You are on page 1of 53

Wilson Disease 1st Edition Edition

Anna Cz■onkowska And Michael L.


Schilsky (Eds.)
Visit to download the full and correct content document:
https://ebookmass.com/product/wilson-disease-1st-edition-edition-anna-czlonkowska-
and-michael-l-schilsky-eds/
HANDBOOK OF CLINICAL
NEUROLOGY

Series Editors

MICHAEL J. AMINOFF, FRANÇOIS BOLLER, AND DICK F. SWAAB

VOLUME 142
ELSEVIER
Radarweg 29, PO Box 211, 1000 AE Amsterdam, Netherlands
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States

© 2017 Elsevier B.V. All rights reserved.

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

This book and the individual contributions contained in it are protected under copyright by the Publisher (other than
as may be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our
understanding, changes in research methods, professional practices, or medical treatment may become necessary.

Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any
information, methods, compounds, or experiments described herein. In using such information or methods they
should be mindful of their own safety and the safety of others, including parties for whom they have a professional
responsibility.

With respect to any drug or pharmaceutical products identified, readers are advised to check the most current
information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to
verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the
responsibility of practitioners, relying on their own experience and knowledge of their patients, to make diagnoses,
to determine dosages and the best treatment for each individual patient, and to take all appropriate safety precautions.

To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for
any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any
use or operation of any methods, products, instructions, or ideas contained in the material herein.

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library

Library of Congress Cataloging-in-Publication Data


A catalog record for this book is available from the Library of Congress

ISBN: 978-0-444-63625-6

For information on all Elsevier publications


visit our website at https://www.elsevier.com/books-and-journals

Publisher: Mara Conner


Editorial Project Manager: Kristi Anderson
Production Project Manager: Sujatha Thirugnana Sambandam
Cover Designer: Alan Studholme

Typeset by SPi Global, India


Handbook of Clinical Neurology 3rd Series

Available titles
Vol. 79, The human hypothalamus: basic and clinical aspects, Part I, D.F. Swaab, ed. ISBN 9780444513571
Vol. 80, The human hypothalamus: basic and clinical aspects, Part II, D.F. Swaab, ed. ISBN 9780444514905
Vol. 81, Pain, F. Cervero and T.S. Jensen, eds. ISBN 9780444519016
Vol. 82, Motor neurone disorders and related diseases, A.A. Eisen and P.J. Shaw, eds. ISBN 9780444518941
Vol. 83, Parkinson’s disease and related disorders, Part I, W.C. Koller and E. Melamed, eds. ISBN 9780444519009
Vol. 84, Parkinson’s disease and related disorders, Part II, W.C. Koller and E. Melamed, eds. ISBN 9780444528933
Vol. 85, HIV/AIDS and the nervous system, P. Portegies and J. Berger, eds. ISBN 9780444520104
Vol. 86, Myopathies, F.L. Mastaglia and D. Hilton Jones, eds. ISBN 9780444518996
Vol. 87, Malformations of the nervous system, H.B. Sarnat and P. Curatolo, eds. ISBN 9780444518965
Vol. 88, Neuropsychology and behavioural neurology, G. Goldenberg and B.C. Miller, eds. ISBN 9780444518972
Vol. 89, Dementias, C. Duyckaerts and I. Litvan, eds. ISBN 9780444518989
Vol. 90, Disorders of consciousness, G.B. Young and E.F.M. Wijdicks, eds. ISBN 9780444518958
Vol. 91, Neuromuscular junction disorders, A.G. Engel, ed. ISBN 9780444520081
Vol. 92, Stroke – Part I: Basic and epidemiological aspects, M. Fisher, ed. ISBN 9780444520036
Vol. 93, Stroke – Part II: Clinical manifestations and pathogenesis, M. Fisher, ed. ISBN 9780444520043
Vol. 94, Stroke – Part III: Investigations and management, M. Fisher, ed. ISBN 9780444520050
Vol. 95, History of neurology, S. Finger, F. Boller and K.L. Tyler, eds. ISBN 9780444520081
Vol. 96, Bacterial infections of the central nervous system, K.L. Roos and A.R. Tunkel, eds. ISBN 9780444520159
Vol. 97, Headache, G. Nappi and M.A. Moskowitz, eds. ISBN 9780444521392
Vol. 98, Sleep disorders Part I, P. Montagna and S. Chokroverty, eds. ISBN 9780444520067
Vol. 99, Sleep disorders Part II, P. Montagna and S. Chokroverty, eds. ISBN 9780444520074
Vol. 100, Hyperkinetic movement disorders, W.J. Weiner and E. Tolosa, eds. ISBN 9780444520142
Vol. 101, Muscular dystrophies, A. Amato and R.C. Griggs, eds. ISBN 9780080450315
Vol. 102, Neuro-ophthalmology, C. Kennard and R.J. Leigh, eds. ISBN 9780444529039
Vol. 103, Ataxic disorders, S.H. Subramony and A. Durr, eds. ISBN 9780444518927
Vol. 104, Neuro-oncology Part I, W. Grisold and R. Sofietti, eds. ISBN 9780444521385
Vol. 105, Neuro-oncology Part II, W. Grisold and R. Sofietti, eds. ISBN 9780444535023
Vol. 106, Neurobiology of psychiatric disorders, T. Schlaepfer and C.B. Nemeroff, eds. ISBN 9780444520029
Vol. 107, Epilepsy Part I, H. Stefan and W.H. Theodore, eds. ISBN 9780444528988
Vol. 108, Epilepsy Part II, H. Stefan and W.H. Theodore, eds. ISBN 9780444528995
Vol. 109, Spinal cord injury, J. Verhaagen and J.W. McDonald III, eds. ISBN 9780444521378
Vol. 110, Neurological rehabilitation, M. Barnes and D.C. Good, eds. ISBN 9780444529015
Vol. 111, Pediatric neurology Part I, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444528919
Vol. 112, Pediatric neurology Part II, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444529107
Vol. 113, Pediatric neurology Part III, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444595652
Vol. 114, Neuroparasitology and tropical neurology, H.H. Garcia, H.B. Tanowitz and O.H. Del Brutto, eds.
ISBN 9780444534903
Vol. 115, Peripheral nerve disorders, G. Said and C. Krarup, eds. ISBN 9780444529022
Vol. 116, Brain stimulation, A.M. Lozano and M. Hallett, eds. ISBN 9780444534972
Vol. 117, Autonomic nervous system, R.M. Buijs and D.F. Swaab, eds. ISBN 9780444534910
Vol. 118, Ethical and legal issues in neurology, J.L. Bernat and H.R. Beresford, eds. ISBN 9780444535016
Vol. 119, Neurologic aspects of systemic disease Part I, J. Biller and J.M. Ferro, eds. ISBN 9780702040863
Vol. 120, Neurologic aspects of systemic disease Part II, J. Biller and J.M. Ferro, eds. ISBN 9780702040870
Vol. 121, Neurologic aspects of systemic disease Part III, J. Biller and J.M. Ferro, eds. ISBN 9780702040887
Vol. 122, Multiple sclerosis and related disorders, D.S. Goodin, ed. ISBN 9780444520012
Vol. 123, Neurovirology, A.C. Tselis and J. Booss, eds. ISBN 9780444534880
vi AVAILABLE TITLES (Continued)
Vol. 124, Clinical neuroendocrinology, E. Fliers, M. Korbonits and J.A. Romijn, eds. ISBN 9780444596024
Vol. 125, Alcohol and the nervous system, E.V. Sullivan and A. Pfefferbaum, eds. ISBN 9780444626196
Vol. 126, Diabetes and the nervous system, D.W. Zochodne and R.A. Malik, eds. ISBN 9780444534804
Vol. 127, Traumatic brain injury Part I, J.H. Grafman and A.M. Salazar, eds. ISBN 9780444528926
Vol. 128, Traumatic brain injury Part II, J.H. Grafman and A.M. Salazar, eds. ISBN 9780444635211
Vol. 129, The human auditory system: Fundamental organization and clinical disorders, G.G. Celesia
and G. Hickok, eds. ISBN 9780444626301
Vol. 130, Neurology of sexual and bladder disorders, D.B. Vodušek and F. Boller, eds. ISBN 9780444632470
Vol. 131, Occupational neurology, M. Lotti and M.L. Bleecker, eds. ISBN 9780444626271
Vol. 132, Neurocutaneous syndromes, M.P. Islam and E.S. Roach, eds. ISBN 9780444627025
Vol. 133, Autoimmune neurology, S.J. Pittock and A. Vincent, eds. ISBN 9780444634320
Vol. 134, Gliomas, M.S. Berger and M. Weller, eds. ISBN 9780128029978
Vol. 135, Neuroimaging Part I, J.C. Masdeu and R.G. González, eds. ISBN 9780444534859
Vol. 136, Neuroimaging Part II, J.C. Masdeu and R.G. González, eds. ISBN 9780444534866
Vol. 137, Neuro-otology, J.M. Furman and T. Lempert, eds. ISBN 9780444634375
Vol. 138, Neuroepidemiology, C. Rosano, M.A. Ikram and M. Ganguli, eds. ISBN 9780128029732
Vol. 139, Functional neurologic disorders, M. Hallett, J. Stone and A. Carson, eds. ISBN 9780128017722
Vol. 140, Critical care neurology Part I, E.F.M. Wijdicks and A.H. Kramer, eds. ISBN 9780444636003
Vol. 141, Critical care neurology Part II, E.F.M. Wijdicks and A.H. Kramer, eds. ISBN 9780444635990

All volumes in the 3rd Series of the Handbook of Clinical Neurology are published electronically, on Science Direct:
http://www.sciencedirect.com/science/handbooks/00729752.
Foreword

It is a pleasure to present this volume of the Handbook of Clinical Neurology (HCN), which is dedicated to Wilson
disease. There are plenty of eponyms in medicine and particularly in neurology, but very few HCN volumes include a
person’s name in their title and this is only the third in the present series, after Parkinson and Huntington. Kinnier
Wilson (1878–1937) fully deserves this honor. In 1912, he published in Brain what had been his MD thesis:
“Progressive lenticular degeneration: a familial nervous system disease associated with cirrhosis of the liver.” As is
almost always the case, similar conditions associating liver and brain disease had been described previously, going
as far back as Morgagni and, more recently, by Westphal and Strumpell. However, the Wilson monograph included
the main clinical and pathologic features of the disease, and it opened the way to a new chapter in the history of
neurology, namely that of extrapyramidal system disorders, a term Wilson introduced.
The volume opens with a chapter describing in detail the history of the disease written by one of its protagonists,
John M. Walshe, who in 1956 proposed the use of penicillamine, one of the first disease-modifying drugs in neurology.
The volume includes chapters dealing with the epidemiology and genetics of the disease. It focuses on the two main
organs affected, the brain and the liver, with detailed descriptions of the pathology and pathogenesis of the disease,
including animal models, as well as its diagnosis and the various available forms of management and treatment. Novel
perspectives are discussed in a chapter that introduces a new chelating agent, tetrathiomolybdate, as well as nonchelat-
ing drugs currently under clinical investigation. As is appropriate for such a rare yet highly complex hereditary disease,
the volume concludes with a chapter on the groups that advocate for and support patients with Wilson disease.
We have been fortunate to have as volume editors two distinguished scholars and clinicians, Anna Członkowska,
Professor at the Second Department of Neurology, Institute of Psychiatry and Neurology in Warsaw, and Michael L.
Schilsky, Associate Professor of Medicine and Surgery and Medical Director of Adult Liver Transplantation at the
Yale-New Haven Transplantation Center. Both have been on the forefront of research on Wilson disease for many
years. They have assembled a truly international group of authors with acknowledged expertise and together they have
produced this authoritative, comprehensive, and up-to-date volume. Its availability electronically on Elsevier’s Science
Direct site as well as in print format should ensure its ready accessibility and facilitate searches for specific information.
We are grateful to them and to all the contributors for their efforts in creating such an invaluable resource. As series
editors we read and commented on each of the chapters with great interest. We are therefore confident that both
clinicians and researchers in many different medical disciplines will find much in this volume to appeal to them.
And last, but not least, we thank Elsevier, our publisher – and in particular Michael Parkinson in Scotland, and Mara
Conner and Kristi Anderson in San Diego – for their unfailing and expert assistance in the development and production
of this volume.
Michael J. Aminoff
François Boller
Dick F. Swaab
Preface

There is almost no other treatable disorder with as wide a clinical spectrum for presentation as Wilson disease. From
child to septuagenarian at diagnosis, patients may be asymptomatic or present with advanced cirrhosis or liver failure,
with a movement disorder ranging from mild tremor or bradykinesia to severe dystonia, or with an affective disorder or
psychosis. Although this is a volume in a series focused on neurologic and neuropsychiatric disorders, it was inescap-
able that we cross between disciplines and consider the full spectrum of the disease, the underlying hepatic disease that
accompanies the disorder, the diverse neurologic presentations, and the fascinating history of the discovery of its basis
and treatment.
Recent advances in molecular genetics testing allow easier and faster diagnosis of Wilson disease, for which treat-
ment may prevent, stop, or even reverse tissue injury. Also introduced more recently are imaging techniques that help
estimate the stage of the disease at the time of diagnosis and which are helpful for monitoring the progress of treatment.
With a better understanding of the disease pathogenesis, there are new opportunities for safer and even more effective
therapy.
The aim of this text was to provide a thorough examination of Wilson disease in an easy-to-digest format. To accom-
plish this, we engaged a group of international experts from Europe and North America and asked for their help in
covering a wide range of relevant topics important for all caregivers of patients with Wilson disease, whether pediatric
or adult physicians, neurologists, psychiatrists, or hepatologists.
The text begins with a historic perspective, and then provides overviews of disease pathogenesis and its underlying
genetic basis, the effects of the disease on both the central nervous system and liver, its diagnosis, and the range of
clinical disease and treatments. Unusually, we have included the viewpoint of a lay patient organization on what is
important from a patient and advocacy perspective.
We thank all our contributors for their time and help in putting together this volume. A number of revisions were
necessary to make the handbook more cohesive, and we appreciate their patience and forbearance, which made this
possible.
We would like particularly to acknowledge John M. Walshe for his historic perspective and his contributions to the
field, and Mary L. Graper for her advocacy for all patients with Wilson disease. We especially thank all of our
coworkers, with whom we have worked over the years to assist hundreds of patients, broaden our personal experience,
and share it with others. We are also grateful for the cooperation of our patients and their families who put their trust in
us and from whom we continue to learn.
Anna Członkowska and Michael L. Schilsky
Contributors

A. Ahmad First Faculty of Medicine and General University


Section of Transplantation and Immunology, Hospital in Prague, Prague, Czech Republic and Institute
Department of Surgery, Yale University School of of Neuroradiology, University Medicine Goettingen,
Medicine, New Haven, CT, USA Goettingen, Germany


A. Ala K. Dziezyc
Department of Clinical and Experimental Medicine, Second Department of Neurology, Institute of Psychiatry
Faculty of Health and Medical Sciences, University of and Neurology, Warsaw, Poland
Surrey and Department of Gastroenterology and
Hepatology, Royal Surrey County Hospital NHS P. Ferenci
Foundation Trust, Guildford, Surrey, UK Department of Internal Medicine 3, Gastroenterology
and Hepatology, Medical University of Vienna, Vienna,
O. Bandmann Austria
Sheffield Institute for Translational Neuroscience
(SITraN), University of Sheffield, Sheffield, UK M.L. Graper
Wilson Disease Association, Milwaukee, WI, USA
S. Boga
Department of Gastroenterology, Sisli Etfal Education S.H. Hahn
and Research Hospital, Istanbul, Turkey Division of Genetic Medicine, Department of Pediatrics,
University of Washington School of Medicine, Seattle
R. Brůha Children’s Hospital, Seattle, WA, USA
Fourth Department of Internal Medicine, Charles
University in Prague, First Faculty of Medicine and General D. Huster
University Hospital in Prague, Prague, Czech Republic Department of Gastroenterology and Oncology,
Deaconess Hospital Leipzig, Leipzig, Germany
G. Chabik
Second Department of Neurology, Institute of Psychiatry T. Litwin
and Neurology, Warsaw, Poland Second Department of Neurology, Institute of Psychiatry
and Neurology, Warsaw, Poland
I.J. Chang
Division of Medical Genetics, Department of Medicine, C. Lo
University of Washington School of Medicine, Seattle, Sheffield Institute for Translational Neuroscience
WA, USA (SITraN), University of Sheffield, Sheffield, UK

A. Członkowska V. Medici
Second Department of Neurology, Institute of Psychiatry and Division of Gastroenterology and Hepatology,
Neurology and Department of Experimental and Clinical Department of Internal Medicine, University of
Pharmacology, Medical University of Warsaw, Poland California Davis, Sacramento, CA, USA

P. Dušek J. Mikol
Department of Neurology and Center of Clinical Pathology Department, Paris Diderot University, Paris,
Neuroscience, Charles University in Prague, France
xii CONTRIBUTORS
J. Pfeiffenberger P. Socha
Department of Gastroenterology and Hepatology, Departments of Gastroenterology, Hepatology,
University Hospital of Heidelberg, Heidelberg, Nutritional Disorders and Pediatrics, The Children’s
Germany Memorial Health Institute, Warsaw, Poland

A. Poujois W. Stremmel
French National Reference Centre for Wilson Disease, Department of Gastroenterology and Hepatology,
Neurology Department, Lariboisière Hospital, Paris, University Hospital of Heidelberg, Heidelberg,
France Germany

M. Pronicki E. Torrazza-Perez
Department of Pathology, The Children’s Memorial Division of Gastroenterology and Hepatology,
Health Institute, Warsaw, Poland Department of Internal Medicine, University of New
Mexico, Albuquerque, USA
E.A. Roberts
Departments of Paediatrics, Medicine and Pharmacology J.M. Walshe
and Toxicology, University of Toronto, Toronto, Canada Formerly of Addenbrookes Hospital, Cambridge and the
Middlesex Hospital, London, UK
C. Rupp
Department of Internal Medicine, University Hospital K.-H. Weiss
Heidelberg, Heidelberg, Germany Department of Gastroenterology and Hepatology,
University Hospital of Heidelberg, Heidelberg, Germany
I.F. Scheiber
Department of Parasitology, Faculty of Science, Charles F. Woimant
University, Prague, Czech Republic French National Reference Centre for Wilson Disease,
Neurology Department, Lariboisière Hospital, Paris,
M.L. Schilsky France
Section of Digestive Diseases and Transplantation and
Immunology, Department of Medicine and Surgery, Yale P. Zimbrean
University School of Medicine, New Haven, CT, USA Departments of Psychiatry and Surgery (Transplant),
Yale University, New Haven, CT, USA
J. Seniów
Second Department of Neurology, Institute of Psychiatry
and Neurology, Warsaw, Poland
Handbook of Clinical Neurology, Vol. 142 (3rd series)
Wilson Disease
A. Członkowska and M.L. Schilsky, Editors
http://dx.doi.org/10.1016/B978-0-444-63625-6.00001-X
© 2017 Elsevier B.V. All rights reserved

Chapter 1

History of Wilson disease: a personal account


JOHN M. WALSHE*
Formerly of Addenbrookes Hospital, Cambridge and the Middlesex Hospital, London, UK

Abstract
This chapter focuses on the historic aspects of the development of much of our current knowledge of the
diagnosis and treatment of Wilson disease. Included are descriptions of the clinical signs of neurologic and
hepatic disease, the natural history of disease progression, studies of disease pathogenesis and a unique
perspective on the development of diagnostic testing and pharmacological therapy.

“Begin at the beginning,” the King said very gravely, they later became associated (Kayser, 1902). It is surpris-
“then go on until the end, then stop.” ing, in view of the very detailed nature of his descriptions
Alice’s Adventures Through the Looking Glass, of his patients, that Wilson did not observe the corneal
Lewis Carroll rings and indeed he refused to admit of their relationship
to his disease for 10 years after his original publication
This admirable advice for any story teller is honored (Wilson, 1922). These rings were for many years thought
more often in the breach than the observance. The diffi- to pathognomonic of Wilson disease but it is now known
culty remains, where is the beginning and does the story that they can be found in other forms of liver disease,
have any definitive end? With the history of disease the such as primary biliary cirrhosis and chronic cholestasis
probability is that there is, in all likelihood, no end. (Fleming et al., 1975).
Knowledge and theories will continue to develop almost Only 1 year after Wilson’s original publication in
indefinitely. Well then, is there a beginning? Probably, Brain, the Austrian pathologist, Rumpel, reported find-
but exactly where that is, is not always obvious. In this ing an excess of copper in the liver of a patient dying
case, it might seem self-evident that the story begins with of Wilson disease, an observation whose importance
Wilson’s original article in Brain in 1912, when he was completely missed (Rumpel, 1913). He also sug-
described four cases of what he called “progressive len- gested that there was excess of silver in the eyes.
ticular degeneration: a familial nervous disease associ- Another important observation which passed unno-
ated with cirrhosis of the liver.” (Fig. 1.1) In addition ticed was by Bramwell, whose house physician Wilson
he found four cases in the literature which fitted his had once been (Bramwell, 1916). Bramwell described
description, one of which was dated back to 1890. Fur- a family in which four siblings died, between the ages
thermore, there is a case report in Frerichs’ book of of 9 and 14 years, of rapid-onset liver failure and sug-
1860 that was almost certainly a case of Wilson disease. gested that this might be related to Wilson disease. It
Indeed, going back even further, Morgagni, in 1761, would be interesting to see if any descendants of this fam-
described several cases of liver disease associated with ily still exist so that they could be tested to see if they
nervous symptoms which may possibly have been cases carry, in single dose, the mutation for Wilson disease.
of Wilson disease. The next important addition to knowledge came when
It is interesting that one of the defining physical signs Hall (1921) showed that this disease was inherited in a
of Wilson disease, the Kayser–Fleischer corneal rings, recessive mode, later confirmed in more detail by Bearn
had been described 10 years before the disease to which (1960), who studied 30 families he saw in New York

*Correspondence to: J.M. Walshe, MD, ScD, FRCP, 58 High Street Hemingford Grey, Huntingdon PE 28 9BN, UK.
E-mail: penicillamine@waitrose.com
2 J.M. WALSHE
copper excreted in the urine. This important observation
was followed up in 1951 by Cumings himself in London
and by Denny Brown and Porter (1951) in Boston. Both
teams showed that courses of BAL resulted in significant
improvement in their patients’ neurologic symptoms.
However it soon became apparent that patients needed
repeated courses of the drug and subsequent courses
were less effective than the initial one. In addition the
injections were painful and associated with a wide vari-
ety of toxic reactions. This was clearly not an ideal
treatment.
Fig. 1.1. Dr. John Walshe (right) with Dr. James Kinner The picture changed when, in 1956, I reported that
Wilson, son of neurologist Samuel Alexander Kinner Wilson, penicillamine promoted a much larger excretion of cop-
whose 1912 publication on hepatolenticular degeneration was per in the urine than either BAL or the other medical che-
being celebrated at the Centennial Symposium on Wilson’s
lating agent ethylenediamine tetraacetic acid (EDTA)
disease in London, October 5–6, 2012.
(Walshe, 1956, 1960). Penicillamine is a breakdown
product of penicillin and is excreted in the urine of all
whose ancestry could be traced back to Eastern Europe or patients treated with penicillin, an observation I had
Southern Italy. made some years earlier when studying changes in amino
It must be noted that in Wilson’s original publication acid metabolism in patients with liver damage (Walshe,
of only 4 patients, he described almost all of the signs and 1956). This idea came to me when working in the Liver
symptoms which we now know are characteristic of this Unit under Dr. Charles Davidson, at the Boston City
disease (Wilson, 1912). He carried out his own patho- Hospital. Having seen one of Professor Denny Brown’s
logic studies and even traveled to Switzerland to collect Wilson disease patients who was not prospering on treat-
the brain of a patient dying of his disease. Wilson did not ment with BAL, it occurred to me that penicillamine,
believe that the liver pathology was a significant factor in with its –SH and –NH3 groups, might have the right
the natural history of the disease, although one of his own chemical structure to chelate copper and promote its
patients actually died of bleeding esophageal varices. excretion in the urine. Dr. Davidson was able to obtain
Over the next 30 years there were only minor for me 2 grams of penicillamine from Professor Sheehan
advances in the understanding of the disease, whose at the Massachusetts Institute of Technology. Having
course remained remorseless and invariably fatal. In taken 1 gram myself to prove its safety, I administered
1922 Siemerling and Oloff described the association of the second gram to Professor Denny Brown’s patient
sunflower cataracts with Kayser–Fleischer rings and and recorded the very satisfactory cupresis so induced.
noted the similarity to lesions caused by intraocular dam- With this simple procedure the start of penicillamine
age with intraocular copper fragments. Vogt (1929) and therapy was launched.
Haurowitz (1930) reported finding excess copper in the In those far-off days there were no ethical committees
brain and liver of Wilson disease patients – an observa- to be approached for permission; life was a lot easier
tion which was not followed up. when trying out new ideas. By 1960 I was able to publish
In 1945 Sir Rudolph Peters and his team in the first account of a patient with Wilson disease improv-
Oxford were able to report their earlier work in the devel- ing significantly as a result of this new therapy (Walshe,
opment of the antiarsenical drug, dimercaprol (British 1960). This was confirmed by similar studies by
anti-Lewisite, BAL). This was developed during the Sec- Scheinberg and Sternlieb, also in 1960.
ond World War to combat anticipated attack by Hitler Whilst these important advances in the treatment of
with the arsenical gas Lewisite. The importance of this this disease were progressing, so also was our under-
became apparent in 1948 when Cumings showed that standing of its pathogenesis. In 1948 Holmberg and
Wilson disease was indeed due to accumulation of excess Laurell described the finding of a copper-carrying pro-
copper in the brain and liver of all patients dying of this tein in the plasma which they named ceruloplasmin. In
disease and postulated that treatment with BAL might 1952, working independently in New York, both Bearn
halt the progress of the illness (Cumings, 1951). By coin- and Kunkel and Scheinberg and Gitlin showed that
cidence, at the same time, Mandelbrote et al. (1948) patients with Wilson disease all had low or absent con-
reported their findings on the effect of BAL on copper centrations of this protein (Bearn and Kunkel,1952;
excretion on a wide spectrum of patients with neurologic Scheinberg and Gitlin, 1952). Shortly after this
diseases; one of these patients had Wilson disease and in Cartwright and his team (1954) in Salt Lake City pub-
this particular patient there was a great increase in the lished an important article on copper metabolism in
HISTORY OF WILSON DISEASE: A PERSONAL ACCOUNT 3
Wilson disease, an article which inspired my own interest undergone cholecystectomy for gallstones, that the dif-
in this malady, in which they discussed the various ther- ference in copper excretion in the bile was of an order
apies then available and their shortcomings. Despite of magnitude between normal individuals and patients
these real advances Uzman, working in Denny Brown’s (Gibbs and Walshe, 1980). Furthermore, by studying
department in Boston, believed that Wilson disease was the difference in urine copper concentration in pigment
due to an abnormality of peptide metabolism and that stones the mean figure for copper in patients was
copper deposition was a side-effect and not the directly 50 mg/g compared with the figure of 1072.5 mg/g in con-
causative etiology of the disease (Uzman et al., 1956). trols (Walshe, 1998).
This hypothesis was later firmly disproved by Asator The first description of hemolysis and a serious com-
et al. (1976) using a technique more sophisticated than plication of Wilson disease was first described by Profes-
that available to Uzman. Although Uzman was wrong sor Sherlock and her team at the Royal Free Hospital in
on this point, he was able to confirm Bramwell’s hypoth- London (Sherlock et al., 1968), whilst Roche-Sicot and
esis that Wilson disease could present as liver disease in Benhamou (1977) described the first case of massive
children before the onset of neurologic signs (Bramwell, hepatic necrosis and hemolysis, thus proposing the idea
1916; Uzman et al., 1956; Chalmers et al., 1957). of acute fatal Wilson disease. Recently I showed that
The clinical picture of the disease was widened by hemolysis was to be found in 6.9% of 321 patients and
Bearn, who showed that there were abnormalities of that the hemolysis was extravascular, the age of onset
renal function and skeletal lesions in many patients was 12.6 years, and there was a female-to-male ratio of
(Bearn, 1957). He and Kunkel also introduced the idea 2:1 (Walshe, 2013).
of using radiolabeled copper, the short half-isotope, to Again, in the 1960s, in addition to our increased
study copper metabolism in Wilson disease patients understanding of the pathogenesis of Wilson disease
(Bearn and Kunkel, 1954). About the same time the role was an increased range of therapies which favorably
of ceruloplasmin in the pathogenesis was hotly debated influenced the prognosis. In 1961 Schouwink reported
and Uzman and Hood (1952) rightly pointed out that that zinc salts could block the uptake of copper from
the concentrations of ceruloplasmin found in Wilson dis- the intestine and thereby induce, although very slowly,
ease patients was quite variable and frequently over- a negative copper balance and this was later taken up
lapped that found in symptomless carriers of the enthusiastically by Hoogenraad et al. (1979) in the
Wilson disease gene in single dose. Netherlands and by Brewer et al. (1981) in the United
In 1959 Cumings published a book, Heavy Metals States.
and the Brain, in which he tried to cover the entire liter- In an attempt to overcome the toxic side-effects of
ature of Wilson disease to date and this remains an penicillamine, which were becoming apparent by the
invaluable historical source of information for those late 1960s (Walshe, 1968), with the invaluable help of
interested in the history of the disease, as also does Dr. Hal Dixon, I proposed the use of triethylene tetramine
Scheinberg and Sternlieb’s (1984) monograph. 2HCl as an alternative chelating agent and this proved to
Beginning in the 1960s I realized the potential of be an effective therapy and remarkably free of side-
using radiocopper in the investigation of Wilson disease, effects (Walshe, 1982). The next advance was the
working first with Osborn, and later with Potter (Osborn introduction of liver transplantation by Starzl and his
and Walshe, 1965, 1967; Walshe and Potter, 1977). team in 1971 (DuBois et al., 1971). This approach actu-
Together we devised a method of determining the uptake ally cured the Wilson disease so patients no longer
of copper by the liver and its further distribution through- needed chelation treatment, but they did require lifelong
out the body (Osborn and Walshe, 1967). It was thus pos- immunosuppression.
sible to show that in the presymptomatic stages of the The final advance came with the use of ammonium
disease the liver had a high affinity for the metal and tetrathiomolybdate (Walshe, 1986). I first took this
90% of the injected dose was present in the liver within myself for a week to test its safety before giving it to a
24 hours. As the disease progressed the liver uptake of patient who had proved intolerant to penicillamine and
radiocopper became less and less effective and the trientine and had found the side-effects of zinc salts intol-
isotope could be found distributed through many other erable. Within a year it had effectively decoppered her
tissues. However treatment with chelating agents effec- liver and changed the histology from a fatty liver with
tively decoppered the liver and restored its ability to cellular infiltrates to normal (Walshe, 1986). It is of inter-
sequester the metal. But in no patient was it possible to est that Bickel et al. (1957), had tried the use of molyb-
show, as in normal individuals, a peak in the lower abdo- date in the 1950s, but they had not used the tetrathio salt
men which represented copper excreted via the bile in to and their preparation had no anticopper properties so the
the intestine (Walshe and Potter, 1977). Later it was possibilities of this form of therapy were set back some
possible to show, in patients and controls, who had 30 years. They had relied on the observation that sheep
4 J.M. WALSHE
fed on pasture contaminated with molybdenum devel- Bearn AG (1957). Wilson’s disease. An inborn error of metab-
oped copper deficiency but failed to realize that in the olism with multiple manifestations. Am J Med 22: 747–764.
rumen of the sheep the MoO4 was converted, by reducing Bearn AG (1960). A genetical analysis of 30 families with
conditions, to MoS4, an action which does not take place Wilson’s disease (hepatolenticular degeneration). Ann
Hum Genet 24: 33–43.
in the human gut.
Bearn AG, Kunkel HG (1952). Biochemical abnormalities in
The most recent, and perhaps the most important,
Wilson’s disease. J Clin Invest 31: 616.
breakthrough came in 1993 when three separate groups Bearn AG, Kunkel HG (1954). Localisation of 64Cu in serum
(Bull et al., 1993; Tanzi et al., 1993; Yamaguchi et al., fractions folloqwing oral administration in Wilson’s dis-
1993) reported in Nature Genetics that they had ease. Proc Soc Expl Biol 85: 44–48.
identified the Wilson disease gene as a P-type APTase, Bickel H, Neale FC, Hall G (1957). A clinical and biochemical
140-kDa copper-transporting enzyme located on chro- study of hepatolenticular degeneration (Wilson’s disease).
mosome 13q14. There have been no further important QJM 26: 527–558.
advances since then, except for the identification of more Bramwell B (1916). Familial cirrhosis of the liver; four cases
than 500 mutations of this gene. As most patients are of acute fatal cirrhosis in the same family, the patients being
compound heterozygotes the number of permutations respectively nineteen, fourteen and fourteen years of age;
suggested relationship to Wilson’s progressive degenera-
and combinations is enormous and will make
tion of the lenticular nucleus. Edin Med J 17: 90–99.
phenotype–genotype correlations almost impossible.
Brewer GJ, Prasad AS, Cossack ZT et al. (1981). Treatment of
Having brought the story up to date the question Wilson’s disease with oral zinc. Clin Res 29: 758.
remains, what of the future? It would seem that, as far Bull PC, Thomas GR, Romanes JN et al. (1993). The Wilson’s
as natural history and biochemistry and genetics are con- disease gene is a putative copper transporting P-type
cerned, the disease is worked out. However therapy is far ATP-ase similar to the Menkes gene. Nat Genet 5:
from ideal. The initial therapy with BAL still has a place 327–337.
for severely dystonic patients but it has too many Cartwright GE, Hodges RE, Gubler CJ et al. (1954). Studies on
side-effects and its administration is something of an copper metabolism. Hepatolenticular degeneration. J Clin
ordeal for the patient. Penicillamine is effective in many Invest 33: 1487–1501.
cases but also has too many side-effects, mostly immu- Chalmers TC, Iber FL, Uzman LL (1957). Hepatolenticular
degeneration (Wilson’s disease) as a form of idiopathic
nogenic in nature and, in a small percentage of patients,
cirrhosis. N Engl J Med 256: 235–242.
can induce a sudden deterioration which is, unfortu-
Cumings JN (1951). The effect of BAL in hepatodegeneration.
nately, unpredictable (Walshe, 2011). Trientine has far Brain 74: 10–22.
fewer side-effects and is a very useful alternative Cumings JN (1959). Heavy metals and the brain, Blackwell
(Walshe, 1982) but, like all forms of treatment, is some- Scientific Publications, Oxford.
times associated with initial worsening of symptoms, Denny Brown D, Porter H (1951). The effect of BAL (2.3-
which is always a worrying state of affairs. Tetrathiomo- dimercapto propanol) on hepatolenticular degeneration
lybdate is yet another alternative but is not yet in the (Wilson’s disease). N Engl J Med 245: 917–925.
pharmacopeia so is not readily available. Zinc salts are DuBois RS, Rodgerson DO, Martineau G et al. (1971).
effective in blocking copper absorption from the gut Orthopic liver transplantation for Wilson’s disease.
but can only produce a very slow negative copper bal- Lancet 1: 505–508.
Fleming CR, Dickson ER, Hellenhorst ER et al. (1975).
ance and are probably more useful for maintenance than
Pigmented corneal rings in a patient with biliary cirrhosis.
initial therapy. Many zinc salts are gastric irritants and are
Gastroenterology 69: 220–225.
probably contraindicated in patients with prominent Frerichs FT (1860). In: C Murchison (Ed.), Clinical treatise
esophageal varices. Finally there is liver transplantation, on diseases of the liver, Vol. 1. New Sydenham Society,
in effect a cure, but which condemns the patient to London.
long-term immunosuppression with all its possible com- Gibbs K, Walshe JM (1980). Biliary excretion of copper in
plications. Its use suggests a failure of either diagnosis or Wilson’s disease. Lancet 2: 538–539.
initial treatment. One day it may be possible to offer gene Hall HC (1921). La degenerescence hepatolenticulare.
replacement but this is not yet even on the horizon. Maladie de Wilson-pseudosclerose, Mason, Paris.
It remains to be seen what the future holds for patients Haurowitz F (1930). Uber eine anamolides
with Wilson disease. Kupferstoffwechsels. Hoppe-Seyl Z 190: 72–74.
Holmberg CG, Laurell CB (1948). Investigations on serum
copper II. Isolation of the copper containing protein and a
REFERENCES description of properties. Acta Chem Scand 85: 550–556.
Hoogenraad TU, Koevoet R, du Ruyter Korvet EG (1979).
Asator AM, Milne MD, Walshe JM (1976). The urinary excre- Oral zinc sulphate as long term treatment in Wilson’s dis-
tion of peptides and hydroxyproline in Wilson’s disease. ease (hepatolenticular degeneration). Eur Neurol 18:
Clin Sci Mol Med 51: 369–378. 205–211.
HISTORY OF WILSON DISEASE: A PERSONAL ACCOUNT 5
Kayser B (1902). Uber einen Fall von angeborener Tanzi R, Petrukhin K, Chernov I et al. (1993). The Wilson’s
Grunlicherverfarbung der Cornea. Kiln Mbl Augenheilk disease gene is a copper transporting ATPase with homol-
40: 22–25. ogy to the Menkes gene. Nat Genet 5: 44–50.
Mandelbrote BM, Stanier MW, Thompson RHS et al. (1948). Uzman LL, Hood B (1952). The familial nature of the
Studies on copper metabolism in demyelinating diseases. amino-aciduria of Wilson’s disease. Am J Med Sci 223:
Brain 71: 212–228. 392–400.
Morgagni GB (1761). De sedibus et causis morborum, Trans Uzman LL, Iber FL, Chalmers TC et al. (1956). The mecha-
Alexander B, London. nism of copper deposition in the liver in hepatolenticular
Osborn SB, Walshe JM (1965). Studies with radiocopper degeneration (Wilson’s disease). Am J Med Sci 231:
(64Cu) in Wilson’s disease; the dynamics of copper trans- 511–518.
port. Clin Sci 29: 575–581. Vogt A (1929). Kupfer und siber aufgespeichert in Auge, leber
Osborn SB, Walshe JM (1967). Studies with radioactive cop- Milz und Nieren als Symptom der Peudoskerlose. Klin Mbl
per (64Cu, 67Cu) in relation to the natural history of Augenheilk 83: 417–419.
Wilson’s disease. Lancet 1: 346–350. Walshe JM (1956). Penicillamine, a new oral therapy for
Peters RA, Stocken LA, Thompson RHS (1945). BAL. Nature Wilson’s disease. Am J Med 21: 487–495.
156: 616. Walshe JM (1960). The treatment of Wilson’s disease with
Roche-Sicot J, Benhamou JN (1977). Acute intravascular penicillamine. Lancet 1: 188–192.
harmolysis and acute liver failure associated as a first man- Walshe JM (1968). Toxic reactions to penicillamine in patients
ifestation of Wilson’s disease. Ann Intern Med 86: with Wilson’s disease. Postgrad Med J 6–8. Suppl.
301–303. Walshe JM (1982). Triethylene tetramine dihydrochloride: a
Rumpel A (1913). Ueber das Wesen und die Bedeutung der new chelating agent for copper. In: FE Karch (Ed.),
Leberveranderungen und der Pigmentierungen bei den Orphan Drugs, Marcel Dekker, New York.
damit verbundenen Fallen von pseudosclerose (Westphal- Walshe JM (1986). Tetrathiomolybdate (MoS4) as an anti
Strumpell). Deutche Zeitschrift Nervenheilkde 49: 54–73. copper agent in man. In: IH Scheinberg, JM Walshe
Scheinberg IH, Gitlin D (1952). Deficiency of ceruloplasmin (Eds.), Orphan Diseases and Orphan Drugs. Manchester
in patients with hepatolenticular degeneration (Wilson’s University Press, Manchester.
disease). Science 116: 484–485. Walshe JM (1998). Wilson disease: gall stone copper follow-
Scheinberg IH, Sternlieb I (1960). Long term management of ing liver transplantation. Ann Clin Biochem 35: 681–682.
hepatolenticular degeneration (Wilson’s disease). Am Walshe JM (2011). Penicillamine neurotoxicity: a hypothesis.
J Med 29: 316–333. ISRN Neurology. Article ID 464572.
Scheinberg IH, Sternlieb I (1984). Wilson’s disease, Saunders, Walshe JM (2013). The acute haemolytic syndrome in
Philadelphia. Wilson disease – a review of 22 patients. Q J Med 106:
Schouwink G (1961). De hepatocerebrale degeneratie, met 1003–1008.
een onderzoek nar zinkstofwisseling. In: University of Walshe JM, Potter G (1977). The pattern of the whole body
Amsterdam, Thesis, MD. distribution of radioactive copper (67Cu, 64Cu) in
Sherlock S, McIntyre N, Clink HM et al. (1968). Wilson’s disease and various control groups. Q J Med
Haemolytic anaemia in Wilson’s disease. In: S Bergsma, 46: 445–462.
IH Scheinberg, I Sternlieb (Eds.), Wilson’s disease. Wilson SAK (1912). Progressive lenticular degeneration; a
Birth defects, original article series, Vol. IV. National familial nervous disease associated with cirrhosis of the
Foundation - March of Dimes, New York, pp. 99–102. liver. Brain 34: 295–507.
Siemerling E, Oloff H (1922). Pseudosclerose (Westphal– Wilson SAK (1922). La maladie de Wilson. In: P Marie (Ed.),
Strumpell) mit Cornealring (KayserFleischer) und doppel- Question Neurologique d’Actualitie. Mason, Paris.
seitger Scheinkatarakt dienurbie Seitlicherbeuchtung Yamaguchi Y, Heiny M, Gitlib JD (1993). Isolation and char-
sichtbar ist und die, der nach Verletzung durch kupfersplit- acterisation of human liver cDNA as a candidate gene for
terentstehenden Katarakt ahnlich ist. Klin Wschr 1: Wilson’s disease. Biochem Biophys Res Commun 197:
1087–1089. 271–277.
Handbook of Clinical Neurology, Vol. 142 (3rd series)
Wilson Disease
A. Członkowska and M.L. Schilsky, Editors
http://dx.doi.org/10.1016/B978-0-444-63625-6.00002-1
© 2017 Elsevier B.V. All rights reserved

Chapter 2

Epidemiology and introduction to the clinical presentation


of Wilson disease

CHRISTINE LO AND OLIVER BANDMANN*


Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK

Abstract
Our understanding of the epidemiology of Wilson disease has steadily grown since Sternlieb and
Scheinberg’s first prevalence estimate of 5 per million individuals in 1968. Increasingly sophisticated genetic
techniques have led to revised genetic prevalence estimates of 142 per million. Various population isolates
exist where the prevalence of Wilson disease is higher still, the highest being 885 per million from within the
mountainous region of Rucar in Romania. In Sardinia, where the prevalence of Wilson disease has been cal-
culated at 370 per million births, six mutations account for around 85% of Wilson disease chromosomes
identified. Significant variation in the patterns of presentation may however exist, even between individuals
carrying the same mutations. At either extremes of presentation are an 8-month-old infant with abnormal liver
function tests and individuals diagnosed in their eighth decade of life. Three main patterns of presentation
have been recognized – hepatic, neurologic, and psychiatric – prompting their presentation to a diverse range
of specialists. Deviations in the family history from the anticipated autosomal-recessive mode of inheritance,
with apparent “pseudodominance” and mechanisms of inheritance that include uniparental isodisomy (the
inheritance of both chromosomal copies from a single parent), may all further cloud the diagnosis. It can
therefore take the efforts of an astute clinician with a high clinical index of suspicion to clinch the diagnosis
of this eminently treatable condition.

INTRODUCTION
of population isolates in which a higher prevalence
Sternlieb and Scheinberg (1968) first estimated the prev- was likely.
alence of Wilson disease in 1968 to be 5 per million indi- Park and coworkers (1991), concerned by the potential
viduals. Subsequent work by Bachmann et al. (1979a, b) for a significant number of undiagnosed individuals with
at the Leipzig Centre for Wilson’s Disease in East Wilson disease, sought to determine the prevalence of
Germany between 1949 and 1977 calculated a birth prev- Wilson disease in Scotland. Examining computerized hos-
alence of Wilson disease of 29 per million. Saito (1981) pital statistical records, death certificates, and the results of
reported a similar birth prevalence of 33 per million in a postal questionnaire sent to relevant clinicians, they iden-
1981. With this expanded data set and taking into tified 21 patients with Wilson disease alive at the point of
account mortality figures from the USA pertaining to analysis in a population of 5 090 700 individuals, equating
the number of deaths from Wilson disease, which they to a prevalence of 4 per million. The authors re-examined
assumed to be half the true number, Scheinberg and the data presented by Bachmann et al. (1979a, b) from East
Sternlieb (1984) revised their worldwide prevalence esti- Germany and calculated a revised prevalence rate of 4.6
mate to 30 per million with a heterozygote carrier fre- per million, which was comparable with their own figure
quency of 1 in 90, whilst acknowledging the existence (Park et al., 1991). This rate was still higher than that of

*Correspondence to: Oliver Bandmann, SITraN, 385a Glossop Road, Sheffield S10 2HQ, UK. Tel: +44-114-2222262,
E-mail: o.bandmann@sheffield.ac.uk
8 C. LO AND O. BANDMANN
2.7 per million reported by Przuntek and Hoffmann (1987) were similarly disappointing. They achieved greater suc-
from West Germany, notwithstanding their less than ideal cess when blood samples from a slightly older population
response rate of 45%. Reilly et al. (1993) adopted a meth- of 24 165 children aged 6 months to 9 years old were used
odologic approach similar to that taken by Park et al. in to measure ceruloplasmin with a combination of oxidase
order to determine the prevalence of Wilson disease in activity, particle-coated fluorescence immunoassay, and
the Republic of Ireland. Twenty-six cases of Wilson specific monoclonal antibody. Three patients with Wil-
disease over a 19-year period were identified, 5 of son disease were identified, the youngest patient being
which had died of an illness consistent with the disease 8 months old, equating to a prevalence of 124 per million.
before its formal diagnosis. An adjusted birth incidence The authors suggested that, considered in combination
rate of 17 per million was calculated for the period with existing screening programs, serum ceruloplasmin
of 1950–1969, corresponding to a gene frequency of level stability and ease of blood collection meant that
0.41% and heterozygote incidence of 0.82%. To allow the age of 3 years old was the most opportune time to
for the maximal degree of anticipated consanguinity, undertake any population-wide ceruloplasmin-based
the gene frequency and heterozygote incidence esti- mass screening in children (Yamaguchi et al., 1999).
mates were revised to 0.36% and 0.72% respectively, In apparent support of their supposition, a presympto-
providing minimum disease estimates. matic 32-month-old boy was the sole positive case iden-
In this chapter we will provide an overview of clinical tified through a pilot study in which ceruloplasmin levels
genetic and biochemical studies to date and examine in dried blood spots were assessed in 3667 asymptomatic
their roles in further delineating the epidemiology of Korean children aged between 3 months and 15 years old
Wilson disease. We will explore factors which may help (Hahn et al., 2002). A similar study, with a 2.6-fold
to explain the disparity between the number of individ- higher prevalence of 717 per million, screened the dried
uals predicted to be affected on the basis of screening blood spots of 2789 children, identifying 2 aged
studies and those diagnosed clinically. Finally, we will 30 and 39 months old, with markedly low ceruloplasmin
review the clinical course of Wilson disease and the levels (0.24 and 0.40 mg/dL compared to an average
diverse ways in which it may present. of 12.4 mg/dL). The diagnoses were later confirmed
genetically by the identification of compound heterozy-
gous A803T/2871delC and R778L/G1035V mutations
BIOCHEMICAL SCREENING FOR THE
(Ohura et al., 1999).
STUDY OF PREVALENCE
Noninvasive techniques to assess the amount of uri-
Many of the early epidemiologic studies are thought to nary holoceruloplasmin protein have also been developed.
have underestimated the prevalence of Wilson disease, Screening of urine from 48 819 children led to the identi-
not least due to their reliance on symptomatic patients fication of 36 children with very low urinary and serum
having received the correct diagnosis. Considerable holoceruloplasmin levels, 2 of whom additionally demon-
interest lies in the early diagnosis of presymptomatic strated low serum copper and increased urinary copper
individuals with Wilson disease so that the early instiga- excretion. Both children were otherwise asymptomatic
tion of treatment may prevent future complications. The with no signs on physical examination. Yet on further
UK NHS newborn blood spot screening program is genetic testing they were found to be compound heterozy-
already utilized to screen for six inherited metabolic dis- gotes for mutations recognized in Japanese patients with
eases. A number of pilot studies have investigated the Wilson disease (Owada et al., 2002).
possibility of mass screening for Wilson disease. Serum The calculated prevalence of 41 per million was less
copper is typically reduced in Wilson disease. However, than half that derived from another Japanese study, where
its utility as a target agent for dried blood spot screening a further 11 362 children underwent measurement of uri-
is limited by its environmental abundance and presence nary ceruloplasmin concentration. An immunologic latex
in unpredictable amounts in dried filter paper itself agglutination assay kit was applied to an automated ana-
(Hahn, 2014). Instead an enzyme-linked immunosorbent lyzer and results were shown to correlate closely with uri-
assay using a monoclonal antibody specify to holoceru- nary holoceruloplasmin level measurements. Three
loplasmin (ceruloplasmin bound with copper) has been successive screening stages led to the identification of
developed to measure ceruloplasmin in dried blood spots 9 children as positive for low urinary ceruloplasmin levels
(Ohura et al., 1999; Yamaguchi et al., 1999; Hahn et al., from an initial 668 positive subjects. A diagnosis of
2002; Kroll et al., 2006). Wilson disease was excluded in 8 children in the context
Screening of 1045 anonymous newborn screening of normal biochemical data. Ultimately, in the absence of
dried blood spots failed to identify any cases of Wilson symptoms or clinical signs, genetic confirmation was
disease (Kroll et al., 2006). The results of screening a necessary to diagnose 1 child, aged 3 years old, as a com-
total of 126 810 newborns by Yamaguchi et al. (1999) pound heterozygote (Nakayama et al., 2008).
EPIDEMIOLOGY AND INTRODUCTION TO THE CLINICAL PRESENTATION OF WILSON DISEASE 9
the mutation compared to 20% being heterozygous.
MODERN GENETICS STUDIES
Homozygotes were reported to more often present with
As techniques in molecular genetics have developed, neurologic symptoms, though this did not reach statisti-
attempts have been made to better characterize the under- cal significance. However, compared to patients without
lying ATP7B mutations in patients with Wilson disease the H1069Q mutation, homozygotes and heterozygotes
from different populations. In 1999 Curtis et al. studied were 3.5 and 2.13 times more likely to present neurolog-
Wilson disease mutations in Britain in 52 patients by ically (Stapelbroek et al., 2004). Mirroring studies by
screening parts of the ATP7B gene using single-strand Shah et al. (1997) and Gromadzka et al. (2006), patients
conformational polymorphism (SSCP) and subsequent heterozygote for the H1069Q mutation were found to
sequencing. At the time SSCP screening of three exons present at a slightly earlier age than homozygotes but pre-
was predicted to identify around 60% of mutations. sented at a later age than patients harboring non-H1069Q
Indeed, complete or partial genetic characterization mutations (Stapelbroek et al., 2004).
was achieved in 37 individuals, with 4 patients having A greater disparity in the existence of heterozygous
a homozygous ATP7B mutation, 18 being compound and homozygous H1069Q mutations was found in
heterozygotes, and 15 in whom a mutation in only one Romanian patients, where an allelic frequency of
allele was detected (Curtis et al., 1999). An earlier study 38.1% was reported, accounting for 34.2% of mutations
had similarly managed to characterize around 60% of in the heterozygous and 21.1% in the homozygous state
disease alleles (Nanji et al., 1997). A significant (Iacob et al., 2012). In a German cohort described
improvement in the overall mutation detection frequency by Merle et al. in 2010, patients with homozygous
to 98% was reported in a subsequent study from the UK H1069Q mutations occurred at an even greater fre-
by Coffey et al. in 2013. Genetic confirmation was quency, being identified in 50.9% of patients with a fur-
achieved in 177 out of 181 patients with Wilson disease ther 25.4% of patients being compound heterozygotes
diagnosed on the basis of clinical and biochemical find- with one H1069Q mutant allele.
ings. Of the remaining 4 patients, 2 were found to have a Though the H1069Q mutation is considered to be
single ATP7B mutation whilst, in the others, no muta- the most common mutation in Eastern and Northern
tions were identified (Coffey et al., 2013). The improved European populations, this is not necessarily the case
detection rate was largely due to improved mutation tech- worldwide. Indeed, in a Japanese study by Nanji et al.
niques as well as the analysis of the entire ATP7B coding in 1997, out of 21 unrelated families the H1069Q muta-
region. tion was not detected in a single case. Instead, the R778L
Though population estimates vary, in Europe the most mutation occurred at the greatest frequency, accounting
common mutation observed is the H1069Q missense for 12% of the identified Wilson disease mutations.
mutation (Shah et al., 1997; Stapelbroek et al., 2004; The R778L mutation has also been reported in Taiwanese
Gomes and Dedoussis, 2015). Ivanova-Somlenskya families with Wilson disease (Chuang et al., 1996), as
et al. (1999) described a cohort of 40 unrelated patients well as in Korean patients (Kim et al., 1998) and in the
with Wilson disease from Slavonic families from the Chinese Han population, in whom the allele frequency
European part of Russia to have H1069Q mutations, is considerably higher, at 45.6% (Liu et al., 2004).
representing 48.7% of Wilson disease chromosomes. A Spanish study identified the M645R missense muta-
Tarnacka et al. in 2000 described 148 Polish patients with tion as the most frequent mutation; its detection in the
Wilson disease from 95 families. The H1069Q mutation heterozygous state was reported in 22 out of 40 unrelated
was found in 57% of chromosomes studied appearing in patients with Wilson disease (Margarit et al., 2005). In
the homozygous and heterozygous states in 39.9% and Costa Rica another missense mutation, the N1270S
30.4% of cases respectively. Though they attempted mutation, also observed in Sicilian and continental Ital-
genotype–phenotype correlation, no relationship was ian and Turkish populations, was found to represent
found either with symptomatology or age of onset. In 61% of all mutations (Tanzi et al., 1993; Figus et al.,
contrast, a study by Shah et al. (1997) reported a younger 1995; Shah et al., 1997).
age of onset in individuals heterozygous for the H1069Q Whilst interpatient variability may in part be
mutation at 15.4 years of age compared to 20 in homozy- explained by differences in the underlying genotype,
gotes. Through their use of the Hardy–Weinberg equilib- studies have examined rates of intrafamilial concordance
rium, they estimated that the ratio of H1069Q between individuals sharing similar genetic characteris-
heterozygotes to homozygotes was in the order of 3.26. tics. In a study involving 73 index cases from 73 unre-
In a study of 70 symptomatic Dutch patients from lated families and 95 of their siblings, with an overall
59 unrelated families with predominant hepatic presenta- H1069Q allelic frequency of 77%, an 86% rate of con-
tion, the H1069Q mutation was reported to account for cordance of presenting symptoms was noted among indi-
33% of alleles, with 23% of patients homozygous for viduals first presenting with hepatic symptoms. A lower
10 C. LO AND O. BANDMANN
concordance rate of 66% was noted between index cases 200 unrelated habitants revealed a carrier frequency of
presenting primarily with neurologic symptoms and their p.Q289X and 398delT mutations of 1 in 11 (Dedoussis
symptomatic siblings (Chabik et al., 2014). et al., 2005).
In a survey of Wilson disease in Iceland over a 40-year The relative genetic homogeneity within the Sardin-
period, 8 patients, from two kindreds, were diagnosed ian population is thought to arise from its marked isola-
with Wilson disease. All patients were found to be homo- tion and history of inbreeding propagating a possible
zygous for the same 2010del7 mutation, presumably founder effect (Loudianos et al., 1999b). Figus et al.
inherited from a shared common progenitor. Despite (1995) postulated a higher prevalence of Wilson disease
their common genotype and primarily neurologic symp- within the Sardinian population in 1995 based on their
tomatology, differences in their presentation were noted, identification of 10–12 new cases per year. Sixteen
with 3 out of the 8 manifesting with psychiatric symp- different haplotypes associated with Wilson disease
toms (Thomas et al., 1995). Differing phenotypes were chromosomes were identified in 39 individuals of Sar-
also reported by Wang et al. (2011) in two siblings, both dinian descent, of which haplotype IX (5 10 3 3) was
compound heterozygous for the I1148T missense muta- the most common, observed at a frequency of 55% of
tion and the S105Stop mutation. Whilst neuropsychiatric Wilson disease chromosomes. Further analysis of the
symptoms were manifested by the older proband, her promoter and the 5’ UTR of the Wilson disease gene
younger brother’s presentation was of the hepatic type. sequence from patients with the most common haplotype
Członkowska et al. (2009) furthermore described phe- revealed a single mutation consisting of a 15-nt deletion
notypic discordance in two pairs of monozygotic twins. from position –441 to position –427 relative to the trans-
In the first pair of twins, both sisters were compound het- lation start site, which accounted for 60.5% of the studied
erozygous for the H1069Q and N404Kfs mutations and Wilson disease chromosomes (Loudianos et al., 1999b).
were found to have evidence of previous hepatitis These mutations are uncommon in patients of European
B infection, with active infection having been excluded. ancestry outside of Sardinia (Cullen et al., 2003). How-
The proposita had presented relatively late at the age of ever, screening of 5290 newborns in Sardinia revealed
38, with a 4-year history of fatigue followed by progres- the presence of 122 individuals heterozygous for the –
sive hepatic decompensation and then the onset on neu- 441/–427 deletion, equating to an allelic frequency of
ropsychiatric symptoms. In contrast, her twin was 1.15%. With the inclusion of an allelic frequency of
asymptomatic, with normal brain and liver imaging, 0.77%, accounting for non- –441/–427 deletions, an
the only biochemical abnormality detected being overall Wilson disease mutation frequency of 1.92%
decreased serum levels of copper and ceruloplasmin. was inferred. Assuming Hardy–Weinberg equilibrium,
The second pair of twins described was homozygous the prevalence of Wilson disease was calculated at 370
for the common H1069Q mutation. The results of their per million births, one of the highest in the world and
diagnostic evaluation at the age of 28 were similar. Imag- considerably higher than that first quoted by
ing revealed T2-weighted changes on brain magnetic res- Bachmann et al. (1979b) (Zappu et al., 2008). Yet it is
onance imaging (MRI) and hepatosplenomegaly on comparable with the estimated prevalence of 366 per mil-
ultrasound scanning. Laboratory investigations identi- lion calculated from a homozygosity index of 0.476 and
fied evidence of abnormal copper metabolism, liver inbreeding coefficient of 7.8  10–4 and applied to a set
function test derangement, leukopenia, and thrombocy- of 178 Sardinian individuals with clinically and molecu-
topenia. Whilst one twin had presented with neurologic larly diagnosed Wilson disease (Gialluisi et al., 2013). As
symptoms, the other described problems with menstrual six mutations account for around 85% of Wilson disease
irregularity and recurrent nose bleeding but barring the chromosomes within the Sardinian population, further
finding of Kayser–Fleischer rings on examination was efforts have included the targeted screening for these
otherwise devoid of neurologic signs (Członkowska mutations using automated TaqMan screening (Zappu
et al., 2009). et al., 2010). The identification of heterozygotes through
this initial screening process is followed by screening for
the remaining 16 mutations detected in the Sardinian
POPULATION ISOLATES
population, yielding a sensitivity of 94.6% with a spec-
Various population isolates exist in which Wilson disease ificity of 100% and potentially resulting in the diagnosis
occurs at a greater incidence and prevalence than would of an additional 5 cases per year (Lovicu et al., 2003;
otherwise be expected based on worldwide figures. In a Zappu et al., 2008, 2010).
small mountain village next to Heraklion city in the A similarly high prevalence of Wilson disease of 385
island of Crete, the reported incidence of clinically per million was reported for the island of Gran Canaria,
and/or biochemically diagnosed Wilson disease was 6 where the rare Leu708Pro mutation was observed in
out of 90 births over a 25-year period. Screening of 18 out of 24 individuals with Wilson disease; 12 in the
EPIDEMIOLOGY AND INTRODUCTION TO THE CLINICAL PRESENTATION OF WILSON DISEASE 11
homozygous state, 4 as compound heterozygotes in There is a significant discrepancy between the number
conjunction with another established Wilson disease of individuals predicted on the basis of genetic studies to
mutation, and 2 further individuals who only had one be affected with Wilson disease and those clinically diag-
identifiable ATP7B mutation. A further 3 individuals nosed. Reduced penetrance of ATP7B mutations may
who were compound heterozygous for previously offer an alternative explanation for the lower than
established Wilson disease mutations were detected expected number of individuals diagnosed clinically
and no mutations were identified in 3 affected with Wilson disease. Indeed, Członkowska et al.
individuals from the same pedigree. Despite their com- (2008) described the indolent course of Wilson disease
mon genotype, affected individuals with homozygous first diagnosed in a woman at the age of 54 when she
Leu708Pro mutations had marked phenotypic variability was assessed during familial screening. At the time of
(Garcia-Villarreal et al., 2000). her initial assessment she was asymptomatic, the only
The same group who initiated newborn screening of sign being Kayser–Fleischer rings, and she elected to
the Sardinian population also screened 396 newborns refuse treatment. Genetic confirmation was later
on the similarly isolated Greek island of Kalymnos. achieved when she was found to be homozygous for
The screening of 60% of newborns between 1999 and the H1069Q mutation. It was not until the age of 74 that
2003 identified 18 newborns heterozygous for the she started to demonstrate evidence of liver dysfunction
H1069Q mutation, 9 heterozygous for the R969Q muta- with a slight elevation in her liver enzymes and reduction
tion, and 1 compound heterozygote. The allelic frequen- in serum albumin. Thirty years after her original diagno-
cies of the H1069Q and R969Q mutations were 2.4% and sis, at the age of 84, she remained symptom free
1.3% respectively, yielding a carrier rate for the two (Członkowska et al., 2008).
mutations of 7% and, assuming Hardy–Weinberg equi- Identification of such individuals may still be war-
librium, an even higher prevalence than in Sardinia of ranted with respect to the risk of transmission to potential
135 per million births (Zappu et al., 2008). children that they may have. Of greatest concern is the
The highest prevalence of genetically confirmed potential for there to be a large number of patients
Wilson disease ever reported is 885 per million from affected by Wilson disease who remain undiagnosed
within the mountainous region of Rucar in Romania. and without effective treatment, having either never pre-
Its isolation and tradition of intraclan marriage are sented to their medical practitioner or having presented
thought to have contributed to substantial levels of con- with seemingly atypical features (Coffey et al., 2013;
sanguinity. Screening for ATP7B mutations in 50 individ- Bandmann et al., 2015). The absence of a “typical” fam-
uals from two large families, spanning six generations, ily history suggestive of autosomal-recessive inheritance
identified compound heterozygous H1069Q/M769H– may also dissuade a clinician from considering the diag-
mutations in 5 symptomatic adults and 2 clinically nosis of Wilson disease. It is important to be aware of less
asymptomatic children. Symptomatic individuals dem- common mechanisms of inheritance, including the inher-
onstrated strong phenotypic concordance with an age itance of three different ATP7B mutations, the potential
of symptom onset of 18  1 years. Initial symptoms for Wilson disease in successive generations, and the role
included dysarthria and dysphagia and, in all symptom- of segmental uniparental isodisomy, as will now be
atic individuals, the presence of Kayser–Fleischer rings outlined.
was observed (Cocos et al., 2014). The majority of patients with Wilson disease are com-
pound heterozygotes with one mutation on each ATP7B
allele (Schilsky, 2014a). There are nevertheless a number
GENETIC VERSUS CLINICAL
of patients who carry three different mutations.
PREVALENCE
Dedoussis et al. (2005) described a 15-year-old boy with
Sequencing of all 21 ATP7B exons in over 1000 DNA Wilson disease from a consanguineous pedigree who
samples and exons 8, 14, and 18 in over 5000 samples was compound heterozygous for the Q289X, I1148T,
by Coffey et al. led to a revised figure of 0.040 or 1:25 and G1176R mutations; the latter two mutations cosegre-
as the frequency of heterozygote mutation carriers in gating in cis (i.e., in the same copy of the gene). Although
the UK. Even with the exclusion of four class 2 his clinically asymptomatic younger brother carried a
single-nucleotide variants in the absence of in silico wild-type allele alongside the I1148T/G1176R muta-
(computer-simulated) evidence of pathogenicity, the fre- tions, biochemical indices demonstrated significantly
quency of individuals predicted to carry two pathogenic lower copper and ceruloplasmin levels.
ATP7B mutations was 142 per million, over four times In an ethnic Han Chinese population, with one out of
the often-quoted prevalence figure of 30 per million 65 families reporting consanguinity, Mak et al. (2008)
for Wilson disease (Scheinberg and Sternlieb, 1984; identified 4 patients who carried the Q1142H nonsense
Coffey et al., 2013). and I1148T missense mutations in cis, with another
12 C. LO AND O. BANDMANN
mutation on the trans allele (i.e., in the other copy of the the strong suspicion of consanguinity, later confirmed
gene). All mutations had previously been identified inde- when the mother and father were found to be first
pendently as disease-causing mutations (Loudianos et al., cousins.
1998, 1999a). Due to the frequency of their cosegregation Based on the much-quoted heterozygote carrier fre-
in cis, the authors recommended further parental genotyp- quency of 1 in 90 (Scheinberg and Sternlieb, 1984),
ing or exon sequencing upon the simultaneous discovery Bennett et al. (2013) calculated the probability of an indi-
of Q1142H and I1148T mutations, in order to distinguish vidual with Wilson disease having an affected grandchild
individuals bearing two mutations from those bearing to be 0.003%. Yet such a family did they describe, with
three (Mak et al., 2008). the proband’s mother, maternal aunt, and son having Wil-
Wang et al. (2011) subsequently described 2 patients son disease; this was due to all their reproductive partners
who presented with neuropsychiatric symptoms and having been heterozygote carriers. Dziezyc et al. (2014)
were found to carry three mutations. Fascinatingly, hav- identified nine nonconsanguineous families comprising
ing previously been reported to cosegregate with another 12 affected offspring from 9 probands from their screen
mutation in cis, the I1148T and Q1142H mutations were of 294 individuals. A risk of 4.08% of Wilson disease in
identified on the same allele, a finding also reported in the offspring of probands was calculated, higher than the
another patient by Coffey et al. (2013). The I381S/ figure of 0.5% that had previously been estimated (Ala
I1184T mutation and the N41S/I1021V mutations have et al., 2007; Dziezyc et al., 2014).
also been reported to cosegregate in cis (Coffey In the study by Coffey et al. (2013), three families
et al., 2013). with “pseudodominant” Wilson disease were identified.
As an autosomal-recessively inherited condition, In one family, the proband and his wife remained con-
Wilson disease would normally not be expected in sub- cerned about the genetic risk to their asymptomatic chil-
sequent generations within a family. The phenomenon of dren in spite of a very low risk having been quoted to
“pseudodominance” is one that has been observed in them. Rather surprisingly, a novel variant, predicted by
other conditions, including Friedreich’s ataxia (Lamont in silico analysis to be pathogenic, was identified in
et al., 1997) and ataxia with ocular apraxia type 2 the proband’s wife, leading to the subsequent diagnosis
(Schols et al., 2008). Cases of Wilson disease in two suc- of one of their two children as a compound heterozygote
cessive generations have also been reported. Loudianos for the V1234F and R778W mutations (Coffey et al.,
et al. (2013) described two families from Sardinia. In the 2013). Taken together, the comparatively frequent obser-
first family, Wilson disease was diagnosed after the pro- vation of families with Wilson disease in subsequent gen-
band presented following a suicide attempt. She was erations in different populations lends support to the
found to be compound heterozygous for the R919W assumption that heterozygote carrier status for ATP7B
and H1069Q mutations. Screening of her triplet siblings mutations may be more common than previously
led to the diagnosis of her dizygotic sibling with Wilson thought.
disease, carrying both the R919W and T993W muta- Segmental uniparental isodisomy has also been pro-
tions. Their mother carried the R919W mutation in addi- posed as a mechanism by which the direct transmission
tion to a wild-type allele. A posthumous diagnosis was of a normally monogenically inherited disorder may
made in their father, who was postulated to have trans- occur between a single parent and his or her offspring
mitted the H1069Q and T993W mutations, having died (Loudianos et al., 1999a). Uniparental isodisomy for
at the age of 39 with liver failure. The affected mother of any chromosome is estimated to affect up to 1 in 3500
another family was homozygous for the –441/–427 dele- births and occurs when both copies of a chromosome
tion, the most common regional mutation. Both of her or part thereof are inherited from a single parent with
monozygotic twins had inherited an allele with no contribution from the other. From their cohort of
the –441/–427 deletion as well as the G869R mutation, 181 index cases with Wilson disease, Coffey et al.
which they inherited from their father. Despite being (2013) reported the finding of 2 patients homozygous
compound heterozygotes, the twins were clinically for different mutations, in the absence of biparental
asymptomatic when they were diagnosed at the age of homologue contribution. Multiple microsatellite markers
33 (Loudianos et al., 2013). subsequently confirmed segmental isodisomy giving rise
A similar pattern of inheritance, whereby an affected to autozygosity for mutations inherited from one parent,
parent unknowingly had children with a heterozygous with potential significance with respect to genetic
carrier resulting in a child with Wilson disease, was counseling of individuals (Coffey et al., 2013).
reported in three out of four French families described Heterozygote carriers are often clinically asymptom-
by Dufernez et al. (2013). In the fourth family, the iden- atic. However, detailed examination may reveal subtle
tification of both the mother and her affected child as abnormalities. Ultrastructural examination of liver tissue
being homozygous for the same T569del mutation raised using electron microscopy has revealed alterations in the
EPIDEMIOLOGY AND INTRODUCTION TO THE CLINICAL PRESENTATION OF WILSON DISEASE 13
mitochondria and endoplasmic reticulum of heterozy- H1069Q mutation was also identified. The authors spec-
gous individuals, in keeping with copper toxicity ulated on the unusual occurrence of two rare conditions
(Lough and Wiglesworth, 1976). Biochemical studies within a single family and postulated a potential interplay
have reported various abnormalities of copper metabo- between the mutations that might account for an unusu-
lism, including decreased levels of serum ceruloplasmin ally late presentation of Alagille syndrome in the absence
and copper and an increase in the biologic halftime for of bile duct paucity typically observed on liver biopsy
the clearance of the isotope 65Cu from the plasma pool (Amson et al., 2012).
(Marecek and Nevsimalova, 1984; Lyon et al., 1995;
Merli et al., 1998; Tarnacka et al., 2009).
CLINICAL COURSE
Relative exchangeable copper determination may be
better at distinguishing patients with Wilson disease from Three main patterns of presentation have been recog-
heterozygotes and controls (Trocello et al., 2014). In a nized: hepatic, neurologic, and psychiatric, the symp-
study involving 12 heterozygous carriers, without path- toms of the latter two groups sometimes being
ologic changes on plain MRI, significantly higher ratios combined. The presentations observed by clinicians
of Glx/Cr and Lip/Cr in 1H magnetic resonance spectros- may be skewed by the specialties to which patients pre-
copy in the pallidum and thalami were reported com- sent. Unsurprisingly, neurologists report a greater pro-
pared to controls, suggesting possible asymptomatic portion of patients presenting with neurologic
copper deposition (Tarnacka et al., 2009). Electroen- symptoms (69.1%) compared to hepatic symptoms
cephalographic abnormalities have also been described (14.9%), whereas the opposite is true with patients pre-
in heterozygous children. However, such studies, per- senting to gastroenterologists, where hepatic features
formed prior to the discovery of the ATP7B gene, relied predominate (68.1%) (Taly et al., 2007; Weiss et al.,
upon the inferred heterozygous carrier state in immediate 2011). In a registry of 627 patients with Wilson disease
relatives of affected individuals. Out of 16 presumed seen at a neurology department in Poland, 510 of whom
heterozygotes, neurologic abnormalities were detected were symptomatic at the point of diagnosis, a male pre-
in 5 children. Abnormalities on electroencephalography ponderance was observed (55%). Neurologic and hepatic
were detected in 12 children (75%), 5 of which demon- symptoms and signs were seen more frequently in men
strated epileptic features which did not correlate with (60%) and women (57%) respectively. Overall, symp-
clinical symptomatology (Marecek and Nevsimalova, toms and signs were found to precede a diagnosis of
1984). The findings were replicated in a subsequent Wilson disease by around 2 years. The mean age of
study but interestingly appeared to resolve by adulthood, symptom onset in the hepatic group was about 4 years
at which time no significant pathologic electroencepha- earlier than in the group with neuropsychiatric symptoms
lographic abnormalities were detected above that of the (23.7  8 years versus 28.0  8 years) (Litwin et al.,
control group (Nevsimalova et al., 1986). 2012). Similar findings were reported in an independent
The potential for heterozygous mutations in the German cohort of 163 patients with Wilson disease, 137
Wilson disease gene to influence other, apparently unre- of whom were symptomatic. Though the mean age of
lated disease processes has been suggested. Cocco et al. symptom onset was younger than that described in the
(2009) described a 37-year-old man with liver cirrhosis Polish cohort, hepatic symptoms were again found to
secondary to chronic hepatitis C presenting with enceph- precede neurologic symptoms (15.5 compared to
alopathy, in whom treatment with penicillamine and tri- 20.2 years of age). The interval between symptom onset
hexyphenidyl resulted in a marked improvement of his and diagnosis was reported to be almost three times lon-
neuropsychiatric symptoms. The patient was later found ger in patients presenting with neurologic symptoms
to be a heterozygous carrier of the G1111A missense compared to hepatic symptoms (44.4 months vs.
mutation, his response to treatment suggesting a subtle 14.4 months) (Merle et al., 2007).
defect in brain copper metabolism (Cocco et al., 2009). Individuals may present with a spectrum of hepatic
In another case, screening of an otherwise asymptom- symptoms, with 25.4% of patients described by
atic 18-year-old man, the sibling of a patient recently Gheorghe et al. (2004) having clinically asymptomatic
diagnosed with Wilson disease, revealed deranged liver disease, 21.8% fulminant hepatic failure, and 52.8%
function tests. Indices of copper metabolism were normal chronic liver disease at presentation . Untreated, hepatic
and further investigation led to a diagnosis of Alagille symptoms may remain self-limiting, yet may also pro-
syndrome, a rare autosomal dominantly inherited disor- gress and precede the development of neurologic
der of embryogenesis, affecting multiple organ systems, sequelae. Reassuringly, in treated cases, the prognosis
in which abnormalities in bile duct formation led to liver of hepatic Wilson disease is good (Schilsky, 2014b).
dysfunction. The associated JAG1 gene mutation was Of patients presenting with neurologic symptoms,
found to have arisen de novo but, interestingly, one improvement in 53.5–58.2%, stability in 22.4–27.3%,
14 C. LO AND O. BANDMANN
and worsening in 3.6–24.1% may be expected after Although classically considered to be associated with
pharmacologic treatment (Merle et al., 2007; Bruha an onset in the second or third decade of life, an increas-
et al., 2011). Psychiatric symptoms have been reported ing number of cases of late-onset Wilson disease are
in up to 51% of patients upon their initial presentation being reported (Członkowska and Rodo, 1981; Ala
and 72% of patients established on treatment, although et al., 2005; Ferenci et al., 2007; Sohtaoglu et al.,
a pure psychiatric presentation is rare (Dening and 2007; Członkowska et al., 2008). Two siblings, diag-
Berrios, 1989; Svetel et al., 2009). The most commonly nosed in their eighth decade of life, were described by
identified symptoms on the neuropsychiatric inventory Ala et al. (2005). The elder sibling had presented at the
include anxiety, depression, irritability, and apathy age of 72 with a 5-year history of progressive neurologic
(Svetel et al., 2009). Cognitive changes, when present, disability. Her younger brother had had a mild hand
may be subtle and prone to influence by changes in tremor at the age of 45 that had been treated with a
affect (Lang et al., 1990). Normalization may occur beta-blocker. He was diagnosed at the age of 70 during
in response to treatment (Rosselli et al., 1987). Whilst screening for Wilson disease; his only symptom was mild
symptomatic individuals typically present with hepatic, gait abnormalities. Both siblings were found to be com-
neurologic, or psychiatric manifestations of Wilson dis- pound heterozygous for the E1064A and H1069Q muta-
ease, less common presentations have been recognized, tions with stabilization of their clinical course following
including presentation with an osseomuscular pheno- treatment (Ala et al., 2005).
type as well as renal, cardiovascular, endocrine, hema- Another similar case, described by Sohtaoglu et al.
tologic, and dermatologic involvement. The varied (2007), was of a woman who developed slurred speech,
presentations of Wilson disease and the role of genetic tremor, and postural instability at the age of 75, though
and environmental modifiers will be detailed in subse- her first symptoms of mild tremor and forgetfulness
quent chapters. had begun 8 years earlier. The salient message from
With the ability to screen for mutations in the ATP7B her case was of a marked deterioration following the ini-
gene, a diagnosis of Wilson disease is possible even in tiation of penicillamine, prompting the authors to caution
the absence of symptoms or definitive biochemical with respect to its use in older patients with neurologic
abnormalities. Diagnosis is therefore possible in the pre- Wilson disease.
symptomatic stage, even in the newborn. Shimizu et al.
(1997) described an asymptomatic 8-month-old boy who
CONCLUSION
was found on opportunistic screening to have low ceru-
loplasmin levels in the context of otherwise normal bio- Considerable advances have been made since the first esti-
chemical parameters. No signs were demonstrated on mate of the prevalence of Wilson disease in 1968
clinical examination. A diagnosis of Wilson disease (Scheinberg and Sternlieb, 1984). As diagnostic tech-
was made on the basis of DNA sequencing analysis, niques have become increasingly sophisticated, so too
which revealed him to be homozygous for the frameshift has our ability to identify the causative mutations in indi-
mutation 2302insC (Shimizu et al., 1997). A child of the viduals with Wilsondisease. Nonetheless, work from our
same age was also reported to be the youngest individual own recent genetic prevalence study and others has raised
to present with evidence of liver dysfunction, detected concerns that Wilson disease may be considerably more
during a hospital admission with diarrhea. Clinical exam- common than previously thought (Ohura et al., 1999;
ination was normal, yet low ceruloplasmin levels raised Coffey et al., 2013; Gialluisi et al., 2013). Considerable
the suspicion of Wilson disease. Other causes of elevated phenotypic variation, the potential for reduced penetrance,
alanine aminotransferase and aspartate aminotransferase and the diversity in the number of mutations and mecha-
were excluded. Genetic testing identified him to be com- nisms of inheritance may all complicate the diagnosis.
pound heterozygous for the missense mutations A874V Thus, it is necessary to maintain an index of suspicion in
and N1270S (Abuduxikuer et al., 2015). A similar pre- individuals presenting with symptoms consistent with a
sentation in a 9-month-old boy was reported by Kim diagnosis of Wilson disease, even though they may not
et al. (2013). Biochemical evidence of liver dysfunction conform to the stereotypic presentation of this disorder.
was investigated further with percutaneous liver biopsy.
Elevated hepatic copper content (748 mg/g dry weight of REFERENCES
liver tissue) was accompanied by histologic findings of Abuduxikuer K, Li LT, Qiu YL et al. (2015). Wilson disease
mild lobular activity, mild portoperiportal activity, and with hepatic presentation in an eight-month-old boy. World
periportal fibrosis. Molecular analysis confirmed the J Gastroenterol 21: 8981–8984.
presence of the missense mutation G1186S and the Ala A, Borjigin J, Rochwarger A et al. (2005). Wilson disease
frameshift mutation c.4006delA on separate alleles in septuagenarian siblings: raising the bar for diagnosis.
(Kim et al., 2013). Hepatology 41: 668–670.
EPIDEMIOLOGY AND INTRODUCTION TO THE CLINICAL PRESENTATION OF WILSON DISEASE 15
Ala A, Walker AP, Ashkan K et al. (2007). Wilson’s disease. Dening TR, Berrios GE (1989). Wilson’s disease. Psychiatric
Lancet 369: 397–408. symptoms in 195 cases. Arch Gen Psychiatry 46:
Amson M, Lamoureux E, Hilzenrat N et al. (2012). Alagille 1126–1134.
syndrome and Wilson disease in siblings: a diagnostic Dufernez F, Lachaux A, Chappuis P et al. (2013). Wilson
conundrum. Can J Gastroenterol 26: 330–332. disease in offspring of affected patients: report of four
Bachmann H, Lossner J, Biesold D (1979a). Wilson’s disease French families. Clin Res Hepatol Gastroenterol 37:
in the German Democratic Republic. I. Genetics and epide- 240–245.
miology. Z Gesamte Inn Med 34: 744–748. Dziezyc K, Litwin T, Chabik G et al. (2014). Families with
Bachmann H, Lossner J, Gruss B et al. (1979b). The epidemi- Wilson’s disease in subsequent generations: clinical and
ology of Wilson’s disease in the German Democratic genetic analysis. Mov Disord 29: 1828–1832.
Republic and current problems from the viewpoint of pop- Ferenci P, Członkowska A, Merle U et al. (2007). Late-onset
ulation genetics. Psychiatr Neurol Med Psychol (Leipz) 31: Wilson’s disease. Gastroenterology 132: 1294–1298.
393–400. Figus A, Angius A, Loudianos G et al. (1995). Molecular
Bandmann O, Weiss KH, Kaler SG (2015). Wilson’s disease pathology and haplotype analysis of Wilson disease in
and other neurological copper disorders. Lancet Neurol Mediterranean populations. Am J Hum Genet 57:
14: 103–113. 1318–1324.
Bennett JT, Schwarz KB, Swanson PD et al. (2013). An excep- Garcia-Villarreal L, Daniels S, Shaw SH et al. (2000). High
tional family with three consecutive generations affected prevalence of the very rare Wilson disease gene mutation
by Wilson disease. JIMD Rep 10: 1–4. Leu708Pro in the Island of Gran Canaria (Canary
Bruha R, Marecek Z, Pospisilova L et al. (2011). Long-term Islands, Spain): a genetic and clinical study. Hepatology
follow-up of Wilson disease: natural history, treatment, 32: 1329–1336.
mutations analysis and phenotypic correlation. Liver Int Gheorghe L, Popescu I, Iacob S et al. (2004). Wilson’s disease:
31: 83–91. a challenge of diagnosis. The 5-year experience of a tertiary
Chabik G, Litwin T, Członkowska A (2014). Concordance centre. Rom J Gastroenterol 13: 179–185.
rates of Wilson’s disease phenotype among siblings. Gialluisi A, Incollu S, Pippucci T et al. (2013). The homozy-
J Inherit Metab Dis 37: 131–135. gosity index (HI) approach reveals high allele frequency
Chuang LM, Wu HP, Jang MH et al. (1996). High frequency of for Wilson disease in the Sardinian population. Eur
two mutations in codon 778 in exon 8 of the ATP7B gene in J Hum Genet 21: 1308–1311.
Taiwanese families with Wilson disease. J Med Genet 33: Gomes A, Dedoussis GV (2015). Geographic distribution
521–523. of ATP7B mutations in Wilson disease. Ann Hum Biol:
Cocco GA, Loudianos G, Pes GM et al. (2009). “Acquired” 1–8.
hepatocerebral degeneration in a patient heterozygote car- Gromadzka G, Schmidt HH, Genschel J et al. (2006).
rier for a novel mutation in ATP7B gene. Mov Disord 24: p.H1069Q mutation in ATP7B and biochemical parameters
1706–1708. of copper metabolism and clinical manifestation of
Cocos R, Sendroiu A, Schipor S et al. (2014). Genotype– Wilson’s disease. Mov Disord 21: 245–248.
phenotype correlations in a mountain population commu- Hahn SH (2014). Population screening for Wilson’s disease.
nity with high prevalence of Wilson’s disease: genetic Ann N Y Acad Sci 1315: 64–69.
and clinical homogeneity. PLoS One 9. e98520. Hahn SH, Lee SY, Jang YJ et al. (2002). Pilot study of mass
Coffey AJ, Durkie M, Hague S et al. (2013). A genetic study of screening for Wilson’s disease in Korea. Mol Genet
Wilson’s disease in the United Kingdom. Brain 136: Metab 76: 133–136.
1476–1487. Iacob R, Iacob S, Nastase A et al. (2012). The His1069Gln
Cullen LM, Prat L, Cox DW (2003). Genetic variation in mutation in the ATP7B gene in Romanian patients with
the promoter and 5’ UTR of the copper transporter, Wilson’s disease referred to a tertiary gastroenterology
ATP7B, in patients with Wilson disease. Clin Genet 64: center. J Gastrointestin Liver Dis 21: 181–185.
429–432. Ivanova-Smolenskaya IA, Ovchinnikov IV, Karabanov AV
Curtis D, Durkie M, Balac P et al. (1999). A study of et al. (1999). The His1069Gln mutation in the ATP7B gene
Wilson disease mutations in Britain. Hum Mutat 14: in Russian patients with Wilson disease. J Med Genet 36:
304–311. 174.
Członkowska A, Rodo M (1981). Late onset of Wilson’s dis- Kim EK, Yoo OJ, Song KY et al. (1998). Identification of three
ease. Report of a family. Arch Neurol 38: 729–730. novel mutations and a high frequency of the Arg778Leu
Członkowska A, Rodo M, Gromadzka G (2008). Late onset mutation in Korean patients with Wilson disease. Hum
Wilson’s disease: therapeutic implications. Mov Disord Mutat 11: 275–278.
23: 896–898. Kim JW, Kim JH, Seo JK et al. (2013). Genetically confirmed
Członkowska A, Gromadzka G, Chabik G (2009). Wilson disease in a 9-month old boy with elevations of ami-
Monozygotic female twins discordant for phenotype of notransferases. World J Hepatol 5: 156–159.
Wilson’s disease. Mov Disord 24: 1066–1069. Kroll CA, Ferber MJ, Dawson BD et al. (2006). Retrospective
Dedoussis GV, Genschel J, Sialvera TE et al. (2005). Wilson determination of ceruloplasmin in newborn screening
disease: high prevalence in a mountainous area of Crete. blood spots of patients with Wilson disease. Mol Genet
Ann Hum Genet 69: 268–274. Metab 89: 134–138.
16 C. LO AND O. BANDMANN
Lamont PJ, Davis MB, Wood NW (1997). Identification and Nakayama K, Kubota M, Katoh Y et al. (2008). Early and pre-
sizing of the GAA trinucleotide repeat expansion of symptomatic detection of Wilson’s disease at the manda-
Friedreich’s ataxia in 56 patients. Clinical and genetic cor- tory 3-year-old medical health care examination in
relates. Brain 120 (Pt 4): 673–680. Hokkaido Prefecture with the use of a novel automated uri-
Lang C, Muller D, Claus D et al. (1990). Neuropsychological nary ceruloplasmin assay. Mol Genet Metab 94: 363–367.
findings in treated Wilson’s disease. Acta Neurol Scand Nanji MS, Nguyen VT, Kawasoe JH et al. (1997). Haplotype
81: 75–81. and mutation analysis in Japanese patients with Wilson
Litwin T, Gromadzka G, Członkowska A (2012). Gender dif- disease. Am J Hum Genet 60: 1423–1429.
ferences in Wilson’s disease. J Neurol Sci 312: 31–35. Nevsimalova S, Marecek Z, Roth B (1986). An EEG study of
Liu XQ, Zhang YF, Liu TT et al. (2004). Correlation of ATP7B Wilson’s disease. Findings in patients and heterozygous rel-
genotype with phenotype in Chinese patients with Wilson atives. Electroencephalogr Clin Neurophysiol 64: 191–198.
disease. World J Gastroenterol 10: 590–593. Ohura T, Abukawa D, Shiraishi H et al. (1999). Pilot study of
Loudianos G, Dessi V, Lovicu M et al. (1998). Haplotype and screening for Wilson disease using dried blood spots
mutation analysis in Greek patients with Wilson disease. obtained from children seen at outpatient clinics.
Eur J Hum Genet 6: 487–491. J Inherit Metab Dis 22: 74–80.
Loudianos G, Dessi V, Lovicu M et al. (1999a). Mutation anal- Owada M, Suzuki K, Fukushi M et al. (2002). Mass screening
ysis in patients of Mediterranean descent with Wilson dis- for Wilson’s disease by measuring urinary holoceruloplas-
ease: identification of 19 novel mutations. J Med Genet 36: min. J Pediatr 140: 614–616.
833–836. Park RH, McCabe P, Fell GS et al. (1991). Wilson’s disease in
Loudianos G, Dessi V, Lovicu M et al. (1999b). Molecular Scotland. Gut 32: 1541–1545.
characterization of Wilson disease in the Sardinian Przuntek H, Hoffmann E (1987). Epidemiologic study of
population – evidence of a founder effect. Hum Mutat Wilson’s disease in West Germany. Nervenarzt 58: 150–157.
14: 294–303. Reilly M, Daly L, Hutchinson M (1993). An epidemiological
Loudianos G, Zappu A, Lepori MB et al. (2013). Wilson’s dis- study of Wilson’s disease in the Republic of Ireland.
ease in two consecutive generations: the detection of three J Neurol Neurosurg Psychiatry 56: 298–300.
mutated alleles in the ATP7B gene in two Sardinian fam- Rosselli M, Lorenzana P, Rosselli A et al. (1987). Wilson’s
ilies. Dig Liver Dis 45: 342–345. disease, a reversible dementia: case report. J Clin Exp
Lough J, Wiglesworth FW (1976). Wilson disease. Neuropsychol 9: 399–406.
Comparative ultrastructure in a sibship of nine. Arch Saito T (1981). An assessment of efficiency in potential
Pathol Lab Med 100: 659–663. screening for Wilson’s disease. J Epidemiol Community
Lovicu M, Dessi V, Zappu A et al. (2003). Efficient strategy Health 35: 274–280.
for molecular diagnosis of Wilson disease in the Scheinberg IH, Sternlieb I (1984). Wilson’s disease, W.B.
Sardinian population. Clin Chem 49: 496–498. Saunders, Philadelphia.
Lyon TD, Fell GS, Gaffney D et al. (1995). Use of a stable cop- Schilsky ML (2014a). A century for progress in the diagnosis
per isotope (65Cu) in the differential diagnosis of Wilson’s of Wilson disease. J Trace Elem Med Biol 28: 492–494.
disease. Clin Sci (Lond) 88: 727–732. Schilsky ML (2014b). Long-term outcome for Wilson disease:
Mak CM, Lam CW, Tam S et al. (2008). Mutational analysis of 85% good. Clin Gastroenterol Hepatol 12: 690–691.
65 Wilson disease patients in Hong Kong Chinese: identi- Schols L, Arning L, Schule R et al. (2008). “Pseudodominant
fication of 17 novel mutations and its genetic heterogene- inheritance” of ataxia with ocular apraxia type 2 (AOA2).
ity. J Hum Genet 53: 55–63. J Neurol 255: 495–501.
Marecek Z, Nevsimalova S (1984). Biochemical and clinical Shah AB, Chernov I, Zhang HT et al. (1997). Identification
changes in Wilson’s disease heterozygotes. J Inherit and analysis of mutations in the Wilson disease gene
Metab Dis 7: 41–45. (ATP7B): population frequencies, genotype–phenotype
Margarit E, Bach V, Gomez D et al. (2005). Mutation analysis correlation, and functional analyses. Am J Hum Genet
of Wilson disease in the Spanish population – identification 61: 317–328.
of a prevalent substitution and eight novel mutations in the Shimizu N, Nakazono H, Watanabe A et al. (1997). Molecular
ATP7B gene. Clin Genet 68: 61–68. diagnosis of Wilson’s disease. Lancet 349: 1811–1812.
Merle U, Schaefer M, Ferenci P et al. (2007). Clinical presen- Sohtaoglu M, Ergin H, Ozekmekci S et al. (2007). Patient with
tation, diagnosis and long-term outcome of Wilson’s dis- late-onset Wilson’s disease: deterioration with penicilla-
ease: a cohort study. Gut 56: 115–120. mine. Mov Disord 22: 290–291.
Merle U, Weiss KH, Eisenbach C et al. (2010). Truncating Stapelbroek JM, Bollen CW, van Amstel JK et al. (2004). The
mutations in the Wilson disease gene ATP7B are associ- H1069Q mutation in ATP7B is associated with late and
ated with very low serum ceruloplasmin oxidase activity neurologic presentation in Wilson disease: results of a
and an early onset of Wilson disease. BMC Gastroenterol meta-analysis. J Hepatol 41: 758–763.
10: 8. Sternlieb I, Scheinberg IH (1968). Prevention of Wilson’s dis-
Merli M, Patriarca M, Loudianos G et al. (1998). Use of the ease in asymptomatic patients. N Engl J Med 278: 352–359.
stable isotope 65Cu test for the screening of Wilson’s dis- Svetel M, Potrebic A, Pekmezovic T et al. (2009).
ease in a family with two affected members. Ital Neuropsychiatric aspects of treated Wilson’s disease.
J Gastroenterol Hepatol 30: 270–275. Parkinsonism Relat Disord 15: 772–775.
EPIDEMIOLOGY AND INTRODUCTION TO THE CLINICAL PRESENTATION OF WILSON DISEASE 17
Taly AB, Meenakshi-Sundaram S, Sinha S et al. (2007). Wang LH, Huang YQ, Shang X et al. (2011). Mutation anal-
Wilson disease: description of 282 patients evaluated over ysis of 73 southern Chinese Wilson’s disease patients: iden-
3 decades. Medicine (Baltimore) 86: 112–121. tification of 10 novel mutations and its clinical correlation.
Tanzi RE, Petrukhin K, Chernov I et al. (1993). The Wilson J Hum Genet 56: 660–665.
disease gene is a copper transporting ATPase with homol- Weiss KH, Gotthardt DN, Klemm D et al. (2011). Zinc mono-
ogy to the Menkes disease gene. Nat Genet 5: 344–350. therapy is not as effective as chelating agents in treatment of
Tarnacka B, Gromadzka G, Rodo M et al. (2000). Frequency of Wilson disease. Gastroenterology 140: 1189–1198. e1181.
His1069Gln and Gly1267Lys mutations in Polish Wilson’s Yamaguchi Y, Aoki T, Arashima S et al. (1999). Mass screen-
disease population. Eur J Neurol 7: 495–498. ing for Wilson’s disease: results and recommendations.
Tarnacka B, Szeszkowski W, Buettner J et al. (2009). Pediatr Int 41: 405–408.
Heterozygous carriers for Wilson’s disease – magnetic Zappu A, Magli O, Lepori MB et al. (2008). High incidence
spectroscopy changes in the brain. Metab Brain Dis 24: and allelic homogeneity of Wilson disease in 2 isolated
463–468. populations: a prerequisite for efficient disease prevention
Thomas GR, Jensson O, Gudmundsson G et al. (1995). Wilson programs. J Pediatr Gastroenterol Nutr 47: 334–338.
disease in Iceland: a clinical and genetic study. Am J Hum Zappu A, Lepori MB, Incollu S et al. (2010). Development of
Genet 56: 1140–1146. TaqMan allelic specific discrimination assay for detection
Trocello JM, El Balkhi S, Woimant F et al. (2014). Relative of the most common Sardinian Wilson’s disease mutations.
exchangeable copper: a promising tool for family screening Implications for genetic screening. Mol Cell Probes 24:
in Wilson disease. Mov Disord 29: 558–562. 233–235.
Handbook of Clinical Neurology, Vol. 142 (3rd series)
Wilson Disease
A. Członkowska and M.L. Schilsky, Editors
http://dx.doi.org/10.1016/B978-0-444-63625-6.00003-3
© 2017 Elsevier B.V. All rights reserved

Chapter 3

The genetics of Wilson disease


IRENE J. CHANG1 AND SI HOUN HAHN2*
1
Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
2
Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle Children’s Hospital,
Seattle, WA, USA

Abstract
Wilson disease (WD) is an autosomal-recessive disorder of hepatocellular copper deposition caused by
pathogenic variants in the copper-transporting gene, ATP7B. Early detection and treatment are critical
to prevent lifelong neuropsychiatric, hepatic, and systemic disabilities. Due to the marked heterogeneity
in age of onset and clinical presentation, the diagnosis of Wilson disease remains challenging to physicians
today. Direct sequencing of the ATP7B gene is the most sensitive and widely used confirmatory testing
method, and concurrent biochemical testing improves diagnostic accuracy. More than 600 pathogenic var-
iants in ATP7B have been identified, with single-nucleotide missense and nonsense mutations being the
most common, followed by insertions/deletions, and, rarely, splice site mutations. The prevalence of Wil-
son disease varies by geographic region, with higher frequency of certain mutations occurring in specific
ethnic groups. Wilson disease has poor genotype–phenotype correlation, although a few possible modi-
fiers have been proposed. Improving molecular genetic studies continue to advance our understanding of
the pathogenesis, diagnosis, and screening for Wilson disease.

INTRODUCTION excluded simply due to a misleading family history con-


sistent with an autosomal-dominant inheritance pattern.
In this chapter, we will discuss the inheritance, gene fre-
Furthermore, recent studies have also identified Wilson
quency, variants, genotype–phenotype correlation, and
disease due to atypical forms of inheritance, such as
modifiers of the ATP7B gene, and the clinical molecular
the presence of three concurrent mutations in a single
diagnosis and population screening for Wilson disease.
patient or segmental uniparental disomy (Coffey et al.,
2013). Uniparental disomy occurs when both homologs
INHERITANCE of a chromosome originate from a single parent. These
findings have implications for clinical practice and
Wilson disease is a monogenic autosomal-recessive con-
genetic counseling, as clinicians may need to consider
dition and carriers do not manifest any symptoms.
genotyping asymptomatic parents or obtaining full
Autosomal-recessive conditions are not usually present
sequencing of ATP7B to confirm that pathogenic variants
in consecutive generations, but may occur in populations
occur in trans.
with particularly high carrier frequency of Wilson dis-
ease (Wu et al., 2015). Our group and others have
ATP7B GENE AND ATPASE
reported the presence of Wilson disease in two or more
successive generations within the same family, reflecting Wilson disease is caused by homozygous or compound
a “pseudo-dominant” inheritance (Dziezyc et al., 2011, heterozygous mutations in the ATP7B gene (OMIM#
2014; Bennett et al., 2013; H. Park et al., 2015). There- 606882), which encodes a transmembrane copper-
fore, the diagnosis of Wilson disease should not be transporting P-type ATPase of the same name. Currently,

*Correspondence to: Si Houn Hahn, MD, PhD, Department of Pediatrics, University of Washington School of Medicine, Seattle
Children’s Hospital, Seattle WA 98105, USA. Tel: +1-206-987-7610, Fax: +1-206-987-5329, E-mail: sihahn@uw.edu
20 I.J. CHANG AND S.H. HAHN

Fig. 3.1. Schematic representation of copper-induced relocalization of ATP7A and ATP7B. The left side of the diagram represents
an enterocyte and the right side represents a hepatocyte. On both sides, copper enters the cell through copper transporter 1 (CTR1)
and is escorted by copper chaperone antioxidant protein 1 (ATOX1) to ATP7A or ATP7B in the trans-Golgi network (TGN). When
copper levels rise above a certain threshold, ATP7A and ATP7B excrete copper into the plasma on the basolateral side of the
enterocyte and into the bile on the apical side of the hepatocyte. Defects in localization of ATP7B may lead to copper accumulation
at the (1) TGN due to unresponsiveness, (2) cell periphery, and (3) endoplasmic reticulum (ER) due to misfolding. (Reproduced
from de Bie et al., 2007.)

ATP7B is the only identified gene known to cause Wil copper into vesicles and excretes it via exocytosis across
son disease (Bull et al., 1993; Petrukhin et al., 1993; the apical canalicular membrane into bile (Bull et al.,
Tanzi et al., 1993). Mutations in the ATP7B gene have 1993; Tanzi et al., 1993; Yamaguchi et al., 1999; Cater
been reported in almost all exons. Previous studies have et al., 2007). Due to the binary role of the ATP7B trans-
reported individuals with both biochemical and clinical porter in both the synthesis and excretion of copper,
diagnosis of Wilson disease in the absence of two defects in its function lead to copper accumulation and
ATP7B mutations, raising the possibility of a second the progressive features of Wilson disease (Fig. 3.1).
causative gene (Lovicu et al., 2006; Kenney and Cox,
2007; S. Park et al., 2007; Mak and Lam, 2008; MOLECULAR STRUCTURE OF ATP7B
Nicastro et al., 2010; Coffey et al., 2013). Nonetheless,
ATP7B is located on 13q14.3 and contains 20 introns and
ATP7B remains the only known gene responsible for
21 exons, for a total genomic length of 80 kb (Bull et al.,
Wilson disease.
1993; Petrukhin et al., 1993; Tanzi et al., 1993). The gene
Human dietary intake of copper is about
is synthesized in the endoplasmic reticulum, then relo-
1.5–2.5 mg/day, which is absorbed in the stomach and
cated to the TGN within hepatocytes. ATP7B is most
duodenum, bound to circulating albumin, and trans-
highly expressed in the liver, but is also found in the kid-
ported to the liver for regulation and excretion (Culotta
ney, placenta, mammary glands, brain, and lung.
and Scott, 2016). The uptake of copper occurs on the
basolateral side of hepatocytes via copper transporter 1
ATP7B (P-TYPE ATPASE) PROTEIN
(CTR1), as illustrated in Figure 3.1. A specific copper
STRUCTURE AND FUNCTION
chaperone, antioxidant protein 1 (ATOX1), delivers cop-
per to the Wilson disease protein, ATP7B, by copper- ATP7B belongs to class 1B (PIB) of the highly conserved
dependent protein–protein interactions (Walker et al., P-type ATPase superfamily, which is responsible for
2004). Within hepatocytes, ATP7B performs two impor- the transport of copper and other heavy metals across
tant functions in either the trans-Golgi network (TGN) or cellular membranes (Gourdon et al., 2011). The protein
in cytoplasmic vesicles. In the TGN, ATP7B activates contains 1465 amino acids, a phosphatase domain
ceruloplasmin by packaging six copper molecules into (A-domain), phosphorylation domain (P-domain, amino
apoceruloplasmin, which is then secreted into the acid residues 971–1035), nucleotide-binding domain
plasma. In the cytoplasm, ATP7B sequesters excess (N-domain, amino acid residues 1240–1291), and
THE GENETICS OF WILSON DISEASE 21

5’UTR 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

M Tm Tm Tm Tm Tm Tm Tm Tm

GD
Cu PD
D COOH

GV
K

ND
Cu T
Cu G
NH2 Hinge region
Cu Phosphorylation
Cu
domain

Cu N
SEHPL D
TG
ATP binding site

Fig. 3.2. Schematic representation of ATP7B gene and corresponding human ATP7B protein. Top diagram shows 5’UTR pro-
moter region and exons separated by introns. Bottom diagram shows the domain organization of human copper ATPase. Conserved
amino acid motifs are present at the core structure of each functional domain, i.e., TGDN and GDGVND at the A-domain, DKTG at
the P-domain, and SEHPL in the N-domain. M, phospholipidic bilayer of the membrane; Cu, the metal-binding domains of the
trasmembrane cation channel; Tm, transmembrane domains; PD, phosphatase domain. (Reproduced from Fanni et al., 2005.)

M-domain, which comprises eight transmembrane ion Although the mechanism by which the histadine-
channels (Fig. 3.2) (Cater et al., 2004, 2007; containing SEHPL motif affects copper transport
Lenartowicz and Krzeptowski, 2010). remains to be elucidated, it is clear that histidine-to-
Unique amino acid motifs are present at the core struc- glutamate substitution at amino acid 1069 (p.H1069Q)
ture of each domain, such as TGEA at the A-domain, in this motif is the most common cause of Wilson disease
DKTGT at the P-domain, and SEHPL in the in northern Europeans. In the hepatocytes of patients
N-domain. Specifically, the N-terminal metal-binding homozygous for p.H1069Q, ATP7B was found in the
domain (MBD) is composed of six copper-binding sites, endoplasmic reticulum instead of its usual TGN location,
each with the conserved sequence motif GMXCXXC suggesting abnormal protein trafficking (Huster et al.,
(Fatemi and Sarkar, 2002; Sazinsky et al., 2006). These 2003). Insect models with the p.H1069Q mutation in
MBDs play a central role in accepting copper from cop- SF9 cells showed decreased ATP-mediated catalytic
per chaperone ATOX1 through protein–protein interac- phosphorylation but no major protein misfolding, sug-
tions. Previous studies have demonstrated unequal gesting a role for p.H1069Q in the orientation of the
impact of MBDs on ATP7B activity, with MBD 5 and ATP7B catalytic site for ATP binding prior to hydrolysis
6 having stronger effects on the catalytic activation of (Tsivkovskii et al., 2003).
ATP7B than MBDs 1–4 (Lutsenko et al.,1997).
The active transport of copper across membranes is a
VARIANTS IN THE ATP7B GENE
complex process that begins with ATP7B binding copper
at the N-terminal domain and transporting it across cel- More than 600 pathogenic variants in ATP7B have been
lular membranes, using ATP as an energy source identified, with single-nucleotide missense and nonsense
(Fig. 3.2). Next, free copper binds intracellularly to mutations being the most common, followed by inser-
GG motifs in the MBDs, followed by transport on to tions/deletions and splice site mutations (Human Gene
the Cys-Pro-Cys (CPC) sequence motifs in MBD 6. Mutation Database, accessed 29 April 2016; Stenson
Finally, dephosphorylation of acyl-phosphate at the et al., 2014). Other rare genetic mechanisms that have
A-domain discharges copper across the cellular mem- been reported in the literature include whole-exon dele-
brane. Mutations causing copper accumulation may tions, promoter region mutations, three concurrent path-
occur at any of these steps (Huster et al., 2006; ogenic variants, and monogenic disomy (Coffey et al.,
Schushan et al., 2012). 2013; Bandmann et al., 2015). Mutation “hotspots” in
22 I.J. CHANG AND S.H. HAHN
Transmembrane
Cu binding Transmembrane (1-6) ATP binding (7-8)

2 3 4 5 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

p.C271* p.M645R p.R778L p.L795F p.G943D c.3400delC p.N1270S c.4193delC


p.E122fs p.Q457* p.G710S p.A874V p.L1083F p.L1273S p.Q1399R
p.c.1708-1G>C p.N1270S
c.2299insC p.P992H p.A1135Q p.Q1399R
c.-441_-427del
p.G691R p.I1002T p.G1266R p.S774R
p.S774R p.A1003V
p.Y670*
p.W779X p.H1069Q c.3903+6C>T
p.L708P p.V845fs p.E1064K
p.K832R p.G1351*
p.M769fs p.G1061E p.V1146M
p.L1371P
p.W779* p.A11140V
p.R969Q
p.A1003T p.A1003T
p.P992L
p.P992H
p.T977M
p.N958fs
Fig. 3.3. Schematic of the ATP7B gene with common mutation sites, including p.H1069Q (rs76151636), p.R778L (rs28942074),
p.E1064K (rs376910645), c.3400delC, and p.Ala1135fs (rs137853281). Please refer to Table 3.1 for more details.

ATP7B have also been reported to vary by geographic et al., 2007; van den Berghe et al., 2009). Other preva-
region (see regional gene frequency section, below). lent mutations, such as protein-truncating nonsense
The majority of pathogenic mutations are located in mutations (13% of known point mutations) (Merle
the M- and N-domains in presymptomatic patients or et al., 2010) and frameshift mutations (Vrabelova
in those with hepatic symptoms (S. Park et al., 2007). et al., 2005), are predicted to cause decay of mRNA
The common mutations in ATP7B seen in various popu- (Mendell et al., 2004; Chang et al., 2007) or a severely
lations are listed in Figure 3.3. truncated protein, resulting in absent or diminished levels
The p.H1069Q mutation is one of the most common of protein. It is therefore expected that most patients with
mutations, with a population allelic frequency of Wilson disease have absent or significantly reduced
10–40% (30–70% among Caucasians). Most patients levels of ATP7B.
are compound heterozygotes, carrying different muta-
tions on each copy of the chromosome (Usta et al., REGIONAL GENE FREQUENCY
2014). The p.H1069Q mutation occurs when histidine
The prevalence of Wilson disease varies by geographic
of the conserved SEHPL motif in the N-domain of
region, with higher prevalence of specific mutations
ATP7B is replaced by glutamic acid, resulting in
reported in certain populations (Ferenci, 2006) (see
N-domain protein misfolding, abnormal phosphoryla-
Chapter 2 for more details). A list of the common
tion in the P-domain, and decreased ATP binding affinity
regional variants of ATP7B mutations and geographic
(Rodriguez-Granillo et al., 2008). This mutation also
clustering of mutations are shown in Table 3.1 and
leads to decreased heat stability and abnormal localiza-
Figure 3.4, respectively.
tion of the protein to the TGN (Ralle et al., 2010).
Other common mutations in ATP7B include p.
GENOTYPE–PHENOTYPE CORRELATION
E1064A, p.R778L, p.G943S, and p.M769V. Mutations
in p.E1064A, also found in the SEHPL motif, completely Direct genotype–phenotype relationships in Wilson dis-
disable ATP binding affinity but do not result in protein ease have been difficult to establish, despite several stud-
misfolding, transport abnormalities, or thermal instabil- ies examining correlation (Panagiotakaki et al., 2004;
ity. The p.R778L mutation affects transmembrane trans- Vrabelova et al., 2005; Nicastro et al., 2010; Cocoş
port of copper (Dmitriev et al., 2011). The p.G943S and et al., 2014; Usta et al., 2014). The numerous
p.M769V mutations result in defective copper metabo- low-frequency and compound heterozygous nature of
lism but preserved ceruloplasmin levels (Okada Wilson disease obfuscate the process of characterizing
et al., 2010). its numerous genetic variants and their clinical conse-
A substantial proportion of Wilson disease-associated quences. Descriptions of phenotypes are limited to age
missense mutations, including p.H1069Q and p.R778L, of onset and presenting symptoms, both of which may
result in markedly decreased level of the protein caused be affected by inaccurate diagnostic criteria, delayed
by enhanced degradation (Payne et al., 1998; de Bie diagnosis, and practitioner selection bias. Therefore,
Table 3.1
Regional distribution of common Wilson disease mutations by geographic location

Prevalent mutations

Region AF (%) Protein Nucleotide RS Exon Type Domain

Europe
Austria (Ferenci, 2006) 34.1 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
6.4 p.Gly710Ser c.2128G > A 8 Missense TM2
3.6 p.Met769fs c.2298_2299insC rs137853287 8 Premature stop TM4
Benelux (Ferenci, 2006) 53 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
Bulgaria (Todorov et al., 2005) 58.8 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
Canary Islands 64 p.Leu708Pro c.2123 T > C 8 Missense TM2
(García Villarreal et al., 2000)
Czech Republic 57 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Vrabelova et al., 2005)
Denmark 18 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Møller et al., 2011)
16 p.Trp779* c.2336G > A rs137853283 8 Nonsense TM4
France 15 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Bost et al., 2012)
Germany 47.9 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Ferenci, 2006)
Germany (East, former) 63 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Caca et al., 2000)
Greece 35 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Panagiotakaki et al., 2004;
Dedoussis et al., 2005; Gomes
and Dedoussis, 2016)
12 p.Arg969Gln c.2906G > A rs774028495 13 Missense TM6
Hungary 42.9 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Firneisz et al., 2002; Folhoffer et al., 2007)
Iceland 100 p.Tyr670* c.2007_2013del 7 Nonsense TM1
(Thomas et al., 1995a; Hofer et al., 2012)
Italy 17.5 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Loudianos et al., 1999)
9 p.Val845fs c.2530delA rs755709270 10 Premature stop Td
6 p.Met769fs c.2298_2299insC rs137853287 8 Premature stop TM4
Netherlands 33 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Stapelbroek et al., 2004)

Continued
Table 3.1
Continued

Prevalent mutations

Region AF (%) Protein Nucleotide RS Exon Type Domain

Poland 72 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop


(Gromadzka et al., 2005)
7.3 p.Ala1135fs c.3400delC rs137853281 15 Premature stop ATP loop
3.7 p.Gln1351* c.4051C > T 20 Nonsense
Romania (Iacob et al., 2012) 38.1 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
Russia 49 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Ivanova-Smolenskaya et al., 1997)
Sardinia 60.5 c.-441_-427del 5prime Unknown Promoter
(Figus et al., 1995)
8.5 p.Met822fs c.2463delC 10 Deletion TM4/Td
7.9 p.Val1146Met c.3436G > A 16 Missense ATP loop
Serbia 38.4 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Tomic et al., 2013)
11.6 p.Met769fs c.2304dupC 8 Missense TM4
9.3 p.Ala1003Thr c.3007G > A rs1801247 13 Missense TM6/Ph
Spain 27 p.Met645Arg c.1934 T > G rs121907998 6 Missense Cu6/TM1
(Margarit et al., 2005)
Sweden 38 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Shah et al., 1997)
Turkey 17.4 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Ferenci, 2006; Simsek Papur et al., 2013)
5.3 p.Gly710Ser c.2128G > A rs772595172 8 Missense TM2
4.53 p.Gln457* c.1369C > T 3 Nonsense Cu4/Cu5
UK 19 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Coffey et al., 2013)
8 p.Met769Val c.2305A > G 8 Missense TM4
Yugoslavia (former) 48.9 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Loudianos et al., 1999)
11.4 p.Met769fs c.2298_2299insC rs137853287 8 Premature stop TM4
Asia
China 31 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4
(Gu et al., 2003; Z.-Y. Wu et al., 2003;
Wang et al., 2011; Wei et al., 2014)
10 p.Pro992Leu c.2975C > T rs201038679 13 Missense TM6/Ph
9.6 p.Ile1148Thr c.3443 T > C rs60431989 16 Missense ATP loop
3.3 p.Thr935Met c.2804C > T 12 Missense TM5
19 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4
North India
(S. Kumar et al., 2006; Gupta et al., 2007)
12 p.Ile1102Thr c.3305 T > C rs560952220 15 Missense ATP loop
9 p.Pro992His c.2975C > A 13 Missense TM6/Ph
South India 11 p.Ala1003Val c.3008C > T 13 Missense TM6/Ph
(Santhosh et al., 2006;
S. S. Kumar et al., 2012)
11 p.Cys271* c.813C > A rs572147914 2 Nonsense Cu3
9 p.Pro768Leu c.2303C > T 8 Missense TM4
9 p.Arg969Gln c.2906G > A rs121907996 13 Missense TM6
East India 16 p.Cys271* c.813C > A rs572147914 2 Nonsense Cu3
(Gupta et al., 2005)
11 p.Gly1061Glu c.3182G > A 14 Missense ATP loop
8.5 c.1708-1G > C rs137853280 5 Splice Cu6
West India 20 p.Cys271* c.813C > A rs572147914 2 Nonsense Cu3
(Aggarwal and Bhatt, 2013;
Aggarwal et al., 2013)
11 p.Glu122fs c.365_366delins 2 Ins/Del Cu1
TTCGAAGC
6 p.Thr977Met c.2930C > T rs72552255 13 Missense TM6
6 p.Leu795Phe c.2383C > T 9 Missense TM4/Td
Japan 17.95 p.Asn958fs c.2871delC 13 Premature stop TM5/TM6
(Okada et al., 2000; Tatsumi et al., 2010)
16.7 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4
10.5 c.1708-5 T > G 5 Splice Cu6
Korea 37.9 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4
(E. K. Kim et al., 1998; Yoo, 2002;
G.-H. Kim et al., 2008; Song et al., 2012)
12.1 p.Asn1270Ser c.3809A > G rs121907990 18 Missense ATP hinge
9.4 p.Ala874Val c.2621C > T rs376355660 11 Missense TM5
8 p.Leu1083Phe c.3247C > T 15 Missense ATP loop
Lebanon 44.7 p. Ala1003Thr c.2299insC rs137853287 8 Missense TM4
(Usta et al., 2014)
Saudi Arabia 32 p.Gln1399Arg c.4196A > G 21 Missense After TM8
(Al Jumah et al., 2004; Majumdar et al., 2004)
16 p.Ser774Arg c.2230 T > C rs535217574 21 Missense TM3
Taiwan 29.6 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4
(Lee et al., 2000; Wan et al., 2006)
8.9 p.Pro992Leu c.2975C > T rs201038679 13 Missense TM6
4.8 p.Gly943Asp c.2828G > A 12 Missense TM5

Continued
Table 3.1
Continued

Prevalent mutations

Region AF (%) Protein Nucleotide RS Exon Type Domain

Thailand 10.52 p.Arg778Leu c.2332C > T rs28942074 8 Missense TM4


(Panichareon et al., 2011)
7.89 p.Leu1371Pro c.4112 T > C 20 Missense TM8
Iran 19 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Zali et al., 2011)
Africa
Egypt 42.2 IVS18 + 6 T > C c.3903 + 6C > T rs2282057 18 Splice
(Abdelghaffar et al., 2008;
Abdel Ghaffar et al., 2011)
40.6 p.Ala11140Val c.3419C > T 16 Missense ATP loop
26.5 p.Lys832Arg c.2495A > G rs1061472 10 Missense TM4/Td
Americas
USA 40.3 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Kuppala et al., 2009)
1.9 p.Asn1270Ser c.3809A > G rs121907990 18 Missense ATP hinge
1.9 p.Gly1266Arg c.3796G > A rs121907992 18 Missense ATP hinge
Brazil 37.1 p.His1069Gln c.3207C > A rs76151636 14 Missense ATP loop
(Deguti et al., 2004; Machado et al., 2008;
Bem et al., 2013)
31.25 p.Ala1135fs c.3400delC rs137853281 15 Premature stop ATP loop
11.4 p.Ala1135GlnfsX13 c.3402delC rs137853281 15 Premature stop ATP loop
p.Leu708Pro c.2123 T > C 8 Missense TM2
Costa Rica 61 p.Asn1270Ser c.3809A > G rs121907990 18 Missense ATP hinge
(Shah et al., 1997)
Venezuela 26.9 p.Ala1135GlnfsX13 c.3402delC rs137853281 15 Premature stop ATP loop
(Paradisi et al., 2015)
9.6 p.Gly691Arg c.2071G > A 7 Missense TM2

AF, allelic frequency; RS, Reference single nucleotide polymorphism (SNP) cluster identification number.
THE GENETICS OF WILSON DISEASE 27

Fig. 3.4. Prevalence of ATP7B mutation by geographic region; the darker the gradient, the higher the allelic frequency. (Repro-
duced from Gomes and Dedoussis, 2016, with permission from Taylor and Francis.)

the marked variability in phenotype of Wilson disease is partial preservation of copper-transporting function, per-
likely attributable to an amalgamation of genetic, meta- haps explaining the milder phenotypes associated with
bolic, and environmental factors (Leggio et al., 2006). certain mutations (Rodriguez-Granillo et al., 2008;
The most consistent genotype–phenotype correlation Dmitriev et al., 2011; Huster et al., 2012). Individuals
in Wilson disease is that the most severe, early-onset dis- with the R778L mutation have been shown to have an
ease with predominantly hepatic presentation is associ- earlier onset of disease and predominantly hepatic pre-
ated with mutations causing absent ATPase activity. sentation (Z. Y. Wu et al., 2003). In contrast, individuals
Convincing studies have demonstrated fulminant hepatic with the H1069Q mutation have a mean onset of symp-
disease in mouse models such as the toxic milk (tx) toms between 20–22 years old and predominantly neuro-
mouse and the Jackson tx mouse (txj), which harbor point logic phenotype (Stapelbroek et al., 2004; Kalita et al.,
mutations causing loss of ATP7B function, but not affect- 2010). There is also some evidence that Kayser–
ing ATP7B synthesis (Theophilos et al., 1996; Coronado Fleischer rings are more common in H1069Q homozy-
et al., 2001; La Fontaine et al., 2001; Huster et al., 2006). gous patients in Hungary at time of diagnosis than in
Genetic polymorphisms in ATP7B, other genes, and compound heterozygous individuals (Folhoffer
epigenetic factors have been shown to impact disease et al., 2007).
phenotype by affecting ATP7B protein structure and Moreover, pathogenic variants may affect ATP7B tar-
function. Of the over 600 mutations associated with Wil- geting from the TGN to cytosolic vesicles. For instance,
son disease, the majority are missense mutations that the p.Met875Val mutation results in a less stable protein
completely inactivate the copper-transporting function and causes reversible ATP7B localization defects. Under
of ATP7B (Lutsenko, 2014). In general, individuals with a low-copper environment, the p.Gly875Arg variant is
protein-truncating mutations have earlier onset of disease sequestered in the endoplasmic reticulum. However,
due to decreased protein stability and quantity (Merle addition of exogenous copper to the cellular growth
et al., 2010). However, other studies have demonstrated medium stabilizes the protein, allowing it to complete
28 I.J. CHANG AND S.H. HAHN
its intended journey to the TGN and overcoming its raise the suspicion for epigenetic modifiers in Wilson
disease-causing phenotype. Theoretically, patients with disease. See Chapter 4 for more details about the genetic
this specific variant may be more sensitive to dietary cop- and environmental modifiers of Wilson disease.
per deficiency (Gupta et al., 2011).
The timing and location of copper buildup can also
CLINICAL MOLECULAR DIAGNOSIS
preferentially alter the hepatic transcriptome, based
on homozygous ATP7B–/– mouse models. Proteomic The current gold standard of diagnosis for Wilson disease
analyses of mRNA profiles at each of these disease is direct Sanger sequencing of the ATP7B gene or molec-
stages reflect unique patterns (Huster et al., 2006; ular testing for previously identified familial mutations.
Ralle et al., 2010). In the initial stage, mRNA for pro- Historically, most pathogenic variants in ATP7B were
teins responsible for cell cycle regulation, splicing, identified using a combination of polymerase chain reac-
and cholesterol synthesis is present (Burkhead et al., tion (PCR)/restriction fragment length polymorphism
2011). This leads to early accumulation of copper (RFLP), single-strand conformation polymorphism
bound to metallothioneins in the cytosol and free copper (SSCP), denaturing gradient gel electrophoresis
in the nuclei. In the progressive stage, mRNA changes (DGGE), temporal temperature gradient electrophoresis
throughout the cell are present, including the endoplas- (TTGE), denaturing high-performance liquid chroma-
mic reticulum, mitochondria, and endocytic pathways, tography (DHPLC), and Sanger sequencing
causing copper to pathologically accumulate within (Loudianos et al., 1999; Shimizu et al., 1999; Margarit
hepatocytes. In the later stages, mRNA for lysosomal et al., 2005; Vrabelova et al., 2005; G. H. Kim et al.,
and endosomal proteins is upregulated. In these final 2008). The critical demerits of this complex tiered
stages, copper concentrations decrease in the cytosol approach are that the detection rate is not high enough
and nuclei, and accumulate in the membranous cellular to find most mutations and the turnaround time is often
compartment, causing bile duct proliferation and extended. Although regional clusters of specific muta-
hepatic neoplastic changes. Therefore, the location of tions have been well described, a customized screening
copper accumulation may convey more specific prog- approach taking into account these regional variants
nostic information about disease progression rather may be complicated by ethnically diverse populations
than total copper levels. and inaccurate clinical information provided with
Other studies have compared homozygotes to com- samples. Biochemical results are often imprecise, as ele-
pound heterozygotes of the same mutation to establish vations in urinary copper excretion tend to occur late in
genotype–phenotype correlations. A study of 76 mem- the disease process and fewer than 40% of presympto-
bers of a large, consanguineous Lebanese family showed matic patients excrete copper less than 100 mg/day
an association between c.2299insC and hepatic disease (Sternlieb and Scheinberg, 1968; Nakayama et al.,
and between the p.Ala1003Thr mutation and neurologic 2008). For these reasons, direct sequencing of the ATP7B
disease (Usta et al., 2014). gene has become the preferred standard and provides the
Other candidate polymorphisms that are thought to greatest yield in clinical molecular diagnosis. Please refer
modify the clinical phenotype of Wilson disease include to Chapter 14 for details about the diagnosis of Wilson
MTHFR (Gromadzka et al., 2005), COMMD1 (Weiss, disease.
2006), ATOX1 (Simon, 2008), XIAP (Weiss et al., Starting the diagnostic process with molecular testing
2010), PNPLA3 and hepatic steatosis (St€attermayer may significantly reduce the need for invasive liver
et al., 2012), and DMT1 (Przybyłkowski et al., 2014), biopsy. Liver copper content alone was found to be insuf-
although none of these genes has been demonstrated to ficient to exclude Wilson disease, as levels may not be
have significant diagnostic or predictive value. elevated in some affected patients. Based on several
Significant phenotypic variation of Wilson disease previous studies, biallelic pathogenic variants were
exists between individuals with the same mutation, indi- identified in about 80% of patients with biochemical
viduals within the same family, and even between mono- and clinical tests suggestive of Wilson disease. Currently
zygotic twins (Członkowska et al., 2009; Kegley et al., available screening tests may not definitively rule out the
2010). While some studies have documented high intra- disease, and no single test could permit de novo diagno-
familial concordance of clinical symptoms and biochem- sis. Of note, many patients may not possess the charac-
ical results (Hofer et al., 2012; Chabik et al., 2014; teristic findings and may present when their clinical
Ferenci et al., 2015), others have reported a wide range disease is relatively mild. Inappropriate treatment for
in age of onset and presenting symptoms amongst sib- false-positive cases has the potential of inducing copper
lings (Ala et al., 2007; Taly et al., 2007) and families car- deficiency, which can result in hematologic and neuro-
rying the same mutation (Takeshita et al., 2002). Indeed, logic sequelae (N. Kumar et al., 2003). These findings
disparate clinical presentations in monozygotic twins reinforce the need for reliable clinical diagnostic criteria
THE GENETICS OF WILSON DISEASE 29
and underscore the benefits of DNA testing prior to inva- 3-year-old child (Wilson et al., 2000). Mutation analysis
sive procedures (Ferenci, 2005). has also confirmed late-onset disease, including the case
Multiplex PCR is used to amplify all 21 exons and of two siblings in their 70s – the oldest reported patients
splice sites of ATP7B, including promoter regions. so far at time of diagnosis (Nanji et al., 1997; Gupta et al.,
Although the large deletions or duplications cannot be 2005; Perri et al., 2005; Weitzman et al., 2014).
detected with this conventional Sanger sequencing ATP7B mutation analysis makes an important con-
method, the chance of these being present in Wilson tribution to clinical practice. Unfortunately, systematic
disease appears low (Stenson et al., 2012). If clinical genetic testing for Wilson disease is still difficult and
suspicion is still high with only one pathogenic variant fairly expensive due to the plethora of different muta-
identified, then multiplex ligation-dependent probe tions, the occurrence of regulatory mutations in non-
amplification (MLPA) test should be considered. Micro- coding sequence, the large size of the gene, and the
array-based comparative genomic hybridization is limitations of currently available methods. However,
another option to evaluate partial or full gene deletions technical advances allowing high-throughput screen-
or duplications with higher sensitivity. Cases with only ing could be applied to the disease (Bost et al.,
one pathogenic variant present should be carefully 2012; Lepori et al., 2012). This new apparatus can
reviewed in the context of other biochemical and clinical sequence six million basepairs of DNA per hour with
findings. Molecular genetic testing using direct mutation accuracy greater than 99%. Such advances might per-
analysis is very effective in identifying affected patients mit specialized laboratories to detect all variants by
and presymptomatic siblings of probands (Manolaki sequencing the entire genomic Wilson disease gene
et al., 2009). from patients, including not only the translated exons,
Wilson disease is an autosomal-recessive disorder, but also the important noncoding sequences that are
which means that there is a 25% chance that a full sibling not normally investigated.
of the index case is also affected. Once homozygous or Interpretation of variants of uncertain significance has
compound heterozygous mutations in ATP7B have been become a major challenge for accurate interpretation,
established in the index patient, mutation detection genetic counseling, and prevention. Screening family
becomes valuable in family screening. The same geno- members may help with the interpretation of variants
type in asymptomatic family members confirms diagno- of uncertain significance, but not all variants can be
sis of the disease, thus allowing for early treatment before resolved with this approach. Functional analysis is often
the onset of complications. In family members in whom necessary; however, no clinical functional analysis is
clinical and biochemical features are uncertain, the dem- currently available. A computational approach to predict
onstration of either heterozygous (carrier) or wild-type significance of mutations is often helpful, but a further
gene sequence prevents unnecessary treatment (Chang concrete model is required to demonstrate the efficacy
et al., 2007). in guiding clinical decisions.
If the proband has secured a diagnosis of Wilson dis-
ease on the basis of clinical and biochemical evidence,
POPULATION SCREENING
but testing for ATP7B mutations is not available, family
screening can be done by haplotype analysis of polymor- The purpose of newborn screening is to identify treatable
phic markers flanking the disease gene (Thomas et al., congenital conditions that can affect a child’s long-term
1995b; Gupta et al., 2005; Przybyłkowski et al., 2014). health and development. Recent tandem mass spectrom-
In this instance, the rare possibility of recombination etry (MS/MS) applications have markedly expanded the
events (typically 0.5–5% of cases) needs to be consid- ability to screen for >50 metabolic diseases from a single
ered. The rate of recombination is dependent on which dried blood spot. In addition to the original Wilson–
flanking markers are studied. Microsatellite or single- Jungner classic screening criteria (Wilson and Jungner,
nucleotide polymorphisms in the ATP7B lateral wing 1968), the American College of Medical Genetics con-
are used for haplotyping, which is useful for screening vened the Newborn Screening Expert Group to develop
relatives of patients with previously identified familial a uniform screening panel in 2006 (American College of
mutations. False-positive results may occur if hap- Medical Genetics Newborn Screening Expert Group,
lotyping is used on patients with low-probability gene 2006). Of the primary tenants, Wilson disease is an ideal
recombinations. target for screening, given its relatively high prevalence
Genetic testing for ATP7B mutations can be valuable and availability of effective treatment (Hahn et al., 2002;
to confirm a diagnosis of Wilson disease, especially Roberts et al., 2008). Unfortunately, despite extensive
when presentation is unusual (Caprai et al., 2006). Atten- discussion on the need for population screening, no
tion has been drawn to this situation by the molecular cost-effective biomarkers or methods for early detection
confirmation of early-onset hepatic disease in a have been developed for Wilson disease yet. Several
30 I.J. CHANG AND S.H. HAHN
small pilot studies have been conducted using cerulo- REFERENCES
plasmin as a biomarker for screening, with limited find-
Abdel Ghaffar TY, Elsayed SM, Elnaghy S et al. (2011).
ings (Yamaguchi et al., 1999; Hahn et al., 2002; Owada
Phenotypic and genetic characterization of a cohort
et al., 2002; Schilsky and Shneider, 2002; Kroll et al., of pediatric Wilson disease patients. BMC Pediatr 11
2006). Ceruloplasmin alone is not sufficient to screen (1): 56.
for Wilson disease in newborns, as a substantial number Abdelghaffar TY, Elsayed SM, Elsobky E et al. (2008).
of newborns present with physiologically low cerulo- Mutational analysis of ATP7B gene in Egyptian children
plasmin. Ceruloplasmin assay around 3 years of age with Wilson disease: 12 novel mutations. J Hum Genet
may be the most appropriate population-screening 53 (8): 681–687.
method, but mandatory health checkups at this age are Aggarwal A, Bhatt M (2013). Update on Wilson disease. Int
not universally available in the USA and worldwide. Rev Neurobiol 110: 313–348.
Many treatable congenital disorders are caused by Aggarwal A, Chandhok G, Todorov T et al. (2013). Wilson
disease mutation pattern with genotype–phenotype correla-
mutations that result in absent or diminished levels of
tions from Western India: confirmation of p.C271* as a
proteins; thus, protein biomarkers have enormous poten-
common Indian mutation and identification of 14 novel
tial in the diagnosis/screening of congenital disorders. mutations. Ann Hum Genet 77 (4): 299–307.
Liquid chromatography mass spectrometry with multiple Al Jumah M, Majumdar R, Al Rajeh S et al. (2004). A clinical
reaction monitoring (LC-MRM-MS) has emerged as a and genetic study of 56 Saudi Wilson disease patients: iden-
robust technology that enables highly precise, specific, tification of Saudi-specific mutations. Eur J Neurol 11 (2):
multiplex quantification of signature proteotypic pep- 121–124.
tides as stoichiometric surrogates of biomarker proteins. Ala A, Walker AP, Ashkan K et al. (2007). Wilson’s disease.
Our lab is currently exploring the use of peptide Lancet (London, England) 369 (9559): 397–408.
immunoaffinity enrichment (Whiteaker et al., 2010, American College of Medical Genetics Newborn Screening
2011) to quantify ATP7B in dried blood spot (DBS). Expert Group (2006). Newborn screening: toward a uni-
form screening panel and system – executive summary.
These promising proof-of-concept data open up the pos-
Pediatrics 117 (5 Pt 2): S296–S307.
sibility of screening for Wilson disease in newborns. Fur-
Bandmann O, Weiss KH, Kaler SG (2015). Wilson’s disease
ther clinical validation on a large-scale study will be and other neurological copper disorders. Lancet Neurol
required to determine the efficacy of the assay. 14 (1): 103–113.
Bem RS, Raskin S, Muzzillo DA, de et al. (2013). Wilson’s
disease in Southern Brazil: genotype–phenotype correla-
CONCLUSION
tion and description of two novel mutations in ATP7B
Wilson disease is an autosomal-recessive disease due to gene. Arq Neuropsiquiatr 71 (8): 503–507.
pathogenic mutations in ATP7B. ATP7B is the only iden- Bennett JT, Schwarz KB, Swanson PD et al. (2013). An excep-
tified gene known to cause Wilson disease, and encodes a tional family with three consecutive generations affected
transmembrane copper-transporting ATPase of the same by Wilson disease. JIMD Reports 10 (Chapter 206): 1–4.
Bost M, Piguet-Lacroix G, Parant F et al. (2012). Molecular
name. While biochemical testing and clinical criteria
analysis of Wilson patients: direct sequencing and MLPA
may assist in the early diagnosis and treatment, the cur-
analysis in the ATP7B gene and Atox1 and COMMD1 gene
rent gold standard for Wilson disease diagnosis is direct analysis. J Trace Elem Med Biol 26 (2–3): 97–101.
Sanger sequencing of ATP7B or molecular testing for Bull PC, Thomas GR, Rommens JM et al. (1993). The Wilson
known familial mutations. Genotype–phenotype correla- disease gene is a putative copper transporting P-type
tions have been studied extensively but direct causations ATPase similar to the Menkes gene. Nat Genet 5 (4):
remain nebulous. Modifier genes may affect the pene- 327–337.
trance and phenotypes but a large-scale study for clinical Burkhead JL, Ralle M, Wilmarth P et al. (2011). Elevated cop-
validation is warranted. The overall worldwide preva- per remodels hepatic RNA processing machinery in the
lence of Wilson disease is 1 in 30 000 individuals, with mouse model of Wilson’s disease. J Mol Biol 406 (1):
significant geographic variation. The most common 44–58.
Caca K, Ferenci P, Kuhn HJ et al. (2000). High prevalence of
mutation in Northern America and Europe is the mis-
the ATP7B-H1069Q mutation in Wilson disease patients
sense mutation p.H1069Q and the most common muta-
from Eastern Germany. J Hepatol 32: 134.
tion in East Asian populations is the missense p.R778L. Caprai S, Loudianos G, Massei F et al. (2006). Direct diagnosis
Ceruloplasmin alone is insufficient to screen for Wilson of Wilson disease by molecular genetics. J Pediatr 148 (1):
disease in newborns. While peptide immunoaffinity 138–140.
assays show promise for newborn screening, further Cater MA, Forbes J, La Fontaine S et al. (2004). Intracellular
large-scale clinical studies are required to determine effi- trafficking of the human Wilson protein: the role of the six
cacy of these population-based screening methods for N-terminal metal-binding sites. Biochem J 380 (3):
Wilson’s disease. 805–813.
THE GENETICS OF WILSON DISEASE 31
Cater MA, La Fontaine S, Mercer JFB (2007). Copper bind- Ferenci P, Litwin T, Seniow J et al. (2015). Encephalopathy in
ing to the N-terminal metal-binding sites or the CPC motif Wilson disease: copper toxicity or liver failure? J Clin Exp
is not essential for copper-induced trafficking of the Hepatol 5: S88–S95.
human Wilson protein (ATP7B). Biochem J 401 (1): Figus A, Arigius A, Loudianos G et al. (1995). Molecular
143–153. pathology and haplotype analysis of Wilson disease in
Chabik G, Litwin T, Członkowska A (2014). Concordance Mediterranean populations. Am J Hum Genet 57 (6):
rates of Wilson’s disease phenotype among siblings. 1318–1324.
J Inherit Metab Dis 37 (1): 131–135. Firneisz G, Lakatos PL, Szalay F et al. (2002). Common muta-
Chang Y-F, Imam JS, Wilkinson MF (2007). The nonsense- tions of ATP7B in Wilson disease patients from Hungary.
mediated decay RNA surveillance pathway. Annu Rev Am J Med Genet 108 (1): 23–28.
Biochem 76 (1): 51–74. Folhoffer A, Ferenci P, Csak T et al. (2007). Novel mutations
Cocoş R, Sendroiu A, Schipor S et al. (2014). Genotype– of the ATP7B gene among 109 Hungarian patients with
phenotype correlations in a mountain population com- Wilson’s disease. Eur J Gastroenterol Hepatol 19 (2):
munity with high prevalence of Wilson’s disease: 105–111.
genetic and clinical homogeneity. PLoS One 9 (6): Garcı́a Villarreal L, Daniels S, Shaw SH et al. (2000). High
e98520. prevalence of the very rare Wilson disease gene mutation
Coffey AJ, Durkie M, Hague S et al. (2013). A genetic study of Leu708Pro in the island of Gran Canaria (Canary
Wilson’s disease in the United Kingdom. Brain 136 (Pt 5): Islands, Spain): a genetic and clinical study. Hepatology
1476–1487. (Baltimore, Md) 32 (6): 1329–1336.
Coronado V, Nanji M, Cox DW (2001). The Jackson toxic milk Gomes A, Dedoussis GV (2016). Geographic distribution of
mouse as a model for copper loading. Mamm Genome 12: ATP7B mutations in Wilson disease. Ann Hum Biol 43
793–795. (1): 1–8.
Culotta V, Scott RA (2016). Metals in Cells, John Wiley, Gourdon P, Liu XY, Skjorringe T et al. (2011). Crystal struc-
Chichester. ture of a copper-transporting PIB-type ATPase. Nature 475
Członkowska A, Gromadzka G, Chabik G (2009). (7354): 59–64.
Monozygotic female twins discordant for phenotype of Gromadzka G, Schmidt HH, Genschel J et al. (2005).
Wilson’s disease. Mov Disord 24 (7): 1066–1069. Frameshift and nonsense mutations in the gene for
de Bie P, van de Sluis B, Burstein E et al. (2007). Distinct ATPase7B are associated with severe impairment of cop-
Wilson’s disease mutations in ATP7B are associated with per metabolism and with an early clinical manifestation
enhanced binding to COMMD1 and reduced stability of of Wilson’s disease. Clin Genet 68 (6): 524–532.
ATP7B. Gastroenterology 133 (4): 1316–1326. Gu Y-H, Kodama H, Du SL et al. (2003). Mutation spectrum
Dedoussis GVZ, Genschel J, Sialvera TE et al. (2005). Wilson and polymorphisms in ATP7B identified on direct
disease: high prevalence in a mountainous area of Crete. sequencing of all exons in Chinese Han and Hui ethnic
Ann Hum Genet 69 (Pt 3): 268–274. patients with Wilson’s disease. Clin Genet 64 (6): 479–484.
Deguti MM, Genschel J, Cancado EL et al. (2004). Wilson dis- Gupta A, Aikath D, Neogi R et al. (2005). Molecular pathogen-
ease: novel mutations in the ATP7B gene and clinical cor- esis of Wilson disease: haplotype analysis, detection of
relation in Brazilian patients. Hum Mutat 23 (4): 398. prevalent mutations and genotype–phenotype correlation
Dmitriev OY, Bhattacharjee A, Nokhrin S et al. (2011). in Indian patients. Hum Genet 118 (1): 49–57.
Difference in stability of the N-domain underlies distinct Gupta A, Chattopadhyay I, Dey S et al. (2007). Molecular
intracellular properties of the E1064A and H1069Q pathogenesis of Wilson disease among Indians: a perspec-
mutants of copper-transporting ATPase ATP7B. J Biol tive on mutation spectrum in ATP7B gene, prevalent
Chem 286 (18): 16355–16362. defects, clinical heterogeneity and implication towards
Dziezyc K, Groamadzka G, Czlonkowska A (2011). Wilson’s diagnosis. Cell Mol Neurobiol 27 (8): 1023–1033.
disease in consecutive generations of one family. Park Rel Gupta A, Bhattacharjee A, Dmitriev OY et al. (2011). Cellular
Dis 17: 577–578. copper levels determine the phenotype of the Arg875 var-
Dziezyc K, Litwin T, Chabik G et al. (2014). Families with iant of ATP7B/Wilson disease protein. Proc Natl Acad Sci
Wilson’s disease in subsequenr generations: clinical and U S A 108 (13): 5390–5395.
genetic analysis. Mov Dis 29: 1828–1832. Hahn SH, Lee SY, Jang YJ et al. (2002). Pilot study of mass
Fanni D, Pilloni L, Orru S et al. (2005). Expression of ATP7B screening for Wilson’s disease in Korea. Mol Genet
in normal human liver. Eur J Histochem 49 (4): 371–378. Metab 76 (2): 133–136.
Fatemi N, Sarkar B (2002). Molecular mechanism of copper Hofer H, Willheim-Polli C, Knoflauch P et al. (2012).
transport in Wilson disease. Environ Health Perspect 110 Identification of a novel Wilson disease gene mutation fre-
(Suppl 5): 695–698. quent in Upper Austria: a genetic and clinical study. J Hum
Ferenci P (2005). Wilson’s disease. Clin Gastroenterol Genet 57 (9): 564–567.
Hepatol 3 (8): 726–733. Huster D, Hoppert M, Lutsenko S et al. (2003). Defective cel-
Ferenci P (2006). Regional distribution of mutations of the lular localization of mutant ATP7B in Wilson’s disease
ATP7B gene in patients with Wilson disease: impact on patients and hepatoma cell lines. Gastroenterology 124
genetic testing. Hum Genet 120 (2): 151–159. (2): 335–345.
32 I.J. CHANG AND S.H. HAHN
Huster D, Finegold MJ, Morgan CT et al. (2006). Consequences mutation and evidence of haplotype-mutation association.
of copper accumulation in the livers of the Atp7b–/– (Wilson J Hum Genet 45 (5): 275–279.
disease gene) knockout mice. Am J Pathol 168 (2): 423–434. Leggio L, Addolorato G, Loudianos G et al. (2006). Genotype–
Huster D, Kuhne A, Bhattacharjee A et al. (2012). Diverse phenotype correlation of the Wilson disease ATP7B gene.
functional properties of Wilson disease ATP7B variants. Am J Med Genet A 140A (8): 933.
Gastroenterology 142 (4): 947–956.e5. Lenartowicz M, Krzeptowski W (2010). Structure and func-
Iacob R, Iacob S, Nastase A et al. (2012). The His1069Gln tion of ATP7A and ATP7B proteins – Cu-transporting
mutation in the ATP7B gene in Romanian patients with ATPases. Postepy Biochem 56: 317–327.
Wilson’s disease referred to a tertiary gastroenterology Lepori M-B, Zappu A, Incollu S et al. (2012). Mutation anal-
center. J Gastrointestin Liver Dis 21 (2): 181–185. ysis of the ATP7B gene in a new group of Wilson’s disease
Ivanova-Smolenskaya I, Karabanov AV, Illarioshkin SN et al. patients: contribution to diagnosis. Mol Cell Probes 26 (4):
(1997). 5-29-06 Molecular analysis in Russian families 147–150.
with Wilson’s disease. J Neurol Sci 150: S314. Loudianos G, Dessi V, Lovicu M et al. (1999). Mutation anal-
Kalita J, Somarajan BI, Misra UK et al. (2010). R778L, ysis in patients of Mediterranean descent with Wilson dis-
H1069Q, and I1102T mutation study in neurologic ease: identification of 19 novel mutations. J Med Genet 36
Wilson disease. Neurol India 58 (4): 627–630. (11): 833–836.
Kegley KM, Sellers MA, Ferber MJ et al. (2010). Fulminant Lovicu M, Dessi V, Lepori MB et al. (2006). The canine
Wilson’s disease requiring liver transplantation in one copper toxicosis gene MURR1 is not implicated in the path-
monozygotic twin despite identical genetic mutation. Am ogenesis of Wilson disease. J Gastroenterol 41 (6):
J Transplant 10 (5): 1325–1329. 582–587.
Kenney SM, Cox DW (2007). Sequence variation database for Lutsenko S (2014). Modifying factors and phenotypic diver-
the Wilson disease copper transporter, ATP7B. Hum Mutat sity in Wilson’s disease. Ann N Y Acad Sci 1315 (1):
28 (12): 1171–1177. 56–63.
Kim EK, Yoo OJ, Song KY et al. (1998). Identification of three Lutsenko S, Petrukhin K, Cooper MJ et al. (1997). N-terminal
novel mutations and a high frequency of the Arg778Leu domains of human copper-transporting adenosine tripho-
mutation in Korean patients with Wilson disease. Hum sphatases (the Wilson’s and Menkes disease proteins) bind
Mutat 11 (4): 275–278. copper selectively in vivo and in vitro with stoichiometry of
Kim G-H, Jang JY, Park JY et al. (2008). Estimation of one copper per metal-binding repeat. J Biol Chem 272 (30):
Wilson’s disease incidence and carrier frequency in the 18939–18944.
Korean population by screening ATP7B major mutations Machado AAC, Deguti MM, Genschel J et al. (2008).
in newborn filter papers using the SYBR green intercalator Neurological manifestations and ATP7B mutations in
method based on the amplification refractory mutation sys- Wilson’s disease. Parkinsonism Relat Disord 14 (3):
tem. Genet Test 12 (3): 395–399. 246–249.
Kroll CA, Ferber MJ, Dawson BD et al. (2006). Retrospective Majumdar R, Al-Jumah M, Zaidan R (2004). A rare homozy-
determination of ceruloplasmin in newborn screening gous missense mutation in ATP7B exon 19 in a case of
blood spots of patients with Wilson disease. Mol Genet Wilson disease. Eur Neurol 51 (1): 52–54.
Metab 89 (1–2): 134–138. Mak CM, Lam C-W (2008). Diagnosis of Wilson’s disease: a
Kumar N, Gross JB, Ahlskog JE (2003). Myelopathy due to comprehensive review. Crit Rev Clin Lab Sci 45 (3):
copper deficiency. Neurology 61 (2): 273–274. 263–290.
Kumar S, Thapa B, Kaur G et al. (2006). Analysis of most com- Manolaki N, Nikolopoulou G, Daikos GL et al. (2009). Wilson
mon mutations R778G, R778L, R778W, I1102T and disease in children: analysis of 57 cases. J Pediatr
H1069Q in Indian Wilson disease patients: correlation Gastroenterol Nutr 48 (1): 72–77.
between genotype/phenotype/copper ATPase activity. Margarit E, Bach V, Gomez D et al. (2005). Mutation analysis
Mol Cell Biochem 294 (1–2): 1–10. of Wilson disease in the Spanish population - identification
Kumar SS, Kurian G, Eapen CE et al. (2012). Genetics of of a prevalent substitution and eight novel mutations in the
Wilson’s disease: a clinical perspective. Indian ATP7B gene. Clin Genet 68 (1): 61–68.
J Gastroenterol 31 (6): 285–293. Mendell JT, Sharifi NA, Meyers JL et al. (2004). Nonsense
Kuppala D, Deng J, Brewer GJ et al. (2009). Wilson disease surveillance regulates expression of diverse classes of
mutations in the American population: identification mammalian transcripts and mutes genomic noise. Nat
of five novel mutations in ATP7B. Open Hepatol J 1 (1): Genet 36 (10): 1073–1078.
1–4. Merle U, Weiss KH, Eisenbach C et al. (2010). Truncating
La Fontaine S, Theophilos MB, Firth SD et al. (2001). Effect of mutations in the Wilson disease gene ATP7B are associ-
the toxic milk mutation (tx) on the function and ated with very low serum ceruloplasmin oxidase activity
intracellular localization of Wnd, the murine homologue and an early onset of Wilson disease. BMC Gastroenterol
of the Wilson copper ATPase. Hum Mol Genet 10 (4): 10 (1): 8.
361–370. Møller LB, Horn N, Jeppesen TD et al. (2011). Clinical presen-
Lee CC, Wu JY, Tsai FJ et al. (2000). Molecular analysis of tation and mutations in Danish patients with Wilson dis-
Wilson disease in Taiwan: identification of one novel ease. Eur J Hum Genet 19 (9): 935–941.
THE GENETICS OF WILSON DISEASE 33
Nakayama K, Kubota M, Katoh Y et al. (2008). Early and pre- Ralle M, Huster D, Vogt S et al. (2010). Wilson disease at a
symptomatic detection of Wilson’s disease at the manda- single cell level: intracellular copper trafficking activates
tory 3-year-old medical health care examination in compartment-specific responses in hepatocytes. J Biol
Hokkaido Prefecture with the use of a novel automated uri- Chem 285 (40): 30875–30883.
nary ceruloplasmin assay. Mol Genet Metab 94 (3): Roberts EA, Schilsky MLAmerican Association for Study of
363–367. Liver Diseases (AASLD) (2008). Diagnosis and treatment
Nanji MS, Nguyen VT, Kawasoe JH et al. (1997). Haplotype of Wilson disease: an update. Hepatology (Baltimore, Md)
and mutation analysis in Japanese patients with Wilson dis- 47 (6): 2089–2111.
ease. Am J Hum Genet 60 (6): 1423–1429. Rodriguez-Granillo A, Sedlak E, Wittung-Stafshede P (2008).
Nicastro E, Ranucci G, Vajro P et al. (2010). Re-evaluation of Stability and ATP binding of the nucleotide-binding
the diagnostic criteria for Wilson disease in children with domain of the Wilson disease protein: effect of the common
mild liver disease. Hepatology (Baltimore, Md) 52 (6): H1069Q mutation. J Mol Biol 383 (5): 1097–1111.
1948–1956. Santhosh S, Shaji RV, Eapen CE et al. (2006). ATP7B muta-
Okada T, Shiono Y, Hayashi H et al. (2000). Mutational anal- tions in families in a predominantly Southern Indian cohort
ysis of ATP7B and genotype–phenotype correlation in of Wilson’s disease patients. Indian J Gastroenterol 25 (6):
Japanese with Wilson’s disease. Hum Mutat 15 (5): 277–282.
454–462. Sazinsky MH, Mandal AK, Arguello JM et al. (2006).
Okada T, Shinono Y, Kaneko Y et al. (2010). High prevalence Structure of the ATP binding domain from the
of fulminant hepatic failure among patients with mutant Archaeoglobus fulgidus Cu + -ATPase. J Biol Chem 281
alleles for truncation of ATP7B in Wilson’s disease. (16): 11161–11166.
Scand J Gastroenterol 45 (10): 1232–1237. Schilsky ML, Shneider B (2002). Population screening for
Owada M, Suzuki K, Fukushi M et al. (2002). Mass screening Wilson’s disease. J Pediatr 140 (5): 499–501.
for Wilson’s disease by measuring urinary holoceruloplas- Schushan M, Bhattacharjee A, Ben-Tai N et al. (2012).
min. J Pediatr 140 (5): 614–616. A structural model of the copper ATPase ATP7B to facilitate
Panagiotakaki E, Tzetis M, Manolaki N et al. (2004). analysis of Wilson disease-causing mutations and studies of
Genotype–phenotype correlations for a wide spectrum of the transport mechanism. Metallomics : Integrated Biometal
mutations in the Wilson disease gene (ATP7B). Am Science 4 (7): 669–678.
J Med Genet A 131A (2): 168–173. Shah AB, Chernov I, Zhang HT et al. (1997). Identification
Panichareon B, Taweechue K, Thongnoppakhun W et al. and analysis of mutations in the Wilson disease gene
(2011). Six novel ATP7B mutations in Thai patients with (ATP7B): population frequencies, genotype–phenotype
Wilson disease. Eur J Med Genet 54 (2): 103–107. correlation, and functional analyses. Am J Hum Genet
Paradisi I, De Freitas L, Arias S (2015). Most frequent muta- 61 (2): 317–328.
tion c.3402delC (p.Ala1135GlnfsX13) among Wilson dis- Shimizu N, Nakazono H, Takeshita Y et al. (1999). Molecular
ease patients in Venezuela has a wide distribution and two analysis and diagnosis in Japanese patients with Wilson’s
old origins. Eur J Med Genet 58 (2): 59–65. disease. Pediatr Int 41 (4): 409–413.
Park S, Park JY, Kim GH et al. (2007). Identification of novel Simon I (2008). Analysis of the human Atox 1 homologue in
ATP7Bgene mutations and their functional roles in Korean Wilson patients. World J Gastroenterol 14 (15): 2383.
patients with Wilson disease. Hum Mutat 28 (11): Simsek Papur O, Akman SA, Cakmur R et al. (2013). Mutation
1108–1113. analysis of ATP7B gene in Turkish Wilson disease
Park H, Park DK, Kim MS et al. (2015). Pseudo-dominant patients: identification of five novel mutations. Eur
inheritance in Wilson’s disease. Neurol Sci 37 (1): J Med Genet 56 (4): 175–179.
153–155. Song M-J, Lee ST, Lee MK et al. (2012). Estimation of carrier
Payne AS, Kelly EJ, Gitlin JD (1998). Functional expression of frequencies of six autosomal-recessive Mendelian disor-
the Wilson disease protein reveals mislocalization and ders in the Korean population. J Hum Genet 57 (2):
impaired copper-dependent trafficking of the common 139–144.
H1069Q mutation. Proc Natl Acad Sci U S A 95 (18): Stapelbroek JM, Bollen CW, van Amstel JK et al. (2004). The
10854–10859. H1069Q mutation in ATP7B is associated with late and
Perri RE, Hahn SH, Ferber MJ et al. (2005). Wilson disease – neurologic presentation in Wilson disease: results of a
keeping the bar for diagnosis raised. Hepatology meta-analysis. J Hepatol 41 (5): 758–763.
(Baltimore, Md) 42 (4): 974. St€attermayer AF, Rutter K, Beinhardt S et al. (2012). Genetic
Petrukhin K, Fischer SG, Pirastu M et al. (1993). Mapping, factors associated with histologic features of the liver and
cloning and genetic characterization of the region contain- treatment outcome in chronic hepatitis C patients.
ing the Wilson disease gene. Nat Genet 5 (4): 338–343. Z Gastroenterol 50 (05): P51.
Przybyłkowski A, Gromadzka G, Członkowska A (2014). Stenson PD, Ball EV, Mort M et al. (2012). The Human Gene
Polymorphisms of metal transporter genes DMT1 and Mutation Database (HGMD) and its exploitation in the
ATP7A in Wilson’s disease. J Trace Elem Med Biol 28 fields of personalized genomics and molecular evolution.
(1): 8–12. Curr Protoc Bioinformatics. Chapter 1: Unit 1.13.
34 I.J. CHANG AND S.H. HAHN
Stenson PD, Mort M, Ball EV et al. (2014). The Human Gene a key role in the transfer of copper from Atox1. J Biol
Mutation Database: building a comprehensive mutation Chem 279 (15): 15376–15384.
repository for clinical and molecular genetics, diagnostic Wan L, Tsai CH, Tsai Y et al. (2006). Mutation analysis of
testing and personalized genomic medicine. Hum Genet Taiwanese Wilson disease patients. Biochem Biophys
133 (1): 1–9. Res Commun 345 (2): 734–738.
Sternlieb I, Scheinberg IH (1968). Prevention of Wilson’s Wang L-H, Huang YQ, Shang X et al. (2011). Mutation anal-
disease in asymptomatic patients. New Engl J Med 278: ysis of 73 southern Chinese Wilson’s disease patients: iden-
352–359. tification of 10 novel mutations and its clinical correlation.
Takeshita Y, Shimizu N, Yamaguchi Y et al. (2002). Two fam- J Hum Genet 56 (9): 660–665.
ilies with Wilson disease in which siblings showed differ- Wei Z, Huang Y, Liu A et al. (2014). Mutational characteriza-
ent phenotypes. J Hum Genet 47 (10): 0543–0547. tion of ATP7B gene in 103 Wilson’s disease patients from
Taly AB, Meenakshi-Sundaram S, Sinha S et al. (2007). Southern China: identification of three novel mutations.
Wilson disease: description of 282 patients evaluated over Neuroreport 25 (14): 1075–1080.
3 decades. Medicine 86 (2): 112–121. Weiss KH (2006). Copper toxicosis gene MURR1 is not chan-
Tanzi RE, Petrukhin K, Chernov I et al. (1993). The Wilson ged in Wilson disease patients with normal blood cerulo-
disease gene is a copper transporting ATPase with homol- plasmin levels. World J Gastroenterol 12 (14): 2239.
ogy to the Menkes disease gene. Nat Genet 5 (4): 344–350. Weiss KH, Runz H, Noe B et al. (2010). Genetic analysis of
Tatsumi Y, Hattori A, Hayashi H et al. (2010). Current state of BIRC4/XIAP as a putative modifier gene of Wilson dis-
Wilson disease patients in central Japan. Intern Med ease. J Inherit Metab Dis 33 (3): 233–240.
(Tokyo, Japan) 49 (9): 809–815. Weitzman E, Pappo O, Weiss P et al. (2014). Late onset
Theophilos MB, Cox DW, Mercer JF (1996). The toxic milk fulminant Wilson’s disease: a case report and review of
mouse is a murine model of Wilson disease. Hum Mol the literature. World J Gastroenterol 20 (46): 17656–17660.
Genet 5 (10): 1619–1624. Whiteaker JR, Zhao L, Anderson L et al. (2010). An
Thomas GR, Jensson O, Gudmundsson G et al. (1995a). automated and multiplexed method for high throughput
Wilson disease in Iceland: a clinical and genetic study. peptide immunoaffinity enrichment and multiple reac-
Am J Hum Genet 56 (5): 1140–1146. tion monitoring mass spectrometry-based quantification
Thomas GR, Roberts EA, Walshe JM et al. (1995b). of protein biomarkers. Mol Cell Proteomics 9 (1):
Haplotypes and mutations in Wilson disease. Am J Hum 184–196.
Genet 56 (6): 1315–1319. Whiteaker JR, Lin C, Kennedy J et al. (2011). A targeted
Todorov T, Savov A, Jelev H et al. (2005). Spectrum of muta- proteomics-based pipeline for verification of biomarkers
tions in the Wilson disease gene (ATP7B) in the Bulgarian in plasma. Nat Biotechnol 29 (7): 625–634.
population. Clin Genet 68 (5): 474–476. Wilson JMG, Jungner G (1968). Principles and practice of
Tomic A, Dobricic V, Novakovic I et al. (2013). Mutational screening for disease. Public Health Paper no. 34, World
analysis of ATP7B gene and the genotype–phenotype cor- Health Organization, Geneva.
relation in patients with Wilson’s disease in Serbia. Wilson DC, Phillips MJ, Cox DW et al. (2000). Severe hepatic
Vojnosanit Pregl 70 (5): 457–462. Wilson’s disease in preschool-aged children. J Pediatr 137
Tsivkovskii R, Efremov RG, Lutsenko S (2003). The role of (5): 719–722.
the invariant His-1069 in folding and function of the Wu Z-Y, Lin MT, Murong SX et al. (2003). Molecular diag-
Wilson’s disease protein, the human copper-transporting nosis and prophylactic therapy for presymptomatic
ATPase ATP7B. J Biol Chem 278: 13302–13308. Chinese patients with Wilson disease. Arch Neurol 60
Usta J, Wehbeh A, Rida K et al. (2014). Phenotype–genotype (5): 737–741.
correlation in Wilson disease in a large Lebanese family: Wu F, Wang J, Pu C et al. (2015). Wilson’s disease: a compre-
association of c.2299insC with hepatic and of p. hensive review of the molecular mechanisms. Int J Mol Sci
Ala1003Thr with neurologic phenotype. PLoS One 9 16 (3): 6419–6431.
(11): e109727. Yamaguchi Y, Aoki T, Arashima S et al. (1999). Mass screen-
van den Berghe PVE, Stapelbroek JM, Krieger E et al. (2009). ing for Wilson’s disease: results and recommendations.
Reduced expression of ATP7B affected by Wilson Pediatr Int 41 (4): 405–408.
disease-causing mutations is rescued by pharmacological Yoo HW (2002). Identification of novel mutations and the
folding chaperones 4-phenylbutyrate and curcumin. three most common mutations in the human ATP7B gene
Hepatology (Baltimore, Md) 50 (6): 1783–1795. of Korean patients with Wilson disease. Genet Med 4
Vrabelova S, Letocha O, Borsky M et al. (2005). Mutation (6 Suppl): 43S–48S.
analysis of the ATP7B gene and genotype/phenotype cor- Zali N, Mohebbi SR, Estaghamat S et al. (2011). Prevalence of
relation in 227 patients with Wilson disease. Mol Genet ATP7B gene mutations in Iranian patients with Wilson dis-
Metab 86 (1–2): 277–285. ease. Hepatitis Monthly 11 (11): 890–894.
Walker JM, Huster D, Ralle M et al. (2004). The N-terminal
metal-binding site 2 of the Wilson’s disease protein plays
Handbook of Clinical Neurology, Vol. 142 (3rd series)
Wilson Disease
A. Członkowska and M.L. Schilsky, Editors
http://dx.doi.org/10.1016/B978-0-444-63625-6.00004-5
© 2017 Elsevier B.V. All rights reserved

Chapter 4

Genetic and environmental modifiers of Wilson disease


VALENTINA MEDICI1* AND KARL-HEINZ WEISS2
1
Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California Davis,
Sacramento, CA, USA
2
Department of Gastroenterology and Hepatology, University Hospital of Heidelberg, Heidelberg, Germany

Abstract
Wilson disease (WD) is characterized by remarkable variety in its phenotypic presentation. Patients with
WD can present with hepatic, neurologic, and psychiatric symptoms combined in different and unpredict-
able ways. Importantly, no convincing phenotype–genotype correlation has ever been identified, opening
the possibility that other genes, aside from ATPase copper-transporting beta (ATP7B), are involved in the
pathogenesis of this condition. In addition, modifier genes, or genes that can affect the expression of other
genes, may be involved. Clinical and basic science data indicate that environmental and dietary factors can
potentially modify gene expression in WD and, consequently, its clinical presentation and course. In par-
ticular, previously studied genes include copper metabolism domain-containing 1 (COMMD1), antioxi-
dant 1 copper chaperone (ATOX1), X-linked inhibitor of apoptosis (XIAP), apolipoprotein E (APOE),
hemochromatosis (HFE), and 5,10-methylenetetrahydrofolate reductase (MTHFR). Dietary factors
include iron and methyl group donors which could affect methionine metabolism and epigenetic mecha-
nisms of gene expression regulation. Most of the work conducted in this field is in its initial stages but it has
the potential to change the diagnosis and treatment of WD.

INTRODUCTION1 COMMD1 GENE


The major knowledge gap in Wilson disease (WD) is the An interesting candidate for a modifier gene is copper
lack of mechanistic understanding of the phenotype diver- metabolism domain-containing 1 (COMMD1). Defi-
sity and the responses to treatment despite the genetic ciency of COMMD1 causes canine copper toxicosis of
inheritance of ATPase copper-transporting beta (ATP7B) Bedlington terriers (Twedt et al., 1979; van de Sluis
mutations. Proteins other than ATP7B contribute to the et al., 2002; Klomp et al., 2003) that resembles WD,
molecular mechanism of copper excretion and mutations although ceruloplasmin levels are not decreased
or polymorphisms of these proteins might contribute to (Su et al., 1982) and there are no evident neurologic
WD, perhaps explaining the highly variable clinical pre- symptoms. The human orthologous gene was identified
sentation (Steindl et al., 1997; Riordan and Williams, on chromosome 2p13-16 and is distinct from the ATP7B
2001; Ala and Schilsky, 2011) and course of this disease. gene locus (van de Sluis et al., 1999).
This chapter will review the published data on concomi- As several studies confirmed a physical interaction of
tant genes and modifier genes as well as environmental COMMD1 with ATP7B (Tao et al., 2003; de Bie et al.,
factors that can have a role in WD phenotypic expression. 2006; Donadio et al., 2007), cohorts of WD patients were

1
Abbreviations used in the chapter are listed at the end of the chapter before References section.

*Correspondence to: Valentina Medici, MD, Department of Internal Medicine, Division of Gastroenterology and Hepatology,
University of California Davis, 4150 V St., Suite 3500, Sacramento CA 95817, USA. Tel: +1-916-734-3751, E-mail:
vmedici@ucdavis.edu
36 V. MEDICI AND K.-H. WEISS
screened for alterations in COMMD1. So far no exonic Correspondingly, reduced copper levels are found in
mutations of COMMD1 have been found in patients cells and tissues of XIAP-deficient knockout mice
with WD (Stuehler et al., 2004) or other rare human cop- (Burstein et al., 2004). Four nonsynonymous coding
per overload diseases like Indian childhood cirrhosis, single-nucleotide polymorphisms (SNPs) have previously
endemic Tyrolean infantile cirrhosis, or idiopathic cop- been described in the coding region of the XIAP gene
per toxicosis (Muller et al., 2003). Studies on known (Salzer et al., 2008; Serre et al., 2008); their physiologic
polymorphisms of COMMD1 in patients with WD role however remains unclear. In 98 WD patients
revealed conflicting results. In WD patients homozygous (Weiss et al., 2010), overall allele frequency did not differ
for the most common ATP7B mutation H1069Q, an significantly from previously reported panels. For all
association between a COMMD1 polymorphism and SNPs, statistical analysis did not reveal any correlation
onset of neurologic and hepatic symptoms was reported. to age of onset, clinical presentation (neurologic vs.
Onset of disease was significantly earlier for heterozy- hepatic vs. mixed vs. asymptomatic), or presentation as
gous patients at codon Asn 164 (GAT/GAC) than for fulminant WD (Weiss et al., 2010).
wild-type (GAT/GAT) (Stuehler et al., 2004). This could
only partially be confirmed by others (Weiss et al., 2006; APOE GENE
Bost et al., 2012).
Similarly to WD, several studies suggest that copper dys-
function and ATP7B variants influence the phenotypes of
ATOX1 GENE
neurodegenerative disorders such as Alzheimer’s disease
The human homologue antioxidant 1 copper chaperone (Squitti et al., 2013). The presence of apolipoprotein E
(ATOX1) is an 8-kDa cytosolic protein that contains a (APOE) allele ε4 is associated with an increased vulner-
single copy of the highly conserved MxCxxC motif in ability of the brain to disease effects, whereas the pres-
the amino-terminus which is repeated sixfold in ATP7B. ence of APOE genotype ε3/3 appears to provide
This metallochaperone interacts directly with ATP7B moderate neuroprotection. In line with this concept,
(Hamza et al., 1999) and can regulate its copper occu- Schiefermeier et al. (2000) reported that the homozygous
pancy (Walker et al., 2002). By modulating the amount APOE ε3 genotype is associated with delayed onset of
of copper bound to the protein, ATOX1 has an impact WD signs and symptoms. In an independent cohort,
on intracellular localization (DiDonato et al., 2000), as Litwin et al. (2012b) reported that women with APOE
well as posttranslational modification and enzymatic ε4-positive genotypes presented earlier onset of WD
activity of ATP7B as a copper-dependent transcription symptoms, particularly among ATP7B p.H1069Q homo-
factor involved in cell proliferation (Vanderwerf et al., zygous patients. However, data on an association
2001; Itoh et al., 2008). However, in human cohort stud- between APOE genotype and WD clinical expression
ies, no significant nonsynonymous coding variations in conflict with other authors who do not confirm these
the ATOX1 gene were evident (Simon et al., 2008; findings (Gu et al., 2005; Kocabay et al., 2009).
Bost et al., 2012). A common polymorphism within
ATOX1 (5’UTR -99 T > C) was detected at expected fre- HFE GENE AND METAL TRANSPORTER
quencies of 49–57%. Based on these data, no major role GENES DMT1 AND ATP7A
can be attributed to ATOX1 in the pathophysiology or
Data in patients and in animal models of WD indicate
clinical variation of WD.
iron accumulation may contribute to the phenotype of
this condition. HFE (hemochromatosis) gene mutations
XIAP GENE
are associated with hereditary hemochromatosis and are
XIAP (X-linked inhibitor of apoptosis protein), a related to increased intestinal iron uptake. Two initial
well-characterized antiapoptotic protein, has been pro- case reports described concomitant presence of HFE
posed as a regulator of copper-induced cell injury. XIAP and ATP7B gene polymorphisms with corresponding
binds copper directly, followed by enhanced degradation hepatic iron and copper accumulation (Hafkemeyer
and blunted inhibition of caspases. Therefore, in addition et al., 1994; Walshe and Cox, 1998). A study of
to copper overload, copper binding to XIAP sensitizes 32 WD patients from Sardinia described iron and copper
hepatocytes for apoptosis (Mufti et al., 2006), which sug- metabolism indices, HFE mutations, and liver biopsies
gests an unexpected new pathogenic mechanism for (Sorbello et al., 2010). Six of the studied patients were
copper-induced cell damage (Mufti et al., 2007). heterozygous for the H63D mutation; none presented
It has also been hypothesized that XIAP plays a role C282Y and S65C mutations. Patients with HFE poly-
in maintaining cellular copper homeostasis by interacting morphisms presented with approximately double the
with COMMD1 and promoting its degradation (Burstein hepatic iron concentration compared to patients without
et al., 2004; Prohaska, 2008; Maine et al., 2009). HFE polymorphisms. Long-term anticopper treatment,
GENETIC AND ENVIRONMENTAL MODIFIERS OF WILSON DISEASE 37
both zinc salts and chelation, was associated with hyperhomocysteinemia has not been described in WD,
improved alanine aminotransferase (ALT) levels and the possibility that homocysteine or aberrant methionine
decreased hepatic iron concentration in HFE wild-type metabolism can affect the phenotype is interesting, as
patients, whereas ALT levels and hepatic iron remained homocysteine can pass the blood–brain barrier and can
stable in patients with HFE polymorphism. HFE muta- also have neurotoxic effects by interacting with copper
tions were also studied in 40 patients with WD compared (White et al., 2001; Linnebank et al., 2006). In addition,
to 295 healthy controls and there was no difference in methionine metabolism is closely related to mechanisms
allele frequencies for the C282Y and H63D mutations of DNA and histone methylation with potential conse-
between the two populations (Erhardt et al., 2002). Sim- quences for gene expression regulation.
ilarly, a larger study on 143 WD patients explored HFE
allele frequencies and did not find any significant differ- HUMAN PRION GENE
ence between WD patients and the general population
(Pfeiffenberger et al., 2012). Prion protein (PRNP) binds copper ion with low affinity
The prevalence of divalent metal transporter 1 (Brown et al., 1997) and may affect copper metabolism,
(DMT1) and ATPase copper-transporting alpha (ATP7A) especially in the central nervous system, where this pro-
mutations was also studied in patients with WD tein is highly expressed (Ford et al., 2002). The interac-
(Przybyłkowski et al., 2014), as DMT1 is involved in tion between copper and PRNP may have a protective
iron transport and ATP7A is involved in copper transport effect on neurons (Gasperini et al., 2015). Given this
in the intestinal epithelium. The study, which included background, Merle et al. (2006) studied the prevalence
more than 100 patients with WD, found an increased fre- of the PRNP polymorphism at codon 129 (M129V) asso-
quency of the DMT1 IVS4 C(+) allele in patients with ciated with age at disease onset and subsequent disease
WD compared to healthy controls, but no differences course in 134 patients with WD. The prevalence of the
according to hepatic or neurologic phenotype. ATP7A M129V genotype was similar in WD patients compared
alleles did not present different prevalence between the to a healthy control population. With respect to their clin-
two studied populations. The rationale for studying these ical presentation, patients with PRNP codon 129 result-
genes resides in two previous findings: increased tran- ing in homozygous methionine (129 M/M) were about
script levels of DMT1 in the brains of patients with 5 years older at disease onset and had neurologic presen-
Parkinson’s disease and in animal models of this condi- tation an average of 7 years later compared to carriers of
tion (Salazar et al., 2008), and increased mRNA tran- PRNP 129 V(+).
script and protein levels of duodenal ATP7A as a
possible compensatory mechanism after copper accumu- GENDER
lation (Przybyłkowski et al., 2013). As pointed out by a study from Litwin et al. (2012a), neu-
ropsychiatric presentation in WD is more common in
MTHFR GENE men than women and women develop these symptoms
at a later age than men. In a complex study on more than
A key enzyme in folate and methionine me- 1000 WD patients, Ferenci (2014) reported that gender
tabolism is 5,10-methylenetetrahydrofolate reductase and age modify disease presentation specifically. Young
(MTHFR), which catalyzes the conversion of 5,10- females present more often with fulminant liver disease
methylenetetrahydrofolate to 5-methyltetrahydrofolate, (Ferenci et al., 2011) and males present more frequently
a co-substrate for homocysteine remethylation to with neurologic disease, whereas neurologic disease is
methionine. Mutations of MTHFR are associated with rare among children with WD (Ferenci, 2014). These
increased homocysteine levels, which may contribute findings may be related to hormonal changes but also
to greater severity of WD or to its phenotypic variability. to differences in iron accumulation and metabolism
Two-hundred and forty-five patients with WD were gen- (by menstrual iron loss), which are discussed below.
otyped for the two MTHFR polymorphisms, C677T and
A1298C, and genotype–phenotype correlations were
IRON
studied. The C667T genotype was more frequent than
expected according to Hardy–Weinberg equilibrium. Studies in animal models and in humans show evidence
The C677T allele was associated more frequently with that copper accumulation and low ceruloplasmin levels
hepatic onset. The A1298C allele was associated with as well as long-term treatment of WD are associated with
younger age at presentation. MTHFR polymorphisms iron accumulation. There is a close relation between iron
were not associated with any difference in copper metab- and copper accumulation primarily derived from the fact
olism (Gromadzka et al., 2011). Even though these initial that ceruloplasmin is a potent ferroxidase, catalyzing the
observations were not explored in other populations and conversion of Fe2+ to Fe3+, and this activity is required
38 V. MEDICI AND K.-H. WEISS
for iron cellular uptake (Attieh et al., 1999). Low cerulo- SAH is an inhibitor of almost all methylation reactions.
plasmin levels as presented in WD may thus result in iron SAHH is a bidirectional enzyme that regulates the pro-
accumulation. The other mechanism possibly underlying duction of homocysteine in the forward direction, while
iron accumulation in WD is related to long-term anticop- excess homocysteine promotes SAH in the reverse direc-
per treatment or overtreatment, probably associated with tion. Thus, SAHH may play a critical role in the regula-
reduced copper bioavailability, worsening hypocerulo- tion of methylation pathways; its inhibition could result
plasminemia, and, ultimately, iron overload (Schilsky, in an increase of its substrate SAH, with resultant inhibi-
2001). In a genetic animal model of WD, the Long– tion of gene methylation reactions, thereby potentially
Evans Cinnamon (LEC) rats, iron deprivation or phlebot- regulating the expression of genes involved in liver
omies prevented the development of acute liver failure injury. Dietary methyl groups are provided by either
and hepatocellular carcinoma, despite absence of hepatic betaine or choline which are abundant in spinach, beet,
copper treatment (Kato et al., 1996). In WD patients trea- cereals or eggs, liver, almonds, and broccoli.
ted with penicillamine for 3–8.5 years, there was evi- Copper is a major regulator of methionine metabolism
dence of hepatic iron accumulation and reduced through its effect on SAHH (Bethin et al., 1995). Copper
hepatic copper content over time. Phlebotomies in 2 inhibits SAHH in a noncompetitive manner since its
patients achieved lower serum ferritin and ALT levels binding to this enzyme results in the release of NAD+
(Shiono et al., 2001). Iron parameters were studied for cofactors (Li et al., 2007). Sahh hepatic transcript levels
a group of 40 patients with WD compared to healthy con- are downregulated in a murine model of WD character-
trol subjects. WD patients presented higher serum ferritin ized by spontaneous hepatic copper accumulation
levels than controls and a subgroup of WD patients with (Medici et al., 2013; Le et al., 2014). In addition, mater-
low ceruloplasmin presented higher ferritin levels than nal provision of methyl groups in the form of choline
those with normal ceruloplasmin (Erhardt et al., 2002). was able to increase Sahh transcripts to control levels,
A larger study of hepatic iron concentrations on indicating maternal diet can have an effect on gene
follow-up liver biopsies in patients with WD confirmed expression and epigenetic mechanisms of phenotype reg-
penicillamine treatment was associated with 3–10 times ulation in WD (Medici et al. 2014).
increased hepatic iron content compared to baseline,
whereas zinc treatment was not associated with signifi- OTHER DIETARY FACTORS
cant increase in hepatic iron concentration (Medici
Another study on the LEC rat examined the effects of die-
et al., 2007). However, iron studies were conducted on
tary fatty acids on peroxidative stress and histologic dam-
a subsequent larger study of 143 patients with WD
age. Polyunsaturated fatty acids, both n-6 and n-3 type,
(Pfeiffenberger et al., 2012). Interestingly, serum ferritin
were associated with fewer histologic features of hepati-
was lower in males with WD who also presented lower
tis and lower liver enzymes. Cyclooxygenase-2 hepatic
ceruloplasmin levels. Only 3 out of 27 patients who
transcript levels, a marker of inflammation, were down-
underwent liver biopsy presented hepatic iron concentra-
regulated in the group of rats receiving polyunsaturated
tion higher than the upper limit, all of them on chelating
fatty acids (Du et al., 2004).
agents at the time of the biopsy. Collectively, the direct
Another study explored the effects of mild zinc defi-
and indirect evidence points to the fact that iron may con-
ciency on the same animal model and the major finding
tribute to the phenotype of WD and monitoring of iron
was that LEC rats exposed to a zinc-deficient diet for
metabolism parameters should be considered in the
4 weeks developed acute liver failure at a younger age
long-term management of these patients.
compared to control rats (94 vs. 136 days of age)
(Saito et al., 2007). A diet rich in histidine was associated
METHYL GROUPS with reduced hepatic copper levels by 47% and increased
urinary copper excretion in LEC rats, with correspond-
Hepatic methionine metabolism regulates DNA and
ing reduced activity of the antioxidant enzyme Cu,
histone methylation reactions with potential conse-
Zn-superoxide dismutase (Xu et al., 2003). LEC rats
quences on gene expression regulation and disease
fed a soy protein-rich diet starting at 6 weeks of age pre-
presentation and course. The central players in methio-
sented 80% higher hepatic copper concentration and
nine metabolism are S-adenosylmethionine (SAM),
reduced survival compared to control mice (Yonezawa
S-adenosylhomocysteine (SAH), and the bidirectional
et al., 2003).
SAH hydrolase (SAHH). SAM is irreversibly converted
to SAH by donating its methyl moiety to DNA methyl-
SUMMARYAND CONCLUSION
transferases. The SAM-to-SAH ratio is considered a
useful index of methylation capacity, as SAM is the uni- It is becoming evident from clinical and basic science
versal methyl donor for methyltransferase reactions and research that the phenotype of WD is affected by
GENETIC AND ENVIRONMENTAL MODIFIERS OF WILSON DISEASE 39
genetic, epigenetic, and environmental factors. This can Du C, Fujii Y, Ito M et al. (2004). Dietary polyunsaturated
have implications for the neonatal screening, early diag- fatty acids suppress acute hepatitis, alter gene expression
nosis, and treatment of this condition. The fact that and prolong survival of female Long–Evans Cinnamon
maternal diet can affect the onset and progression of rats, a model of Wilson disease. J Nutr Biochem 15:
273–280.
disease supports the indication of neonatal screening
Erhardt A, Hoffmann A, Hefter H et al. (2002). HFE gene
of WD. In addition, current anticopper treatments could
mutations and iron metabolism in Wilson’s disease.
be optimized by modifying dietary factors and could be Liver 22: 474–478.
tailored according to the presence of certain mutations Ferenci P (2014). Phenotype–genotype correlations in
and gene modifiers, ultimately improving disease patients with Wilson’s disease. Ann N Y Acad Sci
course and prognosis. 1315: 1–5.
Ferenci P, Weiss KH, Członkowska A et al. (2011). Gender
influences the clinical presentation of Wilson disease
ABBREVIATIONS (WD). Gastroenterology 140: S939.
APOE, apolipoprotein E; ATOX1, antioxidant 1 Ford MJ, Burton LJ, Morris RJ et al. (2002). Selective expres-
sion of prion protein in peripheral tissues of the adult
copper chaperone; ATP7A, ATPase copper-transporting
mouse. Neuroscience 113: 177–192.
alpha; ATP7B, ATPase copper-transporting beta;
Gasperini L, Meneghetti E, Pastore B et al. (2015). Prion pro-
COMMD1, copper metabolism domain-containing 1; tein and copper cooperatively protect neurons by modulat-
DMT1, divalent metal transporter 1; HFE, hemochromato- ing NMDA receptor through S-nitrosylation. Antioxid
sis; LEC, Long–Evans Cinnamon; MTHFR, 5,10- Redox Signal 22: 772–784.
methylenetetrahydrofolate reductase; PRNP, prion protein; Gromadzka G, Rudnicka M, Chabik G et al. (2011). Genetic
SAH, S-adenosylhomocysteine; SAHH, S-adenosyl- variability in the methylenetetrahydrofolate reductase gene
homocysteine hydrolase; SAM, S-adenosylmethionine; (MTHFR) affects clinical expression of Wilson’s disease.
WD, Wilson disease. J Hepatol 55: 913–919.
Gu YH, Kodama H, Du SL (2005). Apolipoprotein E genotype
REFERENCES analysis in Chinese Han ethnic children with Wilson’s dis-
ease, with a concentration on those homozygous for
Ala A, Schilsky M (2011). Genetic modifiers of liver injury in R778L. Brain Dev 27: 551–553.
hereditary liver disease. Semin Liver Dis 31: 208–214. Hafkemeyer P, Schupp M, Storch M et al. (1994). Excessive
Attieh ZK, Mukhopadhyay CK, Seshadri V et al. (1999). iron storage in a patient with Wilson’s disease. Clin
Ceruloplasmin ferroxidase activity stimulates cellular iron Investig 72: 134–136.
uptake by a trivalent cation-specific transport mechanism. Hamza I, Schaefer M, Klomp LW et al. (1999). Interaction of
J Biol Chem 274: 1116–1123. the copper chaperone HAH1 with the Wilson disease pro-
Bethin KE, Cimato TR, Ettinger MJ (1995). Copper binding to tein is essential for copper homeostasis. Proc Natl Acad Sci
mouse liver S-adenosylhomocysteine hydrolase and the U S A 96: 13363–13368.
effects of copper on its levels. J Biol Chem 270: Itoh S, Kim HW, Nakagawa O et al. (2008). Novel role of
20703–20711. antioxidant-1 (Atox1) as a copper-dependent transcription
Bost M, Piguet-Lacroix G, Parant F et al. (2012). Molecular factor involved in cell proliferation. J Biol Chem 283:
analysis of Wilson patients: direct sequencing and MLPA 9157–9167.
analysis in the ATP7B gene and Atox1 and COMMD1 gene Kato J, Kobune M, Kohgo Y et al. (1996). Hepatic iron depri-
analysis. J Trace Elem Med Biol 26: 97–101. vation prevents spontaneous development of fulminant
Brown DR, Qin K, Herms JW et al. (1997). The cellular hepatitis and liver cancer in Long–Evans Cinnamon rats.
prion protein binds copper in vivo. Nature 18–25; 390: J Clin Invest 98: 923–929.
684–687. Klomp AE, van de Sluis B, Klomp LW et al. (2003). The ubiq-
Burstein E, Ganesh L, Dick RD et al. (2004). A novel role for uitously expressed MURR1 protein is absent in canine cop-
XIAP in copper homeostasis through regulation of per toxicosis. J Hepatol 39: 703–709.
MURR1. Embo J 23: 244–254. Kocabay G, Tutuncu Y, Yilmaz H et al. (2009). Impact of apo-
de Bie P, Burstein E, van de Sluis B et al. (2006). Several lipoprotein E genotypes on phenotypic expression in Turkish
COMMD proteins interact with ATP7B; possible candidate patients with Wilson’s disease. Scand J Gastroenterol 44:
genes for hepatic copper overload disorders with unknown 1270–1271.
etiology. Eur J Gastroenterol Hepatol 18: A52. Le A, Shibata NM, French SW et al. (2014). Characterization
DiDonato M, Hsu HF, Narindrasorasak S et al. (2000). of timed changes in hepatic copper concentrations, methi-
Copper-induced conformational changes in the N-terminal onine metabolism, gene expression, and global DNA meth-
domain of the Wilson disease copper-transporting ylation in the Jackson Toxic Milk mouse model of Wilson
ATPase. Biochemistry 39: 1890–1896. disease. Int J Mol Sci 15: 8004–8023.
Donadio S, De Keukeleire J, Micoud J et al. (2007). Cellular Li M, Li Y, Chen J et al. (2007). Copper ions inhibit
localization and trafficking of endogenous ATP7B and S-adenosylhomocysteine hydrolase by causing dissocia-
COMMD1 proteins. Febs J 274: 106. tion of NAD + cofactor. Biochemistry 46: 11451–11458.
Another random document with
no related content on Scribd:
1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or expense
to the user, provide a copy, a means of exporting a copy, or a means
of obtaining a copy upon request, of the work in its original “Plain
Vanilla ASCII” or other form. Any alternate format must include the
full Project Gutenberg™ License as specified in paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™ works
unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or providing


access to or distributing Project Gutenberg™ electronic works
provided that:

• You pay a royalty fee of 20% of the gross profits you derive from
the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”

• You provide a full refund of any money paid by a user who


notifies you in writing (or by e-mail) within 30 days of receipt that
s/he does not agree to the terms of the full Project Gutenberg™
License. You must require such a user to return or destroy all
copies of the works possessed in a physical medium and
discontinue all use of and all access to other copies of Project
Gutenberg™ works.

• You provide, in accordance with paragraph 1.F.3, a full refund of


any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.

• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.

1.E.9. If you wish to charge a fee or distribute a Project Gutenberg™


electronic work or group of works on different terms than are set
forth in this agreement, you must obtain permission in writing from
the Project Gutenberg Literary Archive Foundation, the manager of
the Project Gutenberg™ trademark. Contact the Foundation as set
forth in Section 3 below.

1.F.

1.F.1. Project Gutenberg volunteers and employees expend


considerable effort to identify, do copyright research on, transcribe
and proofread works not protected by U.S. copyright law in creating
the Project Gutenberg™ collection. Despite these efforts, Project
Gutenberg™ electronic works, and the medium on which they may
be stored, may contain “Defects,” such as, but not limited to,
incomplete, inaccurate or corrupt data, transcription errors, a
copyright or other intellectual property infringement, a defective or
damaged disk or other medium, a computer virus, or computer
codes that damage or cannot be read by your equipment.

1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES - Except


for the “Right of Replacement or Refund” described in paragraph
1.F.3, the Project Gutenberg Literary Archive Foundation, the owner
of the Project Gutenberg™ trademark, and any other party
distributing a Project Gutenberg™ electronic work under this
agreement, disclaim all liability to you for damages, costs and
expenses, including legal fees. YOU AGREE THAT YOU HAVE NO
REMEDIES FOR NEGLIGENCE, STRICT LIABILITY, BREACH OF
WARRANTY OR BREACH OF CONTRACT EXCEPT THOSE
PROVIDED IN PARAGRAPH 1.F.3. YOU AGREE THAT THE
FOUNDATION, THE TRADEMARK OWNER, AND ANY
DISTRIBUTOR UNDER THIS AGREEMENT WILL NOT BE LIABLE
TO YOU FOR ACTUAL, DIRECT, INDIRECT, CONSEQUENTIAL,
PUNITIVE OR INCIDENTAL DAMAGES EVEN IF YOU GIVE
NOTICE OF THE POSSIBILITY OF SUCH DAMAGE.

1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If you


discover a defect in this electronic work within 90 days of receiving it,
you can receive a refund of the money (if any) you paid for it by
sending a written explanation to the person you received the work
from. If you received the work on a physical medium, you must
return the medium with your written explanation. The person or entity
that provided you with the defective work may elect to provide a
replacement copy in lieu of a refund. If you received the work
electronically, the person or entity providing it to you may choose to
give you a second opportunity to receive the work electronically in
lieu of a refund. If the second copy is also defective, you may
demand a refund in writing without further opportunities to fix the
problem.

1.F.4. Except for the limited right of replacement or refund set forth in
paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.

1.F.5. Some states do not allow disclaimers of certain implied


warranties or the exclusion or limitation of certain types of damages.
If any disclaimer or limitation set forth in this agreement violates the
law of the state applicable to this agreement, the agreement shall be
interpreted to make the maximum disclaimer or limitation permitted
by the applicable state law. The invalidity or unenforceability of any
provision of this agreement shall not void the remaining provisions.
1.F.6. INDEMNITY - You agree to indemnify and hold the
Foundation, the trademark owner, any agent or employee of the
Foundation, anyone providing copies of Project Gutenberg™
electronic works in accordance with this agreement, and any
volunteers associated with the production, promotion and distribution
of Project Gutenberg™ electronic works, harmless from all liability,
costs and expenses, including legal fees, that arise directly or
indirectly from any of the following which you do or cause to occur:
(a) distribution of this or any Project Gutenberg™ work, (b)
alteration, modification, or additions or deletions to any Project
Gutenberg™ work, and (c) any Defect you cause.

Section 2. Information about the Mission of


Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new computers.
It exists because of the efforts of hundreds of volunteers and
donations from people in all walks of life.

Volunteers and financial support to provide volunteers with the


assistance they need are critical to reaching Project Gutenberg™’s
goals and ensuring that the Project Gutenberg™ collection will
remain freely available for generations to come. In 2001, the Project
Gutenberg Literary Archive Foundation was created to provide a
secure and permanent future for Project Gutenberg™ and future
generations. To learn more about the Project Gutenberg Literary
Archive Foundation and how your efforts and donations can help,
see Sections 3 and 4 and the Foundation information page at
www.gutenberg.org.

Section 3. Information about the Project


Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-profit
501(c)(3) educational corporation organized under the laws of the
state of Mississippi and granted tax exempt status by the Internal
Revenue Service. The Foundation’s EIN or federal tax identification
number is 64-6221541. Contributions to the Project Gutenberg
Literary Archive Foundation are tax deductible to the full extent
permitted by U.S. federal laws and your state’s laws.

The Foundation’s business office is located at 809 North 1500 West,


Salt Lake City, UT 84116, (801) 596-1887. Email contact links and up
to date contact information can be found at the Foundation’s website
and official page at www.gutenberg.org/contact

Section 4. Information about Donations to


the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission of
increasing the number of public domain and licensed works that can
be freely distributed in machine-readable form accessible by the
widest array of equipment including outdated equipment. Many small
donations ($1 to $5,000) are particularly important to maintaining tax
exempt status with the IRS.

The Foundation is committed to complying with the laws regulating


charities and charitable donations in all 50 states of the United
States. Compliance requirements are not uniform and it takes a
considerable effort, much paperwork and many fees to meet and
keep up with these requirements. We do not solicit donations in
locations where we have not received written confirmation of
compliance. To SEND DONATIONS or determine the status of
compliance for any particular state visit www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states where


we have not met the solicitation requirements, we know of no
prohibition against accepting unsolicited donations from donors in
such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot make


any statements concerning tax treatment of donations received from
outside the United States. U.S. laws alone swamp our small staff.

Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.

Section 5. General Information About Project


Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could be
freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose network of
volunteer support.

Project Gutenberg™ eBooks are often created from several printed


editions, all of which are confirmed as not protected by copyright in
the U.S. unless a copyright notice is included. Thus, we do not
necessarily keep eBooks in compliance with any particular paper
edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg Literary
Archive Foundation, how to help produce our new eBooks, and how
to subscribe to our email newsletter to hear about new eBooks.

You might also like