Health Sciences Center, Oklahoma City, OK, USA ocular diseases are genetically based, the contribu-
Harold Hamm Diabetes Center, University of tion of environmental factors in the progression of
Oklahoma Health Sciences Center, the dystrophies remains to be elucidated. Currently,
Oklahoma City, OK, USA over 100 pathogenic mutations spanning all 14
e-mail: Jian-xing-ma@ouhsc.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 415
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_61
416 W. Wu et al.

exons of the RPE65 gene have been identified in heterozygotes (WT/KO). It is well documented
the human population to cause retinal degenera- that one copy of the functional RPE65 gene is
tion. Almost all of these mutations are recessively sufficient to maintain normal retinal health and
inherited and affect its isomerase function. In vision in both human and rodent models [5]. We
2011, Bowne’s group uncovered the first patho- demonstrated that when exposed under the same
genic dominant-acting mutation of RPE65 [2]. A day-light luminance intensity of 2000 lux, com-
single-nucleotide substitution at exon 13 of the pared to the WT/KO heterozygotes, the WT/KI
RPE65 gene, a c.1430A > G, caused a missense heterozygote mice displayed reduced total retina
mutation, an aspartic acid residue at position 477 thickness and reduced scotopic ERG amplitude
to glycine (D477G). Subsequently, the D477G beginning at 15 months of age. Molecularly we
mutation was identified in other patients with demonstrated that D477G causes atypical
adRP of unknown etiology [8]. protein-­protein interactions with WT-RPE65
Unlike typical RP patients, RPE65-D477G leading to early RPE65 protein degradation (data
carriers exhibit various retinal degeneration phe- not shown). In this study, we ascertained that the
notypes, including the simultaneous loss of retinal dysfunction in the RPE65 WT/KI mouse
peripheral and central vision, which may involve model is due to D477G being dominant negative.
choroidal atrophy and often include macula and/ We also identified a link between environmental
or RPE perturbations [6]. The onset of visual light and vision impairment caused by genetic
impairment among the D477G carriers occurs at mutations.
various ages, and the degree of retinal degenera-
tion is often independent of the extent of the
decline in visual function [7]. Three groups have, 2 Materials and Methods
in total, generated five D477G knock-in (KI)
mouse models to study the pathophysiology of 2.1 Animals
the mutant in vivo [9]. As with the heterogeneous
clinical features of the D477G adRP patients, the All animal experiments were approved by the
reported impacts of the mutation were diverse. Institutional Animal Care and Use Committee of
As yet, none of the WT/KI mouse models have the University of Oklahoma Health Sciences
recapitulated the visual and morphological Center (Oklahoma City, OK) and performed fol-
defects found in the human patients or provided a lowing the guidelines of the Association for
satisfactory explanation for the observed D477G Research in Vision and Ophthalmology Statement
dominant pattern of human inheritance. Thus, the for the Use of Animals in Ophthalmic and Vision
molecular mechanisms for retinal dysfunction Research. The RPE65 D477G KI mouse model
and degeneration caused by the D477G mutation was generated by our group previously [15].
remain inconclusive. RPE65 knockout (KO) mice were a kind gift
Here we report retinal dysfunction and mor- from Dr. Michael Redmond (National Eye
phological changes in WT/KI that resembled the Institute, Bethesda, MD) [14]. WT/KI mice were
phenotypes observed in patients. We discovered obtained by crossing homozygous RPE65 KO
that ambient light influenced the occurrence of with WT C57BL/6 J mice. All mice were in the
retinal dysfunction in the WT/KI mice. We C57BL/6 J background and were maintained in
addressed the dominant-negative nature of the standard housing conditions with 12/12 h, food
mutation by comparing WT/KI to RPE65 KO and water ad libitum.
Environmental Light Has an Essential Effect on the Disease Expression in a Dominant RPE65 Mutation 417

2.1.1 Physiological Day-Light 3 Results


Exposure
For chronic light exposure at physiological lumi- 3.1 DayL-Exposed RPE65 WT/KI
nance of 2 K lux, the experimental mice were Mice Exhibited Both Retinal
first kept at regular vivarium dim-light (DimL, Function and Morphological
~100 Lux) to around 3 months of age. At age Changes
3 months, the WT/KI and the WT/KO were
grouped randomly to be maintained in 12 h day-­ Previously we and others have detected no visual
light (DayL, ~2 K Lux)/12 h dark or in 12 h function differences between the WT/KI and the
DimL/12 h dark conditions. wild-type (WT/WT) mice when raised under
dim-light (DimL) intensity (~100 lux) [9]. Here
we evaluated visual function in the WT/KI and
2.2 ERG WT/KO mice exposed to day-light (DayL) lumi-
nance of 2 K lux and found that starting at
Full-field ERG was recorded in the WT/KI and 15 months of age, compared to the WT/KO, the
WT/KO mice, maintained under either DimL or WT/KI mice exposed to DayL displayed reduced
DayL, every 3 months, as previously described scotopic ERG a- and b-wave amplitudes (Fig. 1).
[13]. All ERGs were recorded with the Espion E3 Previous studies reported that WT/KI mice reared
system Ganzfeld Color Dome system (Diagnosys under DimL showed normal retina morphology
LLC, Lowell, MA). [10]. Here we found that, compared to the WT/
KO, WT/KI mice maintained under DayL lumi-
nance displayed retina degeneration in the form
2.3 Histology of decreased total retina thickness (Fig. 2a–e), a
phenotype seen in human D477G patients [6].
As previously described, enucleated eyes were
fixed in Davidson’s fixative and stored in 70%
ethanol for subsequent serial dehydration and 4 Discussion
embedment in paraffin [11]. Eyes were sectioned
(5 μm thickness) on a standard microtome and In this study, we manipulated the environmental
then annealed onto glass slides at 60 °C. Following luminance to expose the retinal pathogenic effect
hematoxylin/eosin staining of tissue sections, the of D477G. By directly comparing phenotypic
Olympus Provis Ax-70 light microscope assessed changes of the WT/KI to WT/KO mice, we pro-
gross retinal morphology. vided functional and molecular (data not shown)
evidences supporting the dominant-negative
2.3.1 Spectral Domain Optical nature of the D477G mutant. WT/KI raised under
Coherence Tomography DayL showed reduced retinal ERG amplitudes
(SD-OCT) (Fig. 1) and architectural defects (Fig. 2d). Our
Mice were anesthetized, and their pupils were results suggested that the adverse physiological
dilated. Artificial tears (Systane Ultra, Alcon, consequence of D477G mutant co-expressed
Fort Worth, TX) were used to maintain corneal with WT-RPE65 far exceeded that of losing a
hydration and clarity. The retinal thickness was single allele of WT-RPE65 (Figs. 1b and 2a, b),
measured using a spectral-domain optical coher- which demonstrated that the retinal dysfunction
ence tomography (SD-OCT) device (Bioptigen caused by WT/KI is not haploinsufficiency.
Inc. Durham, NC) following a previously estab- Moreover, we identified ambient light as a causal
lished method [12]. factor that could induce the progression of IR.
418 W. Wu et al.

Fig. 1 ERG declined in WT/KI exposed to DayL lumi- 15 months. (b) Quantitative evaluation of scotopic
nance. (a) Representative dark-adapted scotopic single-­ response amplitudes of the WT/KI and WT/KO. Data are
flash ERG traces at 4 cd.s/m2 in mice of indicated presented as mean ± SEM. n = 8 (*P ≤ 0.05 **P ≤ 0.01,
genotypes maintained under DayL or DimL at age in one-way ANOVA with Tukey’s post hoc comparison)

Fig. 2 Total retina thickness thinning observed in DayL-­ total retina thickness in 15-month-old mice of indicated
exposed WT/KI mice. (a–d) Representative images of genotypes exposed to DimL or DayL luminance. Data are
H&E-stained paraffin-embedded eye sections from mice presented as mean ± SEM. n = 6 (**P ≤ 0.01, in one-way
of indicated genotypes. (e) SD-OCT quantification of ANOVA with Tukey’s post hoc comparison)

Acknowledgments The authors are grateful to the


OUSHC animal husbandry team for their diligence and References
devotion to the well-being of all our mice. This study is
supported by the National Institutes of Health (NIH) 1. Aoun M, Passerini I, Chiurazzi P, Karali M, De
grants (EY018659, EY019309, EY012231, EY028949, Rienzo I, Sartor G, et al. Inherited retinal diseases due
EY032930, EY032931). to RPE65 variants: from genetic diagnostic manage-
ment to therapy. Int J Mol Sci. 2021;22(13):7207.
Environmental Light Has an Essential Effect on the Disease Expression in a Dominant RPE65 Mutation 419

2. Bowne SJ, Humphries MM, Sullivan LS, Kenna PF, variant associated with autosomal dominant retinitis
Tam LCS, Kiang AS, et al. A dominant mutation in pigmentosa. Genes (Basel). 2020;11(12):1420.
RPE65 identified by whole-exome sequencing causes 10. Li Y, Furhang R, Ray A, Duncan T, Soucy J, Mahdi R,
retinitis pigmentosa with choroidal involvement. Eur et al. Aberrant RNA splicing is the major pathogenic
J Hum Genet. 2011;19(10):1074–81. effect in a knock-in mouse model of the dominantly
3. Cai X, Conley SM, Naash MI. RPE65: role in the inherited c.1430A>G human RPE65 mutation. Hum
visual cycle, human retinal disease, and gene therapy. Mutat. 2019;40(4):426–43.
Ophthalmic Genet. 2009;30(2):57–62. 11. Ma X, Takahashi Y, Wu W, Liang W, Chen J,
4. Cremers FPM, Boon CJF, Bujakowska K, Zeitz Chakraborty D, et al. ADAM17 mediates ectodomain
C. Special issue introduction: inherited retinal dis- shedding of the soluble VLDL receptor fragment in the
ease: novel candidate genes, genotype-phenotype retinal epithelium. J Biol Chem. 2021;297(4):101185.
correlations, and inheritance models. Genes (Basel). 12. Qiu F, Ma X, Shin Y-H, Chen J, Chen Q, Zhou K,
2018;9(4):215. et al. Pathogenic role of human C-reactive protein in
5. Galvin JA, Fishman GA, Stone EM, Koenekoop diabetic retinopathy. Clin Sci. 2020;134(13):1613–29.
RK. Evaluation of genotype-phenotype associations 13. Qiu F, Meng T, Chen Q, Zhou K, Shao Y, Matlock G,
in leber congenital amaurosis. Retina (Philadelphia, et al. Fenofibrate-loaded biodegradable nanoparticles
Pa). 2005;25(7):919–29. for the treatment of experimental diabetic retinopathy
6. Hull S, Mukherjee R, Holder GE, Moore AT, and neovascular age-related macular degeneration.
Webster AR. The clinical features of retinal disease Mol Pharm. 2019;16(5):1958–70.
due to a dominant mutation in RPE65. Mol Vis. 14. Redmond TM, Yu S, Lee E, Bok D, Hamasaki D,
2016;22:626–35. Chen N, et al. Rpe65 is necessary for production
7. Jauregui R, Park KS, Tsang SH. Two-year progres- of 11-cis-vitamin A in the retinal visual cycle. Nat
sion analysis of RPE65 autosomal dominant retinitis Genet. 1998;20(4):344–51.
pigmentosa. Ophthalmic Genet. 2018;39(4):544–9. 15. Shin Y, Moiseyev G, Chakraborty D, Ma J-X. A
8. Kenna PF, Humphries MM, Kiang A-S, Brabet P, dominant mutation in Rpe65, D477G, delays dark
Guillou L, Ozaki E, et al. Advanced late-onset retini- adaptation and disturbs the visual cycle in the mutant
tis pigmentosa with dominant-acting D477G RPE65 knock-in mice. Am J Pathol. 2017;187(3):517–27.
mutation is responsive to oral synthetic retinoid ther- 16. Thompson DA, Gyürüs P, Fleischer LL, Bingham
apy. BMJ Open Ophthalmol. 2020;5(1):e000462-e. EL, McHenry CL, Apfelstedt-Sylla E, et al. Genetics
9. Kiang AS, Kenna PF, Humphries MM, Ozaki E, and phenotypes of RPE65 mutations in inherited
Koenekoop RK, Campbell M, et al. Properties and ther- retinal degeneration. Invest Ophthalmol Vis Sci.
apeutic implications of an enigmatic D477G RPE65 2000;41(13):4293–9.
Microglia Preserve Visual Function
in a Mouse Model of Retinitis
Pigmentosa with Rhodopsin-P23H
Mutant

Chen Yu and Daniel R. Saban

Abstract Keywords

Microglia · Macrophages · Subretinal space ·


Most forms of outer retinal degenerative dis-
Photoreceptor · Retinal degeneration ·
eases involve the ectopic accumulation of
Retinitis pigmentosa · Rhodopsin · P23H
microglia/macrophages in the subretinal
space, including retinitis pigmentosa.
However, their role in the loss of photorecep-
tor function during retinal degeneration
Abbreviations
remains unknown. Here, we examined the
effect of conditional microglial depletion on
CNS central nervous system
photoreceptor numbers and visual function in
DAM disease-associated microglia
mice with the rhodopsin P23H mutation, a
ERG electroretinogram
dominant form of retinitis pigmentosa in
ONL outer nuclear layer
humans. We found that microglial depletion
RPE retinal pigment epithelium
led to an elevated level of rhodopsin and
increased photoreceptor layer thickness.
However, overall electrophysiological func-
tions of the retina were reduced with microg- 1 Introduction
lial depletion. Therefore, these results identify
an essential role of microglia specially in pre- Microglia, the tissue-resident macrophages in the
serving visual function in outer retinal central nervous system (CNS), are the major
degeneration. immune cells and serve as immune sentinels of
the CNS, including the retina [1, 2]. In particular
to outer retinal degenerative diseases in both ani-
C. Yu
Department of Ophthalmology, Duke University mal models and patients, microglia/macrophages
School of Medicine, Durham, NC, USA are found accumulating at the site of degenera-
D. R. Saban (*) tion, referred to as the subretinal space [3, 4].
Department of Ophthalmology, Duke University However, studies of microglia function in disease
School of Medicine, Durham, NC, USA are often confounded by the presence of infiltrat-
Departments of Ophthalmology and Immunology, ing of monocyte-derived cells [5–7]. Given the
Duke University School of Medicine, differences in ontogeny and residency between
Durham, NC, USA microglia and infiltrated monocyte-derived mac-
e-mail: daniel.saban@duke.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 421
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_62
422 C. Yu and D. R. Saban

rophages [8], appreciations of their functional (DT; Sigma-Aldrich) at P21 and P28, and used
differences have been emerging in many degen- for experiment at P60. The same genotype litter-
erative settings of the CNS. Recently, we identi- mates that did not receive DT were used as con-
fied that microglia are the predominant cells at trols for experiments.
the subretinal space, which clear photoreceptor
debris and protect the retinal pigment epithelium
(RPE) from damage caused by degeneration [9]. 2.3 Electroretinogram (ERG)
However, it remains unclear about their role in
preservation of photoreceptors and vision in ERG was performed as previously described
degeneration. Here, we performed microglia-­ [11]. Briefly, mice were dark-adapted for at least
specific depletion to understand their impact on 4 h, and pupils were dilated with 0.5% (wt/vol)
photoreceptor function in a genetic mouse model tropicamide and 1.25% (wt/vol) phenylephrine
of retinitis pigmentosa with rhodopsin-P23H and anesthetized with a mixture of ketamine
mutant (RhoP23H mice) [10], which is a dominant (100 mg/kg) and xylazine (10 mg/kg). Scotopic
form of retinitis pigmentosa in humans. ERG responses were recorded using an Espion
E2 system (Diagnosys LLC, Lowell, MA), at
increasing flash intensities (2.5 × 10−5, 5 × 10−5,
2 Materials and Methods 5 × 10−4, 5 × 10−3, 0.05, 0.5, 5, 50, and 500 cd·s/
m2). Recordings consisted of single-flash presen-
2.1 Animals tations, repeated 0–15 times to verify the response
reliability and improve the signal-to-noise ratio.
Cx3cr1YFP-Cre-ER (stock No. 021160) and
Rosa26iDTR (stock No. 006148) were originally
purchased from the Jackson Laboratories. RhoP23H 2.4 Tissue Processing
mice were generated as described previously for Histology
[10]. These mice were crossed to generate and Immunoblotting
RhoP23H/+; Cx3cr1YFP-Cre-ER/+; Rosa26iDTR/+ (P23H-­
iDTR) mice for microglia-specific depletion. All For histology of plastic sections, euthanized mice
mice herein did not carry rd8 mutation and were were perfused with PBS and then fixed via trans-
bred and housed at a barrier-free and specific-­ cardial perfusion with 0.1% cacodylate buffer
pathogen-­free facility at Duke University School (pH = 7.2) containing 2% paraformaldehyde and
of Medicine. All procedures were carried out in 2% glutaraldehyde. The eye tissues were post-
accordance with the guidelines of the Association fixed in the same fixative for at least 24 h and
for Research in Vision and Ophthalmology state- then processed in a solution of 2% osmium
ment for the “Use of Animals in Ophthalmic and tetroxide in 0.1% cacodylate buffer, following by
Vision Research” and approved by the dehydration with gradient ethanol from 50% to
Institutional Animal Care and Use Committee at 100% and propylene oxide, and infiltration of
Duke University. propylene oxide: epoxy 812 compound with the
ratio of 1:1 overnight under the vacuum. Samples
were further processed with pure epoxy 812
2.2 Microglia-Conditional compound the next day and embedded in fresh
Depletion epoxy 812 compound resins at 65 °C overnight.
Semi-thin cross sections with 0.5 μm thickness
Tamoxifen (Sigma-Aldrich, 75 mg/kg dissolved were stained with 1% methylene blue.
in corn oil) was intraperitoneally injected at post- For immunoblotting, dissected retinas were
natal day 4 (P4) and 7 (P7), respectively. After sonicated with 2% SDS in PBS containing prote-
tamoxifen treatment, mice were intraperitoneally ase inhibitor mixture. Lysates were centrifuged at
injected with two doses of 0.5 μg diphtheria toxin 14,800 g for 5 min, and then supernatants were
Microglia Preserve Visual Function in a Mouse Model of Retinitis Pigmentosa with Rhodopsin-P23H Mutant 423

collected. Equal volumes of supernatants were 3.2 Effect of Microglial Depletion


loaded to SDS/PAGE. Proteins were transferred on Photoreceptor Cell
to PVDF membrane (Bio-Rad) and probed with Numbers
primary antibodies against rhodopsin (4D2) and
HLA and goat secondary antibodies conjugated To compare rod photoreceptor levels, we first
with Alexa Fluor 680 or 800 (Invitrogen). Protein analyzed the protein level of rhodopsin in retinal
levels were detected using an Odyssey infrared lysates. Immunoblotting revealed a ~25%
imaging system (LI-COR Bioscience). increase in normalized rhodopsin levels in the
microglia depletion setting (Fig. 1b, c). To test
the effect of microglial depletion on photorecep-
2.5 Statistics tor cell numbers, we measured the thickness of
outer nuclear layer (ONL) in central retinal cross
Graphical data presented in this study are pre- sections across the optic nerve. We found that
sented as the means ± SDs. Data normality and microglial depletion resulted in a thicker photo-
homogeneity of variance were assessed to fit the receptor layer compared with non-depleted con-
assumptions of tests, and transformation was per- trol group (Fig. 1d), which is consistent with
formed when necessary. Two-group comparisons elevated rhodopsin level. Therefore, these data
were analyzed using unpaired Student’s two-­ suggest that microglial clearance contributes to
tailed t test. Multiple-factor comparisons were removal of photoreceptors in retinitis pigmen-
analyzed using ANOVA with Tukey’s post hoc tosa. However, microglial depletion also led to
test. A p-value of <0.05 was considered statisti- the accumulation of dead photoreceptors in this
cally significant. Statistical graphs were gener- model as shown previously [9]. In addition, we
ated using GraphPad Prism version 9.1.2 (La found that the length of outer segment appears
Jolla). shortened with microglial depletion compared
with control, suggesting a compromised function
of these photoreceptors in the absence of microg-
3 Results lia (Fig. 1e).

3.1 Conditional Microglial


Depletion in RhoP23H Mice 3.3 Effect of Microglial Depletion
on Visual Function
To investigate the effect of microglia on photo-
receptors in retinal degeneration, we utilized Next, we examined visual function of these mice
lineage tracing technique in P23H-iDTR mice by measuring their scotopic and photopic ERG
to specifically deplete microglia. Briefly, the responses. Interestingly, despite increased photo-
expression of DTR driven by a macrophage pro- receptor cell number with microglial depletion,
moter of Cx3cr1 was induced by tamoxifen scotopic a-waves were similar between two
treatments at P4 and P7. During 2-week wash- groups, which suggests the presence of nonfunc-
out period, short-­lived monocytes were replaced tional photoreceptors or reduced function of indi-
by Cx3cr1− bone marrow stem cells and lost vidual rod photoreceptor with microglial
DTR expression, while long-lived microglia depletion (Fig. 2a). Moreover, amplitudes of sco-
maintained the expression of DTR. Therefore, topic b-waves were reduced with microglial
by administrating DT, we were able to deplete depletion, indicating deficiency in responses of
microglia but spare monocyte-derived cells. We postsynaptic neurons to rod photoreceptors
achieved approximately 70% depletion effi- (Fig. 2b). We did not detect any significant
ciency by analyzing the abundance of subretinal changes in either photopic a-waves or b-waves at
microglia at P60, 1 month since last DT admin- P60 (Fig. 2c, d). Collectively, our findings dem-
istration (Fig. 1a). onstrate that microglia depletion leads to impaired
424 C. Yu and D. R. Saban

Fig. 1 Microglial depletion led to the inefficient clear- depletion. Rhodopsin levels were normalized to the load-
ance of photoreceptors in RhoP23H mice. (a) Quantifications ing control HLA and shown as relative to control (n = 5
of depletion efficiency of subretinal microglia between per group). (d) Graph showing ONL thickness with dis-
control (ctrl, n = 4) and depleted (dep, n = 8) P23H-iDTR tance to the optic nerve. Ctrl: n = 10; dep: n = 15 [9]. (e)
mice [9]. (b) Representative immunoblots showing Representative images of retinal cross sections showing
increased rhodopsin level with microglial depletion as the shrinkage of outer segments with microglial depletion
indicated. (c) Quantifications of normalized rhodopsin as indicated by arrows [9]. Scale bar = 100 μm. *p < 0.05;
(Rho) protein levels between control and microglial **p < 0.01; ***p < 0.001

Fig. 2 Microglial depletion impaired dark-adapted vision respectively. N = 5 mice per group. **p < 0.01. ns not
in RhoP23H mice. Graphs showing a-waves and b-waves of significant
scotopic (a and b) and photopic ERG responses (c and d),

visual function derived from rod but not cone CR3-dependent clearance to protect photorecep-
photoreceptors in the rhodopsin-P23H mouse tors in rd10 mouse model of retinitis pigmentosa
model of retinitis pigmentosa. [12]. Together with our previous findings on their
protection to RPE cells [9], these results indicate
a critical role of clearance by microglia to limit
4 Discussion deleterious materials released from cell death and
to restore retinal homeostasis in degeneration.
In this study, we applied conditional microglia We showed that microglial depletion leads to a
depletion and demonstrated that absence of deterioration of ERG responses in RhoP23H mice,
microglia accelerates loss of both functional pho- especially scotopic b-waves. Reduced b-waves
toreceptors and vision in retinitis pigmentosa. may result from lack of direct protection or func-
The increased rhodopsin protein level and photo- tional support by microglia on postsynaptic neu-
receptor layer by microglial depletion appear to rons or photoreceptors [13]. Moreover, microglial
result from the absence of dead cell clearance. A depletion had negligible consequences on scoto-
recent study showed that microglia utilize C3 and pic a-waves in our study, which could be
Microglia Preserve Visual Function in a Mouse Model of Retinitis Pigmentosa with Rhodopsin-P23H Mutant 425

explained by at least two reasons. First, the extra degeneration, and age-related macular degeneration.
Exp Eye Res. 2003;76(4):463–71.
photoreceptors are not likely all dead and do not 5. O’Koren EG, Mathew R, Saban DR. Fate mapping
completely lose their functions. Some of them reveals that microglia and recruited monocyte-derived
may be in the process of losing functions but still macrophages are definitively distinguishable by phe-
partially contribute to the ERG responses. notype in the retina. Sci Rep. 2016;6:20636.
6. Sennlaub F, Auvynet C, Calippe B, Lavalette S,
Second, microglia repopulate themselves in the Poupel L, Hu SJ, et al. CCR2(+) monocytes infiltrate
brain and retina after depletion [14, 15]. atrophic lesions in age-related macular disease and
Repopulating microglia since depletion at P28 mediate photoreceptor degeneration in experimental
may further minimize the difference on scotopic subretinal inflammation in Cx3cr1 deficient mice.
EMBO Mol Med. 2013;5(11):1775–93.
a-waves observed in this study. 7. Lad EM, Cousins SW, Van Arnam JS, Proia
Recent single-cell RNA-sequencing data AD. Abundance of infiltrating CD163+ cells in the
reveal that these subretinal microglia undergo retina of postmortem eyes with dry and neovascular
transcriptional reprogramming [9, 16], which are age-related macular degeneration. Graefes Arch Clin
Exp Ophthalmol. 2015;253(11):1941–5.
similar with disease-associated microglia (DAM) 8. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P,
in other CNS degenerative conditions [17]. Our Gokhan S, et al. Fate mapping analysis reveals that
strategy demonstrated a protective net effect of adult microglia derive from primitive macrophages.
microglial depletion and eliminated about ~70% Science. 2010;330(6005):841–5.
9. O’Koren EG, Yu C, Klingeborn M, Wong AYW,
subretinal microglia. Given their proximity to Prigge CL, Mathew R, et al. Microglial function is
degenerating photoreceptors and the RPE, we distinct in different anatomical locations during
speculate that the phenotypes of microglial retinal homeostasis and degeneration. Immunity.
depletion are largely due to loss of these cells 2019;50(3):723–37 e7.
10. Sakami S, Maeda T, Bereta G, Okano K, Golczak M,
specifically in the subretinal microglia. However, Sumaroka A, et al. Probing mechanisms of photo-
we cannot rule out the direct contribution of receptor degeneration in a new mouse model of the
microglia in the inner retina. Further studies are common form of autosomal dominant retinitis pig-
needed to better understand the precise role of mentosa due to P23H opsin mutations. J Biol Chem.
2011;286(12):10551–67.
subretinal microglia in retinal degeneration. 11. Herrmann R, Lobanova ES, Hammond T, Kessler C,
Burns ME, Frishman LJ, et al. Phosducin regulates
Acknowledgments This work was funded by the transmission at the photoreceptor-to-ON-bipolar cell
National Institutes of Health grants R01EY030906 and synapse. J Neurosci. 2010;30(9):3239–53.
R01EY021798 from the National Eye Institute, Bright 12. Silverman SM, Ma W, Wang X, Zhao L, Wong
Focus Foundation MDR grant, Research to Prevent WT. C3- and CR3-dependent microglial clearance
Blindness (Unrestricted, Duke Eye Center), NIH/NEI protects photoreceptors in retinitis pigmentosa. J Exp
Core grant P30EY005722 (Duke Eye Center). We would Med. 2019;216(8):1925–43.
like to thank William J. Spencer and Mikael Klingeborn 13. Wang X, Zhao L, Zhang J, Fariss RN, Ma W,
for their technical supports. Kretschmer F, et al. Requirement for microglia for the
maintenance of synaptic function and integrity in the
mature retina. J Neurosci. 2016;36(9):2827–42.
14. Huang Y, Xu Z, Xiong S, Sun F, Qin G, Hu G, et al.
References Repopulated microglia are solely derived from the
proliferation of residual microglia after acute deple-
1. Yu C, Roubeix C, Sennlaub F, Saban DR. Microglia tion. Nat Neurosci. 2018;21(4):530–40.
versus monocytes: distinct roles in degen- 15. Huang Y, Xu Z, Xiong S, Qin G, Sun F, Yang J, et al.
erative diseases of the retina. Trends Neurosci. Dual extra-retinal origins of microglia in the model of
2020;43(6):433–49. retinal microglia repopulation. Cell Discov. 2018;4:9.
2. Reyes NJ, O’Koren EG, Saban DR. New insights into 16. Yu C, Saban DR. Identification of a unique sub-
mononuclear phagocyte biology from the visual sys- retinal microglia type in retinal degeneration
tem. Nat Rev Immunol. 2017;17(5):322–32. using single cell RNA-Seq. Adv Exp Med Biol.
3. Thanos S. Sick photoreceptors attract activated 2019;1185:181–6.
microglia from the ganglion cell layer: a model to 17. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-­
study the inflammatory cascades in rats with inherited Natan O, Dvir-Szternfeld R, Ulland TK, et al. A
retinal dystrophy. Brain Res. 1992;588(1):21–8. unique microglia type associated with ­ restricting
4. Gupta N, Brown KE, Milam AH. Activated microg- development of Alzheimer’s disease. Cell.
lia in human retinitis pigmentosa, late-onset retinal 2017;169(7):1276–90 e17.
Part IX
Mechanisms
of Degeneration – Metabolism
Measuring the Release of Lactate
from Wild-Type and rd1 Mouse
Retina

Yiyi Chen, Laimdota Zizmare,


Christoph Trautwein,
and François Paquet-Durand

Abstract even higher energy demand and metabolic rate


compared to healthy retina. We conclude that
The retina has the highest energy consumption the use of lactate measurement can be a reli-
of any tissue in the human body. Remarkably, able and simple readout to evaluate ongoing
to satisfy its energy demand, the retina appears retinal metabolism.
to rely mostly on aerobic glycolysis, which
results in the production and release of large Keywords
amounts of lactate. In the present study, we
compared two different methods to assess lac- Aerobic glycolysis · Retinal metabolism ·
tate release from in vitro organotypic retinal Retina degeneration · Warburg effect ·
explants cultured under entirely controlled, NMR Spectroscopy
serum-free conditions. We used a standard
lactate assay kit and 1H-nuclear magnetic res-
onance (NMR) spectroscopy-based analysis. 1 Introduction
We found that during the culturing of retinal
explants derived from wild-type mice, lactate The metabolism of the retina is characterized by
was released in large amounts and that the two a very high energy demand [1, 2] most of which
different methods agreed well with each other. likely stems from photoreceptors and their need
When comparing wild-type retina with degen- to maintain the so-called dark current [3, 4]. Most
erating rd1 mouse retina, we found the latter of the ATP generated in the retina likely comes
to release significantly higher amounts of lac- from the relatively energy-inefficient glycolysis,
tate. Hence, degenerating retina may have an even though oxygen for the more energy-efficient
oxidative phosphorylation would be available.
This phenomenon was recognized by Otto
Y. Chen · F. Paquet-Durand (*) Warburg already 100 years ago and termed “aero-
Cell Death Mechanism Group, Institute for bic glycolysis” [5–7]. The by-product of glycoly-
Ophthalmic Research, University of Tübingen,
Tübingen, Germany sis, lactate, is a highly dynamic metabolite that
e-mail: Francois.paquet-durand@klinikum.uni- may shuttle through the retinal pigment epithe-
tuebingen.de lium (RPE), to the bloodstream. In the retina high
L. Zizmare · C. Trautwein lactate levels (13 mM) were detected at the inter-
Werner Siemens Imaging Center, Department of photoreceptor matrix, while 3.8 mM lactate was
Preclinical Imaging and Radiopharmacy, University measured near the RPE surface [8].
of Tübingen, Tübingen, Germany

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 429
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_63
430 Y. Chen et al.

Given the significance of lactate for retinal and antibiotics, as previously described [11–
energy metabolism, we set out to explore the extra- 13]. Briefly, after removing the sclera, the retina
cellular lactate released from organotypic retinal was placed on cell culture inserts (Corning,
explant cultures using two different ­methodologies: Kennebunk, ME; Cat. No.: 3412), with the gan-
[1] enzymatic lactate conversion and colorimetric glion cell layer facing up and cultured in 1 ml
detection and [2] 1H-nuclear magnetic reso- R16 complete medium (CM) in a humidified
nance (NMR) spectroscopy-based metabolomics incubator with 5% CO2 at 37 °C. The whole cul-
analysis. With both methods we found a strong and ture medium was collected and replaced by 1 ml
continuous lactate excretion from the cultured ret- of fresh CM every 2 days. The culture was
ina. Moreover, retina derived from the Pde6b ended at P15 and the retinal tissue was fixed
mutant, photoreceptor degeneration rd1 mouse [9], with 4% PFA and embedded in cryomatrix for
released significantly larger amounts of lactate into subsequent cryosectioning. For immunohisto-
the surrounding medium when compared with chemistry, cryosections were rinsed with PBS
wild-type (WT). Given the stability of lactate, we three times at room temperature. Then, cell
demonstrate its potential usage in in vitro organo- nuclei were stained with DAPI and sections
typic retinal explant cultures as a biomarker for were mounted with Vectashield. Finally, fluo-
metabolic activity under both physiological and rescence images were generated by a Zeiss
pathophysiological conditions. microscope equipped with a Z1 Aptome (Zeiss,
Oberkochen, Germany).

2 Materials and Methods


2.3 Lactate Measurements
2.1 Animals
Lactate in culture media were measured spec-
C3H HeA Pde6brd1/rd1 (rd1) and congenic C3H trophotometrically using the L-lactate assay kit
HeA Pde6b+/+ wild-type (WT) mice were used I from Eton Bioscience Inc. (San Diego, CA,
[10]. Animals were housed under standard white USA; Cat. No.: 120001100P). In this assay the
cyclic illumination, had a free access to food and enzyme lactate dehydrogenase converts lactate
water, and were used irrespective of gender. The and NAD+ into pyruvate and NADH. The
procedures were reviewed and approved by the detection is based on the subsequent reduction
Tübingen University review board (AK02-19 M), of the tetrazolium salt iodonitrotetrazolium
and in compliance with the association for (INT) in a NADH-­coupled enzyme reaction to
research in vision and ophthalmology (ARVO) formazan, which exhibits an absorbance maxi-
statement for the use of animals in ophthalmic mum at 490 nm. Briefly, L-lactate standards
and visual research. Postnatal (P) day 9 animals and culture media samples were added into the
were sacrificed by decapitation, and both eyes corresponding wells of a 96-well plate. 50 μl of
were used for the explant preparation. A total of L-lactate assay solution was added to each
17 animals was used in this study, yielding 34 well. Subsequently, the 96-well plate was incu-
retinal explant cultures. bated at 37 °C for 30 min. Then, the reaction
was stopped by addition of 50 μl 0.5 M acetic
acid to each well, followed by brief and gentle
2.2 Organotypic Retinal Explant agitation. Finally, the absorbance was mea-
Cultures sured by a Tecan Spark10M microplate reader
(Tecan, Männedorf, Switzerland). The lactate
Retinal explants were derived from P9 mice and concentration was proportional to the absor-
were cultured in R16 medium (Gibco, Paisley, bance value at 490 nm and was calculated
Ireland; Cat. No.: 07491252A) free of serum based on the lactate standard curve.
Measuring the Release of Lactate from Wild-Type and rd1 Mouse Retina 431

2.4 Lactate Measurements Based 3 Results


on 1H-NMR
3.1 Steady Secretion of Lactate
400 μL of retina culture media was added to from Mouse Retina
400 μL of ultrapure methanol, thoroughly mixed
and centrifuged at 30,000× g for 30 min to remove To measure lactate release from retinal explants,
remaining solid particles that would disturb the we used the supernatant medium, which was col-
magnetic field homogeneity. The clear supernatant lected every 2 days at P11, P13, and P15 during
was transferred to a new PTFE tube, and the solu- media change. Media samples were split and ana-
tion was evaporated to dryness overnight in a vac- lyzed either with the standard lactate assay kit or
uum concentrator. Dry metabolite pellet was by using 1H-NMR spectroscopy. Notably, the
resuspended in 45 μL 200 mM phosphate serum-free R16 medium did not contain any lac-
(K2HPO4) buffer containing 200 μM NaN3 tate initially. After 2 days in culture, the lactate
(pH = 7.4). Due to the very high salt content, an content was strongly increased, a phenomenon
internal standard could not be used for absolute that was detected with both lactate assays (P11
quantification of metabolites; instead, integral lactate assay kit, 2.90 ± 0.20 mM, p < 0.001; 1H-­
areas were used for relative quantification. The NMR, 62.48 ± 8.32 standard area (sa), p < 0.001).
suspension was centrifuged at 30,000× g for Between P11 and P13, lactate continued to accu-
10 min, and 40 μL of the supernatant was trans- mulate in the culture media with an even higher
ferred to 1.7 mm autosampler-compatible NMR release rate (P13 lactate assay kit: 3.32 ± 0.17 mM;
tubes (Bruker BioSpin, Germany). Spectra were 1
H-NMR 64.15 ± 3.99 sa). By P15, lactate release
acquired on a 600 MHz ultra-shielded NMR spec- continued increasing abundantly (lactate assay
trometer (Avance III HD, Bruker BioSpin, kit 3.72 ± 0.79 mM; 1H-NMR 81.87 ± 7.02 sa,
Germany) with a 1.7 mm room temperature triple p < 0.01) (Fig. 1).
resonance probe at 298 K. A 1H-NMR 15 min
Carr-Purcell-Meiboom-Gill (CPMG) experiment
with residual macromolecule and water suppres- 3.2 Increased Lactate Release
sion was used. Spectra were processed in Bruker from rd1 Mouse Retina
TopSpin 3.6.1 software, and lactate integral area
was calculated by Chenomx NMR Suite 8.5 We then employed the lactate assay to compare
software. lactate release from a healthy wild-type (WT)
and degenerating rd1 mouse retina. In rd1 retina
a rapid degeneration of photoreceptors occurs
2.5 Statistics starting at around P11. By P15 around 80% of the
rd1 photoreceptors had been lost (Fig. 2a). We
Data were analyzed using Microsoft Excel and followed the temporal changes in lactate release
GraphPad Prism 9.1. Values are given as in culture media from rd1 and WT retina over a
mean ± SD. The data were compared using two-­ period of 6 days, from P9 to P15 (Fig. 2b). Retina
way ANOVA and subsequent Tukey’s multiple from rd1 mice displayed an overall increased
comparison tests. Significance levels as indicated release of lactate compared to WT retina
by asterisks were *p < 0.05; **p < 0.01, and (p < 0.001). Noticeably, a considerably higher
***p < 0.001. release rate was observed from rd1 mice during
the first 2 days (P9–P11) of culturing. Later on,
432 Y. Chen et al.

Fig. 1 Release of lactate from wild-type retina into the lected in triplicate for the lactate assay kit and in five rep-
R16 culture medium. The line graphs depict extracellular licates for the 1H-NMR spectroscopy analysis of
lactate levels in the organotypic retinal explant culture extracellular lactate. Data points indicate mean values and
medium measured by lactate assay kit (a) and 1H-NMR standard deviation; statistical analysis was performed
spectroscopy (b). Retinal explants were cultured from P9 using one-way ANOVA and Tukey’s multiple comparison
to P15 in complete R16 medium (CM). The CM was col- test

Fig. 2 Comparison of lactate release from wild-type and pared to aged-matched WT. (b) Lactate release from WT
rd1 mouse retina. (a) Histological cross section of retinal and rd1 retina explant cultures was measured by lactate
tissues from rd1 and wild-type (WT) mice. From P11 to assay kit. Data points shown are mean values and standard
P15, rd1 mice went through a rapid degeneration of pho- deviation for n = 3. Statistical analysis was performed
toreceptors such that, at P15, the outer nuclear layer using two-way ANOVA and Sidak’s multiple comparison
(ONL) was markedly thinner than at P11 or when com- test

from P13 to P15, lactate release rate slowed even higher than that of WT. Since the retina is
down, yet the overall production was still higher exceptional in its abundant use of aerobic gly-
than in WT. colysis, measuring lactate release provides a
unique opportunity to deduce retinal metabolic
activity.
4 Discussion Even though the fact that the retina releases
enormous amounts of lactate in the presence of
In this study, we explored the extracellular release oxygen was discovered already 100 years ago
of lactate from organotypic retinal explant cul- [5], the precise reasons for this phenomenon are
tures. We used a conventional enzymatic lactate still not completely understood. Moreover, it is
assay and 1H-NMR spectroscopy-based detection not clear which retinal cell type is responsible for
to show that WT retina releases large amounts of aerobic glycolysis and lactate production. While
lactate into the surrounding medium. Remarkably, photoreceptors harbor large numbers of mito-
lactate release from degenerating rd1 retina was chondria in their inner segments – suggesting that
Measuring the Release of Lactate from Wild-Type and rd1 Mouse Retina 433

they should perform oxidative phosphorylation – calibration (e.g., by the ERETIC approach [17])
a recent study indicated that photoreceptors were to fully quantify also NMR data.
using mostly glycolysis to satisfy their large In conclusion, we showed two reliable meth-
energy demands [14]. In explant cultures the ods for measuring extracellular lactate in an in
strong excretion of lactate between P9 and P15 vitro organotypic retinal culture system. This cul-
may at least partially be related to high develop- ture system could be extremely useful in further
mental requirements. The fact that rd1 retinal retinal energy metabolism studies as it allows
explants release approximately twice as much manipulating the retina under entirely controlled
lactate compared to WT retina suggests a higher conditions. Given the simplicity, stability, and
glycolytic rate in the mutant. This in turn could cost-effectiveness of lactate measurement, extra-
mean that dying rd1 photoreceptors have a cellular lactate can be a readily quantifiable read-
strongly increased energy demand and could help out for evaluating cellular metabolic changes
to explain their rapid degeneration phenotype. even in a particular disease condition.
The relative decrease of lactate release observed
in rd1 retina from P13 to P15 may then be due to Acknowledgments We are grateful for excellent techni-
the fact that by this time most photoreceptor cells cal support from Norman Rieger. This study was sup-
ported by grants from the Tistou and Charlotte Kerstan
have already been lost. Foundation and the Chinese Scholarship Council. This
Compared to other metabolites, lactate is work was supported by Prof. Dr. Bernd J. Pichler and
abundant, stable, and can be easily detected [15]. Werner Siemens Foundation for the access to the NMR
It has been reported that lactate is 10–50 times spectroscopy equipment.
more plentiful than pyruvate in cells, serving as
bridge between glycolysis and mitochondria
[16]. With its long and extensive studied history, References
lactate is an attractive surrogate marker for study-
1. Ames A 3rd, Li YY, Heher EC, Kimble CR. Energy
ing retinal energy metabolism. At a systemic metabolism of rabbit retina as related to function: high
level, lactate measurement has been long-­ cost of Na+ transport. J Neurosci. 1992;12(3):840–53.
established in clinical practice as a blood marker 2. Wong-Riley MT. Energy metabolism of the visual
of tissue hypoxia. The significant difference system. Eye Brain. 2010;2:99–116.
3. Dowling JE. The retina: an approachable part of the
observed in our study between WT and rd1 retina brain. Harvard University Press; 1987.
indicates that also in RD patient blood samples, 4. Okawa H, Sampath AP, Laughlin SB, Fain GL. ATP
lactate measurements may allow detecting and consumption by mammalian rod photoreceptors in
monitoring retinal disease state. darkness and in light. Curr Biol. 2008;18(24):1917–21.
5. Warburg O. The metabolism of carcinoma cells. J
Our study confirmed the validity and reliabil- Cancer Res. 1925;9(1):148–63.
ity of both lactate kit and 1H-NMR spectroscopy-­ 6. Petit L, Ma S, Cipi J, Cheng SY, Zieger M, Hay N,
based measurements. Moreover, our data et al. Aerobic glycolysis is essential for normal rod
indicated lactate as a stable and reliable readout function and controls secondary cone death in retinitis
pigmentosa. Cell Rep. 2018;23(9):2629–42.
when comparing to other metabolites, such as 7. Vander Heiden MG, Cantley LC, Thompson
ATP. Besides the commercial availability, the CB. Understanding the Warburg effect: the meta-
assay kit requires no additional time-consuming bolic requirements of cell proliferation. Science.
steps. The divergence between the two methods 2009;324(5930):1029–33.
8. Adler AJ, Southwick RE. Distribution of Glucose
could result from limits in sensitivity and vari- and Lactate in the Interphotoreceptor Matrix.
ability of the enzyme activity-based colorimetric Ophthalmic Research. 1992;24(4):243–252. https://
approach. However, when attempting to quantify doi.org/10.1159/000267174
more than one specific metabolite, the NMR 9. Arango-Gonzalez B, Trifunović D, Sahaboglu A,
Kranz K, Michalakis S, Farinelli P, et al. Identification
spectroscopy-based approach can give a full view of a common non-apoptotic cell death mecha-
on the present metabolic readout. Furthermore, nism in hereditary retinal degeneration. PLoS One.
future NMR assays should incorporate external 2014;9(11):e112142.
434 Y. Chen et al.

10. Sanyal S, Bal AK. Comparative light and electron 15. Seymour CW, Carlbom D, Cooke CR, Watkins TR,
microscopic study of retinal histogenesis in normal Bulger EM, Rea TD, et al. Temperature and time sta-
and rd mutant mice. Z Anat Entwicklungsgesch. bility of whole blood lactate: implications for feasi-
1973;142(2):219–38. bility of pre-hospital measurement. BMC Res Notes.
11. Caffe AR, Ahuja P, Holmqvist B, Azadi S, Forsell 2011;4(1):169.
J, Holmqvist I, et al. Mouse retina explants after 16. Brooks GA. The science and translation of lactate
long-term culture in serum free medium. J Chem shuttle theory. Cell Metab. 2018;27(4):757–85.
Neuroanat. 2001;22(4):263–73. 17. Akoka S, Barantin L, Trierweiler M. Concentration
12. Sahaboglu A, Tanimoto N, Bolz S, Garrido MG, measurement by proton NMR using the ERETIC
Ueffing M, Seeliger MW, et al. Knockout of method. Anal Chem. 1999;71(13):2554–7.
PARG110 confers resistance to cGMP-induced tox-
icity in mammalian photoreceptors. Cell Death Dis.
2014;5(5):e1234-e.
13. Belhadj S, Tolone A, Christensen G, Das S, Chen Y,
Paquet-Durand F. Long-term, serum-free cultivation
of organotypic mouse retina explants with intact reti-
nal pigment epithelium. J Vis Exp. 2020;165 https://
doi.org/10.3791/61868.
14. Kanow MA, Giarmarco MM, Jankowski CS, Tsantilas
K, Engel AL, Du J, et al. Biochemical adaptations
of the retina and retinal pigment epithelium support
a metabolic ecosystem in the vertebrate eye. elife.
2017;6:e28899.
Aerobic Glycolysis
in Photoreceptors Supports
Energy Demand in the Absence
of Mitochondrial Coupling

Daniel T. Hass, Celia M. Bisbach, Martin Sadilek,


Ian R. Sweet, and James B. Hurley

Abstract Keywords

Metabolism is adapted to meet energetic Photoreceptors · Metabolic flux · Aerobic


needs. Based on the amount of ATP required glycolysis · Mitochondria · Uncoupling ·
to maintain plasma membrane potential, pho- Retina
toreceptor energy demands must be high. The
available evidence suggests that photorecep-
tors primarily generate metabolic energy
through aerobic glycolysis, though this evi- Abbreviations
dence is based primarily on protein expression
and not measurement of metabolic flux. ATP Adenosine triphosphate
Aerobic glycolysis can be validated by mea- GC-MS Gas chromatography-mass
suring flux of glucose to lactate. Aerobic gly- spectrometry
colysis is also inefficient and thus an GCR Glucose consumption rate
unexpected adaptation for photoreceptors to HK2 Hexokinase 2
make. We measured metabolic rates to deter- LPR Lactate production rate
mine the energy-generating pathways that NADH Nicotinamide adenine dinucleotide
support photoreceptor metabolism. We found NADPH Nicotinamide adenine dinucleotide
that photoreceptors indeed perform aerobic phosphate
glycolysis and this is associated with mito- OCR Oxygen consumption rate
chondrial uncoupling. PKM2 Pyruvate kinase M2

1 Introduction
D. T. Hass (*) · C. M. Bisbach · J. B. Hurley
Department of Biochemistry, University of
Washington, Seattle, WA, USA When glucose enters cells, glycolysis breaks it
e-mail: dhass@uw.edu down to pyruvate. In mitochondria pyruvate can
M. Sadilek be oxidized to CO2, and in the cytosol it is
Department of Chemistry, University of Washington, reduced to lactate in a process called aerobic
Seattle, WA, USA glycolysis [1]. Oxidation in mitochondria yields
I. R. Sweet approximately 16 times more ATP per glucose
UW Medicine - Diabetes Institute, University of than conversion to lactate. Energy demand is
Washington, Seattle, WA, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 435
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_64
436 D. T. Hass et al.

high in the retina and it relies on glucose to sur- cervical dislocation and dissected retinas in
vive [2, 3]. Mitochondrial pyruvate oxidation is a Hank’s buffered salt solution.
more efficient way to meet energy demands, yet
at physioxia most of energy in the retina instead
comes from aerobic glycolysis [4]. 2.2 Ex Vivo Incubations
Hexokinase 2 (HK2) and pyruvate kinase M2
(PKM2) are associated with aerobic glycolysis in Following dissection, retinas were placed in
cancer, and both are expressed in photoreceptors, Krebs-Ringer bicarbonate buffer (KRB; 98.5 mM
suggesting that rods and cones are the major sites NaCl, 4.9 mM KCl, 1.2 mM KH2PO4 1.2 mM
of aerobic glycolysis [5–7]. However when these MgSO4-7H2O, 20 mM HEPES, 2.6 mM CaCl-­
genes are knocked out, aerobic lactate production 2H2O, 25.9 mM NaHCO3) with 5 mM glucose, at
decreases at most by 25% [8, 9]. If the associa- 37 °C and 95% air/5% CO2. Extracellular flux
tion between HK2, PKM2, and aerobic glycoly- was measured in 5 mM unlabeled glucose, in
sis is not causal, photoreceptor expression of which tissues were incubated for 90 min at 37 °C
these enzymes is not sufficient evidence that they and 5% CO2 (media sampling times are indicated
are the source of aerobic glycolysis in the retina. in figures). Intracellular fluxes were measured in
This study tests whether photoreceptors are tissue incubated in KRB with 5 mM 4-2H glucose
sites of aerobic lactate production. We approach at 37 °C and 95% air/5% CO2.
this goal by comparing glycolytic flux and O2
consumption (OCR) in retinas from C57BL/6J
(B6) mice and C57BL/6J; AIPL1−/− mice 2.3 Glucose Assay
(AIPL1−/−). AIPL1−/− mice are model of Leber’s
congenital amaurosis, in which photoreceptors We measured media glucose with a coupled
degenerate by 1 month of age [10]. By contrast- enzymatic assay wherein glucose phosphoryla-
ing metabolism in retinas with or without photo- tion and oxidation is coupled to NADP+ reduc-
receptors, we find that photoreceptors are a major tion [11]. NADPH absorbs light at 340 nm. We
contributor to aerobic glycolysis but not the incubated samples and 0–7 mM standards in the
exclusive source of lactate. We also find that pho- assay buffer: 50 mM Tris, 1 mM MgCl2, 500 μM
toreceptor aerobic glycolysis is associated with NADP+, 500 μM ATP, 0.2 U/mL hexokinase,
mitochondrial uncoupling. Uncoupling is a sign 0.08 U/mL glucose-6-phosphate dehydrogenase,
of dysfunction in many cells, but in photorecep- pH 8.1. We measured A340 over time until A340
tors it may be a normal mode of metabolism. reached steady state using a Bio-Tek Synergy 4
plate-reader. Other measures of glucose were
performed using an Accu-Chek Performa blood
2 Materials and Methods glucometer (Roche).

2.1 Animals and Euthanasia


2.4 Lactate Assay
This study was in accordance with National
Research Council’s Guide for the Care and Use We measured media lactate with a coupled
of Laboratory Animals (8th ed). All protocols enzymatic assay where lactate dehydrogenase
were approved by the UW IACUC. We used converts lactate and NAD+ to pyruvate and
2–4 month-old male and female B6 and AIPL1−/− NADH [11]. Pyruvate is consumed by the assay
mice. Mice were housed at 25 °C, with a 12-h buffer, drawing the reaction to completion.
light cycle and ad libitum access to water and NADH absorbs light at 340 nm. We incubated
normal rodent chow. Immediately prior to mea- equal volumes of samples and 0–7 mM standards
sures of metabolic flux, we euthanized mice via in assay buffer (300 mM glycine, 166 mM
Aerobic Glycolysis in Photoreceptors Supports Energy Demand in the Absence of Mitochondrial Coupling 437

hydrazine, 2.5 mM NAD+, 8 U/mL lactate 2.7 Measurement of O2


dehydrogenase) and determined A340 with a Bio- Consumption Rate (OCR)
Tek Synergy 4 plate-reader. Steady-state A340 was
used standards to determine media [lactate]. We determined OCR in a perifusion flow-­culture
system [13]. Tissues were placed in chambers
between Cytopore beads (Amersham
2.5 Photoreceptor Contributions Biosciences) and porous frits and then perifused
to Retina Metabolism in KRB with 5 mM glucose, 1x Antibiotic-­
Antimycotic, 1 mg/mL fatty acid-free albumin,
LPR: Lactate Production Rate, GCR: Glucose 21% O2, 5% CO2, and 74% N2. Media was
Consumption Rate passed through a bubble trap before exposure to
LPRphotoreceptors = LPRC57BL6/J – LPR AIPL1 tissues. Outflow media was in contact with a
GCRphotoreceptors = GCRC57BL6/J – GCR AIPL1 glass wall coated with a thin layer of oxygen-
Aerobic glycolysis = LPR/GCR sensitive polymerized Pt(II) Meso-
tetra(pentafluorophenyl)porphine dye (Frontier
Note: The maximum molar ratio of lactate to Scientific). Following a 405 nm light pulse, the
glucose is 2, which is thus maximum possible dye-coated glass emits a phosphorescent signal
value for LPR/GCR. We omitted values >2 and detected at 650 nm. The decay lifetime is depen-
negative values of LPR/GCR because these dent on oxygen tension. The media flow rate and
results are likely consequences of technical the quantitative relationship between dye phos-
imprecision in data collection (pipetting errors or phorescent decay and oxygen concentration
the influence of measurements taken near the were used to determine OCR.
assay’s limit of detection). The statistical signifi-
cance of the result is maintained regardless of
whether we drop either or both of these exclusion 2.8 Statistical Analysis
criteria.
Data were graphed and analyzed in Prism v9.0
(GraphPad Software). We set the significance
2.6 Metabolite Extraction and Gas threshold at p < 0.05. Displayed p-values repre-
Chromatography-Mass sent the result of a parametric two-tailed student’s
Spectrometry t-test. To compare fluxes we performed linear
regressions of the data and compared slopes with
4-2H-glucose flux was determined using gas an extra-sum-of-squares F-test.
chromatography-mass spectrometry (GC-MS).
We extracted metabolites with 10 μM methylsuc-
cinate in 80% methanol. We derivatized metabo- 3 Results
lites from single retinas in 20 mg/mL
methoxyamine in pyridine (10 μL) and with tert-­ We compared glucose consumption (GCR) and
butyl dimethylsilyl-N-methyltrifluoroacetamide lactate production rates (LPR) in retina explants
(10 μL). Samples were analyzed using an Agilent freshly from B6 and AIPL1−/− mice in 5 mM glu-
7890/5975C GC-MS system operated in selec- cose and 21% O2. Retinas lacking photoreceptors
tive ion monitoring mode to detect ions specific consumed glucose 59% and produced lactate 65%
to metabolites of interest eluting at specific reten- more slowly than retinas with photoreceptors
tion times. Extracted ion chromatogram peak (Fig. 1a, b). This suggests photoreceptors consume
areas were used to quantify the metabolite con- the most glucose and produce the most lactate.
centration utilizing a calibration curve generated Rather than an “all or none” contribution of photo-
for analytes of interest. We corrected for natural receptors there was residual lactate production in
isotopologue abundance using IsoCor [12].
438 D. T. Hass et al.

AIPL1−/− retinas, suggesting that other cells also consumption (OCR). We determined whether
contribute to aerobic glycolysis (Fig. 1b). OCR in photoreceptor mitochondria generates
We quantified aerobic glycolysis in B6 and ATP by measuring mitochondrial “uncoupling”
AIPL1−/− retinas by normalizing the LPR to (residual respiration when ATP synthesis in
GCR. This normalization accounts for differ- inhibited). Oligomycin inhibits ATP synthesis.
ences in retina tissue amount. At most a glucose Uncoupling (OCRoligomycin/OCRbasal) is the
molecule can make two lactate molecules. The reciprocal of the respiratory control ratio, a
LPR/GCR, or “Lactate/Glucose,” is lower in common metric of mitochondrial function [16].
AIPL1−/− than B6 retinas, indicating photorecep- Oligomycin completely inhibits respiration in
tors contribute to aerobic glycolysis (Fig. 1c). fully coupled mitochondria, and yields a “%
Assuming the difference between glucose con- uncoupling” of 0. In AIPL1−/− retinas, oligomycin
sumption and lactate production between B6 and inhibits 65% of OCR, but in B6 retinas it inhibits
AIPL1−/− retinas represents photoreceptors, the only 45%, showing that mitochondria in B6
“photoreceptor” ratio of lactate/glucose is retinas are more uncoupled. We subtracted
1.9 ± 0.2, nearing the theoretical maximum molar AIPL1−/− retina OCR from B6 OCR to estimate
ratio of 2. the effect on photoreceptor mitochondria. The
We next measured intracellular flux of 2H difference curve (blue) suggests that oligomycin
from 4-2H-glucose. 2H from glucose travels with has little effect on photoreceptor mitochondria and
glucose breakdown products in glycolysis until that that electron transport in photoreceptor
the glyceraldehyde-3-phosphate dehydrogenase mitochondria is mostly uncoupled. Thus, aerobic
reaction; it is transferred to NAD+ to make glycolysis and lactate production may be espe-
NAD2H. 2H then is transferred either to lactate by cially active in photoreceptors to compensate for
lactate dehydrogenase or to malate by malate poor mitochondrial ATP synthesis.
dehydrogenase (Fig. 1d) [14]. The rate of 2H
accumulation on fractional lactate labeling is
63% lower in AIPL1−/− retinas (Fig. 1e). This 4 Discussion
change in 2H labeling could result from a decrease
in the proportion of glucose molecules converted Predicted energetic requirements for
to lactate, in glucose consumption, or both. If photoreceptors suggest that retinas lacking rods
glucose consumption is slowed in AIPL1−/− reti- should require less metabolic energy than retinas
nas, 2H accumulation on malate would be slowed with rods [3]. Glycolytic differences between
to 63% of control. 2H-accumulation rate on rod-­sufficient (B6) and rod-deficient (AIPL1−/−)
malate is only 44% lower in AIPL1−/− retinas retinas confirm this. Our findings suggest
(Fig. 1f). Since 2H-lactate production is decreased photoreceptors convert a high proportion of glu-
more dramatically than 2H-malate production, cose to lactate, a sign of aerobic glycolysis.
the proportion of glucose converted to lactate We also show that photoreceptor mitochondria
must be less in retinas lacking photoreceptors. are uncoupled and do not generate substantial
Independent measurements of extracellular ATP (Fig. 1g, h). These OCR measurements
and intracellular 2H flux lead to the same conclu- replicate our earlier findings [17], and since that
sions: AIPL1−/− retinas consume less glucose report we learned that retinas synthesize and
than normal retinas, and for every glucose mole- export succinate [18]. Succinate release sacri-
cule that is oxidized, less becomes lactate. This fices a source of reducing power mitochondria
strongly suggests that photoreceptors are the could use if they were coupled, and is consistent
main site for aerobic glycolysis in the retina. with mitochondrial uncoupling in photorecep-
Anaerobic glycolysis occurs when tors. Our combined data suggest that since mito-
mitochondria do not generate sufficient ATP chondria are uncoupled, aerobic glycolysis may
from mitochondrial activity [15]. This happens be the only remaining metabolic pathway to
when ATP synthesis is not coupled to O2
Aerobic Glycolysis in Photoreceptors Supports Energy Demand in the Absence of Mitochondrial Coupling 439

A B C 0.0059
1500 1500 2.0

Lactate / Glucose
1.5
Glucose (nmol)

Lactate (nmol)
1000 1000

1.0

500 500
C57BL6/J 0.5
AIPL1-/-
0 0 0.0
0 20 40 60 80 100 0 20 40 60 80 100

-/-
J
L6

L1
Time (mintues) Time (mintues)

IP
57

A
C
D E F
15 C57BL6/J 15

% m1 Lactate AIPL1-/-

% m1 Malate
10 10

5 5

0 0
0 20 40 60 80 100 120 0 20 40 60 80 100 120
Time (seconds) Time (seconds)

G H
Oligomycin Antimycin A KCN
3.0 1.0
OCR (nmol O2/min/retina)

(Oligomycin / Basal OCR)

C57BL6/J
2.5 0.8
AIPL1-/- 0.0026
Uncoupling

2.0
"Photoreceptors" 0.6
1.5
0.4
1.0

0.5 0.2

0.0 0.0
0 50 100 150
-/-
/J

L1
L6

Time (min)
IP
B
57

A
C

Fig. 1 Glycolytic and oxidative metabolism in the incubated in 5 mM 4-2H glucose. (g) Retina OCR before
C57BL/6J and C57BL/6J; AIPL1−/− retina. (a) Glucose and after treatment with 30 μM oligomycin, to isolate
depletion and (b) lactate production in retinas iso- uncoupled respiration and 1 μM antimycin A and 3 mM
lated from C57BL/6J and AIPL1−/− mice (n = 5–7). (c) KCN, to fully inhibit mitochondrial respiration (n = 4).
Lactate production rate over glucose consumption rate in The mean difference of these OCR traces is in blue. (h)
a larger data set (n = 11–18). (d) Scheme for 2H labeling Ratio of mitochondrial respiration with oligomycin to
of metabolites, created on BioRender.com. (e) 2H labeling basal mitochondrial respiration with glucose. Error bars:
of lactate and (f) malate in C57BL/6J and AIPL1−/− retinas SEM. Blue dotted lines are estimates of photoreceptor-­
specific metabolism (c, h)

make sufficient ATP to meet photoreceptor be the sole cause of metabolic differences
energetic needs (Fig. 2). between B6 and AIPL1−/− retinas. There are
There is a critical caveat to the interpretation structural differences in AIPL1−/− retina neurons,
of our findings. A lack of photoreceptors may not and glia become more “activated,” or “reactive”
440 D. T. Hass et al.

Fig. 2 Model of the relationship between mitochondrial from NADH to H2O does not fuel ATP synthesis because
uncoupling and aerobic glycolysis in photoreceptors. To of H+ leak. This prevents ATP synthesis. Mitochondria do
meet ATP demand, glucose enters photoreceptors and not generate sufficient ATP for rods to meet ATP demand,
generates ATP by glycolysis. A portion of pyruvate from so pyruvate from glycolysis becomes lactate and is
glycolysis enters mitochondria and is oxidized by the exported. This allows rapid glycolysis to generate suffi-
TCA cycle. This generates NADH, but electron transport cient ATP. (Created with BioRender.com)

2. Winkler BS. Glycolytic and oxidative metabolism


[10]. Transcriptomic analysis of reactive Müller in relation to retinal function. J Gen Physiol.
glia shows increased expression of glycolytic 1981;77(6):667–92.
genes [19], and activated microglia are more gly- 3. Okawa H, Sampath AP, Laughlin SB, Fain GL. ATP
consumption by mammalian rod photoreceptors in
colytic in culture [20, 21]. However, if Müller darkness and in light. Curr Biol. 2008;18(24):1917–21.
glial or microglial glycolysis increases in 4. Ames A, Li Y-Y, Heher EC, Kimble CR. Energy
AIPL1−/− mice, it is insufficient to mask the more metabolism of rabbit retina as related to function: high
dominant decrease in glucose use and decrease in cost of Na+ transport. J Neurosci. 1992;12(3):940–53.
5. Lindsay KJ, Du J, Sloat SR, Contreras L, Linton
lactate/glucose ratio. Increased glycolysis associ- JD, Turner SJ, et al. Pyruvate kinase and aspartate-­
ated with gliosis may account for residual lactate glutamate carrier distributions reveal key metabolic
production we observe from AIPL1−/− retinas links between neurons and glia in retina. Proc Natl
(Fig. 1b). Acad Sci U S A. 2014;111(43):15579.
6. Ferguson EC, Rathmell JC. New roles for pyruvate
kinase M2: working out the Warburg effect. Trends
Biochem Sci. 2008;33(8):359–62.
References 7. Wolf A, Agnihotri S, Micallef J, Mukherjee J, Sabha
N, Cairns R, et al. Hexokinase 2 is a key mediator
1. Warburg O. The metabolism of carcinoma cells. J of aerobic glycolysis and promotes tumor growth
Cancer Res. 1925;9(1):148–63. in human glioblastoma multiforme. J Exp Med.
2011;208(2):313–26.
Aerobic Glycolysis in Photoreceptors Supports Energy Demand in the Absence of Mitochondrial Coupling 441

8. Petit L, Ma S, Cipi J, Cheng S-Y, Zieger M, Hay N, 15. Pasteur L. Expériences et vues nouvelles sur la nature
et al. Aerobic glycolysis is essential for normal rod des fermentations. Comptes rendus l’Académie des
function and controls secondary cone death in retinitis Sci. 1861;52:1260–4.
pigmentosa. Cell Rep. 2018;23(9):2629. 16. Brand M, Nicholls D. Assessing mitochondrial
9. Chinchore Y, Begaj T, Wu D, Drokhlyansky E, dysfunction in cells. Biochem J. 2011;435(2):297–312.
Cepko C. Glycolytic reliance promotes anabolism in 17. Du J, Rountree A, Cleghorn WM, Contreras L,
photoreceptors. elife. 2017;6:e25946. Lindsay KJ, Sadilek M, et al. Phototransduction
10. Singh RK, Kolandaivelu S, Ramamurthy V. Early influences metabolic flux and nucleotide metabolism
alteration of retinal neurons in Aipl1−/− animals. in mouse retina. J Biol Chem. 2016;291(9):4698–710.
Invest Ophthalmol Vis Sci. 2014;55(5):3081–92. 18. Bisbach CM, Hass DT, Robbings BM, Rountree AM,
11. Lowry OH, Passonneau JV. A flexible system of Sadilek M, Sweet IR, et al. Succinate can shuttle
enzymatic analysis. Elsevier; 1972. reducing power from the hypoxic retina to the O2-rich
12. Millard P, Delépine B, Guionnet M, Heuillet M, pigment epithelium. Cell Rep. 2020;31(5):107606.
Bellvert F, Létisse F. IsoCor: isotope correction 19. Roesch K, Stadler MB, Cepko CL. Gene expression
for high-resolution MS labeling experiments. changes within Müller glial cells in retinitis
Bioinformatics. 2019;35(21):4484–7. pigmentosa. Mol Vis. 2012;18:1197.
13. Neal A, Rountree A, Philips C, Kavanagh T, Williams 20. Scholz R, Sobotka M, Caramoy A, Stempfl T, Moehle
DP, Newham P, et al. Quantification of low-level drug C, Langmann T. Minocycline counter-regulates pro-­
effects using real-time, in vitro measurement of oxygen inflammatory microglia responses in the retina and
consumption rate. Toxicol Sci. 2015;148(2):594–602. protects from degeneration. J Neuroinflammation.
14. Lewis CA, Parker SJ, Fiske BP, Mccloskey D, Gui DY, 2015;12(1):1–14.
Green CR, et al. Article tracing compartmentalized 21. Wang L, Pavlou S, Du X, Bhuckory M, Xu H, Chen
NADPH metabolism in the cytosol and mitochondria M. Glucose transporter 1 critically controls microglial
of mammalian cells. Mol Cell. 2014;55:253–63. activation through facilitating glycolysis. Mol
Neurodegener. 2019;14(1):1–15.
Redox Status in Retinitis Pigment
osa

L. Olivares-González, S. Velasco, I. Campillo,


J. M. Millán, and R. Rodrigo

Abstract cytokines released by the activated immune


cells (e.g., macrophages or microglia). The
Retinitis pigmentosa (RP) is the most role of oxidative stress is very important in
common form of inherited retinal dystrophy the retina. Rods are the main consumers of
characterized by the progressive loss of oxygen (O2), so they are constantly exposed
vision. It is a rare disease. Despite being a to oxidative stress and lipid peroxidation.
genetic disease, its progression is influenced According to the oxidative hypothesis, after
by oxidative damage and chemokines and rod death in the early stages of the disease,
O2 would accumulate in large quantities in
the retina, producing hyperoxia and favoring
L. Olivares-González · S. Velasco · I. Campillo
Pathophysiology and Therapies for Vision Disorders, the accumulation of reactive oxygen species
Principe Felipe Research Center (CIPF), and reactive nitrogen species that would
Valencia, Spain cause oxidative damage to lipids, proteins,
Joint Unit on Rare Diseases CIPF-La Fe, and DNA, exacerbating the process of retinal
Valencia, Spain degeneration. Evidence shows alterations in
e-mail: lolivares@cipf.es; svelasco@cipf.es;
the antioxidant-oxidant state in patients and
icampillo@cipf.es
in animal models of RP. In recent years,
J. M. Millán
therapeutic approaches aimed at reducing
Joint Unit on Rare Diseases CIPF-La Fe,
Valencia, Spain oxidative stress have emerged as useful
therapies to slow down the progression of
Rare Diseases Networking Biomedical Research
Centre (CIBERER), Madrid, Spain RP. We focus this review on oxidative stress
and its relationship with the progression of
Molecular, Cellular and Genomic Biomedicine,
Health Research Institute La Fe, Valencia, Spain RP.
e-mail: millan_jos@gva.es
R. Rodrigo (*) Keywords
Pathophysiology and Therapies for Vision Disorders,
Principe Felipe Research Center (CIPF), Retinitis pigmentosa · Hyperoxia · Oxidative
Valencia, Spain
damage · Antioxidant response ·
Joint Unit on Rare Diseases CIPF-La Fe, Inflammation · Cell death
Valencia, Spain
Rare Diseases Networking Biomedical Research
Centre (CIBERER), Madrid, Spain
e-mail: rrodrigo@cipf.es

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 443
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_65
444 L. Olivares-González et al.

Abbreviations cium accumulation or endoplasmic reticulum


stress, etc. [6–9].
DHA docosahexaenoic acid
HIF1 hypoxia-inducible factor 1
IL interleukins 2 Oxidative Stress
IRDs inherited retinal dystrophies
NUT nutraceuticals Oxidative stress occurs because of an alteration
O2 oxygen between the formation of reactive oxygen (ROS)
ROS reactive oxygen species and nitrogen species and the ability of the anti-
RP retinitis pigmentosa oxidant systems to eliminate them [10].
TNFα tumor necrosis factor alpha Retinal photoreceptor cells are metabolically
very active cells with a weak antioxidant system,
so they are very sensitive to oxidative damage
1 Introduction and to changes in O2 concentration [11–13]. Rods
make up 95% of the photoreceptor cells. Rod
Inherited retinal dystrophies (IRDs) comprise a death in the early stages of RP could lead to a
heterogeneous group of diseases that lead to pro- decrease in O2 consumption that reaches the ret-
gressive loss of vision [1]. IRDs are mainly ina (oxidative hypothesis). This would cause a
caused by dysfunction or loss of photoreceptor situation of retinal hyperoxia that would produce
cells (cones and rods) affecting 1 in 2000–3000 oxidative damage including ROS generation;
individuals [2]. IRDs can affect rods, cones, and oxidation of macromolecules such as lipids
both, thus classifying themselves as rod-­ (malondialdehyde), proteins (protein carbonyl-
dominated dystrophies, cone-dominated dystro- ation), and nucleic acids (8-oxoguanine, 8-oxo-
phies, and generalized dystrophies involving ­dG); or the deregulation of antioxidant effectors
both rods and cones [3]. They are clinically and (antioxidant enzymes) contributing to the cone
genetically heterogeneous. To date, more than degeneration observed in RP models [2, 14–18]
300 genes are known, and different mutations in (Fig. 1).
the same gene can lead to different phenotypes Inducible hypoxia factor alpha (HIF1α) is a
[4]. IRDs include retinitis pigmentosa (RP), hypoxic signaling protein that regulates many
Leber’s congenital amaurosis, Stargardt’s macu- genes under conditions of low O2 concentration.
lar dystrophy, and choroideremia [5]. It appears to be important in O2-dependent retinal
RP is the most common IRD. It is a rare diseases [19, 20]. We previously observed that
disease characterized by the progressive loss of HIF1α protein and its HIF1α target genes vascu-
rods and cones, with a prevalence of 1 in 4000 lar endothelial growth factor, glucose transporter
individuals [5]. RP patients lose night and 1, and endothelin-1 were decreased in rd10 mice
peripheral vision in the early stages of the dis- across the degeneration suggesting a hyperoxic
ease, due to rod death by genetic mutations. In situation in the retina. Stabilization of HIF1α by
advanced stages of the disease, cone death dimethyloxalylglycine increased gene expression
occurs resulting in loss of central and color of HIF1α target, reduced the photoreceptor cell
vision. Rod death causes the release of different loss and microglia activation, and ameliorated
substances to the extracellular environment as the antioxidant response in rd10 mice [16].
free radicals or inflammatory molecules, which Supporting this hypothesis and an altered
would generate a toxic microenvironment that redox status, oxidative stress markers and reduced
may affect cone survival, exacerbating the pro- antioxidant response have been observed in bio-
cess of degeneration [2, 6]. Several factors seem logical samples of RP patients [15, 21, 22]. In
to participate in the progression of IRDs includ- response to these changes, vascular pruning and
ing inflammation, oxidative stress, guanosine vessel attenuation would occur promoting cone
monophosphate deregulation, intracellular cal- death and a situation of hypoxia in the inner ret-
Redox Status in Retinitis Pigmentosa 445

Fig. 1 Schematic representation of the oxidative stress processes underlying photoreceptor degeneration in retinitis
pigmentosa

ina [17, 23]. In this sense, changes in the concen- (DHA), lutein, zeaxanthin, progesterone, and
tration and distribution of O2 across different RP lipoic acid have been selected for this purpose.
stages have been also detected in the retina of In recent years, several studies have documented
animal models [18, 24, 25] and RP patients [26, the beneficial effects of these compounds by
27], reflecting a gradual loss of O2 metabolism. reducing cell death and improving retinal func-
tion in animal models of IRDs [30, 31].
Supplementation with vitamins A and E has
3 Antioxidant Therapies been assayed in RP [32, 33]. However, it seems
with Nutraceuticals that only the combination of vitamin A plus
DHA present some beneficial effect on RP pro-
Several antioxidant strategies have been carried gression [34]. Lutein and zeaxanthin are macu-
out to reduce oxidative damage during IRD pro- lar pigments acting to protect cells from
gression. In particular, the oral supplementation oxidative damage, and death. They also improve
with substances called nutraceuticals seems to photoreceptor function, probably by reducing
be a very promising treatment. Functional or endoplasmic reticulum oxidative stress in ani-
nutraceutical foods are natural substances that mal models of IRDs [30, 35, 36]. Progesterone
exhibit antioxidant or/and anti-inflammatory is capable of preventing photoreceptor cell loss
properties. They have been already used as puta- and reactive gliosis maybe by reducing lipid
tive therapeutic strategies against different reti- peroxidation in rd1 and rd10 mice, two RP mod-
nal diseases [16, 22, 28, 29]. Compounds such els [37, 38]. Other compounds such as lipoic
as vitamin A, vitamin E, docosahexaenoic acid acid and curcumin can also reduce oxidative
446 L. Olivares-González et al.

stress and promote cell survival in RP models and DNA derived from the oxidative stress gener-
[23, 39, 40]. ated after rod death can contribute to activate
Recently, it has been described the oral microglia, and trigger the inflammatory process,
supplementation with a formulation of which, if prolonged over time, may also contrib-
nutraceuticals containing folic acid, vitamin A, ute to the process of retinal degeneration [43, 44].
vitamin B6, copper, zinc, selenium, lutein, and Despite the advances in the knowledge of the
zeaxanthin (called NUT), improved retinal redox, mechanisms involved in photoreceptor cell death,
photoreceptor survival, and visual function in the great diversity of the findings shows the com-
rd10 mice [41]. In addition, NUT also showed plexity of IRDs. However, evidence suggests that
anti-­inflammatory effects by decreasing microglia both oxidative stress and inflammation are
activation, reactive gliosis, and cytokines (IL-6, involved in the progression of IRDs. Strategies to
IL-1β, or TNFα) [41]. target oxidative stress or inflammation could
slow down, at least in part, the progression of
IRDs regardless of the genetic defect. Therefore,
4 Oxidative Stress the design of combined strategies to deal with
and Inflammation both processes could be an interesting approach.

Oxidative stress and inflammation are closely


related pathophysiological processes. Both cel- References
lular processes contribute to the pathogenesis of
RP. During the progression of RP, a chronic and 1. Hartong DT, Berson EL, Dryja TP. Retinitis
pigmentosa. Lancet (London, Engl).
sustained inflammation has been observed in ani- 2006;368(9549):1795–809.
mal models of RP and patients. The inflammation 2. Wooff Y, Man SM, Aggio-Bruce R, Natoli R,
leads to microglia activation, release of pro-­ Fernando N. IL-1 family members mediate cell death,
inflammatory cytokines such as the TNFα, IL6, inflammation and angiogenesis in retinal degenerative
diseases. Front Immunol. 2019;10:1618.
IL1β, etc., reactive gliosis, free radical produc- 3. Broadgate S, Yu J, Downes SM, Halford
tion, etc. [28]. S. Unravelling the genetics of inherited retinal
Compounds with typically anti-inflammatory dystrophies: past, present and future. Prog Retin Eye
characteristics such as progesterone, lipoic acid Res. 2017;59:53–96.
4. RetNet. Retinal Information Network. Available
[31, 39], and curcumin [42] also have antioxidant from: https://sph.uth.edu/retnet/home.htm
properties reducing oxidative stress. Therefore, 5. Chang B. Animal models of retinitis Pigmentosa (RP).
the combination of antioxidant and inflammatory In: Chan C-C, editor. Animal models of ophthalmic
compounds would be a comprehensive strategy diseases. Cham: Springer International Publishing;
2016. p. 101–16.
for tackling retinal degenerative diseases. 6. Campochiaro PA, Mir TA. The mechanism of cone
cell death in retinitis Pigmentosa. Prog Retin Eye Res.
2018;62:24–37.
5 Conclusions and Future 7. Kruk J, Kubasik-Kladna K, Aboul-Enein HY. The role
oxidative stress in the pathogenesis of eye diseases:
Perspectives current status and a dual role of physical activity. Mini
Rev Med Chem. 2015;16(3):241–57.
IRDs, including RP as genetic diseases, leads to 8. Viringipurampeer IA, Bashar AE, Gregory-Evans CY,
blindness; however, there are different underly- Moritz OL, Gregory-Evans K. Targeting inflamma-
tion in emerging therapies for genetic retinal disease.
ing cellular processes such as oxidative stress, Int J Inflamm. 2013;2013:581751.
inflammation, cyclic guanosine monophosphate 9. Yoshida N, Ikeda Y, Notomi S, Ishikawa K, Murakami
deregulation, intracellular calcium accumulation, Y, Hisatomi T, et al. Clinical evidence of sustained
and endoplasmic reticulum stress that can exac- chronic inflammatory reaction in retinitis pigmentosa.
Ophthalmology. 2013;120(1):100–5.
erbate the death of photoreceptor cells [6–9]. On 10. Sies H. Oxidative stress: concept and some practical
the other hand, the oxidation of lipids, proteins, aspects. Antioxidants (Basel). 2020;9(9):852.
Redox Status in Retinitis Pigmentosa 447

11. Arjunan P, Lin X, Tang Z, Du Y, Kumar A, Liu L, 26. Lang M, Harris A. Vascular dysfunction in retinitis
et al. VEGF-B is a potent antioxidant. Proc Natl Acad pigmentosa. Acta Ophthalmol. 2019;97(7):660–4.
Sci U S A. 2018;115(41):10351–6. 27. Türksever C, Valmaggia C, Orgül S, Schorderet
12. De La Paz MA, Anderson RE. Lipid peroxidation DF, Flammer J, Todorova MG. Retinal vessel
in rod outer segments. Role of hydroxyl radical and oxygen saturation and its correlation with structural
lipid hydroperoxides. Invest Ophthalmol Vis Sci. changes in retinitis pigmentosa. Acta Ophthalmol.
1992;33(7):2091–6. 2014;92(5):454–60.
13. Glickman RD, Lam KW. Oxidation of ascorbic acid 28. Olivares-González L, Velasco S, Campillo I, Rodrigo
as an indicator of photooxidative stress in the eye. R. Retinal inflammation, cell death and inherited
Photochem Photobiol. 1992;55(2):191–6. retinal dystrophies. Int J Mol Sci. 2021;22(4):2096.
14. Martínez-Fernández de la Cámara C, Hernández-­ 29. Valero-Vello M, Peris-Martínez C, García-Medina
Pinto AM, Olivares-González L, Cuevas-Martín C, JJ, Sanz-González SM, Ramírez AI, Fernández-
Sánchez-Aragó M, Hervás D, et al. Adalimumab Albarral JA, et al. Searching for the antioxidant,
reduces photoreceptor cell death in A mouse model of anti-­inflammatory, and neuroprotective potential of
retinal degeneration. Sci Rep. 2015;5:11764. natural food and nutritional supplements for ocular
15. Murakami Y, Ikeda Y, Yoshida N, Notomi S, Hisatomi health in the mediterranean population. Foods.
T, Oka S, et al. MutT homolog-1 attenuates oxidative 2021;10(6):1231.
DNA damage and delays photoreceptor cell death 30. Dias MF, Joo K, Kemp JA, Fialho SL, da Silva Cunha
in inherited retinal degeneration. Am J Pathol. A Jr, Woo SJ, et al. Molecular genetics and emerging
2012;181(4):1378–86. therapies for retinitis pigmentosa: basic research
16. Olivares-González L, Martínez-Fernández de la and clinical perspectives. Prog Retin Eye Res.
Cámara C, Hervás D, Millán JM, Rodrigo R. HIF-1α 2018;63:107–31.
stabilization reduces retinal degeneration in a 31. Hernandez-Rabaza V, Lopez-Pedrajas R, Almansa
mouse model of retinitis pigmentosa. FASEB J. I. Progesterone, lipoic acid, and sulforaphane as
2018;32(5):2438–51. promising antioxidants for retinal diseases: a review.
17. Shen J, Yang X, Dong A, Petters RM, Peng YW, Wong Antioxidants (Basel). 2019;8(3):53.
F, et al. Oxidative damage is a potential cause of cone 32. Berson EL, Rosner B, Sandberg MA, Hayes KC,
cell death in retinitis pigmentosa. J Cell Physiol. Nicholson BW, Weigel-DiFranco C, et al. A randomized
2005;203(3):457–64. trial of vitamin A and vitamin E supplementation for
18. Yu DY, Cringle S, Valter K, Walsh N, Lee D, Stone retinitis pigmentosa. Arch Ophthalmol (Chicago, Ill:
J. Photoreceptor death, trophic factor expression, 1960). 1993;111(6):761–72.
retinal oxygen status, and photoreceptor function 33. Li T, Sandberg MA, Pawlyk BS, Rosner B,
in the P23H rat. Invest Ophthalmol Vis Sci. Hayes KC, Dryja TP, et al. Effect of vitamin A
2004;45(6):2013–9. supplementation on rhodopsin mutants threonine-17
19. Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, → methionine and proline-347 → serine in transgenic
et al. HIFalpha targeted for VHL-mediated destruction mice and in cell cultures. Proc Natl Acad Sci U S A.
by proline hydroxylation: implications for O2 sensing. 1998;95(20):11933–8.
Science (New York, NY). 2001;292(5516):464–8. 34. Berson EL, Rosner B, Sandberg MA, Weigel-­
20. Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). DiFranco C, Moser A, Brockhurst RJ, et al. Further
Mol Pharmacol. 2006;70(5):1469–80. evaluation of docosahexaenoic acid in patients with
21. Martínez-Fernández de la Cámara C, Salom D, retinitis pigmentosa receiving vitamin A treatment:
Sequedo MD, Hervás D, Marín-Lambíes C, Aller E, subgroup analyses. Arch Ophthalmol (Chicago, Ill:
et al. Altered antioxidant-oxidant status in the aqueous 1960). 2004;122(9):1306–14.
humor and peripheral blood of patients with retinitis 35. Thomson LR, Toyoda Y, Langner A, Delori FC,
pigmentosa. PloS One. 2013;8(9):e74223. Garnett KM, Craft N, et al. Elevated retinal zeaxanthin
22. Martínez-Fernández de la Cámara C, Sequedo MD, and prevention of light-induced photoreceptor
Gómez-Pinedo U, Jaijo T, Aller E, García-Tárraga cell death in quail. Investig Ophthalmol Vis Sci.
P, et al. Phosphodiesterase inhibition induces retinal 2002;43(11):3538–49.
degeneration, oxidative stress and inflammation in 36. Yang F, Yu J, Ke F, Lan M, Li D, Tan K, et al. Curcumin
cone-enriched cultures of porcine retina. Exp Eye alleviates diabetic retinopathy in experimental
Res. 2013;111:122–33. diabetic rats. Ophthalmic Res. 2018;60(1):43–54.
23. Komeima K, Rogers BS, Lu L, Campochiaro 37. Pajovic S, Saicic ZS, Spasic MB, Petrovic VM,
PA. Antioxidants reduce cone cell death in a model Martinovic JV. Effect of progesterone and estradiol
of retinitis pigmentosa. Proc Natl Acad Sci U S A. benzoate on superoxide dismutase activity in the brain
2006;103(30):11300–5. of male rats. Experientia. 1996;52(3):221–4.
24. Yu DY, Cringle SJ. Retinal degeneration and local 38. Stein DG, Wright DW, Kellermann AL. Does
oxygen metabolism. Exp Eye Res. 2005;80(6):745–51. progesterone have neuroprotective properties? Ann
25. Yu DY, Cringle SJ, Su EN, Yu PK. Intraretinal Emerg Med. 2008;51(2):164–72.
oxygen levels before and after photoreceptor 39. Ramírez-Lamelas DT, Benlloch-Navarro S, López-­
loss in the RCS rat. Invest Ophthalmol Vis Sci. Pedrajas R, Gimeno-Hernández R, Olivar T, Silvestre
2000;41(12):3999–4006. D, et al. Lipoic acid and progesterone alone or in
448 L. Olivares-González et al.

combination ameliorate retinal degeneration in an 42. Wang Y, Yin Z, Gao L, Sun D, Hu X, Xue L, et al.
experimental model of hereditary retinal degeneration. Curcumin delays retinal degeneration by regulating
Front Pharmacol. 2018;9:469. microglia activation in the retina of rd1 mice. Cell
40. Sanz MM, Johnson LE, Ahuja S, Ekstrom PA, Romero Physiol Biochem. 2017;44(2):479–93.
J, van Veen T. Significant photoreceptor rescue by 43. Rojo AI, McBean G, Cindric M, Egea J, Lopez MG,
treatment with a combination of antioxidants in an Rada P, et al. Redox control of microglial function:
animal model for retinal degeneration. Neuroscience. molecular mechanisms and functional significance.
2007;145(3):1120–9. Antioxid Redox Signal. 2014;21(12):1766–801.
41. Olivares-González L, Velasco S, Campillo I, Salom 44. Roy A, Jana A, Yatish K, Freidt MB, Fung YK,
D, González-García E, Soriano Del Castillo J, et al. Martinson JA, et al. Reactive oxygen species up-­
Nutraceutical supplementation ameliorates visual regulate CD11b in microglia via nitric oxide:
function, retinal degeneration, and redox status in implications for neurodegenerative diseases. Free
rd10 Mice. Antioxidants (Basel). 2021;10(7):1033. Radic Biol Med. 2008;45(5):686–99.
Perspectives on Retinal Dolichol
Metabolism, and Visual Deficits
in Dolichol Metabolism-Associated
Inherited Disorders

Sriganesh Ramachandra Rao, Steven J. Pittler,


and Steven J. Fliesler

Abstract ciated with Dol metabolism disorders, and


identify the need for generation and character-
De novo synthesis of dolichol (Dol) and doli- ization of suitable animal models of these dis-
chyl phosphate (Dol-P) is essential for protein orders to elucidate the underlying molecular
glycosylation. Herein, we provide a brief and cellular mechanisms of the associated
overview of Dol and Dol-P synthesis and the retinopathies.
maintenance of their cellular content. Retinal
Dol metabolism and the requirement of Dol-­ Keywords
linked oligosaccharide synthesis in the neural
retina also are discussed. There are recently Dolichol · Dolichyl phosphate · Retinitis
discovered and an emerging class of rare con- pigmentosa · Vision defects · Protein glyco-
genital disorders that affect Dol metabolism, sylation · DHDDS · NUS1 · SRD5A3 ·
involving the genes DHDDS, NUS1, SRD5A3, DOLPP1 · DOLK
and DOLK. Further understanding of these
congenital disorders is evolving, based upon
studies utilizing yeast and murine models, as 1 Introduction
well as clinical reports of these rare disorders.
We summarize the known visual deficits asso- The mevalonate pathway generates a broad spec-
trum of isoprenoid metabolites essential for life.
This includes cyclic isoprenoids such as choles-
S. Ramachandra Rao · S. J. Fliesler (*)
Departments of Ophthalmology and Biochemistry, terol and other sterols, and linear isoprenoids
and Neuroscience Graduate Program, Jacobs School such as dolichol (Dol) and its derivatives, and
of Medicine and Biomedical Sciences, State ubiquinone. Several of these key isoprenoid
University of New York, Buffalo, NY, USA metabolites are directly involved in protein post-
Research Service, VA Western NY Healthcare translational modifications (e.g., protein prenyl-
System, Buffalo, NY, USA ation). Farnesyl pyrophosphate (FPP) and
e-mail: sramacha@buffalo.edu; fliesler@buffalo.edu;
steven.fliesler@va.gov isopentenyl pyrophosphate (IPP) are key meval-
onate pathway intermediates required for the
S. J. Pittler
Department of Optometry and Vision Science, Vision synthesis of squalene, sterols, as well as Dol (see
Science Research Center, School of Optometry, Fig. 1). This article focuses on Dol metabolism
University of Alabama at Birmingham, and its role and requirement in retinal physiol-
Birmingham, AL, USA ogy, specifically as pertains to protein
e-mail: pittler@uab.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 449
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_66
450 S. Ramachandra Rao et al.

g­lycosylation. We will briefly discuss genetic lower rates [8]. Free Dol is phosphorylated to
disorders pertaining to Dol metabolism and the form Dol-P by a CTP-dependent, ER-resident
associated visual deficits. enzyme called dolichol kinase (DOLK) [9]. The
The committed step in Dol synthesis is cata- N-linked oligosaccharide synthesis begins on the
lyzed by the heterotetrameric cis-prenyl transfer- cytosolic face of ER, before the Dol-P-linked oli-
ase (CPT) complex, constituted by gosaccharide (with five mannose residues) is
dehydrodolichyl diphosphate synthase (DHDDS, flipped across the ER membrane to the ER
OMIM# 608172) and Nogo-B receptor (NgBR; lumen via the activity of scramblases/flippases
NUS1, OMIM# 610463) (Fig. 1). The mamma- that are yet to be identified and characterized
lian CPT complex catalyzes the cis-condensation [10–13] (Fig. 1). An oligosaccharyl
of an FPP (C15) molecule with multiple molecules transferase (DPAGT1, dolichyl phosphate
of IPP (C5), to form polyprenol pyrophosphate N-acetylglucosaminephosphotransferase 1) cata-
(C90-C100) [1–4]. Polyprenol pyrophosphate then lyzes the transfer of oligosaccharides onto aspar-
undergoes reduction to generate dolichyl pyro- agine glycosylation consensus sites of nascent
phosphate (Dol-PP), catalyzed by steroid-5α-­ peptides in the rough ER lumen. This regenerates
reductase 3 (SRD5A3) [5]. However, the obligate Dol-PP on the rough ER luminal face, the sub-
glycan carrier required for protein glycosylation strate for DOLPP1, and regenerates Dol-P. Dol-P
is dolichyl phosphate (Dol-P) [6, 7]. Cellular is then flipped to the cytoplasmic side of the ER
Dol-P homeostasis is maintained by the action of for subsequent rounds of oligosaccharide synthe-
dolichol diphosphatase 1 (DOLPP1) on Dol-PP, sis [14].
which generates Dol-P. DOLPP1 also can cata- Contribution of dietary Dol to tissue Dol con-
lyze the conversion of Dol-P to Dol, albeit at tent is negligible [15–17]. Although serum Dol is

Fig. 1 Schematic representation of Dol-P synthesis and dehydrodolichyl diphosphate synthase, NUS1 Nogo-B
Dol homeostasis. The interplay between Dol homeostasis receptor (NgBR), SRD5A3 steroid-5α-reductase 3,
and protein glycosylation has been represented. Visual DOLPP1 dolichyl diphosphatase 1, DOLK dolichol
deficits associated with congenital disorders of Dol kinase, OST oligosaccharyltransferase, CTP cytidine
metabolism have been provided. Abbreviations: DHDDS triphosphate
Perspectives on Retinal Dolichol Metabolism, and Visual Deficits in Dolichol Metabolism-Associated… 451

associated with high-density lipoproteins (HDL) ing and incorporation of rhodopsin into rod outer
[18–23], there is no receptor-mediated endocytic segment (ROS) membranes. Similarly, point
mechanism for cellular/tissue Dol uptake in vivo. mutation of either of the two glycosylation con-
On the other hand, low-density lipoprotein (LDL) sensus sites of rhodopsin leads to autosomal
particles loaded with Dol may undergo receptor-­ dominant retinitis pigmentosa, with rapid retinal
mediated endocytosis, but such Dol accumulates degeneration [43–45]. Below, we briefly discuss
in the lysosomes [24]. However, uptake of LDL-­ an emerging class of inherited disorders, involv-
borne cholesterol (via LDL receptor-mediated ing mutations in genes (DHDDS, NUS1, SRD5A3,
uptake), or pharmacological inhibition of the DOLK, and DOLPP1) pertinent to Dol and Dol-P
mevalonate pathway at the level of 3-hydroxy-­3- homeostasis, wherein the underlying mechanism
methylglutaryl coenzyme-A reductase of retinopathy is presumed to be driven by defec-
(HMGCR), using statins, decreases tissue Dol tive protein glycosylation.
synthesis [25–28]. Therefore, tissues including
the neural retina depend on de novo synthesis of
Dol for Dol-dependent cellular processes. 3 Inherited Disorders Affecting
Dolichol Metabolism

2 Dolichol Synthesis 3.1 DHDDS and NUS1-CDG


and Dolichol-Linked
Oligosaccharides Dol-P, the obligate glycan carrier, is indispens-
in the Neural Retina able for protein N-glycosylation, as well as pro-
tein O- and C-mannosylation, O-glucosylation,
The de novo synthesis of Dol and its derivatives and glycosylphosphatidylinositol (GPI) anchor
in the retina was first assessed in vitro by incubat- synthesis [5–7, 46]. The first described cases of
ing frog retinas with [3H]acetate, the de novo pre- DHDDS-associated retinopathy were classified
cursor to acetyl-CoA in the mevalonate pathway as a non-syndromic autosomal recessive form of
[29] (Fig. 1). The specific activity of radiolabeled retinitis pigmentosa, called RP59 (OMIM#
Dol was determined by radio-HPLC. The major- 613861). They involved a founder missense
ity of the [3H]acetate was incorporated into squa- mutation (K42E) in DHDDS [47, 48]. Other
lene, rather than into sterols. Dol synthesis was DHDDS mutations also have been described in
found to be 60-fold slower than that of choles- RP59 patients (e.g., T206A, R98W both found
terol synthesis [29]. [3H]acetate was incorporated heterozygously with the K42E mutation) [49–
into squalene, as well as cholesterol, and Dol [29, 53]. Since de novo synthesized Dol-P is required
30]. This in vitro experiment demonstrated the by all eukaryotic cells for the assembly of
capacity of the neural retina to synthesize Dol de N-glycan chains and cell viability, RP59 and
novo. Other in vitro studies, utilizing bovine, NUS1-associated retinopathies have been classi-
chick, and rodent retinas, demonstrated an essen- fied as “congenital disorders of glycosylation”
tial role for Dol-P in incorporation of mannose (CDGs) [47, 48, 54]. Curiously, the clinical fea-
into retinal glycoproteins [31–37]. Statin treat- tures of RP59 are restricted to the eye/retina (i.e.,
ment also affects protein prenylation in the neural RP59 is non-syndromic); the retina-specific func-
retina, and leads to retinal degeneration [38, 39]. tions of DHDDS are yet unknown [47, 48, 53]
Further, in vivo [40] and in vitro [41, 42] admin- (Fig. 1). More recently, mutations in DHDDS
istration of tunicamycin, a DPAGT1 oligosac- have been implicated in early-onset encephalopa-
charyltransferase (OST) inhibitor, on adult thy [50, 55–57]. The proposed mechanism of
amphibian retinas, demonstrated the importance CPT-associated retinopathies involves defective
of protein N-glycosylation in maintaining the rod and cone opsin N-glycosylation leading to
structure and function of the neural retina, and loss of photoreceptors [47, 48]. This hypothesis
demonstrated glycosylation-dependent traffick- was based upon studies of a dhdds knockdown
452 S. Ramachandra Rao et al.

zebrafish model [48, 58]. We generated the first ders [54, 62, 63, 66–69]. Foveal lesions have
validated murine animal models to determine the been reported in patients with mutations in NUS1
link between CPT activity and retinal protein gly- [54] (Fig. 1). Defects in other cellular processes
cosylation and morphology/function, by knock- mediated by CPT, or their interacting protein
ing in the RP59-associated K42E DHDDS partners, cannot be ruled out; however, it is diffi-
mutation, as well as selectively ablating (knock- cult to envision such effects being retina specific.
out) Dhdds in rod photoreceptors and RPE cells It may be interesting to investigate the role of
[4, 59, 60]. Surprisingly, rod-specific knockout of dolichol in photoreceptor membrane, and if such
Dhdds led to rapid retinal degeneration within functions are compromised by the reduction in
6 weeks of age [4]; however, the observed degen- chain length, as observed in RP59 [49].
eration did not lead to obvious protein Understanding the roles of DHDDS and NgBR in
N-glycosylation defects, despite marked reduc- Dol-dependent protein glycosylation, specifi-
tion in retinal Dol levels. The global K42E cally in retinal photoreceptors, may provide criti-
knock-in mouse model of RP59 exhibits no overt cal insights regarding the developmental role of
degenerative phenotype up to at least 12 months the CPT complex in photoreceptor glycosylation
of age [49, 59]; however, a mild, age-dependent as well as in phagolysosome biology.
phenotype has been observed at later time points
(unpublished results). Further, RP59 patient
fibroblasts and serum transferrin profiles also do 3.2 SRD5A3-CDG
not exhibit obvious protein N-glycosylation
defects [50, 56]. These findings argue against the The reduction of polyprenol pyrophosphate to
hypothesis implicating the primary involvement Dol-PP in the ER was demonstrated using rat
of defective protein N-glycosylation [4] in RP59. liver microsome preparations [70]. The involve-
We have not observed retinal degeneration as ment of SRD5A3 (steroid-5α-reductase 3) in the
occurs when N-glycosylation is specifically dis- catalytic reduction of the terminal isoprene unit
rupted [40–42, 45, 61]. In contrast, RP59 patient-­ (the alpha isoprene residue) of CPT-synthesized
derived fibroblasts exhibit accumulation of lipid polyprenol pyrophosphate to Dol-PP was
droplets and signatures of lysosome storage dis- described recently [5] (Fig. 1). Deletion of the
orders [56, 57]. The disease mechanism underly- yeast ortholog of SRD5A3, dfg10–100, leads to
ing RP59 remains to be elucidated, and to be accumulation of polyprenol pyrophosphate and
studied in relation to other genes involved in Dol decrease in Dol-PP, with accompanying protein
synthesis pathway, to better define how Dol glycosylation defects [5]. Such glycosylation
homeostasis is achieved and maintained in the defects in dfg10–100 could be rescued by co-­
neural retina. transfection with human SRD5A3. Murine
NgBR (the NUS1 gene product) is a heterotet- global knockout of Srd5a3 leads to embryonic
rametric partner of DHDDS, required for Dol lethality by E13.5 days, with reduction of Dol
synthesis. However, NgBR appears to have a levels, and accumulation of polyprenol pyro-
broad interactome based on its various functions phosphate. Targeted deletion of Srd5a3 in the
[62–65]. NgBR plays a critical role in lysosome cerebellum in mice, using a conditional allele
function and lipid trafficking, by stabilizing the model, leads to frank protein glycosylation
Niemann-Pick type C-2 complex (NPC2), which defects [71]. Patient-­ associated mutations in
is required for trafficking of free cholesterol from SRD5A3 lead to a CDG [5, 72, 73] (OMIM#
lysosomes to the ER. Deletion of Nus1 causes a 612379). Ocular manifestations of SRD5A3
Niemann-Pick type C disease-like lysosomal mutation include retinal dystrophy, optic atro-
storage disorder [63, 66, 67]. Yeast and mamma- phy, nystagmus, and myopia (Fig. 1). Other sys-
lian models involving Nus1 mutations or knock- temic defects involve ichthyosis (skin defects),
out clearly exhibit protein glycosylation defects, cerebellar ataxia, muscular hypotonia, and cog-
and classic signatures of lysosomal storage disor- nitive deficits [5, 72–74].
Perspectives on Retinal Dolichol Metabolism, and Visual Deficits in Dolichol Metabolism-Associated… 453

3.3 DOLPP1-CDG from yeast and mouse models of human disor-


ders classified as CDGs, associated with Dol
DOLPP1 belongs to the PAP2 (phosphatidic acid metabolism. We have further accounted for the
phosphatase 2)-like superfamily of proteins [75]. reported biochemical and vision-related pheno-
The classic enzymatic active sites of the PAP2-­ types in patients with mutations associated with
like family of proteins are fully conserved genes involved in Dol metabolism. While muta-
between DOLPP1 and its yeast ortholog, CWH8 tions in most of the Dol metabolism-associated
[76, 77]. Both Dol-PP and Dol-P serve as enzy- genes (NUS1, SRD5A3, DOLK, DOLPP1) clearly
matic substrates (Fig. 1). DOLPP1 activity lead to protein glycosylation defects, DHDDS-­
increases during brain development, and requires associated mutations surprisingly do not.
divalent cations, especially Mn2+ [78, 79]. Mutations in Dol metabolism-associated genes
Mutation of the yeast gene cwh8 leads to protein are emerging as a new class of congenital disor-
hypoglycosylation, which could be rescued by ders with significant ocular manifestations. To
reconstitution of DOLPP1 [80, 81]. To date, there fully elucidate the role of Dol metabolism in the
are no published clinical reports pertaining to neural retina, and to understand the disease
DOLPP1 mutations. mechanisms underlying the above-discussed
congenital disorders, we propose that there is a
need for the generation and characterization of
3.4 DOLK-CDG animal models with retinal cell type-specific
deletion of these genes and further modeling of
DOLK belongs to cytidylyltransferase family of identified mutations. Such rigorous modeling of
CTP-dependent kinase enzymes [75] (Fig. 1). these rare metabolic disorders will facilitate bet-
DOLK is an ER-resident protein; the highest ter understanding of the disease mechanisms,
level of DOLK activity is exhibited in the brain providing a rational basis for development of tar-
during development [9, 82]. DOLK transcript geted therapeutic interventions, as well as pro-
and protein expression have been documented in viding a more thorough understanding of
all cell types of the neural retina [46, 83]. Deletion Dol-linked protein glycosylation in the neural
of sec59, the yeast ortholog of DOLK, also leads retina.
to protein hypoglycosylation and lipid droplet
accumulation [9, 46, 84] (Fig. 1). Patients with
DOLK mutations (OMIM# 610768) are clini- References
cally diagnosed by a CDG type 1 transferrin gly-
cosylation pattern, with elevated disialo- and 1. Edani BH, Grabinska KA, Zhang R, Park EJ, Siciliano
B, Surmacz L, et al. Structural elucidation of the cis-­
asialo-transferrin glycoforms [46, 85, 86]. Also, prenyltransferase NgBR/DHDDS complex reveals
patients harboring DOLK mutations (OMIM# insights in regulation of protein glycosylation. Proc.
610768) exhibit systemic defects, such as cardio- Natl. Acad. Sci. U. S. A. 2020;117(34):20794–802.
myopathy, hepatic and renal complications, mus- 2. Bar-El ML, Vankova P, Yeheskel A, Simhaev L,
Engel H, Man P, et al. Structural basis of hetero-
cular hypotonia with underlying tetrameric assembly and disease mutations in the
dystroglycanopathy, and ichthyosis, along with human cis-prenyltransferase complex. Nat. Commun.
visual system defects, including nystagmus and 2020;11(1):5273.
strabismus (Fig. 1) [46, 86]. 3. Ramachandra Rao S, Fliesler SJ. Cholesterol homeo-
stasis in the vertebrate retina: biology and pathobiol-
ogy. J. Lipid Res. 2021;62:100057.
4. Ramachandra Rao S, Skelton LA, Wu F, Onysk A,
4 Conclusions Spolnik G, Danikiewicz W, et al. Retinal degeneration
caused by rod-specific dhdds ablation occurs without
concomitant inhibition of protein N-glycosylation.
Herein, we have briefly reviewed the role and iScience. 2020;23(6):101198.
requirement of Dol metabolism in the neural ret- 5. Cantagrel V, Lefeber DJ, Ng BG, Guan Z, Silhavy JL,
ina, and have described relevant observations Bielas SL, et al. SRD5A3 is required for converting
454 S. Ramachandra Rao et al.

polyprenol to dolichol and is mutated in a congenital tatin on liver and plasma levels of cholesterol, doli-
glycosylation disorder. Cell. 2010;142(2):203–17. chol and ubiquinol in hypercholesterolemic rats. Ital.
6. Leloir LF. The role of dolichol in protein glycosyl- J. Biochem. 1995;44(1):1–9.
ation. Adv. Exp. Med. Biol. 1977;83:9–19. 22. Kuriyama M, Yoshidome H, Nakahara K, Nakagawa
7. Parodi AJ. Synthesis of glycosyl-dolichol derivatives H, Fujiyama J, Take H, et al. Blood dolichols in
in bakers’ yeast and their role in protein glycosyl- a patient with abetalipoproteinaemia. Ann. Clin.
ation. Eur. J. Biochem. 1977;75(1):171–80. Biochem. 1999;36(Pt 2):176–9.
8. Scher MG, Waechter CJ. Brain dolichyl pyrophosphate 23. Shiota Y, Kiyota K, Kobayashi T, Kano S, Kawamura
phosphatase. Solubilization, characterization, and dif- M, Matsushima T, et al. Distribution of dolichol in
ferentiation from dolichyl monophosphate phospha- the serum and relationships between serum dolichol
tase activity. J. Biol. Chem. 1984;259(23):14580–5. levels and various laboratory test values. Biol. Pharm.
9. Shridas P, Waechter CJ. Human dolichol kinase, a Bull. 2008;31(3):340–7.
polytopic endoplasmic reticulum membrane protein 24. Van Dessel G, Van Meirvenne H, Lagrou A, Hilderson
with a cytoplasmically oriented CTP-binding site. H, Dierick W. Uptake of dolichol by Vero cells.
J. Biol. Chem. 2006;281(42):31696–704. Biochem. Cell Biol. 1992;70(6):475–80.
10. Verchere A, Cowton A, Jenni A, Rauch M, Haner 25. Borgford TJ, Hurta RA, Tough DF, Burton
R, Graumann J, et al. Complexity of the eukaryotic DN. Regulation of 3-hydroxy-3-methylglutaryl coen-
dolichol-­linked oligosaccharide scramblase suggested zyme A reductase and lipid metabolism in a concana-
by activity correlation profiling mass spectrometry. valin A-resistant Chinese hamster ovary cell line.
Sci. Rep. 2021;11(1):1411. Arch. Biochem. Biophys. 1986;244(2):502–16.
11. Sanyal S, Menon AK. Stereoselective transbilayer 26. Dull BJ, McCarthy RD, Kilara A. The modulating
translocation of mannosyl phosphoryl dolichol by an effect of an inhibitor of cholesterol genesis pres-
endoplasmic reticulum flippase. Proc. Natl. Acad. Sci. ent in bovine milk upon the synthesis of choles-
U. S. A. 2010;107(25):11289–94. terol, dolichol and ubiquinone. Atherosclerosis.
12. Sanyal S, Menon AK. Specific transbilayer transloca- 1983;49(3):231–9.
tion of dolichol-linked oligosaccharides by an endo- 27. Filipovic I, Menzel B. Action of low-density lipo-
plasmic reticulum flippase. Proc. Natl. Acad. Sci. protein and compactin, a competitive inhibitor of
U. S. A. 2009;106(3):767–72. 3-hydroxy-3-methylglutaryl-CoA reductase, on the
13. Sanyal S, Frank CG, Menon AK. Distinct flippases synthesis of dolichol-linked oligosaccharides and
translocate glycerophospholipids and oligosaccharide low-density-lipoprotein receptor in human skin fibro-
diphosphate dolichols across the endoplasmic reticu- blasts. Biochem. J. 1981;196(2):625–8.
lum. Biochemistry. 2008;47(30):7937–46. 28. Appelkvist EL, Edlund C, Low P, Schedin S, Kalen
14. Rush JS, Gao N, Lehrman MA, Waechter A, Dallner G. Effects of inhibitors of hydroxymeth-
CJ. Recycling of dolichyl monophosphate to the cyto- ylglutaryl coenzyme A reductase on coenzyme Q
plasmic leaflet of the endoplasmic reticulum after the and dolichol biosynthesis. Clin. Investig. 1993;71(8
cleavage of dolichyl pyrophosphate on the lumenal Suppl):S97–102.
monolayer. J. Biol. Chem. 2008;283(7):4087–93. 29. Keller RK, Fliesler SJ, Nellis SW. Isoprenoid bio-
15. Keller RK, Jehle E, Adair WL Jr. The origin of doli- synthesis in the retina. Quantitation of the sterol
chol in the liver of the rat. Determination of the dietary and dolichol biosynthetic pathways. J. Biol. Chem.
contribution. J. Biol. Chem. 1982;257(15):8985–9. 1988;263(5):2250–4.
16. Rip JW, Carroll KK. Distribution, metabolism and 30. Fliesler SJ, Florman R, Rapp LM, Pittler SJ, Keller
excretion of [1-14C]dolichol injected intravenously RK. In vivo biosynthesis of cholesterol in the rat ret-
into rats. Biochem. J. 1985;227(3):705–10. ina. FEBS Lett. 1993;335(2):234–8.
17. Crick DC, Carroll KK. Absorption and distribution 31. Kean EL. GDP-mannose-polyprenyl phosphate
of [1-14C]dolichol intubated into rats. Biochem. Cell mannosyltransferases of the retina. J. Biol. Chem.
Biol. 1987;65(4):317–20. 1977;252(16):5622–9.
18. Elmberger PG, Engfeldt P, Dallner G. Presence of 32. Kean EL. Mannosyl transferases of the retina: man-
dolichol and its derivatives in human blood. J. Lipid nolipid and complex glycan biosynthesis. I. Kinetic
Res. 1988;29(12):1651–62. properties; product identification. Exp. Eye Res.
19. Elmberger PG, Kalen A, Brunk UT, Dallner 1977;25(4):405–17.
G. Discharge of newly-synthesized dolichol and ubi- 33. Kean EL. Activation by dolichol
quinone with lipoproteins to rat liver perfusate and to phosphate-­ mannose of the biosynthesis of
the bile. Lipids. 1989;24(11):919–30. N-acetylglucosaminylpyrophosphoryl polyprenols by
20. Van Dessel G, Lagrou A, Hilderson HJ, Dierick the retina. J. Biol. Chem. 1982;257(14):7952–4.
W. The shuttling of dolichol between VLDL and HDL: 34. Kean EL. Influence of metal ions on the biosynthesis
involvement of a protein factor from lipoprotein-­ of N-acetylglucosaminyl polyprenols by the retina.
deficient human serum. Biochim. Biophys. Acta. Biochim. Biophys. Acta. 1983;750(2):268–73.
1993;1166(1):64–72. 35. Plantner JJ, Kean EL. The dolichol pathway in the
21. Marinari UM, Pronzato MA, Dapino D, Gazzo P, retina: oligosaccharide-lipid biosynthesis. Exp. Eye
Traverso N, Cottalasso D, et al. Effects of simvas- Res. 1988;46(5):785–800.
Perspectives on Retinal Dolichol Metabolism, and Visual Deficits in Dolichol Metabolism-Associated… 455

36. Kean EL. The dolichol pathway in the retina and 51. Venturini G, Koskiniemi-Kuendig H, Harper S,
its involvement in the glycosylation of rhodopsin. Berson EL, Rivolta C. Two specific mutations are
Biochim. Biophys. Acta. 1999;1473(2–3):272–85. prevalent causes of recessive retinitis pigmentosa in
37. O'Brien PJ. Incorporation of mannose into rho- North American patients of Jewish ancestry. Genet.
dopsin in isolated bovine retina. Exp. Eye Res. Med. 2015;17(4):285–90.
1977;24(5):449–58. 52. Biswas P, Duncan JL, Maranhao B, Kozak I, Branham
38. Pittler SJ, Fliesler SJ, Fisher PL, Keller PK, Rapp K, Gabriel L, et al. Genetic analysis of 10 pedigrees
LM. In vivo requirement of protein prenylation for with inherited retinal degeneration by exome sequenc-
maintenance of retinal cytoarchitecture and photore- ing and phenotype-genotype association. Physiol.
ceptor structure. J. Cell Biol. 1995;130(2):431–9. Genomics. 2017;49(4):216–29.
39. Pittler SJ, Fliesler SJ, Rapp LM. Novel morphological 53. Kimchi A, Khateb S, Wen R, Guan Z, Obolensky
changes in rat retina induced by intravitreal injection A, Beryozkin A, et al. Nonsyndromic retini-
of lovastatin. Exp. Eye Res. 1992;54(1):149–52. tis Pigmentosa in the Ashkenazi Jewish popula-
40. Fliesler SJ, Rapp LM, Hollyfield JG. Photoreceptor-­ tion: genetic and clinical aspects. Ophthalmology.
specific degeneration caused by tunicamycin. Nature. 2018;125(5):725–34.
1984;311(5986):575–7. 54. Park EJ, Grabinska KA, Guan Z, Stranecky V,
41. Fliesler SJ, Basinger SF. Tunicamycin blocks the Hartmannova H, Hodanova K, et al. Mutation of
incorporation of opsin into retinal rod outer seg- Nogo-B receptor, a subunit of cis-prenyltransferase,
ment membranes. Proc. Natl. Acad. Sci. U. S. A. causes a congenital disorder of glycosylation. Cell
1985;82(4):1116–20. Metab. 2014;20(3):448–57.
42. Fliesler SJ, Rayborn ME, Hollyfield JG. Membrane 55. Piccolo G, Amadori E, Vari MS, Marchese F, Riva
morphogenesis in retinal rod outer segments: inhibi- A, Ghirotto V, et al. Complex neurological phe-
tion by tunicamycin. J. Cell Biol. 1985;100(2):574–87. notype associated with a De Novo DHDDS muta-
43. Tam BM, Moritz OL. The role of rhodopsin gly- tion in a boy with intellectual disability, refractory
cosylation in protein folding, trafficking, and epilepsy, and movement disorder. J Pediatr Genet.
light-sensitive retinal degeneration. J. Neurosci. 2021;10(3):236–8.
2009;29(48):15145–54. 56. Galosi S, Edani BH, Martinelli S, Hansikova H,
44. Kim KJ, Kim C, Bok J, Kim KS, Lee EJ, Park SP, et al. Eklund EA, Caputi C, et al. De novo DHDDS variants
Spectrum of rhodopsin mutations in Korean patients cause a neurodevelopmental and neurodegenerative
with retinitis pigmentosa. Mol. Vis. 2011;17:844–53. disorder with myoclonus. Brain; 2021.
45. Murray AR, Vuong L, Brobst D, Fliesler SJ, Peachey 57. Courage C, Oliver KL, Park EJ, Cameron JM,
NS, Gorbatyuk MS, et al. Glycosylation of rhodop- Grabinska KA, Muona M, et al. Progressive myoc-
sin is necessary for its stability and incorporation into lonus epilepsies-residual unsolved cases have marked
photoreceptor outer segment discs. Hum. Mol. Genet. genetic heterogeneity including dolichol-dependent
2015;24(10):2709–23. protein glycosylation pathway genes. Am. J. Hum.
46. Lefeber DJ, de Brouwer AP, Morava E, Riemersma Genet. 2021;108(4):722–38.
M, Schuurs-Hoeijmakers JH, Absmanner B, et al. 58. Wen R, Dallman JE, Li Y, Zuchner SL, Vance JM,
Autosomal recessive dilated cardiomyopathy due to Pericak-Vance MA, et al. Knock-down DHDDS
DOLK mutations results from abnormal dystroglycan expression induces photoreceptor degeneration in
O-mannosylation. PLoS Genet. 2011;7(12):e1002427. zebrafish. Adv. Exp. Med. Biol. 2014;801:543–50.
47. Zelinger L, Banin E, Obolensky A, Mizrahi-­ 59. Ramachandra Rao S, Fliesler SJ, Kotla P, Nguyen
Meissonnier L, Beryozkin A, Bandah-Rozenfeld D, MN, Pittler SJ. Lack of overt retinal degeneration in
et al. A missense mutation in DHDDS, encoding dehy- a K42E Dhdds knock-in mouse model of RP59. Cell.
drodolichyl diphosphate synthase, is associated with 2020;9(4):4.
autosomal-recessive retinitis pigmentosa in Ashkenazi 60. DeRamus ML, Davis SJ, Rao SR, Nyankerh C, Stacks
Jews. Am. J. Hum. Genet. 2011;88(2):207–15. D, Kraft TW, et al. Selective ablation of dehydrodoli-
48. Züchner S, Dallman J, Wen R, Beecham G, Naj A, chyl diphosphate synthase in murine retinal pigment
Farooq A, et al. Whole-exome sequencing links a vari- epithelium (RPE) causes RPE atrophy and retinal
ant in DHDDS to retinitis pigmentosa. Am. J. Hum. degeneration. Cell. 2020;9(3):3.
Genet. 2011;88(2):201–6. 61. Murray AR, Fliesler SJ, Al-Ubaidi MR. Rhodopsin:
49. Wen R, Lam BL, Guan Z. Aberrant dolichol chain the functional significance of asn-linked glycosyl-
lengths as biomarkers for retinitis pigmentosa caused ation and other post-translational modifications.
by impaired dolichol biosynthesis. J. Lipid Res. Ophthalmic Genet. 2009;30(3):109–20.
2013;54(12):3516–22. 62. Harrison KD, Park EJ, Gao N, Kuo A, Rush JS,
50. Sabry S, Vuillaumier-Barrot S, Mintet E, Fasseu M, Waechter CJ, et al. Nogo-B receptor is neces-
Valayannopoulos V, Heron D, et al. A case of fatal sary for cellular dolichol biosynthesis and protein
type I congenital disorders of glycosylation (CDG N-glycosylation. EMBO J. 2011;30(12):2490–500.
I) associated with low dehydrodolichol diphosphate 63. Harrison KD, Miao RQ, Fernandez-Hernando C,
synthase (DHDDS) activity. Orphanet J. Rare Dis. Suarez Y, Davalos A, Sessa WC. Nogo-B receptor sta-
2016;11(1):84. bilizes Niemann-Pick type C2 protein and ­regulates
456 S. Ramachandra Rao et al.

intracellular cholesterol trafficking. Cell Metab. conserved domain database. Nucleic Acids Res.
2009;10(3):208–18. 2015;43(Database issue):D222–6.
64. Zhao B, Hu W, Kumar S, Gonyo P, Rana U, Liu Z, 76. Ishikawa K, Mihara Y, Gondoh K, Suzuki E, Asano
et al. The Nogo-B receptor promotes Ras plasma Y. X-ray structures of a novel acid phosphatase
membrane localization and activation. Oncogene. from Escherichia blattae and its complex with
2017;36(24):3406–16. the transition-state analog molybdate. EMBO J.
65. Yang YD, Li MM, Xu G, Feng L, Zhang EL, Chen J, 2000;19(11):2412–23.
et al. Nogo-B receptor directs mitochondria-­associated 77. Lu S, Wang J, Chitsaz F, Derbyshire MK, Geer
membranes to regulate vascular smooth muscle cell RC, Gonzales NR, et al. CDD/SPARCLE: the con-
proliferation. Int. J. Mol. Sci. 2019;20(9):9. served domain database in 2020. Nucleic Acids Res.
66. Yu SH, Wang T, Wiggins K, Louie RJ, Merino EF, 2020;48(D1):D265–D8.
Skinner C, et al. Lysosomal cholesterol accumula- 78. Scher MG, Sumbilla CM, Waechter CJ. Dolichyl
tion contributes to the movement phenotypes asso- phosphate metabolism in brain. Developmental
ciated with NUS1 haploinsufficiency. Genet. Med. increase in polyisoprenyl phosphate phosphatase
2021;23(7):1305–14. activity. J. Biol. Chem. 1985;260(25):13742–6.
67. Ory DS. Getting a “hold” on NPC2. Cell Metab. 79. Bhat NR, Frank DW, Wolf MJ, Waechter
2009;10(3):161–2. CJ. Developmental changes in enzymes involved in
68. Park EJ, Grabinska KA, Guan Z, Sessa WC. NgBR dolichyl phosphate metabolism in cultured embryonic
is essential for endothelial cell glycosyl- rat brain cells. J. Neurochem. 1991;56(1):339–44.
ation and vascular development. EMBO Rep. 80. Rush JS, Cho SK, Jiang S, Hofmann SL, Waechter
2016;17(2):167–77. CJ. Identification and characterization of a cDNA
69. Zhang P, Cui D, Liao P, Yuan X, Yang N, Zhen Y, encoding a dolichyl pyrophosphate phosphatase
et al. Case report: clinical features of a Chinese boy located in the endoplasmic reticulum of mammalian
with epileptic seizures and intellectual disabilities cells. J. Biol. Chem. 2002;277(47):45226–34.
who carries a truncated NUS1 variant. Front. Pediatr. 81. Fernandez F, Rush JS, Toke DA, Han GS, Quinn
2021;9:725231. JE, Carman GM, et al. The CWH8 gene encodes a
70. Sagami H, Kurisaki A, Ogura K. Formation of doli- dolichyl pyrophosphate phosphatase with a lumi-
chol from dehydrodolichol is catalyzed by NADPH-­ nally oriented active site in the endoplasmic reticu-
dependent reductase localized in microsomes of rat lum of Saccharomyces cerevisiae. J. Biol. Chem.
liver. J. Biol. Chem. 1993;268(14):10109–13. 2001;276(44):41455–64.
71. Medina-Cano D, Ucuncu E, Nguyen LS, Nicouleau 82. Volpe JJ, Sakakihara Y, Rust RS. Dolichol kinase and
M, Lipecka J, Bizot JC, et al. High N-glycan multi- the regulation of dolichyl phosphate levels in develop-
plicity is critical for neuronal adhesion and sensitizes ing brain. Brain Res. 1987;428(2):193–200.
the developing cerebellum to N-glycosylation defect. 83. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog
Elife. 2018;7:7. C, Oksvold P, Mardinoglu A, et al. Proteomics.
72. Taylor RL, Arno G, Poulter JA, Khan KN, Morarji J, Tissue-based map of the human proteome. Science.
Hull S, et al. Association of steroid 5alpha-­reductase 2015;347(6220):1260419.
type 3 congenital disorder of glycosylation with 84. William James A, Ravi C, Srinivasan M, Nachiappan
early-onset retinal dystrophy. JAMA Ophthalmol. V. Crosstalk between protein N-glycosylation and
2017;135(4):339–47. lipid metabolism in Saccharomyces cerevisiae. Sci.
73. Grundahl JE, Guan Z, Rust S, Reunert J, Muller B, Rep. 2019;9(1):14485.
Du Chesne I, et al. Life with too much polyprenol: 85. Helander A, Stodberg T, Jaeken J, Matthijs G,
polyprenol reductase deficiency. Mol. Genet. Metab. Eriksson M, Eggertsen G. Dolichol kinase deficiency
2012;105(4):642–51. (DOLK-CDG) with a purely neurological presenta-
74. Wheeler PG, Ng BG, Sanford L, Sutton VR, tion caused by a novel mutation. Mol. Genet. Metab.
Bartholomew DW, Pastore MT, et al. SRD5A3-CDG: 2013;110(3):342–4.
expanding the phenotype of a congenital disorder of 86. Kranz C, Jungeblut C, Denecke J, Erlekotte A,
glycosylation with emphasis on adult onset features. Sohlbach C, Debus V, et al. A defect in dolichol
Am. J. Med. Genet. A. 2016;170(12):3165–71. phosphate biosynthesis causes a new inherited disor-
75. Marchler-Bauer A, Derbyshire MK, Gonzales der with death in early infancy. Am. J. Hum. Genet.
NR, Lu S, Chitsaz F, Geer LY, et al. CDD: NCBI’s 2007;80(3):433–40.
Retinal Metabolic Profile on IMPG2
Deficiency Mice with Subretinal
Lesions

Rong Xu, Yekai Wang, Jianhai Du,


and Ezequiel M. Salido

Abstract energy metabolism in mice retinas of IMPG2


KO mice with subretinal lesion.
The interphotoreceptor matrix (IPM) is the
extracellular matrix between the photorecep- Keywords
tors and the retinal pigment epithelium (RPE).
The IPM has two proteoglycans: the IPM pro- Interphotoreceptor matrix · Metabolomics ·
teoglycans 1 and 2 (IMPG1 and IMPG2, Vitelliform macular dystrophy · IMPG2 ·
respectively). Patients with mutations on Retina · Metabolite
IMPG2 develop subretinal vitelliform lesions
that affect vision. We previously created an
IMPG2 knockout (KO) mice model that gen- 1 Introduction
erates subretinal lesions similar to those found
in humans. These subretinal lesions in IMPG2 In between the photoreceptors and interfacing
KO mice retinas are, in part, composed of mis- with the retinal pigment epithelium (RPE) lies an
localized IMPG1. In addition, IMPG2 KO extracellular matrix called interphotoreceptor
mice show microscopic IMPG1 material accu- matrix (IPM). Activities crucial to vision occur
mulation between the RPE and the photore- within IPM, including the trafficking of nutrients
ceptor outer segments. In this work we discuss and metabolites [12]. The IPM has two unique
the possibility that material accumulation on proteoglycans: the IPM proteoglycans 1 and 2
IMPG2 KO mice retinas affects photoreceptor (IMPG1 and IMPG2, respectively), also known
metabolism. To further investigate this idea, as SPACR and SPACRCAN or IPM150 and
we used targeted metabolomics to profile reti- IPM200, respectively [9, 12]. Humans with
nal metabolome on IMPG2 KO mice. The mutations in IMPG2 develop, over time, vision
metabolite set enrichment analysis showed loss characterized by subretinal vitelliform
reduced glutamate metabolism, urea cycle, lesions, predominantly in the macula, named
and galactose metabolism suggesting affected vitelliform macular dystrophy (VMD) [5, 15,
21]. Previously, our lab created an animal model
lacking IMPG2 that generates subretinal lesions
R. Xu · Y. Wang · J. Du · E. M. Salido (*) similar to those found in humans. In the absence
Departments of Biochemistry and molecular of IMPG2, abnormal accumulation of material
medicine, and Ophthalmology and Visual Sciences, constituted by IMPG1 and chondroitin sulfate
West Virginia University, Morgantown, WV, USA was found between the photoreceptors and RPE.
e-mail: ezequiel.salido@hsc.wvu.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 457
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_67
458 R. Xu et al.

Over time, these mutant mice develop subretinal cold HBSS solution for retina isolation. Once
lesions detectable by optical coherence tomogra- collected in microtubes, two retinal tissues were
phy (OCT), glial reactivity, and migration of snap-frozen in liquid N2. Five animals from the
microglia focalized in the area of the subretinal same litter were used. Two IMPG2 KO mice with
lesion [16]. confirmed subretinal lesions in both eyes by opti-
The primordial nutrition of the photoreceptors cal coherence tomography (OCT) and three
comes from the RPE [6, 11]. All nutrients and IMPG2 heterozygous mice were used as control.
metabolites that flow between photoreceptors Total of N = 4 retinas for IMPG2 KO and N = 6
and RPE must pass through the IPM between retinas for the control group.
these cells. We think that pathological diseases
that alter the functional-structural properties of
the IPM can impair molecules exchange. This 2.3 Mass Spectrometry Sample
work uses a mice model with disrupted IPM to Preparation and Metabolite
evaluate possible metabolic impairment of the Analysis by LC-MS/MS
retina. To address this, we used targeted metabo-
lomics using mass spectrometry (MS) coupled Samples were processed and analyzed as
with liquid chromatography (LC) to evaluate 241 described in Grenell et al. [7] and Li et al. [14].
metabolites in mutant and control mice retinas. Briefly, retinas were homogenized in pre-chilled
80% methanol to extract metabolites. The extracts
were dried and reconstituted for LC-MS/MS
2 Materials and Methods analysis. Metabolite extracts were analyzed by a
Shimadzu LC Nexera X2 UHPLC coupled with a
2.1 Animals QTRAP 5500 LC-MS/MS (AB Sciex). An
ACQUITY UPLC BEH Amide analytic column
The IMPG2 KO animal model used in this study (2.1 × 50 mm, 1.7 μm) (Waters Corp., Milford,
was generated by Crispr-Cas9 technology as MA) was used for chromatographic separation.
described by Salido and Ramamurthy [16]. The mobile phase was (A) water with 10 mM
Briefly, heterozygote animals were backcrossed ammonium acetate (pH 8.9) and (B) acetonitrile/
with C57BL/6 J (The Jackson Laboratory). All water (95/5) with 10 mM ammonium acetate
animals were generated at the Transgenic Core of (pH 8.2) (all solvents were LC-MS Optima grade
West Virginia University and did not contain rd1 from Fisher Scientific). The total run time was
or rd8 alleles. Mice of both sexes at 8 months old 11 min. With a flow rate of 0.5 ml/min. and an
were used in this experiment. The animals were injection volume of 5 μl. The gradient elution was
maintained under 12-h light/dark cycles with 95–61% B in 6 min, 61–44% B at 8 min, 61–27%
food and water provided ad libitum. All experi- B at 8.2 min, and 27–95% B at 9 min. The column
mental procedures involving animals in this study was equilibrated with 95% B at the end of each
were approved by the Institutional Animal Care run. The source and collision gas was N2. The ion
and Use Committee of West Virginia University. source conditions in positive and negative mode
were curtain gas (CUR) = 25 psi, collision gas
(CAD) = high, ion spray voltage (IS) = 3800/-
2.2 Retina Collection 3800 volts, temperature (TEM) = 500 °C, ion
source gas 1 (GS1) = 50 psi, and ion source gas 2
Retinas were collected as described in Wang (GS2) = 40 psi. Each metabolite was tuned with
et al. [20]. Briefly, mice were euthanized and standards for optimal transitions. 13C-nicotinic
eyes enucleated and placed on wet filter papers acid (Toronto Research Chemicals) was used as
presoaked with cold HBSS on ice. The anterior the internal standard. The extracted MRM peaks
half of the eye was removed, and the posterior were integrated using MultiQuant 3.0.3 software
half was moved and submerged in a drop of 50 μl (AB Sciex, Concord, ON, CA).
Retinal Metabolic Profile on IMPG2 Deficiency Mice with Subretinal Lesions 459

2.4 Statistical Analysis datasets shows two distinct groups with different
metabolic profiles (Fig. 1).
Differences between means were determined by To identify significantly changed metabolites,
unpaired two-tailed T-tests. The principal compo- we perform a T-test statistical analysis over all
nent analysis and partial least squares-­ the target metabolites analyzed between groups.
discriminant analysis (PLS-DA) (P < 0.05 on raw This study identified 11 different significant
data, unpair) were analyzed using MetaboAnalyst metabolites described in Table 1. All identified
5.0 (http://www.metaboanalyst.ca/). Enrichment metabolites are significantly reduced in the KO
analysis for significantly altered metabolic path- group with respect to the control. In order to visu-
ways was performed with MetaboAnalysts. alize and evaluate metabolites changes from indi-
vidual samples, we performed a fold change
hierarchical cluster analysis for each sample
3 Results using the top 25 most statistically different sam-
ples by T-test (Fig. 2). The heatmap represents
To evaluate the metabolic homeostasis of retinas the fold change for each sample using a color
with altered IPM and subretinal lesions, we per- code.
formed a metabolomics analysis comparing reti- To identify biologically meaningful pathways
nas from heterozygous IMPG2 KO mice as a affected on retinas from mutant mice, we per-
control group and homozygous IMPG2 KO mice. form a metabolite set enrichment analysis
We first evaluate for general differences between (Fig. 3). The top three most significantly enriched
control and KO groups using a PLS-DA analysis. metabolite sets are glutamate metabolism, urea
The score plot analysis on individual samples cycle, and galactose metabolism.

Fig. 1 Distinctive
metabolic profiles
between control and
IMPG2 KO retinas.
Score plot of retina
metabolites separated
into two different groups
by PLS-DA. The
explained variances are
shown in brackets
460 R. Xu et al.

Table 1 Individual statistically significant molecules by T-test


Important molecules identified by T-tests
Metabolite t.stat p.value FDR FC log2(FC)
1 cGMP 4.429 0.0022 0.205 1.35 0.433
2 Malate 3.784 0.0054 0.236 1.20 0.266
3 Nicotinamide 3.386 0.0096 0.236 1.39 0.475
4 Alanine 3.344 0.0102 0.236 1.18 0.244
5 Taurine 3.167 0.0132 0.246 1.31 0.387
6 Myo-inositol 2.752 0.0250 0.304 1.13 0.172
7 Erythritol 2.744 0.0253 0.304 1.30 0.381
8 UDP 2.722 0.0262 0.304 1.30 0.382
9 3-Aminoisobutyrate 2.481 0.0380 0.363 1.22 0.285
10 GMP 2.464 0.0391 0.363 1.36 0.441
11 Aspartic acid 2.308 0.0498 0.421 1.18 0.240
Fold change (FC) obtained as control/KO. t.stat t-statistic, FDR false discovery rate

Fig. 2 Metabolites’ hierarchical cluster analysis for each are fold change of ion intensity of the KO vs. control.
sample shown as a heatmap. The data were clustered for N = 10 from five animals in total
similar patterns using MetaboAnalyst 5.0 software. Data
Retinal Metabolic Profile on IMPG2 Deficiency Mice with Subretinal Lesions 461

Fig. 3 Most enriched affected metabolic pathways on IMPG2 KO retinas. Top metabolite patterns plot listed based on
lower P-value and colored coded. The enrichment ratio is visualized as the size of the circles

4 Discussion RPE generate a negative impact on the photore-


ceptors’ health [6]. Mice with IMPG2 deletion
In this work, we identified glutamate metabolism, develop extracellular matrix material accumula-
urea cycle, and galactose metabolism as the most tion and subretinal lesions between the photore-
affected pathways in the retina of mutant mice. ceptors and the RPE. At 8 months old, the retina
These findings suggest that retinas with altered function of these mice is significantly reduced
IPM and subretinal lesions create changes in the without major changes in the retina. We hypoth-
retina metabolic homeostasis. esize that the observed decrease in GMP and
cGMP are directly related to a decrease in the
photoreceptor outer segment health. A detailed
4.1 Altered analysis of photoreceptors outer segments’ length
IPMInterphotoreceptor matrix needs to be done to confirm this hypothesis.
(IPM) and Retina Metabolism The neural retina consumes glucose, aspartate,
glutamate, citrate, nicotinamide, and other
The photoreceptors cells are the most abundant nutrients to sustain its active glycolysis, mito-
cell type in the retina as well as one of the cells chondrial oxidative metabolism, and neurotrans-
with higher demand for nutrients in the body mitter synthesis [14]. We consider that the
[14]. Nutrient deficits between these cells and decrease in the glutamate metabolism, urea cycle,
462 R. Xu et al.

and galactose metabolism is directly linked to performed in younger IMPG2 KO mice before
photoreceptor nutrient stress product of the subretinal lesion formation. In addition, 8-month-­
affected IPM. On the other hand, carnitine plays old mutant mice present microglia migration and
a vital role in transporting long-chain CoA into Müller cell reactivity [16]. A major limitation in
the mitochondria for oxidation [20]. In this sense, the data interpretation arises due to the impossi-
the only three positively enriched metabolites in bility to separate the metabolic changes origi-
the mutant retinas are acetylcarnitine, spermine, nated from the immune response from those
and 2-methylbutyroylcarnitine indicating dys- changes inherent to nutrient stress. Hence, test
regulation of lipid metabolism. metabolomics in younger mice will help to
reduce the influence of the immune response.

4.2 Experimental Limitations


and Future Directions 4.3 Subretinal Lesions, Nutrient
Stress, and Macular
The data gathered in this work help to understand Degeneration
the photoreceptor function reduction in IMPG2
KO mice; nevertheless, it possesses several limi- The extracellular matrices tend to lose their
tations that need to be addressed in further work. mechanical and hydration properties over time [3].
In this work, heterozygous IMPG2 littermate During aging, the retina metabolism changes [20].
mice have been used as a control reference. Unstudied age-related changes in the IPM may
Heterozygous mice may present unknown defects contribute to changes in vision observed with
in the IPM structure that also affect the molecules aging [2]. Patients suffering from age-­related mac-
interchange thought the IPM. In this case, the dif- ular degeneration (AMD) can accumulate material
ferences found in this work between IMPG2 KO within the IPM called subretinal drusenoid depos-
and control are underestimated. Further its (SDDs) similar to those found in IMPG2 KO
experiments using wild-type mice as control
­ mice. SDDs are thought as independent risk fac-
could yield greater and new significant metabo- tors for late AMD. Geographic atrophy and cho-
lite differences between groups. On the other roidal neovascularization are particularly
hand, this work used both eyes from the same associated with SDD [1, 8, 17, 18]. Furthermore,
animal as separate data points. Further experi- patients with SDDs without signs of AMD have an
ments need to pool both retinas in one sample increased risk of disease development [10, 19]. In
and increase the sample group size to five for addition, the photoreceptors from patients with
each group. AMD exhibit an increased expression of glyco-
The photoreceptors and the RPE work in lytic genes in addition to an altered metabolic
combination to interchange nutrients and marker profile suggesting nutrient stress during
metabolic waste. The RPE metabolic homeostasis disease [4, 13]. In this sense, retina metabolism
can potentially be affected by defects in the IPM. disbalance and subretinal lesions developed in
In future studies, we will evaluate the metabolism IMPG2 KO animals place these mutant mice as a
of the RPE along with the retina. promising tool to study the role of subretinal
Mice without IMPG2 generate subretinal lesions in the AMD context.
lesions after 5 months of age but possess material
accumulation between photoreceptors and RPE Acknowledgments The Knight Templar Eye Foundation
as young as 45 days old. To dissect the influence (EMS) and West Virginia Lions and Lions Club
International Foundation supported this work. We thank
of the subretinal lesions and the micro-material Scott Rhodes and Benjamin Mitchell for support in the
accumulation, metabolomics analysis could be maintenance of animal models and genotyping.
Retinal Metabolic Profile on IMPG2 Deficiency Mice with Subretinal Lesions 463

References 12. Ishikawa M, Sawada Y, Yoshitomi T. Structure


and function of the interphotoreceptor matrix
surrounding retinal photoreceptor cells. Exp Eye Res.
1. Alten F, Eter N. Current knowledge on reticular
2015;133:3–18.
pseudodrusen in age-related macular degeneration. Br
13. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Tucker
J Ophthalmol. 2015;99:717–22.
J, Marc RE. Retinal remodeling and metabolic
2. Al-Ubaidi MR, Naash MI, Conley SM. A perspective
alterations in human AMD. Front Cell Neurosci.
on the role of the extracellular matrix in progressive
2016;10:103.
retinal degenerative disorders. Invest Ophthalmol Vis
14. Li B, Zhang T, Liu W, Wang Y, Xu R, Zeng S, Zhang
Sci. 2013;54:8119–24.
R, Zhu S, Gillies MC, Zhu L, Du J. Metabolic features
3. Birch HL. Extracellular matrix and ageing. Subcell
of mouse and human retinas: rods versus cones,
Biochem. 2018;90:169–90.
macula versus periphery, retina versus RPE. iScience.
4. Cheng SY, Cipi J, Ma S, Hafler BP, Kanadia RN, Brush
2020;23:101672.
RS, Agbaga MP, Punzo C. Altered photoreceptor
15. Meunier I, Manes G, Bocquet B, Marquette V,
metabolism in mouse causes late stage age-related
Baudoin C, Puech B, Defoort-Dhellemmes S, Audo
macular degeneration-like pathologies. Proc Natl
I, Verdet R, Arndt C, Zanlonghi X, Le Meur G,
Acad Sci U S A. 2020;117:13094–104.
Dhaenens CM, Hamel CP. Frequency and clinical
5. Chowers I, Tiosano L, Audo I, Grunin M, Boon
pattern of vitelliform macular dystrophy caused by
CJ. Adult-onset foveomacular vitelliform dystrophy: a
mutations of interphotoreceptor matrix IMPG1 and
fresh perspective. Prog Retin Eye Res. 2015;47:64–85.
IMPG2 genes. Ophthalmology. 2014;121:2406–14.
6. Du J, Zhu S, Lim RR, Chao JR. Proline metabolism
16. Salido EM, Ramamurthy V. Proteoglycan IMPG2
and transport in retinal health and disease. Amino
shapes the Interphotoreceptor matrix and modulates
Acids. 2021;53:1789.
vision. J Neurosci. 2020;40:4059–72.
7. Grenell A, Wang Y, Yam M, Swarup A, Dilan TL,
17. Sivaprasad S, Bird A, Nitiahpapand R,
Hauer A, Linton JD, Philp NJ, Gregor E, Zhu S,
Nicholson L, Hykin P, Chatziralli I, Moorfields
Shi Q, Murphy J, Guan T, Lohner D, Kolandaivelu
UCLAMDC. Perspectives on reticular pseudodrusen
S, Ramamurthy V, Goldberg AFX, Hurley JB, Du
in age-related macular degeneration. Surv
J. Loss of MPC1 reprograms retinal metabolism to
Ophthalmol. 2016;61:521–37.
impair visual function. Proc Natl Acad Sci U S A.
18. Spaide RF, Ooto S, Curcio CA. Subretinal drusenoid
2019;116:3530–5.
deposits AKA pseudodrusen. Surv Ophthalmol.
8. Hogg RE. Reticular pseudodrusen in age-related
2018;63:782–815.
macular degeneration. Optom Vis Sci. 2014;91:854–9.
19. Spaide RF, Yannuzzi L, Freund KB, Mullins R,
9. Hollyfield JG, Rayborn ME, Midura RJ, Shadrach
Stone E. EYES WITH SUBRETINAL DRUSENOID
KG, Acharya S. Chondroitin sulfate proteoglycan
DEPOSITS AND NO DRUSEN: progression of
core proteins in the interphotoreceptor matrix:
macular findings. Retina. 2019;39:12–26.
a comparative study using biochemical and
20. Wang Y, Grenell A, Zhong F, Yam M, Hauer A,
immunohistochemical analysis. Exp Eye Res.
Gregor E, Zhu S, Lohner D, Zhu J, Du J. Metabolic
1999;69:311–22.
signature of the aging eye in mice. Neurobiol Aging.
10. Huisingh C, McGwin G Jr, Neely D, Zarubina
2018;71:223–33.
A, Clark M, Zhang Y, Curcio CA, Owsley C. The
21. Wilde C, Lakshmanan A, Patel M, Morales MU,
association between subretinal Drusenoid deposits in
Dhar-Munshi S, Amoaku WM. Prevalence of reticular
older adults in Normal macular health and incident
pseudodrusen in newly presenting adult onset
age-­related macular degeneration. Invest Ophthalmol
foveomacular vitelliform dystrophy. Eye (Lond).
Vis Sci. 2016;57:739–45.
2016;30:817–24.
11. Hurley JB, Lindsay KJ, Du J. Glucose, lactate, and
shuttling of metabolites in vertebrate retinas. J
Neurosci Res. 2015;93:1079–92.
Part X
Neuroprotection
Glutathione Coating of Liposomes
Enhances the Delivery
of Hydrophilic Cargo to the Inner
Nuclear Layer in Retinal Cultures

Gustav Christensen and François Paquet-Durand

Abstract in the retinal inner vasculature, but further


studies are needed for verification.
To successfully deliver intracellular
compounds to retinal cells, a delivery system Keywords
based on purified lipids, self-assembled into
Liposomes · Retinal explant cultures ·
synthetic vesicles called liposomes, can be
Blood-retinal barrier · Glutathione
used. Liposomes have the potential to target
distinct tissues and cells in the body by molec-
ular targeting moieties conjugated to their sur-
face. To enhance liposome delivery to neurons, 1 Introduction
glutathione has formerly been used as target-
ing moiety. It is unclear whether and how the Liposomes are lipid-based submicron vesicles
glutathione conjugation improves the (20–1000 nm) able to encapsulate drug molecules.
liposome-­induced uptake to cells within the Liposomal encapsulation may prolong drug
retina. To explore this, glutathione-liposomes release and decrease drug treatment intervals,
were prepared and loaded with a fluorescent and can be used to target specific cell types or
tracer, which was added to organotypic retinal tissues [2, 3, 5, 7]. Typically, coating the
explant cultures derived from mice. The fluo- liposomes with highly hydrophilic polymers,
rescence in the tissue was analyzed from his- e.g., poly(ethylene glycol) (PEG), is done to
tological sections using fluorescent shield the liposome surface, so as to limit immune
microscopy. Comparisons were done with responses [9]. Liposomes with glutathione
liposomes without a targeting device and conjugated to the end of the PEG chains have
cysteine-­conjugated liposomes. A significant previously been used to target neurons in the
increase (p ≤ 0.05) of fluorescent signal was brain, and have shown an enhanced permeation
observed from the inner nuclear layer of reti- through the blood-brain barrier [6, 8]. Not much
nas exposed to glutathione-conjugated lipo- is known about the specific mechanism of uptake,
somes. Qualitatively, this might be attributed however. One hypothesis is that glutathione, due
to the accumulation of glutathione-liposomes to preferred uptake in brain endothelial cells,
allows the liposomes to be taken up from the
G. Christensen · F. Paquet-Durand (*) plasma side and transport the encapsulated cargo
Institute for Ophthalmic Research, University of to the brain [8]. Whether this would be the same
Tübingen, Tübingen, Germany for the retina is not well understood even though
e-mail: francois.paquet-durand@uni-tuebingen.de

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 467
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_68
468 G. Christensen and F. Paquet-Durand

glutathione-­conjugated liposomes have also 3 Methods


shown improved delivery of neuroprotective
compounds to the in vivo retina in mouse models 3.1 Liposome Preparation
of retinal degeneration, when compared to and Encapsulation
administration of the free drug [12].
Here, we performed an exploratory study to Liposomes were prepared using a published
test the uptake of glutathione-conjugated lipo- protocol [13], which will be briefly summa-
somes in organotypic retinal explant cultures rized here. The lipids POPC, cholesterol, and
derived from wild-type mice. As controls, we DSPE-­mPEG2000 or DSPE-PEG2000-maleimide,
used liposomes with unconjugated PEG chains dissolved in chloroform, were mixed in a
(mPEG), as well as liposomes with cysteine con- round-bottom flask in the molar ratio of 63.3:
jugations. We analyzed where in the tissue the 31.7: 5 (total mass = 1 mg of dried material).
different types of liposomes accumulated prefer- The chloroform was removed from the mixture
entially, and which cell types showed enhanced on a rotary evaporator (model RC600, KNF
uptake of a fluorescent tracer. Neuberger, Trenton, NJ, USA), operating at a
speed of 100 rpm and a pressure of 300 mbar
for at least 1 h. Afterwards, a thin film of dried
2 Materials lipids remained on the bottom of the curved
flask. 1 mL of calcein (2 mM in PBS) was
1 - P a l m i t o y l - 2 - o l e o y l - s n - g l y c e r o - 3 -­ added to the lipids, and the flask vortexed, until
phosphocholine (POPC), 3β-hydroxy-5-­ all the lipids were visibly dissolved. The mix-
cholesten (cholesterol), 1,2-distearoyl- ture was extruded using a mini-extruder (Avanti
s n - g l y c e r o - 3 -­­p h o s p h o e t h a n o l a m i n e - N - Polar Lipids, Alabaster, AL, USA) through a
[(polyethylene glycol)-2000] (ammonium salt) 100 nm porous polycarbonate membrane
(DSPE-mPEG2000), 1,2-distearoyl-sn-glycero- between 2 filter supports back and forth at least
3-­­phosphoethanolamine-N-[maleimide(polyethy 11 times. For samples containing DSPE-
lene glycol)-2000] (ammonium salt) (DSPE- PEG2000-maleimide, 20 μL of either L-cysteine
PEG2000-maleimide), anhydrous chloroform (≥ or reduced glutathione (1 mg/mL in maleimide-
99%) with 0.5–1% ethanol, paraformaldehyde, thiol reaction buffer) was added and incubated
sucrose, disodium hydrogen phosphate dihy- with the samples for 2 h at room temperature to
drate, sodium dihydrogen phosphate monohy- produce cysteine-coated liposomes (Lp-Cys) or
drate, sodium chloride, hydrogen chloride, glutathione-coated liposomes (Lp-Glu), respec-
sodium hydroxide, Tris(2-­carboxyethyl)phos- tively. Alternatively, 20 μL of PBS was added to
phine hydrochloride (TCEP), L-cysteine, reduced the liposomes with DSPC-­ mPEG2000
glutathione, and calcein were all obtained from (Lp-Control). Excess L-cysteine or glutathione
Sigma-Aldrich (Darmstadt, Germany). and nonencapsulated calcein were removed
Buffers: Phosphate-buffered saline (PBS): from the solutions by dialysis against 150 mM
10 mM disodium hydrogen phosphate dihydrate, NaCl (Slide-A-Lyzer Dialysis Cassettes, 10 K
1.8 mM sodium dihydrogen phosphate monohy- MWCO, cellulose membrane, Thermo Fisher
drate, and 137 mM sodium chloride in miliQ Scientific, Waltham, MA, USA). Afterwards,
water. Maleimide-thiol reaction buffer: 10 mM the liposomes were sterile filtered (Millex-GP,
TCEP in miliQ water. All buffers were adjusted 0.22 μm, polyethersulfon, Merck Millipore,
to pH 7.4 with HCl and NaOH using an elec- Darmstadt, Germany) and stored at 2–8 °C until
tronic pH meter. further use.
Glutathione Coating of Liposomes Enhances the Delivery of Hydrophilic Cargo to the Inner Nuclear Layer… 469

3.2 Liposome Uptake following retinal layers: ganglion cell layer


in Organotypic Retinal (GCL), inner plexiform layer (IPL), inner nuclear
Explant Cultures layer (INL), outer nuclear layer (ONL), and the
outer and inner photoreceptor segments using the
Organotypic retinal explants derived from wild-­ acquisition software (ZEN 2.6, Zeiss). This sig-
type mice at postnatal day (P) 13 were cultured nal was divided by the area of the corresponding
following an established protocol [1, 10]. Briefly, layer and compared across the three types of lipo-
the eyes were enucleated and the isolated retinas somes tested. GraphPad Prism 9 (GraphPad
placed in a Transwell insert (0.4 μm, polycarbon- Software, San Diego, CA, USA) was used to
ate, Corning-Costar, New York, NY, USA) in a present the data and perform statistical tests.
6-well plate with the attaching RPE layer facing
down. Serum-free R16 medium (Thermo Fisher
Scientific, Waltham, MA, USA) was added from 4 Results and Discussion
underneath the retinal cultures and replaced every
second day. At P15, a 20 μL solution of lipo- To assess how different targeting components,
somes (100 μg/mL) with encapsulated calcein conjugated to the surface of dye-loaded lipo-
(see above) was added dropwise on the top of the somes, would affect their distribution in the ret-
culture. This has been done previously to exam- ina, glutathione-coated, cysteine-coated, and
ine the effect of nanoparticles entering the retina PEGylated control liposomes were tested and
from the vitreous side as they would during an compared. Since not much was known about how
intravitreal injection [4, 11]. After 6 h of incuba- glutathione coating of liposomes benefits drug
tion, the retinas were fixed in a 4% paraformalde- delivery to the retina, we focused on how gluta-
hyde/PBS solution for 45 min, followed by thione affected cellular uptake and distribution
cryoprotection in solutions of incremental within the retina. For comparison, we used also
amounts of sucrose (10% sucrose for 10 min, cysteine conjugation because this molecule
20% sucrose for 20 min, and 30% sucrose at shares structural similarities with glutathione
2–8 °C overnight) and submerged in blocks of (Fig. 1a), thus allowing to assess the specificity
embedding medium (Tissue-Tek of glutathione. The retinal explants were cultured
O.C.T. Compound, Sakura Finetek Europe, for a period of 2 days between explantation and
Alphen aan den Rijn, Netherlands), which was treatment, so as to allow proper assessment of
then frozen in liquid nitrogen. The frozen blocks retinal culture quality and viability. Liposomes
were sectioned on a cryostat (NX50, Thermo loaded with the green dye calcein were then
Fisher, Waltham, MA, USA) to produce 14 μm added to retinas derived from mice and incubated
sections on Superfrost Plus object slides for 6 h (Fig. 1b). A relatively short incubation
(R. Langenbrinck, Emmendingen, Germany) time was chosen since longer culturing periods
from the center of the retinal culture (in the range were deemed to less well represent native condi-
of 1700–2900 μm from the edge). The sections tions (e.g., ganglion and endothelial cells in cul-
were washed in PBS, stained with DAPI ture are degenerating over a time course of
(Vectashield with DAPI, Vector laboratories, 4 days). Another reason for the short incubation
Burlingame, CA, USA), and imaged with fluo- time was that, over longer periods of time, cal-
rescence microscope (Axio Imager Z2 with cein might (passively or actively) redistribute
ApoTome function, Zeiss, Oberkochen, between cells, which would make it harder to dis-
Germany). A CCD camera with a 20X objective tinguish specific effects of the targeting compo-
was used. A blue filter was used to measure the nents. After the incubation period, the tissues
DAPI signal (Ex./Em. 353/465 nm), and a green were sectioned and imaged, using the same
filter was used to measure the calcein signal (Ex./ parameters across all samples (Fig. 1c). The aver-
Em. 493/517 nm). The signal intensity from the age calcein signal was measured from different
green channel was measured coming from the retinal layers (Fig. 1c iv). Since the liposomes
470 G. Christensen and F. Paquet-Durand

Fig. 1 Distribution of cysteine- and glutathione-­ loaded with calcein were added on top of the cultures
conjugated liposomes in organotypic retinal explant (from the ganglion cell layer side) at postnatal day 15, and
cultures the cultures were fixed after a 6-h incubation period. (c)
(a) Structure of liposome built from 1-palmitoyl-2-oleoyl- Calcein signal measured from cross sections obtained
sn-glycero-3-phosphocholine (POPC), cholesterol, and from retinal explant cultures, where (i) Lp-control/cal-
1,2-distearoyl-sn-glycero-3-­­p hosphoethanolamine cein, (ii) Lp-cysteine/calcein, or (iii) Lp-glutathione/cal-
(DSPE) with 2 kDa poly(ethylene glycol) (2000PEG) cein was added. Scale bar = 50 μm. (iv) Averaged calcein
conjugated to either of three different chemical groups signal quantified across retinal layers and expressed as
(R): either R = methoxyl group (OMe) in the formulation mean ± SD for n = 3–5. *p < 0.05. Images shown in C are
Lp-control, R = cysteine in Lp-cysteine, or R = glutathi- representative for explant cultures obtained from n = 3–5
one in Lp-glutathione. (b) Experimental design. animals. Statistical analysis: One-way ANOVA with
Organotypic retinal explant cultures derived from mice Tukey’s post hoc test
were isolated and cultured at postnatal day 13. Liposomes

were added to the cultures from the side closest signal coming from the INL could be detected in
to the ganglion cell layer, we saw the most abun- cultures to which Lp-glutathione was added
dant fluorescence signal there, while signals from (Fig. 1c iv), indicating that the glutathione conju-
the deeper retinal layers were gradually decreas- gation might assist liposomes in reaching deeper
ing. An exception was the inner/outer segments into the retinal tissue. The large green areas might
(IOS), where the signal appeared to be stronger relate to the inner retinal vasculature, suggesting
than signal from the inner or outer nuclear that Lp-glutathione was accumulating in the
layers. endothelial cells within the retina. In the explant
Remarkably, Lp-glutathione seemed to be culture system, the outer vasculature is removed
localized in large depots between the INL and prior to culturing. Thus, if indeed conjugated
ONL (Fig. 1c iii), and a significant increase in the liposomes were targeting endothelial cells, gluta-
Glutathione Coating of Liposomes Enhances the Delivery of Hydrophilic Cargo to the Inner Nuclear Layer… 471

thione conjugation could potentially help to T, Reinisalo M, Itkonen J, Toropainen E, Casteleijn


M, Kidron H, Antopolsky M, Vellonen K-S, Ruponen
deliver cargo across the inner blood-retinal M, Urtti A. Pharmacokinetic aspects of retinal drug
­barrier. Alternatively, the accumulated fluorescent delivery. Prog Retin Eye Res. 2017;57:134–85.
signal could relate to horizontal cells, which 6. Hu Y, Gaillard PJ, de Lange ECM, Hammarlund-­
reside in the INL, in close proximity to the Udenaes M. Targeted brain delivery of methotrexate
by glutathione PEGylated liposomes: how can
OPL. In the future, it will be interesting to label the formulation make a difference? Eur J Pharm
cells with distinct markers in the retinal tissue Biopharm. 2019;139:197–204.
sections (e.g., RBPMS expressed in certain types 7. Khiev D, Mohamed ZA, Vichare R, Paulson R, Bhatia
of ganglion cells, CD31 expressed in the capillary S, Mohapatra S, Lobo GP, Valapala M, Kerur N,
Passaglia CL, Mohapatra SS, Biswal MR. Emerging
endothelium, or calbindin expressed in horizontal nano-formulations and nanomedicines applications
cells) to investigate potential co-localization with for ocular drug delivery. Nano. 2021;11(1)
the liposome-delivered dye. 8. Maussang D, Rip J, van Kregten J, van den Heuvel A,
van der Pol SMA, van der Boom B, Reijerkerk A, Chen
L, de Boer M, Gaillard PJ, de Vries HE. Glutathione
Acknowledgments We are grateful for excellent
conjugation dose-dependently increases brain-­
technical support from Norman Rieger. This study was
specific liposomal drug delivery in vitro and in vivo.
supported by grants from the Tistou and Charlotte Kerstan
Drug Discov Today Technol. 2016;20:59.
Foundation and the Deutsche Forschungsgemeinschaft
9. Nosova AS, Koloskova OO, Nikonova AA, Simonova
(DFG; PA1751/10-1).
VA, Smirnov VV, Kudlay D, Khaitov MR. Diversity
of PEGylation methods of liposomes and their
influence on RNA delivery. Med Chem Comm.
References 2019;10(3):369–77.
10. Prajapati M, Christensen G, Paquet-Durand F,
Loftsson T. Cytotoxicity of β-cyclodextrins in retinal
1. Belhadj S, Tolone A, Christensen G, Das S, Chen Y,
explants for intravitreal drug formulations. Molecules.
Paquet-Durand F. Long-term, serum-free cultivation
2021;26(5)
of Organotypic mouse retina explants with intact
11. Sen M, Al-Amin M, Kicková E, Sadeghi A, Puranen
retinal pigment epithelium. J Vis Exp. 2020;
J, Urtti A, Caliceti P, Salmaso S, Arango-Gonzalez
2. Blazaki S, Pachis K, Tzatzarakis M, Tsilimbaris M,
B, Ueffing M. Retinal neuroprotection by controlled
Antimisiaris SG. Novel liposome aggregate platform
release of a VCP inhibitor from self-assembled
(LAP) system for sustained retention of drugs in
nanoparticles. J Control Release. 2021;339:307–20.
the posterior ocular segment following intravitreal
12. Vighi E, Trifunović D, Veiga-Crespo P, Rentsch A,
injection. Int J Pharm. 2020;576:118987.
Hoffmann D, Sahaboglu A, Strasser T, Kulkarni M,
3. Bourlaist CL, Chevanne F, Ropert P, Bretagne G, Acar
Bertolotti E, van den Heuvel A, Peters T, Reijerkerk
L, Zia H, Sado PA, Needham T, Leverge R. Release
A, Euler T, Ueffing M, Schwede F, Genieser H-G,
kinetics of liposome-encapsulated ganciclovir after
Gaillard P, Marigo V, Ekström P, Paquet-Durand
intravitreal injection in rabbits. J Microencapsul.
F. Combination of cGMP analogue and drug delivery
1996;13(4):473–80.
system provides functional protection in hereditary
4. Christensen G, Barut L, Urimi D, Schipper N, Paquet-­
retinal degeneration. Proc Natl Acad Sci U S A.
Durand F. Investigating ex vivo animal models to
2018;115(13)
test the performance of intravitreal liposomal drug
13. Zhang H. Thin-film hydration followed by extrusion
delivery systems. Pharmaceutics. 2021;13(7)
method for liposome preparation. In: D’Souza GGM,
5. del Amo EM, Rimpelä A-K, Heikkinen E, Kari
editor. Liposomes: methods and protocols. New York:
OK, Ramsay E, Lajunen T, Schmitt M, Pelkonen L,
Springer; 2017. p. 17–22.
Bhattacharya M, Richardson D, Subrizi A, Turunen
Modification of Müller Glial Cell
Fate and Proliferation with the Use
of Small Molecules

Marcus J. Hooper

Abstract 1 Introduction

In recent years, reprogramming Müller glia The death of postmitotic retinal neurons causes
by overexpressing Ascl1 and other transcrip- retinal degeneration, a primary cause of
tion factors has shown promise for the regen- irreversible blindness. Current treatment
eration of postmitotic retinal neurons, strategies for retinal degeneration do not restore
primarily bipolar cells, following injury. lost neurons or visual function, but delay
Müller glial proliferation and efficiency of degeneration. Regeneration of lost neurons is an
neuronal differentiation can be modified by ideal solution to this problem. Regeneration of
the use of small molecules in various sys- retinal neurons in mammals is a largely
tems. The molecules and pathways studied unexplored but growing field, due to promising
thus far share remarkable consistency with developments in recent years [1–3]. In zebrafish,
astrocytes. In this mini review, we provide an retinal glia, called Müller glia, are responsible for
overview on the modulation of Müller glial regeneration of all retinal cell types following
proliferation and cell fate using small mole- ablation by various types of injury. The response
cules in injury and reprogramming. We also to retinal damage induces regenerative programs,
compare these observations to what has been which include proneural and developmental
observed in astrocytes. transcription factors.
In the mammalian retina, however, the natural
Keywords response to damage does not lead to robust
regeneration, and critical factors such as Ascl1
Müller glia · Reprogramming · Drug · Small
are not induced. In order to replace lost neurons
molecule · Neuron · Glia · Neuronal repro-
effectively in disease, further understanding of
gramming · Pharmacological · Proliferation
Müller glial cell reprogramming is necessary.
With the eventual goal of replacing lost neurons
via glial cell conversion, this review is written to
summarize findings about pathways that influence
M. J. Hooper (*)
Department of Biological Structure, University of Müller glial cell fate and proliferation, in
Washington, Seattle, WA, USA reprogramming and injury. As Müller glia share
e-mail: mjhooper@uw.edu many characteristics with astrocytes, we also

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 473
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_69
474 M. J. Hooper

compare to studies in astrocyte reprogramming injury, such as that induced with NMDA, is often
to highlight pathways that are conserved for glial used as a means to induce retinal ganglion cell
cell conversion. death in the retina. Much has been done in exam-
ining the role of pharmacological agents and spe-
cific pathways in the chick retina after retinal
2 Drugs and Pathways Shown NMDA-induced injury. Interestingly, in this sys-
to Affect MG Cell Fate After tem, Fgf signaling is able to partially compensate
Injury for the signals induced in injury. Because a lot of
work has been done in this system, using consis-
Glial cells undergo several changes in response tent experimental approaches, we have summa-
to injury, and injury is often necessary to induce rized these findings in Table 1. Pharmacological
neurogenesis in regenerative systems. Neurotoxic treatments that have been shown to alter Müller

Table 1 Pharmacological treatments found to alter Müller glial fate or proliferation following injury
Pathway Compound(s) Target Effect References
BMP DMH1 ALK2 ↓ proliferation ⬖ [4]
FGF SU5402 FGFR VEGFR ↓ proliferation ⬖ [5]
PDGFR
Gelatinases and SB-3CT MMP2/9 ↑ proliferation ⬖◨ [6]
metalloproteinases
Gelatinases and MMP2i II MMP2 ↑ proliferation ⬖◨ [6]
metalloproteinases
GR Dexamethasone Mono−/dimeric ↓ proliferation ⬖◨ [7]
GR (agonist)
GR Compound A Monomeric GR ↓ proliferation ⬖◨ [7]
(agonist)
GR RU486 GR ↑ proliferation ⬖ [7]
Hedgehog SAG Smo (agonist) ↑ proliferation ⬖◨ (8)
Hedgehog GANT61 Gli1 + Gli2 DNA ↓ proliferation ⬖ [8]
binding
Hedgehog GANT58 Gli1 DNA binding ↓ proliferation ⬖ [8]
Hedgehog KAAD-cyclopamine Smo ↓ proliferation ⬖ [8]
MAPK U0126 MEK1/MEK2 ↓ proliferation ⬖ [5]
mTOR Rapamycin mTOR ↓ proliferation ⬖◨ [9]
NF-kB Sulfasalazine IκBα ↑ proliferation ⬖ [10]
NF-kB Prostaglandin J2 Nf-kB P50, ↑ proliferation ⬖ [10]
PPARγ ligand
NF-kB SC75741 Nf-kB DNA ↑ proliferation ⬖ [10]
binding
Notch DAPT y-secretase ↓ proliferation, ↑neuronal [11]
survival ⬖
RAR TTNBP RAR (agonist) ↑ proliferation ⬖ [12]
RAR BMS 483 RAR ↓ proliferation ⬖ [12]
STAT sc144 Gp130 ↓ proliferation ◨, ↑ [13]
neuronal differentiation ⬖
STAT Stattic Stat3 ↓ proliferation ◨ [13]
Tgfb SIS3 Smad3 ↑ proliferation ⬖ [4]
Tgfb SB431542 ALK4,5,7 ↑ proliferation ⬖ [4]
Wnt XAV939 Tankyrase 1/2 ↓ proliferation ⬖, ↑ Ascl1 [14]
expression
Wnt Wnt-c59 Porcupine ↓ proliferation ⬖ [14]
Wnt 1-Azakenpaullone, Gsk3B (Wnt ↑ proliferation ⬖ [14]
CHIR99021, BIO agonist)
The compound, relevant pathway, and specific target are shown. ⬖, with injury; ◨, with Fgf2 treatment
Modification of Müller Glial Cell Fate and Proliferation with the Use of Small Molecules 475

glial cell fate and proliferation following injury consistently influences proliferation is activation
include those that affect sonic hedgehog (SHH), of SHH signaling, the activation of which consis-
mTOR, Wnt, MapK, glucocorticoid receptors tently increases cell division in glia. Activation of
(GR), NF-kB, retinoic acid receptors (RAR), the SHH pathway in combination with Oct4
Notch, Tgfb, and gelatinases. There are a few increases cell proliferation and may improve
inferences that can be made from these studies. electrophysiological responses of reprogrammed
Firstly, Müller glial proliferation and response to neurons [16]. Consistent with observations in
injury can be prevented by targeting many path- Müller glia, activation of GR signaling using
ways. This may mean that these pathways are dexamethasone inhibits proliferation, while
highly integrated or dependent on one another. inhibiting GR signaling using RU-486 increases
Second, thus far, modulation of Müller glial phe- proliferation [17]. As in Müller glia, inhibition of
notype using small molecules in vivo depends on mTOR, using rapamycin in spinal cord astro-
an initial injury or induction of Fgf signaling. cytes, also reduces proliferation after injury [18].
Third, use of single small molecule treatments Gamma secretase (Notch) inhibition in astrocytes
does not seem to alter cell diversity of Müller using DAPT significantly increases the efficiency
glia-derived neurons with most candidates tested of reprogramming as it does in Müller glia [19].
thus far. Lastly, many treatments seem to alter To our knowledge, STAT3 inhibition hasn’t yet
cell proliferation (thymidine analog uptake), been reported to increase neurogenesis in astro-
while few treatments increase the efficiency of cyte reprogramming, but consistent with obser-
reprogramming Müller glia to neuronal fates, vations in Müller glia, a recent report shows that
with some notable exceptions. STAT3 inhibition inhibiting STAT3 increases neuronal gene expres-
has also been found to increase neurogenesis fol- sion, including Tuj1 in rat neural stem cells [20].
lowing injury in mice [2]. This observation makes STAT3 has also been shown to be important for
sense, in that STAT3 has been established as a glial scar formation in spinal cord injury [21].
critical transcription factor in glial cell differen-
tiation and activity. In mice, histone deacetylase
(HDAC) inhibition has also been shown to 3.1 Effects of Combination
increase Müller glial neuronal differentiation Treatments in Astrocytes
(Otx2+ bipolar cells) when coupled with Ascl1
overexpression [1]. As mentioned above, studies have been done in
astrocyte reprogramming that claim efficient
reprogramming of astrocytes to neurons using
3 Emphasis on Pathways combinations of small molecules. Often, these
Common with Astrocytes include modulators of the pathways mentioned
above, including Wnt, SHH, Notch, Tgfb, and
Many of the pathways that have effects on Müller HDACs. It is common for these studies to use
glial reprogramming have also been shown to several combinations of drugs and, after an effect
have similar effects in astrocytes. These are of is observed, remove individual compounds to see
particular interest, as they are repeatable across which are necessary for the observed effect. One
labs, and consistently deliver similar effects on particular study showed that a combination of
cell fate in different species and systems, in vitro nine molecules reprogrammed astrocytes to neu-
and in vivo. As mentioned above, Fgf signaling is rons. Removing any of the following reduced
important for the proliferative response to injury efficiency of reprogramming, in order of effect
in Müller glia, and the same is true in astrocytes. size: DAPT, CHIR99021, SB431542,
MEK and MapK are downstream of Fgf activa- LDN193189, SAG + purmorphamine, and val-
tion, and inhibition with U0126 has also been proic acid [19]. SHH activation with purmor-
shown to inhibit a proliferative phenotype in phamine is commonly included as a part of drug
astrocytes after injury [15]. Another pathway that cocktails in astrocyte to neuron reprogramming
476 M. J. Hooper

schemes [19]. The results of another study show positively or negatively. Fortunately, several
that transcription factor-mediated reprogram- pathways relevant in Müller glial reprogramming
ming of astrocytes can be enhanced by a combi- are also found to impact the same activity in
nation of small molecules, including astrocytes following injury or in reprogramming.
5-aza-2′-deoxycytidine, valproic acid, SB431542, Few studies, however, have shown an increase in
LDN193189, CT99021, and purmorphamine the efficiency of neuronal differentiation, and
[22]. It is important to note, however, that these fewer still have identified factors that increase the
candidate molecule screens are generally tar- diversity or maturation of reprogrammed neu-
geted toward modulating signaling pathways of rons. Despite these drawbacks, small molecules
known developmental importance, and a large-­ allow an efficient means to activate and inhibit
scale unbiased screen for reprogramming factors and study relevant pathways, and due to ease of
might yield additional candidates. delivery may be useful in examining effects of
combinations of treatments. Indeed the literature
does suggest that combinations of small mole-
3.2 Non-cell Autonomous Effects cules can lead to either more efficient or qualita-
In Vivo tively different outcomes when reprogramming
glia. These combination treatments often involve
In recent years, studies have shown an interaction complex treatment paradigms and are not repli-
between Müller glia and microglia after inflam- cated across organisms and tissue types, further
matory challenge or injury [23]. Studies in chick increasing complexity. Additionally, these types
and mouse retina have shown that microglial of studies are rarely done in a large-scale, sys-
ablation using small molecules, including tematic manner, and the readout is often cell
plx5622, influences Müller glia cell fate during counts using immunohistochemistry, which is
reprogramming in vivo [24]. Further, the effect of laborious and can be subject to biases. This sug-
Nf-kB inhibition on Müller glial reprogramming gests the need for a more efficient, quantitative,
and proliferation in chick is eliminated in the and reproducible system. Lastly, recent studies
absence of microglia, further supporting the rel- show that the interaction between microglia and
evance of non-cell autonomous pharmacological Müller glia is important for influencing Müller
intervention on the success of glial to neuron cell response to injury and cell fate. For this rea-
reprogramming in vivo [10]. Like Müller glia, son, future in vivo reprogramming strategies will
astrocyte proliferation and response to injury are likely benefit from considering their role.
also affected by non-cell autonomous effects,
caused by infiltrating monocytes [25].
References

4 Discussion 1. Jorstad NL, Wilken MS, Grimes WN, Wohl SG,


VandenBosch LS, Yoshimatsu T, et al. Stimulation of
functional neuronal regeneration from Müller glia in
Reprogramming glial cells to adopt neuronal adult mice. Nature. 2017 Aug 3;548(7665):103–7.
fates is an attractive method to regenerate postmi- 2. Jorstad NL, Wilken MS, Todd L, Finkbeiner C,
totic neurons in neurodegenerative disorders. Nakamura P, Radulovich N, et al. STAT signaling
modifies Ascl1 chromatin binding and limits neural
Müller glial reprogramming holds promise for regeneration from Müller glia in adult mouse retina.
retinal regeneration. There have been recent suc- Cell Rep. 2020;30(7):2195–208.e5.
cesses of transcription factor-mediated repro- 3. Todd L, Hooper MJ, Haugan AK, Finkbeiner C,
gramming of Müller glial cells. Small molecule Jorstad N, Radulovich N, et al. Efficient stimulation
of retinal regeneration from Müller glia in adult mice
treatments may further improve or complement using combinations of proneural bHLH transcription
reprogramming strategies in the long term. Many factors. Cell Rep. 2021 Oct 19;37(3):109857.
studies find that single small molecule treatments 4. Todd L, Palazzo I, Squires N, Mendonca N, Fischer
are capable of influencing glial cell proliferation AJ. BMP- and TGFβ-signaling regulate the formation
Modification of Müller Glial Cell Fate and Proliferation with the Use of Small Molecules 477

of Müller glia-derived progenitor cells in the avian 15. Neary JT, Kang Y, Shi Y-F. Cell cycle regulation
retina. Glia. 2017;65(10):1640–55. of astrocytes by extracellular nucleotides and
5. Fischer AJ, Scott MA, Tuten W. Mitogen-activated fibroblast growth factor-2. Purinergic Signal. 2005
protein kinase-signaling stimulates Müller glia to Dec;1(4):329–36.
­proliferate in acutely damaged chicken retina. Glia. 16. Yang H, Liu C, Fan H, Chen B, Huang D, Zhang
2009 Jan 15;57(2):166–81. L, et al. Sonic hedgehog effectively improves Oct4-­
6. Campbell WA 4th, Deshmukh A, Blum S, Todd L, mediated reprogramming of astrocytes into neural
Mendonca N, Weist J, et al. Matrix-metalloproteinase stem cells. Mol Ther. 2019 Aug 7;27(8):1467–82.
expression and gelatinase activity in the avian retina 17. Crossin KL, Tai MH, Krushel LA, Mauro VP, Edelman
and their influence on Müller glia proliferation. Exp GM. Glucocorticoid receptor pathways are involved
Neurol. 2019 Oct;320:112984. in the inhibition of astrocyte proliferation. Proc Natl
7. Gallina D, Zelinka C, Fischer AJ. Glucocorticoid Acad Sci U S A. 1997 Mar 18;94(6):2687–92.
receptors in the retina, Müller glia and the formation 18. Goldshmit Y, Kanner S, Zacs M, Frisca F, Pinto
of Müller glia-derived progenitors. Development. AR, Currie PD, et al. Rapamycin increases neuronal
2014 Sep;141(17):3340–51. survival, reduces inflammation and astrocyte
8. Todd L, Fischer AJ. Hedgehog signaling stimulates proliferation after spinal cord injury. Mol Cell
the formation of proliferating Müller glia-derived Neurosci. 2015 Sep;68:82–91.
progenitor cells in the chick retina. Development. 19. Zhang L, Yin J-C, Yeh H, Ma N-X, Lee G, Chen XA,
2015 Aug 1;142(15):2610–22. et al. Small molecules efficiently reprogram human
9. Zelinka CP, Volkov L, Goodman ZA, Todd L, Palazzo Astroglial cells into functional neurons. Cell Stem
I, Bishop WA, et al. mTor signaling is required for Cell. 2015 Dec 3;17(6):735–47.
the formation of proliferating Müller glia-derived 20. Li T, Zhao X, Duan J, Cui S, Zhu K, Wan Y, et al.
progenitor cells in the chick retina. Development. Targeted inhibition of STAT3 in neural stem cells
2016 Jun 1;143(11):1859–73. promotes neuronal differentiation and functional
10. Palazzo I, Deistler K, Hoang TV, Blackshaw S, recovery in rats with spinal cord injury. Exp Ther
Fischer AJ. NF-κB signaling regulates the formation Med. 2021 Jul;22(1):711.
of proliferating Müller glia-derived progenitor cells 21. Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen
in the avian retina. Development [Internet]. 2020 TK, et al. STAT3 is a critical regulator of astrogliosis
147(10). Available from: https://doi.org/10.1242/ and scar formation after spinal cord injury. J Neurosci.
dev.183418 2008 Jul 9;28(28):7231–43.
11. Ghai K, Zelinka C, Fischer AJ. Notch signaling 22. di Val Cervo PR, Romanov RA, Spigolon G, Masini
influences neuroprotective and proliferative D, Martin-Montanez E, Toledo EM, et al. Induction of
properties of mature Müller glia. J Neurosci. 2010 functional dopamine neurons from human astrocytes
Feb 24;30(8):3101–12. in vitro and mouse astrocytes in a Parkinson’s disease
12. Todd L, Suarez L, Quinn C, Fischer AJ. Retinoic acid-­ model. Nat Biotechnol. 2017;35(5):444–52.
signaling regulates the proliferative and neurogenic 23. Wang M, Ma W, Zhao L, Fariss RN, Wong
capacity of Müller glia-derived progenitor cells in the WT. Adaptive Müller cell responses to microglial
avian retina. Stem Cells. 2018 Mar;36(3):392–405. activation mediate neuroprotection and coordinate
13. Todd L, Squires N, Suarez L, Fischer AJ. Jak/Stat inflammation in the retina. J Neuroinflammation.
signaling regulates the proliferation and neurogenic 2011 Dec 7;8:173.
potential of Müller glia-derived progenitor cells in the 24. Todd L, Finkbeiner C, Wong CK, Hooper MJ,
avian retina. Sci Rep. 2016 Oct 19;6:35703. Reh TA. Microglia suppress Ascl1-induced
14. Gallina D, Palazzo I, Steffenson L, Todd L, Fischer retinal regeneration in mice. Cell Rep. 2020 Dec
AJ. Wnt/β-catenin-signaling and the formation of 15;33(11):108507.
Müller glia-derived progenitors in the chick retina 25. Frik J, Merl-Pham J, Plesnila N, Mattugini N, Kjell J,
[Internet]. Vol. 76, Developmental Neurobiology. Kraska J, et al. Cross-talk between monocyte invasion
2016. pp. 983–1002. Available from: https://doi. and astrocyte proliferation regulates scarring in brain
org/10.1002/dneu.22370 injury. EMBO Rep [Internet]. 2018 19(5). Available
from: https://doi.org/10.15252/embr.201745294
A Potential Neuroprotective Role
for Pyruvate Kinase 2 in Retinal
Degeneration

Jiaming Zhou, Michel Rasmussen,


and Per Ekström

Abstract Keywords

Retinitis pigmentosa (RP) is an inherited dis- PKM2 · Retinal degeneration · Cell death ·
order that results in vision impairment that TUNEL · Organotypic retinal explant culture
specific therapeutic strategies are not avail-
able. However, it is widely regarded that the
cGMP system, including cGMP and its inter- 1 Introduction
actor cGMP-dependent protein kinase (PKG),
acts as a crucial effector during retinal degen- Retinitis pigmentosa (RP) is one of the most
eration. We have previously identified a list of common forms of inherited retinal degeneration,
cGMP-PKG-dependent genes in the context which refers to genetic disorders in retinal photo-
of RP, and in this study, we further validated receptors, leading to severe vision problems. The
one of the targets, namely, pyruvate kinase 2 prevalence of this retinal disorder reaches 1 in
(PKM2), and investigated the potential role of 3000–4000 [5], and there is currently in principle
PKM2 for the photoreceptors’ well-being dur- no effective treatment available, likely because of
ing RP. With the aid of organotypic retinal the extensively heterogeneous mutations of
explant cultures, we pharmacologically disease-­leading genes [4]. Work in our laboratory
manipulated the PKM2 activities in different has previously shown that a neurodegenerative
RP mouse models via the addition of TEPP-46 effect of elevated cGMP at least in part works via
(a PKM2 activator) and found that activation increasing the activity of its dependent protein
of PKM2 alleviates the progress of photore- kinase G (cGMP-dependent protein kinase;
ceptor death in the rd10 mouse model. This PKG) leading to over-phosphorylation within
observation provides supportive evidence that photoreceptors [7].
PKM2 may serve as a novel potential molecu- The subtype of pyruvate kinase (PKM2) has
lar target in RP. been shown to express within photoreceptors
[10]. Our previous transcriptome research [13],
where we modulated the cGMP-PKG system in
J. Zhou (*) · M. Rasmussen · P. Ekström
Ophthalmology, Department of Clinical Sciences,
retinal explants from the healthy wild-type (wt)
Lund University, Lund, Sweden model, showed a lower expression of PKM2 after
e-mail: jiaming.zhou@med.lu.se; PKG activation in wt retinas. This pointed to the
michel.rasmussen@med.lu.se; possibility that PKM2 may be inhibited by the
per.ekstrom@med.lu.se

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 479
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_70
480 J. Zhou et al.

high cGMP-PKG within diseased photorecep- 2.2 Organotypic Retinal Explant


tors. To this end, we know that PKG inhibition Culture
via a liposome-formulated cGMP analogue can
protect degenerating photoreceptors and preserve This followed our previous protocol as in Vighi
their function after systemic administration [12]. et al. [12]. The culturing paradigm used is termed
We speculated that since PKG appears to decrease P9 + 2 + 8 in vivo and means that the animals
PKM2 in the degenerating photoreceptors, direct were sacrificed at postnatal day 9 (P9), after
activation of PKM2 during the degeneration may which they were cultured for 2 days without
have neuroprotective effects. manipulation followed by 8 days with PKM2
To acquire a deeper understanding of this, we activation (TEPP-46 (HY-18657),
here utilized a manipulation of the PKM2 activity MedChemExpress), with medium changes every
in the retina via a pharmacological method. To do second day during the whole culturing period. In
so we added 6-((3-aminophenyl)methyl)- parallel, untreated counterparts served as con-
4-methyl-2-methylsulfinylthieno [3, trols. The culturing paradigm thus means that the
4]-pyrrolo[1,3-d]pyridazin-5-one (TEPP-46, a endpoint resembles P19.
PKM2 activator) [6] during organotypic retinal
explant culturing with tissue from either rd2 [11]
or rd10 strains [2], two RP models where photo- 2.3 Cryosectioning
receptor cGMP is also high [1]. According to our
hypothesis, we expected to have a lower expres- At the culture endpoint P19, the samples were
sion of PKM2 in these disease models and that collected for cryosectioning. All explants were
activation of this enzyme may alleviate the prog- treated with 4% formaldehyde for 2 h, washed
ress of cell death. 4 × 15 min in phosphate-buffered saline (PBS),
Our investigations show that rd10 retinas ben- and cryoprotected in PBS + 10% sucrose over-
efitted with an apparent lower amount of photore- night at +4 °C and then with PBS + 25% sucrose
ceptor death from PKM2 activation, while no for 2 h. After embedding, 12-μm-thick retinal
difference could be observed in the rd2 strain cross sections were cut and collected from an
with the same treatment. Given the slow progress HM560 cryotome (Microm, Walldorf, Germany).
of photoreceptor loss in the rd2 strain, a positive
effect of long-term PKM2 activation in rd2 can-
not be excluded, though. 2.4 TUNEL Assay

Fluorescent terminal deoxynucleotidyl transfer-


2 Materials and Methods ase dUTP nick end labeling (TUNEL) assay
(11687495910, Roche Diagnostics, Mannheim,
2.1 Experiment with Animals Germany) was performed as by manufacturer’s
instructions and used on cryosections from the
The mouse strains rd2 (C3H) and rd10 different conditions to evaluate cell death. The
(C57BL/6) were used in our study. Animals number of TUNEL-positive cells in the
were kept under standard white cyclic lighting, photoreceptor-­bearing outer nuclear layer (ONL)
with ad libitum access to food and water, and was calculated as shown previously [7]. For each
used irrespective of sex. All procedures had situation, three to four sections from five to six
been evaluated by the local animal care and eth- animals were analyzed to generate an average
ics committees and performed in accordance value. The same sections were used for the analy-
with permit 2124-20. All efforts were made to sis of ONL thickness, which was done via mea-
keep the number of animals used and their suf- surements using the ZEN2 software (blue edition)
fering to a minimum. tool for image analysis.
A Potential Neuroprotective Role for Pyruvate Kinase 2 in Retinal Degeneration 481

2.5 Microscopy and Image tective potential of PKM2 during RP progression


Processing in this model. For the rd2 strain, there were
changes in the same directions on the ONL thick-
A Zeiss Imager Z1 Apotome Microscope (Zeiss, ness by the PKM2 activation, but these were
Oberkochen, Germany), with a Zeiss Axiocam smaller and did not reach statistical significance.
digital camera, was used for microscopy observa- The rd2 retina has a peak of cell death at around
tions. Image generation and contrast enhance- P18, but cell death is seen also several days after
ment were performed identically for all images that [1]. Given that our explant culturing protocol
via the ZEN2 software (blue edition). starts at P9 and ends at P19, we might thus have
missed a fully developed protective effect that
may require longer treatment to be clearly
2.6 Statistics detected. The higher percentage of dying photo-
receptors in the rd10 retina at around P18 [1]
All t-tests in this report were performed via could then explain why the effect came out
GraphPad Prism 9, and a p-value lower than 0.05 clearer in that model. Alternatively, since the dif-
was regarded as significant. ferent gene mutations of rd10 and rd2 are likely
to result in differences in for instance exact path-
way activation patterns, PKM2 could be of less
3 Results protective importance in rd2, either throughout
the degeneration or in the period studied.
In this study, we manipulated the PKM2 activi- Our results go very well together with those of
ties within the photoreceptors in two RP models Rajala et al. [10], who observed an increased
by adding TEPP-46 during the organotypic reti- number of degenerating cells within the ONL in
nal explant cultures. For the rd10 model, we retinas from their mouse model with genetically
found that the number of TUNEL-positive cells deleted PKM2 expression, indicating its impor-
decreased compared to the untreated counterpart tance for photoreceptor viability. Interestingly,
(Fig. 1c) and that this was complemented with an they also observed that PKM2 deletion lowered
increase of ONL thickness (Fig. 1d–f). By con- the expression of glucose transporter-1 (Glut1),
trast, we could not observe any significant differ- which has the important function to deliver glu-
ences in either photoreceptor death or ONL cose to photoreceptors [9]. The role for PKM2
thickness in the explants with PKM2 activation may thus be to stabilize photoreceptors via the
from the rd2 model compared to their untreated maintenance of energy supply, a situation that
counterparts (Fig. 1a–b, e–f). However, we note appears vital as photoreceptors are regarded as
that the activator treatment of the rd2 explants one of the most metabolically active cells within
gave thickness values with a mean that was the body [3]. In fact, the general outcome of our
higher than for corresponding untreated controls previous transcriptome analysis [13] was that an
(activator 54 ± 3; control 42 ± 1, unit: μm). The overactive cGMP system could be an important
rd2 outcome thus at least partially pointed toward negative regulator of retinal metabolic events.
a similar effect by the activator treatment as in the Since increased cGMP levels can be linked to
rd10 case, but without attaining statistical many types of RP [8], it is thus possible that dis-
significance. turbed actions of PKM2, or other enzymes related
to energy metabolism, lie at the heart of the RP
pathology.
4 Discussion Many questions with respect to PKM2 and
photoreceptor degeneration remain to be
The lower counts of degenerating photoreceptors answered, like if the retinal substrates for this
in rd10 after PKM2 activation were accompanied enzyme are the same as in other tissues, and if
by a thicker ONL, which indicated a neuropro- this changes in disease. Moreover, it will be of
482 J. Zhou et al.

Fig. 1 Activation of PKM2 showed a decreased number marizing the TUNEL-positive ONL cell counts and ONL
of dying photoreceptors in rd10 explants as well as thickness of the different experimental groups, respec-
resulted in a thicker ONL, while the same treatment did tively. Two t-tests, namely, the comparison between rd2
not change these parameters in rd2 explants. TUNEL-­ with PKM2 activator vs rd2 untreated and rd10 with
positive cells are shown in green, and DAPI (blue) was PKM2 activator vs rd10 untreated was performed in e and
used as nuclear counterstain. (a–d) rd2 untreated control, f. ONL outer nuclear layer, INL inner nuclear layer,
rd2 treated with PKM2 activator, rd10 untreated control, GCL ganglion cell layer. N = 4–5 biological replicates
rd10 treated with PKM2 activator. (e–f) Bar chart sum-
A Potential Neuroprotective Role for Pyruvate Kinase 2 in Retinal Degeneration 483

interest to see if treatment of wt explants with a 3. Country M. Retinal metabolism: a comparative look
at energetics in the retina. Brain Res. 2017;1672:50–7.
pharmacological PKM2 inhibitor will mimic the 4. Daiger S, Sullivan L, Bowne S. Genes and muta-
outcome of genetic deletion of the enzyme, as in tions causing retinitis pigmentosa. Clin Genet.
Rajala et al. [10], i.e., and lead to photoreceptor 2013;84:132–41.
death. If so, this would not only strengthen the 5. Haim M. The epidemiology of retinitis pigmentosa
in Denmark. Acta Ophthalmol Scand. 2002;80:1–34.
connection between PKM2 and retinal degenera- 6. Jiang J, Boxer M, Vander Heiden M, et al. Evaluation
tion but also by inference again point to PKM2 as of thieno[3,2-b]pyrrole[3,2-d]pyridazinones as
a molecular target for RP therapy. activators of the tumor cell specific M2 isoform
All in all, the outcomes from our study shed of pyruvate kinase. Bioorganic Med Chem Lett.
2010;20:3387–93.
light on a potential novel molecular target that 7. Paquet-Durand F, Hauck S, van Veen T, et al.
may exert a direct effect on RP. Modulation of PKG activity causes photoreceptor cell death in
PKM2, therefore, appears suitable for further two retinitis pigmentosa models. J Neurochem.
exploration in other RP models in the future to 2009;108:796–810.
8. Power M, Das S, Schütze K, et al. Cellular mecha-
gain deeper insights. nisms of hereditary photoreceptor degeneration –
focus on cGMP. Prog Retin Eye Res. 2020;74:100772.
Acknowledgments This study was funded by the 9. Pragallapati S, Manyam R. Glucose transporter
European Union’s Horizon 2020 research and innovation 1 in health and disease. J Oral Maxillofac Pathol.
program under the Marie Sklodowska-Curie grant agree- 2019;23:443.
ment No 765441 (transMed; H2020-­ MSCA-­ 765441), 10. Rajala A, Wang Y, Brush R. Pyruvate kinase M2 regu-
Stiftelsen för Synskadade i f.d. Malmöhus län, lates photoreceptor structure, function, and viability.
Kronprinsessan Margaretas Arbetsnämnd för synskadade, Cell Death Dis. 2018;9:240.
and Ögonfonden. 11. Travis G, Brennan M, Danielson P, et al. Identification
of a photoreceptor-specific mRNA encoded by the
gene responsible for retinal degeneration slow (rds).
Nature. 1989;338:70–3.
References 12. Vighi E, Trifunović D, Veiga-Crespo P. Combination
of cGMP analogue and drug delivery system provides
1. Arango-Gonzalez B, Trifunović D, Sahaboglu A, functional protection in hereditary retinal degenera-
et al. Identification of a common non-apoptotic cell tion. Proc Natl Acad Sci U S A. 2018;115:2997–3006.
death mechanism in hereditary retinal degeneration. 13. Zhou J, Rasmussen M, Ekström P. cGMP-PKG
PLoS One. 2014;9:112142. dependent transcriptome in normal and degenerating
2. Chang B, Hawes N, Hurd R, et al. Retinal degenera- retinas: novel insights into the retinitis pigmentosa
tion mutants in the mouse. Vis Res. 2002;42:517–25. pathology. Exp Eye Res. 2021;212:108752.
Part XI
Photoreceptors
Critical Role of VEGF as a Direct
Regulator of Photoreceptor
Function

Jianyan Hu, Meili Zhu, Dai Li, Qiang Wu,


and Yun-Zheng Le

Abstract toreceptor function with electroretinography


(ERG) in mice. In our case, rVEGF caused a
Vascular endothelial growth factor (VEGF or significant reduction of scotopic ERG a-wave
VEGF-A), a major pathogenic factor for dia- and b-wave amplitudes and photopic ERG
betic and hypoxic blood–retina barrier (BRB) b-wave amplitudes in a dose-dependent
diseases, has been shown to act as a direct manner in dark-adapted wild-type (WT) mice,
functional regulator for neurons in the periph- shortly after the intravitreal delivery of rVEGF
eral and central nerve systems. To determine if in dark. However, the effect of rVEGF on
VEGF plays a direct role in regulating retinal photoreceptor function was nullified in adult
neuronal function, we established specific Akita diabetic mice. Our data strongly suggest
experimental procedures and examined the that VEGF is a direct regulator of photoreceptor
effect of recombinant VEGF (rVEGF) on pho- function and VEGF upregulation contributes

J. Hu Q. Wu
Department of Medicine/Endocrinology, University Department of Ophthalmology, Shanghai Jiao Tong
of Oklahoma Health Sciences Center, University Affiliated Sixth People’s Hospital,
Oklahoma City, OK, USA Shanghai, China
Department of Ophthalmology, Shanghai Jiao Tong Shanghai Key Laboratory of Diabetes Mellitus,
University Affiliated Sixth People’s Hospital, Shanghai, China
Shanghai, China e-mail: qiang.wu@shsmu.edu.cn
M. Zhu Y.-Z. Le (*)
Department of Medicine/Endocrinology, University Department of Medicine/Endocrinology, University
of Oklahoma Health Sciences Center, of Oklahoma Health Sciences Center,
Oklahoma City, OK, USA Oklahoma City, OK, USA
D. Li Department of Cell Biology, University of Oklahoma
Department of Medicine/Endocrinology, University Health Sciences Center, Oklahoma City, OK, USA
of Oklahoma Health Sciences Center,
Department of Ophthalmology, University of
Oklahoma City, OK, USA
Oklahoma Health Sciences Center,
School of Optometry, Hubei University of Science Oklahoma City, OK, USA
and Technology, Xianning, China
Harold Hamm Diabetes Center, University of
Oklahoma Health Sciences Center,
Oklahoma City, OK, USA
e-mail: Yun-Le@ouhsc.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 487
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_71
488 J. Hu et al.

significantly to the diabetes-induced reduction 2 Critical Factors


of photoreceptor function. In this chapter, we in Experimental Design
will discuss the relevant background, key
experimental procedures and results, and clin- Although there were many investigations on the
ical significance of our work. effect of rVEGF on retinal function with ERG,
the experimental limitations in these studies
Keywords made it impossible to obtain information on the
role of VEGF as a direct functional regulator for
VEGF · Photoreceptor function · ERG · retinal neurons. This is due to the fact that VEGF
Diabetes · Hypoxia is a secreted multifunctional protein that could
play many roles once it is introduced into the
retina. Any experiment that does not measure the
1 Introduction immediate effect of VEGF is unlikely to reveal
the direct role of VEGF in modulating photore-
A major accomplishment in the treatment of ceptor function. To address this issue, we made
blood–retina barrier (BRB) diseases in recent modifications to our experimental procedures
decades is the identification of vascular endothe- with the following special arrangement: we dark-­
lial growth factor (VEGF) as a major pathogenic adapted the mice overnight, intravitreally injected
factor for BRB breakdown in diabetic retinopa- rVEGF under long-wave illumination, and per-
thy (DR), retinopathy of prematurity (ROP), and formed ERG 15–20 min post-intravitreal injec-
age-related macular degeneration (AMD) [1–3, tion. The waiting period is necessary for rVEGF
17, 18]. However, VEGF is known to play a role to migrate to photoreceptor regions. Using a
as a neuronal functional regulator in the brain method established by us previously [21], we
and peripheral neurons, through major VEGF detected migration of bright 10-kDa fluorescein
receptors and co-receptors. It has been shown isothiocyanate (FITC)–dextran, which had a
that VEGF was capable of reducing excessive comparable MW as that of VEGF, to the photore-
excitation of hypoglossal motor neurons by ceptor region 20 min after intravitreal injection
downregulating stimulus-evoked depolarization, (Fig. 1a). This suggests the feasibility of this time
a critical mechanism to prevent amyotrophic lat- frame. These experimental settings allow us to
eral sclerosis [13]. VEGF has also been shown to measure the direct effect of VEGF on photore-
reduce synaptic responses significantly in rat ceptor function, which is conceptually identical
hippocampal neurons through VEGF receptor-2 to the exposure of rVEGF briefly to brain slices
(VEGFR2) signaling [9, 12]. VEGFR2 also that in the initial identification of VEGF as a
plays a role in activating P2X2/3 in primary sen- direct functional regulator of hippocampal neu-
sory neurons to mediate neuropathic pain trans- rons [12].
mission [10]. VEGF co-receptor neuropilin 1
(NRP1) is involved in downregulating synaptic
transmission in hippocampal synapses [5]. As 3 Effect of VEGF
both VEGFR1 and VEGFR2 are present in the on Photoreceptor Function
retina, including photoreceptors [7, 8, 15, 16], it
is possible that VEGF receptors and co-receptors To determine the direct effect of VEGF on
could mediate photoreceptor function in the ret- photoreceptor function, we intravitreally injected
ina. To explore the direct role of VEGF in modu- human rVEGF (0.3 μg/eye) to 1.5-month-old
lating retinal neuronal function, we established overnight dark-adapted C57BL6 background
specific experimental procedures and used elec- mice under long-wavelength illumination.
troretinography (ERG) approach to measure Twenty minutes after intravitreal rVEGF injec-
photoreceptor function shortly after intravitreal tion, scotopic ERG was recorded with flash
recombinant VEGF (rVEGF) delivery. intensities from 0.002 to 400 cd·s/m2. Figure 1b
Critical Role of VEGF as a Direct Regulator of Photoreceptor Function 489

Fig. 1 Measurement of direct effect of intravitreally real injection. (b) Representative scotopic ERG tracers
injected rVEGF-induced reduction of photoreceptor func- showing VEGF-induced reduction of ERG amplitudes in
tion with ERG. (a) Frozen retinal section showing the overnight dark-adapted WT mice, 20 min after intravitreal
migration of 10 kDa FITC-dextran (bright), which has a injection of human rVEGF (0.3 μg/eye) in dark. Flash
comparable MW to rVEGF, in mice 20 min after intravit- intensity: 200 cd·s/m2

demonstrated representative scotopic ERG trac- photopic ERG b-wave amplitudes, was nullified
ers of rVEGF downregulated both scotopic ERG in 5-month-old Akita spontaneous diabetic mice
a-wave and b-wave amplitudes significantly. (Figures 3 and 4 in [8]). To ascertain if VEGF
However, rVEGF did not affect the time for was upregulated in 5-month-old Akita spontane-
trough or peak in the ERG. The effect of rVEGF ous diabetes mice, we localized VEGF with
on scotopic ERG appeared dose-dependent immunohistochemistry and found that VEGF
(Figure 1 in [8]). Likewise, rVEGF also had an levels were elevated in the area of photoreceptor
dose-dependent effect on depressing cone photo- inner segments, Müller glia, outer nuclear layer
receptor function, as shown in photopic ERG (ONL), outer plexiform layer (OPL), inner
(Figure 2 in [8]). As our experiments were nuclear layer (INL), and ganglion cell layer
designed to measure the direct effect of rVEGF (Figure 5 in [8]), where VEGFR2 was also pres-
on photoreceptor function, our data strongly sug- ent (Figure 6 in [8]) [7]. In short, our results
gest that VEGF is a direct functional regulator of strongly suggest that VEGF is a contributing fac-
photoreceptors. tor for diabetes-induced reduction of photorecep-
tor function. Finally, hypoxia-induced VEGF
upregulation may also play a similar role in
4 Contribution of VEGF depressing photoreceptor function in AMD and
on Reducing Photoreceptor other hypoxic retinal diseases. We are actively
Function in Diabetes investigating this possibility using an animal
and Hypoxia chamber with regulatable oxygen content.

Reduction of photoreceptor function is a


characteristic of DR (Figures 3 and 4 in [8]). We 5 Potential Mechanism
therefore hypothesized that VEGF upregulation of VEGF-Mediated Direct
is a contributing factor for the reduction of Effect on Photoreceptor
photoreceptor function in DR. To test this Function
hypothesis, we used Akita spontaneous diabetic
mice, an acceptable model for experimental DR Our work on the direct role of VEGF on
[4], and examined the direct effect of intravitreally photoreceptor function reveals a major knowledge
injected rVEGF on photoreceptor function with gap in retinal neurobiology for the first time. At
ERG. Compared with that in age-matched WT present, the mechanism of VEGF-induced
controls, the effect of rVEGF on scotopic ERG reduction of visual function remains largely
a-wave and b-wave amplitudes, as well as on unknown. As there was an elevated presence of
490 J. Hu et al.

VEGF in ­photoreceptor inner segments in aging development of new therapeutics for these
diabetic mice (Figure 5 in [8]), it is likely that leading causes of blindness.
VEGF may regulate photoreceptor function under
pathological stresses. Based on the observation of Acknowledgments Our work was supported by NIH
ciliary neurotrophic factor- or diabetes-induced grants R01EY26970, P30EY021725, and P30GM122744;
National Natural Science Foundation of China grants
depression of visual function [6, 11, 14, 19, 20], 81070738 and 81300775; grants from Presbyterian Health
VEGF might modulate photoreceptor function by Foundation and Oklahoma Center for Advancement of
altering phototransduction or visual cycle Science and Technology; and endowments from Mr.
machinery. VEGF elevation in DR and hypoxia Harold Hamm and Choctaw Nation of Oklahoma.
could serve as a compensatory or protective
mechanism for photoreceptors under these Conflicts of Interest Authors declare no conflicts of
interest.
stresses. VEGF has been shown to regulate synap-
tic transmission through VEGF receptor-mediated
signaling in the rat hippocampus [9]. A strong
presence of VEGFR2 in photoreceptor synaptic References
terminal area suggests a possibility for VEGFR2- 1. Aiello LP, Avery RL, Arrigg PG, Keyt BA, Jampel HD,
mediated alteration of photoreceptor synaptic Shah ST, Pasquale LR, Thieme H, Iwamoto MA, Park
transmission (Figure 6 in [8]). More work with JE, et al. Vascular endothelial growth factor in ocular
photoreceptor-­specific VEGFR2 knockout mice fluid of patients with diabetic retinopathy and other
retinal disorders. N Engl J Med. 1994;331:1480–7.
will allow us to address this issue and to reveal the 2. Alon T, Hemo I, Itin A, Pe’er J, Stone J, Keshet
potential mechanisms of VEGF-regulated neuro- E. Vascular endothelial growth factor acts as a sur-
nal function. vival factor for newly formed retinal vessels and has
implications for retinopathy of prematurity. Nat Med.
1995;1:1024–8.
3. Bai Y, Ma JX, Guo J, Wang J, Zhu M, Chen Y, Le
6 Clinical Relevance YZ. Muller cell-derived VEGF is a significant
contributor to retinal neovascularization. J Pathol.
Comparison of the effect of rVEGF on 2009;219:446–54.
4. Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans
photoreceptor function between Akita diabetic RS, Krady JK, Levison SW, Gardner TW, Bronson
mice and WT controls suggests that VEGF is a SK. The Ins2Akita mouse as a model of early retinal
major contributing factor for diabetes-induced complications in diabetes. Invest Ophthalmol Vis Sci.
reduction of photoreceptor function, which not 2005;46:2210–8.
5. Bouzioukh F, Daoudal G, Falk J, Debanne D, Rougon
only reveals a major knowledge gap in DR field G, Castellani V. Semaphorin3A regulates synaptic
for the first time but also provides the mechanistic function of differentiated hippocampal neurons. Eur
basis for investigating diabetes-altered visual J Neurosci. 2006;23:2247–54.
function. Our work also produces the critical 6. Chen J, Shao Y, Sasore T, Moiseyev G, Zhou K,
Ma X, Du Y, Ma JX. Interphotoreceptor retinol-
knowledge background related to the therapeutic binding protein ameliorates diabetes-induced
effect(s) of anti-VEGF drugs in DR, AMD, and retinal dysfunction and neurodegeneration through
other hypoxic retinal disorders. In addition, our rhodopsin. Diabetes. 2021;70:788–99.
finding could serve as the scientific basis for the 7. Fu S, Dong S, Zhu M, Sherry DM, Wang C, You Z,
Haigh JJ, Le YZ. Müller glia are a major cellular
development of improved patient care procedures source of survival signals for retinal neurons in
during anti-­VEGF therapies. Since VEGF family diabetes. Diabetes. 2015;64:3554–63.
cytokines and their soluble receptors and 8. Hu J, Zhu M, Li D, Wu Q, Le YZ. VEGF as a direct
co-receptors are potential therapeutic agents/ functional regulator of photoreceptors and contributing
factor to diabetes-induced alteration of photoreceptor
targets for BRB breakdown in DR, wet AMD, function. Biomolecules. 2021;11:988–97.
retinopathy of prematurity, and other hypoxic 9. Kim BW, Choi M, Kim YS, Park H, Lee HR, Yun
vascular diseases, further research into the CO, Kim EJ, Choi JS, Kim S, Rhim H, Kaang BK,
mechanism and direct role of VEGF family Son H. Vascular endothelial growth factor (VEGF)
signaling regulates hippocampal neurons by elevation
cytokines and receptors in the modulation of of intracellular calcium and activation of calcium/
retinal neuronal function will be necessary to the
Critical Role of VEGF as a Direct Regulator of Photoreceptor Function 491

calmodulin protein kinase II and mammalian target of 15. Saint-Geniez M, Maharaj AS, Walshe TE, Tucker
rapamycin. Cell Signal. 2008;20:714–25. BA, Sekiyama E, Kurihara T, Darland DC, Young
10. Lin J, Li G, Den X, Xu C, Liu S, Gao Y, Liu H, Zhang MJ, D'Amore PA. Endogenous VEGF is required for
J, Li X, Liang S. VEGF and its receptor-2 involved in visual function: evidence for a survival role on muller
neuropathic pain transmission mediated by P2X(2)(/) cells and photoreceptors. PLoS One. 2008;3:e3554.
(3) receptor of primary sensory neurons. Brain Res 16. Shih SC, Ju M, Liu N, Smith LE. Selective stimulation
Bull. 2010;83:284–91. of VEGFR-1 prevents oxygen-induced retinal
11. Malechka VV, Moiseyev G, Takahashi Y, Shin Y, Ma vascular degeneration in retinopathy of prematurity. J
JX. Impaired rhodopsin generation in the rat model of Clin Invest. 2003;112:50–7.
diabetic retinopathy. Am J Pathol. 2017;187:2222–31. 17. Wang J, Xu X, Elliott MH, Zhu M, Le YZ. Muller
12. McCloskey DP, Croll SD, Scharfman HE. Depression cell-derived VEGF is essential for diabetes-induced
of synaptic transmission by vascular endothelial retinal inflammation and vascular leakage. Diabetes.
growth factor in adult rat hippocampus and evidence 2010;59:2297–305.
for increased efficacy after chronic seizures. J 18. Wang JJ, Zhu M, Le YZ. Functions of Muller cell-­
Neurosci. 2005;25:8889–97. derived vascular endothelial growth factor in diabetic
13. McCloskey DP, Hintz TM, Scharfman retinopathy. World J Diabetes. 2015;6:726–33.
HE. Modulation of vascular endothelial growth 19. Wen R, Tao W, Li Y, Sieving PA. CNTF and retina.
factor (VEGF) expression in motor neurons and Prog Retin Eye Res. 2012;31:136–51.
its electrophysiological effects. Brain Res Bull. 20. Wen R, Song Y, Kjellstrom S, Tanikawa A, Liu Y, Li Y,
2008;76:36–44. Zhao L, Bush RA, Laties AM, Sieving PA. Regulation
14. McGill TJ, Prusky GT, Douglas RM, Yasumura D, of rod phototransduction machinery by ciliary
Matthes MT, Nune G, Donohue-Rolfe K, Yang H, neurotrophic factor. J Neurosci. 2006;26:13523–30.
Niculescu D, Hauswirth WW, Girman SV, Lund RD, 21. Xu HZ, Le YZ. Significance of outer blood-retina
Duncan JL, LaVail MM. Intraocular CNTF reduces barrier breakdown in diabetes and ischemia. Invest
vision in normal rats in a dose-dependent manner. Ophthalmol Vis Sci. 2011;52:2160–4.
Invest Ophthalmol Vis Sci. 2007;48:5756–66.
Lysine Ubiquitylation Drives
Rhodopsin Protein Turnover

Allen P. F. Chen, Leon Chea, Eun-Jin Lee,


and Jonathan H. Lin

Abstract domain of rhodopsin. We previously found that


P23H rhodopsin was misfolded, ubiquitinyl-
Rhodopsin is a G-protein-coupled receptor that ated, and rapidly degraded. Here, we investi-
is specifically and abundantly expressed in rod gated the role of lysine residues on P23H
photoreceptors. Over 150 rhodopsin mutations rhodopsin ubiquitinylation and turnover. We
cause autosomal dominant retinitis pigmentosa transfected HEK293 cells with a P23H human
(adRP). The most common mutation in the rhodopsin construct where all 11 lysine residues
United States is the conversion of proline to his- were mutated to arginine (K-null P23H). We
tidine at position 23 (P23H) in the N-terminal found that the K-null P23H rhodopsin was sig-
nificantly less ubiquitylated than intact P23H
rhodopsin. We found that K-null P23H protein
A. P. F. Chen
Medical Scientist Training Program, Renaissance turnover was significantly slower compared to
School of Medicine at Stony Brook University, P23H rhodopsin through cycloheximide chase
Stony Brook, NY, USA analysis. Finally, we also generated a wild-type
e-mail: allen.chen@stonybrookmedicine.edu
rhodopsin construct where all lysines were con-
L. Chea · J. H. Lin (*) verted to arginine and found significantly
Department of Ophthalmology, Palo Alto, CA, USA
reduced ubiquitylation. Our findings identify
Department of Pathology, Stanford University, ubiquitinylation of lysine residues as an impor-
Palo Alto, CA, USA
tant posttranslational modification involved in
VA Palo Alto Healthcare System, P23H rhodopsin protein degradation.
Palo Alto, CA, USA
e-mail: l1chea@stanford.edu;
Jonathan.H.Lin@stanford.edu Keywords
E.-J. Lee
Department of Ophthalmology, Palo Alto, CA, USA Rhodopsin · Ubiquitylation · Lysine ·
ER-associated degradation · P23H
Department of Pathology, Stanford University,
Palo Alto, CA, USA
VA Palo Alto Healthcare System,
Palo Alto, CA, USA 1 Introduction
USC ROSKI Eye Institute and Department of
Ophthalmology, Keck School of Medicine, University Rhodopsin is a light-sensitive G-protein-coupled
of Southern California, Los Angeles, CA, USA receptor that is highly expressed in rod photore-
e-mail: eunjin76@stanford.edu ceptors where it compromises >90% of proteins

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 493
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_72
494 A. P. F. Chen et al.

in the rod outer segment [7]. A proline to histi- 4–15% Mini-PROTEAN TGX gels (Bio-Rad).
dine missense mutation at amino acid 23 of rho- For primary antibodies, anti-rhodopsin 1D4
dopsin (P23H) is the most common retinitis (1:1000, Santa Cruz Biotechnologies) and anti-
pigmentosa rhodopsin mutation in North America ubiquitin PD41 (1:1000, Santa Cruz
[6]. The P23H mutation causes rhodopsin protein Biotechnologies) were used and incubated at
misfolding in the endoplasmic reticulum (ER) 4 °C (24 h). Membranes were washed in TBS
[11] to undergo endoplasmic reticulum-­with 0.1% Tween-20, followed by incubation
associated degradation (ERAD) [4, 5]. with horseradish peroxidase-­coupled secondary
ERAD is a protein quality control pathway that antibody in 5% nonfat milk in washing buffer
targets misfolded ER client proteins for retrotrans- for 1 h. Immunoreactive bands were detected
location from the ER, ubiquitinylation, and protea- with the SuperSignal West chemiluminescent
somal degradation [12]. Ubiquitin is a 73-amino substrate (Pierce). ImageJ software was used to
acid polypeptide that is covalently conjugated most for protein quantification.
commonly at lysine residues of target proteins to Immunoprecipitation was performed as pre-
mark them for degradation [9]. Prior studies found viously described [4]. 1D4 anti-rhodopsin anti-
that P23H rhodopsin is robustly degraded and ubiq- body (Santa Cruz Biotechnologies) was added
uitinylated in vitro and in retina [3–5, 8]. Here, we to equal amounts of total protein-Dynabeads
performed site-­directed mutagenesis of lysine resi- Protein G (Life Technologies) and incubated for
dues on P23H rhodopsin to evaluate their roles in 30 min at 4 °C. Antibody–bead complex was
ubiquitinylation and protein turnover in vitro. crosslinked using dimethyl pimelimidate
(ThermoFisher). Aliquoted protein mixtures
were incubated overnight at 4 °C with rotation.
2 Materials and Methods Beads were then heated in 1X LDS (lithium
dodecyl sulfate) sample buffer with suspended
2.1 Cell Culture and Transfection beads at a total volume of 40 μL at 95 °C for
rhodopsin elution fraction.
HEK293FT cells were maintained in 10-cm cul-
ture plates at 37 °C at 5% CO2 in DMEM
(Mediatech) supplemented with 1% penicillin/ 2.3 Cycloheximide (CHX) Chase
streptomycin (Invitrogen) and 5% fetal bovine Assay
serum (Invitrogen). Wild-type and P23H human
rhodopsin were expressed by transient transfection Cycloheximide (10 μg/uL) was added to
of a pcDNA3.1 plasmid vector driven by HEK293FT cells after 24 h incubation of trans-
Cytomegalovirus (CMV) enhancer–promoter fection. Cells were harvested 3, 6, or 12 h after
(Invitrogen). FuGENE 6 transfection reagent addition of cycloheximide. CHX-treated cells
(Promega) was used [3, 5]. PrimeStar Mutagenesis were cultured on Poly-D Lysine-coated plates
site-directed primers were used to modify amino (EMD Millipore).
acid lysine to arginine through in-frame modifica-
tion of 5′-AAG-3′ to 5′-AGG-3′ in pcDNA3.1
(Takara Bio Inc). HEK239FT cells were trans- 2.4 Statistical Analysis
fected 24 h prior to lysis.
Representative graphical results were shown as
a mean with standard error of the mean (SEM).
2.2 Immunoblotting Analysis Commercial software program (GraphPad
and Immunoprecipitation Prism 9) was used to determine P values
through two-­way ANOVA or Student’s two-
Immunoblotting analysis and quantification tailed t-test. P value <0.05 was considered
were performed as previously published [4]. significant.
Equal amounts of protein were applied onto
Lysine Ubiquitylation Drives Rhodopsin Protein Turnover 495

3 Results CHX treatment (purple asterisk *P < 0.05,


Fig. 1e). This finding indicates that K-null P23H
3.1 P23H Rhodopsin Is Less has a greater t1/2 than P23H and supports that
Ubiquitylated When Lysine Is lysine ubiquitinylation drives P23H rhodopsin
Replaced with Arginine protein turnover.

Human rhodopsin is a 348-amino acid membrane


protein, with 11 lysines distributed across intra- 3.2 K-null WT Rhodopsin Is Less
discal/ER luminal (1 lysine), cytosolic (9 lysines), Ubiquitylated Than WT
and transmembrane (1 lysine) domains (Fig. 1a). Rhodopsin
To investigate the role of lysines in P23H rhodop-
sin ubiquitylation and degradation, we performed We also generated a WT human rhodopsin
site-directed mutagenesis to convert all 11 lysine expression construct where all 11 lysines were
residues into arginine (K-to-R) (K-null P23H) converted to arginines (K-null WT) (Fig. 1a, b).
(Fig. 1a, b) [2]. Arginine is positively charged Immunoblot analysis showed comparable levels
like lysine but cannot form an isopeptide bond of rhodopsin immunoreactive bands in both WT
with ubiquitin. First, to determine if the change in and K-null WT transfected cell lysates (Fig. 2a).
lysine residues affected rhodopsin protein expres- However, when we immunopurified WT or
sion, we visualized immunoblots of cell lysates K-null WT rhodopsin protein from total lysates,
transfected with P23H rhodopsin or K-null P23H we saw significantly less ubiquitinylation of
rhodopsin (Fig. 1c). We saw no difference in the K-null WT rhodopsin compared to WT rhodop-
levels of monomeric (35–50 kDa) or total rho- sin (Fig. 2b). This finding indicates that lysines
dopsin protein levels between P23H and K-null are also ubiquitinylated on WT rhodopsin protein
P23H (P > 0.05, Fig. 1c). This finding indicated and likely drive normal rhodopsin turnover.
that the K-to-R conversions did not interfere with
rhodopsin protein synthesis and folding in vitro.
Next, we examined how mutating all lysine resi- 4 Discussion
dues affected P23H rhodopsin ubiquitylation. We
immunopurified comparable amounts of K-null In this study, we show that ubiquitylation and
P23H and P23H rhodopsin protein (Fig. 1d). We turnover of P23H rhodopsin is lysine-dependent.
noted visibly less ubiquitylation signal using We show that the K-null P23H rhodopsin was
anti-ubiquitin antibody P4D1 on the immunopu- significantly less ubiquitylated and more stable
rified K-null P23H protein compared to P23H compared to P23H rhodopsin with intact lysine
rhodopsin (Fig. 1d). These findings indicated that residues. Single K-to-R substitutions did not alter
P23H rhodopsin undergoes ubiquitinylation on P23H rhodopsin ubiquitinylation levels (data not
its lysines. shown), and this suggests that multiple lysines on
We next examined if reduced ubiquitinylation rhodopsin are likely covalently modified. Future
of K-null P23H protein affected its half-life (t1/2). studies can identify on the combination of lysine
We collected transfected cell lysates 3, 6, and residues of P23H rhodopsin that are covalently
12 h after cycloheximide (CHX) treatment and conjugated to ubiquitin to drive its degradation.
quantified total rhodopsin protein levels at each In the absence of lysines, K-null P23H rho-
time point (Fig. 1e). WT rhodopsin protein levels dopsin was still degraded albeit at a slower pace
remained stable at all time points in our CHX (Fig. 1e). This may indicate redundant and
experiment. P23H rhodopsin protein levels compensatory protein degradation systems to
­
dropped quickly after CHX treatment with a remove P23H rhodopsin. Previously, we found
t1/2 ~ 3–6 h similar to prior studies [11]. By con- that P23H rhodopsin could be degraded through
trast, K-null P23H protein levels remained sig- proteasome and autophagy pathways [5]. In vivo,
nificantly elevated compared to P23H at 6 h after P23H rhodopsin triggered ER stress and acti-
496 A. P. F. Chen et al.

Fig. 1 Role of lysine residues on P23H rhodopsin ubiqui- Lysine residues are in red. The proline 23 converted to
tylation and turnover. (a) Schematic representation of histidine (P23H) is in blue. (b) Table lists pcDNA3.1
human rhodopsin protein with 348-amino acid sequence. expression constructs of wild-type (WT) rhodopsin; WT
Lysine Ubiquitylation Drives Rhodopsin Protein Turnover 497

Fig. 2 Role of lysine


residues on wild-type
rhodopsin
ubiquitylation. (a) Cells
were transfected with
WT or K-null WT
rhodopsin and
immunoblotted for
rhodopsin. (b)
Rhodopsin was
immunoprecipitated
from cells from (a) and
immunoblotted for
rhodopsin (top). The
membrane was stripped
and re-probed for
ubiquitin (bottom)

vated the Unfolded Protein Response, which in to ubiquitinylate P23H rhodopsin could enhance
turn regulates many anabolic and catabolic mech- its protein clearance by autophagy or proteasome
anisms (translation, autophagy, ERAD, etc.) to and potentially benefit retinitis pigmentosa
reduce cellular levels of misfolded ER client pro- patients carrying these mutations.
teins [1, 4]. Recently, modulation of autophagy
or proteasome activity in P23H rhodopsin mice Acknowledgments This study was supported by NIH
reduced retinal degeneration [10, 13]. Strategies awards R01EY027335, CIRM DISC2-10973 award, and
VA Merit I01BX002284.

Fig. 1 (continued) rhodopsin with all lysines converted to arginine (K-null); P23H rhodopsin; and P23H rhodopsin with
all lysines converted to arginine (K-null P23H). (c) Protein lysates were prepared from HEK293 cells transfected with
P23H or K-null P23H (n = 6 independent experimental replicates). Monomeric and total rhodopsin protein were
detected by immunoblotting, quantified, and normalized to loading control, Actin. Data represent mean ± SEM,
Student’s t-test, *P < 0.05. (d) Left panels show lysates of cells transfected with P23H or K-null P23H and immunoblot-
ted for rhodopsin, ubiquitin, or actin (loading control). On right, rhodopsin was immunoprecipitated from cells express-
ing P23H or K-null P23H and immunoblotted for rhodopsin (top right). The membrane was stripped and re-probed for
ubiquitin (bottom right). (e) Cells were transfected with WT, P23H, or K-null P23H. Cycloheximide (CHX) was added
after 24 h. Lysates were collected as indicated after CHX addition and a representative immunoblot for rhodopsin or
actin (loading control) is shown. Rhodopsin protein levels after CHX treatment were quantified from three experimental
repeats, normalized to actin, and are graphed relative to protein levels at start of CHX treatment. Shown are rhodopsin
protein levels in WT vs K-null P23H (red asterisk), WT vs P23H (blue asterisk), and P23H vs K-null P23H (purple
asterisk). Data represent mean ± SEM, two-way ANOVA, *P < 0.05,**P < 0.005
498 A. P. F. Chen et al.

References 7. Hargrave PA. Rhodopsin structure, function,


and topography the Friedenwald lecture. Invest
Ophthalmol Vis Sci. 2001;42(1):3–9.
1. Alavi MV, Chiang WC, Kroeger H, Yasumura D,
8. Illing ME, Rajan RS, Bence NF, Kopito RR. A rho-
Matthes MT, Iwawaki T, LaVail MM, Gould DB, Lin
dopsin mutant linked to autosomal dominant reti-
JH. In vivo visualization of endoplasmic reticulum
nitis pigmentosa is prone to aggregate and interacts
stress in the retina using the ERAI reporter mouse.
with the ubiquitin proteasome system. J Biol Chem.
Invest Ophthalmol Vis Sci. 2015;56(11):6961–70.
2002;277(37):34150–60.
2. Batonnet S, Leibovitch MP, Tintignac L, Leibovitch
9. Komander D, Rape M. The ubiquitin code. Annu Rev
SA. Critical role for lysine 133 in the nuclear
Biochem. 2012;81:203–29.
ubiquitin-­mediated degradation of MyoD. J Biol
10. Lobanova ES, Finkelstein S, Li J, Travis AM, Hao
Chem. 2004;279(7):5413–20.
Y, Klingeborn M, Skiba NP, Deshaies RJ, Arshavsky
3. Chiang WC, Hiramatsu N, Messah C, Kroeger H, Lin
VY. Increased proteasomal activity supports photore-
JH. Selective activation of ATF6 and PERK endo-
ceptor survival in inherited retinal degeneration. Nat
plasmic reticulum stress signaling pathways prevent
Commun. 2018;9(1):1738.
mutant rhodopsin accumulation. Invest Ophthalmol
11. Sung CH, Davenport CM, Hennessey JC, Maumenee
Vis Sci. 2012a;53(11):7159–66.
IH, Jacobson SG, Heckenlively JR, Nowakowski R,
4. Chiang WC, Kroeger H, Sakami S, Messah C,
Fishman G, Gouras P, Nathans J. Rhodopsin muta-
Yasumura D, Matthes MT, Coppinger JA, Palczewski
tions in autosomal dominant retinitis pigmentosa.
K, LaVail MM, Lin JH. Robust endoplasmic reticulum-­
Proc Natl Acad Sci U S A. 1991;88(15):6481–5.
associated degradation of rhodopsin precedes retinal
12. Wu X, Rapoport TA. Mechanistic insights into
degeneration. Mol Neurobiol. 2015;52(1):679–95.
ER-associated protein degradation. Curr Opin Cell
5. Chiang WC, Messah C, Lin JH. IRE1 directs protea-
Biol. 2018;53:22–8.
somal and lysosomal degradation of misfolded rho-
13. Yao J, Qiu Y, Frontera E, Jia L, Khan NW,
dopsin. Mol Biol Cell. 2012b;23(5):758–70.
Klionsky DJ, Ferguson TA, Thompson DA, Zacks
6. Dryja TP, McGee TL, Reichel E, Hahn LB, Cowley
DN. Inhibiting autophagy reduces retinal degen-
GS, Yandell DW, Sandberg MA, Berson EL. A point
eration caused by protein misfolding. Autophagy.
mutation of the rhodopsin gene in one form of retinitis
2018;14(7):1226–38.
pigmentosa. Nature. 1990;343(6256):364–6.
In Silico Prediction
of MYO1C-­Rhodopsin Interactions
and Its Significance in Protein
Localization and Visual Function

Glenn P. Lobo, Rakesh Radhakrishnan,


Matthias Leung, Andrew Gruesen, Hans-­
Joachim Knölker, Frederik J. van Kuijk,
and Sandra R. Montezuma

Abstract this study, using modeling and docking


analysis, we aimed to identify the protein–
Rods and cones are photoreceptor neurons in protein interaction sites between MYO1C and
the retina that are required for visual sensation Rhodopsin to establish a hypothesis that a
in vertebrates, where proper protein localiza- physical interaction between these proteins is
tion and compartmentalization are critical for necessary for the proper trafficking of
phototransduction and visual function. In rhodopsin and visual function.
human retinal diseases, improper protein
transport to the outer segment (OS) or mislo- Keywords
calization of proteins to the inner segment (IS)
Motor protein · Myosin 1C · Photoreceptor ·
could lead to impaired visual responses and
Rhodopsin · Retina · Protein localization ·
photoreceptor cell degeneration, causing a
Outer segments · Visual function
loss of visual function. We showed involve-
ment of an unconventional motor protein,
MYO1C, in the proper localization of rhodop-
sin to the OS, where loss of MYO1C in a 1 Introduction
mammalian model caused mislocalization of
rhodopsin to IS and cell bodies, leading to All proteins destined for the photoreceptor outer
progressively severe retinal phenotypes. In segment (OS) are synthesized in the inner
segment (IS) compartment, so they must be
G. P. Lobo (*) · R. Radhakrishnan · M. Leung · properly trafficked to reach their final functional
A. Gruesen · F. J. van Kuijk · S. R. Montezuma destination. The proper trafficking of proteins to
Department of Ophthalmology and Visual specific cellular compartments of the
Neurosciences, University of Minnesota, photoreceptor cell are critical for effective
Minneapolis, MN, USA
e-mail: lobo0023@umn.edu; rakeshr@umn.edu; phototransduction and visual function [1].
leung132@umn.edu; grues020@umn.edu; vankuijk@ Specific sorting of proteins within the
umn.edu; smontezu@umn.edu photoreceptor cell requires trafficking and plasma
H.-J. Knölker membrane targeting/localization sequences that
Faculty of Chemistry, Technische Universität serve as “biochemical zip codes.” Wherein, rapid
Dresden, Dresden, Germany and efficient transport and proper localization of
e-mail: hans-joachim.knoelker@tu-dresden.de

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 499
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_73
500 G. P. Lobo et al.

the proteins from the endoplasmic reticulum ing and docking analysis and cell culture, we
(ER) to the proximal OS is essential for the aimed to investigate the hypothesis that rhodop-
photoreceptor cell function and survival. Thus, sin is a cargo for MYO1C, which is critical for its
making the photoreceptors among the most trafficking to the photoreceptor OS, in the sup-
interesting cells to study intracellular protein port of visual function.
sorting, trafficking, and localization in retinal
health and disease conditions [2]. A fundamental
question in this regard is how the post-­ 2 Materials and Methods
biosynthesis transport of plasma membrane-­
associated proteins occurs and what mechanisms 2.1 Phylogenetic Sequence
target these proteins to the OS for disc assembly Analysis
and/or their participation in the phototransduc-
tion cascade. Thus, understanding and identify- The protein sequence of unconventional Myosin
ing the yet unknown mechanisms involved in family proteins was obtained from the NCBI
regulating and maintaining accurate protein database and an alignment and a phylogenetic
transport and compartmentalization in photore- analysis was conducted using MEGA version X
ceptors is important. The G-protein coupled [10]. The available MYO1C structures were
receptor (GPCR) Type II Opsin, Rhodopsin, con- derived from the truncated protein and alphafold
stitutes the majority (approximately 80%) of the protein structure database (https://alphafold.ebi.
OS resident proteins, and is the major player in ac.uk/), which is an AI system that predicts a
vertebrate phototransduction [3–8]. Our recent complete structure of unsolved protein structures.
work identified MYO1C as an unexpected pro- To understand the possible composition, the pre-
tein for proper rhodopsin localization to rod pho- dicted complete MYO1C structure was obtained
toreceptor OS [9]. In co-immunoprecipitation in a PDB file format and visualized and color-­
assays using retinal lysates from mice, we identi- coded in PyMOL Molecular Graphics System,
fied opsins as novel cargo for MYO1C. In the Version 2.0 Schrödinger, LLC.
absence of MYO1C, while rhodopsin was still
able to localize to OS, retinas of Myo1c−/− mice
showed progressively severe rhodopsin mislocal- 2.2 Protein–Protein Interaction
ization and retention in the photoreceptor IS and Bioinformatics Tools
cell bodies [9]. Most importantly, the global
genetic deletion of Myo1c only affected the ret- The complete structure of human MYO1C and
ina, while other systemic organs analyzed Rhodopsin was obtained from the human protein
remained unaffected. Our study identified for the sequences MYO1C (UNIPROT ID O00159) and
first time that an unconventional motor protein, RHO (UNIPROT ID P08100) against the avail-
MYO1C, is an essential component within mam- able crystal structure of human MYO1C
malian photoreceptors, where it plays a canonical N-terminus PDB 4BYF and mouse MYOIC
role in promoting the proper localization of rod C-terminus PDB 4R8G and Bovine RHO com-
opsin and in maintaining the normal visual func- plete structure 1HZX. Using the MEMDOCK
tion of the eye [9]. While the mechanisms facili- server (http://bioinfo3d.cs.tau.ac.il/Memdock;
tating the binding of MYO1C with Rhodopsin in [11]), the interactions between human MYO1C
photoreceptor cell function are still under investi- and RHO were predicted by protein–protein
gation, our computational-based studies suggest docking. The active site of this putative interac-
the binding of MYO1C with Rhodopsin likely tion was visualized by the PyMOL Molecular
occurs within the C-terminal sites of both pro- Graphics System, Version 2.0 Schrödinger.
teins, where rhodopsin has a conserved VXPX Docking analysis was further confirmed using a
motif that is necessary for its membrane localiza- second docking server, HADDOCK 2.4 [12],
tion. In this study, using a combination of model- which showed similar results. The human
In Silico Prediction of MYO1C-Rhodopsin Interactions and Its Significance in Protein Localization and… 501

MYO1C gene mutation residues that lead to hear- the alpha helix and the beta sheets (Fig. 1c). It
ing impairments and the Rhodopsin gene muta- performs numerous functions, including facilitat-
tions that cause Retinitis Pigmentosa (RP) are ing myosin association, anchoring myosins for
available in the Human Gene Mutation Database movement relative to actin filaments, and defin-
(http://www.hgmd.cf.ac.uk/). ing binding to diverse payloads [13]. Although
cells may create certain types of movement by
polymerizing actin, many cellular motions rely
2.3 In Vitro Transfections on interactions between actin filaments and myo-
and Co-localization Studies sin and ATPase that travels along actin filaments
by connecting ATP hydrolysis to conformational
COS1 cells were cultured on coverslips in changes [14]. As a result, this sort of enzyme that
DMEM (Cat. No. 10566024, Thermo Fisher transfers chemical energy into mechanical energy
Scientific, Waltham, MA) supplemented with is known as a mechanochemical enzyme or, more
10% FBS (Cat. No. F2442, Sigma-Aldrich, St. commonly, a motor protein. Myosin is the motor,
Louis, MO) and 1X Penicillin-Streptomycin anti- actin filaments are the rails that myosin travels
biotic solution (Cat. No. 30-002-CI, Corning, along, and ATP is the fuel that drives
Glendale, AZ). The pmCherry Myo1c and movement [15].
Rhodopsin GFP (Cat. No. RG211328, Origene,
Rockville, MD) plasmids were transfected into
COS1 cells using FuGENE® HD Transfection 3.2 MYO1C Interaction
Reagent (Cat. No. E2311, Promega, Madison, with Rhodopsin
WI) as per the manufacturer’s instructions. After
12 h transfection, the media was replaced with Protein–protein interactions in vesicle trafficking
fresh complete DMEM media, and after 48 h of are critical for sustaining orientation-based vesi-
incubation, cells were fixed in 4% PFA (buffered cle movement in three-dimensional cytoplasmic
in PBS) and mounted with DAPI (Vector content. Transmembrane protein like rhodopsin
Laboratories, Newark, CA) on microscope slides has many hindrances for direct physical interac-
and images were captured using a Keyence tions with cytosolic proteins (Fig. 2a). It is known
microscope (Keyence Corporation of America, that the spanning of transmembrane residues cov-
Itasca, IL) in 60X oil immersion objective. ered by lipid membrane nullifies the accessibility
by potential interaction partners, restricting the
interactions to the cytosolic overhangs of resi-
3 Results dues (Fig. 2a). The MYO1C protein is an unusual
Myosin family member with an evolutionarily
3.1 Myosin Family Controlling conserved motor domain for ATP hydrolysis and
Cytosolic Dynamics two IQ domains and one TH1 domain at its
C-terminus, according to the Uniprot database.
Myosins are a superfamily of over 20 classes that IQ domains are around 25 amino acids long,
connect and translocate along actin filaments which helps in interacting with calmodulin in the
using the energy obtained from ATP hydrolysis absence of Ca2+. Whereas the TH1 domain, also
[13]. They typically consist of three domains: (1) known as the Class1 tail homology domain, has
a conserved head responsible for actin binding, an embedded pleckstrin-homology (PH) domain
ATPase activity, and movement generation; (2) a capable of attaching to lipid membranes [16]
short neck that typically interacts with myosin (Figs. 1 and 3a, b). While the mechanisms facili-
light chains; and (3) a variable tail that commonly tating MYO1C–Rhodopsin binding in photore-
binds the motor “cargo” and determines the ceptor function are still being investigated, our in
motor’s functional specificity (Fig. 1a, b). silico studies suggest that MYO1C–Rhodopsin
Secondary structure of MYO1C is composed of binding most likely occurs within the C-terminus
502 G. P. Lobo et al.

Fig. 1 Phylogenic tree of Myosin. (a) MEGA-X software lar evolutionary analysis [10]. (b) Domains and residue
version 10.2.6 was used to align the Myosin family pro- position of MYO1C. (c) The structure of MYO1C, with
tein sequences, and a phylogenetic tree was constructed. the alpha-helix in cyan and the beta sheets in magenta
MEGA version X was used for phylogenetic and molecu-

Fig. 2 Homology modeling of MYO1C. (a) The bovine MYO1C. The complete structure of both MYO1C and
rhodopsin structure in lipid bilayer and possible sites of Rhodopsin was obtained from https://alphafold.ebi.ac.
anchoring are shown in distortion view from http://mem- uk/. [19]
protmd.bioch.ox.ac.uk [18]. (b) The predicted structure of
In Silico Prediction of MYO1C-Rhodopsin Interactions and Its Significance in Protein Localization and… 503

sites of both proteins, where rhodopsin has a con- motor domain for ATP hydrolysis and actin fila-
served VXPX motif that is required for mem- ment binding, while the C-terminus domains IQ
brane localization [17]. Our in silico modeling and TH1 help in protein and lipid binding
investigations further indicated that a putative (Fig. 1b). Interestingly, our docking analysis of
interaction between MYO1C and Rhodopsin MYO1C and Rhodopsin revealed putative con-
occurs at their C-terminal (Fig. 3a). At the IQ tact sites in which MYO1C had active amino acid
domains of MYO1C and serine and threonine residues from region 785 to 858. The two docu-
residues near the VXPX area of rhodopsin, they mented MYO1C mutations (K823N and E831K)
create hydrogen bonds and salt bridges. The previously identified in patients with deafness
VXPX region of rhodopsin’s cytosolic overhang and Rhodopsin C-terminal mutations (P34S/L,
makes it more accessible for protein–protein V345M, and Q344ter) fall within this predicted
interaction and tethering for vesicle trafficking MYO1C–RHO binding domain, explicitly sug-
(Fig. 3c). MYO1C comprises an N-terminus

Fig. 3 Protein docking and predicted active sites for patients with deafness, and Rhodopsin C-terminal muta-
MYO1C to Rhodopsin interaction. (a) MYO1C and tions (P34S/L, V345M, and Q344ter) fall within the pre-
Rhodopsin structure with predicted interaction sites are dicted MYO1C–RHO binding domain. (d) Protein–protein
shown in cartoon view. Detailed domains of MYO1C are interactions in COS1 cell line, where MYO1C tagged
indicated. The putative active binding site of MYO1C– with pmCherry (red) and Rhodopsin tagged with EGFP
Rhodopsin interaction (shown in the red circle) was (green), were overexpressed. This analysis shows co-­
obtained in the PyMOL Molecular Graphics System, localization patterns (merge, yellow) between the MYO1C
Version 2.0 Schrödinger, LLC. The protein–protein dock- and Rhodopsin and possible sites of co-localization
ing analysis was performed using MEMDOCK. (b, c) The (boxed and in the zoom panel) in the plasma membrane of
representative figure showing the domains, sites, and transfected cells, which are visible in the accompanying
genetic point mutations in MYO1C and Rhodopsin. Two intensity profiles
mutations in MYO1C (E831K and K823N) found in
504 G. P. Lobo et al.

gesting the relevance of this interaction in human Acknowledgments This work was supported by the
National Institute of Health—National Eye Institute
eye and ear pathologies (Fig. 3a–c). (NIH-NEI) grants R21EY025034 and R01EY030889 to
G.P.L. This project was also supported in part by a SCTR-
NIH/NCATS grant (5UL1TR001450) and University of
3.3 Co-Localization of MYO1C Minnesota Start-up funds to G.P.L.
with Rhodopsin in COS1 Cells

Our co-transfection experiments in COS1 cells References


demonstrated co-localization patterns of wild-­
1. Imanishi Y. Protein Sorting in Healthy and Diseased
type MYO1C with Rhodopsin at the plasma Photoreceptors. Annual Review of Vision Science.
membrane with a positive Pearson correlation 2019;5(1):73–98.
(Fig. 3d). However, to completely understand the 2. Bales KL, Gross AK. Aberrant protein trafficking
significance and implications of these protein– in retinal degenerations: The initial phase of
retinal remodeling. Experimental Eye Research.
protein interactions between MYO1C and 2016;150:71-80.
Rhodopsin in human retinal disorders, these pre- 3. Jastrzebska B, Fotiadis D, Jang GF, Stenkamp RE,
dictive interactions between MYO1C and Engel A, Palczewski K. Functional and structural
Rhodopsin must be further investigated. characterization of rhodopsin oligomers. J Biol Chem.
2006;281(17):11917–11922.
4. Jastrzebska B. GPCR: G protein complexes—the
fundamental signaling assembly. Amino Acids.
4 Discussion 2013;45(6):1303–1314.
5. Lobo GP, Ebke LA, Au A, Hagstrom SA. TULP1
Missense mutations induces the endoplasmic retic-
Understanding the mechanisms responsible for ulum unfolded protein response stress complex
protein localization and trafficking in photore- (ER-UPR). Adv Exp Med Biol. 2016a;854:223–30.
ceptor cells is crucial for understanding visual 6. Lobo GP, Au A, Kiser PD, Hagstrom
function and disorders associated with hereditary SA. Involvement of Endoplasmic reticulum stress
in tulp1 induced retinal degeneration. PLOS ONE.
retinal cell degeneration. The unexpected discov- 2016b;11(3):e0151806–10.
ery of an unconventional motor protein, MYO1C, 7. Nemet I, Ropelewski P, Imanishi Y. (2015). Trafficking
as an important component of rhodopsin (RHO) of GPCRs: rhodopsin trafficking and mistrafficking.
localization to rod OS reveals a completely Prog Mol Biol Transl Sci. 2015;132:39–71.
8. Orban T, Jastrzebska B, Palczewski K. Structural
undiscovered function for motor proteins in pro- approaches to understanding retinal proteins
tein transport and emphasizes the importance of needed for vision. Current Opinion in Cell Biology.
the myosin-tail homology domain in cargo/protein 2014;27:32–43.
localization, particularly in photoreceptor cells. 9. Solanki AK, et al. Loss of Motor Protein MYO1C
Causes Rhodopsin Mislocalization and Results in
Future in vitro and in vivo studies are therefore Impaired Visual Function. Cells. 2021;10(6):1322.
warranted to further explore the physiological 10. Tamura K, Stecher G, Kumar S. MEGA11:
role of MYO1C in Rhodopsin trafficking for Molecular Evolutionary Genetics Analysis Version
visual function. In the future, it will be intriguing 11 Abstract Molecular Biology and Evolution.
2021;38(7):3022–3027.
to understand the molecular mechanism that 11. Hurwitz N, Schneidman-Duhovny D, Wolfson
favor MYO1C interaction with the C-terminus HJ. Memdock: an α-helical membrane protein docking
end of Rhodopsin and how disruption of these algorithm. Bioinformatics. 2016;32(16):2444-2450.
interactions might be causative of protein mis- 12. van Zundert GCP, et al. The HADDOCK2.2
Web Server: User-Friendly Integrative Modeling
trafficking in photoreceptors and retinal of Biomolecular Complexes. J of Mol. Biol.
dysfunction. 2016;428(4):720–725.
In Silico Prediction of MYO1C-Rhodopsin Interactions and Its Significance in Protein Localization and… 505

13. Sellers JR. Myosins: a diverse superfamily Biochimica Putative Pleckstrin Homology Domain. Molecular
et Biophysica Acta (BBA) - Molecular Cell Research. Biology of the Cell. 2006;17(11):4856–4865.
2000;1496(1):3–22. 17. Mazelova J, et al. Ciliary targeting motif VxPx directs
14. Svitkina T. The Actin Cytoskeleton and Actin-Based assembly of a trafficking module through Arf4. The
Motility. Cold Spring Harbor Perspectives in Biology. EMBO Journal. 2009;28(3):183–192.
2018;10(1):a018267. 18. Newport TD, Sansom MSP, Stansfeld PJ. The
15. Mosby LS, et al. Myosin II Filament Dynamics MemProtMD database: a resource for membrane-­
in Actin Networks Revealed with Interferometric embedded protein structures and their lipid interactions.
Scattering Microscopy. Biophysical Journal. Nucleic Acids Research. 2019;47(D1) D390–D397.
2020;118(8):1946–1957. 19. Richard JJ, et al. Highly accurate protein
16. Hokanson DE, Laakso JM, Lin T, Sept D, Ostap structure prediction with AlphaFold. Nature.
EM. Myo1c Binds Phosphoinositides through a 2021;596(7873):583–589.
A Ciliary Branched Actin Network
Drives Photoreceptor Disc
Morphogenesis

William J. Spencer and Vadim Y. Arshavsky

Abstract many studies focusing on the role of actin dur-


ing disc morphogenesis.
The light-detecting organelle of the photorecep-
tor cell is a modified primary cilium, called the Keywords
outer segment. The outer segment houses hun-
dreds of light-sensitive membrane, “discs,” that Actin cytoskeleton · Cilia · Outer segment ·
are continuously renewed by the constant for- Photoreceptor · Vision
mation of new discs at the outer segment base
and the phagocytosis of old ones from outer seg-
ment tips by the retinal pigment epithelium. In 1 Filamentous Actin Resides
this chapter, we describe how an actin cytoskel- Within the Photoreceptor’s
eton network, residing precisely at the site of Cilium at the Site of Disc
disc formation, provides the driving force that Formation
pushes out the ciliary plasma membrane to form
each disc evagination that subsequently can Primary cilia were first shown to contain actin
mature into a bona fide disc. We highlight the nearly 40 years ago in quail oviduct cells [16]. A
functions of actin-binding proteins, particularly couple of years later, it was shown by anti-actin
PCARE and Arp2/3, that are known to partici- immunogold electron microscopy that a rela-
pate in disc formation. Finally, we describe a tively large patch of actin resides within the cil-
working model of disc formation built upon the ium of frog photoreceptor cells, at the site of disc
formation [7]. More specifically, actin was found
within the first newly forming disc evagination,
W. J. Spencer (*) whereas it is completely absent from mature
Department of Ophthalmology, Duke University, discs inside the outer segment. This result was
Durham, NC, USA corroborated in photoreceptors of numerous ver-
Duke Eye Center, Durham, NC, USA tebrate species including humans [4, 6, 20]. The
Upstate Medical University, Syracuse, NY, USA actin present at the site of disc formation was
e-mail: will.spencer@duke.edu; shown to be in a filamentous form by labeling it
spencerw@upstate.edu with myosin subfragment-1, which specifically
V. Y. Arshavsky interacts with filamentous actin making it visible
Department of Ophthalmology, Duke University, by electron microscopy [1, 5]. Furthermore, dec-
Durham, NC, USA orating actin filaments with myosin subfragment-
e-mail: vadim.arshavsky@duke.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 507
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_74
508 W. J. Spencer and V. Y. Arshavsky

1­ allows differentiation of the growing end from growth of the several disc evaginations at the
the non-growing end of an actin filament, which base of the outer segment. To interpret this result,
appear by electron microscopy as barbs or points, one must consider that each photoreceptor cell
respectively. The barbed, growing ends of the has several newly forming disc evaginations
actin filaments could be observed oriented toward present at the base of the outer segment at any
the ciliary plasma membrane, while the pointed, given time; for example, ~5 disc evaginations
non-growing ends were oriented toward the axo- observed in frog rods [2]. These evaginations are
neme. This means that these actin filaments continuous with the ciliary plasma membrane,
began to form at the axoneme and grew toward enabling them to accept membrane material
the ciliary plasma membrane. delivered through the connecting cilium so that
they can be expanded to their final size (i.e., the
diameter of the outer segment). Once the expan-
2 Inhibition of Actin sion of an immature disc is completed, it can
Polymerization Prevents enclose inside the outer segment as a fully
Initiation of Disc Formation matured disc, thereby allowing space for a new
disc evagination to form and the cycle to con-
In a classical study by Williams et al. [22], the tinue. Photoreceptor cells make a new, fully
functional significance of the actin filaments matured disc every ~20 min, which is about 18 of
present at the site of disc formation in rods and them in 6 h (Fig. 1, left). The treatment of retinas
cones was shown by treating frog retinas with for 6 h with cytochalasin D halts the formation of
cytochalasin D. This toxin tightly binds the new, discrete disc evaginations, without prevent-
barbed end of actin filaments, thereby blocking ing the trafficking of protein and lipid material to
addition of actin monomers and ultimately inhib- the cilium. As a result, 6 hours’ worth of protein
iting actin polymerization. Treating retinas with and lipid material delivered through the cilium
cytochalasin D for 6 h caused the massive over- was directed into the ~5 disc evaginations that

Fig. 1 Actin polymerization is required for the formation new disc evaginations without impairing the flow of mem-
of new disc evaginations. Left: A cartoon depicting the brane material through the cilium. This causes 6 hours’
site of photoreceptor disc formation. Photoreceptors make worth of disc membrane material (highlighted in purple)
~18 new discs every 6 h – visualized by purple highlight. to be directed into the several disc evaginations already
Right: A photoreceptor that has been incubated with the present at the start of treatment, ultimately causing them
actin polymerization inhibitor cytochalasin D for 6 h. to overgrow
Inhibiting actin polymerization prevents the formation of
A Ciliary Branched Actin Network Drives Photoreceptor Disc Morphogenesis 509

were already present at the time of treatment acute cytochalasin D treatment because the
rather than being allocated into ~18 new discs knockout was permanent, resulting in a never-­
(Fig. 1, right). As a result, the immature disc ending supply of membrane material. This work
evaginations at the base of the outer segment put forth a new model of disc formation whereby
overgrew several fold larger than normal. branched actin polymerization, nucleated by
The effect of cytochalasin D treatment on disc Arp2/3, provides the force that pushes out the
formation was corroborated in several subse- ciliary membrane to initiate disc formation. This
quent studies and shown to be a direct effect [10, mechanism is functionally analogous to the role
12, 14, 19, 21]. Most recently it was shown that of Arp2/3 in driving actin polymerization that
the overgrown disc evaginations present after protrudes the plasma membrane to form the lead-
cytochalasin D treatment can be enclosed [19]. ing edge of migrating cells, called the lamellipo-
This result implies that disc enclosure does not dia [3].
require a normal disc diameter or the action of
the actin cytoskeleton.
4 PCARE Recruits Actin
Polymerization Machinery
3 The Arp2/3 Complex Initiates to the Cilium
Actin Polymerization During
Disc Formation The Arp2/3 complex is ubiquitously expressed
by eukaryotic cells and requires other proteins to
The experiments treating retinas with cytochala- direct its action to the correct cellular location.
sin D showed that actin polymerization is Recently, a breakthrough was made in how
required for disc formation, but the molecular Arp2/3 activity is directed to the site of disc for-
players involved in the process were unknown mation in photoreceptor cells. A photoreceptor-­
until recently. To identify these molecules, specific protein called PCARE (photoreceptor
Spencer et al. [19] isolated proteins present at the cilium actin regulator; originally C2orf71) was
site of disc morphogenesis and identified them by shown to localize to the site of disc formation and
mass spectrometry. They identified a large num- recruit the actin nucleation-promoting factor,
ber of actin binding proteins, including the WASF3 to this site [9]. Nucleation-promoting
branched actin nucleating protein complex, factors are essential cofactors for Arp2/3 and pro-
Arp2/3. Because actin nucleation is the essential, vide a means to regulate branched actin polymer-
rate limiting step in the formation of actin net- ization at the right time and place [17].
works, the authors generated a conditional, rod-­ Highlighting the sufficiency of PCARE to drive
specific knockout of the Arp2/3 complex to actin polymerization in the cilia, PCARE reliably
determine its role in disc formation. The knock- targets to the cilia in cultured cells and, when co-­
out of the Arp2/3 complex from rods correlated expressed with WASF3, it causes the formation
with a loss of the actin filaments at the site of disc of actin filaments within the cilium [9, 15]. The
formation. Analogous to the case of cytochalasin loss of PCARE in rods causes a phenotype simi-
D treatment, the loss of this actin network blocked lar to that of the Arp2/3 knockout [13].
the formation of new disc evaginations without Collectively, these data show that PCARE
stopping the delivery of protein and lipid material recruits the molecular machinery that initiates
to the cilium. This caused the immature disc branched actin polymerization at the site of disc
evaginations already present at the time of knock- formation to create new disc evaginations.
out to overgrow into massive membrane whorls, A couple of questions remain regarding
which completely disrupted outer segment struc- PCARE’s function in photoreceptors. Because
ture, eventually leading to photoreceptor cell PCARE localizes throughout the entire length of
death. The extent of disc overgrowth in the the cilium in cultured cells [9, 15], it remains to
Arp2/3 knockout was much greater than upon be determined how it precisely localizes to the
510 W. J. Spencer and V. Y. Arshavsky

site of disc formation rather than the whole length directly to the site of disc formation and recruits
of the outer segment. Furthermore, WASF3 is WASF3. WASF3 and Arp2/3 work together to
known to require the activity of the small GTPase, nucleate branched actin filaments that polymer-
Rac1 [8], yet mice containing a rod specific ize from the axoneme toward the ciliary plasma
knockout of Rac1 have completely normal disc membrane. In step 2, continued branched actin
morphogenesis [11]. Future studies are required polymerization provides the force that pushes
to determine if WASF3 has a direct role in disc out the ciliary membrane to form a broad mem-
formation and whether any small GTPase is brane protrusion called a “lip” in early literature
involved in the process. [7]. In step 3, the branched actin filaments depo-
lymerize, allowing the newly formed immature
disc evagination to flatten. In step 4, the
5 A Cycle of Actin branched actin filaments have completely depo-
Polymerization lymerized to allow the immature disc evagina-
and Depolymerization tion to become perfectly flat and the cycle to
Initiates Disc Formation repeat itself. At any given time, there are several
immature disc evaginations at the base of the
These findings can be summarized in the current outer segment (see Spencer et al. [18] for a
model about the role of actin during disc mor- detailed description and 3D animation of this
phogenesis (Fig. 2). In step 1, PCARE localizes process).

Fig. 2 The actin dynamics cycle that initiates disc forma- structural component of these filaments. In step 3, while
tion. A cycle of actin polymerization/disassembly begins the membrane remains protruded, actin filaments are dis-
in step 1 when PCARE (photoreceptor cilium actin regu- assembled and retracted allowing the evaginated mem-
lator) recruits an actin nucleation-promoting factor, like brane to flatten. In step 4, actin has completely retracted,
WASF3, to the site of disc morphogenesis. This actin and the initiation step of disc formation is complete.
nucleation-promoting factor associates with the Arp2/3 Meanwhile, the newly forming disc continues to elongate
complex to nucleate branched actin filaments (F-actin). In through ongoing addition of membrane material.
step 2, continued actin polymerization pushes the ciliary Reprinted with permission from Spencer et al. [18]
membrane outward. The Arp2/3 complex remains as a
A Ciliary Branched Actin Network Drives Photoreceptor Disc Morphogenesis 511

Acknowledgments This work was supported by NIH from photo-oxidative stress without affecting their
grants EY012859, EY030451, and EY005722; the structure or function. Proc Natl Acad Sci U S A.
Knights Templar Eye Foundation; and Unrestricted Grant 2009;106:9397–402.
from Research to Prevent Blindness. 12. Kaplan MW. Disk membrane initiation and inser-
tion are not required for axial disk displacement in
Xenopus laevis rod outer segments. Curr Eye Res.
1998;17:73–8.
References 13. Kevany BM, Zhang N, Jastrzebska B, et al. Animals
deficient in C2Orf71, an autosomal recessive reti-
1. Arikawa K, Williams DS. Organization of actin fila- nitis pigmentosa-associated locus, develop severe
ments and immunocolocalization of alpha-actinin in early-onset retinal degeneration. Hum Mol Genet.
the connecting cilium of rat photoreceptors. J Comp 2015;24:2627–40.
Neurol. 1989;288:640–6. 14. Nemet I, Tian G, Imanishi Y. Submembrane assem-
2. Besharse JC, Hollyfield JG, Rayborn ME. Turnover bly and renewal of rod photoreceptor cGMP-gated
of rod photoreceptor outer segments. II. Membrane channel: insight into the actin-dependent pro-
addition and loss in relationship to light. J Cell Biol. cess of outer segment morphogenesis. J Neurosci.
1977;75:507–27. 2014;34:8164–74.
3. Blanchoin L, Boujemaa-Paterski R, Sykes C, et al. 15. Nishimura DY, Baye LM, Perveen R, et al.
Actin dynamics, architecture, and mechanics in cell Discovery and functional analysis of a retinitis
motility. Physiol Rev. 2014;94:235–63. pigmentosa gene, C2ORF71. Am J Hum Genet.
4. Chaitin MH, Bok D. Immunoferritin localization of 2010;86:686–95.
actin in retinal photoreceptors. Invest Ophthalmol Vis 16. Sandoz D, Gounon P, Karsenti E, et al.
Sci. 1986;27:1764–7. Immunocytochemical localization of tubulin,
5. Chaitin MH, Burnside B. Actin filament polarity at actin, and myosin in axonemes of ciliated cells
the site of rod outer segment disk morphogenesis. from quail oviduct. Proc Natl Acad Sci U S A.
Invest Ophthalmol Vis Sci. 1989;30:2461–9. 1982;79:3198–202.
6. Chaitin MH, Carlsen RB, Samara GJ. Immunogold 17. Soderling SH. Grab your partner with both hands:
localization of actin in developing photoreceptor cytoskeletal remodeling by Arp2/3 signaling. Sci
cilia of normal and rds mutant mice. Exp Eye Res. Signal. 2009;2:pe5.
1988;47:437–46. 18. Spencer WJ, Lewis TR, Pearring JN, et al.
7. Chaitin MH, Schneider BG, Hall MO, et al. Actin in Photoreceptor discs: built like Ectosomes. Trends Cell
the photoreceptor connecting cilium: immunocyto- Biol. 2020;30:904–15.
chemical localization to the site of outer segment disk 19. Spencer WJ, Lewis TR, Phan S, et al. Photoreceptor
formation. J Cell Biol. 1984;99:239–47. disc membranes are formed through an Arp2/3-­
8. Chen Z, Borek D, Padrick SB, et al. Structure and dependent lamellipodium-like mechanism. Proc Natl
control of the actin regulatory WAVE complex. Acad Sci U S A. 2019;116:27043–52.
Nature. 2010;468:533–8. 20. Vaughan DK, Fisher SK. The distribution of F-actin
9. Corral-Serrano JC, Lamers IJC, van Reeuwijk J, et al. in cells isolated from vertebrate retinas. Exp Eye Res.
PCARE and WASF3 regulate ciliary F-actin assem- 1987;44:393–406.
bly that is required for the initiation of photoreceptor 21. Vaughan DK, Fisher SK. Cytochalasin D disrupts
outer segment disk formation. Proc Natl Acad Sci U S outer segment disc morphogenesis in situ in rabbit
A. 2020;117:9922–31. retina. Invest Ophthalmol Vis Sci. 1989;30:339–42.
10. Hale IL, Fisher SK, Matsumoto B. The actin network 22. Williams DS, Linberg KA, Vaughan DK, et al.
in the ciliary stalk of photoreceptors functions in Disruption of microfilament organization and deregu-
the generation of new outer segment discs. J Comp lation of disk membrane morphogenesis by cytochala-
Neurol. 1996;376:128–42. sin D in rod and cone photoreceptors. J Comp Neurol.
11. Haruta M, Bush RA, Kjellstrom S, et al. Depleting 1988;272:161–76.
Rac1 in mouse rod photoreceptors protects them
Part XII
RPE
Revisiting the Daily Timing of POS
Phagocytosis

Antonio E. Paniagua, Harjas S. Sabharwal,


Kausalya Kethu, Andrew W. Chang,
and David S. Williams

Abstract Keywords

Retinal pigment epithelium (RPE) cells daily Phagocytosis · Phagosome · RPE ·


ingest the tips of the photoreceptor outer seg- Photoreceptor outer segment · Daily rhythm
ments (POSs), with phagosome number vary-
ing throughout a 24-h cycle. A major focus in
the literature has been on a peak in phagosome 1 Introduction
concentration shortly after lights-on.
Moreover, this peak has frequently been Electron micrographs of photoreceptor outer
inferred to represent a peak in POS tip inges- segment (POS) phagosomes were first published
tion. Here, we have reviewed old and new lit- in the early 1960s [1]. Subsequent autoradiography
erature on the daily cycle of phagosome experiments demonstrated that these structures
number in the RPE and conclude that there is were indeed phagosomes, originating from the
more variation in the timing of phagosome POS tips, as part of the continual renewal of POS
concentration peaks than is currently acknowl- disk membranes [2, 3]. The entire POS is renewed
edged. We also discuss that phagosome quan- in ~10 days in the rat, mouse, and monkey and
tity is affected by the rate of phagosome 40–50 days in frogs [3–5]. Given these renewal
degradation as well as the rate of ingestion; rates, the relative sizes of the POSs and
given that phagosome half-life may not be phagosomes, and the average number of
constant throughout the daily cycle, maximal photoreceptors that interface a single retinal
POS ingestion may not necessarily coincide pigment epithelium (RPE) cell [4, 6], each RPE
with a peak in phagosome concentration. cell degrades 50–300 phagosomes daily in a
monkey retina, and 200–400 in a mouse retina.
The ingestion and degradation of the POS tips is
essential for photoreceptor cell viability, as any
impairment in either POS ingestion or degradation
A. E. Paniagua (*) · H. S. Sabharwal · K. Kethu · leads to retinal degeneration [7, 8].
A. W. Chang · D. S. Williams (*) Phagocytosis involves two consecutive
Departments of Ophthalmology and Neurobiology,
Stein Eye Institute, David Geffen School of Medicine processes: ingestion of the POS tip, followed by
at UCLA, Los Angeles, CA, USA degradation of the phagosome. Although
e-mail: paniagua@ucla.edu; hsabharwal@ucla.edu; reference to “disk shedding” that precedes
krkethu@g.ucla.edu; andrewchang0731@ucla.edu; ingestion of shed POS particles is common in the
dswilliams@ucla.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 515
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_75
516 A. E. Paniagua et al.

literature, there is no convincing evidence of shed similar size, one after lights-on and another after
packets of disk membranes awaiting ingestion. lights-off [15].
Loss of the POS tip requires normal attachment Studies on two other animals also found two
to the RPE [9] and appears to transpire only as daily peaks in phagosome numbers. First, in the
part of ingestion by the RPE. Phagosome mouse, a main peak after lights-on was accompa-
degradation occurs entirely within the RPE [10]. nied by a small elevation in phagosome number
In 1976, LaVail reported a daily peak in during the night (about double that during the lat-
phagosome number shortly after the onset of ter part of the day) [5]. Then, the duplex retina of
light in rats [11]. A similar peak was described in a tree squirrel was reported to have a similar dou-
frogs [12]. Although subsequent studies with ble peak, although the second peak was not evi-
other species showed different types of daily dent until the middle of the night [16].
rhythms in phagosome number, the prevailing While these studies were consistent with the
dogma is that phagosome number is highest after hypothesis of phagosomes from rod outer seg-
lights-on. Moreover, the daily variation in phago- ments (OSs) after lights-on and cone OSs after
some number has often been interpreted to indi- lights-off, they were followed by studies, from
cate a comparable variation in ingestion, but this the early 1980s, that did not support this
assumes that phagosome degradation is coupled hypothesis.
directly with ingestion so that phagosome half-­ Fisher and colleagues found that in the
life remains constant. We are questioning these domestic cat, a crepuscular feline that has a
assumptions. Here, we have reviewed the litera- duplex retina, phagosome concentration is
ture with respect to the daily rhythm of phago- elevated after lights-on and during the latter part
some number. We discuss limitations in the of the dark phase. Notably, rod and cone
conclusions that can be drawn from the reported phagosomes, which could be distinguished based
results. on their initial location, were present during both
periods [17]. The same group also studied the
tree shrew, which is a diurnal mammal with
2 Daily Cycle of Phagosome mostly cone photoreceptors; their results showed
Number that a single peak in phagosome concentration
occurred in the morning [18]. Thus, the cat and
The 1976 studies on rat and frog [11, 12] tree shrew studies indicated that peaks in
concerned phagosomes mainly from rods. phagosome concentration could not be explained
Shortly after that, a study on the all-cone retina generally by differences in the timing of rod and
of a lizard found that, in contrast, phagosome cone phagosomes.
number peaked after lights-off [13]. Young A study of four Rhesus macaques was also
thus proposed that the disks from each inconsistent with the hypothesis of rod phago-
photoreceptor type are ingested when the cell somes at lights-on, cone phagosomes at lights-­off.
becomes less functional: rods at lights-on, Retinas were fixed at 1 or 5 h after lights-on or
cones at lights-off. lights-off. Phagosome number was found to vary
This idea was supported by his subsequent little among the timepoints in the foveal RPE,
studies on rods and cones in duplex retinas. In the where cone density is highest. However, in extra-
goldfish, which has five times more rods than foveal RPE, where there are 20 times more rods
cones, a large peak of phagosome number than cones, phagosomes were most abundant at
occurred after lights-on and was attributed to rod 5 h after lights-off, with a secondary peak in
disks, while a smaller one occurred after lights-­ phagosome number at 1 h after lights-on [19]. This
off and was attributed to cone disks [14]. In the study suffers from the limited number of individu-
chicken retina, where cones make up the majority als analyzed, but so far, it represents the only his-
of photoreceptors, there were also two peaks of
Revisiting the Daily Timing of POS Phagocytosis 517

tological analysis of phagosome number at move up to 100 μm from the apical side, where
different times of day in the RPE of any primate. they are ingested, to the basal side, where their
More recently, specific antibodies that degradation is completed. Therefore, the apical
distinguish rod and cone phagosomes from each phagosomes represent newly formed phago-
other were used to determine the rod or cone somes, while more mature phagosomes localize
origin of phagosomes at different times of day. In toward the basal region [23–25]. The opossum
the Nile rat, a diurnal rodent with a 2:1 ratio of has two peaks in phagosome number: the main
rods to cones, a morning peak in phagosomes one after lights-on and a second one during the
from both rods and cones was observed, together night. Interestingly, when phagosomes are classi-
with a small elevation in phagosome number fied according to their location along the apical–
during the night [20]. A study from Besharse’s basal axis of the RPE, the authors observed that
lab, on zebrafish and mouse, then showed two the night peak is mainly formed by apical phago-
similar peaks in phagosome number, one shortly somes, while the morning peak contains mostly
after lights-on and another shortly after lights-off. basal phagosomes; the number of new (apical)
Moreover, at all times and in both species, the phagosomes is similar at night and after
phagosomes originated from both rods and cones lights-on.
[21]. Young’s findings in the chick retina [15]
In conclusion, the daily cycle of phagosome present a similar situation. The POS phagosome
number commonly includes an elevation during peak after lights-on was formed by a greater
the dark phase, as well as after lights-on, with proportion of old phagosomes (in which the disk
both periods containing phagosomes from both membranes showed perturbed organization by
rods and cones. electron microscopy) compared with the peak
after lights-off.
These observations, based on a distinction
3 Ingestion and Degradation between new and old phagosomes, indicate that
Rates the relationship between POS ingestion and deg-
radation differs between the two time periods.
The number of phagosomes at any given time of Without having a direct measure of at least one of
day is a consequence of not only the rate of inges- the processes, however, it is unclear how it dif-
tion but also the rate of phagosome degradation. fers. For example, the increase in basal phago-
An increase in phagosome number is a result of a somes after lights-on could result from a decrease
higher ingestion rate than degradation rate, and a in ingestion coupled with a decrease in the rate of
decrease in phagosome number occurs when degradation, so that most of the phagosomes may
degradation rate overcomes the ingestion rate. have been formed before lights-on and then
However, a change in phagosome number has degraded slowly (i.e., an increase in phagosome
often been interpreted as a change in the rate of half-life). In this case, an increase in POS phago-
ingestion, despite the lack of evidence on whether some number would occur when POS tip inges-
the rate of phagosome degradation remains con- tion has decreased. However, an alternative
stant or varies. The ability to distinguish early explanation is that an increase in ingestion after
phagosomes from late phagosomes is helpful in lights-on was met by an increase in the rate of
addressing whether the relationship between maturation of new to old phagosomes.
POS ingestion and degradation is constant or dif- Further insight into the process of phagosome
fers among different times of day. degradation could be gained by studying phago-
Herman and Steinberg [22] analyzed RPE somes with markers that indicate different states
phagosomes in the opossum, a nocturnal marsu- of maturation. For example, the degradation of
pial with a rod dominant retina. The large height rhodopsin (RHO), the main protein present in
of its RPE cells makes this animal a useful model phagosomes from rods, has been shown to occur
to study phagosome maturation, as phagosomes in at least two different steps that can be identi-
518 A. E. Paniagua et al.

fied with different antibodies against tion, changes in POS length are affected by the
RHO. Immunolabeling with RHO mAb 1D4, rate of disk membrane morphogenesis as well as
which recognizes a C-terminal epitope, is limited POS tip ingestion. So, while these studies on liv-
to new phagosomes; phagosomes lose this label- ing retinas provide important insight, they too
ing quite quickly, while continuing to be labeled have their limitations. Also, unfortunately, nei-
with other RHO antibodies [24, 26]. This ther report included measurements from
approach has been used to examine defects in nighttime.
phagosome degradation [8, 25].

5 Conclusion
4 POS Tip Ingestion
Earlier studies indicated a pattern of phagocytosis
Although variation in POS tip ingestion, of rod OS tips after lights-on and phagocytosis of
according to a daily cycle, cannot be inferred cone OS tips after lights-off. However, later
directly from a cycle in phagosome numbers, studies identified inconsistencies. Studies
there is evidence for such a daily variation. The reported rod and cone OS phagosomes occurring
initial stage of ingestion of a POS tip involves at the same time, as well as elevated numbers of
binding to the RPE apical surface, which is both rod and cone OS phagosomes after lights-on
facilitated by the integrin, αvβ5 [27]. In β5 and during the night.
integrin-deficient mice, POS phagosomes are Variation in POS phagosome number clearly
still evident in the RPE, but there is a smaller demonstrates a daily cycle in POS tip phagocyto-
daily variation in the number of phagosomes sis. However, a better understanding of the rela-
(~two-fold, compared with ~five-­fold in wild- tionship between POS tip ingestion and the rate
type), with the number after lights-­on reduced of degradation of the ensuing phagosome is still
(by about half of that in wild-type) [28]. Since the needed to be able to account for the mechanisms
primary effect of the β5 integrin-­deficiency is underlying this daily variation in POS phago-
presumably on POS tip ingestion, rather than some number—despite the publication of the first
degradation, this observation supports a daily reports on this topic occurring 45 years ago.
variation in POS tip ingestion.
With the relatively recent development of Acknowledgments The authors are supported by a
retinal live imaging methods, attempts have been BrightFocus Foundation postdoctoral fellowship (A.E.P.,
M2021004F) and NIH R01EY027442 grant.
made to detect correlates to the ingestion of the
POS tip. Changes in cone OS tip reflection, deter-
mined by adaptive optics-optical coherence
tomography (AO-OCT) of human retinas, were References
presented to indicate a change in cone OS length
1. Dowling JE, Gibbons IR. The fine structure of the
and thus removal of the cone OS tip. A transient pigment epithelium in the albino rat. J Cell Biol.
loss in the cone OS tip reflection occurred more 1962;14:459–74.
frequently during the morning than during the 2. Young RW. The renewal of photoreceptor cell outer
segments. J Cell Biol. 1967;33:61–72.
afternoon or the evening. It was argued that this
3. Young RW, Bok D. Participation of the retinal pigment
change in reflection is an optical signature of epithelium in the rod outer segment renewal process. J
cone OS tip loss [29]. Another study measured Cell Biol. 1969;42:392–403.
rod OS length in live mice, using OCT, at differ- 4. Young RW. The renewal of rod and cone outer
segments in the rhesus monkey. J Cell Biol.
ent times of day. A 10% shortening of rod OS
1971;49:303–18.
length was detected over a 4.5-h period begin- 5. Besharse JC, Hollyfield JG. Turnover of mouse
ning 0.5 h before lights-on [30]. Of course, just as photoreceptor outer segments in constant
phagosome number is affected by the rate of light and darkness. Invest Ophthalmol Vis Sci.
1979;18:1019–24.
phagosome degradation as well as POS tip inges-
Revisiting the Daily Timing of POS Phagocytosis 519

6. Volland S, Esteve-Rudd J, Hoo J, Yee C, Williams 20. Bobu C, Hicks D. Regulation of retinal photoreceptor
DS. A comparison of some organizational phagocytosis in a diurnal mammal by circadian clocks
characteristics of the mouse central retina and the and ambient lighting. Invest Ophthalmol Vis Sci.
human macula. PLoS One. 2015;10:e0125631. 2009;50:3495–502.
7. Gal A, Li Y, Thompson DA, Weir J, Orth U, Jacobson 21. Lewis TR, Kundinger SR, Link BA, Insinna C,
SG, et al. Mutations in MERTK, the human orthologue Besharse JC. Kif17 phosphorylation regulates
of the RCS rat retinal dystrophy gene, cause retinitis photoreceptor outer segment turnover. BMC Cell
pigmentosa. Nat Genet. 2000;26:270–1. Biol. 2018;19:25.
8. Esteve-Rudd J, Hazim RA, Diemer T, Paniagua AE, 22. Herman KG, Steinberg RH. Phagosome movement
Volland S, Umapathy A, et al. Defective phagosome and the diurnal pattern of phagocytosis in the tapetal
motility and degradation in cell nonautonomous RPE retinal pigment epithelium of the opossum. Invest
pathogenesis of a dominant macular degeneration. Ophthalmol Vis Sci. 1982;23:277–90.
Proc Natl Acad Sci U S A. 2018;115:5468–73. 23. Gibbs D, Kitamoto J, Williams DS. Abnormal
9. Williams DS, Fisher SK. Prevention of rod phagocytosis by retinal pigmented epithelium that
disk shedding by detachment from the retinal lacks myosin VIIa, the Usher syndrome 1B protein.
pigment epithelium. Invest Ophthalmol Vis Sci. Proc Natl Acad Sci U S A. 2003;100:6481–6.
1987;28:184–7. 24. Wavre-Shapton ST, Meschede IP, Seabra MC,
10. Lakkaraju A, Umapathy A, Tan LX, Daniele L, Philp Futter CE. Phagosome maturation during
NJ, Boesze-Battaglia K, et al. The cell biology of endosome interaction revealed by partial rhodopsin
the retinal pigment epithelium. Prog Retin Eye Res. processing in retinal pigment epithelium. J Cell Sci.
2020:100846. 2014;127:3852–61.
11. LaVail MM. Rod outer segment disk shedding in 25. Jiang M, Esteve-Rudd J, Lopes VS, Diemer
rat retina: relationship to cyclic lighting. Science. T, Lillo C, Rump A, et al. Microtubule motors
1976;194:1071–4. transport phagosomes in the RPE, and lack of
12. Basinger S, Hoffman R, Matthes M. Photoreceptor KLC1 leads to AMD-­like pathogenesis. J Cell Biol.
shedding is initiated by light in the frog retina. 2015;210:595–611.
Science. 1976;194:1074–6. 26. Esteve-Rudd J, Lopes VS, Jiang M, Williams DS. In
13. Young RW. The daily rhythm of shedding and vivo and in vitro monitoring of phagosome maturation
degradation of cone outer segment membranes in the in retinal pigment epithelium cells. Adv Exp Med
lizard retina. J Ultrastruct Res. 1977;61:172–85. Biol. 2014;801:85–90.
14. O'Day WT, Young RW. Rhythmic daily shedding 27. Finnemann SC, Bonilha VL, Marmorstein AD,
of outer-segment membranes by visual cells in the Rodriguez-Boulan E. Phagocytosis of rod outer
goldfish. J Cell Biol. 1978;76:593–604. segments by retinal pigment epithelial cells
15. Young RW. The daily rhythm of shedding and requires alpha(v)beta5 integrin for binding but
degradation of rod and cone outer segment not for internalization. Proc Natl Acad Sci U S A.
membranes in the chick retina. Invest Ophthalmol Vis 1997;94:12932–7.
Sci. 1978;17:105–16. 28. Nandrot EF, Kim Y, Brodie SE, Huang X,
16. Tabor GA, Fisher SK, Anderson DH. Rod and cone Sheppard D, Finnemann SC. Loss of synchronized
disc shedding in light-entrained tree squirrels. Exp retinal phagocytosis and age-related blindness in
Eye Res. 1980;30:545–57. mice lacking alphavbeta5 integrin. J Exp Med.
17. Fisher SK, Pfeffer BA, Anderson DH. Both rod and 2004;200:1539–45.
cone disc shedding are related to light onset in the cat. 29. Kocaoglu OP, Liu Z, Zhang F, Kurokawa K, Jonnal RS,
Invest Ophthalmol Vis Sci. 1983;24:844–56. Miller DT. Photoreceptor disc shedding in the living
18. Immel JH, Fisher SK. Cone photoreceptor shedding human eye. Biomed Opt Express. 2016;7:4554–68.
in the tree shrew (Tupaia belangerii). Cell Tissue Res. 30. Zhang P, Shibata B, Peinado G, Zawadzki RJ,
1985;239:667–75. FitzGerald P, Pugh EN Jr. Measurement of diurnal
19. Anderson DH, Fisher SK, Erickson PA, Tabor variation in rod outer segment length in vivo in mice
GA. Rod and cone disc shedding in the rhesus with the OCT optoretinogram. Invest Ophthalmol Vis
monkey retina: a quantitative study. Exp Eye Res. Sci. 2020;61:9.
1980;30:559–74.
Inhibition of Bacterial
Peptidoglycan Cytopathy
by Retina Pigment Epithelial
PGRP2 Amidase

Marlyn P. Langford, Laura A. Perilloux-Lyons,


and A. Scott Kavanaugh

Abstract late NOD-mediated cytopathic activity. The


failure of RPE NAMAA to degrade pro-­
Peptidoglycan (PGN) recognition protein 2 inflammatory PGN may play a role in bacte-
(PGRP2; N-acetylmuramyl-l-alanine amidase rial retinopathies.
(NAMAA)) activity in corneal epithelial cells
is thought to inhibit corneal inflammation by Keywords
reducing the PGN-induced cytokines. PGRP2
has not been reported in human retinal pig- Bacteria · Cytopathy · Meso-diaminopimelic
ment epithelial (RPE) cells. RPE cell lysate acid · Muramyl dipeptide · N-acetylmuramyl-­
NAMAA activity was measured densitometri- l-alanine amidase · NOD agonist ·
cally via cleavage of FITC-tagged muramyl Retinopathy
dipeptide (FITCMDP). RPE lysate degradation
of the cytopathic activity of nucleotide-­
binding oligomerization domain (NOD)
receptor agonists was assessed by caspase-3 1 Introduction
activation and DNA ladder detection and
quantitation. PGRP2/NAMAA protein was Retinopathy is not associated with most bacterial
detected in RPE cells by immunofluorescent infections, but central serous chorioretinopathy
antibody assay. RPE lysate NAMAA cleaved (CSCR) has been reported in association with
FITC
MDP in a dose- and time-dependent man- latent Mycobacterium tuberculosis (MTB) [1–3]
ner. RPE lysate selectively inhibited PGN or Helicobacter pylori infection [4], supporting
cytopathic activity of NOD1 agonists contain- the concept that chronic retinal infection and/or
ing d-γ-glutamyl-meso-diaminopimelic acid shedding of bacterial cell wall peptidoglycan
and NOD2 containing l-alanyl-d-­ (PGN) may play a role in CSCR.
isoglutamine. The results suggest RPE PGRP2 Mammalian cells have two intracellular
amidase selectively degrades PGN that stimu- PGN sensors, nucleotide-binding oligomeriza-
tion domain receptors (NOD1 and NOD2) and
four peptidoglycan (PGN) recognition proteins
M. P. Langford (*) · L. A. Perilloux-Lyons ·
A. S. Kavanaugh (PGRP1–4), that can modulate the inflamma-
Department of Ophthalmology, Louisiana State tory response during bacterial infection [5].
University Health Sciences Center, Shreveport, LA, NOD1 activation by PGN containing γ-d-
USA glutamyl-­ meso-diaminopimelic acid and
e-mail: marlyn.langford@lsuhs.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 521
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_76
522 M. P. Langford et al.

NOD2 activation by PGN containing reacted with 1:1000 dilution of rabbit (poly-
N-acetylmuramyl-l-alanyl-d-­i soglutamine clonal) anti-PGRP2 antibody (Fischer
induce pro-inflammatory cytokines [6, 7]. Scientific), and incubated for 6 h. Unreacted
While NODs exhibit no antibacterial activity, anti-PGRP2 antibody was removed and the RPE
PGRP 1, 3, and 4 have bactericidal a­ ctivity and cells rinsed 3× with PBS. Secondary FITC-
PGRP2 is an N-acetylmuramyl-l-alanine ami- tagged goat anti-­ rabbit IgG (1:1000 dilution;
dase (NAMAA) that cleaves the lactyl bond Jackson Laboratories, West Grove, PA) incu-
between N-acetyl muramic acid and l-alanine bated overnight at 4 °C on treated and control
of PGN [8, 9]. NAMAA reduces NOD- slides. The PGRP2 positive RPE cells were
dependent pro-inflammatory activity of bacte- visualized by fluorescent microscopy using the
rial PGN and is thought to protect against method previously described [11].
bacterial-induced inflammation by suppressing
the induction of pro-inflammatory cytokines
[9]. The objective of the current investigation
was to determine if human retinal pigmented Table 1 Peptidoglycan NOD agonists
epithelial (RPE) cells express PGRP2 and NOD Cell
exhibit NAMAA activity against PGN NOD Acronym Peptidoglycan agonist type deatha
agonists. iE-DAP d-γ-glutamyl-meso- 1 24 h
diaminopimelic acid
C12-iE-­ Lauroyl-d-γ-glutamyl- 1 4h
DAP meso-­diaminopimelic acid
2 Materials and Methods Tri-DAP l-alanyl-d-γ-glutamyl-­­ 1 6h
meso-diaminopimelic
2.1 Cell Culture acid
iE-Lys γ-d-glutamyl-l-lysine, 1 >24
Negative control
Human retinal pigmented epithelial (RPE; ARE-­ M-Tri-­ MurNAc-l-alanyl-d-γ-­­ 1/2 6h
19) cells and rabbit kidney (RK13) cells DAP glutamyl-meso-­
(American Type Culture Collection, Manassas, diaminopimelic acid
VA) were grown in 6-well culture dishes PGNEc Insoluble preparation of 1/2 24 h
(Starstedt, Inc., Fischer Scientific, fishersci.com) PGN from E. coli
MDP Muramyl dipeptide; 2 4h
in Dulbecco’s medium (Sigma-Aldrich, St. MurNAc-l-alanyl-d-
Louis, MO) supplemented with 10% calf serum isoglutamine
and antibiotics as previously reported [10]. DD-MDP MurNAc-d-alanyl-d- 2 >24
isoglutamine; Negative
control
L18-MDP Stearoyl-l-alanyl-d- 2 4h
2.2 Peptidoglycan Reagents isoglutamine
Dx-MDP 4-(2-acetamido-2-deoxy-β-­­ 2 6h
Fluorescein isothiocyanate (FITC)-labeled mur- d-glucopyranosyl-­
amyl dipeptide (FITCMDP) as well as five Nod1, MurNAc-­l-alanyl-d-­­
glutamate amide
seven Nod2, and two Nod1/2 agonists (InvivoGen,
M-Tri-­ MurNAc-l-alanyl-d- 2 4h
San Diego, CA) were dissolved in sterile water Lys isoglutaminyl-­Lys
(see Table 1). MBT Murabutide; 2 12 h
MurNAc-l-alanyl-d-
GlnOBu
MFT Liposomal muramyl 2 8h
2.3 Immunofluorescent Antibody
tripeptide [Mifamurtide]
Assay PGN Sa Insoluble preparation of 1/2 12 h
PGN from S. aureus
Slide cultures of RPE cells were fixed by 15 min a
Cell death, incubation period needed to detect >25%
exposure to 70% ethanol, rinsed with PBS (3×), nonviable RK13 cells at 1 μg/mL dose
Inhibition of Bacterial Peptidoglycan Cytopathy by Retina Pigment Epithelial PGRP2 Amidase 523

2.4 Cleavage of FITCMDP by RPE 2.6 Statistical Analysis


Lysate
The mean and standard deviation were deter-
RPE cells grown in 6-well tissue culture plates mined between triplicate experiments and statis-
for 24–48 h were rinsed with PBS (3×), cells har- tical inferences were based upon Student t-test
vested by plastic spatula into 150 μL PBS/cul- analysis; p-values <0.05 were considered
ture, pipetted into Eppendorf vials, and frozen significant.
(−80 °C). FITCMDP was added to PBS or RPE cell
lysate and incubated for up to 72 h at 37 °C. Five
microliters of sucrose (40% w/v) was added to all 3 Results
RPE lysate/FITCMDP and FITCMDP/PBS samples
and vortexed before 30 μL of each was added to a 3.1 Detection of RPE PGRP2
1% agarose gel lane and electrophoresed at and NAMAA Activity
65 mV for 1.5 h. The gel was scanned under UV
light and digital images were obtained using a PGRP2 (NAMAA) was detected in RPE cells
Bio-Rad gel documentation system and Quantity (Fig. 1A). FITCMDP incubation with RPE cell
One software (Bio-Rad, Hercules, CA). A densi- lysate (60 μg) for 24 h resulted in a > 75% shift in
tometric analysis of the gel signals was generated FITC
MDP signal (Fig. 1B) consistent with genera-
using free-download ImageJ software. tion of low molecular weight FITCMurNAc by
NAMAA cleavage (Fig. 1C).

2.5 NOD-Induced Cytopathy


3.2 Concentration and Time-­
Cytopathy, characterized by caspase-3 activation Dependent RPE Lysate
(using the Caspase-3 Assay Colorimetric Kit as NAMAA Cleavage of FITCMDP
per the instructions provided (Sigma)) and poly-
meric DNA fragmentation, was induced by the Incubation of FITCMDP with increasing RPE
1 μg/mL NOD1 and NOD2 agonists as previ- lysate protein concentrations for 5 h at 37 °C
ously reported [12]. The RK13 cell cultures were resulted in incremental increases in free
viewed microscopically through 24 h for the FITC
MurNAc consistent with concentration-­
appearance of dead cells. dependent NAMAA activity (Fig. 2A). Similarly,

A B - + C FITCMDP Degradation by HRPE Lysate


200
RPE FITCMDP
180
FITCMDP
160
140
120
GSU

100
80
60
40
20
0
1

100
111
122
133
144
155
166
12
23
34
45
56
67
78
89

Fraction Number

Fig. 1 (A) Photomicrograph showing the PGRP2 cell lysate (60 μg). (C) Densitometric scan showing the
(NAMAA) in RPE cells. (B) Gel electrophoretic pattern >75% signal shift and the detection of FITCMurNAc
of FITCMDP alone (−) and incubated for 24 h with (+) RPE (arrows) after 24 h incubation
524 M. P. Langford et al.

A. Dose-dependent cleavage B. Time-dependent cleavage


FITC
RPE (µg Protein) MDP
0 5 10 20 40 80 Ctl 0 1 2 3 4 5 6h

Fig. 2 NAMAA dose- and time-dependency. (A) Agarose dent degradation of FITCMDP by 30 μg RPE lysate protein.
gel showing the dependence of FITCMDP degradation on Note the increase in FITCMurNAc cleavage signal (arrow)
RPE lysate protein concentration (mixtures incubated for with increasing RPE lysate concentration and time
5 h at 37 °C). (B) Agarose gel showing the time-depen-

hourly incremental increases in the appearance of selectively inhibit the cytopathic activity of some
free FITCMurNAc were detected through 6 h after NOD1 and NOD2 agonists.
incubation of FITCMDP with RPE lysate consis-
tent with time-dependent degradation of FITCMDP
by RPE NAMAA (Fig. 2B). 4 Discussion

RPE cells were shown to express PGRP2 protein


3.3 RPE Lysate NAMAA Inhibition and cleaved FITCMDP consistent with NAMAA.
of PGN Cytopathy The cleavage of FITCMurNAc from FITCMDP by
RPE lysate occurred in a concentration and time-­
To determine if RPE lysate would inhibit the dependent manner. Unexpectedly, RPE lysate
RK13 cytopathy induced by NOD1 and NOD2 cleavage of FITCMDP did not inhibit its cyto-
specific PGN, RPE lysate or PBS was incubated pathic activity in RK13 cells characterized by
with NOD1 and NOD2 agonist PGNs for 72 h at executioner caspase-3 activation and polymeric
37 °C. The mixtures were applied to cultures of DNA fragmentation. This result can be explained
RK13 cells and incubated until cell death was if NOD2 is activated by the cleaved l-alanyl-d-­
noted in agonist/PBS control. The levels of cas- isoglutamine moiety. Moreover, we observed
pase-­3 activity (Fig. 3A), polymeric DNA lad- that RPE lysate inhibited the cytopathy of most
ders profiles (Fig. 3B), and DNA densitometry NOD1 agonists and some NOD2 agonists. The
(Fig. 3C) were determined. The results suggest results suggest RPE cells express an antibacte-
the cytopathic activity of some NOD1 and NOD2 rial enzymatic activity that selectively cleaves
agonists was inhibited by RPE lysate. Note that PGN NOD agonists. In this regard, Saha et al.
iE-Lys and dd-MDP (inactive NOD1 and NOD2 [13] reported that PGLYRP-2 and NOD2 are
agonists, respectively) did not induce caspase-3 both required for PGN-induced arthritis and
or result in polymeric DNA fragmentation or local inflammation. In addition, PGRP2-deficient
increase DNA ladder density. RPE lysate selec- mice challenged with Pseudomonas aeruginosa
tively inhibited meso-diaminopimelic acid exhibit less severe keratitis and lower induction
NOD1 agonists (C12-iE-DAP, TriDAP, and of pro-inflammatory cytokines than wild-type
MTriDAP) and NOD2 agonists (FITCMDP, L18, mice [14]. Taken together, our in vitro results
M-TriLys, and MBT) induced caspase-3 activa- and these in vivo studies suggest that some
tion (Fig. 3A) as well as DNA fragmentation PGRP2 cleaved PGNs may be inactivated (i.e.,
(Fig. 3B, C ); consistent with endonuclease acti- unable to activate NOD) by RPE NAMAA,
vation. Thus, RPE lysate NAMAA appears to while others may retain cytopathogenic/pro-
Inhibition of Bacterial Peptidoglycan Cytopathy by Retina Pigment Epithelial PGRP2 Amidase 525

A
Caspase-3 Activity
160
140 Control *
(Units/ml) 120
100 *
HRPE *
80 *
60
40
*
20
0

NOD1/2 Agonist NOD2 Agonists

B Control HRPE Lysate Control HRPE Lysate


C12-iE-DAP

C12-iE-DAP

MTriDAP
MTriDAP

DD,MDP

DD,MDP

Dx-MDP
Dx-MDP
MTriLys

MTriLys
iE-DAP

iE-DAP
TriDAP

TriDAP

PGNEc
PGNEc

PGNSa

PGNSa
iE-Lys
iE-Lys

MDP

MBT

MDP

MBT
MFT

MFT
L18

L18
C 70 NOD1 Agonists NOD2 Agonists
Control 70
60 * Control
DNA Density (GSU)

HRPE 60 HRPE
DNA Density (GSU)

50
*
50 *
40 * 40
30 30
* *
20 20
10 10
0 0

meso-Diaminopimelic Acid L-alanyl-D-isoglutamine

Fig. 3 Effect of RPE lysate on inhibition of (A) caspase- and NOD1/2 agonists. *p-values <0.05; significantly dif-
­3 activity, (B) DNA fragmentation, and (C) DNA density ferent from stimulated control
induced in RK13 cell cultures treated with NOD1, NOD2,

inflammatory activity. The selective resistance of References


PGN to RPE NAMAA may play a role in bacte-
ria-associated retinopathy. 1. Vayalambrone D, Ivanova T, Misra A. Atypical cen-
tral serous retinopathy in a patient with latent tuber-
Acknowledgments The authors thank Christopher culosis. BMJ Case Rep. 2012:bcr1120115231. https://
Duggan for his technical assistance and gratefully doi.org/10.1136/bcr.11.2011.5231.
acknowledge the support of Medical Student Research 2. Khan P, Khan L, Anjum N, et al. Central serous cho-
Program (L.A.P.). rioretinopathy secondary to tuberculosis: cause or
526 M. P. Langford et al.

coincidence. BMJ Case Rep. 2017:bcr2016216471. dation with human N-acetylmuramyl-L-alanine ami-
https://doi.org/10.1136/bcr-­2016-­216471. dase. Eur Cytokine Netw. 1997;8:375–82.
3. Kavanaugh AS, Shaukat BA, Pan JT, et al. Bilateral 9. Wang ZM, Li X, Cocklin RR, et al. Human peptido-
chronic central serous chorioretinopathy associated glycan recognition protein-L is an N-acetylmuramoyl-­
with IGRA latent tuberculosis infection: a case report l-alanine amidase. J Biol Chem. 2003;278:49044–52.
and review. New Front Ophthalmol. 2020;6:1. https:// 10. Langford MP, Foreman BD, Srur L, et al. Bilateral
doi.org/10.15761/NFO.1000254. acute pyogenic conjunctivitis with iritis induced by
4. Zavoloka O, Bezditko P, Lahorzhevska I, et al. Clinical unilateral topical application of bacterial peptidogly-
efficiency of helicobacter pylori eradication in the can muramyl dipeptide in adult rabbits. Exp Eye Res.
treatment of patients with acute central serous cho- 2013;116:324–36.
rioretinopathy. Graefes Arch Clin Exp Ophthalmol. 11. Langford MP, Chen D, Welbourne TC, et al. Stereo-­
2016;254:1737–42. isomer specific induction of renal cell apoptosis by
5. Sorbara MT, Philpott DJ. Peptidoglycan: a criti- synthetic muramyl dipeptide (N-acetylmuramyl-­
cal activator of the mammalian immune system l-alanyl-d-isoglutamine). Mol Cell Biochem.
during infection and homeostasis. Immunol Rev. 2002;236:63–73.
2011;243:40–60. 12. Langford MP, McGee DJ, Ta KH, et al. Multiple
6. Laroui H, Yan Y, Narui Y, et al. l-ala-γ-d-Glu-­­ caspases mediate acute renal cell apoptosis induced
meso-diaminopimelic acid (DAP) interacts directly by bacterial cell wall components. Ren Fail.
with leucine-rich region domain of nucleotide-­ 2011;33:192–206.
binding oligomerization domain 1, increasing phos- 13. Saha S, Qi J, Wang S, et al. PGLYRP-2 and Nod2
phorylation activity of receptor-interacting serine/ are both required for peptidoglycan-induced arthri-
threonine-protein kinase 2 and its interaction with tis and local inflammation. Cell Host Microbe.
nucleotide-binding oligomerization domain 1. J Biol 2009;5:137–50.
Chem. 2011;286:31003–13. 14. Gowda RN, Redfern R, Frikeche J, et al. Functions
7. Inohara N, Ogura Y, Fontalba A, et al. Host recogni- of peptidoglycan recognition proteins (Pglyrps) at
tion of bacterial muramyl dipeptide mediated through the ocular surface: bacterial keratitis in gene-targeted
NOD2. Implications for Crohn’s disease. J Biol mice deficient in Pglyrp-2, −3 and −4. PLoS One.
Chem. 2003;278:5509–12. 2015;10(9):e0137129. https://doi.org/10.1371/jour-
8. Hoijer MA, Melief MJ, Debets R, et al. Inflammatory nal.pone.0137129.
properties of peptidoglycan are decreased after degra-
Understanding Ischemic
Retinopathies: The Role
of Succinate and Its Receptor
in Retinal Pigment Epithelium

Bilge Esin Ozturk

Abstract both normal retinal development and prolifer-


ative ischemic retinopathies.
Retinopathy is the general name for all condi-
tion of the eyes in which blood vessels that Keywords
supply oxygen to the retina are damaged.
Ischemic retinopathies · Succinate · G
These include diabetic retinopathy, retinopa-
protein-coupled receptor GPR91 · Retinal
thy of prematurity, hypertensive retinopathy,
pigment epithelium · Hypoxia
and arteriosclerotic retinopathy. Although the
initial trigger that leads to insufficient perfu-
sion of the retina may be different, once a
critical level of ischemia is achieved, all types
1 Introduction
of retinopathies seem to follow a common
sequence—oxidative stress, followed by
1.1 Role of the Retinal Pigment
hypoxia-induced formation of morphologi-
Epithelium (RPE) in the Eye
cally abnormal vessels. This preretinal vascu-
lar growth is the most severe aspect of the
The retinal pigment epithelium is a monolayer of
retinopathy, as the outcome is often retinal
pigment cells that form a part of the blood/retina
detachment and eventually blindness.
barrier. On the apical side, RPE faces the photo-
Regardless of which therapy strategy is fol-
receptor outer segments, consisting of apical
lowed, a deeper understanding of both normal
microvilli that surround the outer segments and
retinal growth and the underlying molecular
establish a structural complex, and on the baso-
mechanisms of retinopathies is needed in
lateral side RPE faces the Burch’s membrane,
order to come up with more effective thera-
which separates RPE from fenestrated epithelium
pies. This chapter focuses on the citric acid
of the choriocapillaris.
cycle intermediate succinate and its G protein-­
As a layer of pigmented cells, one of the main
coupled receptor SUCNR1 in ischemic reti-
functions of the RPE is to absorb the light energy
nopathies, which were identified as potent
focused by the lens on the retina [1, 2]. The RPE
mediators of vessel growth in the settings of
also facilitates the transport of various molecules
and products between the subretinal space and
the blood. This includes transport of ions, water,
B. E. Ozturk (*) and metabolic end products from the subretinal
Department of Ophthalmology, University of
Pittsburgh, Pittsburgh, PA, USA space to the blood [3, 4]. The RPE takes up nutri-
e-mail: beo20@pitt.edu ents such as glucose, retinol, and fatty acids from

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 527
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_77
528 B. E. Ozturk

the blood and delivers these nutrients to photore- guided by the same family of factors in the body.
ceptors. In addition, the RPE is able to secrete a One of the most important of these factors is the
variety of growth factors helping to maintain the class 3 semaphorins (Sema3). Sema3 has been
structural integrity of choriocapillaris found to regulate several cellular processes
­endothelium and photoreceptors. The secretory including neuronal guidance, endothelial and
activity of the RPE plays an important role in tumor cell survival, apoptosis, and migration [7].
establishing the immune privilege of the eye by Other growth factors that take part in neovas-
secreting immunosuppressive factors. With these cularization of the retina include fibroblast
complex different functions, the RPE is essential growth factor (FGF), hepatocyte growth factor
for visual function. A failure of any one of these (HGF), platelet-derived growth factor (PDGF),
functions can lead to degeneration of the retina, and erythropoietin (EPO) [8]. These findings
loss of visual function, and blindness. together suggested a more collaborative orches-
tration of disease progression where several fac-
tors are interrelated with each other. Therefore,
1.2 Molecular Pathways Involved understanding normal retinal vascular growth
in the Regulation of and understanding the events that lead to the ini-
Vascularization in the Retina tial vascular loss and the subsequent events that
aim to restore the oxygen homeostasis is critical
There are a number of important factors that play for developing new therapeutic approaches to
a role in the progression of oxidative stress-­ cure, or prevent several disorders including dia-
related retinopathies. These factors include vas- betic retinopathy, retinopathy of prematurity,
cular endothelial growth factor (VEGF), hypertensive retinopathy, and age-related macu-
insulin-like growth factor (IGF-1), insulin-like lar degeneration (AMD).
growth factor-binding protein 3 (IGFBP-3),
nitro-oxidative stress, trans-arachidonic acids
(TAAs), semaphorin 3 molecules, Omega-3 lip- 2 Succinate and Its Receptor
ids acting as modulators, and succinate signaling SUCNR1
via its succinate receptor SUCNR1 acting in
neuron-­mediated retinal neovascularization. The retina has high content of polyunsaturated
IGF-1 influences angiogenesis and the devel- fatty acids (PUFA) and has the highest oxygen
opment of retinal neovascularization through uptake and glucose oxidation relative to any other
interaction with locally produced factors such as tissue. This phenomenon renders retina more sus-
VEGF, acting as a permissive factor for maxi- ceptible to oxidative stress. Hypoxia is a well-­
mum VEGF stimulation of angiogenesis [5]. It established modulator of retinal vascularization.
was also demonstrated that low IGF-1 levels in The role of hypoxia inducible factor (HIF) and
serum is associated with retinopathy of prema- the hypoxia response genes including VEGF has
turity (ROP), which is associated with poor ini- been extensively studied in retinopathies.
tial vascular development, indicating that IGF-1 Although hypoxia triggered events have been
might be involved in human angiogenesis [5]. classically considered to depend on pathways
IGF-1, together with basic fibroblast growth involving HIF and VEGF, hypoxia can itself cre-
factor (bFGF), and other growth factors have ate a change in metabolites. Thus, exploring the
also been shown to stimulate DNA synthesis major intermediary energy metabolites is one of
and RPE cell proliferation, and are part of the the several approaches to elucidate the mecha-
repertoire of RPE and neuronal responses to nisms involved in regulating the expression of the
injury [6]. factors mentioned above. Recent studies have
It has been long conceived that vascular and revealed a role for succinate and its receptor
neural networks share architectural commonali- SUCNR1, in vascularization that occurs during
ties and interact physically in the body, and it has development and ischemia, both of which are
been realized that blood vessels and neurons are triggered by tissue hypoxia [9].
Understanding Ischemic Retinopathies: The Role of Succinate and Its Receptor in Retinal Pigment… 529

Succinate induces a vaso-proliferative faces choroidal blood. Apical expression of


response in the retina by activating its G protein- SUCNR1 therefore suggests that the receptor is
coupled receptor, SUCNR1 which was initially activated by succinate present in the retinal space.
classified as GPR91 [9]. Succinate is normally Activation of SUCNR1 by succinate results in the
present in the mitochondria as part of the tricar- production and release of several proangiogenic
boxylic acid (Krebs) cycle to produce adenosine molecules, including VEGF, angiopoietin-1, and
triphosphate (ATP). However, differences or angiopoietin-2. Levels of succinate increase sig-
imbalances between the energy state of cells and nificantly in the retina after ischemic events. Even
the oxygen and nutrient supply result in the accu- though the levels of the receptor do not change,
mulation of succinate in the mitochondria and the increase in the agonist levels ensures that the
subsequent translocation from the mitochondria downstream pathway is activated, leading to the
to the cytosol, and the extracellular environment. secretion of proangiogenic factors, and ultimately,
Succinate is increasingly produced and released promotion of neovascularization. This makes
into the extracellular environment during hypoxic SUCNR1 a potentially interesting drug candidate
conditions, most likely due to mitochondrial for the treatment of ischemic retinopathies.
adaptations to intracellular oxidative stress [10,
11]. The conditions that disrupt the energy and
supply state of the cell include hypoxia, diabetes, 3 Conclusion, Therapies,
and solid-tumor cancers [12]. and Prevention
Oxidative stress is known to trigger the release
of the citric acid cycle intermediate succinate in a Although ischemic retinopathies come in multi-
multitude of disorders [13]. The accumulation of ple types and have different origins, their tissue,
succinate due to increased reactive oxygen spe- cellular, and molecular determinants show clear
cies is an indicator of mitochondrial stress, but the similarities. Elucidation of these determinants
effect of succinate has been largely overlooked up may allow the development of novel preventive
to date. However, early studies have successfully therapies or pharmacological cures.
demonstrated that even after short episodes of Current prevention methods include ablative
hypoxia, tissue succinate sharply increases in therapies that destroy tissue in the avascular area
many organs while levels of other tricarboxylic of the retina, which also includes cells that gener-
acid intermediates fall [14]. Recently, SUCNR1 ate VEGF, which promotes aberrant retinal neo-
was shown to promote neovascularization of the vascularization. Despite the therapy, however,
retina in diabetes mellitus, a disease associated visual acuity remains unaffected and peripheral
with oxidative stress, through VEGF [9], which is vision is inevitably lost [2]. Anti-­VEGF therapy
also crucial for neovascularization in AMD [15]. is another strategy to counter ischemic retinopa-
So far, several studies have linked increased suc- thies. In this therapy, a VEGF-specific neutraliz-
cinate levels and consequent SUCNR1 activation ing antibody is used to attenuate VEGF signaling,
with various diseases ranging from diabetes, thereby reducing the recurrence rate of preretinal
hypertension [11], kidney diseases [11, 16], can- neovascularization.
cer, and retinopathies as well [17]. Development of preventive and less-­
The expression of SUCNR1 has been demon- destructive therapies is needed to better deal with
strated in highly vascularized tissues, retinal gan- retinopathies. These therapies should target the
glion cells (RGC) and RPE cells [9, 17]. More initial stages of the disease, and can include sev-
importantly, the receptor was found to be eral strategies such as restricting tissue oxygen-
expressed only in the apical membrane of RPE ation, nutritional supplements that decrease lipid
cells, and not in the basolateral membrane. This peroxidation, administration of EPO and/or
exclusive cellular localization is functionally IGF-­ 1, and the use of Omega-3 fatty acids.
important because the apical membrane faces the Exploring the mechanisms underlying the natural
neural retina, whereas the basolateral membrane vascular growth and retinopathies is required in
530 B. E. Ozturk

order to develop new and effective preventive 2. Chen J, Lois AE, Smith EH. Retinopathy of
prematurity.
therapies. 3. Hamann S. Molecular mechanisms of water transport
Recent studies identified several molecular in the eye. Int Rev Cytol. 2002;215.
mechanisms that contribute to the development of 4. Marmor MF, Tan F. Central serous chorioretinopathy:
ischemic retinopathies. New treatment methods are bilateral multifocal electroretinographic abnormali-
ties. Arch Ophthalmol. 1999;117(2):184–8.
emerging, and these include therapies directed 5. Hellstrom A, Perruzzi C, Ju M, Engström E, Hård
against VEGF. However, a better understanding of A-L, Liu J-L, et al. Low IGF-I suppresses VEGF-­
how oxygen-induced retinopathies progress survival signaling in retinal endothelial cells: direct
through time, and a much clearer picture of how all correlation with clinical retinopathy of prematurity.
Proc Natl Acad Sci. 2001;98(10):5804–8.
the underlying factors are interrelated during the 6. Cao W, Li F, Steinberg RH, LaVail MM. Development
course of retinopathies is required before effective of normal and injury-induced gene expression of
therapies can be developed. aFGF, bFGF, CNTF, BDNF, GFAP and IGF-I in the
Vascularization is an important process for tis- rat retina. Exp Eye Res. 2001;72(5):591–604.
7. Klagsbrun M, Eichmann A. A role for axon guidance
sue development and in restoration of damaged receptors and ligands in blood vessel development
tissues after ischemic injuries. Until recently, the and tumor angiogenesis. Cytokine Growth Factor
main focus of many studies about vascularization Rev. 2005;16(4–5):535–48.
was the direct effects of angiogenic growth fac- 8. Gariano RF, Gardner TW. Retinal angiogenesis in
development and disease. Nature. 2005;438:7070.
tors. Although the central role of VEGF has long Available from: https://www.nature.com/articles/
been recognized in ischemic retinopathies, recent nature04482
research points to an additive effect of a collec- 9. Sapieha P, Sirinyan M, Hamel D, Zaniolo K, Joyal
tion of factors, both angiogenic (VEGF, IGF, and J-S, Cho J-H, et al. The succinate receptor GPR91 in
neurons has a major role in retinal angiogenesis. Nat
EPO) and anti-angiogenic (TSP-1) in nature. Med. 2008;14(10):1067–76. Available from: https://
Moreover, other nutritional factors (PUFA) and www.nature.com/articles/nm.1873.
oxidative and nitro-oxidative stress-dependent 10. Weinberg JM, Venkatachalam MA, Roeser NF, Nissim
mediators (TAA) seem to be interrelated with the I. Mitochondrial dysfunction during hypoxia/reoxy-
genation and its correction by anaerobic metabolism
factors stated above. The gaps and bridges of citric acid cycle intermediates. Proc Natl Acad Sci.
between these factors and pathways are only 2000;97(6):2826–31. Available from: https://www.
recently being identified. pnas.org/content/97/6/2826.
Hypoxia-driven succinate and its receptor 11. Toma I, Kang JJ, Sipos A, Vargas S, Bansal E,
Hanner F, et al. Succinate receptor GPR91 provides
SUCNR1 is one such factor that acts as a potent a direct link between high glucose levels and renin
mediator in retinal angiogenesis, and provides release in murine and rabbit kidney. J Clin Investig.
one of the missing bridges between a hypoxic 2008;118(7):2526–34. Available from: http://www.
retina and the following vascularization pro- jci.org.
12. Hebert SC. Orphan detectors of metabolism. Nature.
cesses. The recent studies on succinate and its 2004;429(6988):143–5. Available from: https://www.
receptor identified new cellular mechanisms and nature.com/articles/429143a.
signaling pathways that may be very important 13. Deen PMT, Robben JH. Succinate receptors in
in the progression of many diseases in different the kidney. J Am Soc Nephrol. 2011;22(8):1416–
22. Available from: https://jasn.asnjournals.org/
organs. Future research on this topic may pro- content/22/8/1416.
vide important information that is clinically 14. Krebs HA. The history of the tricarboxylic acid
applicable to develop new drugs and diagnostic cycle. Perspect Biol Med. 1970;14(1):154–72.
approaches. Available from: https://muse.jhu.edu/article/405199/
summary.
15. Khandhadia S, Lotery A. Oxidation and age-related
macular degeneration: insights from molecular biol-
References ogy. Expert Rev Mol Med. 2010;12:e34. Available
from: https://www.cambridge.org/core/journals/
1. Bok D. The retinal pigment epithelium: a versatile expert-­r eviews-­i n-­m olecular-­m edicine/article/
partner in vision. J Cell Sci. 1993;1993:189. oxidation-­a nd-­a gerelated-­m acular-­d egeneration-­
Understanding Ischemic Retinopathies: The Role of Succinate and Its Receptor in Retinal Pigment… 531

insights-­from-­molecular-­biology/1976C60A7D37A4 17. Gnana-Prakasam JP, Ananth S, Prasad PD, Zhang M,


C2AB0C91EE43779D0C. Atherton SS, Martin PM, et al. Expression and iron-­
16. Ariza AC, Deen PMT, Robben JH. The succinate dependent regulation of succinate receptor GPR91 in
receptor as a novel therapeutic target for oxidative and retinal pigment epithelium. Invest Ophthalmol Vis
metabolic stress-related conditions. Front Endocrinol. Sci. 2011;52(6):3751–8.
2012;0(FEB):22.
The Amphipathic Helix in Visual
Cycle Proteins: A Review

Sheetal Uppal, Eugenia Poliakov,


Susan Gentleman, and T. Michael Redmond

Abstract 1 Introduction

The visual cycle is a complex biological pro- Retinol and its derivatives, collectively termed
cess that involves the sequential action of pro- retinoids, are highly hydrophobic molecules and
teins in the retinal pigment epithelial (RPE) are shielded by several proteins during the visual
cells and photoreceptors to modify and shuttle cycle process so that they can diffuse freely
visual retinoids. A majority of the visual cycle between the photoreceptor and retinal pigment
proteins are membrane proteins, either inte- epithelial (RPE) cells [32]. The RPE cells play a
gral or peripheral membrane proteins. Despite crucial role in the regeneration of the visual chro-
significant progress in understanding their mophore of rhodopsin, 11-cis retinal, thus pro-
physiological function, very limited structural viding a continuous supply to the photoreceptors
information is available for the visual cycle [31]. In the RPE, the visual cycle proteins
proteins. Moreover, the mechanism of mem- (RPE65, LRAT, CRALBP, and RDHs) are
brane interaction is not yet clear in all cases. membrane-­associated to perform their enzymatic
Here, we demonstrate the presence of an function, i.e., production and transport of 11-cis
amphipathic helix in selected RPE visual retinal to photoreceptors [15, 17, 29, 33]. Recent
cycle proteins, using in silico tools, and high- progress in the field of structural biology and
light their role in membrane association and improvements in protein crystallization methods
function. have led to the structural determination of several
visual system proteins; however, the structures of
Keywords all the RPE visual cycle components has not been
determined, which impedes our understanding of
Visual cycle proteins · Membrane association · their underlying mechanisms of membrane
Amphipathic helix · Alpha (α)-helix · association.
Retinoids · RPE65 · LRAT · RDH5 · CRALBP Several reports have highlighted the role of
amphipathic helices as a membrane recognition
and binding motif in peripheral membrane pro-
teins [2, 6, 18]. By definition, amphipathic heli-
S. Uppal · E. Poliakov · S. Gentleman ·
T. M. Redmond (*) ces are secondary structural motifs that have
Laboratory of Retinal Cell and Molecular Biology, hydrophobic and hydrophilic amino acid residues
National Eye Institute, National Institutes of Health, located on the opposite faces of the α-helix.
Bethesda, MD, USA Diverse structural and functional roles have been
e-mail: redmondd@helix.nih.gov

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 533
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_78
534 S. Uppal et al.

identified for membrane-interacting amphipathic 3 Results


helices, and there are several bioinformatic tools
available, such as the HELIQUEST and 3D-HM 3.1 Retinal Pigment Epithelium-­
servers, which can predict the amphipathic seg- Specific 65 kDa (RPE65)
ments in a protein sequence and calculate the Protein
magnitude and orientation of the hydrophobic
moment and charge of the helices [5, 25]. Here, RPE65 is a non-heme iron-containing metallo-
we have examined the RPE-specific visual cycle protein, belonging to the carotenoid cleavage
proteins using in silico tools to predict the puta- dioxygenase (CCD) superfamily [14]. It is a reti-
tive amphipathic α-helix-forming sequences, noid isomerase that carries out the important
including a comparative analysis of their trans–cis isomerization of retinol step in RPE
amphipathic character, and structural visualiza- cells [12, 20, 23, 24]. Despite the absence of any
tions of the predicted amphipathic helices. This hydrophobic transmembrane segments, RPE65
study will provide a better understanding of the associates with the smooth endoplasmic reticu-
role of amphipathic α-helices in the mechanisms lum (sER) membrane to extract the stored all-­
of membrane binding of the different RPE-­ trans retinyl palmitate and produce 11-cis retinol
specific visual cycle proteins. [10, 21]. Understanding the mechanism of
RPE65-membrane binding is of utmost important
in the vision field for the development of small-­
2 Materials and Methods molecule therapeutic compounds to treat RPE65-­
associated retinal dystrophies. Several reports
2.1 Amino Acid Sequence have suggested a role for the crystallographically
Retrieval and Structure unresolved region, comprising of amino acid res-
Visualization idues (aa)108–125 together with the palmi-
toylation of the cysteine residue at the 112th
Sequences of RPE-specific proteins (RPE65, position, as being crucial to membrane associa-
LRAT, CRALBP, and RDHs) were collected tion of RPE65 protein [14, 16, 34, 35]. Our in
from NCBI (http://www.ncbi.nlm.nih.gov/). silico analysis reveals that RPE65 is a soluble
protein and that the crystallographically missing
region can form an amphipathic α-helix with the
2.2 Prediction for Amphipathic C112 residue located in the hydrophobic face,
α-Helix Segments supporting previous observations (Fig. 1). The
in the Protein predicted amphipathic α-helix of RPE65 is highly
hydrophobic in nature with a high hydrophobic
Both the HELIQUEST (https://heliquest.ipmc. moment above 0.6, indicating its strong amphipa-
cnrs.fr/) and EMBOSS:hmoment (https://www. thic character (Table 1).
bioinformatics.nl/cgi-­bin/emboss/hmoment) pro-
grams were employed for prediction and analysis
of segments with a potential for amphipathic 3.2 Lecithin:Retinol
α-helix formation [3, 5]. For prediction, the sliding Acyltransferase
window size of 18 amino acid residues and angle
of rotation of 100° (for α-helix) were used. The Lecithin:retinol acyltransferase (LRAT) is a
sequence with the maximum hydrophobic moment vertebrate-­specific member of N1pc/P60
(μH) was reported for the proteins. Cecropin A, papain-­like thiol protease protein superfamily
Cecropin B, and Melittin were used as a reference localized in the ER membranes of RPE cells [7].
for surface proteins [28]. Rhodopsin, containing It is another crucial enzyme in the visual cycle
seven transmembrane α-helices, was used as a ref- and catalyzes the esterification of the all-trans
erence for transmembrane helices [22]. retinol to produce all-trans retinyl esters, mainly
The Amphipathic Helix in Visual Cycle Proteins: A Review 535

Fig. 1 (a) Helical wheel


representations of AH
sequences of RPE-
specific visual cycle
proteins (RPE65, LRAT,
CRALBP, RDH5,
RDH10, and RDH11).
The numbers in
superscripts correspond
to the residue positions
of the predicted AH
motif. (b) Hydrophobic
moment plot for the
predicted AHs where
each point represents an
18-residue α-helix

all-­trans retinyl palmitate, the substrate for proper positioning of the transmembrane
RPE65. Biochemical studies show that it is a domain (aa196–215) of LRAT.
single-pass integral membrane protein with a
C-terminal luminal orientation [19]. The com-
plete three-­dimensional structure of LRAT pro- 3.3 Cellular Retinaldehyde-­
tein has not been solved; however, the structure Binding Protein
of a LRAT/HRASLS3 chimeric protein contain-
ing the unique 30 amino acids segment of LRAT CRALBP is a soluble 36 kDa retinoid-binding
is available, which provides key structural infor- protein highly expressed in RPE and Muller glial
mation into the function of LRAT. However, the cells of the retina [1, 26]. The three-dimensional
chimeric protein structure does not contain the structure of CRALBP and its interaction with the
transmembrane C-terminal helical region, ligands has been determined by NMR and X-ray
which is proposed to play an important role in crystallography [11, 36]. It has a higher binding
substrate recognition in the phospholipid mem- affinity for 11-cis retinoids compared to RPE65,
brane [7]. We predict that aa179–196 sequence thus allowing the dissociation of 11-cis retinol
in the LRAT protein forms an amphipathic from RPE65 and facilitating its oxidation to 11-cis
α-helix (Fig. 1 and Table 1). This amphipathic retinal by retinol dehydrogenases (RDHs)
region is a part of cytoplasmic-facing topologi- expressed in RPE. Biochemical and structural
cal domain of LRAT, which might facilitate the studies revealed that interaction of the holo-­
536 S. Uppal et al.

Table 1 Comparison of the properties of predicted AH motif of RPE-specific visual cycle proteins
No. of Net
amino Residue Hydrophobic Hydrophobicity charge % non-polar % polar
Protein acids positions moment (μH) (H) (z) residues residues
RPE65 533 108–125 0.664 0.568 2 55.56 44.44
LRAT 230 179–196 0.515 0.372 1 55.56 44.44
CRALBP 317 173–190 0.555 0.683 −3 61.11 38.89
RDH5 318 241–258 0.563 0.606 2 55.56 44.44
RDH10 341 71–88 0.427 0.113 −2 33.33 66.67
RDH11 318 301–318 0.465 0.671 −1 61.11 38.89

CRALBP (bound to 11-cis retinal) with the acidic role in the membrane association of these
phospholipids in the membrane bilayer triggers RDHs, similar to the situation in RDH8.
the release of 11-cis retinal and have identified a
cluster of basic amino acid residues that might
interact with the membrane [27]. Keeping this in 4 Discussion
mind, we examined the CRALBP sequence and
found that aa173–190, an α-helical sequence of Amphipathic α-helices have gained considerable
residues as determined from the crystal structure, attention recently due to their amphiphilic charac-
is predicted to be an amphipathic α-helix with high ter, behaving as a hidden motif in proteins, thus
hydrophobicity (H) value (Fig. 1 and Table 1). facilitating binding to membranes. These AHs do
not have any specific signature sequence and exhibit
a wide variation in amino acid composition and
3.4 Retinol Dehydrogenases length that provides different interfacial properties
and cellular function. As the RPE-­specific visual
Retinol dehydrogenases (RDHs) are NAD+- or cycle proteins require an interfacial function to
NADP+-dependent oxidoreductases that belong access their substrates directly from the lipid–water
to the short-chain dehydrogenases/reductases interface, this prompted us to analyze their
(SDRs) superfamily and contain the conserved sequences for AHs. Our results reveal that predicted
super secondary Rossman fold (βαβ) structure AHs in the RPE-specific visual cycle proteins have
[13, 17]. These enzymes are essential members hydrophobic moments similar to AHs from surface-
of the visual cycle in RPE where they catalyze active peptides and proteins such as the cecropins
the final step, i.e., oxidation of 11-cis retinol to and melittin. The seven transmembrane α-helices of
11-cis retinal. Three RDHs (RDH5, RDH10, rhodopsin protein have low hydrophobic moment
and RDH11) expressed in RPE cells exhibit and high hydrophobicity, which is a typical charac-
11-cis retinol dehydrogenase activity [4, 9, 30]. teristic for transmembrane AHs. Based on the avail-
To date, no structural information is available able literature for the surface-active AHs, we can
for these RDHs and it has been reported that conclude that the predicted AHs of RPE-specific
RDH activity is membrane-associated because visual cycle proteins are surface-exposed and may
of their high hydrophobic nature. A recent report be involved in the membrane binding of the RPE-
demonstrated that the C-terminus of photore- specific visual cycle proteins. We propose that this
ceptor RDH8 consists of an amphipathic study provides a hint toward the role and function of
α-helical structure that contributes to the mem- AHs in visual cycle proteins that should be investi-
brane binding of RDH8 protein [8]. Our in silico gated in further detail.
predictions revealed that all three RPE-
expressed RDHs contain amphipathic α-helices Acknowledgments This study is supported by the
(AHs) as shown in Fig. 1 and Table 1. We Intramural Research Program of the National Eye
Institute, NIH.
assume that these AHs might play an important
The Amphipathic Helix in Visual Cycle Proteins: A Review 537

References 18. Liu A, Sui D, Wu D, et al. The activation loop of


PIP5K functions as a membrane sensor essen-
tial for lipid substrate processing. Sci Adv.
1. Bunt-Milam AH, Saari JC. Immunocytochemical
2016;2:e1600925.
localization of two retinoid-binding proteins in verte-
19. Moise AR, Golczak M, Imanishi Y, et al. Topology
brate retina. J Cell Biol. 1983;97:703–12.
and membrane association of lecithin: retinol acyl-
2. Drin G, Antonny B. Amphipathic helices and mem-
transferase. J Biol Chem. 2007;282:2081–90.
brane curvature. FEBS Lett. 2010;584:1840–7.
20. Moiseyev G, Chen Y, Takahashi Y, et al. RPE65 is the
3. Eisenberg D, Weiss RM, Terwilliger TC. The hydro-
isomerohydrolase in the retinoid visual cycle. Proc
phobic moment detects periodicity in protein hydro-
Natl Acad Sci U S A. 2005;102:12413–8.
phobicity. Proc Natl Acad Sci U S A. 1984;81:140–4.
21. Nikolaeva O, Takahashi Y, Moiseyev G, et al. Purified
4. Farjo KM, Moiseyev G, Takahashi Y, et al. The
RPE65 shows isomerohydrolase activity after reas-
11-cis-retinol dehydrogenase activity of RDH10
sociation with a phospholipid membrane. FEBS J.
and its interaction with visual cycle proteins. Invest
2009;276:3020–30.
Ophthalmol Vis Sci. 2009;50:5089–97.
22. Palczewski K, Kumasaka T, Hori T, et al. Crystal
5. Gautier R, Douguet D, Antonny B, et al. HELIQUEST:
structure of rhodopsin: a G protein-coupled receptor.
a web server to screen sequences with specific alpha-­
Science. 2000;289:739–45.
helical properties. Bioinformatics. 2008;24:2101–2.
23. Redmond TM, Yu S, Lee E, et al. Rpe65 is necessary
6. Gimenez-Andres M, Copic A, Antonny B. The many
for production of 11-cis-vitamin a in the retinal visual
faces of amphipathic helices. Biomol Ther. 2018;8.
cycle. Nat Genet. 1998;20:344–51.
7. Golczak M, Sears AE, Kiser PD, et al. LRAT-­
24. Redmond TM, Poliakov E, Yu S, et al. Mutation of
specific domain facilitates vitamin A metabolism
key residues of RPE65 abolishes its enzymatic role as
by domain swapping in HRASLS3. Nat Chem Biol.
isomerohydrolase in the visual cycle. Proc Natl Acad
2015;11:26–32.
Sci U S A. 2005;102:13658–63.
8. Hadicke A, Coutinho A, Roy S, et al. Membrane bind-
25. Reisser S, Strandberg E, Steinbrecher T, et al. 3D
ing properties of the C-terminal segment of retinol
hydrophobic moment vectors as a tool to characterize
dehydrogenase 8. Biochim Biophys Acta Biomembr.
the surface polarity of amphiphilic peptides. Biophys
2021;1863:183605.
J. 2014;106:2385–94.
9. Haeseleer F, Jang GF, Imanishi Y, et al. Dual-
26. Saari JC. Vitamin A metabolism in rod and cone
substrate specificity short chain retinol dehydro-
visual cycles. Annu Rev Nutr. 2012;32:125–45.
genases from the vertebrate retina. J Biol Chem.
27. Saari JC, Nawrot M, Stenkamp RE, et al. Release
2002;277:45537–46.
of 11-cis-retinal from cellular retinaldehyde-binding
10. Hamel CP, Tsilou E, Pfeffer BA, et al. Molecular
protein by acidic lipids. Mol Vis. 2009;15:844–54.
cloning and expression of RPE65, a novel retinal pig-
28. Sato H, Feix JB. Peptide-membrane interactions and
ment epithelium-specific microsomal protein that is
mechanisms of membrane destruction by amphipa-
post-transcriptionally regulated in vitro. J Biol Chem.
thic alpha-helical antimicrobial peptides. Biochim
1993;268:15751–7.
Biophys Acta. 2006;1758:1245–56.
11. He X, Lobsiger J, Stocker A. Bothnia dystrophy is
29. Sears AE, Palczewski K. Lecithin:retinol acyltrans-
caused by domino-like rearrangements in cellular
ferase: a key enzyme involved in the retinoid (visual)
retinaldehyde-­binding protein mutant R234W. Proc
cycle. Biochemistry. 2016;55:3082–91.
Natl Acad Sci U S A. 2009;106:18545–50.
30. Simon A, Hellman U, Wernstedt C, et al. The retinal
12. Jin M, Li S, Moghrabi WN, et al. Rpe65 is the retinoid
pigment epithelial-specific 11-cis retinol dehydro-
isomerase in bovine retinal pigment epithelium. Cell.
genase belongs to the family of short chain alcohol
2005;122:449–59.
dehydrogenases. J Biol Chem. 1995;270:1107–12.
13. Jornvall H, Persson B, Krook M, et al. Short-chain
31. Strauss O. The retinal pigment epithelium in visual
dehydrogenases/reductases (SDR). Biochemistry.
function. Physiol Rev. 2005;85:845–81.
1995;34:6003–13.
32. Szuts EZ, Harosi FI. Solubility of retinoids in water.
14. Kiser PD, Golczak M, Lodowski DT, et al. Crystal
Arch Biochem Biophys. 1991;287:297–304.
structure of native RPE65, the retinoid isomer-
33. Tsilou E, Hamel CP, Yu S, et al. RPE65, the major
ase of the visual cycle. Proc Natl Acad Sci U S A.
retinal pigment epithelium microsomal membrane
2009;106:17325–30.
protein, associates with phospholipid liposomes. Arch
15. Kiser PD, Golczak M, Maeda A, et al. Key enzymes
Biochem Biophys. 1997;346:21–7.
of the retinoid (visual) cycle in vertebrate retina.
34. Uppal S, Poliakov E, Gentleman S, et al. RPE65 pal-
Biochim Biophys Acta. 2012a;1821:137–51.
mitoylation: a tale of lipid posttranslational modifica-
16. Kiser PD, Farquhar ER, Shi W, et al. Structure of
tion. Adv Exp Med Biol. 2019a;1185:537–41.
RPE65 isomerase in a lipidic matrix reveals roles for
35. Uppal S, Liu T, Poliakov E, et al. The dual roles of
phospholipids and iron in catalysis. Proc Natl Acad
RPE65 S-palmitoylation in membrane association
Sci U S A. 2012b;109:E2747–56.
and visual cycle function. Sci Rep. 2019b;9:5218.
17. Lhor M, Salesse C. Retinol dehydrogenases:
36. Wu Z, Yang Y, Shaw N, et al. Mapping the ligand
membrane-bound enzymes for the visual function.
binding pocket in the cellular retinaldehyde binding
Biochem Cell Biol. 2014;92:510–23.
protein. J Biol Chem. 2003;278:12390–6.
The Retinal Pigment Epithelium:
Cells That Know the Beat!

Elora M. Vanoni and Emeline F. Nandrot

Abstract Keywords

Retinal pigment epithelium · Photoreceptor


The retinal pigment epithelium (RPE) ensures
outer segments · Phagocytosis · Retinal
different functions crucial for photoreceptor
adhesion · Metabolism · Circadian clock ·
survival, and thus for vision, such as photore-
Beta5 integrin · Prpf31 · Animal models
ceptor outer segments (POS) phagocytosis
and retinal adhesion. Both follow a circadian
rhythm with an activity peak occurring respec-
tively 1.5–2 and 3.5 h after light onset. 1 Introduction
Interestingly, we showed that two rodent mod-
els, β5−/− and Prpf31+/− mice, display distinct Photoreceptor outer segments (POS) are in close
alterations in both functions leading to differ- contact with the apical microvilli of the retinal
ent phenotypes. Indeed, the phagocytic peak pigment epithelium (RPE), a monolayer com-
totally disappears in β5 knockout mice but is posed of pigmented, bi-nucleated (most often),
attenuated and shifted in Prpf31+/− mice. and hexagonal-shaped cells in a post-mitotic
Conversely, the retinal adhesion peak only state [35]. Each RPE cell apical microvilli closely
attenuated in β5−/− mice is lost in Prpf31+/− interact with around 22–30 photoreceptors (PRs).
mice. These distinct alterations have different The basal side of this epithelium, constituted of
consequences on retinal homeostasis propor- membrane infoldings, faces the Bruch’s mem-
tional to the observed defects: β5−/− mice pro- brane and the choroid.
gressively lose vision and accumulate RPE RPE cells ensure crucial functions for PRs
lipofuscin deposits, while Prpf31+/− mice survival and thus for vision, such as light absorp-
develop RPE metabolic dysfunctions and tion by melanin pigments, bi-lateral ions, nutri-
gradual structural modifications indicative of ents and trophic factors transport/secretion, and
cellular stress. Hence, animal models are use- recycling of visual cycle molecules. Besides,
ful to understand the importance of the proper POS, which have very few detoxification sys-
regulation of these functions. tems, are constantly submitted to light photons
generating oxidative attacks [43]. To fight these,
POS membranous disks are permanently renewed
E. M. Vanoni · E. F. Nandrot (*) and most oxidized POS tips are eliminated daily
Therapeutics Department, Sorbonne Université, by phagocytosis by RPE cells, which are the bus-
INSERM, CNRS, Institut de la Vision, Paris, France iest and biggest phagocytes in the body [16, 44].
e-mail: emeline.nandrot@inserm.fr

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 539
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_79
540 E. M. Vanoni and E. F. Nandrot

It is important to note that 7–10 days are neces- [17]. MerTK also controls the number of POS
sary for full outer segment replacement. bound to αvβ5 integrin receptors that can be
Moreover, the RPE actively participates to retinal engulfed, possibly by the cleavage of its extracel-
adhesion, ensuring the tightness between RPE lular domain [17, 24]. Gas6 and Protein S,
apical microvilli and the full length of POS, required for MerTK dimerization, activate and
another daily function crucial for PRs survival inhibit phagocytosis, respectively, hence regulat-
[22]. Interestingly, recent studies using different ing MerTK activity [17].
mouse models have shown that the impact of the Moreover, RCS (Royal College of Surgeons)
perturbation of these two rhythmic functions can rats, bearing a genomic deletion encompassing
be very different, allowing us to better under- MerTK, display a total absence of POS phagocy-
stand how RPE cells regulate their activities. tosis [4]. The lack of MerTK causes severe reti-
nal dystrophies in rodents (retinitis pigmentosa)
as well as in humans (rod-cone dystrophies with
2 POS Phagocytosis an early macular implication), highlighting the
importance of POS phagocytosis for global reti-
POS continuously submitted to light photons nal health [1, 7, 9, 13, 20, 25, 40]. However, the
need to be renewed and their oxidized parts elim- absence of integrin receptors leads to a totally
inated. After their internalization, they are different phenotype. Indeed, β5−/− mice have
degraded and components are either recycled in been studied to evaluate the effect of αvβ5
the cell or returned to PRs. Thus, this function is absence on both retina and RPE, β5 only associ-
primordial for PR survival, retinal homeostasis, ating with αv subunits and αvβ5 being solely
and therefore for vision. In vivo, POS phagocyto- expressed at the RPE apical surface [11, 21]. The
sis has been described to be a circadian function absence of αvβ5 integrin receptors directly
with an activity peak 1.5–2 h after light onset for impacts phagocytosis with a 75% decrease in
rods (Fig. 1a) [16, 21]. Indeed, when albino rats RPE primary cultures (Fig. 1a). Besides, the
were placed in constant darkness, POS phagocy- phagocytic peak observed in wild-type (WT)
tosis stays synchronized with a slow progressive control mice, as well as associated FAK and
shift to a 25 h period, a rhythm typical of circa- MerTK phosphorylation, totally disappears in
dian activities [16]. Of note, POS phagocytosis is β5−/− mice and is replaced by a steady-state level
maintained when the retinohypothalamic tract is of activity (Fig. 1a).
severed, thus confirming the local control of the We also studied the phagocytic profile of
rhythmicity of this function [37]. In addition, the Prpf31+/− mice, a model developed to understand
role of the pineal gland has been excluded [36]. the pathogenesis of the second cause of autoso-
The phagocytic machinery is composed of mic dominant retinitis pigmentosa linked to
two main receptors: the αvβ5 integrin and mutations in the Pre-mRNA Processing Factor
MerTK, both located at the RPE apical mem- genes family, including PRPF31 (RP11) [6, 10,
brane. The αvβ5 integrin is associated with the 14]. The decrease in Prpf31 expression levels
extracellular ligand MFG-E8 and is implicated in leads to a 40% phagocytosis defect in RPE pri-
POS binding and phagocytosis synchronization mary cultures (Fig. 1b). Interestingly, a similar
[21, 23]. Its signaling pathways activate MerTK 40% decrease is observed in a shRNA-treated
by phosphorylation via the focal adhesion kinase human (ARPE-19) and rat (unpublished data)
(FAK), thus triggering POS engulfment. RPE cell lines, as well as in RPE cells derived
Conversely, MerTK and both its cognate from induced pluripotent stem cells (RPE-iPSC)
ligands —Gas6 and Protein S— are essential for from RP11 patients, thus confirming that analo-
POS internalization in the RPE [4, 7, 9, 20, 42]. gous mechanisms occur between species [10,
Previously, our team showed that MerTK expres- 27]. Interestingly, mutation correction using the
sion levels change over time in RPE cells and CRISPR-Cas9 technology restores a normal
increase after the phagocytic peak, a time during phagocytosis ratio similar to WT RPE-iPSC [27].
which MerTK receptors are used extensively However, in contrast to what is observed in β5−/−
The Retinal Pigment Epithelium: Cells That Know the Beat! 541

Fig. 1 RPE circadian dysfunctions and associated pheno- right panel) and retinal adhesion (bottom-left panel) were
types in two mouse models. (a) An ~75% phagocytosis disrupted in Prpf31+/− mice (white bars, MUT) when
decrease was detected in β5−/− RPE primary cultures (top-­ compared to wild-type (black bars, WT). RPE ultrastruc-
left panel). The phagocytic peak occurs 2 h after light ture abnormalities were observed by electron microscopy
onset in WT mice (blue bars, WT) and totally disappears in 18-month-old mutants (bottom-right panel, MUT) that
in β5−/− mice (red bars, MUT) (top-right panel). Retinal are absent in wild-type controls (WT). Modified from ©
adhesion is attenuated in β5−/−mice (grey bars, MUT) [21], © [22], © [10], and © [14]; originally published in
when compared to WT mice (black bars, WT) (bottom-­ (a) Journal of Experimental Medicine, https://doi.
left panel). The phagocytic peak is indicated with dashed org/10.1084/jem.20041447 and American Journal of
bars. β5−/− RPE cells accumulate autofluorescent deposits Physiology Cell Physiology, https://doi.org/10.1152/ajp-
(red) in contrast to WT RPE (bottom-right panel). RPE cell.00480.2005, and (b) The American Journal of
nuclei are in blue. (b) An ~40% phagocytosis decrease Pathology, https://doi.org/10.1016/j.ajpath.2014.06.026
was detected in Prpf31+/− RPE primary cultures (top-left and Investigative Ophthalmology and Vision Science,
panel). Circadian rhythms of both POS phagocytosis (top-­ https://doi.org/10.1167/iovs.10-­5194, respectively.

mice, the phagocytic peak is still present but PRs. Interestingly, retinal adhesion also follows a
attenuated and starting earlier (Fig. 1b) [10]. circadian activity with a peak occurring 1.5 h
Thus, the phagocytic rhythmicity is disrupted in after the phagocytic peak [22]. Integrins also
Prpf31+/− mice, but in a different manner. being adhesive receptors, we wondered if the
Interestingly, our team also demonstrated defects αvβ5 integrin would participate in the process. To
in the localization of some of the receptors evaluate retinal adhesion, we mechanically sepa-
involved in POS phagocytosis, including β5 inte- rate the RPE from the retina and quantify the
grin receptors and their ligands MFG-E8 [10]. transfer of RPE melanin pigments and RPE-­
specific RPE65 proteins onto the retina, which
translates the strength of RPE-POS attachment
3 Retinal Adhesion [22].
β5−/− retinae display significantly less melanin
With their extended apical microvilli, RPE cells pigments at all studied timepoints and more par-
maintain at all time close contacts with POS ticularly at the time when retinal adhesion peaks,
referred to as retinal adhesion, which is of the thus implying a crucial role of αvβ5 integrin
utmost importance to guarantee the feeding and receptor in retinal adhesion (Fig. 1a) [22].
trash disposal, as well as overall homeostasis of Moreover, retinal adhesion decreases with age
542 E. M. Vanoni and E. F. Nandrot

and the absence of β5 integrin receptors seems to activator complex includes Clock (Circadian
accelerate this process. These results confirm the locomotor output cycles kaput), Bmal1 (Brain
implication of αvβ5 integrin in retinal adhesion. and muscle ARNT-like protein 1), and Npas2
However, no retinal adhesion difference being (Neuronal PAS domain protein 2), while Per1–3
observed in mice lacking the MFG-E8 phago- (Period) and Cry1–2 (Cryptochrome) belong to
cytic ligand, hence the corresponding adhesion the repressor complex. The secondary transcrip-
ligand/s remain/s unknown at the present time tional loop is constituted of Rev-Erbα (also
[23]. Interestingly, the adhesion peak disappears known as Nr1d1, nuclear receptor subfamily 1
in Prpf31+/− mice but the function remains nor- group D member 1) and Rorα-γ (RAR-related
mal the rest of the time (Fig. 1b) [10]. orphan receptors). Other external proteins regu-
late the whole machinery such as DEC2 or
TWIST1 [18, 32].
4 Potential Link As both POS phagocytosis and retinal adhe-
with the Intracellular sion circadian rhythmicities are altered in
Circadian Clock Prpf31+/− mice (Fig. 1b) [10], we hypothesized
that the RPE circadian clock might be disrupted
As RPE cells display several circadian-regulated in this mouse model. Indeed, the circadian clock
functions, the question is open as to what extent is implicated in the pathogenesis of various dis-
they are related to the RPE intracellular clock. eases such as Alzheimer’s disease, cancers,
Indeed, the RPE and the retina have been hypertension, diabetes or obesity [3, 30, 34].
described to act as peripheral clocks in relation Interestingly, circadian clock disruptions have
with the suprachiasmatic nuclei (SCN) of the been increasingly suggested to lead to some ocu-
hypothalamus. Unlike the other peripheral lar diseases like glaucoma [39]. In addition, a
clocks, retinal photoreceptors synchronize the very recent study showed that Prpf8−/− mice dis-
central circadian clock by receiving and transmit- play lengthened daily locomotor activity periods
ting the light information via the retinohypotha- and disruption of some clock genes expression
lamic tract [31]. In addition, the rd mouse model patterns, reinforcing the hypothesis of more cen-
characterized by an early rod photoreceptors tral intracellular clock defects [33]. We have been
degeneration displays arrhythmic melatonin syn- studying the expression levels of RPE circadian
thesis after the degeneration, confirming the role clock genes and proteins along the light:dark
of photoreceptors as a peripheral clock [38]. In cycle in Prpf31+/− mice. Our current unpublished
addition to photoreceptors, it has been shown that data suggest that circadian clock components
the mouse inner nuclear layer —one of the retinal expression defects exist in mutant RPE compared
layers consisting of the bipolar, horizontal, and to control tissues. In vitro data also confirm that
amacrine cells— can also be considered as an disruption of the circadian clock by siRNA
autonomous circadian clock [29]. At the RPE silencing directly impacts RPE phagocytic
level, a human cell line (HRPE) shows a circa- capacities.
dian rhythmic expression of PER1 and PER2
clock genes [26]. In addition, PER2::LUC RPE
cells continue to show a circadian rhythm of bio- 5 Phenotypes Linked
luminescence production after SCN lesions iso- to Deregulated Circadian
lating them from the central clock [2]. Recently, Functions
a study showed that the transport of lactate to PRs
was under RPE circadian clock control [19]. POS phagocytosis and retinal adhesion deregula-
The circadian clock is a transcriptional feed- tions are associated with some RPE structural
back loop composed of three main components: defects in both β5−/− and Prpf31+/− mice (Fig. 1a,
an activator complex, a repressor complex, and a b) [10, 21]. Indeed, wide-field microscopy exper-
secondary transcriptional loop [5, 8, 28]. The iments showed that aged β5−/− RPE cells contain
The Retinal Pigment Epithelium: Cells That Know the Beat! 543

much higher numbers of vesicular autofluores- 21, 22]. However, the phagocytosis decrease is
cent storage bodies suggestive of lipofuscin accu- more important in β5−/− RPE primary cultures
mulation in contrast to WT tissues (Fig. 1a) [21]. and the rhythmicity of both functions is altered
This is associated with gradual loss of electroret- differently. In addition to RPE functional defects,
inogram responses with time, but no photorecep- both mutant mouse models display RPE struc-
tor degeneration is observed. Additionally, tural defects. While β5−/− RPE cells defects
perturbation of the mitochondrial respiration and resemble some of AMD cardinal signs, Prpf31+/−
ATP production are detected in freshly dissected RPE cells show more widespread ultrastructural
RPE/choroid tissues (unpublished data). On the defects indicative of cellular stress [14]. Indeed,
other hand, Prpf31+/− RPE cells display gross our unpublished data demonstrate overall pertur-
ultrastructural defects with age, namely basal bated RPE energetic metabolism in Prpf31+/− at
infoldings thickening and cytoplasmic vacuoles an early age that is tissue-specific. Implication of
accumulation (Fig. 1b) [14]. These results sug- the mitochondrial and energetic metabolism in
gest the presence of cellular stress in RPE cells. RPE phagocytosis and in aging diseases such as
Our recent studies do show an increase in protein AMD is becoming more widespread and animal
and lipid oxidation with age associated with pro- models can prove being useful to identify cellular
teasome activation, a sign of endoplasmic reticu- targets and molecular mechanisms underlying
lum stress. In young adults, we observe the different steps of pathological development
mitochondrial defects and decreased energy pro- [12, 15, 41].
duction from various cellular sources. Indeed, the
bioenergetic health index is modified and
Prpf31+/− RPE/choroid samples shift their status References
from quiescent to energetic with age, whereas
WT samples remain glycolytic. However, we 1. Audo I, Mohand-Said S, Boulanger-Scemama E, et al.
MERTK mutation update in inherited retinal diseases.
detect neither peritoneal macrophages nor whole Hum Mutat. 2018;39(7):887–913.
body metabolism defects in mutant mice, con- 2. Baba K, Sengupta A, Tosini M, et al. Circadian
firming the tissue-specificity of the phenotype regulation of the PERIOD2::LUCIFERASE biolu-
already showed for the phagocytic defect. minescence rhythm in the mouse retinal pigment
epithelium-­choroid. Mol Vis. 2010;16:2605–11.
3. Bao AM, Swaab DF. The human SCN in health and
neuropsychiatric disorders: postmortem observations.
6 Conclusion and Perspectives In: Gumz ML, editor. Physiology in health and dis-
ease “Circadian clocks: role in health and disease”.
New York: Springer; 2016. p. 117–52.
The alteration of the rhythmicity of two circadian 4. Bok D. The role of the pigment epithelium in the eti-
functions in Prpf31+/− suggests a potential altera- ology of inherited retinal dystrophy in the rat. J Cell
tion of the RPE circadian clock in this mouse Biol. 1985;49:664–82.
model [10]. Our current unpublished results seem 5. Brown SA. Circadian clock-mediated control of stem
cell division and differentiation: beyond night and
to confirm this hypothesis, and also show that cir- day. Development. 2014;141(16):3105–11.
cadian clock alterations directly impact POS 6. Bujakowska K, Maubaret C, Chakarova CF, et al.
phagocytosis in vitro. Our ongoing experiments Study of gene-targeted mouse models of splicing fac-
aim at identifying if these effects are linked to tor gene Prpf31 implicated in human autosomal domi-
nant retinitis pigmentosa (RP). Invest Opthalmol Vis
subcellular mislocalization of the circadian clock Sci. 2009;50:5927.
members or altered transcription of their target 7. Burstyn-Cohen T, Lew ED, Través PG, et al. Genetic
genes. This reinforces the link between the circa- dissection of TAM receptor-ligand interaction in
dian clock and ocular diseases that has already retinal pigment epithelial cell phagocytosis. Neuron.
2012;76:1123–32.
been shown, and there might be some other reti- 8. Chiou Y-Y, Yang Y, Rashid N, et al. Mammalian
nal activities related to the circadian clock that period represses and de-represses transcription by
have not been discovered yet [39]. displacing CLOCK-BMAL1 from promoters in a
β5−/− and Prpf31+/− mice both display POS cryptochrome-dependent manner. Proc Natl Acad Sci
U S A. 2016;113(41):E6072–9.
phagocytosis and retinal adhesion defects [10,
544 E. M. Vanoni and E. F. Nandrot

9. D’Cruz PM, Yasumura D, Weir J, et al. Mutation diurnal retinal phagocytosis. Proc Natl Acad Sci.
of the receptor tyrosine kinase gene Mertk in 2007;104:12005–10.
the retinal dystrophic RCS rat. Hum Mol Genet. 24. Nandrot EF, Silva KE, Scelfo C, et al. Retinal pigment
2000;9(4):645–51. epithelial cells use a MerTK-dependent mechanism to
10. Farkas MH, Lew DS, Sousa ME, et al. Mutations limit the phagocytic particle binding activity of αvβ5
in pre-mRNA processing factors 3, 8, and 31 cause integrin. Biol Cell. 2012;104:326–41.
dysfunction of the retinal pigment epithelium. Am J 25. Parinot C, Nandrot EF. A comprehensive review of
Pathol. 2014;184:2641–52. mutations in the MERTK proto-oncogene. Adv Exp
11. Finnemann SC, Bonilha VL, Marmorstein AD, et al. Med Biol. 2016;854:259–65.
Phagocytosis of rod outer segments by retinal pig- 26. Pavan B, Frigato E, Pozzati S, et al. Circadian
ment epithelial cells requires αvβ5 integrin for bind- clock regulate adenylyl cyclase activity rhythms in
ing but not for internalization. Proc Natl Acad Sci. human RPE cells. Biochem Biophys Res Commun.
1997;94:12932–7. 2006;350(1):169–73.
12. Fisher CR, Ferrington DA. Perspective on AMD 27. Rodrigues A, Slembrouck-Brec A, Nanteau C, et al.
pathobiology: a bioenergetic crisis in the RPE. Invest Modeling PRPF31 retinitis pigmentosa using reti-
Ophthalmol Vis Sci. 2018;59(4):AMD41–7. nal pigment epithelium and organoids combined
13. Gal A, Li Y, Thompson DA, Weir J, et al. Mutations with gene augmentation rescue. NPJ Regen Med.
ins MERTK, the human orthologue of the RCS rat 2022;7(1):39.
retinal dystrophy gene, cause retinitis pigmentosa. 28. Ruan G-X, Zhang D-Q, Zhou T, et al. Circadian orga-
Nat Genet. 2000;26(3):270–1. nization of the mammalian retina. Proc Natl Sci U S
14. Graziotto JJ, Farkas MH, Bujakowska K, et al. Three A. 2006;103(25):9703–8.
gene-targeted mouse models of RNA splicing factor 29. Ruan G-X, Allen GC, Yamazaki S, et al. An autono-
RP show late-onset RPE and retinal degeneration. mous circadian clock in the inner mouse retina by
Invest Opthalmol Vis Sci. 2011;52:190–8. dopamine and GABA. PLoS Biol. 2008;6(10):e249.
15. Klipfel L, Cordonnier M, Thiébault L, et al. A splice 30. Rudic. Circadian rhythms and the circadian clock
variant in SLC16A8 gene leads to lactate transport in the cardiovascular system. In: Gumz ML, editor.
deficit in human iPS cell-derived retinal pigment epi- Physiology in health and disease “Circadian clocks:
thelial cells. Cells. 2021;10(1):179. role in health and disease”. New York, NY: Springer;
16. LaVail MM. Rod outer segment disk shedding in 2016. p. 199–224.
rat retina: relationship to cyclic lighting. Science. 31. Sadun AA, Schaechter JD, Smith LE. A retinohypo-
1976;194:1071–4. thalamic pathway in man: light mediation of circadian
17. Law A-L, Parinot C, Chatagnon J, et al. Cleavage of rhythms. Brain Res. 1984;302(2):371–7.
Mer tyrosine kinase (MerTK) from the cell surface 32. Sato F, Kohsaka A, Bhawal UK, et al. Potential
contributes to the regulation of retinal phagocytosis. Roles of Dec and Bmal1 Genes in Interconnecting
J Biol Chem. 2015;290:4941–52. Circadian Clock and Energy Metabolism. Int J Mol
18. Meier D, Lopez M, Franken P, et al. Twist1 is a TNF- Sci. 2018;19(3):781.
inducible inhibitor of clock mediated activation of 33. Shakhmantsir I, Dooley SJ, Kishore S. RNA splic-
period genes. PLoS One. 2015;10(9):e0137229. ing factors mutations that cause retinitis pigmentosa
19. Milićević N, Ten Brink JB, Ten Asbroeck ALMA, result in circadian dysregulation. J Biol Rhythm.
et al. The circadian clock regulates RPE-mediated 2020;35(1):72–83.
lactate transport via SLC16A1 (MCT1). Exp Eye Res. 34. Situala S, Amador A, Burris TP. The circadian clock
2020;190:107861. as a drug target. In: Gumz ML, editor. Physiology
20. Nandrot E, Dufour EM, Provost AC, et al. in health and disease “Circadian clocks: role in
Homozygous deletion in the coding sequence of the health and disease”. New York, NY: Springer; 2016.
c-mer gene in RCS rats unravels general mecha- p. 335–66.
nisms of physiological cell adhesion and apoptosis. 35. Strauss O. The retinal pigment epithelium in visual
Neurobiol Dis. 2000;7:586–99. function. Physiol Rev. 2005;85:845–81.
21. Nandrot EF, Kim Y, Brodie SE, et al. Loss of syn- 36. Tamai M, Teirstein P, Goldman A, et al. The
chronized retinal phagocytosis and age-related pineal gland does not control rod outer segment
blindness in mice lacking αvβ5 integrin. J Exp Med. shedding and phagocytosis in the rat retina and
2004;200:1539–45. pigment epithelium. Invest Ophthalmol Vis Sci.
22. Nandrot EF, Anand M, Sircar M, et al. Novel 1978;17(6):558–62.
role for alphavbeta5-integrin in retinal adhesion 37. Teirstein PS, Goldman AI, O’Brien PJ. Evidence
and its diurnal peak. Am J Physiol Cell Physiol. for both local and central regulation of rat rod outer
2006;290(4):C1256–62. segment disc shedding. Invest Ophthalmol Vis Sci.
23. Nandrot EF, Anand M, Almeida D, et al. Essential 1980;19(11):1268–73.
role for MFG-E8 as ligand for αvβ5 integrin in
The Retinal Pigment Epithelium: Cells That Know the Beat! 545

38. Tosini G, Menaker M. The clock in the mouse retina: cally impair phagocytosis by retinal pigment epithelial
melatonin synthesis and photoreceptor degeneration. cells. J Biol Chem. 2008;283(36):24770–80.
Brain Res. 1998;789(2):221–8. 42. Vollrath D, Feng W, Duncan JL, et al. Correction of
39. Tosini G, Pozdeyev N, Sakamoto K, et al. The cir- the retinal dystrophy phenotype of the RCS rat by
cadian clock system in the mammalian retina. viral gene transfer of Mertk. Proc Natl Acad Sci U S
BioEssays. 2008;30(7):624–33. A. 2001;98(22):12584–9.
40. Tschernutter M, Jenkins SA, Waseem NH, et al. 43. Winkler BS. An hypothesis to account for the renewal
Clinical characterisation of a family with retinal dys- of outer segments in rod and cone photoreceptor cells:
trophy caused by mutation in the Mertk gene. Br J renewal as a surrogate antioxidant. Invest Ophthalmol
Ophthalmol. 2006;90(6):718–23. Vis Sci. 2008;49:3259–61.
41. Vives-Bauza C, Anand M, Shiraz AK, et al. The age 44. Young RW, Bok D. Participation of the retinal pig-
lipid A2E and mitochondrial dysfunction synergisti- ment epithelium in the rod outer segment renewal pro-
cess. J Cell Biol. 1969;42:392–403.
Part XIII
Stem Cell Models and Therapies
Retinal Organoids: A Human
Model System for Development,
Diseases, and Therapies

Sangeetha Kandoi and Deepak A. Lamba

Abstract peutic approaches to restore or prevent vision


loss in affected individuals.
Inherited retinal degenerations (IRD) encom-
passes a group of heterogeneous disorders Keywords
causing debilitating visual diseases and blind-
ness, affecting more than two million people Stem cells · Organoids · Mini-retinas ·
worldwide, in all age groups. The inheritance Inherited retinal degenerations · Disease-­
patterns vary from autosomal dominant, auto- modeling · Precision medicine
somal recessive, X-linked, and sporadic with
mutations in over 260 genes identified to date.
Despite the significant advances in clinical 1 Introduction
diagnosis, there is no effective treatment avail-
able. Human-induced pluripotent stem cells Embryonic stem cells and human-induced plu-
(hiPSC) derived in vitro 3D retinal organoids ripotent stem cells (hiPSC) are pluripotent stem
offer a powerful preclinical tool to investigate cell populations that have the capacity to gener-
the molecular mechanism(s) of inherited dis- ate cell types from all three germ layers (ecto-
eases. Organoids have the potential for the derm, endoderm, and mesoderm). As opposed to
development of personalized therapies by the conventional two-dimensional monolayer
modeling the disease-specific and patient-­ cell cultures, which are immortalized and often
specific IRD. This mini-review will elaborate limited by the scarcity of primary tissues, iPSCs
on the utility of the advanced culture model can be established from any somatic cell source
system by focusing on staging the in vitro and age groups [19]. Human organoids derived
human retinogenesis, modeling retinal dis- from iPSC are defined as three-dimensional (3D),
eases, and as a tool for testing potential thera- self-organized, genetically stable in vitro tissues.
They are composed of heterogeneous cell types
along with adhesion proteins to provide struc-
S. Kandoi (*) · D. A. Lamba tural support. They are also called “mini-organs”
Department of Ophthalmology, University of that have been widely used to study the molecu-
California San Francisco, San Francisco, CA, USA lar mechanism(s), including the signals that are
Eli & Edythe Broad Center for Regeneration needed in the development and maintenance of
Medicine and Stem Cell Research, University of any organ and for disease modeling [5].
California San Francisco, San Francisco, CA, USA Remarkably, they can model a complex tissue’s
e-mail: sangeetha.kandoi@ucsf.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 549
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_80
550 S. Kandoi and D. A. Lamba

architecture with the temporal and spatial receptors (NRL, NR2E3 ) and bipolar cells (VSX1)
arrangement of cell types, thereby mimicking the at days 90–150; and lastly Müller glial cells
physiologically milieu that exists in vivo. The (RLBP1, GS) by days 160–200. Maturation of
development of organoid technology represents a photoreceptors shows as brush-like projections
promising platform to overcome the drawbacks which are indicative of inner and outer segments
of traditionally used cell lines and the historically (containing opsins) on the laminated retina,
reliant animal models, which do not always effec- and occurs between days 160 and 200. This long
tively recapitulate human physiology and dis- culture process of maintaining the “viable” organ-
eases. Lately, organoids have become mainstream oids is done with minimal manipulation. Recent
in vitro tools, highlighting their value for basic progress has been made in the construction of a
biological research and numerous clinical appli- multi-organoid culture system termed “assemb-
cations including precision medicine, regenera- loid,” a fusion of retinal and brain organoids. This
tive medicine, biobanking, and high-throughput has the potential to enable us to study connectiv-
toxicology studies. ity, ganglion cell axon guidance, and transmission
of visual signals from the developing eye to the
brain [10].
2 Retinal Organoids: Staging cell types across the different devel-
“Mini-retinas” opmental stages of retinal organoids have become
relatively easy with a range of techniques avail-
The generation of iPSCs and their directed dif- able. Some commonly utilized ones include
ferentiation to organoids has meant that the ana- immunohistochemistry (IHC), quantitative real-­
tomical development and the regulatory networks time polymerase chain reaction (qRT-PCR), bulk
that drive the formation of this scarcely available RNA sequencing, and transmission electron
human retinal tissue can now be studied “in a microscopy (TEM). More recently, transcrip-
dish.” The complex process of retinogenesis tomic signatures and gene regulatory networks
involves three sequential processes: (i) specifica- associated with cell types during organoid devel-
tion of the eye field, (ii) morphogenesis of the opment and comparison to human fetal tissue
optic cup, and (iii) differentiation to the different have provided powerful insights using deep-­
retinal cell types [21]. Apart from the pioneering sequencing techniques at the single-cell level
work carried out by Yoshiki Sasai [9], a number (scRNAseq) [24].
of groups have also shown that the retinogenesis Retinal organoids, although a powerful
timeline in vitro through iPSC-derived retinal in vitro model for the human neural retina, have
organoids faithfully matches that of in vivo. some key components missing. Lack of orga-
There are multiple protocols available in the lit- nized accessory tissues and nonneural cell types,
erature for the differentiation of fully matured such as vasculature, macula, fovea (cone-rich
laminated retinal organoids [3, 22, 25]. The most central region of the retina), and immune cells
commonly adopted culture scheme is using a (such as microglia), remains a caveat in this
combination of 2D/3D approaches. A representa- model. Lack of oxygen and nutrient diffusion
tive bright-field microscopy images of a matured deficiency at the interior of the retinal organoids
retinal organoid from hiPSC is shown in Fig. 1. affects the local microenvironment, increasing
Differentiation of seven classes of postmitotic the necrosis of early-differentiated ganglion
retinal cell types (recognized by their markers) cells. Additionally, the well organized monolay-
progresses with the birth-dating similar to the ered retinal pigmented epithelium (RPE), which
human retina in utero. The developmental order physically interfaces and supports photoreceptor
begins with birth of ganglion cells (BRN3) at day function in vivo, is lacking in the retinal organ-
30; horizontal cells (LHX1, ONECUT1), cone oids. RPE instead grows as a separate ectopic
photoreceptors (OTX2, RCVRN, RXRG), and patch of tissue adhering to one corner in the
amacrine cells (HuC/D) at days 40–70; rod photo- ­retinal organoid. While these challenges are not
Retinal Organoids: A Human Model System for Development, Diseases, and Therapies 551

Retinal Differentiation
hiPSC
Neural Induction Medium Retinal Induction Medium 3D-Retinal Induction Medium
2D 3D 2D 3D

D0 D7 D25-28 D30 D180-200

A B C D E
F

Fig. 1 Schematic timeline, culture conditions, and the generate self-forming embryoid bodies (b), embryoid
representative bright-field microscopic images of bodies with optic vesicles and cup-like structures (c),
3D-retinal organoids differentiated from hiPSC using a laminated 3D-organoids (d), and organoids with fully
combination of 2D/3D approach. Undifferentiated hiPSC matured photoreceptors (e and f) displaying outer seg-
(a) directed toward retinal fate using step-wise protocol to ments on the periphery/apical side of the organoids

insurmountable, they require further optimiza- ficking, and impairment of ciliary function lead-
tion in the culture conditions. ing to the degeneration of rods and cones.
Differentiation of iPSCs lines engineered with
a disease-specific mutation, or a patient’s own
3 Modeling Inherited Retinal iPSC’s line, into retinal organoids can be used to
Degenerations Using Retinal decipher the molecular mechanisms and provide
Organoids clues into the pathogenesis of IRD. Studying the
pathophysiology of IRD using retinal organoids
Retinal degenerations include a group of hetero- has been particularly useful over animal models
geneous diseases ranging from monogenic inher- as organoids mimic the human cellular composi-
ited Retinitis Pigmentosa (RP), Leber congenital tion, associates the phenotypes with patient phe-
amaurosis (LCA), and Cone-rod dystrophy (CRD) notypes, and aid in understanding the disease
to genetically complex age-related macular modifiers in the patient’s genome. Some of the
degenerations (AMD). Significant advances have most recent IRDs, modeled using patient iPSC-­
been made to identify the cause of inherited reti- derived retinal organoids carrying the specific
nal degenerations (IRD) in patients using genetic mutation candidates, are shown in Table 1. While
testing, whole-genome sequencing, and next-gen- retinal organoids have been very useful for mod-
eration sequencing [8]. Mutation in over 260 eling early-onset IRDs, a key challenge of retinal
genes has been identified to be the causative fac- organoid technology is the difficulty in modeling
tor for the death of retinal neurons and RPE lead- late-onset diseases where patient’s phenotypes
ing to progressive irreversible vision loss appear in later decades of life. However, compar-
(peripheral and central) and eventual blindness in ing the patient organoids with the control organ-
all age groups [1]. Many of these mutations are oids can still provide some insights into the
located on the genes that encode the sensory mechanistic understanding of pathogenesis.
regions of the photoreceptors and can be feasibly Finally, modeling of complex disorders like
modeled using retinal organoids. These mutations AMD still remains challenging due to the involve-
causes abnormal development of photoreceptors, ment of genetic components, aging-associated
defects in the phototransduction mechanisms, risk factors, and external environmental factors
endoplasmic reticulum stress, protein mis-traf- that contribute to the disease mechanisms.
552 S. Kandoi and D. A. Lamba

Table 1 Patient-specific retinal organoids derived from various IRD for studying the mutation-specific phenotypes and
various therapeutic approaches tested in some studies
Inheritance Therapeutic
IRD Genes (mutation) pattern Retinal organoid phenotype intervention References
RP RP2 (RP2KO, X-linked Rod PR death (D150) and AAV gene Lane et al.
R120X) outer nuclear layer thinning augmentation [16]
(D180)
RP PDE6B Autosomal Elevated cGMP levels, – Gao et al.
recessive impaired synaptic [11]
connections and connecting
cilium in PR (D193, D230)

RP RPGR (frame-shift – Shortened cilium. Defects CRISPR-Cas9-­ Deng et al.


mutation) in PR (morphology, mediated [6]
localization, transcriptional correction
profiling, and
electrophysiological
activity)
LCA4 AIPL1 (Cys89Arg) – Low expression of AIPL1 – Lukovic
and PDE6alpha (weeks 21 et al. [18]
and 27)
LCA7 CRX (I138fs48, Autosomal Defective PR maturation AAV gene Kruczek
K88N) dominant and diminished expression augmentation et al. [14]
of visual opsins
LCA7 CRX (T155ins4, Autosomal Immature PRs (D180) Allele-specific Chirco
K88Q) dominant CRISPR/ et al. [4]
Cas9-based gene
editing
LCA10 CEP20 Autosomal Defective PR ciliation QR-110 (RNA Dulla et al.
(c.2991+1655A>G) recessive oligonucleotide) [7]
Achromatopsia ATF6 (biallelic – Do not form cone structures Small molecule Kroeger
variants—Y567N and and lack cone opsins ATF6 agonist, et al. [13]
R324C) AA147

4 Therapeutic Approaches ferentiating cultures [15]. Additionally, targeting


Using Retinal Organoids the signaling pathways like Notch by treatment
of retinal organoids with small molecule(s)
Massive efforts are being made to utilize this (PF4014, DAPT) promotes the supernumerary
unique human model system for validating a formation of specific cell fates like cones in a
panel of therapeutic interventions. Some of these synchronized fashion, making them abundantly
include cell transplantation, small molecule available for IRD with cone deterioration [12].
screening, antisense oligonucleotides, and Such strategies circumvent the limitations to gen-
genome editing technologies. Since several IRD erate cell types that are sparsely generated.
primarily affects the postmitotic light-sensing Continuous ongoing efforts are being made by
photoreceptors, transplanting the iPSC differenti- independent investigators to demonstrate the sur-
ated photoreceptor precursors in the animal mod- vival and integration of the transplanted photore-
els with dysfunctional/lost photoreceptors has ceptors within the host retina.
been shown to rescue some level of vision [2, Retinal organoids have also been used as a
17]. Genetically engineered hiPSC lines with tool to identify novel therapeutics. For example,
single or multiple fluorescent constructs (green retinal organoids from LCA10 patients with
fluorescent protein (GFP) or mCherry) driven by CEP290 intronic mutation have been used to val-
cell type-specific promotors (e.g., OTX2 or CRX) idate antisense oligonucleotide therapeutics for
allow for the tracking of photoreceptors in dif- pre-mRNA splice modulation and to restore
Retinal Organoids: A Human Model System for Development, Diseases, and Therapies 553

the function [7]. Similarly, a frequently diag- Dr. Lamba, All May See postdoctoral fellowship to Dr.
Kandoi, Research to Prevent Blindness (unrestricted grant
nosed deep-intronic mutation in to Department of Ophthalmology, University of California
­USH2A-­associated retinal degeneration causing San Francisco) and UCSF Vision core grant NIH/NEI P30
the aberrant splicing of truncated nonfunctional EY002162.
protein could potentially be splice corrected
using antisense oligonucleotide on patient-
derived fibroblast [23]. Such approaches can pre- References
serve the survival and function of the cells under
diseased conditions. Likewise, genome editing of 1. Anon RetNet: Summaries [Online]. Available at:
https://sph.uth.edu/retnet/sum-­dis.htm. Accessed 13
the patient-specific iPSC lines with numerous Nov 2021.
IRD is being carried out using CRISPR-Cas9 or 2. Assawachananont J, Mandai M, Okamoto S, et al.
zinc finger nucleases (ZFN) to create an ideally Transplantation of embryonic and induced pluripotent
corrected isogenic control lines or for knocking stem cell-derived 3D retinal sheets into retinal degen-
erative mice. Stem Cell Rep. 2014;2(5):662–74.
down dominant mutant alleles. These approaches 3. Capowski EE, Samimi K, Mayerl SJ, et al.
can strategically be used to validate the develop- Reproducibility and staging of 3D human retinal
ment and amelioration of disease phenotypes organoids across multiple pluripotent stem cell lines.
providing a proof-of-concept [4]. Finally, retinal Development. 2019;146(1).
4. Chirco KR, Chew S, Moore AT, Duncan JL, Lamba
organoids can be very useful as effective drug DA. Allele-specific gene editing to rescue dominant
screening tools for toxicity testing. For example, CRX-associated LCA7 phenotypes in a retinal organ-
a research in our laboratory is currently focused oid model. Stem Cell Rep, vol. 16; 2021. p. 2690.
to validate a previously described NR2E3 inhibi- 5. Corrò C, Novellasdemunt L, Li VSW. A brief his-
tory of organoids. Am J Physiol Cell Physiol.
tors in RHO copy number variant patient-specific 2020;319(1):C151–65.
iPSC-derived retinal organoids [20]. Altogether, 6. Deng W-L, Gao M-L, Lei X-L, et al. Gene correction
the development of therapies using retinal organ- reverses ciliopathy and photoreceptor loss in iPSC-­
oid model systems for numerous IRD provides a derived retinal organoids from retinitis pigmentosa
patients. Stem Cell Rep. 2018;10(4):1267–81.
proof-of-concept to inform the future clinical 7. Dulla K, Aguila M, Lane A, et al. Splice-modulating
therapies. oligonucleotide QR-110 restores CEP290 mRNA and
function in human c.2991+1655A>G LCA10 models.
Mol Ther Nucleic Acids. 2018;12:730–40.
8. Duncan JL, Pierce EA, Laster AM, et al. Inherited ret-
5 Conclusion inal degenerations: current landscape and knowledge
gaps. Transl Vis Sci Technol. 2018;7(4):6.
Significant advances made in the field of stem 9. Eiraku M, Sasai Y. Self-organizing optic-cup morpho-
cell biology over the last decade to generate 3D genesis in three-dimensional culture. Neurosci Res.
2011;71:e127–8.
organoids from patients offer them as a powerful 10. Fligor CM, Lavekar SS, Harkin J, et al. Extension
and efficient preclinical model that can substan- of retinofugal projections in an assembled model of
tially complement or reduce the use of animal human pluripotent stem cell-derived organoids. Stem
models. Addressing the challenges of reproduc- Cell Rep. 2021;16(9):2228–41.
11. Gao M-L, Lei X-L, Han F, et al. Patient-specific
ibility and variability, time taken to mature cer- retinal organoids recapitulate disease features of
tain cell types with a globally standardized late-onset retinitis pigmentosa. Front Cell Dev Biol.
protocol and large-scale systematic analysis of 2020;8:128.
observed results is likely to significantly enhance 12. Kaufman ML, Park KU, Goodson NB, et al.
Transcriptional profiling of murine retinas undergoing
the utility of organoids in the near future. semi-synchronous cone photoreceptor differentiation.
Dev Biol. 2019;453(2):155–67.
Acknowledgments We thank the members of the Lamba 13. Kroeger H, Grandjean JMD, Chiang W-CJ, et al. ATF6
Lab for their valuable discussion and suggestions. The is essential for human cone photoreceptor develop-
writing of this mini-review is supported by the National ment. Proc Natl Acad Sci U S A. 2021;118(39).
Eye Institute (U24 EY029891 and R01EY032197 to Dr. 14. Kruczek K, Qu Z, Gentry J, et al. Gene therapy of
Lamba), Foundation Fighting Blindness research grant to dominant CRX-Leber congenital amaurosis using
554 S. Kandoi and D. A. Lamba

patient stem cell-derived retinal organoids. Stem Cell 21. O’Hara-Wright M, Gonzalez-Cordero A. Retinal
Rep. 2021;16(2):252–63. organoids: a window into human retinal development.
15. Lam PT, Gutierrez C, Del Rio-Tsonis K, Robinson Development. 2020;147(24).
ML. Generation of a retina reporter hiPSC line to 22. Ohlemacher SK, Iglesias CL, Sridhar A, Gamm DM,
label progenitor, ganglion, and photoreceptor cell Meyer JS. Generation of highly enriched popula-
types. Transl Vis Sci Technol. 2020;9(3):21. tions of optic vesicle-like retinal cells from human
16. Lane A, Jovanovic K, Shortall C, et al. Modeling and pluripotent stem cells. Curr Protoc Stem Cell Biol.
rescue of RP2 retinitis pigmentosa using iPSC-derived 2015;32:1H.8.1–1H.8.20.
retinal organoids. Stem Cell Rep. 2020;15(1):67–79. 23. Slijkerman RW, Vaché C, Dona M, et al. Antisense
17. Lin B, McLelland BT, Mathur A, Aramant RB, Seiler oligonucleotide-based splice correction for USH2A-­
MJ. Sheets of human retinal progenitor transplants associated retinal degeneration caused by a frequent
improve vision in rats with severe retinal degenera- deep-intronic mutation. Mol Ther Nucleic Acids.
tion. Exp Eye Res. 2018;174:13–28. 2016;5(10):e381.
18. Lukovic D, Artero Castro A, Kaya KD, et al. Retinal 24. Sridhar A, Hoshino A, Finkbeiner CR, et al. Single-­
organoids derived from hiPSCs of an AIPL1-LCA cell transcriptomic comparison of human fetal ret-
patient maintain cytoarchitecture despite reduced lev- ina, hPSC-derived retinal organoids, and long-term
els of mutant AIPL1. Sci Rep. 2020;10(1):5426. retinal cultures. Cell Rep. 2020;30(5):1644–1659.
19. Malik N, Rao MS. A review of the methods for human e4.
iPSC derivation. Methods Mol Biol. 2013;997:23–33. 25. Wahlin KJ, Maruotti JA, Sripathi SR, et al.
20. Nakamura PA, Shimchuk AA, Tang S, et al. Small Photoreceptor outer segment-like structures in long-­
molecule Photoregulin3 prevents retinal degeneration term 3D retinas from human pluripotent stem cells.
in the RhoP23H mouse model of retinitis pigmentosa. Sci Rep. 2017;7(1):766.
elife. 2017;6.
Modeling Retinitis Pigmentosa
with Patient-Derived iPSCs

Yeh Chwan Leong and Jane C. Sowden

Abstract 1 Introduction

Retinitis pigmentosa (RP) causes blindness in Retinitis pigmentosa (RP) is the most common
1 out of 3000–4000 individuals worldwide. form of inherited retinal diseases (IRDs) with a
Understanding the disease mechanism under- prevalence of 1 in 3000 worldwide [1]. More
lying the death of photoreceptors in RP patient than 80 genes and loci exhibiting autosomal-­
is crucial for the discovery and development dominant, autosomal-recessive, or X-linked
of therapies to prevent and stop the progres- modes of inheritance have been identified to
sion of retinal degeneration. Despite having cause RP [2]. Patients with RP first suffer
provided valuable insight into RP pathology, progressive loss of rod photoreceptor (PR) cells,
several shortcomings of animal models war- resulting in loss of peripheral vision and night
rant the need for a better modeling system. blindness. This is followed by cone PR
This review discusses the current use of degeneration, leading to the loss of central vision
patient-derived induced pluripotent stem cells and eventual legal blindness [3].
(iPSCs) to model RP and its advantages over
animal models. Further improvement to
enhance the representativeness of iPSC RP 2 Challenges in RP Disease
models is also discussed. Modeling

Keywords To date, the molecular mechanisms leading to the


onset of death of rod PR cells remain largely
Retinitis pigmentosa · iPSC · Disease elusive in RP. This is due to several major
modeling · Retina degenerative · Retinal challenges to model the disease, which include
organoids · Photoreceptors · Disease the following.
mechanisms 1. The genetic heterogeneity of RP. Despite the
application of next-generation sequencing
(NGS) technologies, approximately 30% of
Y. C. Leong · J. C. Sowden (*) RP cases have an unidentified genetic
Stem Cells and Regenerative Medicine Section, UCL
Great Ormond Street Institute of Child Health, diagnosis [4]. Known RP genes are responsible
University College London and NIHR Great Ormond for diverse functions in the neural retina and
Street Hospital Biomedical Research Centre, retinal pigment epithelium (RPE), and thus it
London, UK is challenging to attribute the cause of RP to
e-mail: j.sowden@ucl.ac.uk

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 555
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_81
556 Y. C. Leong and J. C. Sowden

specific molecular processes or common 4. Complexity of genetic and epigenetic


pathways. Furthermore, the function of many interaction. The presence of distinct modifier
RP genes has not been fully elucidated and in genes in different genetic backgrounds can have
some cases, missing links between experi- a dramatic effect on the presentation of the RP
mentally identified phenotypes and gene phenotype and its severity. For instance, the
functions are observed and warrant additional simultaneous presence of Rpgr knockout and
research effort [5]. heterozygous hypomorphic mutation of Cep290
2. The lack of faithful animal models. A recent in mice induced early onset of retinal degenera-
review on mouse models of IRDs indicated that tion in comparison to Rpgr knockout alone
only 40% of known IRD genes are currently [10]. Besides, one of a pair of sisters harboring
modeled in mice. Despite being the most fre- USH2A homozygous mutations was reported to
quently used animal model for IRDs, major have earlier onset of RP and a more degenera-
interspecies differences in retinal architecture tive phenotype due to the presence of a frame-
and function exist between mouse and human shift mutation in PDZD7 [11]. In addition,
[6]. For instance, the mouse is nocturnal (with Walia et al. [12] reported two distinct clinical
less exposure to light throughout life ) and has observations of cone–rod dystrophy and
a rod-dominated retinae that lacks a macula. In X-linked RP, respectively, in two brothers har-
addition, the absence of human vision-related boring the same RPGR mutation. Both former
genes, such as EYS and ARMS2, and the dis- cases highlight the additive effect of modifier
crepancy between rodent and human retinal genes and the latter underscores intrafamilial
ageing patterns have also been reported [7]. variability of the RP phenotype. Zhang et al.
These drawbacks become apparent when, for [13] reported reduced severity of a degenerative
example, several Usher syndrome type I mutant retinal phenotype in F2 Nr2e3 rd7/rd7 homozy-
mouse models (such as shaker 1 and deaf cir- gotes generated from outcrosses of B6.Cg-
cler mice) fail to recapitulate the retinal degen- Nr2e3rd7/rd7 mice to AKR/J, CAST/EiJ, and
eration typically observed in Usher patient, NOD.NON-H2nb1 (all with distinct genetic
despite showing deafness and vestibular dys- backgrounds) and subsequent intercross of the
function [8]. Other large animals, such as dog, F1 mice. Follow-­up studies identified Rev-erbα
pig, and non-­human primates, have also been (Nr1d1) as a modifier gene to Nr2e3 with sup-
utilized to investigate the pathogenesis and pressive effects on retinal degeneration [14].
develop therapeutic approaches for RP. Collectively, these findings suggest the pres-
However, these models are cost-prohibitive ence of intricate genetic and epigenetic interac-
and time-­consuming for the generation of suf- tions within the disease network, highlighting
ficient colonies and the manifestation of dis- the importance of modeling RP with models of
ease phenotype owing to slow reproduction the right genetic background, such as the
rate and long life span. Besides, the fovea, a pit patients themselves.
within the macula that contains the most cone
PRs and contributes to the highest visual acu-
ity, is absent in most large animals [9]. 3 Modeling RP with Patient-­
3. Scarcity of human samples and their limitation Derived iPSCs
to study disease progression. The availability
of postmortem RP patient retinal samples is Induced pluripotent stem cells (iPSCs) technology
scarce. Severely degenerative retina is not was first introduced in 2006 [15] and utilized in
ideal for disease modeling due to the rare human cells in 2007 to revert somatic cells back to
presence of cells of interest (i.e. rod PRs). In an embryonic stem cell-like state via the overex-
addition, postmortem patient retina provides pression of OCT4, SOX2, KLF4, and c-MYC tran-
only a snapshot of a particular disease stage scription factors [16]. Since then, it has been
and provides limited information about extensively employed to derive iPSCs from patient
disease progression. cells to serve a wide range of research purposes,
Modeling Retinitis Pigmentosa with Patient-Derived iPSCs 557

including modeling of simple and complex disor- of iPSC banks/depositories in many countries,
ders ranging from monogenic RP to cancers [17]. such as the United States (WiCell), Japan
Patient derived iPSC are often used in conjunction (RIKEN), and the United Kingdom (HipSci), has
with robust differentiation protocols to generate made available an arsenal of patient and control
cells of specific lineages, representing disease- iPSCs to suit different experimental designs [23].
affected cells in the in vivo setting. In IRDs, In particular, it streamlines the acquisition of
numerous retinal differentiation protocols have comprehensively characterized, sex- and age-
been reported to faithfully generate various retinal matched controls and patients of diverse genetic
cell types, including PRs and RPE [18]. Table 1 backgrounds, which under normal circumstances,
shows a summary of published RP disease model- are difficult to obtain [24].
ing studies using patient iPSCs. Notably, many of
these use an insufficient number of iPSC lines to
determine if the reported effects are due to RP 3.2 Factors Important
gene mutations or are –inter-iPSC line variations for the Success of RP
(discussed later) (Table 1). Modeling with Patient iPSCs

In this section, factors crucial for the success of


3.1 Benefits of Patient iPSCs RP disease modeling using patient iPSCs are dis-
for RP Disease Modeling cussed. They are as follows.
1. The generation of retinal organoids that
Comparison of different aspects (molecular, closely recapitulate the complexity of human
cellular, and functional) of patient and healthy retina in vivo. The first modeling study of RP
retinal cells derived from their respective iPSCs in using iPSC was described by the Takahashi
vitro creates a powerful “disease in a dish” model. group involving five patients with mutations
Modeling RP with iPSCs circumvents caveats in RP1, RP9, PRPH2, and RHO [25]. A
met by classical animal models. First, it retains a directed, 2D adherent differentiation protocol
patient’s gene mutation, genetic background, and, was adopted to generate rod PR cells, which
if existing, any potential modifier gene effects, showed some electrophysiological function.
providing a genuine representation of RP ex vivo Subsequently, various 3D platforms have
at the molecular level. iPSCs are capable of divid- been established to generate retinal organoids
ing indefinitely to enable, theoretically, an unlim- that better recapitulate in vivo retinogenesis
ited supply of research material for disease and contain various retinal cell types orga-
modeling [19]. The reproducibility and scalability nized in pseudo-laminated retinal tissue, pro-
of iPSCs, in concert with the implementation of viding a more complex model to study RP
rigorous current good manufacturing practice, is pathology. The 3D organoid models allow
exceptionally important for cell therapy, where long-term maintenance and maturation of PRs
limited cell source is considered one of the big- to acquire subcellular structures, such as the
gest bottlenecks [20]. In addition, the use of connecting cilium, which has enabled the
patient iPSCs bypasses interspecies differences identification of cilia defects in RP modeling
and avoids the application of gene editing proce- studies concerning RPGR and PRPF31 muta-
dures to create transgenic animals carrying the tion [26, 27]. However, further improvements,
desired human RP gene mutation [21]. such as the generation of fully mature outer
Nevertheless, under circumstances where gene segments, light-responsive PRs that generates
editing is required to generate isogenic mutant electrophysiological output, co-culture of
(containing predicted pathogenic RP gene muta- RPE overlaying PRs, and outer segment
tion) or rescued (RP gene mutation corrected) phagocytosis, as well as the inclusion of vas-
lines, iPSCs represent an easier option for manip- cularization, would enhance resemblance to
ulation and screening for mutant clones than ani- the retina in vivo, benefiting the robustness
mal models [22]. Furthermore, the establishment and representativeness of the model.
Table 1 A summary of RP disease modeling studies using patient iPSCs and experimental detail
Latest
Differentiation iPSC cell Reprogramming iPSC time Treatment/
Reference M.O.I. Mutated gene (mutation) protocol Major findings source Feeder approach characterization #Patient #Control point rescue
2D culture
[25] adRP RP1, axonemal [37] Elevation of various Skin Yes Retrovirus IF, karyotypic 5 1 – 150D α-Tocopherol,
microtubule associated stress markers, fibroblast analysis, (~21W) ascorbic acid,
(p.721Lfs722X) including ER stress, teratoma or β-carotene
RP9, pre-mRNA splicing DNA oxidation, and formation
factor (p.H137L) apoptosis
PRPH2, Peripherin 2
(p.W316G)
RHO, Rhodopsin (p.
G188R)
[38] arRP MAK, male germ [39] Identification of Alu Skin Yes Sendai virus IF, karyotypic 1 1 – 150D –
cell-associated kinase insertional mutation fibroblast analysis, (~21W)
(homozygous 353 bp Alu and impairment of teratoma
insertion in exon 9) retina-specific formation
MAK isoform
expression
[40] adRP RHO, Rhodopsin [41] Apoptosis, ER Skin No Lentivirus RT-PCR 1 2 1 33D CRISPR/Cas9
(c.562G>A, p.G188R) stress, and rod fibroblast non-­MAK (~5W) reported in
degeneration RP Burnight,
control Gupta [42]
[43] adRP RHO, Rhodopsin (p. [44] Apoptosis and ER Skin Yes Retrovirus IF, teratoma 2 2 1 isogenic 5W Rapamycin,
E181K) stress; antioxidant fibroblast formation mutant PP242,
treatment line/1 AICAR,
corrected NQDI-1, and
control salubrinal/
line HDAdV
correction
3D culture
[45] arRP USH2A, Usherin (IVS40, [39] Protein misfolding Keratinocyte No Sendai virus RT-PCR, 1 2 1 150D –
p.Arg4192His) and ER stress; teratoma non-­ (~21W)
transplantation formation USH2A
control
Latest
Differentiation iPSC cell Reprogramming iPSC time Treatment/
Reference M.O.I. Mutated gene (mutation) protocol Major findings source Feeder approach characterization #Patient #Control point rescue
[46] arRP REEP6, receptor [47] Identified novel RP Skin No Episomal vector Not applicable 0 1 – 21W –
expression enhancer gene and retina- fibroblast
protein 6 (various, specific isoform
include p.Pro128Leu, p.
Leu135Pro, and p.
Val187Glyfs∗13)
[48] xlRP RP2, retinitis pigmentosa [47] Mislocalization of Skin No Episomal vector IF 1 1 – 21W TRIDs: G418
2 (c.519C>T, p.R120X) ciliary tip kinesins; fibroblast and PTC124
cilia defect; rescue
phenotypes with
TRIDs
[5] arRP TRNT1, tRNA [20] Autophagy defect; Skin No Sendai virus RT-PCR, IF, 3 1 Age-­ 90D –
nucleotidyl transferase, oxidative stress fibroblast karyotypic matched (~13W)
CCA-adding 1 (various, analysis, control
include TaqMan
c.127_129delGAA, scorecard assay
p.Glu43del3 GAA and
c.1252delA,
p.Ser418del1Afs)
[49] xlRP RPGR, retinitis [50] Actin Skin No Episomal vector qPCR, IF, 2 1 Patients 100D Overexpression
pigmentosa GTPase polymerization fibroblast spontaneous are (~14W) of gelsolin in
regulator defect differentiation brothers, hTERT-RPE
(g.ORF15+689−692del4) control is cells
unaffected
son/
nephew
(sex-
matched)

(continued)
Table 1 (continued)
Latest
Differentiation iPSC cell Reprogramming iPSC time Treatment/
Reference M.O.I. Mutated gene (mutation) protocol Major findings source Feeder approach characterization #Patient #Control point rescue
[27] xlRP RPGR, retinitis [51] Aberrant expression Urinary cell/ No Lentivirus/ AP, IF, 3 3 – 82W CRISPR/Cas9
pigmentosa GTPase of key retinal genes; skin episomal vector spontaneous (one patient
regulator (Patient1: various PR, cilia, fibroblast differentiation line)
c.1685_1686delAT; and
Patient2 and 3: ORF15 electrophysiological
c.2234_2235delGA and defects; CRISPR/
c.2403_2404delAG) Cas9 rescue
[26] adRP PRPF31, pre-mRNA [51] Pre-mRNA splicing Skin No Sendai virus IF, FACS, 4 3 Age-­ 43W CRISPR/Cas9
processing factor 3 and cilia defects; fibroblast karyotypic matched (one patient
(c.1115_1125del11 cell stress; analysis, SNP control line)
heterozygous or CRISPR/Cas9 array,
c.522_527+10del rescue spontaneous
heterozygous) differentiation,
teratoma
formation
[52] arRP USH2A, Usherin [51] Morphological, PR Urinary cell No Sendai virus IF, karyotypic 1 1 Age- and 86D –
(c.8559-2A > G, and RPE defects; analysis, sex-­ (~12W)
c.9127_9129delTCC) cell stress spontaneous matched
differentiation control
[53] arRP CRB1, crumbs [18] Outer limiting Skin No Lentivirus IF, spontaneous 3 3 Sex-­ 240D –
homolog-1 (various, membrane fibroblast differentiation matched (~34W)
includes c.3122T > C p. disruption;
(Met1041Thr) optimization of
homozygote) AAV transduction
[33] arRP PDE6B, cGMP-­ [47] Late-onset model; Peripheral No Episomal vector AP, IF, 1 1 – 230D –
phosphodiesterase type 6 mislocalization of blood spontaneous (33W)
(c.694G > A PR; impaired mononuclear differentiation
homozygous) cGMP hydrolysis cell
[54] xlRP RP2, retinitis pigmentosa [47]/[18] Thinning of outer Skin No Episomal vector IF 3 1 1 isogenic 300D AAV (one
2 (c.358C>T, p.R120X) nuclear layer and fibroblasts mutant (43W) isogenic line)
cell death; AAV line
rescue
M.O.I. mode of inheritance, adRP autosomal dominant RP, arRP autosomal recessive RP, xlRP X-linked RP, AP alkaline phosphatase, IF immunofluorescence staining, D day, W week
Modeling Retinitis Pigmentosa with Patient-Derived iPSCs 561

2. Varying capabilities of iPSC to generate are diagnosed at around 35 years old [32].
retinal cells have been well documented. Accumulating evidence suggests that reti-
Cowan et al. [28] reported only 8 out of 23 nal organoids acquire a developed state at
iPSC lines successfully generated retinal cells about 30 weeks, beyond which no further
at day 100. The variation is largely attributable maturation can be achieved using current
to genetic variations between donors and/or methodologies [28]. Although early disease
the epigenetic profiles of iPSCs [29]. Further phenotypes have been successfully demon-
understanding of the variability in iPSC pro- strated within the organoid model in various
duction and developing means to bypass the instances (Table 1), this limited maturation
hurdle and to preselect iPSC with high retinal represents a major caveat for the modeling
differentiation potential would be hugely ben- of late-onset RP. To emphasize the impor-
eficial to streamline RP modeling studies. tance of timing, Gao et al. [33] detected dis-
3. Another source of variation comes from the in ease phenotype in patient retinal organoids
vitro retinal differentiation procedure. Most harboring PDE6H mutation only from day
protocols available rely on spontaneous occur- 180 of differentiation. Additional aberra-
rence of 3D retinal organoids [30] and/or tions relating to PDE6H function only
inconsistent microdissection [18] and scrap- became apparent on day 230. Numerous
ing techniques [31] of 3D and 2D cultures, approaches have been tested in neurodegen-
resulting in high variability among organoids. erative studies of non-retinal ­disease to cap-
Improving the consistency of organoid pro- ture or reproduce aging effects by deriving
duction and ensuring consistent differentia- iPSCs from older donors and through
tion pace could help reduce the confounding progerin overexpression [34], telomere
effects due to technical variations. shortening [35], and the exposure to stress
4. Greater understanding of each RP gene agents [36]. Future studies will need to fur-
function can better inform experimental ther advance the authenticity of iPSC-
designs, such as in recent studies of RPGR derived 3D retinal tissue models to realize
and RP2 in ciliogenesis and PRPH31 in pre- the potential of iPSC technology to under-
mRNA splicing [26, 27]. stand RP disease progression and to develop
5. Lastly, while the onset of RP can be as early and test therapies to prevent PR cell death
as 10 years of age, on average, RP patients (Fig. 1).

Fig. 1 Modeling retinitis pigmentosa with patient-derived induced pluripotent stem cells (iPSCs). (Figure was created
in BioRender.com)
562 Y. C. Leong and J. C. Sowden

Acknowledgments This work was supported by 18. Zhong X, et al. Generation of three-dimensional
GOSHCC, NewLife, and the NIHR GOSH BRC. retinal tissue with functional photoreceptors from
human iPSCs. Nat Commun. 2014;5:4047.
19. Ahmad I, et al. Recapitulating developmental
mechanisms for retinal regeneration. Prog Retin Eye
References Res. 2019;76:100824.
20. Wiley LA, et al. cGMP production of patient-specific
1. Hartong DT, et al. Retinitis pigmentosa. Lancet. iPSCs and photoreceptor precursor cells to treat retinal
2006;368(9549):1795–809. degenerative blindness. Sci Rep. 2016;6:30742.
2. Bravo-Gil N, et al. Unravelling the genetic basis 21. Zhang XH, et al. Off-target effects in CRISPR/Cas9-­
of simplex retinitis pigmentosa cases. Sci Rep. mediated genome engineering. Mol Ther Nucleic
2017;7:41937. Acids. 2015;4:e264.
3. Berger W, et al. The molecular basis of human retinal 22. Engle SJ, et al. Best practices for translational disease
and vitreoretinal diseases. Prog Retin Eye Res. modeling using human iPSC-derived neurons.
2010;29(5):335–75. Neuron. 2018;100(4):783–97.
4. Gao FJ, et al. Genetic and clinical findings in a large 23. Kim JH, et al. Report of the international stem cell
cohort of Chinese patients with suspected retinitis banking initiative workshop activity: current hurdles
Pigmentosa. Ophthalmology. 2019;126(11):1549–56. and progress in seed-stock banking of human
5. Sharma TP, et al. Patient-specific induced pluripotent pluripotent stem cells. Stem Cells Transl Med.
stem cells to evaluate the pathophysiology of TRNT1- 2017;6(11):1956–62.
associated retinitis pigmentosa. Stem Cell Res. 24. Haston KM, et al. Clinical trials in a dish: the
2017;21:58–70. potential of pluripotent stem cells to develop therapies
6. Artero Castro A, et al. Deciphering retinal diseases for neurodegenerative diseases. Annu Rev Pharmacol
through the generation of three dimensional stem Toxicol. 2016;56:489–510.
cell-derived organoids: concise review. Stem Cells. 25. Jin ZB, et al. Modeling retinal degeneration using
2019;37(12):1496–504. patient-specific induced pluripotent stem cells. PLoS
7. Kam JH, et al. Fundamental differences in patterns of One. 2011;6(2):e17084.
retinal ageing between primates and mice. Sci Rep. 26. Buskin A, et al. Disrupted alternative splicing
2019;9(1):12574. for genes implicated in splicing and ciliogenesis
8. Fletcher EL, et al. Animal models of retinal disease. causes PRPF31 retinitis pigmentosa. Nat Commun.
Prog Mol Biol Transl Sci. 2011;100:211–86. 2018;9(1):4234.
9. Winkler PA, et al. Large animal models of inherited 27. Deng WL, et al. Gene correction reverses ciliopathy
retinal degenerations: a review. Cells. 2020;9(4):882. and photoreceptor loss in iPSC-derived retinal
10. Rao KN, et al. Ciliopathy-associated protein CEP290 organoids from retinitis pigmentosa patients. Stem
modifies the severity of retinal degeneration due to loss Cell Rep. 2018;10(6):2005.
of RPGR. Hum Mol Genet. 2016;25(10):2005–12. 28. Cowan CS, et al. Cell types of the human retina
11. Ebermann I, et al. PDZD7 is a modifier of retinal and its organoids at single-cell resolution. Cell.
disease and a contributor to digenic Usher syndrome. 2020;182(6):1623–40. e34
J Clin Invest. 2010;120(6):1812–23. 29. Kilpinen H, et al. Corrigendum: common genetic
12. Walia S, et al. Discordant phenotypes in fraternal variation drives molecular heterogeneity in human
twins having an identical mutation in exon iPSCs. Nature. 2017;546(7660):686.
ORF15 of the RPGR gene. Arch Ophthalmol. 30. Gonzalez-Cordero A, et al. Recapitulation of human
2008;126(3):379–84. retinal development from human pluripotent stem
13. Haider NB, et al. Mapping of genetic modifiers of cells generates transplantable populations of cone
Nr2e3 rd7/rd7 that suppress retinal degeneration and photoreceptors. Stem Cell Rep. 2017;9(3):820–37.
restore blue cone cells to normal quantity. Mamm 31. Regent F, et al. A simple and efficient method
Genome. 2008;19(3):145–54. for generating human retinal organoids. Mol Vis.
14. Cruz NM, et al. Modifier genes as therapeutics: the 2020;26:97–105.
nuclear hormone receptor Rev Erb alpha (Nr1d1) 32. Tsujikawa M, et al. Age at onset curves of retinitis
rescues Nr2e3 associated retinal disease. PLoS One. pigmentosa. Arch Ophthalmol. 2008;126(3):337–40.
2014;9(1):e87942. 33. Gao ML, et al. Patient-specific retinal organoids
15. Takahashi K, et al. Induction of pluripotent stem cells recapitulate disease features of late-onset retinitis
from mouse embryonic and adult fibroblast cultures pigmentosa. Front Cell Dev Biol. 2020;8:128.
by defined factors. Cell. 2006;126(4):663–76. 34. Zhang Y, et al. Author correction: hypothalamic stem
16. Takahashi K, et al. Induction of pluripotent stem cells cells control ageing speed partly through exosomal
from adult human fibroblasts by defined factors. Cell. miRNAs. Nature. 2018;560(7719):E33.
2007;131(5):861–72. 35. Vera E, et al. Generating late-onset human iPSC-
17. Rowe RG, et al. Induced pluripotent stem cells in based disease models by inducing neuronal age-
disease modelling and drug discovery. Nat Rev Genet. related phenotypes through telomerase manipulation.
2019;20(7):377–88. Cell Rep. 2016;17(4):1184–92.
Modeling Retinitis Pigmentosa with Patient-Derived iPSCs 563

36. Liu Y, et al. Protective effect of hydrogen on sodium 45. Tucker BA, et al. Patient-specific iPSC-derived
iodate-induced age-related macular degeneration in photoreceptor precursor cells as a means to investigate
mice. Front Aging Neurosci. 2018;10:389. retinitis pigmentosa. elife. 2013;2:e00824.
37. Osakada F, et al. Toward the generation of rod and 46. Arno G, et al. Mutations in REEP6 cause autosomal-­
cone photoreceptors from mouse, monkey and recessive retinitis pigmentosa. Am J Hum Genet.
human embryonic stem cells. Nat Biotechnol. 2016;99(6):1305–15.
2008;26(2):215–24. 47. Nakano T, et al. Self-formation of optic cups and
38. Tucker BA, et al. Exome sequencing and analysis storable stratified neural retina from human ESCs.
of induced pluripotent stem cells identify the cilia-­ Cell Stem Cell. 2012;10(6):771–85.
related gene male germ cell-associated kinase (MAK) 48. Schwarz N, et al. Arl3 and RP2 regulate the
as a cause of retinitis pigmentosa. Proc Natl Acad Sci trafficking of ciliary tip kinesins. Hum Mol Genet.
U S A. 2011;108(34):E569–76. 2017;26(17):3451.
39. Tucker BA, et al. Transplantation of adult mouse iPS 49. Megaw R, et al. Gelsolin dysfunction causes
cell-derived photoreceptor precursors restores retinal photoreceptor loss in induced pluripotent cell and
structure and function in degenerative mice. PLoS animal retinitis pigmentosa models. Nat Commun.
One. 2011;6(4):e18992. 2017;8(1):271.
40. Jin ZB, et al. Integration-free induced pluripotent 50. Mellough CB, et al. IGF-1 signaling plays an
stem cells derived from retinitis pigmentosa patient important role in the formation of three-dimensional
for disease modeling. Stem Cells Transl Med. laminated neural retina and other ocular structures
2012;1(6):503–9. from human embryonic stem cells. Stem Cells.
41. Osakada F, et al. Stepwise differentiation of 2015;33(8):2416–30.
pluripotent stem cells into retinal cells. Nat Protoc. 51. Kuwahara A, et al. Generation of a ciliary margin-like
2009;4(6):811–24. stem cell niche from self-organizing human retinal
42. Burnight ER, et al. Using CRISPR-Cas9 to generate tissue. Nat Commun. 2015;6:6286.
gene-corrected autologous iPSCs for the treatment 52. Guo Y, et al. Modeling retinitis pigmentosa: retinal
of inherited retinal degeneration. Mol Ther. organoids generated from the iPSCs of a patient
2017;25(9):1999–2013. with the USH2A mutation show early developmental
43. Yoshida T, et al. The use of induced pluripotent stem abnormalities. Front Cell Neurosci. 2019;13:361.
cells to reveal pathogenic gene mutations and explore 53. Quinn PM, et al. Human iPSC-derived retinas
treatments for retinitis pigmentosa. Mol Brain. recapitulate the fetal CRB1 CRB2 complex
2014;7:45. formation and demonstrate that photoreceptors and
44. Lamba DA, et al. Generation, purification and Muller glia are targets of AAV5. Stem Cell Rep.
transplantation of photoreceptors derived from 2019;12(5):906–19.
human induced pluripotent stem cells. PLoS One. 54. Lane A, et al. Modeling and rescue of RP2 retinitis
2010;5(1):e8763. pigmentosa using iPSC-derived retinal organoids.
Stem Cell Rep. 2020;15(1):67–79.
Primary Retinal Cell Cultures
as a Model to Study Retina Biology

Germán A. Michelis, Luis E. Politi,


and S. Patricia Becerra

Abstract Keywords

Since its inception, primary retinal cultures Retina · Primary cultures · Cell lines ·
have been an in vitro tool for modeling the Organoids · In vivo models ·
in vivo environment of the retina for biologi- In vitro models
cal studies on development and disease. They
offer simple and controlled experimental
approaches when compared to in vivo models. 1 Introduction
In this review we highlight the strengths and
weaknesses of primary retinal culture models, The selection of a relevant model that best
and the features of dispersed retinal cell resembles the in vivo organism is key for suc-
cultures. cessful research. The use of in vivo animal mod-
els to study the retina is desirable given that they
contain the whole organ. However, they have
several caveats, such as their degree of com-
plexity and interdependence on other organ sys-
tems that indirectly affect the outcome of the
G. A. Michelis
assays targeting the retina. Given the naturally
Section of Protein Structure and Function, LRCMB,
NEI-NIH, occurring variation among individuals, experi-
Bethesda, MD, USA mental designs tend to include a high number of
Department of Biology, Pharmacy and Biochemistry, animals, which in turn impacts on the cost of
Instituto de Investigaciones Bioquímicas (INIBIBB), resources and manpower to maintain an animal
Universidad Nacional del Sur (UNS)-CONICET, colony, as well as on ethical concerns to reduce,
Bahía Blanca, Argentina
refine, and replace the use of research animals
L. E. Politi [29]. The use of in cellulo models (e.g., dissoci-
Department of Biology, Pharmacy and Biochemistry,
ated cells from a tissue and cell lines) offers sev-
Instituto de Investigaciones Bioquímicas (INIBIBB),
Universidad Nacional del Sur (UNS)-CONICET, eral advantages. The in vitro environment allows
Bahía Blanca, Argentina for additional experimental options over those
S. P. Becerra (*) for the in vivo models, given their controlled
Section of Protein Structure and Function, LRCMB, culturing conditions, which are more difficult to
NEI-NIH, achieve with live animals.
Bethesda, MD, USA
e-mail: becerras@nei.nih.gov

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 565
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_82
566 G. A. Michelis et al.

2 Cell Lines retinal cultures have existed for over five


decades [18, 24].
Retinal cell lines arise from primary cell cultures The use of primary retinal cell cultures in
and are used instead of them to study biological experimental design offers several advantages.
processes. Examples include rat R28 [30], human (1) Cell culture quality control, which refers to
ARPE-19 [4], and mouse 661W [28] cell lines. measures taken to control the quality of the
They are composed of a single cell type with low reagents and cells during the maintenance of
internal variability. However, care must be taken these cellular models. (2) Reproducibility, in that
when interpreting the results because cell lines the experimental variables are more reproducible
are transformed to propagate indefinitely, gener- and easier to replicate with cells in culture than
ally with oncogenic pathways that often generate in vivo. (3) Penetration of testing drugs to reach
artifacts that differ from the natural behavior of the target retinal cells in vitro without the risk of
cells in vivo. They can undergo genetic and phe- unintended effects caused by the ocular delivery
notypic changes due to the cumulative mutations itself in vivo. (4) Retinal cell identity. Generally,
with time in culture, causing deviation from the primary cultures are more comparable to in vivo
progenitor cell and requiring regular validation to than cell lines [35].
monitor their genetic variation. The extended life Great strides have been made to improve cul-
span of cell lines is beyond that of primary cells, ture conditions of cells to resemble the environ-
anticipating artificial death induction when lines ment in the retinal tissue in vivo. These advances
are used in retinal cell survival assays [3]. These have allowed the development of organoids that
events in cell lines may not be extrapolated to can survive for almost a year in culture [14, 41].
in vivo outcomes and require additional verifica- Despite the progress in this field, research to
tion by other models. make retinal organoids equivalent to their in vivo
counterparts is ongoing [32, 40]. While the
advantage of the use of primary retinal cell cul-
3 Primary Retinal Cell Cultures tures as models for retinal studies is in their
application to investigate interactions at a subcel-
Primary cultures are prepared from retinal tis- lular and cell-to-cell levels, a disadvantage is that
sue of animals and have a limited life span [35]. there are processes or factors in vivo that are
They are a compromise between the in vivo likely missed in vitro [7]. For example, the lack
models and cell lines and provide a simplified of extracellular cues present in the whole organ-
environment of the retina, enabling the study of ism leads to a short life span in primary cells in
specific biological processes while guaranteeing culture [6].
the identity of the cell components [35]. While
these models overcome hurdles inherent to the
in vivo and cell line alternatives, other limita- 4 Dispersed Cell Cultures
tions arise that need attention at the planning
stage. Due to its clinical relevance and potential The preparation of dispersed cell cultures requires
as a model for the nervous system, the retina is cell dissociation from dissected retinal tissue
ideal to establish primary cell cultures. through mechanical or enzymatic means, fol-
Compared to the brain as a source of neurons, lowed by plating dissociated cells in a culture
the retina is relatively easy to access and has a dish. Table 1 lists primary retinal cell cultures
stratified architecture with a lower number of prepared from different species and by different
cell types [19], lending itself for modeling neu- laboratories. Figure 1 shows representative
ronal behavior [1, 2, 17]. The models of primary images of primary retinal cells in culture.
Primary Retinal Cell Cultures as a Model to Study Retina Biology 567

Table 1 Primary retinal cell cultures from different species

Animal species Retinal cell types in culture Reference


P8 mice Müller glial cells, photoreceptors Kretz et al. [12]
P2–4 Sprague-­Dawley rats Mixed: Glia, photoreceptors, neurons— Sia et al. [31]
Not retinal ganglion cells (RGCs)
P2–4 Sprague-­Dawley rats RGCs Sia et al. [31]
PN2 mice Photoreceptors, Amacrine cells Politi et al. [26]
E5–E14 chicken embryos RGCs Stanke and Fischer [33]
PN1 Long-­Evans rats Müller glial cells, Amacrine cells, Hicks and Courtois [9]
photoreceptors
PN4 C3H/HeA mice Photoreceptors, Amacrines cells, bipolar Zalis et al. [37]
cells, astrocytes, Müller glial cells
PN4–10 Sprague-­Dawley rats RGCs Fischer et al. [5]
E18 Sprague-­Dawley rats Retinal precursors, converted to RGCs James et al. [11]
E3 chickens Retinal precursors James et al. [11]
Adult Sprague-­Dawley rats or adult RGCs Grozdanov et al. [8]
C57/BL6 mice
E18 Wistar rats Photoreceptors, GABAergic neurons, Matteucci et al. [20]
Müller glial cells
Adult human eyes Müller glia, rod photoreceptors Hicks et al. [10]
PN3–5 Hooded-Lister rats RGCS Lilley and Robbins [17]
PN1 Sprague-­Dawley rats RGCs Li et al. [16]
Stage 25+ Xenopus embryos Retinal progenitors McDonough et al. [21]
10–12 weeks human embryos RGCs Zhang et al. [39]
16–18 weeks human embryos Retinal progenitors Wang et al. [36]
Adult tiger salamanders Müller glial cells, photoreceptors Kung et al. [13]

E18 Sprague-­Dawley rats Photoreceptors, Amacrine cells, RGCs Levine et al. [15]
PN22–25 Long-Evans rats Photoreceptors Zayas-­Santiago and Kang
Derwent [38]
Adult zebrafish Photoreceptors, bipolar cells, horizontal McMahon [22]
cells
E17–21 and PN2 Wistar-­Albino rats RGCs Turner [34]
PN1 Sprague-­Dawley rats RGCs Sarthy et al. [27]
PN2 Dutch rabbits Amacrine cells Osborne et al. [25]

5 Future Directions Retinal organoids are gaining increasing attention


by the scientific community given their potential
Primary cultures are mainstay models in laborato- of creating a more accurate representation of the
ries worldwide that will continue to be valuable retina. Retinal organoids and primary cell cultures
for the advancement of our scientific knowledge will prove to be complementary in vitro models to
regarding retina physiology and pathology. address different aspects of scientific questions.
568 G. A. Michelis et al.

Fig. 1 Photomicrographs of dispersed primary rat retinal were labeled with DAPI (blue) and TOPRO-3 (red).
cells at 5 days in culture prepared according to Michelis Neurites were labeled with anti-acetylated tubulin anti-
et al. [23]. Two cellular types are present: photoreceptors body (green)
(unmarked) and amacrine neurons (asterisks). Nuclei

2. Ames A, Nesbett FB. In vitro retina as an experimen-


For example, primary cell cultures serve to answer tal model of the central nervous system. J Neurochem.
1981;37:867–77.
questions associated with subcellular or cell-to-
3. Bullock J, Pagan-Mercado G, Becerra SP. Cell-based
cell events, while tissue-to-tissue interactions are assays to identify novel retinoprotective agents.
better investigated in organoids. Thus, combining MethodsX. 2020;7:101026.
the in vitro models of primary cell cultures and 4. Dunn KC, Aotaki-Keen AE, Putkey FR, Hjelmeland
LM. ARPE-19, a human retinal pigment epithelial
organoids in a complementary and synergistic
cell line with differentiated properties. Exp Eye Res.
fashion will result in a powerful tool to advance 1996;62:155–69.
our knowledge about retina development under 5. Fischer RA, Zhang Y, Risner ML, Li D, Xu Y,
normal or pathological conditions. Sappington RM. Impact of graphene on the efficacy
of neuron culture substrates. Adv Healthc Mater.
2018;7:e1701290.
Acknowledgments This work was supported in part by 6. Freshney RI. Preparation and sterilization. In: Culture
the Intramural Research Program of the National Eye of animal cells: a manual of basic technique. 5th ed.
Institute, NIH (Project #EY000306, for S.P.B.), the New York: Wiley; 2005. p. 145–73.
National Agency for Science and Technology, Argentina 7. Ghareeb AE, Lako M, Steel DH. Coculture techniques
(PICT-2016-0353, for L.E.P.), and the National Research for modeling retinal development and disease and
Council of Argentina (PIP 1­ 1220-­1101-­00827, for L.E.P.). enabling regenerative medicine. Stem Cells Transl
We thank Dr. Ivan Rebustini for carefully proof-reading Med. 2020;9:1531–48.
the manuscript. 8. Grozdanov V, Müller A, Sengottuvel V, Leibinger M,
Fischer D. A method for preparing primary retinal cell
cultures for evaluating the neuroprotective and neuri-
togenic effect of factors on axotomized mature CNS
References neurons. Curr Protoc Neurosci. 2010;53:1. Chapter
3:Unit 3.22.
1. Agranoff BW, Feldman EL, Heacock AM, Schwartz 9. Hicks D, Courtois Y. Fibroblast growth factor stimu-
M. The retina as a biochemical model of cen- lates photoreceptor differentiation in vitro. J Neurosci.
tral nervous system regeneration. Neurochem Int. 1992;12:2022–33.
1980;1C:487–500.
Primary Retinal Cell Cultures as a Model to Study Retina Biology 569

10. Hicks D, Forster V, Dreyfus H, Sahel J. Survival and 27. Sarthy PV, Curtis BM, Catterall WA. Retrograde
regeneration of adult human photoreceptors in vitro. labeling, enrichment, and characterization of reti-
Brain Res. 1994;643:302–5. nal ganglion cells from the neonatal rat. J Neurosci.
11. James J, Das AV, Bhattacharya S, Chacko DM, 1983;3:2532–44.
Zhao X, Ahmad I. In vitro generation of early-born 28. Sayyad Z, Sirohi K, Radha V, Swarup G. 661W is
neurons from late retinal progenitors. J Neurosci. a retinal ganglion precursor-like cell line in which
2003;23:8193–203. glaucoma-­associated optineurin mutants induce cell
12. Kretz A, Marticke JK, Happold CJ, Schmeer C, death selectively. Sci Rep. 2017;7:16855.
Isenmann S. A primary culture technique of adult 29. Schnichels S, Paquet-Durand F, Löscher M, Tsai T,
retina for regeneration studies on adult CNS neurons. Hurst J, Joachim SC, Klettner A. Retina in a dish:
Nat Protoc. 2007;2:131–40. cell cultures, retinal explants and animal models for
13. Kung F, Wang W, Tran TS, Townes-Anderson common diseases of the retina. Prog Retin Eye Res.
E. Sema3A reduces sprouting of adult rod pho- 2021;81:100880.
toreceptors in vitro. Invest Ophthalmol Vis Sci. 30. Seigel GM. Review: R28 retinal precursor cells: the
2017;58:4318–31. first 20 years. Mol Vis. 2014;20:301–6.
14. Lancaster MA, Knoblich JA. Organogenesis in a dish: 31. Sia PI, Wood JPM, Chidlow G, Casson R. Creatine
modeling development and disease using organoid is neuroprotective to retinal neurons in vitro but not
technologies. Science. 2014;345:1247125. in vivo. Invest Ophthalmol Vis Sci. 2019;60:4360–77.
15. Levine EM, Roelink H, Turner J, Reh TA. Sonic 32. Singh RK, Nasonkin IO. Limitations and promise
hedgehog promotes rod photoreceptor ­differentiation of retinal tissue from human pluripotent stem cells
in mammalian retinal cells in vitro. J Neurosci. for developing therapies of blindness. Front Cell
1997;17:6277–88. Neurosci. 2020;14:179.
16. Li N, Shang L, Wang SC, Liao LS, Chen D, Huang 33. Stanke JJ, Fischer AJ. Embryonic retinal cells and
JF, Xiong K. The toxic effect of ALLN on primary support to mature retinal neurons. Invest Ophthalmol
rat retinal neurons. Neurotox Res. 2016;30:392–406. Vis Sci. 2010;51:2208–18.
17. Lilley S, Robbins J. The rat retinal ganglion cell in 34. Turner JE. Promotion of neurite outgrowth and cell
culture: an accessible CNS neurone. J Pharmacol survival in dissociated fetal rat retinal cultures by
Toxicol Methods. 2005;51:209–20. a fraction derived from a brain extract. Brain Res.
18. Liss L, Wolter JR. Human retinal neurons in tissue 1985;350:265–74.
culture. Am J Ophthalmol. 1961;52:834–41. 35. Uysal O, Sevimli T, Sevimli M, Gunes S, Eker
19. Masland RH. The neuronal organization of the retina. SA. Cell and tissue culture: the base of biotechnology.
Neuron. 2012;76:266–80. In: Omics technologies and bio-engineering: towards
20. Matteucci A, Varano M, Gaddini L, Mallozzi C, Villa improving quality of life. London: Academic; 2018.
M, Pricci F, Malchiodi-Albedi F. Neuroprotective p. 391–426.
effects of citicoline in in vitro models of retinal neu- 36. Wang Z, Gao F, Zhang M, Zheng Y, Zhang F, Xu L,
rodegeneration. Int J Mol Sci. 2014;15:6286–97. Cao L, He W. Intravitreal injection of human retinal
21. McDonough MJ, Allen CE, Ng-Sui-Hing NK, Rabe progenitor cells for treatment of retinal degeneration.
BA, Lewis BB, Saha MS. Dissection, culture, and Med Sci Monit. 2020;26:e921184.
analysis of Xenopus laevis embryonic retinal tissue. 37. Zalis MC, Johansson S, Englund-Johansson
J Vis Exp. 2012;70:4377. U. Immunocytochemical profiling of cultured
22. McMahon DG. Modulation of electrical synaptic mouse primary retinal cells. J Histochem Cytochem.
transmission in zebrafish retinal horizontal cells. J 2017;65:223–39.
Neurosci. 1994;14:1722–34. 38. Zayas-Santiago A, Kang Derwent JJ. Preservation of
23. Michelis G, German OL, Villasmil R, Soto T, Rotstein intact adult rat photoreceptors in vitro: study of dis-
NP, Politi L, Becerra SP. Pigment epithelium-derived sociation techniques and the effect of light. Mol Vis.
factor (PEDF) and derived peptides promote survival 2009;15:1–9.
and differentiation of photoreceptors and induce 39. Zhang XM, Li Liu DT, Chiang SW, Choy KW, Pang
neurite-­outgrowth in amacrine neurons. J Neurochem. CP, Lam DS, Yam GH. Immunopanning purification
2021;159:840. and long-term culture of human retinal ganglion cells.
24. Moscona A. Formation of lentoids by dissoci- Mol Vis. 2010;16:2867–72.
ated retinal cells of the chick embryo. Science. 40. Zhang Z, Xu Z, Yuan F, Jin K, Xiang M. Retinal
1957;125:598–9. organoid technology: where are we now? Int J Mol
25. Osborne NN, Beaton DW, Vigny A, Neuhoff Sci. 2021;22:10244.
V. Localization of tyrosine-hydroxylase immunore- 41. Zhong X, Gutierrez C, Xue T, Hampton C, Vergara
active cells in rabbit retinal cultures. Neurosci Lett. MN, Cao LH, Peters A, Park TS, Zambidis ET, Meyer
1984;50:117–20. JS, Gamm DM, Yau KW, Canto-Soler MV. Generation
26. Politi LE, Lehar M, Adler R. Development of neo- of three-dimensional retinal tissue with functional
natal mouse retinal neurons and photoreceptors in photoreceptors from human iPSCs. Nat Commun.
low density cell culture. Invest Ophthalmol Vis Sci. 2014;5:4047.
1988;29:534–43.
Generation of CRB1 RP
Patient-­Derived iPSCs
and a CRISPR/Cas9-­Mediated
Homology-Directed Repair
Strategy for the CRB1 c.2480G>T
Mutation

Bruna Lopes da Costa, Yao Li, Sarah R. Levi,


Stephen H. Tsang, and Peter M. J. Quinn

Abstract CRB1-associated diseases. This phenotypic


modulation may be due to several factors,
Mutations in the Crumbs-homologue-1 including genetic modifiers, deep intronic
(CRB1) gene lead to a spectrum of severe mutations, isoform diversity, and copy number
inherited retinal diseases, including retinitis variations. Induced pluripotent stem cell
pigmentosa (RP). The establishment of a gen- (iPSC)-derived patient retinal organoids are
otype–phenotype correlation in CRB1 patients novel tools that can provide sensitive,
has been difficult due to the substantial vari- quantitative, and scalable phenotypic assays.
ability and phenotypic overlap between CRB1 RP patient iPSC-derived retinal

B. L. da Costa
Department of Biomedical Engineering, Columbia
University, New York, NY, USA
Edward S. Harkness Eye Institute, Department of S. H. Tsang
Ophthalmology, Columbia University Irving Medical Department of Biomedical Engineering, Columbia
Center/New York-Presbyterian Hospital, University, New York, NY, USA
New York, NY, USA
Edward S. Harkness Eye Institute, Department of
Jonas Children’s Vision Care, and Bernard & Shirlee Ophthalmology, Columbia University Irving Medical
Brown Glaucoma Laboratory, Department of Center/New York-Presbyterian Hospital,
Ophthalmology, Columbia University, New York, NY, USA
New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee
Y. Li · S. R. Levi · P. M. J. Quinn (*) Brown Glaucoma Laboratory, Department of
Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University,
Ophthalmology, Columbia University Irving Medical New York, NY, USA
Center/New York-Presbyterian Hospital,
Columbia Stem Cell Initiative, Columbia University,
New York, NY, USA
New York, NY, USA
Jonas Children’s Vision Care, and Bernard & Shirlee
Department of Pathology & Cell Biology, Columbia
Brown Glaucoma Laboratory, Department of
University, New York, NY, USA
Ophthalmology, Columbia University,
New York, NY, USA Institute of Human Nutrition, Columbia University,
e-mail: pq2138@cumc.columbia.edu New York, NY, USA

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 571
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_83
572 B. L. da Costa et al.

organoids have shown reproducible derived retinal organoids have shown reproduc-
phenotypes compared to healthy retinal ible phenotypes compared to health control
organoids. However, having genetically retinal organoids [4]. However, it remains crucial
defined iPSC isogenic controls that take into to account for potential phenotypic modulation in
account potential phenotypic modulation is these iPSC isogenic controls.
crucial. In this study, we generated iPSC from In this study, we generated iPSCs from a
an early-onset CRB1 patient and developed a CRB1 early-onset RP patient and describe a
correction strategy for the c.2480G>T, p. clustered-­regularly interspaced short palindromic
(Gly827Val) CRB1 mutation using CRISPR/ repeats (CRISPR)/CRISPR-associated (Cas)-
Cas9-mediated homology-­directed repair. mediated homology-directed repair (HDR) strat-
egy for the correction of the c.2480G>T, p.
Keywords (Gly827Val) CRB1 mutation.
Crumbs-homologue-1 (CRB1) · Gene editing
· Homology-directed repair (HDR) · Retinitis
pigmentosa (RP) · iPSC-derived retinal 2 Materials and Methods
organoids
2.1 Clinical Evaluation

A male patient with early-onset RP presented to


1 Introduction the Edward S. Harkness Eye Institute at Columbia
University (New York, NY) for examination. The
Mutations in the Crumbs-homologue-1 (CRB1) patient’s diagnosis was genetically confirmed,
gene cause a range of inherited retinal dystro- revealing compound heterozygote mutations
phies (IRDs) that lead to visual impairment or c.2480G>T, p.(Gly827Val) and c.2843G>A, p.
even complete blindness. Approximately 80,000 (Cys948Tyr) in the CRB1 gene. The study was
patients are affected worldwide, with no treat- conducted under Columbia University
ment to date. CRB1-associated IRDs exhibit high Institutional Review Board approval (IRB
phenotypic variability and despite more than 489 AAAF1849) and all procedures were performed
different mutations being identified in the CRB1 according to the tenets of the Declaration of
gene (http://www.LOVD.nl/CRB1), there is still Helsinki. Informed consent was gathered accord-
no clear genotype–phenotype correlation [5]. ing to IRB AAAF1849. Lidocaine was adminis-
Notably, one lab in particular, the Sallum lab, tered to a small region of the skin on the lower
was able to identify a direct relationship between back, and a biopsy skin punch (McKesson,
phenotype severity and the mutation effect on Virginia) was performed.
protein functionality in a small Brazilian CRB1
cohort. However, the phenotypic modulation
seen in CRB1-associated IRDs may be due to 2.2 Cell Culture
several genetic factors, including modifiers, deep
intronic mutations, isoform diversity, and copy Fibroblasts from the CRB1 RP patient were
number variations [3]. reprogrammed into iPSCs using the CytoTune-­
Induced pluripotent stem cell (iPSC)-derived iPS 2.0 Sendai Reprogramming Kit (Thermo
patient retinal organoids can provide sensitive, Fisher Scientific). The generated iPSCs were
quantitative, and scalable phenotypic assays [2]. tested for their quality and pluripotency by stem
Mutations in CRB1 can cause retinitis pigmen- cell marker staining and karyotyping, as previ-
tosa type 12 (RP12), which is characterized by ously described [1, 6]. All iPSC lines were pas-
preservation of para-arteriolar retinal pigment saged using ReleSR (STEMCELL Technologies)
epithelium and progressive visual field loss from every 3–6 days, while being maintained in
early childhood [5]. CRB1 RP patient iPSC-­ mTeSR-plus medium (STEM CELL
Generation of CRB1 RP Patient-Derived iPSCs and a CRISPR/Cas9-Mediated Homology-Directed Repair… 573

Technologies). HEK293 cells were cultured single guide RNAs (sgRNAs) (sgRNA1:
using Dulbecco’s modified Earl’s medium 5 ′ - T T C TA C G T G G A A A AT C G A A A -­3 ′ ,
(Gibco), supplemented with 10% fetal bovine sgRNA2: 5′-GTAGATGTCATCTACATT
serum (Gibco), 1% Glutamax, and 1% (v/v) peni- GG-3′). The sgRNA’s were tested using an
cillin/streptomycin (Gibco). in vitro cleavage assay: Combined 125 ng
sgRNA, 500 ng Cas9, 200 ng template (amplified
HEK293 DNA using primers for the c.2480G>T,
2.3 DNA Extraction and PCR p.(Gly827Val) locus) and incubated at 37 °C for
Primers 4 h and subsequently cleavage products separated
on 2% agarose gel. CRB1 patient iPSCs and
The cells were detached from the wells by HEK293 cells were nucleofected with the P3
trypsin. The cells from each well were washed Primary Cell 4D-Nucleofector X Kit (Lonza) on
with Dulbecco’s phosphate-buffered saline an Amaxa Nucleofector 4D using the DS150 pro-
(DPBS) (without Ca2+ and Mg2+) and resuspended gram, according to the manufacturer’s instruc-
with 50 μL DPBS. The cells were then incubated tions. Briefly, 1 × 105 cells was resuspended in a
at 95 °C for 20 min. Subsequently, after cooling, 20 μL electroporation solution (16.4 μL of the P3
4 μL of 20 mg/mL Proteinase K (Promega) was nucleofector solution and 3.6 μL of the supple-
added to each sample. The samples were then ment) for each reaction, and subsequently 4 μM
incubated at 56 °C for 1 h followed by a 30-min single-stranded oligo donor (ssODN), 1.2 μL of
incubation at 95 °C to stop the Proteinase K Alt-R HDR Enhancer, and 5 μL of ribonucleo-
digestion. This crude extract of DNA is then protein mixture, which consists of 125 pmol
ready for PCR purpose. Primers used for PCR at Cas9 protein and 150 pmol gRNA, were added
the c.2480G>T, p.(Gly827Val) locus and transferred to nucleocuvette well. After
(Forward: 5′-GCTAGAGCGCG- nucleofection, 75 μL of prewarmed cell-specific
GCAGACTAG-3′,Reverse:5′-TGGATTGAGAGA- culture medium (DMEM: HEK293, mTeSR plus:
TGCATTGTTTGTTGG-­3′) and c.2843G>A, p. iPSCs) was added and subsequently cells were
(Cys948Tyr) locus (Forward: transferred to one matrigel-coated well (matrigel
5′-TTGGCATGCATTAGGATTTCACT GGA- coating only for iPSCs) of a 96-well plate con-
3′, Reverse: 5′-CCATCATTCACTGACT- taining 100 μL of cell-specific culture medium
GCAAACTTGT-­3′). with a further 2 μL of Alt-R HDR Enhancer.
Twenty-four hours after nucleofection, medium
was removed and fresh medium without Alt-R
2.4 CRISPR-Mediated Gene HDR Enhancer was added. mTeSR-plus medium
Correction with 10 μM ROCK inhibitor (Selleck Chemical)
was used for the iPSCs. Five days after nucleo-
For HDR of the c.2480G>T, p.(Gly827Val) fection, the cells were collected and genomic
CRB1 mutation, we used the Alt-R CRISPR-­ DNA extracted. Hpy188III (New England
Cas9 System and HDR donor oligo from Biolabs) was used to carry out restriction frag-
Integrated DNA Technologies (IDT): Alt-R S.p. ment length polymorphism (RFLP) assay. To
Cas9 Nuclease V3, Alt-R HDR Enhancer, Alt-R determine editing efficiency, we amplified the
HDR Oligo (5′-CAGTCTTCACAAA- CRB1 c.2480G>T, p.(Gly827Val) locus using
ACCTAGGATTTATTT CTG-CTTCTA- primers, performed dideoxy sequencing, and
CGTGGAAAATCGAG-AAAGGAGATGTCATC- finally determined editing efficiency by ICE
TACATTGGTG-GCCTACCTGACAAGCAA- Analysis: https://ice.synthego.com/#/.
GAGACTG-AACTTA-­3′), Alt-R CRISPR-Cas9
574 B. L. da Costa et al.

3 Results and Discussion tion in chromosome 20, which could incur a pro-
liferative advantage and therefore this clone was
Mutations in the CRB1 gene lead to a spectrum of not taken forward for subsequent analysis.
IRDs [5]. In this study, we reprogrammed fibro- Further, through immunostaining, we confirmed
blasts from a CRB1 patient with early-onset RP that clones 2 and 4 were pluripotent, using mark-
to generate several iPSC clones and confirmed by ers Nanog and Oct3/4 (Fig. 1c), and that they
dideoxy sequencing that they were compound could form all three germ layers: Ectoderm
heterozygote for the c.2480G>T, p.(Gly827Val) (Nestin and Pax6), Mesoderm (Brachyury and
and c.2843G>A, p.(Cys948Tyr) CRB1 mutations NCAM), and Endoderm (FOX2A and Sox17)
(Fig. 1a). Upon verifying the iPSC clones’ (Fig. 1d).
genomic integrity using G-band karyotyping, we Upon validation of these lines, we sought to
found that clones 2 and 4 had a normal karyotype develop isogenic controls using CRISPR/Cas9-­
(Fig. 1b). However, in clone 3, a small population mediated HDR. It is, however, crucial to account
of the iPSCs were found to have a q11.2 duplica- for potential phenotypic modulation within these

Fig. 1 Generation and validation of CRB1 retinitis ing. (c) Pluripotency of iPSC lines was confirmed by
pigmentosa (RP) patient iPSCs. (a) Dideoxy sequencing immunohistochemistry using markers Oct3/4 and Nanog.
confirmed that iPSC clones established from a RP patient (d) Germ layer potential of iPSC was confirmed by immu-
with compound heterozygote mutations c.2480G>T, p. nohistochemistry: Ectoderm (Nestin and Pax6),
(Gly827Val) and c.2843G>A, p.(Cys948Tyr) in the CRB1 Mesoderm (Brachyury and NCAM), and Endoderm
gene had the expected genotype. (b) To verify genome (FOX2A and Sox17). Scale bars, 50 μm
integrity, iPSC lines were analyzed by G-band karyotyp-
Generation of CRB1 RP Patient-Derived iPSCs and a CRISPR/Cas9-Mediated Homology-Directed Repair… 575

Fig. 2 CRISPR/Cas9-mediated homology-directed of ssODN in bulk edited HEK293 cells. In these cells, we
repair (HDR) strategy for the correction of the c.2480G>T, introduced silent mutations to insert Hpy188III, for RFLP
p.(Gly827Val) CRB1 mutation. (a) Schematic of HDR assay, and also to mutate the PAM from sgRNA1. (d)
correction strategy for the c.2480G>T, p.(Gly827Val) RFLP assay in bulk edited HEK293 cells shows success-
CRB1 mutation. (b) In vitro cleavage assay illustrating the ful cleavage. (e) ICE analysis showing a 65% editing effi-
cutting of sgRNA’s 1 and 2. (c) Successful incorporation ciency in bulk edited HEK293 cells

controls, especially due to the high phenotypic Hpy188III for screening of future single clones
variability seen in CRB1-associated IRDs [3, 5]. by RFLP. Further, a silent mutation was intro-
We designed a HDR approach for the correction duced in the protospacer adjacent motif (PAM)
of the c.2480G>T, p.(Gly827Val) CRB1 mutation for sgRNA1 to prevent Cas9 from recutting the
(Fig. 2a). We tested two possible sgRNAs via genomic site. Successfully editing in bulk edited
in vitro cleavage assay (Fig. 2b). SgRNA1 was HEK293 cells was verified 5 days after nucleo-
taken forward and used in conjugation with a fection by dideoxy sequencing (Fig. 2c), RFLP
106-bp ssODN, which included the wild-type (Fig. 2d), and ICE analysis (Fig. 2e). ICE analy-
c.2480G and a silent mutation to introduce sis showed a 65% editing efficiency in bulk
576 B. L. da Costa et al.

edited HEK293 cells. Initial analysis on the bulk References


edited patient iPSCs showed 5% editing effi-
ciency by ICE analysis but no cleavage products 1. Li Y, Tsai YT, Hsu CW, Erol D, Yang J, Wu WH, Davis
RJ, Egli D, Tsang SH. Long-term safety and efficacy
could be seen by RFLP (data not shown). Our of human-induced pluripotent stem cell (iPS) grafts in
next steps are to generate single clone isogenic a preclinical model of retinitis pigmentosa. Mol Med.
controls. Together, the work outlined in this study 2012;18:1312–9.
creates a platform for the thorough analysis of 2. Manafi N, Shokri F, Achberger K, Hirayama M,
Mohammadi MH, Noorizadeh F, Hong J, Liebau S,
CRB1 RP patient iPSC-derived retinal Tsuji T, Quinn PMJ, Mashaghi A. Organoids and organ
organoids. chips in ophthalmology. Ocul Surf. 2020;19:1–15.
3. Motta FL, Salles MV, Costa KA, Filippelli-Silva R,
Acknowledgments S.H.T. and The Jonas Children’s Martin RP, Sallum JMF. The correlation between
Vision Care and Bernard & Shirlee Brown Glaucoma CRB1 variants and the clinical severity of Brazilian
Laboratory are supported by the National Institutes of patients with different inherited retinal dystrophy
Health (P30EY019007, R01EY018213, R01EY024698, phenotypes. Sci Rep. 2017;7:8654.
R01EY026682, R24EY027285, U01EY030580), 4. Quinn PM, Buck TM, Mulder AA, Ohonin C, Alves
National Cancer Institute Core (5P30CA013696), CH, Vos RM, Bialecka M, van Herwaarden T, van Dijk
Foundation Fighting Blindness (TA-NMT-0116-0692- EHC, Talib M, Freund C, Mikkers HMM, Hoeben RC,
COLU), the Research to Prevent Blindness (RPB) Goumans M-J, Boon CJF, Koster AJ, Chuva de Sousa
Physician-Scientist Award. B.L.D. is a recipient of the Lopes SM, Jost CR, Wijnholds J. Human iPSC-­derived
Capes PhD scholarship. P.M.J.Q. is the current recipient retinas recapitulate the fetal CRB1 CRB2 complex
of a Curing Retinal Blindness Foundation (CRBF) grant, formation and demonstrate that photoreceptors and
a Knights Templar Eye Foundation (KTEF) Career Starter Müller glia are targets of AAV5. Stem Cell Rep.
grant, an Uplifting Athletes Young Investigator grant, and 2019;12:906–19.
a New York Stem Cell Foundation (NYSCF)— 5. Talib M, et al. Genotypic and phenotypic characteristics
Druckenmiller Fellowship. of CRB1-associated retinal dystrophies: a long-term
follow-up study. Ophthalmology. 2017;124:884–95.
6. Tso A, Ragi S, Costa BLD, Fehnel A, Li Y, Quinn
Conflict of Interest Stephen H. Tsang receives
PMJ. Generation of human iPSC-derived retinal
financial support from Abeona Therapeutics, Inc. and
organoids for assessment of AAV-mediated gene
Emendo. He is also the founder of Rejuvitas and is on
delivery. Methods Mol Biol. 2021; (in press).
the scientific and clinical advisory board for Nanoscope
Therapeutics.
Inducing Neural Regeneration
from Glia Using Proneural bHLH
Transcription Factors

Levi Todd

Abstract Replacement of lost neurons is being pursued


through both exogenous and endogenous
Endogenous regeneration strategies to replace strategies [33]. In exogenous strategies, either
lost neurons hold great promise for treating stem cells or differentiated neurons are
neurodegenerative disorders. In the majority transplanted into the diseased nervous system
of cases, neural regeneration is induced by with hopes that these cells can wire into existing
converting resident glial cells into neurogenic circuitry and restore lost function. In endogenous
precursors. This review will outline how pro- strategies, tissue resident cells are targeted to
neural bHLH transcription factors can be used serve as a source of new neurons. The primary
to reprogram glia in the brain and retina into a approach in endogenous regeneration is to
source for new neurons. reprogram glial cells into neurogenic precursors.
Glia-to-neuron reprogramming occurs
Keywords naturally in regenerative species [34]. For
example, in the retina of zebrafish, Müller glia
Regeneration · Glia · Astrocyte · NG2 · respond to injury by reinitiating a molecular
Reprogramming · Müller glia · Retina · program akin to retinal progenitor cells [20].
bHLH Transcription factor · These reprogrammed Müller glia then go onto
Neurodegeneration regenerate retinal neurons capable of restoring
lost vision [12]. In mammals, glia do not
spontaneously reprogram after CNS injury but
1 Introduction instead initiate an inflammatory response [7].
Therefore, the goal in regenerative neuroscience
Neurodegenerative diseases result in permanent is to develop strategies in the mammal that can
disability because once neurons are lost they are steer glia into a neurogenic response when neural
not replaced. Cell replacement strategies aim to loss occurs. To achieve this goal, our group and
restore lost neurons and offer therapeutic hope to others have been using developmental
patients in late-stage neurodegenerative diseases. transcription factors (TFs) to revert glia back into
a neurogenic progenitor state [6, 13, 34]. This
L. Todd (*) mini-review will outline strategies using
Department of Biological Structure, University of proneural basic helix-loop-helix (bHLH) TFs to
Washington, induce glia-to-neuron conversion in the brain and
Seattle, WA, USA retina.
e-mail: ljtodd@uw.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 577
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1_84
578 L. Todd

Proneural bHLH TFs are anciently conserved neurogenesis when combined with other factors.
across the animal kingdom and are crucial for Ascl1 in combination with Sox2, Brn2, Lmx1a of
neuronal differentiation. The proneural bHLH Mytl1 can induce neurons (typically GABAergic)
TFs that have been used in glial reprogramming from both astrocytes and NG2 glia [18, 30, 37,
in the mammalian CNS can be grouped into four 38]. Additionally, when cellular stress pathways
subfamilies: (1) Achaete-Scute complex-like are mitigated by overexpression of Nurr1, Ascl1
(Ascl1), (2) Neurogenin (Neurog2), (3) Atonal– is capable of inducing neural conversion from
homolog (Atoh1 and 7), and (4) Neurod cortical astrocytes on its own [28]. In a recent
(Neurod1) [1]. This review will summarize how study, a retrovirus was used to infect dividing glia
each of these classes of proneural factors have to overexpress Ascl1 and Dlx2 in hippocampal
been used to engineer glial reprogramming glia in a model of temporal lobe epilepsy [24].
in vivo in the injured nervous system. The combination of Ascl1 and Dlx2 was suffi-
cient to regenerate GABAergic neurons that were
lost to seizures and these new cells were capable
2 Ascl1 of integrating into circuitry and reducing seizure
frequency [24]. These studies combined with
In development, Ascl1 promotes proliferation findings in the retina demonstrate the therapeutic
and neural differentiation of progenitor cells potential of Ascl1-mediated reprogramming in
across the brain and retina [16, 21]. This neuro- the diseased nervous system.
genic capacity of Ascl1 has been leveraged by
multiple groups to induce progenitor-like charac-
teristics in mature glial cells (see Table 1). In the 3 Neurogenin (Neurog2)
retina, lentivirus-mediated overexpression of
Ascl1 can reprogram Müller glia into a neuro- Neurog2 is expressed in neurogenic progenitors
genic state in vitro [31]. In follow-up studies, a in the brain and retina where it promotes differ-
transgenic mouse was created that allowed for entiation and subtype specification [2, 32].
in vivo inducible expression of Ascl1, specifi- In the brain, Neurog2 is the most efficient
cally in Müller glia [39]. After retinal injury, proneural bHLH at inducing neural conversion
Ascl1 was capable of stimulating neural regen- from glia [11, 19]. Neurog2, while sufficient to
eration from Müller glia in young mice but not in induce neural reprogramming, is a significant
adults [39]. However, combining Ascl1 overex- stressor to cells [11]. Combining Neurog2 with
pression with a histone deacetylase inhibitor to factors to reduce cellular stress pathways or
allow for more open chromatin led to neural inhibit apoptosis significantly enhances glial-to-
regeneration in the adult mouse retina [22]. These neuron conversion [11, 14, 19, 28]. Interestingly,
glial-derived neurons are capable of functionally in the brain, Neurog2 seems to bias glial-induced
integrating into the damaged retinal circuit, sug- neurons to a glutamatergic fate [11, 28]. This is
gesting a future therapeutic avenue for cell resto- consistent with Neurog2’s role in development
ration in diseased retinas [22, 23]. where it biases progenitors to an excitatory fate
In cortical glia, Ascl1 is typically ineffective [15]. These data suggest that the particular bHLH
or only stimulates a small amount of neurogene- TF used will influence the ultimate fate
sis [14, 18, 28]. Although in the midbrain Ascl1 acquisition in vivo. This has been confirmed with
alone has been reported to convert astrocytes into astrocytes in vitro where Ascl1 induces
neurons in vivo [25], this report failed to find GABAergic fates and Neurog2 induces
Brdu+ neurons, suggesting Ascl1 did not induce glutamatergic fates through distinct downstream
a proliferating progenitor state in astrocytes. targets [27]. The ability of Neurog2 to reprogram
Ascl1 can significantly potentiate glial-mediated
Table 1 Studies reporting in vivo glia to neuron reprogramming in brain and retina
Type
TF used Area of CNS reprogrammed Neuron type induced Fate mapping Citation
Ascl1 Retina Müller glia Bipolar Cre-lineage tracing, EdU Jorstad et al. [22, 23] and
labeling Todd et al. [35]
Ascl1 (+Dlx2) Hippocampus Astrocytes, NG2 GABAergic Retrovirus with BrdU labeling Lentini et al. [24]
Ascl1 Midbrain, striatum, Astrocytes GABAergic + glutamatergic AAVs with GFAP promoter Liu et al. [25]
somatosensory neurons
Ascl1 (+Brn2/Myt1l) Striatum Astrocytes NeuN+ Lentivirus with Cre Torper et al. [37]
Ascl1 (+Sox2) Cortex Astrocytes, NG2 Immature neurons Cre-lineage tracing Heinrich et al. [18]
Ascl1 (with Nurr1) Cortex Astrocytes Undetermined AAV virus combined with Mattugini et al. [28]
Cre-mice + EdU
Ascl1 (with Striatum NG2 glia Parvalbumin interneurons AAV virus combined with Pereira et al. [30] and
Lmx1a + Nurr1) Cre-lineage Torper et al. [38]
Atoh1 or 7 (with Retina Müller glia Bipolars, amacrines, ganglion-like Cre-lineage tracing, EdU Todd et al. [36]
Ascl1) neurons labeling
NeuroD1 Cortex Astrocytes, NG2 Astros = glutamatergic Retrovirus Guo et al. [17]
NG2 = glutamatergic and
GABAergic
NeuroD1 Cortex Astrocytes Excitatory and inhibitory neurons AAV with GFAP promoter Chen et al. [10]
Neurog2 Cortex Astrocytes Glutamatergic Retrovirus Gascon et al. [11]
Inducing Neural Regeneration from Glia Using Proneural bHLH Transcription Factors

Neurog2 (with Nurr1) Cortex Astrocytes Pyrimadial neurons AAV virus combined with Mattugini et al. [28]
Cre-mice + Edu
Neurog2 (with Bcl2) Cortex + thalamus Astrocytes Cortical neurons or thalamic Retrovirus + BrdU-labeling Herrero-Navarro et al. [19]
neurons
579
580 L. Todd

glia in the adult retina in vivo has yet to be 5 NeuroD


explored.
Similar to Atonal genes, NeuroD factors are
upregulated in postmitotic cells where they poten-
4 Atonal–Homolog tiate neural differentiation [5]. NeuroD1 delivered
via retrovirus to infect dividing glia was reported
Atonal genes (Atoh1/7) are deeply conserved to reprogram both astrocytes and NG2 glia in a
regulators of neurogenesis in both the retina and mouse model of Alzheimer’s disease [17].
the brain [3, 8]. Unlike Ascl1 and Ngn2, Atoh7 is Interestingly, NeuroD1 induced neurons with a
not expressed in cycling progenitors but is glutamatergic fate from astrocytes but repro-
induced as cells terminally differentiate into neu- grammed NG2 glia to both glutamatergic and
rons [9]. The ability of Atonal factors to repro- GABAergic fate [17]. This suggests that in addi-
gram glia in the brain has yet to be explored. tional to the TF used, the cell type of origin sig-
Although Atoh1 was identified as the most effec- nificantly influences the induced cell fate choice.
tive factor in a recent screen of all human tran- Using Adeno-associated viruses (AAVs) with
scription factors able to direct induced pluripotent a Glial fibrillary acidic protein (GFAP) promoter,
stems cells into neuron [29]. This suggests Atonal NeuroD1 was reported to induce neurons from
factors are an attractive target for neural regener- glia at an unprecedented efficacy rate (>90%
ation strategies in the brain. In the retina, Atoh7 infected astrocytes) after ischemic stroke and
has been demonstrated to neurogenically repro- facilitate functional recovery [10]. While this
gram glia in the medaka fish. Unlike zebrafish, finding potentially highlights the therapeutic rel-
Müller glia in the medaka do not spontaneously evance of this approach, concerns have clouded
reprogram into progenitors in response to injury interpretations of AAV-mediated glial reprogram-
[26]. Interestingly, forced overexpression of ming [6, 40]. A recent report demonstrated that
Atoh7 is sufficient to stimulate neurogenesis despite GFAP being a glial specific marker, an
from Medaka retinal glia [26]. Recently, our AAV using the GFAP promoter to drive NeuroD1
group tested whether Atonal transcription factors surprisingly lead to labeling of endogenous neu-
could aid in glia-to-neuron reprogramming in the rons [40]. Using classical developmental biology
mammalian retina. Atoh7 overexpression in techniques such as EdU labeling, transgenic lin-
Müller glia in vitro induced a progenitor state eage tracing, and retrograde tracing, it was con-
capable of neurogenesis [36]. The ability of vincingly shown that AAVs using the GFAP
Atoh7 to reprogram retinal glia in vivo remains to promoter to drive NeuroD1 labeled already exist-
be determined. However, we found that Atoh1, a ing neurons and not newly generated cells [40].
closely related TF to Atoh7, significantly More concerning, when stringent controls were
enhanced Ascl1-mediated Müller glia repro- used to induce NeuroD1, specifically and reliably
gramming in the adult mouse retina. The combi- in astrocytes, no neural reprogramming was
nation of Ascl1:Atoh1 also redirected the fate of observed [40]. This leaves NeuroD as a factor
induced neurons from what is typically seen with capable of inducing neuronal reprogramming in
Ascl1 [36]. Ascl1 induces Müller glia to take on doubt.
a bipolar neuron fate, while Ascl1:Atoh1 stimu-
lates regeneration of primarily retinal ganglion-­
like cells [36]. This study provides a proof of 6 Outlook
principle that combinations of proneural bHLH
TFs are ideal candidates for improving glial Despite the significant setback of methodological
reprogramming as well as influencing the ulti- concerns regarding AAV-mediated glial repro-
mate neural cell type generated in the retina. graming, multiple lines of evidence suggest that
glia-to-neuron conversion is a bona fide phenom-
enon that has the potential to be harnessed for
therapeutic cell replacement. In vitro, live imag-
Inducing Neural Regeneration from Glia Using Proneural bHLH Transcription Factors 581

ing has definitively shown that astrocytes can be HY. Math1 is essential for genesis of cerebellar
granule neurons. Nature. 1997;390:169–72.
reprogrammed into neural cells by delivery of 4. Berninger B, Costa MR, Koch U, Schroeder T,
neurogenic TFs [4, 11]. In vivo, because neurons Sutor B, Grothe B, Gotz M. Functional properties of
are postmitotic, reprogrammed glial cells that go neurons derived from in vitro reprogrammed postnatal
through a proliferating progenitor state can be astroglia. J Neurosci. 2007;27:8654–64.
5. Bertrand N, Castro DS, Guillemot F. Proneural genes
labeled with BrdU or EdU. Labeling for BrdU+ and the specification of neural cell types. Nat Rev
or EdU+ neurons demonstrate whether neurons Neurosci. 2002;3:517–30.
are newly derived or existing cells being artefac- 6. Blackshaw S, Sanes JR. Turning lead into gold:
tually labeled. Multiple studies have demon- reprogramming retinal cells to cure blindness. J Clin
Invest. 2021;131:e146134.
strated BrdU/EdU labeled neurons after glia 7. Bringmann A, Iandiev I, Pannicke T, Wurm A,
reprogramming, both in the brain and retina [14, Hollborn M, Wiedemann P, Osborne NN, Reichenbach
19, 23, 24, 28, 35, 36]. Furthermore, classical A. Cellular signaling and factors involved in Muller
transgenic glial-lineage tracing with Cre-ERT2 cell gliosis: neuroprotective and detrimental effects.
Prog Retin Eye Res. 2009;28:423–51.
mice have been used to convincingly demonstrate 8. Brown NL, Patel S, Brzezinski J, Glaser T. Math5
the glial-origin of new neurons [18, 20, 22, 28, is required for retinal ganglion cell and optic nerve
36]. Finally, scRNA-seq technologies can be lev- formation. Development. 2001;128:2497–508.
eraged to observe intermediate molecular profiles 9. Brzezinski JAT, Prasov L, Glaser T. Math5 defines
the ganglion cell competence state in a subpopulation
while glia are in the process of reprogramming to of retinal progenitor cells exiting the cell cycle. Dev
neurogenic progenitors. Indeed, this has been Biol. 2012;365:395–413.
observed in both the brain and retina, further con- 10. Chen YC, Ma NX, Pei ZF, Wu Z, Do-Monte FH,
firming the ability to stimulate neural regenera- Keefe S, Yellin E, Chen MS, Yin JC, Lee G, et al. A
NeuroD1 AAV-based gene therapy for functional brain
tion from glia in vivo [20, 23, 36, 41]. The table repair after ischemic injury through in vivo astrocyte-­
included in this review summarizes reprogram- to-­neuron conversion. Mol Ther. 2020;28:217–34.
ming studies, describes which factor was used, 11. Gascon S, Murenu E, Masserdotti G, Ortega F,
where is the CNS, and what lineage-tracing con- Russo GL, Petrik D, Deshpande A, Heinrich C,
Karow M, Robertson SP, et al. Identification and
trols were employed (Table 1). successful negotiation of a metabolic checkpoint
The first report of in vivo reprogramming of in direct neuronal reprogramming. Cell Stem Cell.
glial cells in the mammalian CNS is less than a 2016;18:396–409.
decade old [37]. Since then, the field has rapidly 12. Goldman D. Muller glial cell reprogramming
and retina regeneration. Nat Rev Neurosci.
developed, discovering diverse strategies to 2014;15:431–42.
reprogram glial cells in various regions of the 13. Gotz M, Bocchi R. Neuronal replacement: concepts,
nervous system. This body of work highlights achievements, and call for caution. Curr Opin
exciting future opportunities to repair the dam- Neurobiol. 2021;69:185–92.
14. Grande A, Sumiyoshi K, Lopez-Juarez A, Howard
aged brain and retina. With important method- J, Sakthivel B, Aronow B, Campbell K, Nakafuku
ological concerns now being considered, the field M. Environmental impact on direct neuronal
should now be able to press forward with more reprogramming in vivo in the adult brain. Nat
reliable technical approaches as we rapidly Commun. 2013;4:2373.
15. Guillemot F. Cell fate specification in the mammalian
advance to developing strategies that may one telencephalon. Prog Neurobiol. 2007;83:37–52.
day be used in human diseases. 16. Guillemot F, Hassan BA. Beyond proneural: emerging
functions and regulations of proneural proteins. Curr
Opin Neurobiol. 2017;42:93–101.
17. Guo Z, Zhang L, Wu Z, Chen Y, Wang F, Chen
References G. In vivo direct reprogramming of reactive glial
cells into functional neurons after brain injury and
1. Baker NE, Brown NL. All in the family: proneural in an Alzheimer’s disease model. Cell Stem Cell.
bHLH genes and neuronal diversity. Development. 2014;14:188–202.
2018;145:dev159426. 18. Heinrich C, Bergami M, Gascon S, Lepier A, Vigano
2. Bassett EA, Wallace VA. Cell fate determination in the F, Dimou L, Sutor B, Berninger B, Gotz M. Sox2-­
vertebrate retina. Trends Neurosci. 2012;35:565–73. mediated conversion of NG2 glia into induced
3. Ben-Arie N, Bellen HJ, Armstrong DL, McCall neurons in the injured adult cerebral cortex. Stem Cell
AE, Gordadze PR, Guo Q, Matzuk MM, Zoghbi Rep. 2014;3:1000–14.
582 L. Todd

19. Herrero-Navarro A, Puche-Aroca L, Moreno-Juan V, 30. Pereira M, Birtele M, Shrigley S, Benitez JA, Hedlund
Sempere-Ferrandez A, Espinosa A, Susin R, Torres-­ E, Parmar M, Ottosson DR. Direct reprogramming of
Masjoan L, Leyva-Diaz E, Karow M, Figueres- resident NG2 glia into neurons with properties of fast-­
Onate M, et al. Astrocytes and neurons share spiking parvalbumin-containing interneurons. Stem
region-­specific transcriptional signatures that confer Cell Rep. 2017;9:742–51.
regional identity to neuronal reprogramming. Sci Adv. 31. Pollak J, Wilken MS, Ueki Y, Cox KE, Sullivan JM,
2021;7:eabe8978. Taylor RJ, Levine EM, Reh TA. ASCL1 reprograms
20. Hoang T, Wang J, Boyd P, Wang F, Santiago mouse Muller glia into neurogenic retinal progenitors.
C, Jiang L, Yoo S, Lahne M, Todd LJ, Jia M, Development. 2013;140:2619–31.
et al. Gene regulatory networks controlling 32. Schuurmans C, Armant O, Nieto M, Stenman
vertebrate retinal regeneration. Science. JM, Britz O, Klenin N, Brown C, Langevin LM,
2020;370:eabb8598. Seibt J, Tang H, et al. Sequential phases of cortical
21. Imayoshi I, Kageyama R. bHLH factors in self-­ specification involve Neurogenin-dependent and
renewal, multipotency, and fate choice of neural -independent pathways. EMBO J. 2004;23:2892–902.
progenitor cells. Neuron. 2014;82:9–23. 33. Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang
22. Jorstad NL, Wilken MS, Grimes WN, Wohl SG, K, Goldberg JL, Ali RR, Young M, Xie Y, Temple
VandenBosch LS, Yoshimatsu T, Wong RO, Rieke S. Regenerating eye tissues to preserve and restore
F, Reh TA. Stimulation of functional neuronal vision. Cell Stem Cell. 2018;23:453.
regeneration from Muller glia in adult mice. Nature. 34. Todd L, Reh TA. Comparative biology of vertebrate
2017;548:103–7. retinal regeneration: restoration of vision through
23. Jorstad NL, Wilken MS, Todd L, Finkbeiner C, cellular reprogramming. Cold Spring Harb Perspect
Nakamura P, Radulovich N, Hooper MJ, Chitsazan Biol. 2021;14:a040816.
A, Wilkerson BA, Rieke F, et al. STAT signaling 35. Todd L, Finkbeiner C, Wong CK, Hooper MJ, Reh
modifies Ascl1 chromatin binding and limits neural TA. Microglia suppress Ascl1-induced retinal
regeneration from Muller glia in adult mouse retina. regeneration in mice. Cell Rep. 2020;33:108507.
Cell Rep. 2020;30:2195–2208.e5. 36. Todd L, Hooper MJ, Haugan AK, Finkbeiner C,
24. Lentini C, d’Orange M, Marichal N, Trottmann Jorstad N, Radulovich N, Wong CK, Donaldson PC,
MM, Vignoles R, Foucault L, Verrier C, Massera C, Jenkins W, Chen Q, et al. Efficient stimulation of
Raineteau O, Conzelmann KK, et al. Reprogramming retinal regeneration from Muller glia in adult mice
reactive glia into interneurons reduces chronic seizure using combinations of proneural bHLH transcription
activity in a mouse model of mesial temporal lobe factors. Cell Rep. 2021;37:109857.
epilepsy. Cell Stem Cell. 2021;28:2104–2121.e10. 37. Torper O, Pfisterer U, Wolf DA, Pereira M, Lau
25. Liu Y, Miao Q, Yuan J, Han S, Zhang P, Li S, Rao Z, S, Jakobsson J, Bjorklund A, Grealish S, Parmar
Zhao W, Ye Q, Geng J, et al. Ascl1 converts dorsal M. Generation of induced neurons via direct
midbrain astrocytes into functional neurons in vivo. J conversion in vivo. Proc Natl Acad Sci U S A.
Neurosci. 2015;35:9336–55. 2013;110:7038–43.
26. Lust K, Sinn R, Perez Saturnino A, Centanin L, 38. Torper O, Ottosson DR, Pereira M, Lau S, Cardoso
Wittbrodt J. De novo neurogenesis by targeted T, Grealish S, Parmar M. In vivo reprogramming of
expression of atoh7 to Muller glia cells. Development. striatal NG2 glia into functional neurons that integrate
2016;143:1874–83. into local host circuitry. Cell Rep. 2015;12:474–81.
27. Masserdotti G, Gillotin S, Sutor B, Drechsel D, 39. Ueki Y, Wilken MS, Cox KE, Chipman L, Jorstad
Irmler M, Jorgensen HF, Sass S, Theis FJ, Beckers N, Sternhagen K, Simic M, Ullom K, Nakafuku
J, Berninger B, et al. Transcriptional mechanisms of M, Reh TA. Transgenic expression of the proneural
proneural factors and REST in regulating neuronal transcription factor Ascl1 in Muller glia stimulates
reprogramming of astrocytes. Cell Stem Cell. retinal regeneration in young mice. Proc Natl Acad
2015;17:74–88. Sci U S A. 2015;112:13717–22.
28. Mattugini N, Bocchi R, Scheuss V, Russo GL, Torper 40. Wang LL, Serrano C, Zhong X, Ma S, Zou Y, Zhang
O, Lao CL, Gotz M. Inducing different neuronal CL. Revisiting astrocyte to neuron conversion with
subtypes from astrocytes in the injured mouse cerebral lineage tracing in vivo. Cell. 2021;184:5465–5481.
cortex. Neuron. 2019;103:1086–1095.e5. e16.
29. Ng AHM, Khoshakhlagh P, Rojo Arias JE, Pasquini G, 41. Zamboni M, Llorens-Bobadilla E, Magnusson JP,
Wang K, Swiersy A, Shipman SL, Appleton E, Kiaee Frisen J. A widespread neurogenic potential of
K, Kohman RE, et al. A comprehensive library of neocortical astrocytes is induced by injury. Cell Stem
human transcription factors for cell fate engineering. Cell. 2020;27:605–617.e5.
Nat Biotechnol. 2021;39:510–9.
Index

A Autophagy, 11, 51, 144, 208, 211, 212, 320–322, 344,


AAV gene therapy, 125 385, 386, 404, 495, 497, 559
AAV serotypes, 119–121, 132, 133, 144 Avian myeloblastosis virus (AMV), 109–113
ABCA4-associated retinopathy, 289–294 Axonal degeneration, 224, 226
Abelson helper integration site 1 (AHI1), 178–180 Axonal transport, 223–226
Achromatopsia, 270, 293, 348, 354, 552
Actin cytoskeleton, 313, 509
Adeno-associated virus (AAV), 117–121, 125, 126, 129, B
132, 133, 138, 139, 270, 273, 328, 354, 552, 560, Bacteria, 15, 525
579, 580 Basal laminar deposit (BLamD), 5–7, 51, 68, 209, 337,
ADP-ribosylation factor-like 3 (ARL3), 177–180 339
Aerobic glycolysis, 429, 432, 435–438, 440 Base editing, 104, 105
Age-related macular degeneration (AMD), 4, 7, 9–12, β-catenin, 241–244, 246, 247
17, 22–24, 27–31, 37–46, 50–52, 55–57, 61–65, Beta5 integrin, 518
68, 70, 73–76, 84, 88–92, 133, 160, 161, 165, bHLH transcription factor, 577–581
167, 207–212, 231, 232, 249, 258–260, 298, 311, Biomarkers, 56, 81–84, 90–92, 166, 169, 170, 224, 226,
312, 321–322, 328–331, 339, 340, 359, 361, 362, 259, 430
371–374, 404, 462, 488–490, 528, 529, 543, 551 Bisretinoids, 16–19
Aging, 17, 23, 27, 50, 51, 68, 70, 170, 210, 225, 226, Blindness, v, vii, 7, 37, 41–43, 55, 65, 73, 83, 104, 113,
359, 360, 373, 374, 462, 490, 543, 561 137, 143, 166, 167, 189, 207, 219, 242, 254, 260,
Alpha (α)-helix, 29, 286, 501, 502, 533–536 270, 271, 293, 303, 330, 340, 342, 358, 371, 372,
Amphipathic helix, 533–536 377, 387, 404, 407, 415, 425, 446, 473, 490, 510,
AMV RT Prime Editors, 110 528, 551, 553, 555, 572
Amyloid, 31, 90, 265–267 Blood-retinal barrier (BRB), 200, 231, 241, 242, 244,
Animal models, vi, 23, 31, 65, 92, 121, 136, 138, 139, 246, 249, 250, 304, 471, 488, 490
169, 190, 202, 203, 218, 225, 226, 231, 272, 310, Bruch’s membrane, 5, 12, 27, 31, 41, 51, 61, 90, 208,
322, 343, 366, 368, 372, 390, 391, 421, 445, 446, 310–312, 321, 336, 337, 339, 359–361, 373, 539
452, 453, 457, 458, 543, 550–553, 556, 557, 565
Anti-AAV immune response, 118–121
Anti-diabetic drugs, 76 C
Antioxidant, 10, 16, 19, 75, 444–446 Capsid, 117, 120, 121, 132, 133, 354
Antioxidant response, 444 Carotenoids, 15–20, 534
Apolipoprotein A1 (ApoA1), 56, 391, 392 Cav1 complexes, 250–254
Apoptosis, 16, 57, 82, 91, 169, 198, 199, 202, 224, 225, Cav1.4, 269–271
270, 303–305, 354, 357, 365, 528, 558, 578 Caveolae, 244, 249, 252
Arl3, 283–287 Caveolin-1 (Cav1), 244–246, 249–254
Arl13B, 177, 283, 284, 286, 287 Cavin1/PTRF, 252
ARMS2, 28–31, 64, 328, 556 CD68, 200, 207–212
Aryl hydrocarbon receptor (Ahr), 209 CD163, 208
Astrocyte, 230, 231, 473–476, 578–580 Cell death, 17, 75, 90–92, 169, 191, 198, 199, 216, 218,
ATP Binding Cassette Subfamily A Member 4 (Abca4), 224, 231, 236, 237, 242, 284, 298, 303–305, 321,
17, 18, 167, 289–293 322, 344, 349, 351, 424, 445, 446, 474, 480, 481,
ATP-binding cassette transporter A1 (ABCA1), 56–57 509, 524, 560, 561

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature 583
Switzerland AG 2023
J. D. Ash et al. (eds.), Retinal Degenerative Diseases XIX, Advances in Experimental Medicine
and Biology 1415, https://doi.org/10.1007/978-3-031-27681-1
584 Index

Cell lines, 30, 31, 65, 89, 92, 216, 219, 250, 503, 540, Disease mechanisms, 62, 65, 166, 169, 180, 293, 366,
542, 550, 565, 566 452, 453, 551
Cellular retinaldehyde binding protein (CRALBP), 208, Disease-modeling, 549
534–536 Disease progression, 10, 41, 84, 137, 139, 200, 203, 209,
Cellular stress, 543, 578 259, 300, 310, 344, 366, 369, 405, 528, 556, 561
CEP290, 174–176, 180, 186, 266, 372, 396, 398, 400, Diseasome, 166–170
401, 552 DNA damage, 17, 57, 88
Ceramide, 39, 40, 303–305 Dolichol, 449–453
Ceramide synthases, 304 Dolichyl phosphate (Dol-P), 450, 451, 453
Cholesterol, 11, 51, 56, 57, 328, 391, 449, 451, 452, 468, DOLK, 450, 451, 453
470 DOLPP1, 450, 451, 453
Choroid, 19, 20, 23, 28, 31, 63–65, 68–70, 207, 311, 312, Dominant negative effects, 136, 137
321, 359–362, 539, 543 Drug, 46, 52, 62, 73–76, 149, 151–153, 165, 166,
Choroidal neovascularization (CNV), 22, 23, 27, 56, 68, 168–170, 186, 267, 331, 369, 404, 467–469,
70, 208, 209, 462 474–475, 490, 529, 530, 553, 566
Cilia, 174, 177, 279, 283, 284, 396–402, 507, 509, 557, Drug repurposing, 75, 76
560
Circadian clock, 542, 543
Clusterin (CLU), 216–219 E
Cnga3, 270, 349–351, 354 Elastin, 68–70, 310, 362
Collagen, 68, 310–314 Electroretinography (ERG), 68, 136, 189–191, 202, 259,
Complement, 10–12, 17, 22, 28, 29, 31, 51, 62, 68–70, 271, 273, 290, 291, 293, 343, 344, 348–351, 366,
73, 75, 90, 160, 161, 197, 200, 208, 290, 367, 373, 374, 390, 391, 405, 407, 416, 417,
360–362, 373, 380, 385, 476, 553 422–425, 488, 489
Condensate, 263–267 ELOVL4, 258–260
Cone, 5, 28, 56, 98, 118, 136, 137, 143–146, 176, 177, Endoplasmic reticulum-associated degradation (ERAD),
184, 190–192, 199, 230, 236, 269–273, 290–293, 357, 358, 494, 497
298, 299, 312, 321, 328, 329, 342, 343, 347–351, Endoplasmic reticulum (ER) calcium homeostasis, 354,
354–358, 367, 368, 377, 380, 390, 404–407, 436, 357
444, 451, 508, 516–518, 551, 552, 555, 556 Endoplasmic reticulum stress, 444, 446, 543, 551
Cone photoreceptors, 5, 52, 144, 190–193, 236, 284, Enhanced S-cone Syndrome (ESCS), 189–193
285, 328, 329, 348, 353, 354, 365, 424, 489, 516, Exonic splicing enhancers (ESEs), 184–186
550 Exonic variants, 184, 186
Connecting cilium (CC), 61, 68, 174, 245–247, 265–266, Exosomes, 45, 46, 81, 82, 88, 90, 91
372, 396, 398, 401, 508, 552, 557 Expansion microscopy (ExM), 396–402
Connectomics, 297–300 Expression quantitative trait loci (eQTL), 62–65, 373
Connexins (Cx), 229–231 Extracellular matrix (ECM), 5, 22–24, 28, 31, 51, 62, 68,
Crumbs-homologue-1 (CRB1), 104–106, 200, 560, 90, 216, 309–314, 321, 360–362, 380, 457, 461
572–576 Extracellular vesicles (EVs), 45–46, 81–84, 88–92, 279,
CRX mutations, 136, 139 391, 392
CTRP5/C1QTNF5, 340 Eye development, 328, 330
Cyclic nucleotide-gated (CNG), 269–272, 353–355, 357, Eye diseases, vi, 31, 83, 120, 191, 259, 260, 328, 330, 331
358
Cyclic nucleotide-gated channel, 349
Cytokine, 11, 50, 69, 70, 197–201, 207–210, 216, 218, F
250, 328, 329, 380, 411, 412, 446, 490, 522, 524 Fam161a, 366–369
Cytopathy, 523, 524 Fixed, 38, 40, 242, 293, 378, 399, 401, 405, 417, 422,
430, 469, 470, 501, 516, 522
Fluoxetine, 75
D Free-energy, 158–161
D477G, 416, 417 Functional genomics, 378, 381
Daily rhythm, 516 Fundus autofluorescent spots, 337, 338
DHDDS, 450–452
Diabetes, 50, 76, 331, 404, 489, 490, 529, 542
Disc, 185, 224, 266, 277–280, 285, 338, 339, 389–392, G
401, 500, 507–510 Gap junctions, 229–231, 299
Disc enclosure, 266, 279, 280, 509 Gene augmentation, 104, 105, 135–140, 552
Disc membrane, 17, 266, 269, 390–392, 508 Gene editing, 83, 99, 110, 176, 552, 557
Disc rim, 266, 278–280 Gene expression regulation, 62, 65, 218
Index 585

Gene knock-out mouse model, 336, 339 Intercellular communication, 82, 83, 229
Gene regulatory networks, 137, 550 Interphotoreceptor matrix (IPM), 310, 312, 391, 429,
Gene therapy, vii, 83, 104, 117–121, 125, 131–132, 166, 457–459, 461, 462
169, 231, 232, 270, 303, 328, 366, 369, 377, 404, Intracellular complement factor H (CFH), 10–12, 28, 64,
406 68, 161, 362
Genetics, 10, 22, 23, 28, 30, 31, 51, 55, 57, 61–65, 68, In vitro models, 550, 567, 568
76, 83, 119, 140, 158, 160, 165–169, 173, 174, In vivo models, 23, 530, 565, 566
180, 184, 200, 207, 209, 242, 272, 290, 291, 320, Ion channels, 269, 271, 273
328, 330, 342, 354, 359, 372, 373, 377, 383, 391, IPSC-derived retinal organoids, 99, 550, 551, 553, 572,
410, 416, 422, 444, 446, 450, 479, 483, 500, 503, 576
551, 555–557, 561, 566, 572 Ischemic retinopathies, 529, 530
Genome-wide association studies (GWAS), 61–65
Genotype, 10, 30, 63, 64, 180, 289–294, 337, 379, 384,
422, 574 J
Genotype-phenotype correlation, 166, 180, 290–292 Joubert syndrome (JBTS), 173–180, 285–287
Geographic atrophy (GA), 10, 22, 23, 27, 44, 46, 75,
208, 336, 462
Glaucoma, 83, 84, 113, 165, 224–226, 231, 232, 249, K
311, 313, 321, 371, 373, 404, 542 Knockout, 50, 51, 127, 139, 145, 190, 211, 236, 237,
Glia, 5, 91, 249–253, 297, 310–312, 377–381, 439, 473, 250, 258–260, 270–272, 284, 285, 354, 366, 384,
475, 476, 489, 567, 577–581 390, 396, 416, 452, 490, 509, 510, 556
Glutathione, 467–471 Kv2.1, 269, 271–273
GPR91, 527–530 Kv8.2, 269, 271–273
GPR143, 44–46, 74
G-protein coupled receptor (GPCR), 44, 500
L
Laminin, 310, 312, 313
H Large animal models, 118
HCN1, 269, 271–272 Late-onset retinal degeneration (L-ORD), 336, 339, 340
High density lipoprotein (HDL), 56, 57, 451 Late-stage treatment, 404
Higher-order assembly, 263 LC-MS/MS, 38–42, 251, 360, 391–393, 458
Homology Directed Repair (HDR), 571–576 L-DOPA, 74
HTRA1, 28–31, 64, 328, 361, 362 Leber congenital amaurosis (LCA), 104, 136, 167,
Hyperoxia, 444 174–176, 179, 180, 200, 266, 270, 371, 415, 551
Hypoxia, 51, 433, 444, 489, 490, 529 Lecithin retinol acyl transferase (LRAT), 18, 372,
534–536
Linkage disequilibrium (LD), 28, 62
I Lipid accumulation, 56, 57, 321
Immune system, 10, 68, 91, 201, 344, 361, 362 Lipid homeostasis, 41, 390, 391
Immunization, 68–70 Lipids, 4–6, 10–12, 16, 17, 38–42, 45, 46, 50, 51, 55–57,
Immunoprecipitation (IP), 244, 250–253, 392, 494 62, 68, 76, 81, 82, 88, 145, 223, 249, 252, 257,
IMPG2, 312, 457–459, 461, 462 266, 283, 284, 304, 311, 321, 339, 361, 372, 374,
Induced pluripotent stem cell (iPSC), 31, 51, 65, 98, 99, 101, 380, 389–393, 401, 444–446, 452, 453, 462, 468,
187, 259, 540, 549–553, 556–562, 572–574, 576 501–503, 508, 509, 528, 530, 543
Inflammation, 10, 12, 17, 31, 41, 51, 57, 69, 75, 76, Lipofuscin, 16, 17, 19, 258, 289–291, 321, 322, 543
90–92, 118, 199, 209, 211, 236, 329, 330, 362, Liposome, 279, 467–470
372, 444, 446, 522, 524 Liver X receptor (LXR), 57
Inherited retinal degenerations (IRDs), 83, 165, 173, 217, Luminance, 192, 416, 417
272, 285, 289, 365, 390, 479, 551, 552 Lysine, 28, 494–497
Inherited retinal diseases (IRDs), 98, 110, 136, 149, Lysosomal dysfunction, 320–322
183–187, 278, 347, 555, 572
Inherited retinal dystrophy (IRD), 404, 444–446
Inherited retinopathy (IR), 415 M
Innate immunity, 410, 411 Macrophages, 56, 57, 69, 197, 202, 203, 208–211, 328,
Inner limiting membrane (ILM), 16, 132, 265, 310, 311, 329, 361, 411, 421–423, 543
313 Macula, 9, 10, 15, 16, 19, 27, 31, 44, 61, 63, 118, 167,
Inositol polyphosphate-5-phosphatase E gene (INPP5E), 191, 202, 259, 270, 273, 285, 336, 404, 416, 457,
178–180, 287 550, 556
Integrin, 82, 132, 133, 311, 313, 518, 540–542 Macular pigments (MPs), 15, 16, 19, 445
586 Index

Mass spectrometry (MS), 3–7, 250–254, 362, 391, 458, 509 416, 422, 444, 451–453, 457, 479, 481, 494, 497,
Matrix assisted laser desorption ionization imaging mass 500, 501, 503, 540, 551–553, 556–558, 561, 566,
spectrometry (MALDI IMS), 4–6 572–575
Matrix metalloproteinases (MMPs), 21–24, 216–218 Myeloid cells, 211, 410, 411, 413
Mechanisms, 10, 11, 22, 23, 31, 41, 46, 50, 62, 65, 69, MYO7A, 125–129
70, 73, 82, 83, 90–92, 105, 136, 144–146, 151, Myosin 1C, 499–504
168, 169, 184, 186, 192, 201, 208, 219, 224, 236,
237, 252, 265, 271, 285, 287, 290, 300, 310, 312,
313, 336, 340, 342–344, 348, 351, 358, 366, 368, N
391, 416, 446, 451, 467, 488–490, 497, 500, 501, 32:6 n-3, 259
504, 509, 518, 528, 530, 534, 540, 543, 549, 551, N-acetylmuramyl-L-alanine amidase (NAMAA),
555 522–525
Medical record database, 74, 76 NanoLC-MS/MS, 38–42
Melanin, 44, 539, 541 Network medicine, 166–170
Membrane association, 534, 536 Neural networks, 297, 298, 528
Meso-diaminopimelic acid, 524 Neurodegeneration, 52, 83, 165, 226, 273, 297–300, 373
Metabolic flux, 436 Neuron, 52, 133, 216, 218, 223–226, 230, 237, 243, 269,
Metabolism, 11, 50, 52, 55–57, 68, 74, 91, 144, 209, 249, 271, 297, 299, 300, 304, 309–310, 329, 351, 389,
304, 305, 321, 328, 361, 372, 380, 391, 429, 430, 423, 424, 439, 467, 473, 475, 476, 488, 528, 551,
433, 436, 439, 445, 449–453, 459, 461, 462, 481, 566–568, 577–581
543 Neuronal reprogramming, 580
Metabolite, 4, 207, 305, 321, 429, 431, 433, 437, 439, Neutralizing antibodies (NABs), 118–121, 133, 530
449, 457–462, 528, 529 NG2, 578–580
Metabolomics, 145, 360, 430, 458, 459, 462 Nicotinamide adenine dinucleotide (NAD+), 235–237,
Metformin, 74–76, 290, 404–406 430, 436–438, 536
Microglia, 29, 197–203, 208–212, 231, 328, 329, 331, NMNAT1, 236, 237, 392
380, 411, 421–425, 440, 444, 446, 458, 462, 476, NOD agonist, 522, 524
550 Nonsense mutations, 149, 151, 186, 198, 202
MicroRNA (miRNA), 56, 57, 82–84, 88 Non-syndromic IRD, 173–180
Mini-retinas, 550–551 Norrin, 241–244, 246–248
Mitochondria, ix, 5, 11, 69, 75, 144, 145, 169, 223, 225, N-retinylidene-N-retinylethanolamine (A2E), 17
226, 236, 432, 433, 435, 438, 440, 462, 529 Nuclear receptor, 189
Motor protein, 223, 226, 500, 501, 504 Nuclear Receptor Subfamily 2 Group E Member 3
Mouse, 19, 20, 29–31, 50–52, 56, 57, 68–70, 75, 76, 83, (NR2E3), 139, 189–191, 193, 202, 550, 553, 556
104, 120, 121, 126–128, 136–140, 144, 145, 169, Nuclear receptors, 49, 51, 201, 327, 542
177, 190, 198–202, 208–211, 216, 236, 237, NUS1, 450–453
242–247, 250, 258–260, 264, 265, 270–273, 279,
284–286, 304, 305, 310, 328–331, 336–340,
342–344, 348, 350, 351, 354–357, 359–362, O
365–369, 372–374, 378, 381, 383–387, 390–392, Ocular diseases, 6–7, 62, 81–84, 169, 321, 330, 373, 415,
396–402, 405–407, 409–411, 413, 416–418, 542, 543
422–424, 430–432, 436, 437, 439, 440, 444–446, Optic atrophy 1 (OPA1), 225, 226
452, 458, 459, 461, 462, 468–470, 475, 476, 480, Oral-Facial-Digital 1 (OFD1), 177–180
488–490, 497, 500, 510, 515–518, 524, 540–543, Organoids, 313, 549–551, 553, 561, 566, 568
556, 566, 567, 578, 580, 581 Organotypic retinal explant culture, 430, 432, 468, 470,
Mouse models, 11, 19, 29, 31, 51, 57, 75, 104, 137, 190, 481
198, 200, 202, 203, 208–211, 225, 226, 236, 237, Outer segments (OSs), 5, 46, 50, 56, 98–100, 137, 144,
243, 259, 270, 284, 305, 328, 336, 339, 340, 145, 202, 257, 265, 266, 269–272, 277–280, 284,
342–344, 348, 354, 360, 366–369, 372–374, 384, 285, 299, 312, 342, 343, 366, 368, 372, 384,
404–406, 410, 416, 422–425, 452, 453, 468, 481, 389–393, 401, 423, 451, 461, 470, 494, 499, 500,
540–543, 556, 580 504, 507–510, 516, 518, 527, 540, 550, 551, 557
mRNA splicing, 183, 552, 558, 560, 561 Oxidative damage, 16, 75, 322, 444, 445
Müller glia, 273, 310 Oxidative stress, 11, 16, 22, 23, 41, 50, 57, 68, 90–91,
Muramyl dipeptide (MDP), 522 197, 200, 217, 218, 304, 312, 321, 322, 372, 373,
Mutations, vi, vii, 17, 29, 30, 44, 83, 98–101, 104, 105, 444–446, 529, 559
110–113, 125, 133, 136, 137, 139, 143–145, 149,
151, 158–161, 165–169, 174, 177, 180, 184–186,
190, 191, 198–200, 202, 224, 225, 236, 242, 258, P
270–272, 277–278, 280, 285–287, 291, 305, 312, P23H, 343, 372, 384, 385, 391, 411, 494–497
321, 336, 338, 341–344, 347, 354, 359, 365–367, Palmitoylation, 390, 534
372, 377, 390, 392, 404–406, 410, 411, 413, 415, Pathoconnectome, 299, 300
Index 587

Peripherin-2 (PRPH2), 98–101, 111–113, 167, 277–280, Prpf31, 540–543, 557, 560
557, 558 Pyruvate kinase M2 (PKM2), 436, 479–481, 483
PGC-1alpha, 49–52, 75, 321
Phagocytosis, 10, 17, 29, 46, 50, 55, 199, 201, 202, 207,
218, 321, 390, 515, 518, 539–543, 557 R
Phagosome, 17, 201, 321, 322, 385, 386, 515–518 Race, 28, 44–46, 372
Pharmacological, 231, 232, 236, 271, 451, 474, 476, 480, rd1, 52, 144, 145, 169, 198, 199, 202, 216, 236, 237,
483, 530 348–351, 366, 368, 369, 380, 430–433, 445, 458
Phospholipids, 257, 391, 393, 535, 536 rd10 mouse model, 199, 424
Photo-oxidation, 16–19 RDH5, 63, 64, 535, 536
Photoreceptor function, 193, 349, 405, 422, 445, 462, Reactive oxygen species (ROS), 16, 49–51, 198, 257,
487–490, 501 321, 322, 444, 451, 518, 529
Photoreceptor outer segments (POSs), 5, 10, 16–18, 46, Regeneration, 18, 68, 197, 309–314, 473, 577, 578, 580,
50, 55, 91, 98, 173, 185, 200, 202, 207, 310, 311, 581
321, 337, 499, 515–518, 527, 539–543 Reprogramming, 198, 310, 404, 405, 425, 473–476, 558,
Photoreceptors (PRs), 4, 5, 12, 16, 19, 22, 24, 27, 30, 37, 572, 577–581
44, 46, 49–51, 55–57, 61, 68, 88–91, 100, 104, Retina, ix, 4, 5, 10, 11, 15, 16, 19, 22, 23, 27, 30, 31, 38, 41,
133, 135, 137–139, 143–145, 166, 167, 173, 174, 50, 51, 56, 57, 61–65, 74, 83, 84, 88–92, 99, 100, 104,
177, 180, 184, 190–193, 198–203, 208, 216, 230, 118, 132, 133, 137–139, 144, 145, 150, 165–167,
231, 236, 237, 242, 248, 257, 265, 266, 269–273, 169, 173, 180, 186, 187, 190–192, 197, 202, 207–209,
277–280, 284, 285, 287, 298–299, 303–305, 310, 216, 218, 219, 229–231, 236, 237, 241–250,
312, 321, 328–330, 337, 341, 348, 349, 354, 358, 257–260, 265, 266, 269–273, 284, 285, 289, 290,
365, 366, 368, 372, 377, 378, 380, 383, 384, 297–300, 304, 305, 309, 310, 312, 313, 321, 328, 329,
389–393, 396–402, 404–407, 410, 411, 415, 331, 336–339, 341–343, 348, 355–357, 360, 366,
422–425, 429–433, 436–440, 444–446, 451, 452, 368, 374, 377–381, 383–386, 390–393, 396–401,
457, 458, 461, 462, 469, 479–481, 483, 488, 489, 405, 406, 410–413, 416, 417, 421, 422, 425, 429–433,
493, 499, 500, 504, 507–510, 515, 516, 528, 536, 436–440, 444–445, 451–453, 458, 459, 461, 462,
539, 542, 543, 550, 555, 567 467–470, 473, 474, 476, 479–481, 488, 494, 500,
Photoreceptor transduction, 354 508, 509, 515, 516, 518, 527–530, 535, 540–542,
Pigmentation, 44, 46, 342 550, 552, 555–557, 565–568, 577–581
Poly-ADP ribose polymerase (PARP), 235–237 Retina degeneration, 417
Precision medicine, 550 Retinal adhesion, 540–543
Preclinical models, 120, 553 Retinal and RPE pathology, 339
Primary cultures, 540, 541, 543, 566, 567 Retinal capillaries, 243, 246
Prime editing, 98–101, 104–105, 110, 111, 113 Retinal degenerations, v, vi, ix, 12, 16, 51, 75, 83, 92,
Progressive rod cone degeneration (PRCD), 390–393 104, 125, 139, 165, 167, 169, 173–180, 189–191,
Proliferation, 56, 57, 91, 191, 201, 202, 313, 329, 197, 199–203, 211, 216, 217, 235–237, 259, 272,
473–476, 528, 578 280, 285–287, 298–300, 303, 342–344, 366–369,
Propensity matching, 76 384, 390, 404, 416, 423, 425, 446, 451, 452, 468,
Protein aggregation, 264–266 473, 479, 483, 497, 551–553, 556
Protein glycosylation, 449–453 Retinal degeneration slow (RDS), 98, 372
Protein localization, 504 Retinal degenerative diseases, ix, 22, 159, 160, 166, 189,
Proteins, 10, 11, 16, 17, 23, 28–31, 45, 46, 50, 51, 56, 237, 266, 272, 299, 304, 310, 341, 367, 391, 421,
57, 68, 81–84, 88–92, 104, 106, 125–128, 132, 446
135–137, 139, 145, 146, 150, 151, 153, 158–161, Retinal dystrophies, 31, 104, 174, 180, 200, 287, 312,
167, 173, 174, 177, 180, 183–186, 197, 199–202, 404, 410, 444, 452, 534, 540, 572
208, 211, 216–219, 225, 226, 229, 237, 241, 244, Retinal explant cultures, 185, 430, 470
245, 249–252, 254, 257, 258, 264–266, 270–272, Retinal gene therapy, 121, 131–133, 137
277–278, 280, 283–287, 290, 305, 310, 312, 313, Retinal homeostasis, 10, 11, 88, 92, 235–237, 372, 424, 540
320, 322, 337, 338, 341–344, 348, 349, 353–358, Retinal metabolism, 462
360–362, 366, 368, 369, 372, 380, 383–387, 389, Retinal organoids, 98, 99, 101, 133, 187, 259, 313,
390, 392, 396, 399, 401, 402, 404, 405, 416, 423, 550–553, 557, 561, 566, 567, 572
424, 444, 446, 449, 451–453, 479, 488, 493–497, Retinal pigment epithelial cells, 216–219, 230, 231
499–504, 508, 509, 517, 521, 523, 524, 528, Retinal pigment epithelium (RPE), 4–6, 10–12, 16–20,
533–536, 540–543, 549, 551–553, 558, 559, 572, 22–24, 27, 29–31, 37–39, 41, 44–46, 49–52,
573 55–57, 61–65, 68–70, 74, 75, 84, 88–92, 99, 100,
Protein translation, 383, 385, 386 133, 139, 144, 145, 166, 186, 190, 200, 202,
Proteome, 62, 84, 88–92, 169, 218, 251, 360, 362, 383 207–212, 216–219, 230, 236, 242, 243, 249, 258,
Proteomics, 88–90, 92, 254, 360, 389, 391 265, 273, 289, 310–312, 321, 322, 336–340, 343,
Proteostasis, 387 359, 360, 362, 372, 373, 389, 404, 415, 416, 422,
588 Index

424, 425, 429, 452, 457, 458, 461, 462, 469, Sub-RPE deposits, 23, 337, 339
515–518, 522–525, 527–529, 534–536, 539–543, Succinate, 438, 528–530
550, 551, 555, 557, 560, 572 Super-resolution, 396, 401, 402
Retinal pigment epithelium (RPE) cells, 529 Systems biology, 92, 166
Retinal regeneration, 231, 310, 311, 476
Retinal remodeling, 298, 378, 380
Retinal vascular development, 528 T
Retinal vascular diseases, 331 Tet-On, 137
Retina pigmented epithelium 65kDa protein (RPE65), 16, Thioflavin T (ThT), 264, 265
18, 161, 208, 243, 372, 415–417, 534–536, 541 Tissue Inhibitor of Metalloproteinase-1, 216–219
Retinitis pigmentosa/retinal pigmentosa (RP), v–vii, 56, TLR, 411
83, 98–101, 104, 136, 139, 143–146, 158, 160, TLR2, 409–411, 413
167, 184–186, 189–191, 200, 211, 216, 218, 236, Tolerance, 39, 69, 70, 139
270, 272, 278, 280, 283, 298, 299, 312, 341–343, Transcription factor EB (TFEB), 320–322
348, 365–369, 371–374, 377, 378, 380, 390–392, Transcriptome, 62, 169, 203, 218, 378, 479, 481
404–406, 409–411, 413, 415, 416, 422–425, Transcriptomics, 203, 377–381, 439, 550
444–446, 451, 479–481, 483, 494, 497, 501, 540, Transferase dUTP nick end labeling (TUNEL), 351, 480
551, 552, 555–562, 572, 574, 576 Translational readthrough (TR), 127–129, 149, 151–153
Retinoic acid receptor-related orphan receptors (RORs), Translation termination, 150–151
327–331 Tropism, 132, 133, 139
Retinoids, 201, 202, 208, 354, 534, 535 Txnip, 144–146
Retinopathies, 46, 51, 74, 76, 136, 137, 165, 224, 232,
237, 248, 249, 272, 305, 451, 521, 525, 528–530
Retinopathy, 328–331 U
Reverse cholesterol transport (RCT), 56, 57 Ubiquitin-proteasome system (UPS), 11, 383–385
Rhodopsin (RHO), vi, 16, 89, 136, 159–161, 184, 185, Ubiquitylation, 495–497
257, 266, 279, 285, 299, 342–344, 349, 384, Ultrastructure, 279, 337, 339, 391, 541
390–392, 423, 424, 451, 493–497, 500–504, 517, Uncoupling, 436, 438, 440
534, 536, 558 Usher’s Syndrome type 1B (USH1B), 125, 129
Rhodopsin densities, 390–392 Usher syndrome, 186, 366, 556
Rho-P23H, 236, 342–344
RNA, 56, 75, 81–84, 88, 98, 105, 110, 112, 140, 177,
186, 187, 203, 216, 217, 348, 354, 380, 410, 412, V
550, 552 Variants, ix, 10–12, 28–31, 51, 62–65, 68, 100, 104–106,
ROM1, 277–280, 372 117, 118, 120, 121, 136, 158–161, 167, 173–180,
RORα, 327–331 184–187, 189–191, 285–287, 289–294, 328, 359,
RORβ, 327–331 552, 553
RORγ, 327, 328, 330–331 Vascular endothelial growth factor (VEGF), 23, 45, 46,
RP2, 177, 283, 284, 286, 287, 552, 559–561 51, 73, 250, 331, 444, 487–490, 528–530
Ryanodine receptor (RyR), 354–358 Very-long-chain polyunsaturated fatty acids (VLC-­
PUFAs), 257–260
Vision, 17, 22, 27, 38, 44, 68, 69, 83, 90, 113, 118, 125,
S 126, 143–145, 165–167, 170, 177, 184, 189, 190,
S-cones, 189–193, 329 192–193, 224–226, 230, 236, 242, 258, 259, 264,
Serotonin receptor agonist, 75 267, 270, 271, 273, 290, 303, 304, 309, 321, 328,
Serotype, 117–121, 132–133 338, 342, 348, 351, 354, 365, 378, 383, 384, 390,
Single-cell RNA sequencing (scRNAseq), 378, 380, 381, 400, 404–406, 410, 415, 416, 422, 424, 430, 444,
550 457, 462, 479, 530, 534, 539–541, 551, 552, 555,
Small molecule, 75, 236, 265–267, 475, 476, 534, 552 577
Smoking, 10, 28, 51, 68–70, 322, 372 Vision defects, 272
Spectroscopy, 18, 431, 433 Visual cycle, 10, 16, 18, 207, 290, 311, 372, 380, 490,
Sphingolipid metabolism, 304 534, 536, 539
Splice sites, 99, 175, 180, 184–186 Visual cycle proteins, 534–536
SRD5A3, 450–453 Visual function, 50, 75, 136, 137, 191–193, 293, 351,
Stargardt disease (STGD1), 17, 167, 289–294 389, 391, 416, 417, 421–425, 446, 473, 489, 490,
Stargardt-3 macular dystrophy (STGD3), 258–259 499, 500, 504, 528
Stem cells, 30, 114, 133, 313, 342, 407, 423, 475, 549, Vitelliform macular dystrophy (VMD), 457
553, 572, 577
Stop codons, 29, 105, 150–152, 202, 405
Sub-retinal immune cells, 208–211 W
Subretinal space, 23, 202, 279, 310, 421, 422, 527 Warburg effect, 429

You might also like