Fanconi Anemia - D. Schindler, Et. Al., (Karger, 2007) WW
Fanconi Anemia - D. Schindler, Et. Al., (Karger, 2007) WW
Series Editor
Volume Editors
Bibliographic Indices. This publication is listed in bibliographic services, including Current Contents® and
Index Medicus.
Disclaimer. The statements, options and data contained in this publication are solely those of the individ-
ual authors and contributors and not of the publisher and the editor(s). The appearance of advertisements in the
book is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness,
quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property
resulting from any ideas, methods, instructions or products referred to in the content or advertisements.
Drug Dosage. The authors and the publisher have exerted every effort to ensure that drug selection and
dosage set forth in this text are in accord with current recommendations and practice at the time of publication.
However, in view of ongoing research, changes in government regulations, and the constant flow of information
relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for
any change in indications and dosage and for added warnings and precautions. This is particularly important when
the recommended agent is a new and/or infrequently employed drug.
All rights reserved. No part of this publication may be translated into other languages, reproduced or
utilized in any form or by any means electronic or mechanical, including photocopying, recording, microcopying,
or by any information storage and retrieval system, without permission in writing from the publisher.
IX Editorial
Schmid, M. (Würzburg)
XI Preface
Schindler, D.; Hoehn, H. (Würzburg)
Introductory Remarks
VI
95 Interphase FISH-Assay for the Detection of MDS- and
AML-Associated Chromosomal Imbalances in Native
Bone Marrow and Peripheral Blood Cells
Tönnies, H.; Huber, S.; Volarikova, E.; Gerlach, A.; Neitzel, H. (Berlin)
Contents VII
Editorial
IX
depth treatment of a given subject that can hardly be achieved with the usual
publish-or-perish types of publications. As such, monographs fulfill the impor-
tant task to remind the reader that any type of scientific progress has its roots in
a multifaceted landscape of prior insights and achievements that need to be col-
lected and preserved in order to provide fertile ground for future growth.
I would like to thank all the authors for their interesting contributions, the
Editors Holger Hoehn and Detlev Schindler for their invaluable support and
help in the organization of this book, and the Publisher Thomas Karger for hav-
ing offered the opportunity to reinitiate this book series.
Michael Schmid
Würzburg, January 2007
Editorial X
Preface
XI
types of DNA repair in FA, and with the establishment of a test system for the
Rad51 recombinase in homology-directed DNA repair. Even though it is
impossible to cover all aspects of Fanconi anemia within the space available for
such a volume, we hope that it may serve as useful introduction to a disease that
provides unique insights into the complex network of genomic maintenance
systems which protect us from cancer and premature aging.
We dedicate this volume to George M. Martin, M.D., Professor Emeritus
of Pathology and Genetics at the University of Washington, Seattle, USA, on
the occasion of his 80th birthday. Like Traute Schroeder-Kurth with respect to
Fanconi anemia, George Martin was one of the first scientists to recognize the
intimate relationship between genetic instability, cancer and aging by studying
the Werner progeria syndrome. Very early on he was convinced of the model
character and unique value of the rare human chromosomal breakage syn-
dromes. He emphasized that these rare experiments of nature would provide
unprecedented insights into the complex mechanisms of DNA damage recogni-
tion and repair which are essential for long lived, warm-blooded species such as
ours. George Martin encouraged us to study the human caretaker gene syn-
dromes, including Fanconi anemia, and we owe him much in terms of motiva-
tion, mentorship, and guidance.
Detlev Schindler
Holger Hoehn
Würzburg, January 2007
Preface XII
Introductory Remarks
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 1–8
Abstract
In a field that embraces multiple aspects of both clinical and basic research and that
moves impressively fast, any answers to the questions why, what and how can we learn from
a rare disease like Fanconi anemia (FA) must remain tentative and preliminary. However,
there are very encouraging advances, most notably at the level of understanding the molecu-
lar basis of FA and at the level of treatment via hematopoietic stem cell transplantation. For
the sake of our patients we clearly need to arrive at meaningful genotype-phenotype correla-
tions and individualized risk profiles. This requires prospective and longterm studies carried
out in close cooperation among patients, clinicians and basic scientists. There are a number
of open questions, for example relating to the mechanisms of chromosomal breakage and
DNA repair, to the spectrum of genetic changes that herald and promote the emergence of
leukaemia and solid tumors, and to the emergence of genetically reverted cells in blood and
bone marrow of FA patients. Researchers in the fields of cancer and aging should be encour-
aged to and are likely to benefit from the study of Fanconi anemia, as are our patients from
the welcome results of such studies.
Copyright © 2007 S. Karger AG, Basel
The year 2007 marks the 80th anniversary of the original description by
Guido Fanconi of ‘Familial infantile pernicious-like anemia’, a rare genetic dis-
ease that since carries the name of this eminent physician-scientist [1]. Why
should we study such a rare disease, what can we learn from its myriad clinical
and molecular manifestations, and how should we go about it? I will try to give
some brief answers to these questions even though by necessity these answers
are tentative and preliminary. Despite undoubted progress, there still are many
aspects of this enigmatic disease that we do not fully understand.
Why Study Fanconi Anemia?
First of all, for the sake of the affected patients. Even though there is no
definite cure for FA on the horizon, there is encouraging progress regarding
diagnosis and treatment. Timely diagnosis is crucial both for the institution of
adequate treatment and for the prevention of inadequate medical management
that may result from failure to recognize the hereditary and specific nature of
the disease. If one looks back at the last monograph on Fanconi anemia that I
had the pleasure to edit with my colleagues Arleen Auerbach and Gunter Obe in
the year 1989 [2], the progress that has since been made is impressive and
encouraging. Two major areas stand out: molecular genetics, and hematopoietic
stem cell transplantation. Progress in the understanding of the molecular basis
of Fanconi anemia is astounding indeed: even though genetic heterogeneity of
FA had been convincingly documented prior to 1989 [3, 4], it was not until
1992 that the first FA gene had been cloned and, as of this writing, 12 FA caus-
ing genes have been identified. Several more are in the offing, as there are a
number of patients who apparently do not belong to any of the known comple-
mentation groups [5]. There is also increasing understanding of how the FA
proteins work, how they work together and how they might interact with other
proteins in maintaining a stable genome [6, 7].
Although less spectacular than the gene discovery itself, the virtual explo-
sion of knowledge concerning the molecular basis of Fanconi anemia during the
past 10–15 years translates into immediate benefits to FA patients and their fami-
lies. For example, precise knowledge of the affected gene and the specific type of
mutation in a given patient will increasingly be of prognostic value and serve as a
rational guide for optimized medical management. We clearly need to collect
many more data in order to arrive at clinically useful genotype-phenotype corre-
lations. We should strongly oppose arguments that deem the collection of such
additional data unnecessary once the clinical diagnosis of FA has been established
and confirmed by a chromosome breakage or cell cycle test. The ultimate goal of
such efforts is to arrive at individualized risk profiles for FA patients similar to
what has already been achieved with congenital abnormality scores [8]. Biallelic
mutations in two of the FA genes (FANCD1/BRCA2 and FANCN/PALB2) have so
far been associated with very early childhood cancers [9, 10], and awareness of
such correlations are of undisputed value in the clinical management of these
young tumor patients. Precise knowledge of the disease causing mutations also
improves and facilitates prenatal diagnosis that remains an unwelcome but com-
promise option for some of the FA families. Last but not least, participation in
gene therapy trials requires knowledge of underlying genes and mutations.
Although one may question the extent to which the vastly improved
molecular knowledge has so far been of immediate benefit to the majority of
Schroeder-Kurth 2
FA patients, such reservations do least apply to the progress in the field of
hematopoietic stem cell transplantation (HSCT). The outcomes of matching
donor sibling transplantations have continuously improved such that today early
HSCT is highly recommended in such families [11]. Unrelated donor trans-
plantation still carries a higher risk, but modified conditioning protocols using
fludarabine and T-cell depletion, reduction or complete absence of whole body
irradiation, the introduction of minitransplants and improved post-transplant
care have contributed to much better longterm survival [12]. The serious prob-
lem of posttransplant squamous cell carcinomas remains to be solved [13]. The
message from all this progress is to consider the option of HSCT in each patient
at a much earlier stage of the disease and with much more optimism than we
could have had in the year 1989.
Why, What and How Can We Learn from a Rare Disease Like Fanconi Anemia? 3
non-allelic homologous recombination? [20]. Does the chromosomal position
within the nucleus influence the opportunity for the type of radial formation?
There are many unsolved questions, there still is much to be learned.
One of the most impressive lessons we learned from patients with Fanconi
anemia is the intimate relationship between genetic instability and cancer. FA
patients carry a high risk for acute leukaemia, squamous cell carcinomas and
other tumors [21]. Young FA patients with certain gene mutations have a higher
risk to develop leukaemia. The older a FA patient gets the higher is the risk to
develop a solid tumor. There are impressive case histories describing adult
patients in whom a leukaemia or a solid tumor were the first manifestations of
FA [22, 23]. With the exception of FANCF that is frequently inactivated by an
epigenetic mechanism in various types of tumors, the other FA genes seem to be
relatively intact in most neoplasias as if they were needed for tumor cell growth
[24]. However, monoallelic truncating mutations in at least three of the FA genes
(FANCD1/BRCA2, FANCJ/BRIP1, and FANCN/PALB2) have been shown to
confer variant degrees of breast cancer susceptibility in non-FA patients [25, 26].
Those are important new insights, connecting a rare with a common disease.
In 1971 we postulated that in vivo chromosomal instability leads to multi-
ple somatic mutations which finally may confer a selective advantage to a sin-
gle mutated cell, giving rise to malignant cell growth [27]. By careful bone
marrow studies, this process has been directly observed in FA patients. Specific
chromosomal changes in bone marrow cells appear to herald progression to
malignancy even prior to any clinical manifestation of leukaemia [28]. It was
quite clear in the 1970s that aberrant clones may also appear in peripheral blood
lymphocyte cultures. Over time, these cytogenetically aberrant clones were
subject to clonal attenuation and clonal succession, but they undoubtedly were
bad news for the patient. One of these patients I followed for more than six
years with repeated chromosome studies [29]. He died at age 35 of bronchial
cancer (unpublished). These types of longterm observations are important for
our patients and should be supported as much as possible. There is much to be
learned about the close relationship between FA and cancer for the sake of our
patients, and for our understanding of the origin of cancer [30].
Even though a fundamental change of the high cancer risk in FA is not in
sight, we have already learned that predictive and preventive measures are benefi-
cial for our patients. As already mentioned, regular monitoring for aberrant bone
marrow clones, particularly those involving monosomy 7 and 3q duplications, is
of great value for both physicians and patient in their difficult decision as to
whether and when to proceed with HSCT [28]. Likewise, non-invasive measures
for the early detection of oral cavity and genital area lesions as currently devel-
oped by the group of Ruud Brakenhoff in Amsterdam [31] will gain even greater
importance with more and more patients undergoing and surviving HSCT.
Schroeder-Kurth 4
Another lesson we are beginning to appreciate from Fanconi anemia is the
discovery of reverse mutations in blood cells [32]. One in four or five patients
displays MMC resistant cells among the original MMC sensitive cells. These
patients are mosaic FA patients who have a chance to escape BM failure and,
possibly, the development of leukaemia. Much research has to be done to clar-
ify the underlying mechanisms. The likelihood of ‘natural gene therapy’ leading
to revertant mosaicism appears to be higher in patients belonging to certain
complementation groups and in the presence of compound heterozygosity.
Longterm observations of these mosaic FA patients will ultimately teach us
whether there is more than a temporary benefit of this type of ‘natural gene
therapy’ to the individual, and what the prerequisites are for somatic reversions
to occur. And, of course, could there be ways to enhance the occurrence of such
reversions for the benefit of our patients?
First and foremost, FA research needs the cooperation of the affected fam-
ilies and patients as well as the unrelenting interest and commitment of their
doctors. A well established (and well funded!) FA registry such as pioneered by
Arleen Auerbach in New York is essential for both clinical care and basic
research [33, 34]. It should include regular follow-ups with complete informa-
tion on the natural history of the disease in each individual family. For practical
purposes, such registries should be organized at the regional or national levels,
but they should and must communicate with each other. Today we have every
reason to believe that most FA families are aware of the importance of FA
research for the sake of their children and thus are willing to cooperate with
such registries. Many research projects require the participation and involve-
ment of the patients themselves, and there is little doubt that the affected
patients and families can still teach us a lot about their disease. On the technical
side, new tools such as retroviral complementation, knockdown of FA genes via
RNA interference, or MLPA for the detection of large deletions have con-
tributed both to diagnosis and research. With such technical advances at hand
the time might have come to tackle the old question of what really causes the
oxygen sensitivity of FA cells [35, 36].
We now know that at least some of the FA genes are highly conserved dur-
ing evolution and that they play a major role during DNA replication and
recombination in premeitotic stages and meiosis. Some of the FA genes have
been recognized as very ancient ‘caretaker’ genes that have emerged even prior
to the vertebrate lineage [37]. In addition to valuable insights gained from ver-
tebrate model organisms such as zebrafish, mouse and birds, scientists working
Why, What and How Can We Learn from a Rare Disease Like Fanconi Anemia? 5
on ancient model organisms like yeast, C. elegans and Drosophila should be
invited and encouraged to participate in FA-oriented basic research. Last but
not least, Fanconi anemia should be introduced as a human model system into
basic cancer research. It has already been shown that targeted inactivation of
FA genes in cancer cells may offer specific therapeutic options [38, 39]. I am
convinced that both, our patients and our colleagues involved in cancer
research will benefit from inclusion of Fanconi anemia into current research
paradigms.
Concerning the premature aging phenotype of Fanconi anemia, it is obvi-
ous that truly progeroid features (such as in the Hutchinson-Gilford or Werner
syndromes) are far less conspicuous in FA. The decline of bone marrow func-
tion in FA clearly is a much more progressive and devastating event compared
to what happens during normal aging. This is the likely reason why Martin and
Oshima did not include FA in their presentation of human genetic instability
syndromes with progeroid features [40]. However, squamous cell carcinomas of
the oral cavity and genital area are typical tumors of advanced age that occur
very prematurely in FA patients. Likewise, endocrine abnormalities including
hyperinsulinemia, growth hormone deficiency and hypothyroidism as typical
endocrine abnormalities of older individuals affect more than 80% of FA
patients during young adulthood [41]. Impaired gametogenesis and premature
reproductive aging are additional features of FA patients that are reminiscent of
an accelerated aging process. Collectively, the premature manifestation, in FA
patients, of clinical features also encountered during normative aging seems to
indicate that genetic instability per se may be one of the most significant factors
that contributes to and promotes aging. From this point of view, scientists
involved in aging research might benefit from the study of Fanconi anemia.
Conversely, Fanconi anemia patients may ultimately benefit from the insights
gained from the ongoing efforts of serious and scientifically sound anti-aging
research.
References
1 Lobitz S, Velleuer E: Guido Fanconi (1892–1979): a jack of all trades. Nat Rev Cancer 2006;6:
893–898.
2 Schroeder-Kurth TM, Auerbach AD, Obe G (eds): Fanconi Anemia. Clinical, Cytogenetic and
Experimental Aspects. Berlin, Heidelberg, New York, Springer-Verlag, 1989.
3 Zakrzewski S, Sperling K: Genetic heterogeneity of Fanconi’s anemia demonstrated by somatic
cell hybrids. Hum Genet 1980;56:81–84.
4 Duckworth-Rysiecki G, Cornish K, Clarke CA, Buchwald M: Identification of two complementa-
tion groups in Fanconi anemia. Somat Cell Mol Genet 1985;11:35–41.
5 Hanenberg H, Batish SD, Pollok KE, Vieten L, Verlander PC, et al: Phenotypic correction of pri-
mary Fanconi anemia T cells with retroviral vectors as a diagnostic tool. Exp Hematol 2002;30:
410–420.
Schroeder-Kurth 6
6 Taniguchi T, D’Andrea AD: Molecular pathogenesis of Fanconi anemia: recent progress. Blood
2006;107:4223–4233.
7 Gurtan AM, D’Andrea AD: Dedicated to the core: understanding the Fanconi anemia complex.
DNA Repair (Amst) 2006;5:1119–1125.
8 Rosenberg PS, Huang Y, Alter BP: Individualized risks of first adverse events in patients with
Fanconi anemia. Blood 2004;104:350–355.
9 Alter BP, Rosenberg PS, Brody LC: Clinical and molecular features associated with biallelic
mutations in FANCD1/BRCA2. J Med Genet 2007;44:1–9.
10 Reid S, Schindler D, Hanenberg H, Barker K, Hanks S, et al: Biallelic mutations in PALB2 cause
Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 2006;39:162–164.
11 Rosenberg PS, Alter BP, Socie G, Gluckman E: Secular trends in outcomes for Fanconi anemia
patients who receive transplants: implications for future studies. Biol Blood Marrow Transplant
2005;11:672–679.
12 Wagner JE, Eapen M, Macmillan ML, Harris RE, Pasquini R, et al: Unrelated donor bone marrow
transplantation for the treatment of Fanconi anemia. Blood 2007;109:2256–2262.
13 Alter BP: Fanconi’s anemia, transplantation, and cancer. Pediatr Transplant 2005;9:81–86.
14 Williams DA, Croop J, Kelly P: Gene therapy in the treatment of Fanconi anemia, a progressive
bone marrow failure syndrome. Curr Opin Mol Ther 2005;7:461–466.
15 Jacome A, Navarro S, Casado JA, Rio P, Madero L, et al: A simplified approach to improve the
efficiency and safety of ex vivo hematopoietic gene therapy in Fanconi anemia patients. Hum
Gene Ther 2006;17:245–250.
16 Kelly PF, Radtke S, Kalle C, Balcik B, Bohn K, et al: Stem cell collection and gene transfer in
Fanconi anemia. Mol Ther 2007;15:211–219.
17 Schroeder TM, Anschütz F, Knopp A: Spontane Chromosomenaberrationen bei familiärer
Panmyelopathie. Humangenetik 1964;1:194–196.
18 Schroeder TM: Cytogenetische und cytologische Befunde bei enzymopenischen Panmyelopathien
und Pancytopenien. Humangenetik 1966;2:287–316.
19 Newell AE, Akkari YM, Torimaru Y, Rosenthal A, Reifsteck CA, et al: Interstrand crosslink-
induced radials form between non-homologous chromosomes, but are absent in sex chromosomes.
DNA Repair (Amst) 2004;3:535–542.
20 Erdogan F, Chen W, Kirchhoff M, Kalscheuer VM, Hultschig C, et al: Impact of low copy repeats
on the generation of balanced and unbalanced chromosomal aberrations in mental retardation.
Cytogenet Genome Res 2006;115:247–253.
21 Rosenberg PS, Greene MH, Alter BP: Cancer incidence in persons with Fanconi anemia. Blood
2003;101:822–826.
22 Liu JM, Auerbach AD, Young NS: Fanconi anemia presenting unexpectedly in an adult kindred
with no dysmorphic features. Am J Med 1991;91:555–557.
23 Huck K, Hanenberg H, Gudowius S, Fenk R, et al: Delayed diagnosis and complications of
Fanconi anaemia at advanced age – a paradigm. Br J Haematol 2006;133:188–197.
24 Neveling K, Kalb R, Schindler D: Cancer in Fanconi anemia and Fanconi anemia genes in cancer;
in Schindler D, Hoehn H (eds): Fanconi Anemia, Basel, Karger-Verlag, 2007, pp 59–78.
25 Seal S, Thompson D, Renwick A, Elliott A, Kelly P, et al: Truncating mutations in the Fanconi
anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat Genet 2006;38:
1239–1241.
26 Rahman N, Seal S, Thompson D, Kelly P, Renwick A, et al: PALB2, which encodes a BRCA2-
interacting protein, is a breast cancer susceptibility gene. Nat Genet 2006;39:165–167.
27 Schroeder TM, Kurth R: Spontaneous chromosomal breakage and high incidence of leukemia in
inherited disease. Blood 1971;37:96–112.
28 Tonnies H, Huber S, Kuhl JS, Gerlach A, Ebell W, Neitzel H: Clonal chromosomal aberrations in
bone marrow cells of Fanconi anemia patients: gains of the chromosomal segment 3q26q29 as an
adverse risk factor. Blood 2003;101:3872–3874.
29 Schroeder TM, Drings P, Beilner P, Buchinger G: Clinical and cytogenetic observations during a
six-year period in an adult with Fanconi⬘s anemia. Blut 1976;34:119–132.
30 Mathew CG: Fanconi anaemia genes and susceptibility to cancer. Oncogene 2006;25:5875–5884.
Why, What and How Can We Learn from a Rare Disease Like Fanconi Anemia? 7
31 Bremmer JF, Braakhuis BJ, Ruijter-Schippers HJ, Brink A, Duarte HM, et al: A noninvasive
genetic screening test to detect oral preneoplastic lesions. Lab Invest 2005;85:1481–1488.
32 Lo Ten Foe JR, Kwee ML, Rooimans MA, Oostra AB, Veerman AJ, et al: Somatic mosaicism in
Fanconi anemia: molecular basis and clinical significance. Eur J Hum Genet 1997;5:137–148.
33 Auerbach AD, Schroeder TM: First announcement of the Fanconi Anemia International Registry.
Blood 1982;60:1054.
34 Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, et al: A 20-year perspective on the
International Fanconi Anemia Registry (IFAR). Blood 2003;101:1249–1256.
35 Pagano G, Degan P, d’Ischia M, Kelly FJ, Nobili B, et al: Oxidative stress as a multiple effector in
Fanconi anaemia clinical phenotype. Eur J Haematol 2005;75:93–100.
36 Mukhopadhyay SS, Leung KS, Hicks MJ, Hastings PJ, Youssoufian H, Plon SE: Defective mito-
chondrial peroxiredoxin-3 results in sensitivity to oxidative stress in Fanconi anemia. J Cell Biol
2006;175:225–235.
37 Nanda I, Buwe A, Wizenman A, Takata M, Haaf T, Schartl M, Schmid M: Fanconi anemia genes
in vertebrates: evolutionary conservation, sex-linkage, and embryonic expression of FANCC and
FANCG in avian cells; in Schindler D, Hoehn H (eds): Fanconi Anemia, pp 183–199. Basel,
Karger-Verlag, 2007.
38 van der Heijden MS, Brody JR, Dezentje DA, Gallmeier E, Cunningham SC, et al: In vivo thera-
peutic responses contingent on Fanconi anemia/BRCA2 status of the tumor. Clin Cancer Res
2005;11:7508–7515.
39 Gallmeier E, Calhoun ES, Rago C, Brody JR, Cunningham SC, et al: Targeted disruption of
FANCC and FANCG in human cancer provides a preclinical model for specific therapeutic
options. Gastroenterology 2006;130:2145–2154.
40 Martin GM, Oshima J: Lessons from the human progeroid syndromes. Nature 2000;408:263–266.
41 Wajnrajch MP, Gertner JM, Huma Z, Popvich J, Lin K, et al: Evaluation of growth and hormonal
status in patients referred to the International Fanconi Anemia Registry. Pediatrics 2001;107:
744–754.
Schroeder-Kurth 8
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 9–22
Abstract
We present the clinical case histories of 7 patients with Fanconi Anemia (FA) in order
to illustrate the widely divergent phenotypes and clinical courses of the disease. Moreover,
these case histories demonstrate that androgen therapy and hematopoietic stem cell trans-
plantation are therapeutic options that have proven their worth despite side effects that can be
severe or even fatal. We show that mild forms and severe forms of the disease can occur in
individuals with identical mutations, as in siblings from a single family. We wish to convince
the reader that there is no ‘typical’ FA patient, even though there are a number of congenital
malformations and clinical manifestations whose patterns and combinations should alert the
physician to the diagnosis of Fanconi anemia. Each individual case history teaches us certain
facets but never the entire spectrum of a disease that has as many faces as affected individu-
als. Fanconi anemia is a disease physicians must think of. We will soon reach a point where
parents and patients need be less discouraged and less concerned by the diagnosis of Fanconi
anemia since there is tremendous progress in understanding the causes and treating the con-
sequences of the disease. There is no definitive cure on the horizon, but there is continuous
improvement of the quality of life for many patients. Early diagnosis, up-to-date information,
closely knit surveillance, participation in family support groups, and a mutually trustful
alliance with expert physicians and scientists are the cornerstones of optimal care.
Copyright © 2007 S. Karger AG, Basel
Dietrich/Velleuer 10
are the treatments of choice. For FA patients with incipient leukemia, the only
valid option is immediate HSCT. In order to detect leukemic changes as early as
possible, FA patients should be monitored at regular intervals for clonal chro-
mosome changes arising in their bone marrow cells. There is solid evidence that
a specific pattern of chromosome changes emerges prior to the onset of clini-
cally overt leukemia (see chapter by Neitzel et al.). The emergence of such aber-
rant cells is a warning sign that requires immediate action in terms of
preparation for HSCT. Four German patients have recently been transplanted
after positive bone marrow findings thus sparing them the additional (and often
fatal) complications of overt leukemia.
The following case reports are meant to illustrate the high degree of vari-
ability of phenotype and clinical course encountered among FA patients. Since
by now there are 12 and possibly more genes whose mutational changes lead to
the development of FA, much of the clinical variability can be attributed to this
tremendous amount of genetic and, at the single gene level, mutational hetero-
geneity. In the absence of prospective studies we still are fairly ignorant about
genotype-phenotype correlations, even though tentative evidence for such cor-
relations exists for some of the FA genes (see chapter by Kalb et al.). Despite
recent progress in our understanding of the molecular basis of the disease, there
are many open questions relating to gene and protein function that need to be
answered before we can explain, for example, why manifestations of the disease
can be so varied and different, even among siblings carrying identical muta-
tions. In presenting these case reports, the authors wish to point out that even
though these descriptions are kept in the usual neutral and medical style, these
individuals are exceptional human beings by instructing us not only about med-
ical facts, but also by teaching us humility towards the manifold facets, pleasant
and unpleasant, of human life.
Philipp
Philipp (fig. 1) was delivered via C-section in the 37th week of pregnancy
after sonography at 32 weeks had revealed a hydrocephalus. He weighed
2125 g, his height was 44.5 cm, and his head circumference was 32 cm. In addi-
tion, there were multiple structural and functional defects, including persisting
ductus Botalli, cleft lip and palate (Pierre-Robin sequence), bilateral radius
aplasia, bilateral thumb aplasia, anal stenosis, aqueduct stenosis with hydro-
cephalus, hypospadia, cryptorchism, pulmonary hypertension due to atelecta-
sis, strabism, multiple café-au-lait spots, horseshoe kidney, rib anomalies.
Intubation and assisted ventilation were required during the first week of
life. Subsequent pneumonia was controlled with antibiotics. Philipp had to be
fed via gastric tube, and the anal stenosis had to be corrected surgically. From
day 3 of his life, Philipp received Vojta-type of physical therapy which was very
exhausting for him, possibly due to the at that time still unknown persistence of
the duct of Botalli. His parents felt that Philipp’s declining health was due to
these stressful exercises. Because of increasing intracranial pressure, Philipp
had to be re-intubated at the age of 2 weeks, and a shunting procedure was per-
formed. There was an episode of bloody liquor four days after shunting, but
Philipp’s condition gradually improved. After four weeks the shunt became
infected and had to be removed. An attempt to close the persisting ductus
Botalli by drug therapy failed, but surgical closure was finally successful. At the
age of 4 months, a plate was installed in his palate in preparation for the later
surgical correction of the large cleft. Except for low thyroid function (for which
he was treated with thyroid hormone) laboratory parameters were within nor-
mal limits, including peripheral blood counts. He was discharged from the hos-
pital at age 5 months with the preliminary diagnosis of VACTERL association.
There was an additional episode of increased intracranial pressure at age 8 months
which required treatment. Growth retardation became apparent around age 1.
Because of the bilateral atresia of his auditory canals, Philipp received a hear-
ing device, and glasses were prescribed to improve strabism and poor eye sight.
Also at age 1 his cleft palate was closed.
During the second and third years of his life, his pronounced statomotoric
retardation was treated with physiotherapy. Developmental milestones were
severely retarded. When he entered kindergarten at age 3 Philipp could neither
speak, nor feed himself, nor walk, and he required assistance for upright sitting
and standing. During the course of recurrent infections his thrombocyte counts
were noted to decrease such that at age 5 the possibility of FA was considered
Dietrich/Velleuer 12
and quickly confirmed by cell cycle and chromosome breakage studies. Philipp
belongs to the rare X-linked complementation group FA-B.
At the time of his diagnosis hemoglobin was 5.7 g/dl, thrombocytes were
20,000/l, leukocytes were 3,300/l. His blood counts continued to decrease,
requiring regular thrombocyte and erythrocyte transfusions. A venous port
catheter was implanted, but became infected and had to be replaced. Until age
6, Philipp received a total of 15 erythrocyte concentrates and 35 units of throm-
bocytes. Even though his thrombocyte counts returned to normal shortly after
the transfusions, a week later his counts could be as low as 8,000/l. Further
complications were esophagitis and esophageal bleeding between ages 5 and 6.
Because of his multiple impairments and poor health status, neither his parents
nor the attending physicians considered HSCT as a viable option for Philipp,
who requires full time care. In a recent issue of the German FA family news let-
ter his parents write: ‘If he feels well, Philipp likes to laugh and conveys the
impression of alertness, happiness and interest in his environment. Despite all
the physical, mental and emotional energy it takes to care for him, we get so
much back from this very special child and we would not want to miss him’.
Dominik
Michael
Even though Michael (fig. 1) was born at term, he was small for date,
weighing 2,250 g and measuring 45 cm. He displayed a number of birth defects,
including bilateral radial ray and thumb aplasia, coloboma of the right eye, and
rightsided inguinal hernia. Feeding was difficult with frequent regurgitation
requiring a gastric tube with individual meals lasting up to 2 h. A membranous
narrowing of his esophagus was finally diagnosed as cause of his problems
which improved after surgical correction. But because of continuous difficul-
ties to swallow, Michael required mainly liquid or semi-solid foods until the age
of 23. Failure to thrive prompted an investigation of his growth hormone levels at
age 3, but results were normal. The diagnosis of FA was considered at age 4 and
confirmed by chromosome breakage studies but his complementation group
was never determined. Because of poor vision he required glasses from age 6. A
tendency to bleed and cytopenia became apparent at age 9. Michael was treated
with cortisol for two weeks which he did not tolerate well. There were multiple
episodes of esophageal bleedings which were thought to be due to his low
thrombocyte counts, but esophageal varices were diagnosed at age 23 together
with ‘fibrosis’ of his liver. Attempts to stop the bleeding episodes by sclerosing
the varices were only partly successful. Despite these recurrent medical prob-
lems, Michael did well in school and obtained his baccalaureate and his driving
license. At age 24 Michael was diagnosed with pituitary dysfunction, hypo-
gonadism and hypothyroidism. From there on, he was treated with anabolic
steroids, growth hormone and thyroxin. At age 28 his height was 152 cm and he
Dietrich/Velleuer 14
weighed 46 kg. He developed diabetes mellitus which was controlled with oral
medication. Under continuous androgen therapy his blood counts remained
rather stable, with normocytic anemia (hemoglobin 10.9 g/dl) and thrombocytes
the order of 93,000/l. He required no more than 12 erythrocyte transfusions
during his entire lifetime.
At age 31, Michael had to undergo bilateral cataract extraction, laser coagu-
lation of the retina of his right eye, and surgical resection of a malignant bone
tumor of his right jaw. He nevertheless finished his apprenticeship, obtained a
license as a motor boat pilot, and joined the administrative staff of the local
state opera. A year later he experienced increasing pains of the neck and was
diagnosed with advanced squamous cell carcinoma of the lateral and posterior
oropharyngeal wall extending to the base of his skull that had escaped detection
on prior routine examinations. He underwent a heroic 14 h operation with sub-
sequent radiotherapy. After he had received a total dose of only 8 Gy, the radio-
therapy had to be discontinued because of severe thrombocytopenia which
could not be controlled. Michael died at age 32 from the complications of
surgery, radiotherapy and pneumonia.
Marleen B.
Marleen (fig. 2) was born two weeks prior to her due date with hypoplastic
thumbs and a number of fairly large café-au-lait spots. After her first feeding,
she suffered from intestinal tract obstruction due to annular pancreas requiring
immediate surgical correction. Because of her anomalies, a chromosome analy-
sis was ordered yielding a normal female karyotype. All additional investiga-
tions were non-contributory. A first low blood count was observed at age 6, but,
because of a recent episode of infection, was considered transitory and not fol-
lowed up with additional tests. However, upper respiratory tract infections
increased in frequency and severity over the next year, and repeated blood tests
showed a decrease in all three blood cell lineages such that the diagnosis of FA
was finally considered and confirmed by the appropriate laboratory tests.
Marleen B. belongs to complementation group FA-A and is heterozygous for
the FANCA mutation c.4010⫹(1-18)del. The other mutation is unknown.
Marleen quickly became transfusion dependent, and attempts to prevent
her bone marrow failure by androgen and cortisol treatments were unsuccess-
ful. To make things worse, AML was diagnosed at age 8.5. Although treatment
with thioguanin was successful in eradicating her blast cells, it led to rapid dete-
rioration of her bone marrow function. Aspergillosis of her lungs was treated
with granulocyte transfusions (collected from her relatives), but her condition
remained critical. A matching donor could not be found within the available
time such that her physicians were forced to use her father despite 2 mismatches
of his HLA surface antigens. Surprisingly, the bone marrow of her father
engrafted rapidly and without any complication. Much sooner than anticipated
she could be discharged with stable blood counts and in good general health.
Today, seven years after transplant, she is still healthy and lives a full life. She
opted for surgical correction of one of her thumbs (fig. 2) and trained the other
so well that she has excellent manual function. Because of proven growth hor-
mone deficiency, she was started on growth hormone therapy at age 12 which
led to a regular adolescent growth spurt. A very satisfying story for all involved.
Marleen S.
Marleen (fig. 2) was delivered three weeks past her due date via C-section.
She weighed 2,750 g. During her second day of life she underwent surgery for
esophageal atresia type IIIb that was successfully corrected. There were no
complications, and there was no evidence for any other congenital defect. Her
developmental milestones were normal, and she started to use her first words at
the age of 18 months. However, she was difficult to understand and further
investigations revealed a hearing impairment due to middle ear malfunction.
A tympanoplastic procedure as unilateral correction (on her left side) was
attempted, but without notable success. She therefore received bilateral hearing
aids which she still uses today. Supportive logopedic training improved her
speech and hearing.
Dietrich/Velleuer 16
Somewhat lower than normal blood counts were first noted during prepar-
ation for ear surgery at age 4. A control investigation at age 5 yielded the fol-
lowing results: hemoglobin 10.4 g/dl, leukocytes 3,300/l, thrombocytes
70,000/l. Neutrophils were low with 10%. During the following months,
hemoglobin rose to 12 g/dl and neutrophils to 35%. There were no infections
such that no measures were taken. Chromosome analysis at that time revealed a
normal female karyotype, but there were no chromosome breakage studies.
In the meantime a horseshoe kidney and low-grade microcephaly had been
additionally diagnosed, but it took until age 6 before the possibility of FA was
considered and confirmed by cell cycle studies. Marleen turned out to belong to
group FA-A, carrying the FANCA mutation c.1567-1G⬎C in a homozygous or
heterozygous state. In addition to the findings described above, a thorough phys-
ical examination at age 6.75 years noted a brownish skin color of her trunk and
freckling of her left axilla. Except for slightly decreased blood counts, all other
investigations were within normal limits, including bone marrow cytology. At
that time her height was 119 cm (50th percentile), her weight was 23 kg (between
the 50th and 75th percentile), and the final report emphasizes her excellent
health. A heart murmur was noted at age 12 which turned out to be due to a per-
sisting ductus arteriosus Botalli. Angioplastic closure was uneventful.
Contrary to most other FA patients, Marleen’s blood counts remained sta-
ble over the years such that she needed no further intervention. Annual bone
marrow biopsies gave only normal results without evidence for cytogenetic
changes. At ages 13 and 14 three hundred ml of bone marrow were obtained
under general anesthesia, frozen and deposited as reserve for future autologous
transplantation and/or gene therapy. Interestingly, there was further improve-
ment of her blood parameters in the wake of these procedures. Neither cell
cycle nor chromosome breakage studies provided evidence for somatic
mosaicism. Now at age 15, Marleen leads a normal teenage life. Her only handi-
cap is her hearing impairment which she controls with her hearing aids. She is
an example of a very mild clinical course despite several congenital anomalies.
weights (2.5 kg Sarah Ninja, 2.2 kg Valeska), similar low head circumference
(32.5 and 31 cm, respectively) and similar body length (46 and 47 cm, respec-
tively). Overall, the sisters’ phenotypes were relatively mild with short stature,
moderate microcephaly, thumb anomalies, and pigment spots as the only mani-
festations pointing towards FA (fig. 3).
Sarah Ninja experienced a severe clinical course with repeated life-threatening
episodes of epistaxis, very severe anemia, thrombocytopenia and neutropenia,
leading to early and life-long transfusion dependency and recurrent infections
requiring antibiotics and, from age 15, regular G-CSF treatments. She received
Dietrich/Velleuer 18
Fig. 3. Sarah Ninja at ages 6 (left) and 19 (center); Valeska at age 17 (right).
the diagnosis of aplastic anemia at age five, and the diagnosis of FA at age 7.5.
Her younger sister Valeska showed a much milder course. Other than Sarah Ninja,
who had been treated with transfusions for 2.5 years prior to starting her on
androgens, Valeska received androgen therapy prior to the onset of transfusion
dependency. After two years, her androgens (oxymetholon, 2 mg/kg/day) were
reduced stepwise over a period of several years to a maintenance dose of
0.1mg/kg/day. Although there were signs of virilization, Valeska never experienced
any severe complications of androgen therapy (large liver adenomas) that forced a
stepwise reduction of androgen therapy to a minimum dose of 0.1 mg/kg/day in
Sarah Ninja. Until her final year of life when the appearance of a monosomy 7
clone in her bone marrow mandated preparation for HSCT, Valeska had sufficient
bone marrow function to lead a normal and very active teenage life. In contrast,
from her childhood years on, Sarah Ninja’s quality of life was impaired, with
many intermittent episodes of severe distress and near-fatal medical complica-
tions due to severe bleedings and/or infections. Without the dedicated care of her
family and her physicians she would have fallen victim to her refractory bone
marrow failure at a much younger age. It is very tragic indeed that the younger
sister Valeska, having been much less affected by the disease throughout her life-
time, succumbed to the complications of alternate donor HSCT and thus died at
an even younger age than her more severely affected older sister.
The medical histories of these two siblings illustrate several points:
(1) clinical course and quality of life can be very different among affected indi-
viduals, even among siblings carrying identical mutations, (2) in the case of the
two sisters, the quality of life depended above all on the degree of residual bone
marrow function and/or responsiveness to androgen therapy, (3) whereas bone
marrow failure is, within obvious limits, amenable to dedicated medical and
family care, the appearance of cytogenetically aberrant bone marrow clones, in
particular trisomy 3q and/or monosomy 7, may herald leukemia which leaves
HSCT as the only but decidedly high risk option.
The first patient among our case reports illustrates the most severe form of
Fanconi anemia, with severe pre- and postnatal growth retardation, failure to
thrive, a multitude of malformations and malfunctions, including severe cogni-
tive impairment which otherwise is rare. Such a child requires 24 h medical and
family care, and his or her life expectancy is severely limited by the multi-
system disease. Despite these severe handicaps, his family felt that caring for
Philipp is a demanding but likewise rewarding experience. The other patients
were far less affected, but the majority nevertheless required life-saving surgery
as newborns or at some point later in their lives. Surgery included correction of
esophageal atresia, esophageal membranes, pancreas annulare, duodenal atre-
sia, urethral obstruction, anal atresia, heart, eyes (cataract extraction), middle
ear, and thumbs. Without these surgical interventions, either these patients
would not have survived or their quality of life would have been severely
impaired.
Pharmacological intervention consisted mainly of androgen therapy which
stabilized blood counts in half of the patients, in the case of Valeska as long as
12 years. Growth hormone was helpful in three of the patients with proven
growth hormone deficiency, and G-CSF was evidently useful in cases of severe
granulocytopenia. With the exception of Marleen S. who never required trans-
fusions, blood product substitution became mandatory in the majority of
patients at some point, amounting to more than 700 transfusions in the case of
Sarah Ninja or to only 13 transfusions in the case of Michael who died at age 32
from squamous cell carcinoma, the most feared solid tumor in FA patients.
Programs and methods for early detection and prevention of SCC must be one
of the highest priorities in FA medical research. Michael is also an example of
endocrine deficiencies which seem to develop much sooner in FA patients than
in ‘normal’ aging, including pituitary dysfunction, hypothyroidism, hyper-
gonadotrophic hypogonadism and diabetes mellitus.
The case history of Dominik illustrates that HSCT is the therapy of choice
if a matching sibling donor is available, and that additional medical complica-
tion, like kidney failure, can be overcome once bone marrow function is
restored. AML as the most frequent hematopoietic malignancy in FA struck
Marleen B. at age 8.5, and despite many concerns a partially mismatched trans-
plant from her father engrafted well, a perfect example of the beneficial nature
of a ‘graft versus leukemia reaction’. With additional growth hormone therapy
and hand surgery her quality of life is excellent. This is also the case for
Marleen S. despite the fact that she had to undergo surgery for esophageal atre-
sia, for middle ear problems and for persisting ductus Botalli. Marleen S. is an
example of FA patients whose low blood counts remain stable over many years
Dietrich/Velleuer 20
without any specific therapy, and without evidence for somatic reversion of one
of her constitutional mutations.
That siblings with identical mutations can show divergent phenotypes
and a divergent course of their disease has been noted and reported before
[e.g. reference 4]. In the case of Sarah Ninja and Valeska this divergence may
in part have been due to the protracted diagnosis of Sarah Ninja who had to be
treated for aplastic anemia with blood products very early in life, 2.5 years
prior to starting her own androgen therapy. Being aware of the diagnosis in the
family when the younger sister Valeska began to show signs of incipient bone
marrow failure, she was treated with androgens right away and she did not
respond with the massive adenomatous growth in the liver that forced drastic
reduction of androgen therapy in Sarah Ninja’s case. We still are largely igno-
rant why adult height and weight, bleeding episodes, infections, medical
emergencies and overall quality of life were so different between the two
sisters carrying identical mutations. As postulated for the divergence of
ages-of-onset of chorea Huntington [5] polymorphisms in genes that interact
or are associated with the affected FA gene might provide some explanation
but remain to be identified in the case of FA. Finally, the case history of
Valeska reminds us that despite undoubted progress (see Chapter by Eyrich
et al.) alternate donor HSCT remains a high risk procedure, particularly if
coincident with clonal bone marrow changes, preexisting chronic infections,
and adulthood.
Altogether, we hope this brief presentation of case histories will intro-
duce the non-specialist reader to the impressive spectrum of phenotypic
manifestations and the widely divergent clinical courses of FA. There is no
doubt that medical interventions and devoted parental care have improved the
quality of life for the great majority of FA patients. There is a clear bias in the
literature and in physicians’ minds in favor of severe cases such as repre-
sented by Philipp among our case histories, because children with severe
growth deficits and malformations are easily recognized and diagnosed.
Likewise, our sample of case histories by necessity lacks the description of
cases that lead a normal or close to normal life as adults, and who may be
diagnosed only in case of malignancy, late onset bone marrow failure, or a
sibling affected with FA [e.g. references 6 and 7]. We personally know of at
least two adult individuals without any congenital malformations and com-
pletely or close to normal blood counts who carry the diagnosis of FA (based
upon family history and chromosome breakage studies) but lead a normal
working and family life. Working out genotype-phenotype correlations in
such extreme cases, either mild or severe, possibly by studying polymor-
phisms in FA-related genes, will remain one of the great challenges in FA
research in the years to come.
The authors and editors are deeply grateful to the patients and their families who con-
sented to the publication of their first names, medical histories and photographs with the
explicit wish to inform the readers of this volume about the many faces of Fanconi anemia.
They would like to dedicate this paper to the memory of Michael, Sarah Ninja, Valeska and
all the other FA patients who lost their heroic struggle against a deadly disease but who
remain alive in our hearts.
References
1 Joyce O: Fanconi Anemia. Standards for Clinical Care. Fanconi Anemia Research Fund Inc.,
Eugene, Oregon, 1999.
2 Frohnmayer L, Frohnmeyer D: Fanconi Anemia. A Handbook for Families and Their Physicians.
ed 3. Eugene, Oregon, Fanconi Anemia Research Fund Inc., 2000.
3 Fanconi Anämie. Ein Handbuch für Eltern, Patienten und ihre Ärzte. Translated version and exten-
sion of reference 2. Deutsche Fanconi-Anämie-Hilfe e.V., Unna, 2005.
4 Schoof E, Beck JD, Joenje H, Doerr HG: Growth hormone deficiency in one of two siblings with
Fanconi’s anemia complementation group FA-D. Growth Horm IGF Res 2000;10:290–294.
5 Metzger S, Bauer P, Tomiuk J, Laccone F, Didonato S, et al: The S18Y polymorphism in the
UCHL1 gene is a genetic modifier in Huntington’s disease. Neurogenetics 2006;7:27–30.
6 Kwee ML, van der Kleij JM, van Essen AJ, Begeer JH, Joenje H, et al: An atypical case of Fanconi
anemia in elderly sibs. Am J Med Genet 1997;68:62–66.
7 Huck K, Hanenberg H, Gudowius S, Fenk R, Kalb R, et al: Delayed diagnosis and complications
of Fanconi anemia at advanced age – a paradigm. Br J Haematol 2006;133:188–197.
Ralf Dietrich
Deutsche Fanconi-Anämie-Hilfe e.V.
Böckenweg 4
D–59427 Unna (Germany)
Tel. ⫹49 23 08 21 11 or 23 24, E-Mail [email protected]
Dietrich/Velleuer 22
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 23–38
Abstract
The recessive disease Fanconi anemia (FA) is a prototype chromosome instability syn-
drome which shows a high level of spontaneous and induced chromosomal aberrations in
combination with a significantly increased cancer risk. Thus, the underlying defect must be
directly or indirectly involved in a fundamental cellular task of long-lived mammalian cells,
the maintenance of genomic integrity. In addition to the delineation of the FA clinical and
cellular phenotypes, prominent milestones in FA research include proof of extensive genetic
heterogeneity, identification of (so far) 12 disease genes, and elucidation of the FA pathway
involved in the repair of crosslinks at arrested replication forks. What is referred to as the
FA/BRCA pathway represents only part of a complex network of protein-protein interactions
which is far from being understood. The ultimate milestone in FA research would be the
achievement of an individualized and curative therapy. Currently, hematopoietic stem cell
transplantation is a promising but still high-risk therapy, and gene therapy is still at the exper-
imental stage. Nonetheless, in a significant proportion of patients, a kind of ‘natural gene
therapy’ can be observed which results from intragenic recombination or from compensating
second site mutations. The elucidation of these somatic events and their underlying mecha-
nisms can be considered a milestone in genetic research. The remarkable progress in FA
research during the last 10–15 years was fostered by the foundation of FA patient support
groups in many countries. These support groups are gratefully acknowledged as important
motivational milestones in FA research.
Copyright © 2007 S. Karger AG, Basel
Digweed/Hoehn/Sperling 24
gene located on the X-chromosome. Because the authors noted a high intrafa-
milial correlation for age of onset and for the severity of malformations, they
correctly predicted genetic heterogeneity and stated that ‘apart from the stan-
dard type, an especially mild type with late onset, few malformations, and a rel-
atively benign course seems to exist’ [17]. They could not have been more
correct, since today we are well aware that ‘mild’ mutations exist in some of the
FA genes, and that the phenomenon of revertant mosaicism may mitigate the
clinical course. Around 1982, Arleen Auerbach started the Fanconi Anemia
International Registry in the United States which has made a number of land-
mark contributions to our understanding of the clinical and genetic heterogene-
ity of FA so far [18–22].
A definitive clinical milestone in FA research was the realization that
patients exhibit a high risk for myelodysplastic syndrome (MDS), and for
developing overt malignancies [23]. Leukemia is found in about 10% of
younger patients (especially acute myeloblastic leukemia, AML), and solid
tumors, particularly squamous cell carcinomas of the oral cavity and genital
area have been reported in almost 10% of older patients. This, in combination
with bone marrow failure, explains the significantly reduced life expectancy of
FA patients. The cumulative incidence of solid tumors reaches 30% by the age
of 45 years and remains the major threat to older FA patients [24, 25]. Since
malignant growth is one of the possible endpoints of somatic mutations, the
high cancer risk reflects the genetic defect in FA, which is related to the defec-
tive maintenance of genomic integrity. The discovery of spontaneous chromo-
somal instability as cytogenetic expression of the (at that time still elusive)
genetic defect marks the first of the many experimental milestones of FA
research.
FA complex
First description of FA
Genetic heterogeneity
FANCD1
FANCD2
Oxygen sensitivity
FANCL
FANCF
FANCB
FANCJ
FANCE
FANCM
FANCC
1920 1930 1960 1970 1980 1990 2000 2010
HSCT
Fig. 1. Milestones in FA-research. The figure does not yet include FANCN as the most
recently identified FA gene whose product proved identical to PALB2, a protein that functions
downstream of FANCD2 as ‘partner and localizer of BRCA2’ [26, 77].
cycle and involve newly replicated DNA. Since each chromatid break is the vis-
ible manifestation of a DNA double-strand break, the characteristic chromoso-
mal instability of FA indicates that the underlying defect in this genetic disease
is directly or indirectly involved in a fundamental cellular process, the repair of
DNA double-strand breaks. Almost at the same time, spontaneous chromoso-
mal instability was also observed by James German and colleagues in Bloom
syndrome [28]. While in FA cells the breakpoints of the interchanges are almost
randomly distributed, in Bloom syndrome they preferably involve homologous
regions, indicating the impairment of different DNA repair pathways [29].
Looking back at these early data from today’s vantage point it is quite remark-
able how careful observations at the cytogenetic level were able to predict mol-
ecular differences.
About 10 years after the discovery of spontaneous chromosomal instability
as a cytogenetic hallmark of FA, Sasaki and Tonomura showed that the sponta-
neous chromosome instability in FA is associated with a high rate of induced
chromosomal aberrations after treatment with DNA crosslinking agents [30].
The determination of cellular sensitivity towards crosslinking agents such as
diepoxybutane, mitomycin C, cisplatinum or nitrogen mustard still serves as gold
standard for the confirmation of the clinical diagnosis of FA [31]. Because of
the highly variable clinical phenotype, each putative patient requires confirma-
tion via the demonstration of increased in vitro sensitivity towards crosslinking
Digweed/Hoehn/Sperling 26
agents. As an alternative to chromosome breakage studies, crosslink sensitivity
can also be assessed via cell cycle analysis [32].
The term chromosome instability syndrome now covers an increasing
number of recessive disorders which share a high level of spontaneous and
induced chromosomal aberrations. However, the type of chromosome anom-
alies and the specificity of the clastogens vary considerably among the different
disorders. For instance, in Ataxia telangiectasia and Nijmegen breakage syn-
drome, the break points of the spontaneous translocations and inversions in
lymphocytes preferentially involve the T-cell receptor and immunoglobulin
gene loci. Other than FA, these patients are highly sensitive to ionizing radia-
tion, but they share a common characteristic of all chromosome instability syn-
dromes which is a sharply increased risk of malignancy [8].
Identification of FA Genes
As illustrated on the time scale of figure 1, the early nineties of the 20th
century marked the beginning of a new, molecular area in FA research. The first
FA gene identified in 1992 belonged to complementation group C and was
termed FACC (FA complementation group C complementing). Since then, the
nomenclature has changed and today is based on a truncation of the name
Fanconi and the letter of the respective complementation group. Therefore the
12 FA genes are: FANCA, FANCB (syn. FAAP95), FANCC, FANCD1 (syn.
BRCA2), FANCD2, FANCE, FANCF, FANCG (syn. XRCC9), FANCJ (syn.
BRIP1), FANCL (syn. FAAP43, PHF9), FANCM (syn. FAAP250, KIAA1596)
and FANCN (syn. PALB2). Figure 2 shows the human chromosome map with
the location of the 12 known FA genes. With the exception of FANCC and
FANCG, which are both located on chromosome 9, and FANCA and FANCN,
which are both located on chromosome 16, the other FA genes are spread over
different chromosomes, including FANCB on the X.
The advent of the Human Genome project paved the way for the identifi-
cation of human disease genes by the technique of positional cloning, reported
for the first time in 1986 [39]. Positional cloning is based on the knowledge of
a gene’s location in the genome, the isolation of candidate genes from the crit-
ical region and the identification of mutations in a candidate gene in patient
DNA. A prerequisite for the success of this approach is the availability of many
Digweed/Hoehn/Sperling 28
FANCL FANCD2
(2p16.1) (3p25.3)
FANCE
(6p21–22) FANCC FANCG FANCF
(9q22.3) (9p13) (11p15)
1 2 3 4 5 6 7 8 9 10 11 12
FANCN/
FANCD1/ PALB2 FANCJ/
BRCA2 FANCM (16p12) FANCA BRIP1
(13q12.3) (14q21.3) (16q24.3) (17q22)
FANCB
(Xp22.32)
13 14 15 16 17 18 19 20 21 22 X Y
Fig. 2. Human karyotype map with locations of the 12 known Fanconi anemia genes.
Figure taken from reference [75], with permission.
affected individuals, usually from different families, but belonging to the same
complementation group. Thus, due to the extensive genetic heterogeneity, this
approach has obvious limitations in FA. In contrast, the older technique of
‘functional cloning’ of a disease gene depends on fundamental information
about the basic biochemical defect and – due to our ignorance – is rarely applic-
able. In the pre-genomic era, many different biochemical lesions have been
attributed to FA cells, including alterations affecting DNA ligase activity, the
intracellular distribution of topoisomerase, UV excision repair or cellular
NAD⫹ levels. Today we know that these were only secondary phenomena.
Expression cloning is a form of functional cloning which does not require
knowledge of the primary defect. In the case of FA it is based on the transfec-
tion of cells with a normal cDNA library or, alternatively, fusing patient cells with
so called mini cells containing different human chromosomes/chromosomal
regions. Successfully complemented cells are not longer sensitive to cross-
linkers since they now contain a functional FA cDNA. The next step is the iden-
tification of the complementing cDNA/genomic DNA. This approach led to the
cloning of the first gene underlying Fanconi anemia (FANCC) in 1992 by
Manuel Buchwald’s group in Toronto, opening up the molecular avenues in FA
research [40]. Most of the other FA genes were identified by the expression
cloning approach (FANCA, FANCE, FANCF, FANCG, and FANCD2). FANCA
was also independently identified by positional cloning, as was FANCJ.
Digweed/Hoehn/Sperling 30
group C is most frequent [47]. Interestingly, there is no strong correlation
between a given complementation group and a given clinical phenotype, simply
because different mutations in the same FA gene can lead to strikingly different
phenotypic consequences. As a rule, null mutations lead to more severe and ear-
lier disease manifestation than mutations that result in a partially functional
gene product [5]. There is no doubt that identification of each single FA gene
represents a milestone in FA research, both with respect to the practice of med-
ical genetics and the understanding of FA gene and protein function.
Elucidation of the FA pathway should link the clinical and cellular FA phe-
notype to the mutations in the affected genes. However, recent insights into the
molecular pathophysiology of FA have indicated an enormous degree of com-
plexity in which individual pathways are only parts of a network of protein-
protein interactions. Nonetheless, the most important advances in medical
research are often obtained by reducing complexity to basic principles. In the
case of FA, one major unanswered question is how chromosome instability is
linked to the susceptibility of FA cells to DNA crosslinking agents.
Some important milestones in this research were:
• The proof in 1997 that the FA proteins, A and C, form a nuclear complex
and the subsequent realization that the FA proteins B, E, F, G, L, and M are
also part of this core complex [48, 49].
• The discovery that the FANCD2 protein takes a central position in the FA
pathway. The FANCD2 protein is activated by the addition of a ubiquitin
molecule in response to DNA damage [50].
• The finding that the FANCD2 protein directly interacts with known DNA
repair proteins such as BRCA1 and BRCA2/FANCD1, linking DNA
double-strand-break (DSB) repair with DNA cross-link repair [50, 51].
It is now well established that each of the proteins that are assembled to the
FA core complex is needed for monoubiquitination of FANCD2. The catalytic
function is obviously provided by the FANCL protein with its E3 ubiquitin lig-
ase domain. Thus, all these 8 proteins act upstream of the evolutionarily con-
served FANCD2 protein. The posttranslational modification of FANCD2
appears to be crucial for its activity. Proficient monoubiquitination is easily rec-
ognized by the appearance of a larger FANCD2 isoform (FANCD2-L). Western
blotting thus permits convenient subtyping of FA cell lines, since patients with
defects in any of the core complex genes fail to monoubiquitinate FANCD2 and
display only the small (FANCD2-S) isoform [52]. Activated, intact FANCD2
protein is targeted to discrete nuclear foci. The combination of proteins
Digweed/Hoehn/Sperling 32
rational, individualized therapy. From the patient’s point of view, this would
clearly be the ultimate milestone in FA research. Currently, there is no causal
therapy for the genetic instability underlying the clinical manifestations of FA.
In this situation, prenatal diagnosis is requested by couples at risk [58]. Both
functional studies, e.g. determination of sensitivity towards DNA crosslinking
agents, and direct FA gene mutation analysis can be performed with fetal cells.
Although prenatal diagnosis is an important by-product of FA research,
it merely represents a makeshift solution rather than a genuine therapeutic
milestone.
A number of conventional treatments are available to combat bone marrow
failure, including steroid and cytokine medications, but the first line of therapy
is hematopoietic stem cell transplantation (HSCT), pioneered in FA among oth-
ers by Elaine Gluckman and her coworkers [59]. As indicated graphically in
figure 1, both the number of patients transplanted and the success of HSCT
have greatly increased during recent years, mostly due to improvements of con-
ditioning regimens and graft T-cell depletion [60]. Even with suitable donors
HSCT still carries a relatively high risk. In this context, a key observation made
in the laboratory of Heidemarie Neitzel has great impact on the difficult deci-
sion whether and when to subject a patient to transplantation. Neitzel and col-
leagues found that specific clonal chromosomal aberrations, most significantly
gains of 3q, precede the onset of myelodysplasia and/or acute myelocytic
leukemia [61]. Knowing whether a potentially devastating conversion to malig-
nant cell growth is in the offing is an important step forward in the medical
management of FA patients. In combination with the establishment of a rapid
and sensitive FISH-assay [62], this has important consequences for early inter-
vention and treatment of FA patients. Given a low a priori chance of 1 in 4, and
given the small sizes of present day families, matching sibling donors are avail-
able in only a minority of affected families. In such a precarious situation, pre-
implantation genetic diagnosis (PGD) for the selection of an HLA identical
embryo as potential stem cell donor has been proposed and performed [63, 64].
The first case involving an FA family made headlines worldwide, both positive
and negative (‘designer baby’). Clearly, PGD is neither a desirable milestone in
FA research nor a breakthrough for patient care but rather an expression of the
desperate situation of parents faced with limited therapeutical possibilities.
Both the economical and psychological burden of PGD are tremendous and the
success rate is, for biological reasons, very low such that PGD is unlikely to
have a major impact on the medical care of FA families.
Despite considerable efforts, gene therapy is still at the experimental
stage [65]. Nonetheless, in a significant proportion of FA patients a kind of
‘natural gene therapy’ can be observed. This is based on the observation that
some patients have two sets of peripheral blood cells: MMC sensitive and
Digweed/Hoehn/Sperling 34
support groups. As such, each of these groups clearly represents a most
welcome milestone in the common quest to improve the life expectancy and
quality of life of FA patients.
The elusive gene underlying complementation group FA-I (Levitus et al., 2004) has
been identified as a paralog and binding partner of FANCD2, bringing the total number of
identified FA genes to 13. FANCI was detected as an ATM/ATR kinase target protein
required for resistance to mitomycin C. Like FANCD2, the newly detected FANCI protein is
monoubiquitinated at a highly conserved lysine residue. In the chain of events leading to
DNA crosslink repair, FANCI and FANCD2 appear to form an interdependent complex (‘ID-
complex’) required for mutual ubiquitination, activation and association with chromatin
(Smorgorzewska et al., 2007).
References
Levitus M, Rooimans MA, Steltenpool J, Cool NF, Oostra AB, et al: Heterogeneity in Fanconi anemia:
evidence for 2 new genetic subtypes. Blood 2004;103:2498–2503.
Smogorzewska A, Matsuoka S, Vinciguerra P, McDonald ER III, Hurov KE, et al: Identification of the
FANCI Protein, a monoubiquitinated FANCD2 Paralog required for DNA repair. Cell 2007;
doi:10.1016/j.cell.2007.03.009.
References
1 Vogel F, Motulsky AG: Human genetics; in Vogel F, Motulsky AG (eds): Human Genetics
‘Problems and Approaches’, ed 3. Heidelberg-Berlin, Springer, 2002.
2 Digweed M, Sperling K: Molecular analysis of Fanconi anemia. Bioessays 1998;18:579–585.
3 Joenje H, Ketan JP: The emerging genetic and molecular basis of Fanconi Anemia. Nat Rev Genet
2001;2:446–457.
4 D’Andrea AD, Grompe M: The Fanconi Anemia/BRCA pathway. Nature 2002;3:23–34.
5 Kennedy RD, D’Andrea AD: The Fanconi Anemia/BRCA pathway: new faces in the crowd. Genes
Dev 2005;19:2925–2940.
6 Niedernhofer LJ, Lalai AS, Hoeijmakers HJ: Fanconi Anemia (Cross) linked to DNA repair. Cell
2005;123:1191–1198.
7 Bagby GC, Alter BP: Fanconi Anemia. Semin Hematol 2006;43:147–156.
8 Digweed M: Genomic instability in Fanconi anemia and Nijmegen Breakage Syndrome. Genome
Dyn Stab 2007;1:363–380.
9 Levitus M, Joenje H, de Winter JP: The Fanconi anemia pathway of genomic maintenance. Cell
Oncol 2006;28:3–29.
10 Fanconi G: Familiäre infantile perniziosartige Anämie (perniziöses Blutbild und Konstitution).
Jahrb Kinderh 1927;117:257–280.
11 Glanz A, Fraser FC: Spectrum of anomalies in Fanconi anemia. J Med Genet 1982;19:412–416.
12 Gillio AP, Verlander PC, Batish SD, Giampietro PF, Auerbach AD: Phenotypic consequences of
mutations in the Fanconi anemia FAC gene: an International Fanconi Anemia Registry study.
Blood 1997;90:105–110.
Digweed/Hoehn/Sperling 36
38 Chandra S, Levran O, Jurickova I, Maas C, Kapur R, et al: A rapid method for retrovirus-mediated
identification of complementation groups in Fanconi anemia patients. Mol Ther 2005;12:
976–984.
39 Royer-Pokora B, Kunkel LM, Monaco AP, Goff SC, Newburger PE, et al: Cloning the gene for an
inherited human disorder – chronic granulomatous disease – on the basis of its chromosomal location.
Nature 1986;322:32–38.
40 Strathdee CA, Gavish H, Shannon W, Buchwald M: Cloning of cDNAs for Fanconi’s anemia by
functional complementation. Nature 1992;356:763–767.
41 Meetei AR, Levitus M, Xue Y, Medhurst AL, Zwaan M, et al: X-linked inheritance of Fanconi ane-
mia complementation group B. Nat Genet 2004;36:1219–1224.
42 Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, et al: Biallelic inactivation of BRCA2 in
Fanconi anemia. Science 2002;297:606–609.
43 Joenje H, Levitus M, Waisfisz O, D’Andrea A, Garcia-Higuera I, et al: Complementation analysis
in Fanconi anemia: assignment of the reference FA-H patient to group A. Am J Hum Genet 2000;67:
759–762.
44 De Winter JP, Waisfisz Q, Rooimans MA, van Berkel CGM, Bosnoyan-Collins L, et al:
The Fanconi anemia group G gene FANCG is identical with XRCC9. Nat Genet 1998;20:
281–283.
45 Timmers C, Taniguchi T, Hejna J, Reifsteck C, Lucas L, et al: Positional cloning of a novel Fanconi
anemia gene FANCD2. Mol Cell 2001;7:241–248.
46 Tipping AJ, Pearson T, Morgan NV, Gibson RA, Kuyt LP, et al: Molecular and genealogical evi-
dence for a founder effect in Fanconi anemia families of the Afrikaner population of South Africa.
Proc Natl Acad Sci USA 2001;98:5734–5739.
47 Whitney MA, Saito H, Gibson RA, Moses RE, Grompe M: A common mutation in the FACC gene
causes Fanconi anemia in Ashkenazi-Jewish individuals. Nat Genet 1993;4:202–205.
48 Kupfer GM, Naf D, Suliman A, Pulsipher M, D’Andrea AD: The Fanconi anemia proteins, FAA
and FAC, interact to form a nuclear complex. Nat Genet 1997;17:487–490.
49 De Winter JP, van der Weel L, de Groot J, Stone S, Waisfisz O, et al: The Fanconi anemia protein
FANCF forms a nuclear complex with FANCA, FANCC and FANCG. Hum Mol Genet 2000;9:
2665–2674.
50 Garcia-Higuera I, Taniguchi T, Ganesan S, Meyn MS, Timmers C, et al: Interaction of the Fanconi
anemia proteins and BRCA1 in a common pathway. Mol Cell 2001;7:249–262.
51 Taniguchi T, Garcia-Higuera I, Andreassen PR, Gregory RC, Grompe M, D’Andrea AD: S-phase-
specific interaction of the Fanconi anemia protein, FANCD2, with BRCA1 and RAD51. Blood
2002;100:2414–2420.
52 Shimamura A, D’Andrea AD: Subtyping of Fanconi anemia patients: implications for clinical
management. Blood 2003;102:3459.
53 Wang X, Andreassen PR, D’Andrea AD: Functional interaction of monoubiquitinated FANCD2
and BRCA2/FANCD1 in chromatin. Mol Cell Biol 2004;24:5850–5862.
54 Joenje H, Arwert F, Eriksson AW, de Koning H, Oostra AB: Oxygen-dependence of chromosomal
aberrations in Fanconi’s anemia. Nature 1981;290:143–144.
55 Schindler D, Hoehn H: Fanconi anemia mutation causes cellular susceptibility to ambient oxygen.
Am J Hum Genet 1988;43:429–435.
56 Pagano G, Youssoufian H: Fanconi anemia proteins: major roles in cell protection against oxida-
tive damage. Bioessays 2003;25:589–595.
57 Pagano G, Degan P, d’Ischia M, Kelly FJ, Nobili B, et al: Oxidative stress as a multiple effector in
Fanconi anemia clinical phenotype. Eur J Haematol 2005;75:93–100.
58 Auerbach AD, Adler B, Chaganti RS: Prenatal and postnatal diagnosis and carrier detection of
Fanconi anemia by a cytogenetic method. Pediatrics 1981;67:128–135.
59 Gluckman E, Auerbach AD, Horowitz MM, Sobocinski KA, Ash RC, et al: Bone marrow trans-
plantation in Fanconi anemia. Blood 1995;86:2856–2862.
60 Boulad F, Gillio A, Small TN, George D, Prasad V, et al: Stem cell transplantation for the
treatment of Fanconi anemia using a fludarabine-based cytoreductive regimen and T-cell-
depleted related HLA-mismatched peripheral blood stem cell grafts. Br J Haematol 2000;111:
1153–1157.
Karl Sperling
Institute of Human Genetics, Charité – Universitätsmedizin Berlin
Augustenburger Platz 1
D–13353 Berlin (Germany)
Tel. ⫹49 30 450566081, Fax ⫹49 30 450566904, E-Mail [email protected]
Digweed/Hoehn/Sperling 38
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 39–58
Abstract
This article is meant to introduce the reader to the FA/BRCA pathway, the currently
known human FA genes, and their mutations. We review structure and mutational profiles of
the 12 FA genes identified to date. The FA genes display striking variability of size and muta-
tional load which partly reflect their genomic composition and functional constraints.
Biallelic mutations in three genes (FANCA, FANCC and FANCG) account for over 80% of all
FA patients worldwide, and there are only single or very few patients with mutations in the
more recently discovered FA genes. FANCA as the most frequently affected gene displays the
entire spectrum of genetic alterations, including at least 32% large deletions correlated to
Alu-mediated recombination. Viability of FA-D2 patients appears to depend on the presence
of hypomorphic mutations and residual protein. Gene frequencies in excess of 1:100 have
been observed, mainly for mutations in FANCA and FANCC, in isolated populations with
defined founder mutations. With the exception of complementation groups FA-D1 and FA-N
whose patients overwhelmingly present with malignancies in early childhood, most FA genes
lack strict genotype-phenotype correlations. Rather, the particular type of mutation in a given
gene appears to determine the severity of congenital malformations, the age of onset of bone
marrow failure, and the length of survival. FA patients who reach adulthood may carry mild
mutations or may have developed somatic reversions. Prospective studies are needed for a
more definitive exploration of genotype-phenotype correlations.
Copyright © 2007 S. Karger AG, Basel
Around 80% of all patients suffering from Fanconi anemia (FA) belong to
one of three prevalent complementation groups (FA-A, FA-C and FA-G).
According to the International Fanconi Anemia Registry (IFAR), 57% of the
FA patients have mutations in FANCA, 15% in FANCC and 9% in FANCG, fol-
lowed by 4% in FANCD1 and 3% in FANCD2 [1]. Mutations in the other FA
genes are rare, and only single patients have been described for FANCL and
FANCM. The estimated overall incidence of FA is 1 in 200,000 with a carrier
frequency of 1 in 300 [2]. However, much higher patient and carrier frequen-
cies are encountered in certain populations due to founder and inbreeding
effects (see below).
The initial FA-complementation groups were defined via somatic cell fusion
[3, 4], but a variety of strategies were applied for the identification of the under-
lying genes. Expression cloning was used for the identification of FANCA [5],
FANCC [6], FANCE [7], FANCF [8] and FANCG [9], while positional cloning
independently led to the identification of FANCA [10], but also to the identifica-
tion of FANCD2 [11] and FANCJ [12, 13]. FANCD1 was identified by a classical
candidate gene approach [14], as was FANCN [15]. Biochemical isolation of the
respective proteins led to the identification of FANCB [16], FANCL [17] and
FANCM [18]. The identification of the first FA genes (FANCC, FANCA and
FANCG) provided only few hints to their molecular function. Except for FANCG,
which is identical with XRCC9, the initially discovered FA genes shared neither
significant homologies to other proteins nor to known functional domains. This
changed when Howlett and coworkers reported that the breast cancer associated
protein 2 (BRCA2) carries biallelic mutations in FA patients belonging to com-
plementation group D1 [14]. This finding implicated the likely involvement of at
least some of the FA proteins in DNA repair via homologous recombination. In
addition, some of the subsequently discovered genes contain protein domains
with defined biochemical functions. For example, FANCL or PHF9/Pog has a
RING domain with ubiquitin E3 ligase activity [17], FANCJ (identical to
BRIP1/BACH1) has a DNA helicase domain [13], and FANCM (homolog to Hef)
shows homologies to DNA translocases [18]. Specific functional properties can
also be deduced for two of the most recently discovered FA proteins, FANCJ and
FANCN, since these proteins interact with and contribute to the functions of the
well known breast cancer proteins BRCA1 and BRCA2 [12, 19]. The current
view is that FA proteins participate in a complex molecular crosstalk with a large
number of other proteins involved in the regulation and execution of DNA main-
tenance functions, including homology directed DNA repair [20, 21].
Kalb/Neveling/Herterich/Schindler 40
DNA damage
FA core complex
Interstrand crosslinks
F Stalled replication forks
Double strand breaks
E C
G
A M IR
B ? ATM NBS1
BLM FAAP MRE11
100
L ATR RAD50
RPA TOPIIIa
NMR complex
ub
ub P P
D2 D2 D2
ub
USP1
Nuclear foci
Fig. 1. Current concept for the involvement of FA proteins in the response to DNA
damage. For explanations see text.
The FA genes are scattered widely throughout the human genome and vary
considerably in both size and structure, as summarized in table 1. Sequence
Kalb/Neveling/Herterich/Schindler 42
Table 1. Summary of Fanconi anemia (FA) complementation groups, corresponding genes and proteins
Fanconi Anemia Genes: Structure, Mutations, and Genotype-Phenotype Correlations
Name Frequency, %a
a
Based on the International Fanconi Anemia Registry (IFAR), reviewed in reference [1].
b
NES: nuclear export sequences; NLS: nuclear localization signals.
43
comparisons indicate that some of the FA genes must have arisen fairly recently
in evolution, lacking homologs beyond the vertebrate kingdom. However, there
are also FA genes/proteins that are more ancient and highly conserved. For
example, FANCM is closely related to the archael protein Hef (helicase-associated
endonuclease for fork-structured DNA) [19], and FANCJ belongs to the group
of recQ-like helicases that share highly conserved domain structures. In addi-
tion, homologs have been found for FANCD2 in D. melanogaster, C. elegans
and A. thaliana [11], and for FANCL in D. melanogaster and A. gambiae [18].
This might reflect a basic function of the conserved FA proteins, most likely in
the area of DNA repair [38], which has been complemented by the recruitment
of the other FA genes later in evolution. Since oxidative stress and oxidative
damage appear to activate the FA/BRCA pathway, an attractive hypothesis
posits that warm-blooded and long-lived species require the extended FA fam-
ily of genes as additional protection against DNA damage caused by reactive
oxygen species.
Figure 2 provides a graphic summary of the known FA genes, depicting
relative size and intron/exon structure. In addition, numbers and types of muta-
tions are indicated above and below the respective genes. As such, figure 2
illustrates the impressive degree of structural and mutational variation among
the known FA genes, both at the genomic and the cDNA levels. One of the
smallest FA-specific genomic regions, comprising less than 5 kb and only a sin-
gle exon, contains the entire FANCF gene, whereas FANCC and FANCJ are rel-
atively large genes spanning 218 kb and 180 kb, albeit with widely differing
intron/exon structures.
The occurrence of genetic alterations in a given gene depends on a variety
of factors related to gene structure, including the presence of repetitive ele-
ments and base pair composition such as CpG dinucleotides. Physical size by
itself clearly cannot be correlated with the number of mutations in a given FA
gene. A multitude of mutations have been described in relatively small or
medium sized genes like FANCG and FANCA, while genes with large tran-
scripts like FANCM and FANCD1 appear to be affected by relatively few alter-
ations. With respect to type, quantity and distribution of mutations, FANCA
clearly stands out. It is so far the only FA gene with a high proportion of large
deletions, amounting to at least 32% [39, 40] of all observed mutations in
FANCA. The prevalence of large deletions has been correlated with the pres-
ence of numerous Alu-type sequences, promoting Alu-mediated recombination
[41]. With the advent of the MLPA (multiplex ligation dependent probe ampli-
fication) technique, it is very likely that the proportion of deletions detected in
FANCA and perhaps in other FA genes will increase in the future. As illustrated
by the example shown in figure 3, MLPA uncovers deletions that escape detec-
tion with standard PCR-based techniques of mutation analysis.
Kalb/Neveling/Herterich/Schindler 44
Fig. 2. Relative size, exon/intron structure and mutational spectra of the known FA
genes. In order to accommodate and compare the distribution of mutations, exons were
drawn 50-fold larger than corresponding introns. Mutational changes are shown above and
below the respective genes. Key to symbols: 䊊 missense mutation, 䊉 nonsense mutation, 䉭
small deletion/insertion or duplication, aberrant splicing/splice site mutation, 䉲 large
insertion, large deletion, X translation initiation (unpublished data and references for:
FANCA [45], FANCB [16, 49], FANCC [2, 6, 88], FANCD1 [51, 52], FANCD2 [11], FANCE [7],
FANCF [8], FANCG [86], FANCJ [12, 13], FANCL [17], FANCM [18] and IFAR (Rockefeller
University)).
0.5
0.0
1 4 7 10 13 16 19 22 25 28 31 34 37 40
Exon
Fig. 3. Multiplex ligation dependent probe amplification (MLPA) assay for the detec-
tion of deletions in FANCA. A reduction of the sample to control ratio to approximately 0.5
indicates a deleted region. The example shown is from a compound heterozygous FA-A
patient. One allele carries a large deletion involving exons 15 to 20; the other allele carries a
small deletion in exon 38 that involves only 3 bp (c.3788-3790delTCT) and obviously affects
the primer binding site.
FANCD2, a gene nearly of the same size and structure as FANCA, also con-
tains a high number of repetitive elements including many Alu repeats but so far
there are only two deletions involving a single exon. The mutational record of
the FA genes indeed suggests that evolutionarily conserved genes like
FANCD2, FANCL or FANCM may be more important than the more recent
FANCA or FANCG genes which seem to be highly tolerant of mutations. In
addition to serving as catalytic subunit of the FA core complex and as such
responsible for the monoubiquitination of FANCD2 there may be other, as yet
unknown function(s) of FANCL that might explain the relative paucity of
respective patients and mutations. Much more in depth analysis of FA gene
structure and function will be required to clarify whether the less frequently
affected genes truly have more important functions, or whether their underrep-
resentation among FA patients has other, possibly structural reasons. A case in
point is the highly conserved FANCD2 gene. All FA-D2 patients examined to
date have at least one hypomorphic or leaky mutation resulting in residual lev-
els of protein which implicates lethality of biallelic null mutations [42].
However, patients with mutations in some of the evolutionarily novel genes like
FANCE and FANCB also are exceedingly rare, suggesting that mutational inac-
tivation of these genes might not be as permissive as mutational inactivation of
FANCA, FANCC or FANCG. To add to the complexity, several functions other
than its participation in the FA core complex have been suggested for FANCA,
and yet biallelic null mutations in FANCA are compatible with viability and
defects in this gene are impressively numerous.
Kalb/Neveling/Herterich/Schindler 46
Annotations of Individual FA Genes (in Alphabetical Order)
FANCA codes for a protein of 163 kDa which contains a leucine zipper-like
motif, two overlapping bipartite nuclear localization signals (NLS) [43] and
five functional leucine-rich nuclear export sequences (NES) that contribute to
CRM1 (chromosome region maintenance 1)-dependent nuclear export [44].
Since the first identification in 1996 more than 200 different mutations of all
possible types have been described (see fig. 2) (reviewed in reference [45]).
Large deletions are notably frequent in FANCA with a striking overlap of the
affected exons. Even though the exact breakpoints have not been determined in
all cases, a number of reports indicate a direct correlation between the presence
of Alu repeats and deletion breakpoints [39, 46, 47]. In addition to large dele-
tions, small deletions and insertions/duplications are also prevalent [48]. These
changes often are associated with short direct repeats, homonucleotide tracts,
and hotspot consensus sequences like CpG dinucleotides. Base substitutions
account for less than half of the mutations in FANCA and cause premature ter-
mination, amino acid exchanges, or affecting translation initiation or normal
exon recognition.
FANCB is the only X-linked FA gene. Approximately 60% of the genes
located at Xp22.3 exhibit biallelic expression in females, which however is not
the case for FANCB which undergoes X-chromosomal inactivation (NCBI,
OMIM, *300515). To date, mutations in FANCB have been found in five male
FA patients [16, 49] including a deletion affecting the promotor region and por-
tions of the 5⬘ untranslated regions (exon 1), three microdeletions/-insertions,
and a single splice site mutation. The analysis of three healthy female FANCB
carriers revealed the preferential inactivation of the X-chromosome carrying
the mutated allele in both peripheral blood cells and fibroblasts, suggesting a
proliferative advantage for cells expressing the wildtype allele early in embryo-
genesis [16].
FANCC was the first FA gene identified in 1992 [6]. It gives rise to two
alternative transcripts differing in usage of the first untranslated exon, denoted
as –1 or –1a [41]. At the genomic level, FANCC is one of the larger FA genes
spanning more than 218 kb, but coding for a protein with a molecular weight of
63 kDa. Two putative p53 binding sites of unknown functional significance
have been described in FANCC [50]. Mutations in FANCC are responsible for
10–15% of all FA cases, but surprisingly few private mutations have been
reported. As shown in figure 2, the mutational spectrum is heterogeneous, and
alterations are scattered widely throughout the gene with the exception of a pos-
sible cluster in the C-terminal region.
FANCD1 is identical to the gene coding for the breast cancer associated
protein 2 (BRCA2) [14] whose monoallelic mutations predispose to breast,
ovarian, pancreatic and possibly other cancers. Biallelic mutations in
Kalb/Neveling/Herterich/Schindler 48
have been found in FA-F patients [8]. These alterations appear to cluster in the
5⬘ portion of the gene. However, FANCF has gained special prominence as a
gene that is frequently silenced by hypermethylation of its promotor region in
various types of malignancies [63–65] (see chapter by Neveling et al.).
FANCG is identical to XRCC9 which was identified in Chinese hamster
cells as a gene involved in DNA repair [9, 66]. FANCG has seven tetratricopep-
tide repeat motifs (TRPs) [67] thought to function as a scaffold for mediating
protein-protein interaction. The molecular function of FANCG is not restricted
to the FA core complex, since it has been shown to interact with BRCA2 and
XRCC3 directly and independently of other FA proteins [68]. Mutations in
FANCG are of all types, with the exception of deletions. They are scattered
throughout the gene, with possible preference for the N- and C-terminal regions
(cf. fig. 2).
FANCJ is identical to the BRCA1-interacting protein 1 (BRIP1), also
denoted as BRCA1-associated C-terminal helicase 1 (BACH1) [12, 13].
FANCJ/BRIP1 was originally found as a BRCA1 interacting protein containing
two functional domains, a DNA-dependent ATPase and a 5⬘- to 3⬘-DNA heli-
case [69]. In vitro, BRIP1 has DNA unwinding activity with a high preference
for forked duplex substrates and apparently executes efficient strand displace-
ment from a D loop substrate representing a likely intermediate in DNA repair
via homologous recombination [70]. On the genomic level, FANCJ belongs to
the larger FA genes, but only 8 different mutations have been described to date
with most of the affected patients being homozygous or heterozygous for
c.2392T⬎C (R798X) [12, 13].
FANCL was identified by mass spectroscopy of a previously isolated 43-
kDa FA-associated polypeptide [17]. Sequence analysis revealed identity to the
WD40-repeat containing PHD finger protein-9 (PHF9) which is highly con-
served. FANCL is thought to serve as the catalytic subunit of the FA core com-
plex during monoubiquitination of FANCD2. Consistent with this important
role in the FA/BRCA pathway, there is only a single patient known to date
whose disease is caused by a homo- or hemizygous insertion of 177 bp into a
pyrimidine-rich sequence between intron 10 and exon 11, leading to skipping of
exon 11.
FANCM also belongs to the highly conserved FA genes [18]. It shows sim-
ilarities to the archael protein Hef which displays a helicase and an endonucle-
ase domain. However, the biological relevance of these domains for the current
function of FANCM has yet to be established. Using specific antibodies lack of
FANCM was discovered in a single cell line from a patient previously excluded
from most of the known complementation groups [18]. Genetic work up
revealed compound heterozygous mutations, a maternal nonsense mutation in
exon 13 and a paternal deletion within exon 15.
Kalb/Neveling/Herterich/Schindler 50
Table 2. Recurrent/founder mutations in Fanconi anemia genes
a
n.d.: not determined.
isolated regions. In Italy, for example, two mutations in FANCA, c.790C⬎T and
c.3559insG were detected in otherwise unrelated families within the Campania
region [75]. Founder mutations or mutations sharing the same haplotype have
been reported for several FA complementation groups (FA-A, C, D2, G and J)
among different ethnic backgrounds, and independent of the overall mutation
frequencies of the FA genes.
Not all recurrent mutations can be sufficiently explained by founder effects,
but may represent examples of gene regions vulnerable to genetic change
(‘mutational hotspots’). A case in point is the mutation c.2392C⬎T in FANCJ
which was detected in a few unrelated Inuit patients that share the same haplo-
type. However, the same mutation was also found in patients from diverse popu-
lations and ethnical backgrounds where it was correlated with at least three
different haplotypes [12]. The combination of different haplotypes and the rarity
of FA-J patients imply an independent recurrence of the mutation c.2392C⬎T.
Genotype-Phenotype Correlations
Clinical course and severity of FA vary strongly between and even within
families, suggesting relatively weak, if any, genotype-phenotype correlations.
In particular, the onset of hematological abnormalities and longterm survival
appear to depend on additional factors such as number and severity of infec-
tions, nutritional status, and access to and quality of medical care. Any definite
conclusion about genotype-phenotype correlations must await the results of
prospective studies which are not available to date. A retrospective study com-
paring the haematological profiles of complementation groups FA-A, -C and
-G failed to reveal statistically significant differences in the onset of aplastic
anemia [76]. However, the manifestation of cytopenia was more severe in
patients belonging to complementation group FA-G. In addition, FA-G patients
may develop AML or MDS earlier than patients of the other complementation
groups. Congenital abnormalities were comparable among FA-A and FA-G
patients, but less severe than in certain FA-C patients. The clinical data of
patients belonging to complementation group D2 suggests certain differences
to other FA groups [42]. Compared to other patients listed in the IFAR database,
FA-D2 patients exhibit a higher average frequency of congenital abnormalities.
Their clinical course is additionally marked by early average onset of bone mar-
row failure and early dependency on hematopoietic stem cell transplantation. It
should be noted, however, that this FA-D2 patient cohort contained a number of
patients with identical mutations which complicates the statistical comparison.
An undoubtedly severe phenotype is caused by biallelic mutations in
FANCD1/BRCA2. Many of these patients succumb to their malignancies
Kalb/Neveling/Herterich/Schindler 52
(Wilms tumor, medulloblastoma and leukemias) prior to the onset of pancy-
topenia and thus may never be diagnosed as FA [51, 52]. Because of the unusu-
ally severe and early onset of neoplasia in FA-D1 patients, their phenotype
appears to be distinctive and may be more appropriately labeled FA-like rather
than genuine FA [51, 77].
Genotype-phenotype correlations are further complicated by the obvious
heterogeneity of the mutational spectrum within each FA gene, the private char-
acter of mutations and, of course, the high prevalence of compound heterozygos-
ity increasing the possibility of somatic reversion. Any missense change
involving a non-conserved position within the gene should be formally tested for
its functional significance. The availability of site-directed mutagenesis, retrovi-
ral vectors with high transduction efficiencies, and loss of MMC sensitivity as a
functional marker of complementation facilitate the performance of such ‘con-
firmatory’ tests. Adachi and coworkers [78] have published an exemplary study
in which the functional significance of a large number of FANCA mutations has
been tested. In terms of cost and labor such confirmatory studies tend to exceed
the capacity of the average diagnostic laboratory and might best be performed by
specialized centers. Notwithstanding these difficulties, empirical studies have
already shown that some mutations can be correlated with distinct phenotypes.
The mutation c.1007_3066del in FANCA, for example, is associated with an
increased rate of AML or MDS and a higher frequency of severe congenital
abnormalities [76]. In contrast, the FANCC c.67delG alteration is associated
with milder hematologic symptoms and a lower rate of somatic abnormalities
when compared to the majority of other FA patients. Another example of a mild
mutation is the missense mutation c.2444G⬎A in FANCD2 which seems to be
associated with a relatively mild clinical course [42].
A mild clinical course may either be due to mutations that retain some
degree of protein function or to reverse mosaicism. Mild mutations and/or
reverse mosaicism will be preferentially found among older FA patients or
patients who have escaped the diagnosis of FA because of lack of clinical symp-
toms. A prominent example of such a cryptic clinical course has recently been
described by Huck et al. [79] whose patient lacked somatic abnormalities and
was diagnosed as FA, at the remarkable age of 49 years, only because of an
unusually severe response to chemotherapy for bilateral breast cancer. Concerning
somatic reversion, there are numerous examples of reversions in the FANCA,
FANCC and FANCD2 genes. Somatic reversions preferentially involve com-
pound heterozygous patients and, depending on the cell lineages affected, may
lead to improved blood counts and a mild or protracted clinical course (see
chapter by Hoehn et al.).
An additional level of complexity is introduced by the genetic background
of the respective patients or patient group. A case in point is the mutation
References
1 Taniguchi T, D’Andrea AD: The molecular pathogenesis of Fanconi anemia: recent progress.
Blood 2006;107:4223–4233.
2 Verlander PC, Kaporis A, Liu Q, Zhang Q, Seligsohn U, Auerbach AD: Carrier frequency of the
IVS4 ⫹ 4 A→T mutation of the Fanconi anemia gene FAC in the Ashkenazi Jewish population.
Blood 1995;86:4034–4038.
3 Joenje H, Oostra AB, Wijker M, di Summa FM, van Berkel CG, et al: Evidence for at least eight
Fanconi anemia genes. Am J Hum Genet 1997;61:940–944.
4 Levitus M, Rooimans MA, Steltenpool J, Cool NF, Oostra AB, et al: Heterogeneity in Fanconi ane-
mia: evidence for 2 new genetic subtypes. Blood 2004;103:2498–2503.
5 Lo Ten Foe JR, Rooimans MA, Bosnoyan-Collins L, Alon N, Wijker M, et al: Expression cloning
of a cDNA for the major Fanconi anaemia gene, FAA. Nat Genet 1996;14:320–323.
6 Strathdee CA, Gavish H, Shannon WR, Buchwald M: Cloning of cDNAs for Fanconi’s anaemia by
functional complementation. Nature 1992;356:763–767.
7 de Winter JP, Leveille F, van Berkel CG, Rooimans MA, van Der Weel L, et al: Isolation of a
cDNA representing the Fanconi anemia complementation group E gene. Am J Hum Genet
2000;67:1306–1308.
8 de Winter JP, Rooimans MA, van Der Weel L, van Berkel CG, Alon N, et al: The Fanconi anaemia
gene FANCF encodes a novel protein with homology to ROM. Nat Genet 2000;24:15–16.
9 de Winter JP, Waisfisz Q, Rooimans MA, van Berkel CG, Bosnoyan-Collins L, et al: The Fanconi
anaemia group G gene FANCG is identical with XRCC9. Nat Genet 1998;20:281–283.
10 The Fanconi anemia/breast cancer consortium: Positional cloning of the Fanconi anaemia group A
gene. Nat Genet 1996;14:324–328.
11 Timmers C, Taniguchi T, Hejna J, Reifsteck C, Lucas L, et al: Positional cloning of a novel Fanconi
anemia gene, FANCD2. Mol Cell 2001;7:241–248.
12 Levran O, Attwooll C, Henry RT, Milton KL, Neveling K, et al: The BRCA1-interacting helicase
BRIP1 is deficient in Fanconi anemia. Nat Genet 2005;37:931–933.
13 Levitus M, Waisfisz Q, Godthelp BC, de Vries Y, Hussain S, et al: The DNA helicase BRIP1 is
defective in Fanconi anemia complementation group J. Nat Genet 2005;37:934–935.
14 Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, et al: Biallelic inactivation of BRCA2 in
Fanconi anemia. Science 2002;297:606–609.
15 Reid S, Schindler D, Hanenberg H, Barker K, Hanks S, et al: Biallelic mutations in PALB2
cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 2007;39:
162–164.
Kalb/Neveling/Herterich/Schindler 54
16 Meetei AR, Levitus M, Xue Y, Medhurst AL, Zwaan M, et al: X-linked inheritance of Fanconi
anemia complementation group B. Nat Genet 2004;36:1219–1224.
17 Meetei AR, de Winter JP, Medhurst AL, Wallisch M, Waisfisz Q, et al: A novel ubiquitin ligase is
deficient in Fanconi anemia. Nat Genet 2003;35:165–170.
18 Meetei AR, Medhurst AL, Ling C, Xue Y, Singh TR, et al: A human ortholog of archaeal DNA
repair protein Hef is defective in Fanconi anemia complementation group M. Nat Genet
2005;37:958–963.
19 Xia B, Sheng Q, Nakanashi K, Ohashi A, Wu J, et al: Control of BRCA2 cellular and clinical func-
tions by a nuclear partner, PALB2. Mol Cell 2006;22:719–729.
20 Bagby GC, Alter BP: Fanconi anemia. Semin Hematol 2006;43:147–156.
21 Surralles J, Jackson SP, Jasin M, Kastan MB, West SC, Joenje H: Molecular cross-talk among
chromosome fragility syndromes. Genes Dev 2004;18:1359–1370.
22 Thompson LH, Hinz JM, Yamada NA, Jones NJ: How Fanconi anemia proteins promote the four
Rs: Replication, recombination, repair, and recovery. Environ Mol Mutagen 2005;45:128–142.
23 Kennedy RD, D’Andrea AD: The Fanconi Anemia/BRCA pathway: new faces in the crowd. Genes
Dev 2005;19:2925–2940.
24 Garcia-Higuera I, Taniguchi T, Ganesan S, Meyn MS, Timmers C, et al: Interaction of the Fanconi
anemia proteins and BRCA1 in a common pathway. Mol Cell 2001;7:249–262.
25 Meetei AR, Sechi S, Wallisch M, Yang D, Young MK, et al: A multiprotein nuclear complex con-
nects Fanconi anemia and Bloom syndrome. Mol Cell Biol 2003;23:3417–3426.
26 Wang X, Andreassen PR, D’Andrea AD: Functional interaction of monoubiquitinated FANCD2
and BRCA2/FANCD1 in chromatin. Mol Cell Biol 2004;24:5850–5862.
27 Andreassen PR, D’Andrea AD, Taniguchi T: ATR couples FANCD2 monoubiquitination to the
DNA-damage response. Genes Dev 2004;18:1958–1963.
28 Taniguchi T, Garcia-Higuera I, Andreassen PR, Gregory RC, Grompe M, D’Andrea AD: S-phase-
specific interaction of the Fanconi anemia protein, FANCD2, with BRCA1 and RAD51. Blood
2002;100:2414–2420.
29 Pichierri P, Rosselli F: The DNA crosslink-induced S-phase checkpoint depends on ATR-CHK1
and ATR-NBS1-FANCD2 pathways. EMBO J 2004;23:1178–1187.
30 Nakanishi K, Taniguchi T, Ranganathan V, New HV, Moreau LA, et al: Interaction of FANCD2 and
NBS1 in the DNA damage response. Nat Cell Biol 2002;4:913–920.
31 Pace P, Johnson M, Tan WM, Mosedale G, Sng C, et al: FANCE: the link between Fanconi
anaemia complex assembly and activity. EMBO J 2002;21:3414–3423.
32 Ciccia A, Ling C, Coulthard R, Yan Z, Xue Y, et al: Identification of FAAP24, a Fanconi anemia
core complex protein that interacts with FANCM. Mol Cell 2007;25:331–343.
33 Park WH, Margossian S, Horwitz AA, Simons AM, D’Andrea AD, Parvin JD: Direct DNA bind-
ing activity of the Fanconi anemia D2 protein. J Biol Chem 2005;280:23593–23598.
34 Stiff T, Reis C, Alderton GK, Woodbine L, O’Driscoll M, Jeggo PA: Nbs1 is required for ATR-
dependent phosphorylation events. EMBO J 2005;24:199–208.
35 Sobeck A, Stone S, Costanzo V, de Graaf B, Reuter T, et al: Fanconi anemia proteins are required
to prevent accumulation of replication-associated DNA double-strand breaks. Mol Cell Biol
2006;26:425–437.
36 Taniguchi T, Garcia-Higuera I, Xu B, Andreassen PR, Gregory RC, et al: Convergence of the fan-
coni anemia and ataxia telangiectasia signaling pathways. Cell 2002;109:459–472.
37 Cantor SB, Andreassen PR: Assessing the link between BACH1 and BRCA1 in the FA pathway.
Cell Cycle 2006;5:164–167.
38 Marek LR, Bale AE: Drosophila homologs of FANCD2 and FANCL function in DNA repair.
DNA Repair (Amst) 2006;5:1317–1326.
39 Morgan NV, Tipping AJ, Joenje H, Mathew CG: High frequency of large intragenic deletions in
the Fanconi anemia group A gene. Am J Hum Genet 1999;65:1330–1341.
40 Callen E, Tischkowitz MD, Creus A, Marcos R, Bueren JA, et al: Quantitative PCR analysis
reveals a high incidence of large intragenic deletions in the FANCA gene in Spanish Fanconi ane-
mia patients. Cytogenet Genome Res 2004;104:341–345.
41 Savoia A, Centra M, Ianzano L, de Cillis GP, Zelante L, Buchwald M: Characterization of the 5⬘
region of the Fanconi anaemia group C (FACC) gene. Hum Mol Genet 1995;4:1321–1326.
Kalb/Neveling/Herterich/Schindler 56
64 Narayan G, Arias-Pulido H, Nandula SV, Basso K, Sugirtharaj DD, et al: Promoter hypermethyla-
tion of FANCF: disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res
2004;64:2994–2997.
65 Taniguchi T, Tischkowitz M, Ameziane N, Hodgson SV, Mathew CG, et al: Disruption of the
Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med 2003;9:568–574.
66 Liu N, Lamerdin JE, Tucker JD, Zhou ZQ, Walter CA, et al: The human XRCC9 gene corrects
chromosomal instability and mutagen sensitivities in CHO UV40 cells. Proc Natl Acad Sci USA
1997;94:9232–9237.
67 Blom E, van de Vrugt HJ, de Vries Y, de Winter JP, Arwert F, Joenje H: Multiple TPR motifs char-
acterize the Fanconi anemia FANCG protein. DNA Repair (Amst) 2004;3:77–84.
68 Hussain S, Wilson JB, Blom E, Thompson LH, Sung P, et al: Tetratricopeptide-motif-mediated
interaction of FANCG with recombination proteins XRCC3 and BRCA2. DNA Repair (Amst)
2006;5:629–640.
69 Cantor SB, Bell DW, Ganesan S, Kass EM, Drapkin R, et al: BACH1, a novel helicase-like pro-
tein, interacts directly with BRCA1 and contributes to its DNA repair function. Cell
2001;105:149–160.
70 Gupta R, Sharma S, Sommers JA, Jin Z, Cantor SB, Brosh RM Jr: Analysis of the DNA substrate
specificity of the human BACH1 helicase associated with breast cancer. J Biol Chem
2005;280:25450–25460.
71 Callen E, Casado JA, Tischkowitz MD, Bueren JA, Creus A, et al: A common founder mutation in
FANCA underlies the world’s highest prevalence of Fanconi anemia in Gypsy families from Spain.
Blood 2005;105:1946–1949.
72 Peleg L, Pesso R, Goldman B, Dotan K, Omer M, et al: Bloom syndrome and Fanconi’s anemia:
rate and ethnic origin of mutation carriers in Israel. Isr Med Assoc J 2002;4:95–97.
73 Tipping AJ, Pearson T, Morgan NV, Gibson RA, Kuyt LP, et al: Molecular and genealogical evi-
dence for a founder effect in Fanconi anemia families of the Afrikaner population of South Africa.
Proc Natl Acad Sci USA 2001;98:5734–5739.
74 Morgan NV, Essop F, Demuth I, de Ravel T, Jansen S, et al: A common Fanconi anemia mutation
in black populations of sub-Saharan Africa. Blood 2005;105:3542–3544.
75 Savino M, Borriello A, D’Apolito M, Criscuolo M, Del Vecchio M, et al: Spectrum of FANCA
mutations in Italian Fanconi anemia patients: identification of six novel alleles and phenotypic
characterization of the S858R variant. Hum Mutat 2003;22:338–339.
76 Faivre L, Guardiola P, Lewis C, Dokal I, Ebell W, et al: Association of complementation group and
mutation type with clinical outcome in Fanconi anemia. European Fanconi Anemia Research
Group. Blood 2000;96:4064–4070.
77 Hirsch B, Shimamura A, Moreau L, Baldinger S, Hag-alshiekh M, et al: Association of biallelic
BRCA2/FANCD1 mutations with spontaneous chromosomal instability and solid tumors of child-
hood. Blood 2004;103:2554–2559.
78 Adachi D, Oda T, Yagasaki H, Nakasato K, Taniguchi T, et al: Heterogeneous activation of the
Fanconi anemia pathway by patient-derived FANCA mutants. Hum Mol Genet 2002;11:
3125–3134.
79 Huck K, Hanenberg H, Gudowius S, Fenk R, Kalb R, et al: Delayed diagnosis and complications
of Fanconi anaemia at advanced age – a paradigm. Br J Haematol 2006;133:188–197.
80 Kutler DI, Auerbach AD: Fanconi anemia in Ashkenazi Jews. Fam Cancer 2004;3:241–248.
81 Futaki M, Yamashita T, Yagasaki H, Toda T, Yabe M, et al: The IVS4 ⫹ 4 A to T mutation of the
fanconi anemia gene FANCC is not associated with a severe phenotype in Japanese patients. Blood
2000;95:1493–1498.
82 Tamary H, Bar-Yam R, Shalmon L, Rachavi G, Krostichevsky M, et al: Fanconi anaemia group A
(FANCA) mutations in Israeli non-Ashkenazi Jewish patients. Br J Haematol 2000;111:338–343.
83 Magdalena N, Pilonetto DV, Bitencourt MA, Pereira NF, Ribeiro RC, et al: Frequency of Fanconi
anemia in Brazil and efficacy of screening for the FANCA 3788-3790del mutation. Braz J Med
Biol Res 2005;38:669–673.
84 Whitney MA, Saito H, Jakobs PM, Gibson RA, Moses RE, Grompe M: A common mutation in the
FACC gene causes Fanconi anaemia in Ashkenazi Jews. Nat Genet 1993;4:202–205.
Reinhard Kalb
Department of Human Genetics, University of Würzburg
Biozentrum, Am Hubland
D–97074 Würzburg (Germany)
Tel. ⫹49 931 8884089, Fax ⫹49 931 8884069, E-Mail [email protected]
Kalb/Neveling/Herterich/Schindler 58
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 59–78
Abstract
At the cellular level, defects in Fanconi anemia (FA) genes manifest themselves as
hypersensitivity to DNA damaging agents which can be assessed by increased chromosome
breakage and cell cycle changes. As long-term manifestations of cellular genetic instability,
FA patients are at high risk for the development of a rather narrow spectrum of malignancies.
Neoplasia occurs at a much younger age than in non-FA patients, and chiefly includes acute
myeloid leukemia (AML) and squamous cell carcinomas (SCC). Given the role of FA genes in
the maintenance of genetic stability, one would expect that FA genes are frequently altered in
various kinds of tumors arising in non-FA patients. Much to our surprise and with the possible
exception of pancreatic carcinoma, there appears to be no convincing evidence for a frequent
association between either germline or somatic alterations in FA genes and malignancies aris-
ing in non-FA patients. Another notable exception is the FANCF gene which features pro-
minently among the many genes that are silenced in cancer via promoter CpG island
hypermethylation. However, the map location of FANCF adjacent to the 11p hotspot region of
hypermethylation raises concerns about a strictly causal relationship between FANCF inacti-
vation and tumorigenesis. Altogether, the available evidence suggests that tumor cells just like
normal cells benefit from intact FA genes, possibly by keeping their replication machinery
intact and by optimizing their defense against the adverse effects of reactive oxygen species.
Copyright © 2007 S. Karger AG, Basel
DNA-damage
Fig. 1. Hypothetical role of human caretaker genes in the DNA damage response. For
explanation see text.
Neveling/Kalb/Schindler 60
Table 1. Types of malignancies in human caretaker syndromes
Chemicals like 4-NQO, Werner syndrome WRN helicase soft tissue sarcomas
BrdU Bloom syndrome BLM helicase all types of leukemias
and solid tumors
Rothmund-Thompson recQ-IV helicase soft tissue sarcomas
syndrome
Ionizing radiation Ataxia telangiectasia ATM kinase lymphoma
protection Nijmegen breakage NBS1 lymphoma
syndrome
AT-like syndromes MRE11, RAD50, LIG4 lymphoreticular neoplasia
Reactive oxygen species, Fanconi anemia FANCA, B, C, D1, D2, AML, squamous cell
crosslinking agents E, F, G, I, J, L, M, N carcinoma
(?) Reactive oxygen species breast cancer BRCA1, BRCA2 breast cancer
ovarian cancer ovarian cancer
the emergence of neoplasia. The first observation concerns the age of onset of
the neoplastic change which in contrast to the general population occurs in the
teens [4] or, in the case of biallelic mutations in FANCD1/BRCA2, in infants
less than 5 years of age [5, 6]. The second observation concerns the fact that the
types of malignancies prevalent in FA neither mirror the spectrum of malignan-
cies that is seen in the general population, nor do they mimic the kind of malig-
nancies that are seen in the other caretaker syndromes (cf. table 1). AML and
squamous cell carcinomas of the oropharynx and the genital area dominate in
FA. These types of malignancies are rarely if at all encountered in other care-
taker gene syndromes (cf. table 1). This suggests that susceptibility to certain
types of neoplasms primarily reflects the type of caretaker gene that is defective
in a given patient. Both the cellular and the clinical phenotype of FA indicate a
close connection between the FA proteins, DNA repair and tumorigenesis. This
article summarizes some basic information about the obvious FA–cancer con-
nection and thus focuses on the role of cancer in FA as well as on what is cur-
rently known on disruptions of FA genes in cancer.
Leukemia
The most common malignancy arising in FA is leukemia. The overall
cumulative incidence of leukemia is 37%, the median age of onset is 14 years
[4]. The types of leukemia prevalent in FA differ substantially from what is seen
in the general population: Acute myeloid leukemia (AML) dominates in FA
(94% of all leukemias), whereas acute lymphocytic leukemia (ALL) amounts to
only 6%. The reverse holds for non-FA patients (84% of leukemias in the gen-
eral population are ALL) [4]. Hematological malignancies other than AML or
ALL are relatively rare in FA. Chronic myelo-monocytic leukemia (CMMOL)
or Burkitt lymphoma have been described as single cases [7].
It is important to point out that AML may be the first and only manifes-
tation of FA in otherwise normal patients [9]. All defined FAB subclasses [10]
are represented in FA-associated AML [4], albeit with different proportions
compared to the general population. There are more FA patients in the M4 and
M6 and less in the M1 and M2 subclasses of AML compared to non-FA patients
[4]. Several recent studies show that certain clonal aberrations can affect the
risk of developing AML [8, 11–13]. The most frequently reported acquired
clonal aberrations in FA patients are trisomies of chromosome 1q and
monosomies of chromosome 7. In addition, partial trisomies and tetrasomies of
chromosome 3q are proven risk factors for the development of MDS and AML
[14] (see Chapter by Neitzel et al.).
Solid Tumors
The risk for solid tumors in FA patients exceeds that of the general popula-
tion by nearly the 50-fold [15]. In addition, cancer in FA strikes very early. The
median age is 16 years, whereas it is 68 years in the non-FA population [4]. FA
patients may have combinations of different malignancies [4, 7]. In 25% of FA
cases with cancer, the malignancy was present prior to diagnosis of FA [4], an
extreme example being a 49-year-old patient with bilateral carcinoma of the
breast who developed MDS and AML following chemotherapy [16].
Neveling/Kalb/Schindler 62
Squamous Cell Carcinoma
Five percent of all FA patients summarized by Alter [4] had solid tumors,
the most typical tumors being squamous cell carcinomas of the head and neck
(HNSCC) in both males and females, and of the genital area in females.
HNSCC in FA include mostly tumors of the tongue, gingiva, upper esophagus,
larynx and oropharynx [15]. The risk for HNSCC in FA patients is more than
700 times greater than in the general population [15]. In non-FA patients,
HNSCC normally occurs in men older than 40 years who smoke and drink [17].
In contrast, FA patients with HNSCC are usually young, non-smokers and non-
drinkers. Females are more often affected than males, especially among those
patients in whom the diagnosis of FA was made after the diagnosis of cancer,
but this may reflect the overall survival advantage of female FA patients [15].
The mucous epithelia are common routes for viral infections, especially
for HPV (human papilloma virus) and HSV (herpes simplex virus). Immuno-
suppression associated with bone marrow failure might render patients vulner-
able to the integration of the viral DNA [7]. While HPV infections are proven
triggers for the development of cancer of the cervix uteri, their role in non-
anogenital cancers is still unclear [18]. The expression profile of HPV-associated
HNSCC differs from that in HPV-negative HNSCC: TP53 is mutated in HPV-
negative tumors, but remains functional in HPV-associated HNSCC because of
rapid p53 degradation by the viral protein E6 [18]. The presence of HPV in
tumors may affect the prognosis, as patients with HPV-associated HNSCC have
a lower risk for death than HPV-negative HNSCC patients [18]. Concerning FA,
it has been found that 54% of all patients with genital squamous cell carcino-
mas of cervix, vulva and anus had HPV-associated condylomas before occur-
rence of the tumor [7]. These findings were seemingly confirmed by a second
study showing that 84% of FA-associated squamous cell carcinomas (SCC) had
detectable HPV-DNA, while only 36% of non-FA SCC showed viral infection
[19]. However, other investigators were unable to demonstrate such high rates
of HPV contamination. For example, four cell lines freshly established from FA
HNSCC tumors had no detectable HPV DNA. No differences concerning loss
of heterozygosity pattern, TP53 mutations and TP53 polymorphisms were
found between these FA cell lines and non-FA HNSCC cell lines leading the
authors to conclude that FA-associated HNSCC are not fundamentally different
from sporadic HNSCC, except for their sensitivity to crosslinking agents [20].
In addition, chromosomal changes in oral squamous cell carcinomas of two FA
patients were very similar to what was seen in non-FA oral tumors [21].
Squamous cell carcinomas are aggressive tumors that initially present as minor
mucosal lesions, chronic inflammations or red or white cell spots. Diagnosis is
often delayed leading to poor therapeutic success and no better than 50% 2-year
survival rates [17]. A sensitive brush method for obtaining cells from tongue
Liver Tumors
Liver tumors are frequent in FA and in the great majority of cases arise in
the context of androgen therapy for aplastic anemia. They mostly are adenomas
and rarely hepatocellular carcinomas. The cumulative incidence for these
tumors is 46% by age 50, and the median age of onset is 13 years [4].
Development of liver tumors due to androgen therapy is frequent in FA but not
specific for FA patients. The risk for hepatic carcinomas may depend on the
specific type of androgen used. Oxymetholone and methyltestosterone seem to
be associated more frequently with the development of hepatocellular carcino-
mas (HCC), whereas patients treated with danazol develop adenomas but may
be less affected by hepatic carcinomas [22]. Adenomas usually regress upon
discontinuation of therapy. In rare cases, both HCCs and adenomas can be
found in the same patient.
Other Tumors
Other tumors occasionally found in FA patients include brain and renal
tumors, breast carcinoma, basal cell carcinoma, neuroblastoma, desmoid
tumors, gonadoblastoma, melanoma, neurilemmona and osteogenic sarcoma
[7]. A major factor thought to promote tumor development in FA is oxidative
stress [23–26]. It should also be emphasized that biallelic mutations in two of
the downstream FA genes (FANCD1/BRCA2 and FANCN/PALB2) confer an
exceptionally high risk for early childhood leukemia and a variety of soild
tumors [6, 43, 84].
Neveling/Kalb/Schindler 64
So far, this has been especially reported for the FANCF gene which was found
to be epigenetically inactivated in a number of tumors in non-FA patients
(reviewed in [27]). In order to find evidence for the involvement of any of the
other FA genes in malignancies, a variety of tumors have been examined for
sequence variations in FA genes in non-FA patients. Studies searching for
germline changes including polymorphisms of FA genes as possible predispos-
ing factors of tumorigenesis in non-FA patients are summarized in table 2,
whereas the special case of FANCF will be discussed separately (see below).
Table 2 provides a survey of germline mutations in FA genes that were
found in tumors of non-FA patients. Listed are the respective FA genes, the type
of tumors investigated, the family history and the kind of mutations that were
detected. The question whether a given mutation is likely to be causal for
tumorigenesis is answered by ‘yes’ or ‘no’ depending on the respective authors’
interpretation. The conclusion suggested by the data presented in table 2 is that
there is no convincing evidence for a causal association between germline
changes in FA genes and propensity to tumorigenesis in non-FA patients.
In contrast to table 2, table 3 summarizes studies looking for purely
somatic changes in FA genes in tumor patients without FA germline mutations.
Again, studies involving FANCF are not included but are discussed below. Table 3
shows the respective FA genes analyzed, the types of tumors investigated, the
putative family history, and the kinds of mutations detected. Answers to the
question whether a mutation is likely to be causal for tumorigenesis are indi-
cated according to the respective authors’ interpretation. It should be pointed
out that in table 3 there are more ‘yes’ answers than ‘no’ answers to this ques-
tion. However, most of the ‘yes’ answers had to be marked with a question mark
which underlines the high degree of uncertainty that characterizes the interpre-
tation of most of these studies. Final conclusions must await additional studies,
but what is already clear at this point is that somatic mutations in FA genes,
with the possible exception of pancreatic carcinoma, appear to be surprisingly
infrequent events in various types of human malignancies.
As listed in tables 2 and 3, a number of malignancies in non-FA patients,
most notably AML, pancreatic cancer, carcinoma of the breast and ovary, and
squamous cell carcinoma, have been repeatedly investigated for possible asso-
ciations with FA germline and/or somatic mutations. The following is a brief
discussion of the most important findings of these studies (see also [28]).
AML
AML is the most common hematological malignancy in FA, and FA genes
were investigated in several cases of familial and sporadic non-FA AML.
Occasional mutations and polymorphisms were described in FANCA, FANCC,
FANCD1/BRCA2 and FANCG. Since more than 50% of FA patients belong to
a
ALL: Acute lymphocytic leukemia; AML: acute myeloid leukemia; SCC: squamous cell carcinoma.
Neveling/Kalb/Schindler 66
Table 3. Somatic changes of FA genes in non-FA tumor patients
a
AML: Acute myeloid leukemia; SCC: squamous cell carcinoma.
complementation group FA-A, in many studies the FANCA gene has been the
primary target of investigation. For example, a leukemic cell line of an adult
non-FA AML patient was found to exhibit hypersensitivity towards mitomycin
C (MMC) and diepoxibutane (DEB) combined with a decrease of FANCA,
FANCG and FANCD2-L expression which could be corrected by transduction
with a FANCA containing vector. However, the authors of this study failed to
find any evidence for mutations in the FANCA gene. They therefore proposed
the idea of unidentified factors affecting the cellular localization and binding
activity of FANCA, leading to cytogenetic instability and clonal progression
[29]. In another study four cases of heterozygous germline FANCA deletions
were reported among 101 sporadic AML patients. In all four cases, there was no
evidence for inactivation of the second allele either by mutation or promoter
hypermethylation. Since the frequency of the detected deletions was 35-fold
higher than the expected frequency for germline FANCA deletions, the authors
concluded that deletions leading to reduced expression of FANCA may be
involved in a subset of cases of sporadic AML [30]. The possibility of a con-
nection between tumorigenesis and FA genes was also considered for FANCC:
Rischewski and coworkers described a heterozygous FANCC frameshift muta-
tion in two siblings with T-ALL [31]. No other mutations were found in these
patients, leading the authors to the conclusion that heterozygous carriers of
FANC mutations might have an increased risk for developing AML.
Pancreatic Cancer
FANC gene mutations in pancreatic cancer have been described for
FANCA, FANCC and FANCG. Again, in some cases the authors of these studies
consider a connection between detected mutations and tumorigenesis, but other
studies fail to find evidence for such an association. In particular, FANCC muta-
tions were found repeatedly in sporadic cases of pancreatic cancer, and inher-
ited as well as somatic mutations were reported in patients with early onset
pancreatic cancer. The mutations were hemizygous due to loss of one allele in
the tumor. Additionally, a probably inherited nonsense mutation was found in
FANCG. The authors concluded that their findings may point towards a general
involvement of FA genes in cancer [35]. Mutations in FANCC and FANCG in
sporadic pancreatic cancer were also described in another study, in which
germline truncating and missense FANCC mutations were reported as well as a
single somatic missense mutation in FANCG. The germline FANCC mutations
were associated with loss of heterozygosity (LOH) in the tumor. Again, the
authors suggested that inherited mutations in FANCC may predispose to pan-
creatic cancer [36]. However, in critically reviewing these and other data,
Rogers et al. [37] concluded that germline and somatic changes in FANCC and
FANCG, if at all, may have comparatively low penetrance for the pancreatic
cancer phenotype. The same was suggested regarding FANCA: although several
exonic FANCA variants, including disease-associated variants, were identified
Neveling/Kalb/Schindler 68
in lymphocyte DNA from familial pancreatic cancer patients, these variants
were either found in controls at the same frequency as in familial pancreatic
cancer, or there were no convincing segregation data for these variants. Rogers
et al. therefore maintain that germline FANCA mutations do not contribute to
familial pancreatic cancer susceptibility [38]. In reviewing these studies we
find that there is some degree of suggestive but no definitive evidence for
a causal association between defective FA genes and pancreatic cancer.
Neveling/Kalb/Schindler 70
and BRCA1-related breast cancer, but it was not stated whether this was due
to somatic mutations, defective processing, or epigenetic silencing [87].
Otherwise, there are contradictory data concerning genetic variation of
FANCD2 and breast cancer risk [56, 60]. The overall conclusion from these
studies is that a number of germline mutations have been identified in FA
genes of non-FA patients with early onset breast cancer. However, a likely role
as low-penetrance susceptibility genes has been demonstrated only for alter-
ations in the BRCA-associated genes FANCJ and FANCN.
General Conclusions
Neveling/Kalb/Schindler 72
Table 4. Silencing of FANCF in human malignancies
a
AML: Acute myeloid leukemia; HNSCC: head and neck squamous cell carcinoma; LCLC: large-cell lung
carcinoma; NSCLC: non-small cell lung cancer; SCLC: small-cell lung cancer.
References
1 Park Y, Gerson SL: DNA repair defects in stem cell function and aging. Annu Rev Med
2005;56:495–508.
2 Beckman RA, Loeb LA: Genetic instability in cancer: theory and experiment. Semin Cancer Biol
2005;15:423–435.
3 Thompson LH, Hinz JM, Yamada NA, Jones NJ: How Fanconi anemia proteins promote the four
Rs: replication, recombination, repair, and recovery. Environ Mol Mutagen 2005;45:128–142.
4 Alter BP: Cancer in Fanconi anemia, 1927–2001. Cancer 2003;97:425–440.
5 Meyer S, Fergusson WD, Oostra AB, Medhurst AL, Waisfisz Q, et al: A cross-linker-sensitive
myeloid leukemia cell line from a 2-year-old boy with severe Fanconi anemia and biallelic
FANCD1/BRCA2 mutations. Genes Chromosomes Cancer 2005;42:404–415.
Neveling/Kalb/Schindler 74
6 Reid S, Renwick A, Seal S, Baskcomb L, Barfoot R, et al: Biallelic BRCA2 mutations are associ-
ated with multiple malignancies in childhood including familial Wilms tumour. J Med Genet
2005;42:147–151.
7 Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, et al: A 20-year perspective on the
International Fanconi Anemia Registry (IFAR). Blood 2003;101:1249–1256.
8 Alter BP, Caruso JP, Drachtman RA, Uchida T, Velagaleti GV, Elghetany MT: Fanconi anemia:
myelodysplasia as a predictor of outcome. Cancer Genet Cytogenet 2000;117:125–131.
9 Auerbach AD, Weiner MA, Warburton D, Yeboa K, Lu L, Broxmeyer HE: Acute myeloid leukemia
as the first hematologic manifestation of Fanconi anemia. Am J Hematol 1982;12:289–300.
10 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, et al: Proposals for the classification
of the myelodysplastic syndromes. Br J Haematol 1982;51:189–199.
11 Fundia A, Gorla N, Larripa I: Spontaneous chromosome aberrations in Fanconi’s anemia patients
are located at fragile sites and acute myeloid leukemia breakpoints. Hereditas 1994;120:47–50.
12 Ferti A, Panani A, Dervenoulas J, Raptis SA: Cytogenetic findings in a Fanconi anemia patient
with AML. Cancer Genet Cytogenet 1996;90:182–183.
13 Oliveira NI, Ribeiro EM, Raimondi SC, Bittencourt MA, Pasquini R, Cavalli IJ: Two different
karyotypes with 1q abnormalities in a patient with Fanconi anemia. Leuk Res 2002;26:
1047–1049.
14 Tonnies H, Huber S, Kuhl JS, Gerlach A, Ebell W, Neitzel H: Clonal chromosomal aberrations in
bone marrow cells of Fanconi anemia patients: gains of the chromosomal segment 3q26q29 as an
adverse risk factor. Blood 2003;101:3872–3874.
15 Alter BP, Joenje H, Oostra AB, Pals G: Fanconi anemia: adult head and neck cancer and
hematopoietic mosaicism. Arch Otolaryngol Head Neck Surg 2005;131:635–639.
16 Huck K, Hanenberg H, Gudowius S, Fenk R, Kalb R, et al: Delayed diagnosis and complications
of Fanconi anaemia at advanced age – a paradigm. Br J Haematol 2006;133:188–197.
17 Kutler DI, Auerbach AD, Satagopan J, Giampietro PF, Batish SD, et al: High incidence of head and
neck squamous cell carcinoma in patients with Fanconi anemia. Arch Otolaryngol Head Neck
Surg 2003;129:106–112.
18 Gillison ML, Lowy DR: A causal role for human papillomavirus in head and neck cancer. Lancet
2004;363:1488–1489.
19 Kutler DI, Wreesmann VB, Goberdhan A, Ben-Porat L, Satagopan J, et al: Human papillomavirus
DNA and p53 polymorphisms in squamous cell carcinomas from Fanconi anemia patients. J Natl
Cancer Inst 2003;95:1718–1721.
20 van Zeeburg HJ, Snijders PJ, Pals G, Hermsen MA, Rooimans MA, et al: Generation and molecu-
lar characterization of head and neck squamous cell lines of Fanconi anemia patients. Cancer Res
2005;65:1271–1276.
21 Hermsen MA, Xie Y, Rooimans MA, Meijer GA, Baak JP, et al: Cytogenetic characteristics of oral
squamous cell carcinomas in Fanconi anemia. Fam Cancer 2001;1:39–43.
22 Velazquez I, Alter BP: Androgens and liver tumors: Fanconi’s anemia and non-Fanconi’s condi-
tions. Am J Hematol 2004;77:257–267.
23 Pagano G, Youssoufian H: Fanconi anaemia proteins: major roles in cell protection against oxida-
tive damage. Bioessays 2003;25:589–595.
24 Pagano G, Degan P, d’Ischia M, Kelly FJ, Nobili B, et al: Oxidative stress as a multiple effector in
Fanconi anaemia clinical phenotype. Eur J Haematol 2005;75:93–100.
25 Joenje H, Arwert F, Eriksson AW, de Koning H, Oostra AB: Oxygen-dependence of chromosomal
aberrations in Fanconi’s anaemia. Nature 1981;290:142–143.
26 Schindler D, Hoehn H: Fanconi anemia mutation causes cellular susceptibility to ambient oxygen.
Am J Hum Genet 1988;43:429–435.
27 Taniguchi T, D’Andrea AD: The molecular pathogenesis of Fanconi anemia: recent progress.
Blood 2006;107:4223–4233.
28 Lyakhovich A, Surralles J: Disruption of the Fanconi anemia/BRCA pathway in sporadic cancer.
Cancer Lett 2006;232:99–106.
29 Lensch MW, Tischkowitz M, Christianson TA, Reifsteck CA, Speckhart SA, et al: Acquired
FANCA dysfunction and cytogenetic instability in adult acute myelogenous leukemia. Blood
2003;102:7–16.
Neveling/Kalb/Schindler 76
54 Sigurdson AJ, Hauptmann M, Chatterjee N, Alexander BH, Doody MM, et al: Kin-cohort esti-
mates for familial breast cancer risk in relation to variants in DNA base excision repair, BRCA1
interacting and growth factor genes. BMC Cancer 2004;4:9.
55 Luo L, Lei H, Du Q, von Wachenfeldt A, Kockum I, et al: No mutations in the BACH1 gene in
BRCA1 and BRCA2 negative breast-cancer families linked to 17q22. Int J Cancer 2002;98:638–639.
56 Lewis AG, Flanagan J, Marsh A, Pupo GM, Mann G, et al: Mutation analysis of FANCD2,
BRIP1/BACH1, LMO4 and SFN in familial breast cancer. Breast Cancer Res 2005;7:R1005–R1016.
57 Vahteristo P, Yliannala K, Tamminen A, Eerola H, Blomqvist C, Nevanlinna H: BACH1 Ser919Pro
variant and breast cancer risk. BMC Cancer 2006;6:19.
58 Seal S, Barfoot R, Jayatilake H, Smith P, Renwick A, et al: Evaluation of Fanconi Anemia genes in
familial breast cancer predisposition. Cancer Res 2003;63:8596–8599.
59 Thompson E, Dragovic RL, Stephenson SA, Eccles DM, Campbell IG, Dobrovic A: A novel
duplication polymorphism in the FANCA promoter and its association with breast and ovarian can-
cer. BMC Cancer 2005;5:43.
60 Barroso E, Milne RL, Fernandez LP, Zamora P, Arias JI, et al: FANCD2 associated with sporadic
breast cancer risk. Carcinogenesis 2006;27:1930–1937.
61 Hu N, Li G, Li WJ, Wang C, Goldstein AM, et al: Infrequent mutation in the BRCA2 gene in
esophageal squamous cell carcinoma. Clin Cancer Res 2002;8:1121–1126.
62 Sparano A, Quesnelle KM, Kumar MS, Wang Y, Sylvester AJ, et al: Genome-wide profiling of
oral squamous cell carcinoma by array-based comparative genomic hybridization. Laryngoscope
2006;116:735–741.
63 de Winter JP, Rooimans MA, van Der Weel L, van Berkel CG, Alon N, et al: The Fanconi anaemia
gene FANCF encodes a novel protein with homology to ROM. Nat Genet 2000;24:15–16.
64 Leveille F, Blom E, Medhurst AL, Bier P, Laghmani el H, et al: The Fanconi anemia gene product
FANCF is a flexible adaptor protein. J Biol Chem 2004;279:39421–39430.
65 Taniguchi T, Tischkowitz M, Ameziane N, Hodgson SV, Mathew CG, et al: Disruption of the Fanconi
anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med 2003;9: 568–574.
66 Agrelo R, Cheng WH, Setien F, Ropero S, Espada J, et al: Epigenetic inactivation of the premature
aging Werner syndrome gene in human cancer. Proc Natl Acad Sci USA 2006;103: 8822–8827.
67 de Bustros A, Nelkin BD, Silverman A, Ehrlich G, Poiesz B, Baylin SB: The short arm of chro-
mosome 11 is a ‘hot spot’ for hypermethylation in human neoplasia. Proc Natl Acad Sci USA
1988;85:5693–5697.
68 Hoffmann MJ, Florl AR, Seifert HH, Schulz WA: Multiple mechanisms downregulate CDKN1C
in human bladder cancer. Int J Cancer 2005;114:406–413.
69 Tischkowitz M, Dokal I: Fanconi anaemia and leukaemia - clinical and molecular aspects. Br
J Haematol 2004;126:176–191.
70 Van der Heijden MS, Brody JR, Elghalbzouri-Maghrani E, Zdzienicka MZ, Kern SE: Does
tumorigenesis select for or against mutations of the DNA repair-associated genes BRCA2 and
MRE11?: considerations from somatic mutations in microsatellite unstable (MSI) gastrointestinal
cancers. BMC Genet 2006;7:3.
71 Tischkowitz M, Ameziane N, Waisfisz Q, De Winter JP, Harris R, et al: Bi-allelic silencing of the
Fanconi anaemia gene FANCF in acute myeloid leukaemia. Br J Haematol 2003;123:469–471.
72 Van Der Heijden MS, Brody JR, Kern SE: Functional screen of the Fanconi anemia pathway in
cancer cells by Fancd2 immunoblot. Cancer Biol Ther 2004;3:534–537.
73 Meyer S, White DJ, Will AM, Eden T, Sim A, et al: No evidence of significant silencing of Fanconi
genes FANCF and FANCB or Nijmegen breakage syndrome gene NBS1 by DNA hyper-methylation
in sporadic childhood leukaemia. Br J Haematol 2006;134:61–63.
74 Marsit CJ, Liu M, Nelson HH, Posner M, Suzuki M, Kelsey KT: Inactivation of the Fanconi
anemia/BRCA pathway in lung and oral cancers: implications for treatment and survival.
Oncogene 2004;23:1000–1004.
75 Olopade OI, Wei M: FANCF methylation contributes to chemoselectivity in ovarian cancer.
Cancer Cell 2003;3:417–420.
76 Narayan G, Arias-Pulido H, Nandula SV, Basso K, Sugirtharaj DD, et al: Promoter hypermethyla-
tion of FANCF: disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res
2004;64:2994–2997.
Kornelia Neveling
Department of Human Genetics, University of Würzburg, Biocenter
B107, Am Hubland, 97074 Würzburg (Germany)
Tel. ⫹49 931 888 4089, Fax ⫹49 931 888 4069
E-Mail [email protected]
Neveling/Kalb/Schindler 78
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 79–94
Abstract
Fanconi anemia patients have a high risk for bone marrow failure, aplastic anemia,
myelodysplastic syndrome, and acute myeloid leukemia. Many FA patients acquire chromo-
somal aberrations in their bone marrow (BM) cells. The significance and predictive value of
these somatic aberrations for hematopoietic function and malignant progress are not fully
understood. Therefore, we initiated in cooperation with the ‘Deutsche Fanconi-Anämie-Hilfe
e.V.’ a prospective cytogenetic follow-up in BM cells of FA patients utilizing systematically
molecular cytogenetic techniques. The most frequent clonal aberrations are gains of material
of the long arm of chromosome 3, loss of most of the long arm of chromosome 7 or loss of
one entire copy of chromosome 7, and gains of the long arm of chromosome 1. The available
data suggest that these clonal chromosome aberrations in bone marrow cells of FA patients
represent an important step in the initiation of MDS and AML. Our data of patients with 3q
aberrations revealed that gains of 3q are an adverse risk factor. The overall survival in the 3q
group was extremely poor compared to FA patients without such aberrations. None of the FA
patients with 3q gains survived without undergoing HSCT. Because of the high MDS/AML
risk and the significantly higher mortality in the group of FA patients with 3q aberrations, we
recommend a systematic evaluation of all FA patients by molecular cytogenetics in order to
detect aberrations, including subtle aberrations, as early as possible. Such clonal aberrations
are a very strong clinical warning sign. All available evidence suggests that the finding of
any chromosomal abnormality in bone marrow cells, especially abnormalities involving
chromosomes 3 and 7, warrant very close clinical follow-up, as they may signal the develop-
ment of MDS or AML.
Copyright © 2007 S. Karger AG, Basel
Fanconi anemia patients have a high risk for bone marrow failure, aplastic
anemia (AA), myelodysplastic syndrome (MDS), acute myeloid leukemia
(AML), and, later in life, for epithelial malignancies. The most life-threatening
early event in most complementation groups is bone marrow failure (patients
with bi-allelic BRCA2 mutations, who seem to have early onset solid tumors,
may be exceptions). The hematologic complications of FA have been exten-
sively reviewed [1]. Patients with FA develop bone marrow (BM) failure, typi-
cally during the first decade of life. The actuarial risk of developing BM failure
is 90% by 40 years of age. The actuarial risk of developing hematologic neo-
plasms is 33% by 40 years of age [2]. The median age of patients who develop
cancer is 14 years for acute myelogenous leukemia (AML). Many FA patients
acquire chromosomal aberrations in their bone marrow cells. The significance
and the predictive value of such clonal alterations with respect to hematopoietic
function and malignant progress are not fully understood but are important to
define. This is the aim of our studies.
Neitzel/Kühl/Gerlach/Ebell/Tönnies 80
Table 1. Overview of clonal chromosomal aberrations in bone marrow cells of Fanconi anemia patients
reported in the literature
a
AML: Acute myeloid leukemia; BMF: bone marrow failure; NS: not specified; MDS: myelodysplastic syndrome.
patients. Auerbach and Allen mentioned that ‘considerable evidence shows that
the presence of chromosomally abnormal clones in the BM heightens the risk for
development of leukemia’ [3]. This is in agreement with Butturini et al. [8] who
demonstrated that the detection of a clonal cytogenetic abnormality correlates
with decreased survival of FA patients. In 1993, a provocative question was
Neitzel/Kühl/Gerlach/Ebell/Tönnies 82
formulated [9]: ‘Clonal chromosomal abnormalities in Fanconi’s anemia: what
do they really mean?’ Alter et al. [9] presented three of 11 FA patients with
clonal abnormalities after adequate cytogenetic analysis. The fact that some of
the clonal aberrations displayed transient appearance led those authors to reason
that ‘changing cytogenetic patterns may not be related to leukemic evolution in
patients with a DNA repair defect’. In contrast, in 2000 it was demonstrated that
the five-year probability of survival after detection of a clone is 0.40 compared
to 0.94 without a clone [7]. Notwithstanding these observations, those authors
stated that the morphological changes in the direction of MDS may be more
important than clonal aberrations with respect to the prediction of an adverse
outcome. Along these lines, Maarek et al. emphasized: ‘The significance of
chromosomal changes without apparent evolution to overt AML or MDS will
only be understood when more FA patients are cytogenetically studied and fol-
lowed over a sufficient period of time’ [5]. However, up to now long term follow-up
data are sparse and there is urgent need to collect such data.
All cytogenetic studies in the 70s to 90s have in common that the chromo-
somal aberrations could not be sufficiently defined by conventional cytogenet-
ics alone in a considerable number of patients. Karyotype descriptions denoted
as ‘⫹’, ‘add’, or ‘mar’ are common, indicating that additional material of
unknown origin was detected either as translocation to another chromosome
(⫹; add) or as marker chromosomes (⫹mar) of unidentified origin (table 1).
The results from these early cytogenetic studies in bone marrow cells of
FA patients illustrate the problem of all studies which were based solely on con-
ventional cytogenetics. There is a limited number of analyzable BM meta-
phases, especially in patients with progressive bone marrow failure, and there is
a limited resolution of GTG-banded chromosomes of BM metaphases resulting
in the inability to identify and assign chromosomal changes correctly, espe-
cially when these are subtle and/or mosaic. However, the correct assignment of
the karyotypic changes in BM cells is a prerequisite for any predication with
respect to hematopoietic function and progression to malignancy.
Patientsa,b,c 53 18 35
Age
Median (months) 141 149 125
Range (months) 34–463 95–463 34–442
Gender
Male 28 11 17
Female 25 7 18
Complementation group
FANCA 28 10 18
FANCC 4 4 0
FANCG 8 3 5
unknown 13 1 12
genotypic reversiona 3 1 2
Outcome
MDSb 9 9 0 ⬍0.001
AMLc 5 4 1
MDS⫹AML 14 13 1 ⬍0.001
Alive 44 11 33 0.005
HSCT 20 12 8
a
Spontaneous reversion of the cellular FA phenotype in lymphocytes by back mutation or gene conversion in
compound heterozygotes.
b
MDS was defined according to the FAB classification; 8/9 MDS patients presented 3–26% blasts in the marrow.
c
AML was defined by more than 30% blasts in the marrow.
d
Fisher’s exact test (2-sided).
Neitzel/Kühl/Gerlach/Ebell/Tönnies 84
wcp3 wcp10
3 10 3/12
a b c
wcp3 wcp6
d
b 2/11 3/9 6/12
Neitzel/Kühl/Gerlach/Ebell/Tönnies 86
46,XX
90 46,XX,dup(1q)
46,XX,⫹3q
80
46,XX,⫹3q,⫹3q
Percentage of aberrant cells
70 46,XX,⫹3q,⫹3q,delXp
60
50
40
30
20
10
0
T0 ⫹7 m ⫹10m ⫹15m ⫹46m ⫹50m 19 3/10
(n ⫽56) (n ⫽ 50) (n ⫽51) (n⫽33) (n ⫽50) (n⫽41)
a Interval of the cytogenetic analyses b c
80
70
60 46,XY
45,X
50
45,X,⫹3q
40
30
20
10
0
T0 ⫹12m ⫹36m ⫹45m ⫹50m ⫹52m
(n⫽ 56) (n ⫽ 20) (n ⫽40) (n⫽8) (n⫽8) (n⫽55)
Time scale of cytogenetic analyses
Fig. 4. Another example for the outgrowth of cells with a clonal 3q aberration. The
first aberrant clone was detected 3 years after the first cytogenetic analysis of bone marrow
cells in this FA patient. At this time, more than 70% of the cells revealed the karyotype 45,X
indicating a loss of the Y chromosome. Only 9 months later, all cells carried additional mate-
rial of 3q. The analyses 5 and 7 months later indicated that the clone persisted.
the time of the first BM analysis while in the other 6 patients the monosomy 7
had developed in the 3q aberrant clone as a secondary event. Even though the
number of patients affected with both, a gain of 3q and monosomy 7, is fairly
small, our data imply that gains of 3q might increase the risk for the subsequent
development of monosomy 7.
In order to determine the percentage of aberrant cells in non-dividing BM
and peripheral blood mononuclear cells (PBMC), we performed interphase FISH
analyses using a YAC from 3q27–28. Our results demonstrate that FA patients
with 3q aberrations in up to 70–80% of their BM and PBMC cells have more
than two signals as compared to a maximum of 2–3% in normal control sub-
jects. Since there was a normal karyotype in their T- and B-lymphocytes, our
results suggest that almost all circulating granulocytes must carry the 3q aber-
ration in these patients. In three of the FA patients the 3q aberration was
detected only by screening PBMC by interphase FISH prior to conventional
cytogenetic analysis of BM cells. This proves the high sensitivity and speci-
ficity of interphase FISH as a screening tool for aberrant clones in PBMC (for
details regarding interphase FISH screening of PBMC see the chapter by
Tönnies et al.).
Neitzel/Kühl/Gerlach/Ebell/Tönnies 88
1.0
0.8
Cum risk of MDS/AML
0.6
0.4
0.2
0
0 100 200 300 400 500
Age (months)
Fig. 5. Risk of developing MDS or AML in Fanconi anemia with (—) or without ( )
chromosome 3 aberration (Log Rank: p ⬍ 0.001) as published 2003 by Tonnies et al. [10].
(—) n ⫽ 18, MDS/AML 13, risk 0.90 ⫾ 0.09. ( ) n ⫽ 35, MDS/AML 1, risk 0.08 ⫾ 0.08.
Patients 23 35
Male 16 17
Female 7 18
Overall survival 8/23 (35%) 27/35 (77%)
HSCT 17/23 (74%) 10/35 (29%)
Survival after HSCT 7/17 (41%) 7/10 (70%)
Survival without HSCT 0/5 (0%) 20/25 (80%)
Neitzel/Kühl/Gerlach/Ebell/Tönnies 90
At present, the data of Hirsch et al. [14] and our investigations are the only
studies utilizing molecular cytogenetic techniques for the investigation of
altogether more than 150 FA patients. Both groups report a high incidence of
3q gains which clearly stands out as the most frequent clonal aberration. The
second frequent clonal aberration in FA is monosomy 7 (partial 7q or complete)
followed by 1q gains.
The high frequency of monosomy 7 and the occasional occurrence of
monosomy 5 have led to the suggestion that AML in FA is similar to secondary
AML which is induced by alkylating agents in non-FA patients [15]. Various
alkylating agents are carcinogenic and augment the incidence of cancer in the
general population, and the incidence of AML is dramatically increased in can-
cer patients secondary to chemotherapy with alkylating agents. These patients
often develop nonrandom clonal chromosomal changes in the preleukemic
phase, especially monosomy 7, deletion of 7q, and monosomy 5. There is little
doubt that the presence of such chromosomally abnormal BM clones heightens
the risk for the development of leukemia in non-FA patients with secondary
AML. It therefore appears unlikely that these clonal aberrations should be pre-
dictors for the leukemic process in secondary AML of non-FA patients, but only
innocent bystanders in the malignant progress in patients with DNA repair defi-
ciencies. Thus, the statement made by Alter et al. in 1993 that ‘changing cyto-
genetic patterns may not be related to leukemic evolution in patients with a
DNA repair defect’ may no longer be valid [9].
While monosomy 5 and monosomy 7 are aberrations found in non-FA
patients with secondary myelodysplastic syndrome (MDS), gains of 3q have
not been described in secondary MDS. In AML of adults, 3q abnormalities are
seen in only 2% of patients and they are thought to be extremely rare in primary
childhood AML of non-FA patients. Common chromosomal aberrations in
childhood AML of non-FA patients include a number of balanced transloca-
tions like t(8;21), t(9;11), inv(16), t(11;19). These aberrations have not been
described in FA patients, indicating that AML in FA patients may indeed be
similar to secondary AML. With regard to the obvious rarity of 3q aberrations
in primary childhood AML a recent publication is of special interest. Haltrich
et al. [16] investigated 28 childhood AML cases with FISH and found aberrations
of chromosome 3 in nine out of the 28 patients. Four patients had a trisomy 3,
one patient a tetrasomy of 3q26.3–q28, one patient loss at 3q13–q21, and three
patients had structural rearrangements involving the 3q26 and/or 3q21 site. All
pediatric AML patients with 3q aberrations had a poor outcome [16].
The data of Haltrich et al. [16] thus suggest that it might turn out that 3q
aberrations are risk factors for the development of primary AML in both FA and
non-FA patients. It is noteworthy that the 3q chromosomal region is also
involved in multiple solid tumors indicating that its tumor promoting potential
Conclusions
Acknowledgement
Supported by grants from the Deutsche Fanconi Anämie Hilfe e.V. and the Charité
Research Fund (No.2000-627).
References
1 Alter BP, Greene MH, Velazquez I, Rosenberg PS: Cancer in Fanconi anemia. Blood 2003;101:
2072.
2 Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, et al: A 20-year perspective on the
International Fanconi Anemia Registry (IFAR). Blood 2003;101:1249–1256.
3 Auerbach AD, Allen RG: Leukemia and preleukemia in Fanconi anemia patients. A review of the
literature and report of the International Fanconi Anemia Registry. Cancer Genet Cytogenet 1991;51:
1–12.
4 Butturini A, Gale RP: Long-term bone marrow culture in persons with Fanconi anemia and bone
marrow failure. Blood 1994;83:336–339.
Neitzel/Kühl/Gerlach/Ebell/Tönnies 92
5 Maarek O, Jonveaux P, Le Coniat M, Derre J, Berger R: Fanconi anemia and bone marrow clonal
chromosome abnormalities. Leukemia 1996;10:1700–1704.
6 Alter BP: Fanconi’s anemia and malignancies. Am J Hematol 1996;53:99–110.
7 Alter BP, Caruso JP, Drachtman RA, Uchida T, Velagaleti GV, Elghetany MT: Fanconi anemia:
myelodysplasia as a predictor of outcome. Cancer Genet Cytogenet 2000;117:125–131.
8 Butturini A, Gale RP, Verlander PC, Adler-Brecher B, Gillio AP, Auerbach AD: Hematologic
abnormalities in Fanconi anemia: an International Fanconi Anemia Registry study. Blood 1994;84:
1650–1655.
9 Alter BP, Sealise A, McCombs J, Najfeld V: Clonal chromosomal abnormalities in Fanconi’s
anaemia: what do they really mean? Br J Haematol 1993;85:627–630.
10 Tonnies H, Huber S, Kuhl JS, Gerlach A, Ebell W, Neitzel H: Clonal chromosomal aberrations in
bone marrow cells of Fanconi anemia patients: gains of the chromosomal segment 3q26q29 as an
adverse risk factor. Blood 2003;101:3872–3874.
11 Berger R, Bernheim A, Le Coniat M, Vecchione D, Schaison G: Chromosomal studies of leukemic
and preleukemic Fanconi’s anemia patients: examples of acquired ‘chromosomal amplification’.
Hum Genet 1980;56:59–62.
12 Berger R, Bussel A, Schenmetzler C: Somatic segregation and Fanconi anemia. Clin Genet 1977;11:
409–412.
13 Schaison G, Leverger G, Yildiz C, Berger R, Bernheim A, Gluckman E: Fanconi’s anemia.
Incidence of its development into leukemia. Presse Med 1983;12:1269–1274.
14 Hirsch B, Dolan M, Brandt K, MacMillan ML, Wagner J, et al: Recurring clonal chromosomal
abnormalities in Fanconi Anemia: characterization and association with bone marrow pathology.
15th Annual Fanconi Anemia Research Scientific Symposium, 2004.
15 Bagby GC, Alter BP: Fanconi Anemia. Semin Hematol 2006;43:147–156.
16 Haltrich I, Kost-Alimova M, Kovacs G, Klein G, Fekete G, Imreh S: Multipoint interphase FISH
analysis of chromosome 3 abnormalities in 28 childhood AML patients. Eur J Haematol 2006;76:
124–133.
17 Lisker R, Cobo de Gutierrez A: Cytogenetic studies in Fanconi’s anemia. Description of a case
with bone marrow clonal evolution. Clin Genet 1974;5:72–76.
18 Bourgeois CA, Hill FG: Fanconi anemia leading to acute myelomonocytic leukemia: cytogenetic
studies. Cancer 1977;39:1163–1167.
19 Tanzer J, Frocrain C, Desmarest MC: Anomalies du chromosome 1 dans 3 cas de leucémie com-
pliquant une aplasie Fanconi (FA). Nouv Rev Fr Hématol 1980;22(suppl):XCIII.
20 Obeid DA, Hill FG, Harnden D, Mann JR, Wood BS: Fanconi anemia. Oxymetholone hepatic
tumors, and chromosome aberrations associated with leukemic transition. Cancer 1980;46:
1401–1404.
21 Auerbach AD, Weiner MA, Warburton D, Yeboa K, Lu L, Broxmeyer HE: Acute myeloid leukemia
as the first hematologic manifestation of Fanconi anemia. Am J Hematol 1982;12:289–300.
22 Rotzak R, Kaplinsky N, Chaki R, Blei F, Berkowitz M, et al: Giant marker chromosome in
Fanconi’s anemia transforming into erythroleukemia in an adult. Acta Haematol 1982;67:
214–216.
23 Stivrins TJ, Davis RB, Sanger W, Fritz J, Purtilo DT: Transformation of Fanconi’s anemia to
acute nonlymphocytic leukemia associated with emergence of monosomy 7. Blood 1984;64:
173–176.
24 Nowell P, Bergman G, Besa E, Wilmoth D, Emanuel B: Progressive preleukemia with a chromo-
somally abnormal clone in a kindred with the Estren-Dameshek variant of Fanconi’s anemia.
Blood 1984;64:1135–1138.
25 Woods WG, Nesbit ME, Buckley J, Lampkin BC, McCreadie S, et al: Correlation of chromosome
abnormalities with patient characteristics, histologic subtype, and induction success in children
with acute nonlymphocytic leukemia. J Clin Oncol 1985;3:3–11.
26 Huret JL, Benz E, Guilhot F, Brizard A, Tanzer J: Fluctuation of a clone 46,XX,i(7q) in bone mar-
row in a Fanconi anaemia. Hum Genet 1986;74:98–100.
27 Barton JC, Parmley RT, Carroll AJ, Huang ST, Goodnough LT, et al: Preleukemia in Fanconi’s
anemia: hematopoietic cell multinuclearity, membrane duplication, and dysgranulogenesis.
J Submicrosc Cytol 1987;19:355–364.
Heidemarie Neitzel
Institute of Human Genetics, Charité, Medical University of Berlin
Augustenburger Platz 1
D–13353 Berlin (Germany)
Tel. ⫹49 30 450566411, Fax ⫹49 30 450566933, E-Mail [email protected]
Neitzel/Kühl/Gerlach/Ebell/Tönnies 94
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 95–109
Abstract
Bone marrow (BM) failure in Fanconi anemia (FA) patients followed by myelodysplas-
tic syndrome or acute myeloid leukemia is frequently associated with the occurrence of spe-
cific clonal chromosomal imbalances in BM cells. Our previous data of patients with 3q
aberrations revealed that gains of 3q are strongly associated with a poor prognosis and repre-
sent an adverse risk factor in FA. In routine cytogenetic diagnostics, these clonal aberrations
can be detected after BM aspiration, culturing of bone marrow cells, and karyotyping of
GTG-banded metaphase chromosomes. However, some unbalanced mosaic aberrations in
bone marrow cells are too subtle to be detected by conventional cytogenetics alone. In order
to develop a sensitive and fast detection protocol for routine use, we studied FA cells by inter-
phase fluorescence in situ hybridization (I-FISH) using a YAC panel. We established a sensi-
tive interphase FISH assay for the early detection of prognostically poor clonal chromosome
aberrations in bone marrow and peripheral blood interphase cells of FA patients as a useful
complement to metaphase chromosome analysis of bone marrow cells. The analysis on the
single cell level allows the early and sensitive detection and the monitoring of chromosomal
imbalances as prerequisite for a timely intervention and treatment of affected patients.
Copyright © 2007 S. Karger AG, Basel
Conventional Cytogenetics
The diagnosis of Fanconi anemia was confirmed in all patients by a standard chromo-
some breakage test after mitomycin C treatment. Cell culture and high-resolution chromo-
some preparations from peripheral blood lymphocytes and synchronized bone marrow
metaphases for the detection of numerical and structural chromosome aberrations employed
standard techniques and GTG-banding [15]. Computer-assisted karyotyping of GTG-
banded chromosomes was performed with the Ikaros system (MetaSystems, Altlussheim,
Germany).
Tönnies/Huber/Volarikova/Gerlach/Neitzel 96
Direct Cell Preparations
For interphase cell preparation, native bone marrow (BMD: bone marrow direct prepar-
ation) and peripheral blood (PBD: peripheral blood direct preparation) specimens (0.5 ml)
were incubated without prior culturing in hypotonic potassium chloride solution (0.4%;
10 min at 37⬚C) and fixed in ice-cold methanol/acetic acid (v/v, 3:1) three times. After drop-
ping on slides, cells were aged overnight at room temperature. For long time storage, slides
were kept at –20⬚C. Peripheral blood and bone marrow smears were prepared by standard
protocols.
Molecular Cytogenetics
In addition to conventional cytogenetics where up to 50 bone marrow metaphases were
analyzed, we utilized comparative genomic hybridization (CGH) and fluorescence in situ
hybridization (FISH) with whole chromosome painting probes for the detection and charac-
terization of chromosomal abnormalities.
Results
Sensitivity of I-FISH
To circumvent selective outgrowth of clonal aberrations in vitro and to
gain insights into the in vivo situation of the patient, we decided to analyze
uncultured interphase cells. As a first technical result, comparative analyses of
bone marrow or peripheral blood smears versus directly prepared cells showed
that hybridization quality and success rate were superior when cell suspension
slides (BMD and PBD) rather than smears were used (data not shown).
Therefore, we determined the positive cut-off levels of our YAC-clone probe
sets on bone marrow and peripheral blood direct preparations by hybridizing
70 control samples (56 PBD; 14 BMD). An average of 500 nuclei for each
hybridization were scored, which means that positive cut-off values were deter-
mined based on the evaluation of 70,000 interphase nuclei. The mean percent-
age of 3q positive cells (presence of three green signals; GGG) in BMD
preparations of control subjects was 1% (mean ⫹3 SD; SD ⫽ 0.25), whereas
using PBD the cut-off level was 2.9% (SD ⫽ 0.69). The cut-off values for par-
tial tetrasomy of chromosome 3q, which in FA specimens often results from
clonal progression of a partial trisomy 3q clone, were 0.98 (SD ⫽ 0.27) for
BMD and 0.26 (SD ⫽ 0.08) for PBD. The monosomy 7 cut-off values were
3.59 (SD ⫽ 0.99) in BMD and 0.46 (SD ⫽ 0.14) in PBD using the chromo-
some 7 probe set.
Tönnies/Huber/Volarikova/Gerlach/Neitzel 98
100 BM-CC
90 BMD
Aberrant cells (%)
80 PBD
70
60
50
40
30
20
10
0
Patient A Patient B Patient C Patient C Patient C
(0 month) (⫹5 months) (⫹8 months)
Fig. 1. Percentage of affected cells with partial trisomy 3q obtained by I-FISH with the
chromosome 3 YAC probe on cultivated bone marrow cells (BM-CC), bone marrow direct
preparations (BMD) and peripheral blood direct preparations (PBD) of three patients (A–C).
For patient C, the follow-up period extended over eight months.
direct preparations was higher than the positive cut-off values for the chromo-
some 3 probe set on BMD (1%) and PBD (2.9%).
70
60
50
40
30
20
10
0
1 2 3 4 5 6 7 8 9 10
No. of analysis with matched BMD and PBD samples
Tönnies/Huber/Volarikova/Gerlach/Neitzel 100
100
GGRR [33]
90 GGGRR [33 ⫹ 3q]
80 GGGGRR [33 ⫹3q3q]
Others
70
Cells (%)
60
50
40
30
20
10
0
th
s
th
th
th
th
th
on
on
on
on
on
on
m
m
0
10
10
⫹
⫹
⫹
⫹
Chromosome 3-PBD Chromosome 3-BMD
3 3 3
70
60
50
40
30
20
10
0
on D
m BD
m B
s)
s)
s)
s)
s)
m BD
m BD
m BD
m BD
th
(0 3 P
th
th
th
th
th
5 P
11 P
13 P
16 P
18 P
on
on
on
on
on
#
(⫹ #3
(⫹ #3
(⫹ #3
(⫹ #3
(⫹ #3
100 GGRR [77]
GR [7]
90 Others
80
70
Aberrant cells (%)
60
50
40
30
20
10
0
on D
m BD
m B
s)
s)
s)
s)
s)
m BD
m BD
m BD
m BD
th
(0 7 P
th
th
th
th
th
5 P
11 7 P
13 7 P
16 P
18 P
on
on
on
on
on
#
(⫹ #7
(⫹ #7
(⫹ #7
#
#
(⫹
(⫹
Tönnies/Huber/Volarikova/Gerlach/Neitzel 102
the monosomy 7 in cultured metaphase spreads (45% affected cells) and
BMD interphase cells (68% affected cells). This long term follow-up clearly
illustrates that I-FISH on PBD cells is a powerful tool for the early and sensi-
tive detection of adverse clonal evolution in uncultured peripheral blood cells
of FA patients.
Discussion
Tönnies/Huber/Volarikova/Gerlach/Neitzel 104
estimate of the proportion of genetically altered cells in native bone marrow
preparations (in vivo situation) [25].
The number of affected cells in our test samples was clearly higher than the
positive cut-off values for the chromosome 3 probe set for BMD (1%) and PBD
(2.9%). We therefore conclude that in the case of FA patients who show aneu-
ploidy by conventional cytogenetics of BM cells, the application of I-FISH on
uncultured BMD and PBD cells leads to the accurate confirmation of the chro-
mosomal alterations.
Tönnies/Huber/Volarikova/Gerlach/Neitzel 106
analysis. Kearns et al. [20] examined bone marrow cells from 96 unselected
patients with bone marrow failure syndromes to assess the frequency of unde-
tected aneuploidy for chromosomes 7 and 8 by FISH. They identified
27 patients with cytogenetically undetected monosomy 7 or trisomy 8. Accord-
ing to these authors, undetected aneuploidy exists in bone marrow cells of a
significant percentage of patients with bone marrow failure syndromes. As we
have demonstrated in this report, the problem can be overcome by continued
monitoring of FA patients using I-FISH on PBD and, if available BMD cells.
This approach will assure the early detection of adverse clones with mono-
somy 7 or gain of 3q allowing earlier intervention, such as bone marrow trans-
plantation, to prevent the onset of leukemia in these patients. We therefore
have adopted I-FISH analysis with specific YAC clones for all FA patients as a
standard practice. Using this strategy, 3q aberrations were detected by screen-
ing PBD cells in three other FA patients (data not shown). This observation
emphasizes the high sensitivity and specificity of I-FISH as a screening assay
for the detection of aberrant clones in native peripheral blood mononuclear
cells.
Concluding Remarks
Acknowledgements
We are indebted to all Fanconi anemia patients, their parents, and their clinicians who
supported this study. YAC clones were provided by the Max-Planck Institute of Molecular
Genetics, Berlin, Germany. Supported by grants from the Charité Universitätsmedizin Berlin
and the Deutsche Fanconi-Anämie-Hilfe e.V. (www.fanconi.de).
1 Rosenberg PS, Greene MH, Alter BP: Cancer incidence in persons with Fanconi anemia. Blood
2003;101:822–826.
2 Tischkowitz M, Dokal I: Fanconi anaemia and leukaemia – clinical and molecular aspects. Br
J Haematol 2004;126:176–191.
3 Alter BP, Greene MH, Velazquez I, Rosenberg PS: Cancer in Fanconi anemia. Blood 2003;
101:2072.
4 Berger R, Bernheim A, Le Coniat M, Vecchione D, Schaison G: Chromosomal studies of leukemic
and preleukemic Fanconi’s anemia patients: examples of acquired ‘chromosomal amplification’.
Hum Genet 1980;56:59–62.
5 Auerbach AD, Allen RG: Leukemia and preleukemia in Fanconi anemia patients: a review of the
literature and report of the International Fanconi Anemia Registry. Cancer Genet Cytogenet
1991;51:1–12.
6 Berger R, Le Coniat M, Schaison G: Chromosome abnormalities in bone marrow of Fanconi ane-
mia patients. Cancer Genet Cytogenet 1993;65:47–50.
7 Butturini A, Gale RP, Verlander PC, Adler-Brecher B, Gillio AP, Auerbach AD: Hematologic
abnormalities in Fanconi anemia: an International Fanconi Anemia Registry study. Blood
1994;84:1650–1655.
8 Berger R, Jonveaux P: Clonal chromosome abnormalities in Fanconi anemia. Hematol Cell Ther
1996;38:291–296.
9 Alter BP: Fanconi’s anemia and malignancies. Am J Hematol 1996;53:99–110.
10 Maarek O, Jonveaux P, Le Coniat M, Derre J, Berger R: Fanconi anemia and bone marrow clonal
chromosome abnormalities. Leukemia 1996;10:1700–1704.
11 Alter BP, Caruso JP, Drachtman RA, Uchida T, Velagaleti GV, Elghetany MT: Fanconi anemia:
myelodysplasia as a predictor of outcome. Cancer Genet Cytogenet 2000;117:125–131.
12 Tonnies H, Huber S, Kühl JS, Gerlach A, Ebell W, Neitzel H: Clonal chromosomal aberrations in
bone marrow cells of Fanconi anemia patients: gains of the chromosomal segment 3q26q29 as an
adverse risk factor. Blood 2003;101:3872–3874.
13 Schroeder TM, Kurth R: Spontaneous chromosomal breakage and high incidence of leukemia in
inherited disease. Blood 1971;37:96–112.
14 Schroeder TM: Relationship between chromosomal instability and leukemia. Hematol
Bluttransfus 1974;14:94–96.
15 Barch MJ, Knutsen T, Spurbeck JL: The AGT Cytogenetics Laboratory Manual. Philadelphia,
Lippincott-Raven, 1997.
16 Tonnies H, Stumm M, Wegner RD, Chudoba I, Kalscheuer V, Neitzel H: Comparative genomic
hybridization based strategy for the analysis of different chromosome imbalances detected in con-
ventional cytogenetic diagnostics. Cytogenet Cell Genet 2001;93:188–194.
17 Telenius H, Carter NP, Nordenskjold M, Ponder BA, Yunnacliffe A: Degenerate oligonucleotide-
primed PCR: general amplification of target DNA by a single degenerate primer. Genomics
1992;13:718–725.
18 Flactif M, Lai JL, Preudhomme C, Fenaux P: Fluorescence in situ hybridization improves the
detection of monosomy 7 in myelodysplastic syndromes. Leukemia 1994;8:1012–1018.
19 Thurston VC, Ceperich TM, Vance GH, Heerema NA: Detection of monosomy 7 in bone marrow
by fluorescence in situ hybridization. A study of Fanconi anemia patients and review of the litera-
ture. Cancer Genet Cytogenet 1999;109:154–160.
20 Kearns WG, Sutton JF, Maciejewski JP, Young NS, Liu JM: Genomic instability in bone marrow
failure syndromes. Am J Hematol 2004;76:220–224.
21 Berger R, Bussel A, Schenmetzler C: Somatic segregation and Fanconi anemia. Clin Genet
1977;11:409–412.
22 Tonnies H: Modern molecular cytogenetic techniques in genetic diagnostics. Trends Mol Med
2002;8:246–250.
Tönnies/Huber/Volarikova/Gerlach/Neitzel 108
23 Wilkens L, Komminoth P, Nasarek A, von Wasielewski R, Werner M: Rapid detection of kary-
otype changes in interphase bone marrow cells by oligonucleotide primed in situ hybridization
(PRINS). J Pathol 1997;181:368–373.
24 Velagaleti GV, Tharapel SA, Tharapel AT: Validation of primed in situ labeling (PRINS) for inter-
phase analysis: comparative studies with conventional fluorescence in situ hybridization and chro-
mosome analyses. Cancer Genet Cytogenet 1999;108:100–106.
25 Yan J, Zhang XX, Fetni R, Drouin R: Trisomy 8 and monosomy 7 detected in bone marrow using
primed in situ labeling, fluorescence in situ hybridization, and conventional cytogenetic analyses.
A study of 54 cases with hematological disorders. Cancer Genet Cytogenet 2001;125:30–40.
26 Huret JL, Benz E, Guilhot F, Brizard A, Tanzer J: Fluctuation of a clone 46,XX,i(7q) in bone mar-
row in a Fanconi anaemia. Hum Genet 1986;74:98–100.
27 Ferti A, Panani A, Dervenoulas J, Raptis SA: Cytogenetic findings in a Fanconi anemia patient
with AML. Cancer Genet Cytogenet 1996;90:182–183.
28 Auerbach AD, Weiner MA, Warburton D, Yeboa K, Lu L, Broxmeyer HE: Acute myeloid leukemia
as the first hematologic manifestation of Fanconi anemia. Am J Hematol 1982;12:289–300.
29 Stivrins TJ, Davis RB, Sanger W, Fritz J, Purtilo DT: Transformation of Fanconi’s anemia to acute
nonlymphocytic leukemia associated with emergence of monosomy 7. Blood 1984;64:173–176.
Dr. H. Tönnies
Institute of Human Genetics, Charité, Universitätsmedizin Berlin
Augustenburger Platz 1
D–13353 Berlin (Germany)
Tel. ⫹49 30 450566807, Fax ⫹49 30 450566933, E-Mail [email protected]
Abstract
Chromosome breakage analysis following exposure of cultured cells to DNA-
crosslinking agents has long been considered the ‘gold standard’ for the confirmation or
exclusion of Fanconi anemia (FA). Cells containing DNA damage are arrested and accumu-
late, with a 4c DNA content, near the S/G2-phase border of the cell cycle. As manifestation
of their impaired DNA damage response, FA cells typically express elevated G2-phase cell
fractions which can easily be measured by flow cytometry. In our experience with more than
3,000 such analyses, at most 1 in 10 blood samples submitted for the exclusion or confirma-
tion of FA yields a positive result. Compared to traditional chromosome breakage analysis,
cell cycle testing is less demanding and offers the advantage of speed and low cost. We pre-
fer flow cytometric cell cycle testing for the initial screening of patients, in whom unex-
plained thrombocytopenia, macrocytic aplastic anemia or other clinical findings require the
exclusion of FA. In addition to its application in diagnostic screening, we here show that cell
cycle analysis has become a valuable tool for the determination of clastogen sensitivity (such
as required within the context of complementation studies), for the precise definition of cell
cycle checkpoints, and for the quantitative determination of compartment-specific cell cycle
delay or cell cycle arrest. Cell cycle analysis not only provides a reliable test system for the
initial confirmation or exclusion of FA, but also serves as a highly informative tool for the
comprehensive characterization of the FA cellular phenotype.
Copyright © 2007 S. Karger AG, Basel
The DNA histograms in the left hand panel of figure 1 (shaded grey) repre-
sent 48-h cell cycle distributions of FA fibroblasts exposed to increasing concen-
trations of MMC as indicated on the right. In the range from 10 to 100 nM MMC,
G2-phase accumulations increase from 21.9 to 72.8%, reflecting the innate
MMC sensitivity of FA cells. Via retroviral complementation, the fibroblast
strain shown in figure 1 had been typed as FA-G, since it was complemented by
stable transfer of FANCG cDNA in a bicistronic construct with GFP. Gene trans-
fer was achieved in 61.8–68.4% of cells. The series of cell cycle distributions
shown in the right hand panel of figure 1 represent the isogenic, but comple-
mented counterparts of the distributions shown on the left. Non-complemented
cells remain GFP-negative (shaded grey) whereas complemented cells are GFP-
positive (shaded green). The GFP marker permits the unambiguous discrimin-
ation between complemented and non-complemented cells via electronic gating
on green-positive and exclusion of green-negative cells. The MMC response of
complemented cells (green panels) is far less pronounced than that of FANCG-
defective cells (grey panels). In the concentration range between 10 and 100 nM
MMC, the respective G2-phase accumulations of complemented cells increase
only from 12.0 to 34.4%. Since the cell cycle distributions shown in figure 1 are
all derived from one and the same fibroblast culture of a single FA-G patient, the
striking differences in the response to MMC between the left and right hand pan-
els solely and uniquely reflect the functional competence of the FANCG gene in
protecting cells from the adverse effects of MMC.
Figure 2 illustrates dose-response curves of the G2-phase fractions of non-
complemented vs. complemented FA-G cells over a broader range of MMC
concentrations. In the range of 33–100 nM, differences of the G2-phase frac-
tions between non-complemented and complemented cells from these other-
wise isogenic cultures amount to 30% of the total cell count. This interval
indicates a diagnostic window allowing for optimal discrimination between
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 112
GFP
Negative Positive MMC (nM)
100
G1
10
G2
S
0
100
33
Relative number of cells (%)
0
100
66
0
100
100
0
1 512 1,024 512 1,024
Fluorescence intensity
(Channel number)
60
G2 phase (%)
40
20
0
1 10 100 1,000
MMC concentration (nM)
achieved with much lower concentrations, if G2-phase arrest rather than chro-
mosome breakage is measured. This proves the higher sensitivity of the cell
cycle assay compared to chromosome breakage analysis.
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 114
The horizontal axes of the cell cycle distributions displayed in figures 1
and 3 indicate DNA content as measured by the fluorescence intensity of a dye
that binds stoichiometrically to DNA. Thus, in each of these panels the distrib-
utions of cells within the G0/G1, S and G2/M compartments of the cell cycle
are visualized from left to right. Cells in G0/G1 have a 2c DNA content,
whereas cells in G2/M, having completed semiconservative DNA replication,
have a 4c DNA content. G2- and M-phase cells cannot be discriminated on the
basis of DNA content. In order to answer the question whether the cell cycle
arrest in response to MMC treatments affects both the G2- and the M-phase
compartments of the cell cycle, we included phospho-histone H3 (H3P)-staining
as a cellular protein marker, whose expression is limited to the M phase of
the cell cycle [16]. The simultaneous measurement of H3P and DNA content
permits the discrimination between G2- and M-phase cells. As shown by the
framed DNA histogram sections of figure 3, H3P-positive (mitotic) cells repre-
sent only a small proportion of the respective G2/M peaks. Following MMC
treatments, the proportion of H3P-positive cells changes inversely to the
increase of the G2/M peak. The higher the G2/M peak, the smaller the H3P-
positive M fraction. In a series of eight different non-FA control cell lines, we
observed a G2-phase increase of 5.2 ⫾ 1.6% (mean ⫾ 1 SD) and a H3P
decrease of 0.06 ⫾ 0.14%. In contrast, the G2-phase increase for five different
FA cell lines was 15.6 ⫾ 6.2% (mean ⫾ 1 SD) and the H3P decrease
0.49 ⫾ 0.23%. Thus, an approximately 3-fold increase of the G2/M peak
resulted in an approximately 8-fold decrease of the H3P-positive cell popula-
tion. These observations suggest that the MMC-induced accumulation of cells
with a 4c DNA content must occur prior to entry into the M phase.
This interpretation was strengthened by the experiment shown in the bot-
tom panels of figure 3: a non-FA control cell line was exposed to nocadazol, an
agent that binds to tubulin and thereby prevents microtubule protofilament
polymerization and spindle fiber formation. Nocadazol treatment resulted in a
G2/M-peak increase of 23.5%, which is very similar to FA cells exposed to
MMC. Strikingly however, the proportion of H3P-positive cells in the culture
exposed to nocadazol showed an increase of 15.8%, accounting for 2/3 of the
total G2/M-peak increase. In contrast to MMC, nocadazol clearly induces accu-
mulations of cells that have completed G2 and entered the M phase of the cell
cycle.
This series of experiments proves that the MMC-induced accumulation of
cells occurs prior to the M phase, but after completion or near completion of
DNA replication. Otherwise the accumulated cells would not display a 4c
DNA content. These observations localize the putative crosslink-sensitive
checkpoint into the early portion of the G2 phase, presumably close to the
S/G2 border.
80
G1
60 H3P
1.2% 1.1% CON
40
20 G2
0
80
60
Relative number of cells (%)
20
80
60
0.45% 0.15% FA-D2
40
20
0
80
60
1.4% 0.6% FA-D1
40
20
⫺Nocadazol ⫹Nocadazol
100
80
60 0.7%
16.5% CON
40
20
0
0 512 0 512 1,024
Fluorescence intensity
(channel number)
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 116
Bivariate Flow Cytometry Enhances the Resolution of
Cell Cycle Analysis in FA
S S
50 h 55h 60h
G2⬘
S⬘ S⬙
S⬘
G1⬙ G1⬙
2nd cycle
G1⬘
65 h 80h 96h
4th cycle
G2⬙ G2⬙
S⬙ S⬘⬙
G1⬘⬙ G1⬘⬙
3rd cycle
BRDU/HOECHST fluorescence
Fig. 4. Sequence of cell cycle distributions reveals entry and progression of PHA-
stimulated lymphocytes throughout four consecutive cell cycles. Bivariate scatter plots of
single cells, reflecting their Hoechst-33258- (x-axis) and EB- (y-axis) fluorescence intensi-
ties. Aliquots of BrdU-substituted PHA-stimulated lymphocytes from a healthy donor were
harvested at intervals from 35 to 96 h as indicated, stained, and flow measurements were
recorded to reveal static images of the dynamic progression through four consecutive cell
cycles (cycle one, labeled G0/G1, S, G2; cycle two, G1⬘, S⬘, G2⬘; cycle three, G1⬙, S⬙, G2⬙;
cycle four, G1, S). Modified from [28].
second cycle. The third cell cycle is almost complete after 65 h. At 80 h, cells
have visibly progressed into the G1 phase of a fourth cycle (G1) and transi-
tion through this cycle is almost complete at 96 h. As the percentage increment
of BrdU substitution of DNA decreases from cycle to cycle, the additional
Hoechst-quenching effect becomes smaller with each cycle, which limits the
resolution of the BrdU-Hoechst technique to about four rounds of replication.
In addition to showing the distribution of cells throughout up to four cell
cycles within a single cell culture, the BrdU-Hoechst technique permits the
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 118
FA CON
G2⬘
S G2 G2⬘ S G2 Min. duration
100 Arrest Arrest
80 G2⬘
60
G2⬘
Cells (%)
40
G2
20 S G2
S
10
20 40 60 80 20 40 60 80
Time after stimulation (h) Time after stimulation (h)
Fig. 5. Kinetic display of successive cell cycle compartment transitions. The curves
represent fits of semi-logarithmic plots of the cell fractions in PHA-stimulated lymphocyte
cultures remaining in any given compartment as a function of time after culture initiation (␣-
plots according to Smith and Martin, modified as proposed by Rabinovitch [17]). The left
hand graph represents a PHA-stimulated lymphocyte culture derived from an FA patient, the
right hand graph that of a non-FA control. The distances of intercepts of exit curves with the
x-axis (representing minimum S, G2 and G2⬘ durations) are indicated by headline bars,
the corresponding arrest fractions are represented as distances of intercepts of the same
curves with the y-axis. The respective G2-phase compartments are shaded.
quantitative assessment of the fraction of cells that fails to respond to the mito-
gen (G0/G1 cell fraction). Unlike any other technique, the resolution of suc-
cessive cell cycles permits the characterization of disturbances of cell cycle
progression due to endogenous or exogenous influences. How this technique
can be applied to the study of the FA cellular phenotype is illustrated in the fol-
lowing section.
More than 20 years ago, Kubbies et al. [9] first described the cell kinetic
behavior of FA cells as revealed by the BrdU-Hoechst technique. Because of the
unique insights provided by this technique, we briefly summarize the essence of
these types of experiments in figure 5. During the course of a 96-h study of
PHA-stimulated lymphocytes, 12 harvests at 6-h intervals were performed, the
first 24 h after initiation of the culture and the last at 90 h. Each harvest was
recorded in the way shown in the previous section. Data on the fractions of cells
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 120
FA CON
1,024 1,024
G2⬘ G2⬙
G2 G2⬘
G2
512 512
EB fluorescence
EB fluorescence
0 0
0 512 1,024 0 512 1,024
G2⬘
G2 G2⬙
G2⬘
G2
1,024 1,024
0 0
1,024 1,024
a BRDU/HOECHST fluorescence b BRDU/HOECHST fluorescence
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 122
100
⫹4mo
80
⫺1 mo
G0/G1 (%)
60
⫹5 mo
40
20 ⫺39mo
⫹11 mo
0
0 0.2 0.4 0.6 0.8 1.0
sG2 vs GF
Fig. 7. Scattergram summarizing the results of flow cytometric cell cycle analyses of
blood samples submitted to the Würzburg laboratory for the confirmation or exclusion of
FA. 72-h, BrdU-substituted cultures of PHA-stimulated lymphocytes were subjected to
Hoechst-33258- and EB-fluorescence intensity recording. Cell-cycle-resolved cytograms
were electronically deconvoluted for quantitative assessment of each cell cycle compartment
and the growth and resting cell fractions. The ratio ‘sum of all G2 phases vs. growth fraction’
(sG2 vs. GF) was calculated and plotted against the G0/G1 fraction, with the latter indicating
the degree of lymphocyte activation. The panel represents 394 FA (red diamonds) and 630
non-FA (green circles) cases. Means of either group ⫾ 1 SD are denoted by the large dia-
mond and circle symbols and are given in the text. Blue squares denote successive cell cycle
assays of an FA patient prior to and after successful HSCT.
threshold empirically set at an sG2/GF of 0.285. The few FA symbols that are
scattered in the immediate vicinity below and above this threshold represent
either proven or likely mosaic cases that consist of a mixture of MMC-sensitive
and MMC-resistant (⫽reverted) cells, or they comprise cases with what we
consider ‘mild’ mutations, i.e. retention of partial protein function. With respect
to the sG2/GF parameter, the mean ⫾ 1 SD amounts to 0.174 ⫾ 0.035 for the
non-FA samples, whereas the mean ⫾ 1 SD of the FA samples is 0.456 ⫾ 0.098
(p ⬍ 0.001). Again, these cultures were not treated with MMC such that these
results would be comparable to the analysis of spontaneous chromosome break-
age rates, which, in contrast, are known to vary greatly and which are generally
judged to be of limited diagnostic value. Bivariate flow cytometry of cultures
exposed to MMC provides even better separation with the FA and non-FA clus-
ters drifting further apart (data not shown). Parallel exposure of cultures to
MMC is always included in diagnostic studies, and such additional MMC test-
ing is particularly helpful in cases of mild FA mutations or in mosaic cases (see
contribution by Hoehn et al.).
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 124
Clinically suspected diagnosis of FA
Fibroblast cultures
DNA
in case of continued clinical
RNA
suspicion
Unsuccessful: Successful:
FANCD2 immunoblot mutation analysis
60
Relative number of cells (%)
G2
40
20
S
0
E F G L
80
60
40
20
0
0 512 0 512 0 512 0 512 1,024
Fluorescence intensity (channel number)
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 126
Concluding Remarks
Cell kinetic studies such as originally performed by Kubbies et al. [9] have
shown that the cell cycle disturbance of FA cells consists of both, delay and
arrest within the S and G2 phases of the cell cycle. Flow cytometric measure-
ments uniformly reveal a 4c DNA content of the accumulated cells. Since it has
become clear that defective FA proteins cause accumulation of ICLs and
replication-associated DNA-double strand breaks [21, 22], transient or permanent
delay of cell cycle progression appears as the logical consequence for cells that
have acquired such lesions. There are some seemingly contradictory findings as
to where precisely within the cell cycle the accumulation of damaged cells takes
place. When FA cells were treated with DNA-crosslinking agents after comple-
tion of DNA replication, i.e. during the G2 phase of the cell cycle, Akkari et al.
[23] observed neither G2/M arrest nor increased chromosome breakage. The
authors therefore concluded that crosslink-induced accumulations of FA cells
take place in the late S phase, even though these cells have a 4c DNA content,
which formally classifies them as having entered the G2 phase of the cell cycle.
Since the early cell kinetic studies by Kubbies et al. [9] clearly showed delay
and arrest during both, the S and G2 phases of the cell cycle, and since cell
cycle studies uniformly prove the 4c DNA content of cells that accumulate in
response to crosslinking agents, is comes down to a more or less semantic argu-
ment whether to call these accumulations late S or early G2. There is complete
agreement, however, that the accumulations take place prior to entry into the M
phase of the cell cycle, and the data summarized in figure 3 provide proof for
this conclusion. Our demonstration that M-phase cells accumulate in response
to a spindle damaging, but not in response to a DNA-crosslinking agent clearly
separates the M- and S/G2-phase checkpoints in FA cells. These observations
leave little doubt that the S/G2-phase checkpoint functions normally in FA
cells, as previously demonstrated by the careful studies of Heinrich et al. [14]
and Freie et al. [24]. As pointed out by these authors, spontaneous or crosslink-
induced accumulations of FA cells in the G2 phase of the cell cycle do not
reflect an abnormal cell cycle response per se, but rather represent a completely
normal cellular response to unresolved DNA damage. Normal function of the
S/G2 checkpoint is critical for the prevention of apoptosis or the erroneous re-
replication of DNA [25]. Most importantly, S/G2 checkpoint-mediated arrest of
damaged cells prevents their entry into mitosis, sets the stage for homology-
directed DNA repair, and thereby minimizes the undesired perpetuation of
genetic lesions.
In terms of diagnostic applications, we have outlined our approach and
summarized our experience with cell cycle analysis as an alternative to traditional
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 128
Acknowledgements
We are grateful to Birgit Gottwald and Gitta Emmert, Würzburg, for dedicated cell cul-
ture work. We recognize and appreciate the long-standing and invaluable support by Peter S.
Rabinovitch, M.D., Ph.D., Seattle, with mathematical approaches to cell cycle models and
software development.
References
Detlev Schindler
Department of Human Genetics, University of Würzburg
Biozentrum, Am Hubland, D–97074 Würzburg (Germany)
Tel. ⫹49 931 888 4089, Fax ⫹49 931 888 4069
E-Mail [email protected]
Schindler/Friedl/Gavvovidis/Kalb/Neveling/Linka/Hanenberg/Kubbies/Hoehn 130
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 131–148
Abstract
There are two main approaches to the prenatal confirmation or exclusion of Fanconi
anemia: functional testing and molecular testing. Functional testing involves the determina-
tion of crosslink sensitivity either by chromosome breakage analysis or cell cycle testing.
Indications for functional testing include ultrasonographic findings of radial ray defects in
the absence of a family history of FA, but also testing of at risk pregnancies in families with
a prior affected child where for various reasons there is no information on complementation
group and disease causing mutations. Since laboratories offering functional prenatal testing
mostly use analysis of chromosome breakage, we here summarize our experience with flow-
cytometric testing of MMC-sensitivity in second trimester amniotic fluid cell cultures. We
show that among a series of 20 pregnancies at risk three amniotic fluid cell cultures were
highly sensitive to MMC as evidenced by their strong G2-phase elevations after exposure to
10 ng/ml of the drug. There were no false positives and no false negatives among our series
suggesting single parameter flowcytometry as a speedy and reliable alternative to conven-
tional chromosome breakage studies for the prenatal diagnosis of FA in situations where only
functional testing can be performed. Molecular testing of course is the method of choice but
requires prior knowledge of complementation group and mutations. Indirect genetic testing
is possible if at least the complementation group is known and DNAs from both parents and
an affected child are available. With the availability of retroviral vectors for rapid subtyping,
and owing to advances in high-throughput mutation analysis including MLPA, direct molec-
ular genetic testing is likely to replace functional testing for most but not all risk pregnancies
in the near future. We illustrate the practice of direct prenatal genetic testing with examples
from families belonging to complementation groups FA-A, FA-C, FA-G and FA-D2. Last but
not least we comment on the implications of preimplantation genetic testing (PGD) as a
high-tech but problematic procedure to preselect potential HLA-matched sibling donors.
Copyright © 2007 S. Karger AG, Basel
Fanconi anemia (FA) is an inherited disease that is in principle amenable to
symptomatic and curative treatment. However, present day small families often
lack a matching sibling donor for lifesaving hematopoietic stem cell transplanta-
tion, and alternate donor transplants still carry a relatively high risk. Squamous
cell carcinomas remain a lifelong threat. Despite remarkable therapeutic progress
during recent years, Fanconi anemia still remains a serious and threatening dis-
ease. There are major impairments of the quality of life for the patients them-
selves, but also for their siblings and families [1]. Because of the severity of the
disease, the demanding care, and the limitations of therapeutic interventions par-
ents often feel overwhelmed and unable to care for a second or third affected
child. Given a 25% recurrence risk and the possibility of prenatal diagnosis, a
number of parents decide to use this option in their family planning. In contrast to
many other medical interventions, prenatal diagnosis is not a routine procedure
without impact on the physical and psychological well-being of patients [2, 3]. In
the case of Fanconi anemia use of prenatal diagnosis is a reliable but desperate
option in the face of our inability to provide a comprehensive cure for the disease.
The first prenatal diagnosis of FA was reported by Arleen Auerbach and her
coworkers in 1979 who treated amniotic fluid cell cultures from at risk pregnancies
with diepoxybutane (DEB) and studied chromosome breakage [4, 5]. Increased
breakage rates were observed in affected fetuses and also in heterozygotes, but
postnatal confirmation was obtained in only a single case [6]. Increased breakage
rates of chromosomes prepared from amniotic fluid cell cultures that had been
exposed to DEB were also reported in an affected fetus from South Africa [7]. In a
subsequent series, Auerbach et al. did chromosome breakage studies in 30 preg-
nancies at risk for FA and were able to confirm the correctness of their prenatal
findings by postnatal clinical and cytogenetic analyses [8]. This landmark study
demonstrated that the chromosome breakage test for the confirmation or exclusion
of FA could be used with amniotic fluid cell cultures much in the same way and
with similar high degrees of sensitivity and specificity as had been shown and put
into clinical practice for postnatally derived peripheral blood mononuclear cells.
The first prenatal diagnosis using fetal blood was reported by Shipley et al.
[9]. The umbilical cord blood mononuclear cells of the affected fetus showed
strongly elevated spontaneous and MMC-induced breakage rates. In 1985,
Dallapiccola et al. [10] reported the first prenatal diagnosis using cells from a
chorionic villus biopsy (CVS) with fetal trophoblast cells showing elevated
chromosome breakage rates following DEB treatments. The usefulness of CVS-
derived materials for the very early prenatal diagnosis of FA was quickly confirmed
by French and American investigators [11, 12]. By that time, ultrasonography
had become a standard monitoring procedure in prenatal care. A French group
reported a family with three affected children one of whom had been diagnosed
prenatally because of radial ray defects and elevated chromosome breakage rates
Depending on the family situation, there are two main categories of fami-
lies requesting prenatal confirmation or exclusion of FA. The first category
comprises pregnancies in which radial ray defects are observed as incidental
findings on routine ultrasonographic examination. Even though such radial ray
anomalies can be associated with a myriad of syndromes [19], this type of
anomaly may of course be an important clinical sign of FA. Increased nuchal
translucency as an ultrasonographic sign of an affected fetus has also been
reported [20], as has been abnormal fetal motor behavior [21]. In these situa-
tions, there is no prior evidence for FA in the family, and the only practical way
for confirmation or exclusion of FA is to carry out functional testing of fetal
cells. Functional testing is also required in families with a prior affected child
who had deceased or families who lack information on complementation group
and mutation. To arrive at a timely molecular diagnosis may also be difficult in
pregnancies where parents are not available for testing or in families where the
disease causing genes are large and rather time consuming to type (e.g. FANCA
or FANCD2). Subtyping and mutation analysis during the course of an ongoing
pregnancy has so far only been reported in two families assigned to subtype
FA-C, with FANCC being a relatively small gene where carrier testing and finding
the disease causing mutations is relatively straightforward [22]. The second cat-
egory, and of course optimal situation for the prenatal exclusion or confirma-
tion of FA comprises families with prior affected children in whom the affected
gene and both disease causing mutations are known. In such families, fetal
DNA can be obtained via chorionic villus biopsy early in the course of preg-
nancy, and the results of the molecular analysis, including testing for maternal
cell contamination, will be available within days [18, 23]. With the availability
of rapid subtyping using retroviral vectors [24] and owing to technical advances
of mutation analysis, including high throughput methods and MLPA, prenatal
molecular genetic testing is expected to replace much of the present functional
testing and thus be available for the majority of FA families in the near future.
50 G2
G1 G2
S
0
Relative number of cells (%)
100 c d
MMC MMC
50
G2
G2
0
100 e f
MMC MMC
50 G2
G2
0
1 512 1,024 512 1,024
DNA content
their untreated as opposed to their treated amniotic fluid cell cultures. These
three cases were confirmed as affected either subsequent to termination of
pregnancy or after birth. The data summarized in figure 2 illustrate two further
points: (1) The G2-phase cell fractions of untreated cultures vary considerably,
presumably due to different growth kinetics and variable amounts of G1-phase
tetraploid cells that occur rather frequently in second trimester amniotic fluid
cell cultures. (2) There is an impressive range of the degrees of MMC-response
among FA-negative cell cultures, with cases number 8 and 10 showing no
response at all, but cases 1 and 18 showing some minor degrees of MMC-
sensitivity which, however, is far below of what is observed in affected cases. In
the series of prenatal MMC sensitivity testing summarized in figure 2 there
were no false positive and no false negative cases, suggesting that a reliable
FA
19
17
15
Family number
13
11 FA
9
7
5 FA
3
1
0 10 20 30 40 50 60 70
Cells in G2 phase (%)
As pointed out before, molecular testing is the method of choice for the
prenatal diagnosis of FA, provided that the affected gene and both mutations are
known. If only the complementation group is known, molecular testing can also
be applied as long as DNA from both parents and an affected child is available.
With the rapidly increasing catalogue of flanking and intragenic polymorphic
markers, indirect genotyping has turned into a practical and accurate tool in sit-
uations in which the affected gene is known but the disease causing mutations
remain to be detected. This is especially true for large and polymorphic genes
such as FANCA [32].
Indirect Genotyping
Two children of family S. suffered from FA and their complementation
group was determined as FA-A via retroviral gene transfer. The causal muta-
tions in FANCA, however, were still unknown at the time when a third preg-
nancy was under way. Indirect genotyping with microsatellite markers flanking
D16S413 137 137 137 137 137 143 Exon 16: G501S
4cM D16S3121 79 77 79 77 79 83
FANCA Exon 22: P643A
1cM
D16S3407 198 198 198 198 198 198
D16S303 121 119 121 119 121 121 Exon 40: T1328A
Fig. 3. Prenatal diagnosis via indirect genotyping in family S. with two affected girls.
Left panel: genotyping using microsatellite markers flanking FANCA. Right panel: genotyp-
ing using intragenic single nucleotide polymorphisms. Fetus at risk marked by arrow.
the FANCA gene was carried out using DNA of both parents, the affected chil-
dren, and fetal DNA obtained via chorionic villus biopsy. Using the markers
D16S413, D16S3121, D16S3407, and D16S303 haplotypes were constructed
encompassing the FANCA locus on chromosome 16. As shown in the left panel
of figure 3, the fetus was found to carry the defective paternal but the wildtype
maternal allele, identifying the fetus as heterozygous carrier not being affected
by FA.
The prediction of an unaffected fetus via microsatellite haplotyping was
confirmed by the analysis of intragenic single nucleotide polymorphisms of
FANCA (fig. 3, right panel). Informative polymorphisms were G501S in exon 16,
P643A in exon 22, and T1328A in exon 40. The parents were heterozygous for
one respectively two of these variants. The fetus was shown to carry the wild-
type maternal and the mutated paternal allele. Because of the concordance of
the results of indirect genotyping with both flanking and intragenic markers, the
fetus was confirmed as an unaffected carrier, and there were no signs of FA
after birth.
Direct Genotyping
Prenatal Diagnosis Involving FANCA: The first child of family V. had
been diagnosed with FA because of typical congenital malformations, aplastic
anemia and elevated chromosome breakage. The child had already died by the
1 2 3 4
time of the second pregnancy, a dizygotic twin pregnancy, for which the parents
requested prenatal diagnosis. Using parental blood and frozen materials from
the deceased child, molecular analysis led to the diagnosis of compound het-
erozygosity for the following mutations in FANCA: a paternal 2-bp deletion at
position 1165 in exon 13 leading to a frameshift, and a maternal 18-bp deletion
within intron 40 affecting splicing (4010delG18), resulting in skipping of
exon 40. In the DNA prepared from chorionic villus biopsies of both twins
there was no evidence for either of the parental mutations indicating that the
twins were homozygous for the FANCA wildtype alleles. In order to exclude the
risk of double puncture of the same chorion in dizygotic twins seven polymor-
phic DNA markers were analyzed. The twins were found to differ in six out of
seven markers such that an erroneous puncture could be excluded. In summary,
family V. could be told that neither of their unborn twins was carrier of a
parental FANCA mutation, which excluded their being affected by FA. At birth,
the fraternal twins were healthy and free of signs of FA.
Prenatal Diagnosis Involving FANCG: As shown in the family pedigree
(fig. 4), the first child of family A. had been diagnosed with FA. During the sec-
ond pregnancy amniotic fluid cells were analyzed in an outside laboratory and
chromosome breakage rates were reported as suggestive of FA, even though the
number and quality of the available metaphases was reported as too low for a
reliable diagnosis. Complementation analysis with retroviral vectors assigned
the amniotic fluid cells to complementation group FA-G, and mutation analysis
detected a homozygous 1-bp deletion in exon 13 (1649delC). Homozygosity
was expected because of consanguinity. The mutation leads to a frameshift and
an unstable protein as described in the literature [33]. After induced abortion the
diagnosis was confirmed via flow cytometric measurement of lymphocytes from
Fetus
homozygous
affected
Mother
heterozygous
Father
heterozygous
Fig. 5. Partial sequence of FANCC exon 4 of parents and fetus of family I. Both parents
are heterozygous carriers of an identical 2-bp deletion within exon 4. The fetus was found to
be homozygous for the defective parental allele and thus will be affected.
Fetus
Exon 21 Exon 23
Index patient
Fig. 6. Partial sequence of exons 21–23 of amniocytes (upper panel) and blood lym-
phoblasts (lower panel) showing homozygous skipping of exon 22 in the index patient of
family G. and the heterozygous nature of the mutation in amniocytes of the tested fetus. Note
slight background of exon 22 signals in the index patient.
Maternal mutation
(IVS4-57ins298)
Exon 34
Paternal mutation
(c.3453delCAAA)
mutations. In view of the result of the genetic testing the parents opted for ter-
mination of the pregnancy.
Acknowledgements
The authors wish to thank the Fanconi Anemia families requesting prenatal diagnosis
for their patience and cooperation. Special thanks go to Ralf Dietrich of the German family
support group for sharing information on the experiences and attitudes of American FA fam-
ilies with PGD.
References
1 Hutson SP, Alter BP: Experiences of siblings of patients with Fanconi anemia. Pediatr Blood
Cancer 2006;48:72–79.
2 Schroeder TM: Ethical conflicts in genetic counselling during pregnancies (author’s transl.).
Monatsschr Kinderheilkd 1982;130:71–74.
3 Auerbach AD: Umbilical cord blood transplants for genetic disease: diagnostic and ethical issues
in fetal studies. Blood Cells 1994;20:303–309.
4 Auerbach AD, Warburton D, Bloom AD, Chaganti RS: Preliminary communication: prenatal
detection of the Fanconi Anemia gene by cytogenetic methods. Am J Hum Genet 1979;31:77–81.
5 Auerbach AD, Adler B, Chaganti RS: Prenatal and postnatal diagnosis and carrier detection of
Fanconi anemia by a cytogenetic method. Pediatrics 1981;67:128–135.
6 Voss R, Kohn G, Shaham M, Benzur Z, Arnon J, et al: Prenatal diagnosis of Fanconi anemia. Clin
Genet 1981;20:185–190.
7 Marx MP, Dawson B, Heyns AD: Prenatal diagnosis of Fanconi’s anemia. S Afr Med J 1982;62:
348.
8 Auerbach AD, Sagi M, Adler B: Fanconi anemia: prenatal diagnosis in 30 fetuses at risk.
Pediatrics 1985;76:794–800.
9 Shipley J, Rodeck CH, Garrett C, Galbraith J, Giannelli F: Mitomycin C induced chromosome
damage in fetal blood cultures and prenatal diagnosis of Fanconi’s anaemia. Prenat Diagn 1984;4:
217–221.
10 Dallapiccola B, Doria Lamba Carbone L, Ferranti G, Cristiani ML, Dagna Bricarelli F:
Monitoring of pregnancies at risk for Fanconi’s anemia by chorionic villi sampling. Acta Haematol
1985;73:157–158.
11 Boue J, Deluchat C, Nicolas H, Oury JF, Dumez Y: Prenatal diagnosis of genetic diseases using
chorionic villi. J Genet Hum 1986;34:221–233.
12 Auerbach AD, Min Z, Ghosh R, Pergament E, Verlinsky Y, Nicolas H, Boue J: Clastogen-induced
chromosomal breakage as a marker for first trimester prenatal diagnosis of Fanconi anemia. Hum
Genet 1986;73:86–88.
13 Bonnet-Gajdos M, Vasmant D, Baruchel S, Mamou JF, Boue J, Lasfargues G: Prenatal diagnosis
of Fanconi’s anemia. Presse Med 1985;14:1612.
Abstract
One out of four to five patients with Fanconi anemia experience a reversion or attenu-
ation of their constitutional mutations during their lifetime. If the reversion event takes place
in a bone marrow stem cell or in an early precursor cell of hematopoiesis, peripheral blood
cell counts may gradually recover, leading to improved quality of life. At the beginning of
this process, MMC testing will reveal a mixture of MMC sensitive and MMC resistant blood
lymphocytes, but after several years MMC sensitive cells (the original FA-cells) may be com-
pletely replaced by the progeny of the reverted progenitor cell such that the confirmation of
FA requires testing of patient fibroblasts. Molecular analysis reveals the presence of the dis-
ease causing biallelic mutations in fibroblast-derived DNA whereas MMC-resistant blood
cells show only a single defective allele, explaining their phenotypic reversion. The mech-
anisms leading to revertant mosaicism include intragenic mitotic recombination (crossing
over and gene conversion), back mutation, and compensatory second site mutations.
Evidence for each of these mechanisms has been obtained in mosaic FA-patients, but their
molecular details are not fully understood. Compound heterozygosity facilitates some of
these mechanisms, but reversions have also been observed in homozygous patients. Patients
belonging to subtypes FA-A, FA-C, FA-D1, FA-D2 and FA-L have developed revertant
mosaicism, with subtypes FA-A and FA-D2 being most frequently involved. Even though the
phenomenon of revertant mosaicism has been well documented in FA, there still are many
questions: we do not know whether the progeny of a single reverted blood stem or progenitor
cell would be able to sustain lifelong hematopoiesis, whether revertant mosaicism provides
protection against hematopoietic malignancy, or whether it would be possible to deliberately
increase the rate of somatic reversions in order to improve chances for ‘natural gene therapy’.
Prospective and long-term follow-up studies are needed to answer these questions.
Copyright © 2007 S. Karger AG, Basel
The restoration to normal (or close to normal) function of a gene previ-
ously inactivated by a constitutional mutation is referred to as ‘reversion’. The
coexistence of reverted and wildtype cells within otherwise isogenic organisms
is referred to as ‘reverse’ or ‘revertant’ mosaicism. In contrast to Lo ten Foe
et al. [1] we prefer the term ‘revertant’ which has been defined as ‘a mutant
which has regained, partially or completely, the wildtype phenotype by either a
genetic or nongenetic mechanism of reversion’ [2]. As pointed out by Jonkman
et al. [3], the term ‘reverse’ simply implies a change in the opposite direction,
but does not necessarily denote complete or partial restoration of a defective
function. The phenomenon of revertant mosaicism has been observed in a num-
ber of inherited diseases, including a skin disease (epidermolysis bullosa), a
metabolic disease (tyrosinemia type I), a number of primary immunodeficiency
syndromes (Wiskott-Aldrich syndrome, X-linked severe combined immuno-
deficiency, ADA-deficiency), and in two chromosomal instability syndromes
(Bloom syndrome and Fanconi anemia) (see reviews by Youssoufian and
Pyeritz [4] and Hirschhorn [5]).
In the case of FA, revertant mosaicism has both clinical and theoretical
implications [1, 6]. These implications relate to the altered in vivo behavior of the
reverted cells and depend on the developmental stage, the blood cell lineage
affected by the reversion event, and the clinical situation of the mosaic patient. In
self-renewing tissues, the reversion event may convey a growth advantage to the
reverted cells such that their progeny may outgrow and, ultimately, replace the
defective cells. Such a positive outcome would suggest that gene therapy might be
successful in FA and other stem cell diseases. Because of these positive prospects,
revertant mosaicism has also been referred to as ‘natural gene therapy’ [3, 6–9].
As early as in 1983, the group of Hans Joenje in Amsterdam noted an unusual
response to bifunctional alkylating agents in a 22-year-old patient with Fanconi
anemia. Unexpectedly, only 40% of the patient’s peripheral blood mononuclear
cells proved highly sensitive to these agents as evidenced by strongly increased
chromosome breakage while 60% of the patient’s cells responded like cells
from healthy individuals [10]. The patient’s advanced age and the observation
of two distinctive peripheral blood cell populations with striking differences in
crosslink sensitivity leave little doubt that this patient represents one of the first
well-documented cases of revertant mosaicism in Fanconi anemia.
At the same time, other authors noted that among clinically diagnosed FA
patients between 1 to 3 out of 9 tested blood samples proved MMC- or DEB-
resistant [11, 12]. There was some concern that loss of activity during storage of
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 150
the DEB or MMC stock solutions could give rise to false-negative chromosome
breakage tests [13], but it was generally felt that a subgroup of patients with
clinical manifestations of FA might lack the FA-typical pattern of crosslink sen-
sitivity. The existence of such a subgroup was convincingly documented by
Arleen Auerbach and Traute Schroeder-Kurth in their first report on the
International Fanconi Anemia Registry (IFAR) [14]. Among a series of 162 FA
patients Auerbach and colleagues noted 7 patients who appeared to have two
populations of cells. 60–81% of DEB-treated cells from these patients dis-
played no evidence for chromosome breakage, while the remainder exhibited
high numbers of breaks and exchanges typical of FA cells. Since three of these
cases were from multiplex sibships, the authors came to the conclusion that the
‘phenomenon of two-cell populations does not seem to be related to genetic
heterogeneity in FA’ [14]. Clearly, what the authors had observed but not recog-
nized as such at that time was the phenomenon of revertant mosaicism.
Laboratories which routinely established lymphoid cell lines from FA patients
noted that around 30% of such EBV transformed cell lines that originate from B-
lymphocytes became crosslink resistant during the course of prolonged in vitro
culture. However, it was not until 1997 when a landmark study by the Amsterdam
group confirmed that between 20 and 30% of FA patients show evidence for a
mixture of MMC-sensitive and MMC-resistant peripheral blood lymphocytes [1].
Molecular analysis of these two types of cells provided unambiguous proof of the
concept of revertant mosaicism. In the study by Lo ten Foe and colleagues [1] the
expected biallelic mutations in a given FANC gene were present in DNA from the
MMC-sensitive subpopulation, whereas the MMC-resistant cells were found to
carry only a single defective allele. Since in recessive diseases a single intact copy
of a disease gene suffices for the maintenance of function, the study by Lo ten Foe
et al. [1] provided molecular proof of rescue of defective gene function via somatic
reversion of a constitutional mutation. In the same year, Jonkman and coworkers
published their landmark study on somatic reversion as molecular cause of mRNA
rescue in epidermolysis bullosa, suggesting that gene conversion might be the
mechanism by which the defective function of one of the collagen XVII alleles had
been restored in unaffected skin areas of their patient [15].
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 152
100
0 ng 0ng 0ng 0ng 0ng 0ng
80
60
40
20
0
100
50ng 50ng 50ng 50 ng 50 ng 50 ng
80
60
40
20
0
100
100ng 100ng 100ng 100ng 100ng 100ng
80
60
40
20
0
0
1
2
3
4
5
6
7
8
ⱖ9
10
0
1
2
3
4
5
6
7
8
ⱖ9
10
0
1
2
3
4
5
6
7
8
ⱖ9
10
0
1
2
3
4
5
6
7
8
ⱖ9
10
0
1
2
3
4
5
6
7
8
ⱖ9
10
0
1
2
3
4
5
6
7
8
ⱖ9
10
a b c d e f
Fig. 1. Chromosomal breakage testing of 72-h peripheral blood mononuclear cell cul-
tures without (top row) and with (middle row, 50 ng/ml; bottom row, 100 ng/ml) mitomcycin
C. (a) MMC-resistant: non-FA; (b) intermediate type of MMC sensitivity: questionable diag-
nosis of FA; (c) MMC-sensitive: FA-positive; (d–f) intermediate types of MMC sensitivity:
different grades of FA-mosaicism.
60 60
40 40
20 20
0 0
a 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 b 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8
100 100
80 80
60 60
40 40
20 20
0 0
c 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 d 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 154
analysis of 72-h peripheral blood mononuclear cell cultures [24] FA-negative
samples display a sum of G2/GF ratios below 0.3 while FA-positive samples
have a sum of G2/GF ratios higher than 0.3, reflecting their FA-typical
G2-phase arrest (see contribution by Schindler et al.). As shown in figure 2a,
mosaicism usually develops slowly over a time course of several years, as evi-
denced by the gradual shift of the sum of G2/GF parameter from above to below
the 0.3 threshold. In the example shown in figure 2b, the first measurement in
1997 suggested nearly complete mosaicism, but subsequent measurement in
2004 and 2005 provided evidence for a persisting mixture of FA-positive
and FA-negative cells. Such fluctuations over time are also documented in
figure 2c, where the measurement 1/2004 yielded a majority of FA-negative,
and the repeat measurement only 4 months later (5/2004) a majority of FA-
positive cells.
The example illustrated in figure 2d implies progression of low-grade
mosaicism to complete mosaicism within a time interval of little over a single
year, with evidence for complete replacement of the original FA cells by
reverted cells. Unless such a patient has been followed with repeat investiga-
tions over time, he or she can no longer be recognized as FA-positive by a one
time chromosome breakage or cell cycle study. If in such instances the labor-
atory study yields a normal result but the possibility of FA persists on clinical
grounds, examination of DEB- or MMC-sensitivity of patient fibroblast
cultures is the only way to arrive at a correct diagnosis [25].
50
40
30
20
Number of cells
10
0
70
60 FA
50
40
30
20
10
0
1 2 3 4 5 6
Days after seeding
Fig. 3. Fibroblast growth assay without (black bars) and in the presence of 1g/ml
(light grey bars) or 3 g/ml mitomcyin C (dark grey bars). Upper panel: control culture from
a non-FA individual; lower panel: fibroblast culture derived from an FA patient. Bars indicate
means and SD of daily triplicate cell counts.
patient derived fibroblast cultures, but simple cell counts such as shown in
figure 3 are the easiest way to confirm or exclude an FA cellular phenotype.
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 156
In most of the cases of revertant mosaicism reported in the literature, molecular
confirmation of the somatic reversion event has been achieved by comparison
of the respective types of mutations found in lymphocyte and fibroblast DNA of
a given patient. This assumes that fibroblast-derived DNA reflects the constitu-
tional pattern of mutations, whereas lymphocyte-derived DNA may have been
altered by somatic mutations. So far there is no evidence that would contradict
the assumption of relative stability of the fibroblast as opposed to the clonally
expanding lymphocyte cell pool, notwithstanding the fact that in certain disease
situations there can be clonal expansion of skin cell populations [3]. Molecular
confirmation of revertant mosaicism has so far been documented mostly for the
FANCA and FANCC genes (cf. table 1) which prompts us to illustrate the mole-
cular confirmation of revertant mosaicism in two mosaic patients belonging to
complementation group FA-D2 [28].
Patient code FANC Allele 1 Allele 2 Reversiona Probable Revertant Cellular Ref.a
Genes (target) mechanisma cellsa phenotypea
URD A 3976C⬎T 856C⬎T 856C (WT) back mutation PBLs, LCL mild pancytopenia [8]
STT A intragenic 862G⬎T 862G (WT) back mutation PBLs, LCL mild pancytopenia [8]
deletion
MRB A IVS9-1G⬎T 971T⬎G 971T (WT) back mutation PBLs, LCL mild pancytopenia [8]
EUFA704 A 1615delG 1615delG 1637delA/ comp. mutation PBLs, LCL NR [40]
1641delT
FA67 A 3720-3724del 2546delC 2546C⬎T comp. mutation granulocytes, mild pancytopenia [18]
mononuclear
phagocytes,
LCL
FA98 A 3720-3724del 2546delC 2546C⬎T⫹ comp. mutation PBLs progressive [18]
3720-3724del pancytopenia
NR A 790C⬎T 2585delCT 2585CT gene conversion PBLs, LCL normal CBC [19]
PD839 A 2555⌬T 2670G⬎A 2927G⬎A comp. mutation PBLs, LCL normal CBC [9]
PD845 A 2555⌬T 2670G⬎A 2927G⬎A comp. mutation PBLs, LCL normal CBC [9]
IFAR557-2 A genomic 2815-2816 2815-2816ins back mutation PBLs, LCL mild [6]
deletion at ins19 19 (WT) stem cells pancytopenia
3⬘ end of gene progression/
clonal
evolution
EUFA393 A 3559insG 3559insG 3580insCG comp. mutation PBLs and LCL NR [40]
CTG or 3576
insGCTGC
158
EUFA173 A Del exon 2852G⬎A 2852G (WT) back mutation PBLs and LCL, mild pancytopenia [8]
17-31 respectively
Revertant Mosaicism in Fanconi Anemia: Natural Gene Therapy at Work
EUFA806 C 67delG 1806insA heterozygote mitotic crossover PBLs, LCL normal CBC [1]
segregants
EUFA449 C L554P 67delG 67delG (WT) gene conversion PBLs, LCL mild pancytopenia [1]
RNT C 67delG IVS11- heterozygote mitotic crossover PBLs, LCL progressive [8]
2A⬎G segregants pancytopenia
EUFA506 C 1749T⬎G 1749T⬎G 1748C⬎T comp. mutation PBLs, LCL NR [40]
FA-AML1 D1 8415G⬎T 8732C⬎A 8731T⬎G comp. mutation Leukemic cells resistance to MMC [41]
FAD2-3 D2 1948-16T⬎G 1948-16T⬎G c.1954G⬎A comp. mutation PBL, LCL, resistance to MMC [28]
bone marrow
FAD2-14 D2 1948-6C⬎A 2775_2776 2776CC (WT) back mutation/ PBL, LCL resistance to MMC [28]
CC⬎TT conversion
FAD2-15 D2 1948-6C⬎A 2775_2776 (heterozygote (mitotic PBL resistance to MMC [28]
CC⬎TT segregants?) crossover?)
FAD2-26 D2 3803G⬎A 376A⬎G c.376A (WT) back mutation PBL, LCL resistance to MMC [28]
FAL-1 L 891C⬎G 483delATCAC c.472-2A⬎G comp. mutation PBL, LCL, BM resistance to MMC [28]
EUFA N exon 4 exon 4 stop in-frame fusion comp. mutation LCL resistance to MMC [34]
1341 (R) deletion codon of exons 3 and (Alu-mediated
mutation 5; elimination recombination)
of exon 4
a
BM ⫽ bone marrow cells; CBC ⫽ comprehensive blood counts; Comp. mutation ⫽ compensatory or second site mutation; LCL ⫽ Lymph-
oblastoid cell line; MMC ⫽ mitomycin C; NR ⫽ not reported; PBL ⫽ peripheral blood lymphocytes; Ref. ⫽ reference; WT ⫽ wild type. Tentative
interpretations are shown in parentheses.
159
gDNA cDNA
Exon 29 Exon 29
c.2775_2776CC>TT c.2775_2776CC>TT
a b
Exon 29 Exon 29
c d
Fig. 4. Partial sequence of exon 29 of FANCD2 at both gDNA and cDNA levels.
Panels a and b show the sequence obtained from fibroblasts (constitutional mutations).
Panels c and d depict the sequence obtained from lymphoid cells that had reverted to MMC-
resistance.
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 160
gDNA cDNA
a b
Intron 21 Exon 22 Exon 23 Exon 22
Exon 21
C T T T T TGT TGGT T TGCT TCC TGA AGGAA TGGGTTGGGCA T A GG A A A T GG A A A G T C A CC T T NCNG A A C A A C A C
230 240 250 260 180 190 200 210
c.1948-16T>G
c.1954G>A
c d
Fig. 5. Partial sequence of FANCD2 extending from intron 21 through exon 23 illus-
trating heterozygous restoration of exon 22 sequence in patient lymphocytes. The best
sequencing result showing reappearance of exon 22 sequence was obtained by use of reverse
primers, which explains the reverse sequence orientation in panel d.
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 162
6 200
15
4
11
120
3
9
80
7 2
Hb WBC Platelets
5 1 40
19 8
19 0
92
19 4
19 6
98
00
88
19 0
92
19 4
19 6
98
00
88
19 0
92
19 4
96
20 8
00
8
9
9
9
9
9
9
a b c
19
19
20
19
19
19
20
19
19
19
19
Reversion of 2852G>A in:
Granulocyte T-lymphocyte B-lymphoblast, new
d e f
Fig. 6. Evidence for phenotypic (panels a–c) and genotypic (panels d–f ) reversion of
the constitutional mutation 2852G⬎A in patient EUFA173 (cf. table 1). The horizontal axis in
panels a–c denotes the years 1988–2000 during which time the recovery of blood counts took
place. Panels d–f illustrate FANCA sequence stretches containing the 2852G⬎A change.
was obtained for the other cell lineages tested, assigning the reversion event to
at least an early hematopoietic precursor, if not to a stem cell.
G1 S G2
a b DNA content c
Fibroblast Blood BFU-E
967 A C C C T /G G A C T A C C C T /G G A C T A C C C T /G G A C T 975
323 Thr Lou/Arg Thr Thr Lou/Arg Thr Thr Lou/Arg Thr 325
d e f
18 240
Hemoglobin (g/dl)
16
Thrombocytes
180
(1,000/l))
14
120
12
10 60
8 0
0 1 2 3 4 5 0 1 2 3 4 5
g Time after diagnosis (y) h Time after diagnosis (y)
Fig. 7. Assignment of patient MRB to complementation group FA-A via retroviral com-
plementation (panels a–c). Panels d–f: evidence for multilineage reversion (e and f) of the con-
stitutional base substitution 971T⬎G present in fibroblast-derived DNA (d). Improvement of
blood counts was noted at 3 years after the patient had been diagnosed with FA (panels g and h).
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 164
Molecular Mechanisms of Somatic Reversion
c d c d c d d c
1 2 3 4
and the mitotic recombination events might have been facilitated by the mutu-
ally distant locations of the respective paternal and maternal mutations.
Gene conversion is another conservative mechanism of mitotic recombi-
nation without, however, mutual strand exchange. Proof of such locus-restricted
recombinational events requires the analysis of polymorphic markers flanking
the mutation which retain their parent-of-origin specific patterns. An intact
homologous copy of the mutated region is required in order to serve as template
for the conversion of the mutated to wildtype sequence. Gene conversion can
only function in compound heterozygous patients, and it does not function if
the homologous gene region on the other allele is deleted. Gene conversion as a
probable mechanism of revertant mosaicism was first suggested by Lo ten Foe
et al. [1]. These authors observed two mosaic siblings harboring segregant
alleles with only single mutations, but without evidence for haplotype switch-
ing. Alter et al. [19] also suggested gene conversion as mechanism for revertant
mosaicism in their compound heterozygous patient even though there was no
study of polymorphic markers within or flanking the presumptive site of gene
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 166
conversion. Given the relatively wide spacing of polymorphic markers, and
given the presumptive shortness of conversion tracts in mammals (less than
58 bp; [36]), gene conversion as a causal mechanism of revertant mosaicism is
still difficult to prove. An additional problem is the requirement for somatic
pairing of homologous chromosomes which may be mediated, at least in the
case of FANCA, by a large heterochromatin block on chromosome 16 [37], but
which otherwise has been rarely observed in mitotic cells.
Back mutation is another possible mechanism underlying revertant
mosaicism that, again, is difficult to prove since there is no change of polymor-
phic markers surrounding the back-mutated gene locus. Back mutation has
been invoked to explain mosaicism for blood cells with either high and low
SCE phenotypes in two patients carrying homozygous mutations in the Bloom
syndrome gene [38]. In compound heterozygous FA-A patients, back mutation
has been assumed as probable mechanism of reversion since (1) the homolo-
gous gene region opposite to the reverted mutation was deleted in some of these
patients, and (2) the reverted allele displayed the original wildtype rather than
any random sequence [8]. With two exceptions, all presumptive cases of back
mutation listed in table 1 were observed in FANCA, a gene with a high number
of small and large repetitive elements such that slipped-strand mispairing has
been assumed as major mechanism of mutagenesis in this gene [32]. The first
case of revertant mosaicism in an FA-A patient ascribed to back mutation was
reported by Gregory et al. [6] who observed the reversion to wildtype of a
maternally inherited 19-bp insertion within exon 29 of FANCA. The authors
argued that the insertion creates a tandem repeat sequence in close proximity to
a deletion hot spot consensus sequence with the possible consequence of for-
mation of a loop structure. If this loop is deleted prior to a next round of repli-
cation, the wildtype sequence would be restored. A very similar line of
arguments was followed by Gross et al. [8] who observed that the reversion in
three of their mosaic FA-A patients affected the region of exons 10–11 of
FANCA which is known as a highly mutable region due to an abundance of
repetitive elements and sequence motifs, including palindromic sequences.
Such structures are known to be involved in the breakage and rejoining of
DNA. Gross et al. [8] therefore assumed that back mutation (via mechanisms
such as slipped-strand mispairing and others) would be the most likely explan-
ation for the fact that one of the mutated single base pair alleles in all of their
FA-A patients had reverted precisely to wildtype. They further argued that
random base alterations would have yielded mostly non-conservative amino
acid exchanges such that the resulting protein may have been functionally
impaired. As suggested by experiments in mice [39] selection would favor cells
with reversions to the original nucleotide since only these would assure full
restoration of protein function. Back mutation combined with selection for the
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 168
recently, the newly discovered FANCN gene has been added to the list of genes
that exhibit somatic reversions, at least in cultured lymphoid cells [34]. The
reversion in FANCN was shown to result from Alu-mediated recombination
leading to an in-frame fusion of exons 3 and 5, thereby eliminating one of the
disease causing mutations located in exon 4. Alu-mediated recombination can
therefore be added to the list of mechanisms instrumental in compensatory
mutations [34].
10
0
WBC ANC Hb Platelets
Fig. 9. Comparison of blood cell counts between mosaic and non-mosaic FA patients.
Data plotted from table 3 of Soulier et al. [33]. ANC ⫽ Absolute neutrophil count; Hb ⫽ hemo-
globin; WBC ⫽ white blood cell count; vertical axis in arbitrary units. See text for details.
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 170
References
1 Lo Ten Foe JR, Kwee ML, Rooimans MA, Oostra AB, Veerman AJ, et al: Somatic mosaicism in
Fanconi anemia: molecular basis and clinical significance. Eur J Hum Genet 1997;5:137–148.
2 Rieger R, Michaelis A, Green MM: Glossary of Genetics and Cytogenetics: Classical and
Molecular, ed 4. New York, Springer, 1996.
3 Jonkman MF, Castellanos Nuijts M, van Essen AJ: Natural repair mechanisms in correcting path-
ogenic mutations in inherited skin disorders. Clin Exp Dermatol 2003;28:625–631.
4 Youssoufian H, Pyeritz RE: Mechanisms and consequences of somatic mosaicism in humans. Nat
Genet Rev 2002;3:748–758.
5 Hirschhorn R: In vivo reversion to normal of inherited mutations in humans. J Med Genet
2003;40:721–728.
6 Gregory JJ Jr, Wagner JE, Verlander PC, Levran O, Batish SD, et al: Somatic mosaicism in
Fanconi anemia: evidence of genotypic reversion in lymphohematopoietic stem cells. Proc Natl
Acad Sci USA 2001;98:2532–2537.
7 Youssoufian H: Natural gene therapy and the Darwinian legacy. Nat Genet 1996;13:255–256.
8 Gross M, Hanenberg H, Lobitz S, Friedl R, Herterich S, et al: Reverse mosaicism in Fanconi
anemia: natural gene therapy via molecular self-correction. Cytogenet Genome Res 2002;98:126–135.
9 Mankad A, Taniguchi T, Cox B, Akkari Y, Rathbun RK, et al: Natural gene therapy in monozygotic
twins with Fanconi anemia. Blood 2006;107:3084–3090.
10 Kwee ML, Poll EHA, Van de Kamp JJP, De Koning H, Eriksson AW, Joenje H: Unusual response
to bifunctional alkylating agents in a case of Fanconi anaemia. Hum Genet 1983;64:384–387.
11 Cohen MM, Simpson SJ, Honig GR, Maurer HS, Nicklas JW, Martin AO: The identification of
Fanconi anemia genotypes by clastogenic stress. Am J Hum Genet 1982;34:794–810.
12 Duckworth-Rysiecki G, Hulten M, Mann J, Taylor AM: Clinical and cytogenetic diversity in
Fanconi’s anemia. J Med Genet 1984;21:197–203.
13 Cervenka J, Hirsch BA: Cytogenetic differentiation of Fanconi anemia, ‘idiopathic’ aplastic ane-
mia, and Fanconi anemia heterozygotes. Am J Med Genet 1983;15:211–223.
14 Auerbach AD, Rogatko A, Schroeder-Kurth TM: International Fanconi Anemia Registry: First
Report; in Schroeder-Kurth TM, Auerbach AD, Obe G (eds): Fanconi Anemia, Clinical
Cytogenetic and Experimental Aspects. Berlin/Heidelberg, Springer-Verlag, 1989, pp 3–17.
15 Jonkman MF, Scheffer H, Stulp RP, Pas HH, Nijenhuis M, et al: Revertant mosaicism in epider-
molysis bullosa caused by mitotic gene conversion. Cell 1997;88:543–551.
16 Hopkins NC, Manoharan A, Lammi AT: Spontaneous improvement in Fanconi’s anemia.
Pathology 1991;23:254–255.
17 Kwee ML, van der Kleij JM, van Essen AJ, Begeer JH, Joenje H, et al: An atypical case of Fanconi
anemia in elderly sibs. Am J Med Genet 1997;68:362–366.
18 Hamanoue S, Yagasaki H, Tsuruta T, Oda T, Yabe H, et al: Myeloid lineage-selective growth of
revertant cells in Fanconi anaemia. Br J Haematol 2006;132:630–635.
19 Alter BP, Joenje H, Oostra AB, Pals G: Fanconi anemia: adult head and neck cancer and
hematopoietic mosaicism. Arch Otolaryngol Head Neck Surg 2005;131:635–639.
20 Joenje H, Oostra AB, Zwaan CM, Pals G: Fanconi anemia; in Humana Press Series on Hereditary
Cancer Syndromes. Totowa, Humana Press, 2006; in press.
21 Schroeder-Kurth TM, Auerbach AD, Obe G (eds): Fanconi Anemia, Clinical Cytogenetic and
Experimental Aspects. Berlin/Heidelberg, Springer-Verlag, 1989, pp 1–378.
22 Schroeder-Kurth TM, Zhu TH, Hong Y, Westphal I: Variation in cellular sensitivities among
Fanconi Anemia patients, non-Fanconi Anemia patients, their parents and siblings, and control
probands; in Schroeder-Kurth TM, Auerbach AD, Obe G (eds): Fanconi Anemia, Clinical
Cytogenetic and Experimental Aspects. Berlin/Heidelberg, Springer-Verlag, 1989, pp 106–136.
23 Bakhshi S, Joenje H, Schindler D, Oostra A, Mohamed AN, et al: A case report of a patient with
microcephaly, facial dysmorphism, mitomycin-C-sensitive lymphocytes, and susceptibility to
lymphoma. Cancer Genet Cytogenet 2006;164:168–171.
24 Schindler D, Hoehn H: Flowcytometric testing for syndromes with chromosomal instability, aplas-
tic anemia and related haematological disorders; in Wegner RD (ed): Diagnostic Cytogenetics.
Berlin/Heidelberg, Springer-Verlag, 1999, pp 269–281.
H. Hoehn
Department of Human Genetics, Biocenter
Am Hubland, 97074 Würzburg (Germany)
Tel. ⫹49 931 888 4071, Fax ⫹49 931 888 4434, E-Mail [email protected]
Hoehn/Kalb/Neveling/Friedl/Bechtold/Herterich/Sun/Gruhn/Hanenberg/Schindler 172
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 173–182
Abstract
Bone marrow transplantation from an HLA-identical sibling donor is the treatment of
choice for Fanconi anemia (FA) patients with bone marrow failure. However, with today’s
small size families less than 25% of FA patients have a matching sibling donor. The remain-
ing patients can be treated with stem cell transplantations from alternative donors such as
matched unrelated or haploidentical donors. While results with conventional bone marrow
transplantation continually improved, outcome after alternative donor transplantation remained
poor due to high rates of transplant-related complications, graft rejection and graft-versus-host
disease (GvHD). Recently, advances with less myeloablative and more immunosuppressive
conditioning regimens have demonstrated promising results. Insights into the mechanisms of
immune reconstitution could show that at least in children immune recovery after haploiden-
tical stem cell transplantation is fast and results in complete restoration of normal immune
function within the first year after transplantation. Finally, innovations in the field of stem
cell collection and processing have led to cellular graft compositions which now provide reli-
able engraftment in almost all patients with concomitant low incidence of GvHD. This
review discusses these recent advances in alternative donor transplantations and concludes
that this treatment option should be an early recommendation for FA patients with bone mar-
row failure who lack an HLA-identical sibling donor.
Copyright © 2007 S. Karger AG, Basel
Eyrich/Winkler/Schlegel 174
decline of the incidence of acute or chronic GvHD, probably due to the even
higher number of T cells in peripheral blood stem cell grafts. For years, it has
been believed that T cells in the graft are indispensable to allow for a stable
engraftment in an allogeneic environment. Only patients with an inherent lack
of T cells like children with severe combined immunodeficiency were able to
accept even T-cell depleted, haploidentical grafts without intensive pretrans-
plant conditioning [5]. However, this view changed with introduction of the
‘megadose concept’: high numbers of CD34⫹ stem cells without T-cell support
are able to suppress anti-donor cytotoxic T lymphocyte (CTL) responses and
ensure engraftment even after transplantation beyond HLA-barriers [6].
Innovations in the field of stem cell processing techniques made it possible to
isolate the required large numbers of highly purified hematopoetic progenitor
cells [7]. This approach was rapidly translated into clinical protocols, first in
high-risk adult leukemia patients [8], later also in a pediatric cohort with child-
hood leukemia [9]. The first case report of a successful haploidentical trans-
plantation in a girl with FA was published by Elhasid et al. in 2000 [10].
Interestingly, this report followed exactly the megadose concept with transplan-
tation of large numbers of highly purified hematopoietic progenitor cells and
only minimal T-cell content. Another study, published in the same year reported
29 FA patients transplanted with partially T-cell depleted bone marrow from
alternative donors after receiving cyclophosphamide (40 mg/kg), ATG and total
body irradiation (450–600 cGy) as conditioning regimen [11]. In this cohort,
the probability of survival at 1 year was only 34%. Although a final assessment
about the use of haploidentical stem cell transplantation based on these results
is not possible so far, it is tempting to speculate that the megadose concept will
result in superior engraftment rates with a concomitant low incidence of acute
and chronic GvHD. In fact, Lang et al. reported a series of 25 patients with non-
malignant diseases (excluding FA) where transplantation of large numbers of
positively enriched CD34⫹ hematopoietic stem cells from related or unrelated
donors resulted in a survival rate of 88% without acute GvHD greater than
grade II and limited chronic GvHD in only 8% of the patients [12]. Future stud-
ies have to show whether these encouraging results will also apply to FA
patients. If haploidentical stem cell transplantation in combination with the
megadose concept proves to be sufficiently safe and effective in FA then this
therapeutic option could be offered early on to all FA patients with deterioration
of bone marrow function. Either parents or siblings qualify as potential hap-
loidentical donors such that almost every patient with FA is likely to have at
least one haploidentical donor. However, successful outcome of stem cell trans-
plantation is not only based on selection of the best donor and appropriate stem
cell processing techniques but also on a pretransplant conditioning regimen
which has been optimized for the special needs of FA patients.
The first conditioning regimens used for preparation of patients for bone
marrow transplantation contained mainly total body irradiation (TBI) plus one
or two alkylating agents. These regimens created ‘marrow space’ for the allo-
geneic graft by eradicating host hematopoiesis but also had a profound
immunosuppressive effect. Initial reports with these conditioning regimens in
FA patients showed an unacceptable high toxicity especially due to the use of
cyclophosphamide and its metabolites [3]. Another problem was the high rate
of graft rejection frequently seen in FA patients [13]. These data suggested that
existing protocols for pretransplant conditioning were too toxic albeit not
immunosuppressive enough for FA patients to allow for safe allogeneic engraft-
ment. In 1991 Fischer et al. demonstrated that addition of a monoclonal anti-
body against leukocyte function-associated antigen (LFA-1, CD11a) to the
conditioning regimen significantly reduced rates of graft rejection in patients
with nonmalignant diseases [14]. Furthermore, it could be shown that patients
with primary nonengraftment or graft rejection can be successfully retrans-
planted after a purely immunological reconditioning based on the anti-T-cell
antibody OKT3 and methylprednisolone [15]. In 1997, the first fludarabine-
based protocol with addition of low-dose cyclophosphamide and ATG was pub-
lished [16]. Fludarabine monophosphate is a purine analogue and has a much
more favorable toxicity profile for FA patients compared to alkylating agents or
irradiation. Most of the conditioning regimens published thereafter used low-
dose cyclophosphamide and ATG in combination with either limited thoraco-
abdominal irradiation [11] or fludarabine [17–19] or both [20–22]. All authors
reported a remarkable primary engraftment rate of almost 100% with sec-
ondary graft rejections in 4–25% of patients [17, 19, 21–23]. Most of these lat-
ter patients could be successfully retransplanted. The rates of acute and chronic
GvHD also decreased over the past decade. However, the incidence of GvHD
seems to be more related to donor selection and the degree of T-cell depletion
than to the use of TBI, cyclophosphamide, or fludarabine.
Taken together, over the last two decades considerable improvements in
pretransplant conditioning regimens were achieved. The application of irradia-
tion or alkylating agents which carry a major risk for secondary malignancies in
FA patients could be dramatically reduced to the benefit of less toxic and highly
T-cell suppressive regimens. These advances in patient conditioning not only
resulted in higher rates of engraftment but also paved the way for the use of
more sophisticated grafts like T-cell depleted stem cells from alternative
donors. Recently, a study has been initiated which completely avoids irradiation
and alkylating agents. Prepared by a conditioning regimen with fludarabine and
Eyrich/Winkler/Schlegel 176
therapeutic antibodies against CD45 and CD52, FA patients in this study will be
grafted with bone marrow from matched related or CD34⫹ peripheral blood
stem cells from haploidentical donors (Persis Amrolia, personal communica-
tion). This ongoing study surely forms the preliminary endpoint of a logical
development, which was fostered by experiences with immunological condi-
tioning regimens from the last 25 years.
The high expectations after the first successful stem cell transplantations
from haploidentical donors were attenuated by a high rate of infectious compli-
cations, especially in adults [24]. This was due to the extremely low numbers of
T cells in the graft and a subsequently delayed immune reconstitution.
However, the patterns of immune recovery after transplantation of highly
enriched, CD34⫹ peripheral blood stem cells from haploidentical donors were
initially only poorly understood. Our group has prospectively analyzed the pat-
terns of immune reconstitution in children after haploidentical transplantations.
In a first study, twenty children transplanted with high doses of purified CD34⫹
hematopoietic stem cells were prospectively monitored for their immune recon-
stitution during the first post-transplant year [25]. T, B, and NK cells began to
reconstitute (as a median) on day ⫹72, ⫹68, and ⫹30 respectively (fig. 1).
During extended follow-up, their numbers and proliferative capacity upon
mitogen stimulation continually increased. Early reconstituting T cells were
predominantly of a memory (primed, activated) phenotype. Naive T cells
emerged after approximately 6 months post transplantation. All patients self-
maintained sufficient immunoglobulin levels after day ⫹200. This study
demonstrated for the first time that at least pediatric recipients of highly puri-
fied, haploidentical stem cells are able to reconstitute functioning T-, B- and
NK-cell compartments within the first post-transplant year.
In the subsequent follow-up study, we analyzed the factors governing
the normalization of the initially restricted T-cell receptor repertoire [26].
Normalization of the initially restricted repertoire is of prime importance to the
patient with regard to fighting off potentially life-threatening infections. We
determined the relative contributions of naive and memory T-cell subsets within
the CD4⫹ and CD8⫹ compartments to the evolution of overall TCR-repertoire
complexity following transplantation of CD34⫹ selected peripheral blood prog-
enitors. During the first post-transplant year, sorted CD4/45RA, CD4/45RO,
CD8/45RA and CD8/45RO subsets were analyzed at three monthly intervals for
B-cells/l
800 500
600 400
300
400
200
200
100
50 100 150 200 250 300 350 400 450 50 100 150 200 250 300 350 400 450
a Days post transplantation b Days post transplantation
500
450
400
350
NK-cells/l
300
250
200
150
100
50
Eyrich/Winkler/Schlegel 178
Thymus-dependent T-cell regeneration (de novo generation of naive
T-cells) is a key pathway for immune reconstitution after stem cell transplanta-
tion. In a third study, we prospectively assessed T-cell dynamics and thymic
function in 164 pediatric patients between 1 and 124 months post-transplant by
measuring T-cell-receptor recombination excision circles (TRECs) as a mea-
sure for de novo thymic T-cell emigrants and spontaneous expression of Ki67 in
peripheral T-cell subsets (as a measure for proliferation of T cells in the periph-
eral pool) [27]. We analyzed the impact of recipient age, conditioning regimen,
type of donor and graft, stem cell dose, and graft-versus-host disease (GvHD)
on the onset as well as on the plateau of thymic output. Multivariate analysis
revealed that the onset of thymic recovery was inversely correlated only with
recipient age (p ⬍ 0.0002), whereas the plateau of thymic output was higher
in patients receiving increased (⬎107 CD34⫹/kg BW) stem cell numbers
(p ⬍ 0.0022). Interestingly, donor type, stem cell source and conditioning regi-
men influenced none of the analyzed parameters. Onset and plateau of thymic
activity were found to be independently regulated by different transplant-related
factors.
Taken together our data demonstrated that in children transplantation of
highly purified stem cells results in fast reconstitution of NK-, T-, and B-cell
compartments. These data are in contrast to results from adult patients after
T-cell depleted haploidentical transplantations, whose T-cell reconstitution can
take up to two years or more. They also correlate with the clinical observation
that transplanted children have a lower incidence of infectious complications
than adult patients. One of the major differences between adults and children
with regard to T-cell reconstitution seems to be the enhanced thymic recovery in
children [28]. Thus, transplantation of highly purified haploidentical stem cells
should especially be considered in pediatric patients, since immune recovery in
children is fast and no long-lasting immunodeficiency due to the transplant pro-
cedure has to be expected.
Future Directions
Acknowledgements
This work was supported in part by IZKF Wuerzburg (Z-2/7) and by a grant from the
Childhood Cancer Parent’s Initiative Wuerzburg.
References
Eyrich/Winkler/Schlegel 180
10 Elhasid R, Ben Arush MW, Katz T, Gan Y, Shechter Y, et al: Successful haploidentical bone mar-
row transplantation in Fanconi anemia. Bone Marrow Transplant 2000;26:1221–1223.
11 MacMillan ML, Auerbach AD, Davies SM, Defor TE, Gillio A, et al: Haematopoietic cell trans-
plantation in patients with Fanconi anaemia using alternate donors: results of a total body irradia-
tion dose escalation trial. Br J Haematol 2000;109:121–129.
12 Lang P, Klingebiel T, Bader P, Greil J, Schumm M, et al: Transplantation of highly purified peripheral-
blood CD34⫹ progenitor cells from related and unrelated donors in children with nonmalignant
diseases. Bone Marrow Transplant 2004;33:25–32.
13 Zwaan CM, Van Weel-Sipman MH, Fibbe WE, Oudshoorn M, Vossen JM: Unrelated donor bone
marrow transplantation in Fanconi anaemia: the Leiden experience. Bone Marrow Transplant
1998;21:447–453.
14 Fischer A, Friedrich W, Fasth A, Blanche S, Le Deist F, et al: Reduction of graft failure by a mon-
oclonal antibody (anti-LFA-1 CD11a) after HLA nonidentical bone marrow transplantation in
children with immunodeficiencies, osteopetrosis, and Fanconi’s anemia: a European Group for
Immunodeficiency/European Group for Bone Marrow Transplantation report. Blood 1991;77:
249–256.
15 Schlegel P, Eyrich M, Bader P, Handgretinger R, Lang P, et al: OKT-3 based reconditioning regi-
men for early graft failure in HLA-non-identical stem cell transplants. Br J Haematol 2000;111:
668–673.
16 Kapelushnik J, Or R, Slavin S, Nagler A: A fludarabine-based protocol for bone marrow trans-
plantation in Fanconi’s anemia. Bone Marrow Transplant 1997;20:1109–1110.
17 Motwani J, Lawson SE, Darbyshire PJ: Successful HSCT using nonradiotherapy-based condition-
ing regimens and alternative donors in patients with Fanconi anaemia – experience in a single UK
centre. Bone Marrow Transplant 2005;36:405–410.
18 Rossi G, Giorgiani G, Comoli P, Nobili B, Salvaneschi L, et al: Successful T-cell-depleted, related
haploidentical peripheral blood stem cell transplantation in a patient with Fanconi anaemia using a
fludarabine-based preparative regimen without radiation. Bone Marrow Transplant 2003;31:
437–440.
19 George B, Mathews V, Shaji RV, Srivastava V, Srivastava A, Chandy M: Fludarabine-based condi-
tioning for allogeneic stem cell transplantation for multiply transfused patients with Fanconi’s
anemia. Bone Marrow Transplant 2005;35:341–343.
20 Boulad F, Gillio A, Small TN, George D, Prasad V, et al: Stem cell transplantation for the
treatment of Fanconi anaemia using a fludarabine-based cytoreductive regimen and T-cell-
depleted related HLA-mismatched peripheral blood stem cell grafts. Br J Haematol 2000;111:
1153–1157.
21 Boyer MW, Gross TG, Loechelt B, Leemhuis T, Filipovich A, Harris RE: Low risk of graft-
versus-host disease with transplantation of CD34 selected peripheral blood progenitor cells from
alternative donors for Fanconi anemia. J Pediatr Hematol Oncol 2003;25:890–895.
22 Yabe H, Inoue H, Matsumoto M, Hamanoue S, Koike T, et al: Allogeneic haematopoietic cell
transplantation from alternative donors with a conditioning regimen of low-dose irradiation, flu-
darabine and cyclophosphamide in Fanconi anaemia. Br J Haematol 2006;134:208–212.
23 Torjemane L, Ladeb S, Ben Othman T, Abdelkefi A, Lakhal A, Ben Abdeladhim A: Bone marrow
transplantation from matched related donors for patients with Fanconi anemia using low-dose
busulfan and cyclophosphamide as conditioning. Pediatr Blood Cancer 2006;46:496–500.
24 Aversa F, Tabilio A, Velardi A, Cunningham I, Terenzi A, et al: Treatment of high-risk acute
leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA
haplotype. N Engl J Med 1998;339:1186–1193.
25 Eyrich M, Lang P, Lal S, Bader P, Handgretinger R, et al: A prospective analysis of the pattern of
immune reconstitution following transplantation of HLA-disparate hematopoietic stem cells from
parental donors. Br J Haematol 2001;114:422–432.
26 Eyrich M, Croner T, Leiler C, Lang P, Bader P, et al: Distinct contributions of CD4⫹ and CD8⫹
naive and memory T-cell subsets to overall TCR-repertoire complexity following transplantation
of T-cell depleted CD34 selected hematopoietic progenitor cells from unrelated donors. Blood
2002;100:1915–1918.
Matthias Eyrich
University of Würzburg
Children’s Hospital, Pediatric Stem Cell Transplantation Program
Josef-Schneider-Strasse 2, D30
D–97080 Würzburg (Germany)
Tel. ⫹49 931 201 27640, Fax ⫹49 931 201 27649
E-Mail [email protected]
Eyrich/Winkler/Schlegel 182
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 183–199
Abstract
Orthologs of the human Fanconi anemia (FANC) genes have been identified in several
vertebrate and invertebrate model organisms, indicating variously conserved functions of the
FANC protein complex. In particular, the analysis of chicken DT40 cells has made important
contributions to the functional characterization of FANC genes. Orthologs for most human
FANC genes have been found in the chicken genome which is considerably smaller than the
human genome. Even though a Fanconi anemia-like phenotype has not been described in
avian species, FANC-deficient chicken cells display the same sensitivity to DNA crosslinking
agents as mammalian cells. In addition to a brief review of FANC gene orthologs in verte-
brates and lower organisms, we here show that the chicken FANCC and FANCG genes are
highly expressed in multiple avian embryonic tissues, whereas in adult birds strong expres-
sion was only observed in gonads which underlines the putative function of these genes dur-
ing premeiotic DNA replication and meiotic recombination. We further show that the avian
homologs of two important members of the FANC gene family, FANCC and FANCG, are
located on the chicken Z sex chromosome. We tested the sensitivity of chicken embryonic
fibroblasts from males (ZZ) and females (ZW) towards mitomycin C and observed a gender
difference. This observation is consistent with the absence of Z chromosome dosage com-
pensation in birds and supports the essential role of FANCC and FANCG in the cellular
defense against DNA crosslinking agents.
Copyright © 2007 S. Karger AG, Basel
Cells of all FA patients are highly susceptible to the induction of chromo-
some breaks with DNA crosslinking agents such as diepoxybutane (DEB) or
mitomycin C (MMC). This indicates that mutations in the different FANC genes
cause a similar DNA repair defect(s) [1–4]. Sensitivity to crosslinking agents is
a cytological ‘FA hallmark’, which is widely used for diagnostic testing of FA.
Somatic cell fusion and biochemical analyses have delineated 13 different FA
complementation groups (FA-A, B, C, D1, D2, E, F, G, I, J, L, M, and N). With
the exception of FANCI, the disease genes underlying these groups have already
been cloned [5–7].
Orthologs of the centrally important FANCD2 protein have been detected
in such diverged organisms as Arabidopsis, Caenorhabditis, Drosophila, fugu
and zebrafish [8–10], indicating likely conservation of FANCD2 function. In
this report we review recent findings on the FA pathway in different vertebrates.
Special attention is given to birds which phylogenetically are closest to mam-
mals. Since they can be easily manipulated by homologous recombination,
chicken DT40 cells are increasingly used in FA research. An interesting addi-
tional aspect of the avian cell model in FA research is the increased susceptibil-
ity of FA cells to oxidative stress because, for various reasons, birds appear to
be less or more resistant than mammals to the detrimental effects of reactive
oxygen species, in general [11, 12].
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 184
and other HR proteins like FANCJ [18, 19]. So far, there are only few studies
that directly test recombination activity, for example by introducing a plasmid
substrate in normal and FA cell extracts [20] or in murine cells with a targeted
mutation at the Fanca locus [21]. However, there are a number of studies that
have evaluated FANC protein function using cell-free assays with Xenopus egg
extracts [22], knockdown zebrafish embryos [10] and chicken DT40 knockout
cells [23]. Collectively, these results suggest a role(s) for FANC proteins in
DSB repair most likely through error-free HR. As such, FANC proteins appear
to be important for maintaining genomic stability [24]. This guardian role is
consistent with evolutionary conservation of at least part of the FA pathway.
Besides their crucial role in HR-mediated DSB repair, FANC proteins have
been functionally linked to oxygen metabolism, cell cycle regulation, hemato-
poiesis, and apoptosis. Human FA cells show increased susceptibility to oxida-
tive stress which suggests a role for FANC proteins in the protection against the
endogenous production of reactive oxygen species (ROS). In spite of a higher
metabolic rate, body temperature and blood sugar level, which all contribute to
increased DNA damage after oxidative stress, birds produce relatively fewer
ROS than mammals [11, 12]. This suggests that birds must be endowed with
preventive or protective mechanisms that may also include the FA family of
genes.
The phenotypes of Fanca, Fancc, and Fancg knockout mice and even dou-
ble knockouts are very similar, indicating involvement of different Fanc genes
in the same biochemical pathway(s). Although Fanc-deficient mouse fibrob-
lasts are also sensitive to crosslinking agents, unlike humans KO mice are not
affected by bone marrow failure or malignancies [25]. Interestingly, the differ-
ent KO mice all showed hypogonadism and impaired fertility [26], indicating a
functional role for FANC proteins during meiosis.
Fancd2-deficient mice and zebrafish embryos exhibit some malforma-
tions, i.e. microcephaly and microphthalmia that are also observed in FA patients.
Increased apoptosis in these embryos can be partially corrected by injection of
human FANCD2 protein [10]. The zebrafish orthologs of nine human FANC
genes have been cloned and mapped to syntenic regions shared by the zebrafish
and human genomes [27]. Evidently, the FANC gene network and most likely
the FA pathway already existed in a common ancestor of teleost fish and
tetrapods. Because of a genome duplication event in the ray fin fish lineage [28,
29] most zebrafish orthologs of human genes exist as two paralogous gene
copies. However, none of the zebrafish Fanc genes has such a second copy,
During the course of evolution, the avian genome has been subject to
extensive structural reorganization. Modern bird genomes are compartmental-
ized into macro- and micro-chromosomes. Although the size of the chicken
genome amounts to only 40% of the human genome it is endowed with almost
the same gene content [36]. It is therefore not surprising that orthologs of
almost all human FANC genes were found in chicken.
The first well characterized FANC gene in chicken was FANCD1 (BRCA2)
[37, 38], which maps to GGA chromosome 1. Most other avian FANC genes
have been isolated from the highly recombinogenic chicken B cell line DT40
[39, 40]. The overall amino acid sequence similarity between human and
chicken FANC orthologs is not particularly high, amounting to 39% for FANCC,
40% for FANCD1 (BRCA1), 49% for FANCG, 54% for FANCJ (BRIP1) and
57% for FANCD2. Despite this overall low level of sequence conservation
between avian and mammalian FANC genes, functionally important domains,
including the PHD finger domain in FANCL, the helicase and degenerated
nuclease domains in FANCM, the helicase domain in FANCJ, the RAD51-binding
BRC repeats in FANCD1, and to some extent the tetratricopeptide motif in
FANCG are highly conserved in the chicken genome. This is consistent with the
view that these domains act as scaffolds to co-ordinate the functions of different
FANC proteins. One remarkable species difference is the absence of a specific
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 186
L
B
NC
NC
FA
M F
D1
FA
NC C
FA FAN
NC
FA
1 2 3 4 5 6 7
D2
A
NC
NC
FA
FA
8 9 10 11 12 13 14
J
NC
FA
15 16 17 18 19 20 21
E
NC
FA
22 23 24 25 26 27 28
C G
NC NC
29 30 31 32 33 34 35
FA FA
36 37 38 Z Z
phosphorylation site in the chicken FANCJ protein. In human cells this site is
critical for interaction of FANCJ with BRCA1 [39].
The availability of the complete chicken genome sequence [36] facilitates
the in silico identification of avian orthologs of human disease genes. By BLAST
searches of the chicken genome (http://www.ncbi.nlm.nih.gov/mapview/;
http://www.ensembl.org/Gallus_gallus) with human FANC gene sequences, we
have mapped orthologs of the human FANC genes to chicken chromosomes
(fig. 1). With exception of FANCE and FANCJ which are represented by several
contigs in the current version of the chicken genome, the chicken and human
FANC orthologs lie in syntenic regions.
In order to confirm the in silico data, we localized FANCC and FANCG by
fluorescence in-situ hybridization (FISH) on chicken metaphase spreads.
Z-linkage of these genes is not unexpected because hemizygous (ZW) chicken
DT40 cells are known to contain only single copies of FANCC and FANCG
[23, 41]. Indeed, we found specific FISH signals for both genes (fig. 2a and b)
on the short arm of the chicken Z sex chromosome with FANCC close to the
FANCG
6.6
p 4.4
Z W Z W
a Ch FANCG FANCC
q 2.3
2.0
Z Z Z Z c GGA Z
b Ch FANCC
d PstI
Fig. 2. Physical mapping of chicken FANCG and FANCC to the Z sex chromosome. (a)
FANCG hybridization signals (green FITC fluorescence) on the Z short arm. The ZW cut-
outs are counterstained with both DAPI (left) and propidium iodide (right). The bright PI flu-
orescence of terminal heterochromatin allows the distinction between the long and short arm
of the metacentric Z. (b) FANCC signals appear close to the centromere of the Z short arm.
(c) Ideogram of the chicken Z chromosome illustrating the location of both genes. (d)
Southern hybridization of FANCG cDNA to PstI-digested male and female chicken genomic
DNA. Note the increased hybridization intensity of bands in males compared to females.
Size markers in kb are indicated on the left.
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 188
600 Male
50 Wild Female
FANCG– 500
40
Total aberrations
Total aberrations
400
30
300
20
200
10 100
0 0
Control 20ng/ml 40ng/ml Control 150ng/ml 250ng/ml 350ng/ml
a MMC concentrations b MMC concentrations
Fig. 3. (a) Frequency of aberrations in normal DT40 (blue bars) and fancgDT40
knockout (purple bars) cells after treatment with the indicated MMC concentrations (for
details, see [23]). Note the increased level of MMC-induced DNA damage in fancg deficient
cells. (b) Number of chromosome aberrations in diploid chicken embryonic fibroblasts of
both sexes (male: blue bar and female: red bar) exposed to the indicated MMC concentra-
tions. Note the significantly higher chromosome aberrations in female. 200 metaphases were
screened for each experiment.
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 190
mutants supports a role for FA proteins in facilitating HR but not global TLS
during crosslink repair. Taken together, FANC-deficient DT40 cells have clearly
improved our understanding of the molecular mechanisms in the FA pathway.
female cells than in homogametic (ZZ) male cells (fig. 3b). The increased sen-
sitivity of female cells is particularly evident when the cells are exposed to rel-
atively high MMC concentrations. At low MMC concentration the gender
difference was less pronounced, however the SCE frequency was markedly
increased in female compared to male chicken cells (data not shown).
As far as we can ascertain, the sex-specific cellular response to MMC
treatments appears to be unique for birds. Although the FANCB gene is
X-linked in mammals, this should not cause a sex-specific DNA damage response
because of X-inactivation in females. It is tempting to speculate that because of
their higher FANCC and FANCG gene dosage male birds may be better pro-
tected from oxidative damage through reducing ROS production. In most ratites
(flight-less birds) the Z and W sex chromosomes are still very similar in size
and gene content [61]. Thus, in contrast to modern birds female ratites most
likely will have two copies of FANCC and FANCG. It will therefore be interest-
ing to test whether sensitivity to MMC treatment and oxidative stress differs
between male and female cells in ratites as we here show for modern birds.
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 192
Fancc HH19 Fancc HH26 Fancg HH24
during development. Mouse studies revealed that Fancc, Fancg and Fanca are
preferentially expressed in highly proliferating and embryonic tissues [62–64].
The importance of Fanc genes during early development was also demonstrated
in zebrafish embryos where Fancd2 prevented inappropriate apoptosis in neural
cells and other highly proliferating tissues [10].
In order to explore the activity of FANC genes during avian development,
we used whole mount RNA in situ hybridization on chicken embryos at differ-
ent Hamburger Hamilton (HH) stages to delineate the expression patterns of
chicken FANCC and FANCG genes. FANCC mRNA was first detected in
embryos at HH19, whereas FANCG expression started at HH24 (fig. 5). At
HH19 (fig. 5, left image) FANCC was highly expressed in the midline of the
entire dorsal neural tube (including brain and spinal cord), in the eye and otic
vesicle, at the tips of the branchial arches (white arrow), and in the mesenchyme
around the diencephalon. FANCC was also expressed in both limb buds at
HH19 and in the developing paws at later stages (fig. 5, middle image). In the
limb buds, FANCC mRNA was enriched in the zone of polarizing activity
(ZPA) and the apical epidermal ridge (AER). At HH26 and later stages FANCC
was still expressed in the dorsal spinal cord and in the somites (fig. 5, middle
image).
FANCG expression became detectable in HH24 embryos. Compared to
FANCC it was expressed in a broader region of the dorsal neural tube with
mRNA present in the entire dorsal midbrain and telencephalon (fig. 5, right
image). In the branchial arches FANCG was only weakly expressed at the poste-
rior edge of Ba1 and Ba3 (white arrows). In the limb buds FANCG mRNA cov-
ered the entire proximal-distal extent. In contrast to FANCC, FANCG was not
)
lee )
ng )
Lu n (F
Sp y (F
Lu (M
st )
Br (M)
Te s (F
er )
Ki (F)
Sp n (
Liv (M
Br F)
s
(
is
y
e
e
ain
ain
ng
er
ar
dn
dn
Liv
Ov
5.1kb
2.0kb
Fig. 6. Northern blot with total RNAs from different adult male and female chicken
tissues hybridized with chicken FANCG cDNA. Note the prominent signals in the gonads.
The two different bands in testis tissue may be due to alternative splicing.
detectable in eye and otic vesicle. At HH30 and later embryonic stages both
FANCC and FANCG were expressed concomitantly in the urogenital region
(data not shown), possibly indicating a function of these genes during gonad
and kidney development. In contrast to our findings on chicken embryos,
FANCG expression was not detectable by Northern blot analysis in adult
somatic tissues. Only testis and ovary showed high FANCG mRNA levels on
Northern blots (fig. 6). RT-PCR revealed low FANCG transcript levels in adult
somatic tissues and high levels in gonads (data not shown).
The observed expression patterns support an important role for both
FANCC and FANCG during chicken development. Clearly, expression of these
genes is tightly regulated in a tissue- and developmental stage-specific manner.
The FANCC and FANCG expression patterns are partially overlapping, but not
identical. Abundant transcript levels in the developing eye, ear, spinal cord, and
brain suggest a function for central nervous system development. This is con-
sistent with the observation that in addition to other congenital malformations
many FA patients suffer from eye and ear defects. The expression of chicken
FANCC in limb and wing buds is consistent with the typical radial ray defects of
FA patients.
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 194
Strong expression of FANC genes in chicken and mouse gonads, the
impaired fertility of Fanc knockout mice, and the strongly reduced fertility of
FA patients [64, 65] argue in favor of a germ-cell specific function of FA pro-
teins. Interestingly, the function(s) of FANC genes and the FA pathway during
germ cell development and meiotic recombination appear to be highly con-
served throughout vertebrate evolution, whereas the effects of FANC gene defi-
ciency on somatic development appear to vary widely among different species.
Perspective
Orthologs for almost all mammalian FANC genes have been identified in
chicken. Because most FANC orthologs are not found in yeast, genetic analysis
in this convenient eukaryotic model organism cannot be performed. In this
light, the possibility to efficiently disrupt endogenous FANC genes in chicken
DT40 cells has been extremely useful for the investigation of individual FANC
genes, their specific functions, and the FA pathway.
In addition, birds have revealed interesting novel properties of FANC genes
that are not evident in other vertebrates. With the exception of FANCD1/ BRCA2,
monoallelic inactivation of FANC genes does not appear to cause serious health
problems in humans, whereas compound heterozygotes are phenotypically
affected. In birds we have the exceptional situation that the female (ZW) genome
contains only one copy of both FANCC and FANCG. Consequently, female
chicken cells exhibit a higher sensitivity to MMC treatments. Because male
birds have a higher level of the Z-linked FANCC and FANCG proteins, they are
likely to be more resistant to oxidative stress. This is consistent with observa-
tions that in many bird species males are longer-lived than females [66], and also
have higher survival rates [67]. Whether the survival advantage of male birds
results chiefly from their higher dosage of FANC caretaker genes or whether
additional protective factors are involved remains to be elucidated.
The high expression of FANC genes in avian gonads, and the fact that fertil-
ity is compromised in Fanc knockout mice and in FA patients argues for an essen-
tial function of the FANC family of genes in germ cell development, most likely
during recombination of homologous chromosomes during meiotic prophase I.
The DNA replication process in avian gonads must be particularly efficient to
ensure massive sperm production in male birds and ovary maturation in females.
High levels of FANC gene and protein expression may be instrumental in repair-
ing DSBs arising from DNA replication machinery slippage. However, FANC
proteins may not only be involved in DSB repair by promoting homologous
recombination, but may also contribute to the repair of base changes by transle-
sion synthesis [68]. The study of the Fanconi anemia family of genes in vertebrate
Acknowledgements
This work was supported by a grant from German Research Foundation (DFG; Schm
484/20-2).
References
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 196
18 Mirchandani KD, D’Andrea AD: The Fanconi anemia/BRCA pathway: a coordinator of cross-link
repair. Exp Cell Res 2006;312:2647–2653.
19 Kitao H, Yamamoto K, Matsushita N, Ohzeki M, Ishiai M, Takata M: Functional interplay between
BRCA2/FancD1 and FancC in DNA repair. J Biol Chem 2006;281:21312–21320.
20 Thyagarajan B, Campbell C: Elevated homologous recombination activity in Fanconi anemia
fibroblasts. J Biol Chem 1997;72:23328–23333.
21 Nakanishi K, Yang YG, Pierce AJ, Taniguchi T, Digweed M, et al: Human Fanconi anemia
monoubiquitination pathway promotes homologous DNA repair. Proc Natl Acad Sci USA 2005;
102:1110–1115.
22 Sobeck A, Stone S, Costanzo V, de Graaf B, Reuter T, et al: Fanconi anemia proteins are required
to prevent accumulation of replication-associated DNA double-strand breaks. Mol Cell Biol
2006;26:425–437.
23 Yamamoto K, Ishiai M, Matsushita N, Arakawa H, Lamerdin JE, et al: Fanconi anemia FANCG
protein in mitigating radiation- and enzyme-induced DNA double-strand breaks by homologous
recombination in vertebrate cells. Mol Cell Biol 2003;23:5421–5430.
24 Levitus M, Joenje H, de Winter JP: The Fanconi anemia pathway of genomic maintenance. Cell
Oncol 2006;28:3–29.
25 Noll M, Battaile KP, Bateman R, Lax TP, Rathbun K, et al: Fanconi anemia group A and C double-
mutant mice: functional evidence for a multi-protein Fanconi anemia complex. Exp Hematol
2002;30:679–688.
26 Koomen M, Cheng NC, van de Vrugt HJ, Godthelp BC, van der Valk MA, et al: Reduced fertility
and hypersensitivity to mitomycin C characterize Fancg/Xrcc9 null mice. Hum Mol Genet 2002;11:
273–281.
27 Titus TA, Selvig DR, Qin B, Wilson C, Starks AM, et al: The Fanconi anemia gene network is con-
served from zebrafish to human. Gene 2006;371:211–223.
28 Wittbrodt J, Meyer A, Schartl M: More genes in fish? Bioessays 1998;20:511–515.
29 Taylor JS, Braasch I, Frickey T, Meyer A, Van de Peer Y: Genome duplication, a trait shared by
22000 species of ray-finned fish. Genome Res 2003;13:382–390.
30 Leveille F, Blom E, Medhurst AL, Bier P, Laghmani el H, et al: The Fanconi anemia gene product
FANCF is a flexible adaptor protein. J Biol Chem 2004;279:39421–39430.
31 Meetei AR, de Winter JP, Medhurst AL, Wallisch M, Waisfisz Q, et al: A novel ubiquitin ligase is
deficient in Fanconi anemia. Nat Genet 2003;35:165–170.
32 Collis SJ, Barber LJ, Ward JD, Martin JS, Boulton SJ: C. elegans FANCD2 responds to replication
stress and functions in interstrand cross-link repair. DNA Repair (Amst.) 2006;5:1398–1406.
33 Kojic M, Kostrub CF, Buchman AR, Holloman WK: BRCA2 homolog required for proficiency in
DNA repair, recombination, and genome stability in Ustilago maydis. Mol Cell 2002;10:683–691.
34 Mosedale G, Niedzwiedz W, Alpi A, Perrina F, Pereira-Leal JB, et al: The vertebrate Hef ortholog
is a component of the Fanconi anemia tumor-suppressor pathway. Nat Struct Mol Biol 2005;12:
763–771.
35 Meetei AR, Levitus M, Xue Y, Medhurst AL, Zwaan M, et al: X-linked inheritance of Fanconi ane-
mia complementation group B. Nat Genet 2004;36:1219–1224.
36 International chicken genome sequence consortium: Sequence and comparative analysis of the
chicken genome provide unique perspectives on vertebrate evolution. Nature 2004;432:695–716.
37 Takata M, Tachiiri S, Fujimori A, Thompson LH, Miki Y, et al: Conserved domains in the chicken
homologue of BRCA2. Oncogene 2002;21:1130–1134.
38 Warren M, Smith A, Partridge N, Masabanda J, Griffin D, Ashworth A: Structural analysis of the
chicken BRCA2 gene facilitates identification of functional domains and disease causing muta-
tions. Hum Mol Genet 2002;11:841–851.
39 Bridge WL, Vandenberg CJ, Franklin RJ, Hiom K: The BRIP1 helicase functions independently of
BRCA1 in the Fanconi anemia pathway for DNA crosslink repair. Nat Genet 2005;37:953–957.
40 Takata M, Kitao H, Ishia M: Fanconi anemia: genetic analysis of a human disease using chicken
system. Cytogenet Genome Res 117 (2007, in press).
41 Hirano S, Yamamoto K, Ishiai M, Yamazoe M, Seki M, et al: Functional relationships of FANCC
to homologous recombination, translesion synthesis, and BLM. EMBO J 2005;24:418–427.
Nanda/Buwe/Wizenman/Takata/Haaf/Schartl/Schmid 198
63 Abu-Issa R, Eichele G, Youssoufian H: Expression of the Fanconi anemia group A gene (Fanca)
during mouse embryogenesis. Blood 1999;94:818–824.
64 Wong JC, Buchwald M: Disease model: Fanconi anemia. Trends Mol Med 2002;8:139–142.
65 Hölzel M, van Diest PJ, Bier P, Wallisch M, Hoatlin ME, et al: FANCD2 protein is expressed in
proliferating cells of human tissues that are cancer-prone in Fanconi anaemia. J Pathol 2003;201:
198–203.
66 Holmes DJ, Thomson SL, Wu J, Ottinger MA: Reproductive aging in female birds. Exp Gerontol
2003;38:751–756.
67 Payevsky VA, Vysotsky VG, Yefremov VD, Markovets MY, Shapoval AP: Sex-specific survival
rates in birds. Zh obshch Biol 1997;58:5–20.
68 Thompson LH, Hinz JM, Yamada NA, Jones NJ: How Fanconi anemia proteins promote the four
Rs: replication, recombination, repair, and recovery. Environ Mol Mutagen 2005;45:128–142.
Indrajit Nanda
Department of Human Genetics, University of Würzburg
Biozentrum, Am Hubland
D–97074 Würzburg (Germany)
Tel. ⫹49 931 888 4078, Fax ⫹49 931 888 4058
E-Mail [email protected]
Abstract
Cells from patients suffering from the recessive syndrome Fanconi anemia (FA), are
characterized by increased sensitivity to agents that induce DNA interstrand cross-links
(ICLs). This hypersensitivity manifests as a dramatically elevated rate of chromosome
breaks in FA cells when compared to controls and led, more than thirty years ago, to the
suggestion that the repair of ICLs is disturbed in FA. Today, a DNA repair defect as the basis
of FA is widely accepted, however, the exact role of the 12 known genes is still elusive. The
past several years have brought growing evidence that FA cells are compromised in homol-
ogy dependent DNA repair processes. This review will summarize these studies with a
focus on integrated plasmid reporter assays which are used to investigate repair products
after induction of a single defined DNA double-strand break (DSB).
Copyright © 2007 S. Karger AG, Basel
a b c a' d
a b a' d
a b a' d
a d
a b c a' d
a b c a' d a d
The reporter plasmids typically used to evaluate DSB repair contain two dif-
ferently modified, non-functional reporter genes, one of these inactive because of
the insertion of a recognition site for the rare cutting I-SceI endonuclease and the
other consisting of an internal fragment of the gene. Repair of a DSB induced in
one copy of the gene, which exploits the second undamaged copy leads to the
restoration of its function. The most commonly used reporter gene is the green
fluorescent protein (GFP), but reporter plasmids with various drug resistance
Demuth/Digweed 202
genes have also been described. Expression of I-SceI in cells containing a copy of
the reporter introduces a single defined DSB. Subsequent analysis allows the effi-
ciency of repair of the DSB to be quantified. For example, when GFP is used as
the reporter, the HDR pathway restores the GFP function by recombination
between the two non-functional GFP gene copies. Thus the fraction of cells that
successfully employed HDR to repair the DSB can be directly assessed by count-
ing green fluorescent cells. In addition, amplification and analysis of the
sequence originally containing the I-SceI recognition site can allow further deter-
mination of the exact repair pathway used (e.g. HDR, SSA or NHEJ).
Analysis of chicken DT40 cells with targeted mutations in FA genes has
been shown to be a powerful strategy to investigate FA protein function and has
been used by many researchers to analyse DNA repair with plasmid reporters
(for references see table 1). Studies of DT40 cells with knocked-out FA genes,
fancg or fancd2, revealed a mild but clear reduction in HDR whereas DT40
cells mutated in fancj and fancm were not affected in this pathway (table 1).
Even the HDR analysis of the same fancc knock-out DT40 cell line yielded
conflicting results [12–14].
Mouse ES cells, homozygous for a Fanca null mutation or for ‘mild’ hypo-
morphic mutations in the Fancd1/Brca2 gene, displayed reduced HDR compara-
ble to that of HDR-compromised DT40 FA cells [15–17].
Interestingly, in addition to the HDR defect, we found Fanca⫺/⫺ mouse
cells, as well as FA patient derived cells from complementation groups A, G
and D2, to be deficient in a further DSB repair pathway, SSA, a finding also
reported for Brca2 mutated cells [17–19].
The relatively mild reduction in HDR capacity of human FA-A and FA-G
cells stands in strong contrast to the more than 100-fold decrease in HDR of
FANCD1/BRCA2-deficient CAPAN-1 tumour cells [15, 18]. In addition to
FA-A and FA-G cells, we also found reduced HDR in FA-D2 cells, however in
another report the same FA-D2 cell line (PD20) was not deficient in this path-
way [18, 19]. MCF7 cells, depleted for FANCJ by siRNA mediated knock-
down, showed a 7–10-fold reduction in HDR which is contradictory to the data
obtained from the analysis of DT40 fancj cells [14, 20].
Chicken DT40
Demuth/Digweed
Human
FANCA reduced n.d. [18]
FANCG reduced n.d. [18]
FANCD1 (BRCA2)c ⬎100-fold reduced n.d. [15]
FANCD2d reduced n.d. [18]
FANCD2d normal n.d. [19]
FANCJ (BACH1/BRIP1)e 7–10-fold reduced n.d. [20]
a
MMC: Mitomycin C; n.d.: not determined; sp.: spontaneous.
b
Identical cell line.
c
Tumour cell line.
204
d
Identical cell line.
e
siRNA mediated depletion.
Table 2. RAD51 foci formation in Fanconi anemia cells
FA gene(s) disrupted RAD51 foci induction (treatment, RAD51 foci formation Reference
time between treatment and
analysis)a
Human
FANC A/B/C/D1/D2/E/F/G IR (12 Gy, 8 h) reduced in all groups [11]
FANC A/B/C/D1/D2/E/F/G IR (12 Gy, 8 h), reduced in FANCD1 cells [25]
MMC (1 h/2.4 g/ml, 24 h) reduced in FANCD1 cells
FANC A/G/C IR (5 Gy, 8 h) slightly reduced [43]
MMC (1 h/0.1 g/ml, time course) delayed
FANCA IR (8 Gy, 2 h) reduced [44]
FANCD2 IR (12 Gy) normal [19]
MMC (40 ng/ml) normal
FANC A/D1/F/I/J/L IR (12 Gy, 7 h/24 h) reduced in FANCD1 cells [26]
MMC (1 h/2.4 g/ml, 7 h/24 h) reduced in FANCD1 cells
FANCJ (BACH1/BRIP1) MMC normal [20]
HU (1 mM, 18 h) normal
FANCD2 IR (2 and 15 Gy, 8 h) reduced [45]
Rodent
CHO-FANCG IR (8 Gy, 4 h) normal [46]
CHO-BRCA2 IR (12 Gy, 8 h) reduced [47]
MMC (1 h/2.4 g/ml, 8 h) reduced
Brca2 IR (10 Gy, 5 h) reduced [16]
Fanca MMC (1 h/0.1 g/ml, 9 h) reduced [17]
DT40
fancd2 IR (8 Gy) normal [40]
MMC (1 h/0.5 g/ml, time course) normal
fancg IR (8 Gy) normal [41]
MMC (1 h/0.5 g/ml, time course) normal
a
HU: Hydroxyurea; IR: ionizing radiation; MMC: mitomycin C.
genes and FA patient cell lines have been analyzed for the integrity of RAD51
foci formation in response to IR or mitomycin C (MMC) (table 2).
As shown in figure 2, we found IR induced RAD51 foci formation was
attenuated in FA cells of complementation groups FA-A, B, C, D1, D2, E, F and
G [11]. This has been confirmed by several studies (table 2). However, the
analysis of FA cells of the same complementation groups and cells of the newer
complementation groups, FA-I, FA-J and FA-L, revealed a specific defect in
RAD51 foci formation only in FA-D1 cells [25, 26]. Other reports in this con-
text on single cell lines are also somewhat contradictory (table 2).
30
Foci-positive cells (%)
20
10
0
b Gy 0 12 0 12 0 12 0 12
Fig. 2. a RAD51 nuclear foci observed in irradiated control cells. Primary fibroblasts
were irradiated with 12 Gy and were fixed 8 h later, permeabilized and incubated with a pri-
mary rabbit antibody directed towards RAD51 followed by detection with a secondary Cy3-
conjugated goat anti-rabbit Ig. Cells were counterstained with DAPI and examined
microscopically using the appropriate filters. Representative digital images are shown.
b Quantification of RAD51 foci in control and FA fibroblasts. The levels of RAD51 foci-
positive cells are shown for control fibroblasts and fibroblasts from various FA complemen-
tation groups. Open columns represent unirradiated cells, filled columns are cells irradiated
with 12 Gy and processed for immunofluorescence 8 h later.
Demuth/Digweed 206
tested for this feature were concordant in targeting efficiency and HDR of a
plasmid reporter (table 1).
The previously mentioned chicken DT40 cell line is derived from B lym-
phocytes and continuously undergoes immunoglobulin gene conversion in cul-
ture, a feature that can also be used to assess HDR at the sequence level. In
those cases analyzed, results were in agreement with data from plasmid reporter
assays, fancc and fancd2 cells being deficient, and fancm cells showing profi-
ciency, in immunoglobulin conversion (table 1).
Symmetrical exchanges between the two sister chromatids (sister chro-
matid exchanges, SCEs) can be visualized cytogenetically after DNA labeling
with 5-bromodeoxyuridine (BrdU) during the preceding S-phase. SCEs occur
spontaneously but their frequency can be enhanced by treatment with various
DNA damaging agents. The exact mechanism by which SCEs are formed is
not known, however, the interpretation of SCEs as the result of homologous
recombination processes is widely accepted. Indeed, cells lacking key homol-
ogous recombination genes exhibit significantly reduced spontaneous and
DNA-damage induced SCE levels [28]. Although spontaneous and induced
SCE levels have been investigated in FA cells for more than thirty years, the
reported studies are rather inconsistent (e.g. [29–31], see also table 1).
However, cells from Bloom syndrome patients, deficient in a protein, BLM,
which associates with the FA core complex [32], do show consistently exces-
sive SCEs [33].
Concluding Remarks
References
Demuth/Digweed 208
13 Mosedale G, Niedzwiedz W, Alpi A, Perrina F, Pereira-Leal JB, et al: The vertebrate Hef ortholog is a
component of the Fanconi anemia tumor-suppressor pathway. Nat Struct Mol Biol 2005;2:763–771.
14 Bridge WL, Vandenberg CJ, Franklin RJ, Hiom K: The BRIP1 helicase functions independently of
BRCA1 in the Fanconi anemia pathway for DNA crosslink repair. Nat Genet 2005;37:953–957.
15 Moynahan ME, Pierce AJ, Jasin M: BRCA2 is required for homology-directed repair of chromo-
somal breaks. Mol Cell 2001;7:263–272.
16 Tutt A, Bertwistle D, Valentine J, Gabriel A, Swift S, et al: Mutation in Brca2 stimulates error-
prone homology-directed repair of DNA double-strand breaks occurring between repeated
sequences. EMBO J 2001;20:4704–4716.
17 Yang YG, Herceg Z, Nakanishi K, Demuth I, Piccoli C, et al: The Fanconi anemia group A protein
modulates homologous repair of DNA double-strand breaks in mammalian cells. Carcinogenesis
2005;26:1731–1740.
18 Nakanishi K, Yang YG, Pierce AJ, Taniguchi T, Digweed M, et al: Human Fanconi anemia
monoubiquitination pathway promotes homologous DNA repair. Proc Natl Acad Sci USA 2005;
102:1110–1115.
19 Ohashi A, Zdzienicka MZ, Chen J, Couch FJ: Fanconi anemia complementation group D2
(FANCD2) functions independently of BRCA2- and RAD51-associated homologous recombin-
ation in response to DNA damage. J Biol Chem 2005;280:14877–14883.
20 Litman R, Peng M, Jin Z, Zhang F, Zhang J, et al: BACH1 is critical for homologous recombin-
ation and appears to be the Fanconi anemia gene product FANCJ. Cancer Cell 2005;8:255–265.
21 Bishop DK, Ear U, Bhattacharyya A, Calderone C, Beckett M, et al: Xrcc3 is required for assem-
bly of Rad51 complexes in vivo. J Biol Chem 1998;273:21482–21488.
22 Takata M, Sasaki MS, Sonoda E, Fukushima T, Morrison C, et al: The Rad51 paralog Rad51B pro-
motes homologous recombinational repair. Mol Cell Biol 2000;20:6476–6482.
23 O’Regan P, Wilson C, Townsend S, Thacker J: XRCC2 is a nuclear RAD51-like protein required
for damage-dependent RAD51 focus formation without the need for ATP binding. J Biol Chem
2001;276:22148–22153.
24 Bhattacharyya A, Ear US, Koller BH, Weichselbaum RR, Bishop DK: The breast cancer suscepti-
bility gene BRCA1 is required for subnuclear assembly of Rad51 and survival following treatment
with the DNA cross-linking agent cisplatin. J Biol Chem 2000;275:23899–23903.
25 Godthelp BC, Artwert F, Joenje H, Zdzienicka MZ: Impaired DNA damage-induced nuclear Rad51
foci formation uniquely characterizes Fanconi anemia group D1. Oncogene 2002;21:5002–5005.
26 Godthelp BC, Wiegant WW, Waisfisz Q, Medhurst AL, Arwert F, et al: Inducibility of nuclear
Rad51 foci after DNA damage distinguishes all Fanconi anemia complementation groups from
D1/BRCA2. Mutat Res 2006;594:39–48.
27 Yamazoe M, Sonoda E, Hochegger H, Takeda S: Reverse genetic studies of the DNA damage
response in the chicken B lymphocyte line DT40. DNA Repair 2004;3:1175–1185.
28 Sonoda E, Sasaki MS, Morrison C, Yamaguchi-Iwai Y, Takata M, Takeda S: Sister chromatid
exchanges are mediated by homologous recombination in vertebrate cells. Mol Cell Biol 1999;19:
5166–5169.
29 Sperling K, Wegner RD, Riehm H, Obe G: Frequency and distribution of sister-chromatid
exchanges in a case of Fanconi’s anemia. Humangenetik 1975;27:227–230.
30 Fujiwara Y, Kano Y, Yamamoto Y: DNA interstrand cross-linking, repair, and SCE mechanism in
human cells in special reference to Fanconi anemia. Basic Life Sci 1984;29(pt B):787–800.
31 Wilson JB, Johnson MA, Stuckert AP, Trueman KL, May S, et al: The Chinese hamster FANCG/
XRCC9 mutant NM3 fails to express the monoubiquitinated form of the FANCD2 protein, is
hypersensitive to a range of DNA damaging agents and exhibits a normal level of spontaneous sis-
ter chromatid exchange. Carcinogenesis 2001;22:1939–1946.
32 Meetei AR, Sechi S, Wallisch M, Yang D, Young MK, et al: A multiprotein nuclear complex con-
nects Fanconi anemia and Bloom syndrome. Mol Cell Biol 2003;23:3417–3426.
33 Chaganti RS, Schonberg S, German J: A manyfold increase in sister chromatid exchanges in
Bloom’s syndrome lymphocytes. Proc Natl Acad Sci USA 1974;71:4508–4512.
34 Yuan SS, Lee SY, Chen G, Song M, Tomlinson GE, Lee EY: BRCA2 is required for ionizing
radiation-induced assembly of Rad51 complex in vivo. Cancer Res 1999;59:3547–3551.
Ilja Demuth
Institut für Humangenetik, Charité Universitätsmedizin Berlin
Campus Virchow-Klinikum
Augustenburger Platz 1
13353 Berlin (Germany)
Tel. ⫹49 30 450 566306, Fax ⫹49 30 450 566904, E-Mail [email protected]
Demuth/Digweed 210
Schindler D, Hoehn H (eds): Fanconi Anemia. A Paradigmatic Disease for the Understanding of
Cancer and Aging. Monogr Hum Genet. Basel, Karger, 2007, vol 15, pp 211–225
Abstract
One of the proteins essential in homologous recombination is RAD51, a recombinase
involved in nucleoprotein filament formation. After DNA damage, RAD51 colocalizes in
nuclear foci with other proteins involved in DNA repair. This foci formation is regulated by
BRCA2/FANCD1. As Rad51 deficiency is lethal in mice and a human disorder due to
defects in RAD51 does not exist, the aim of the current study was to develop a system to
knock-down human RAD51 in an inducible form. For this purpose, we took advantage of the
regulatable shRNA delivery system developed by Wiznerovicz and Trono [1] based on the
cotransduction of target cells with two lentiviral vectors. The first vector carries expression
cassettes for the shRNA and for a marker gene. The second vector constitutively expresses
the transcription suppressing protein tTRKRAB whose activity can be regulated via doxycy-
cline (DOX). Here, we show the ability of lentiviral vectors expressing shRNAs designed
against human RAD51 to downregulate the protein expression in primary human fibroblasts
and in HeLa cells. This effect on the protein levels of RAD51 was controllable by DOX and
allowed to visualize RAD51 foci formation in cells via a gammaretroviral vector expressing
RAD51 fused to the enhanced green fluorescent protein (EGFP). This inducible system for
visualization of fluorescence-tagged essential cellular proteins might facilitate to study the
role of RAD51 and its interaction with members of the FA/BRCA pathway in response to
defined DNA damages.
Copyright © 2007 S. Karger AG, Basel
Rio/Hanenberg 212
hairpin RNAs (shRNAs) which then are processed to siRNA in the target cells
[21]. In some instances, where the knock-out/down of a target gene is lethal for
the cell, such as RAD51, alternative approaches for the controlled suppression
of protein levels are needed. In the present study, we took advantage of the gene
targeting system developed by Wiznerovicz and Trono [1] using lentiviral vec-
tors for the production of siRNAs in a drug inducible system. In the targeting
vector, the shRNA is expressed under the control of human H1 promoter
located in the 3⬘ SIN LTR. Also included in the 3⬘ LTR are tet operator (tetO)
sequences which directly bind the tetracycline repressor (tTR). Via a second
vector, the KRAB domain of human KOX1 fused to tTR is constitutively
expressed in the target cell. In the absence of doxycycline (DOX), this
tTRKRAB protein will bind to the tetO element and suppress any polymerase II
and III promoters within 3 kb in an orientation independent manner. In the pres-
ence of DOX, tTRKRAB is sequestered away from tetO thereby allowing gene
expression from both promoters, the H1 promoter driving the shRNA and the
internal EF1␣ promoter driving EGFP as a marker gene for the easy detection
of transduced cells and promotor activities. Using this targeting strategy, we
established an inducible RAD51 knock-down system that can be applied to the
study of homologous recombination in human primary cells and cell lines,
including cells derived from FA patients.
siRNA Transfection
Two different siRNAs against RAD51 were used (Invitrogen, Karlsruhe, Germany). A
fluorescent oligo labeled with FITC was used as a control (Invitrogen). HeLa cells were
transfected using lipofectamin 2000 following manufacturer’s instructions (Qiagen, Hilden,
Germany). Transfection efficiency was assessed by flow cytometry analysis of cells trans-
fected with the fluorescent oligo.
Vector Construction
shRNAs against RAD51 were ordered as phosphorylated oligos with ClaI and MluI
overhangs, subsequently annealed and then ligated into the LV-THM vector cut in the same
way using standard protocols.
S11EGRAIN was constructed by introducing the EGFP cDNA into the retroviral vector
S11RAIN (Velleuer and Hanenberg, unpublished) expressing human RAD51 linked via an
EMCV IRES site to the neomycin phosphotransferase (NEO) gene under the control of the
SFFV promotor. The presence of the NEO gene in the vector allowed us to select transduced
cells using G418 (0.5 mg/ml, GIBCO/Invitrogen).
Supernatant Production
Lentiviral Vectors: HEK293T cells were cultivated in Dulbecco’s Modified Eagle’s
Medium (DMEM) supplemented with 10% fetal calf serum, 2 mM L-glutamine, penicillin
Western Blot
Western blot analyses were performed using extracts of fibroblasts, collected by
centrifugation, washed twice in PBS, lysed in 1⫻ lysis buffer (50 nM Tris-HCl, 70 mM
2-mercaptoethanol, and 2% sodium dodecylsulfate (SDS), then boiled for 5 min, and sub-
jected to 4–12% Nupage (Invitrogen). After electrophoresis, proteins were transferred to a
nitrocellulose membrane using a submerged transfer apparatus (BioRad, Hercules, CA),
filled with 25 mM Tris Base, 200 mM glycine, and 20% methanol. After blocking with 5%
non-fat dried milk in TBS-T (50 mM Tris-HCl (pH 8.0), 150 mM NaCl, and 0.1% Tween 20)
the membrane was incubated with the primary antibodies (anti-RAD51, Calbiochem; anti-GFP
or anti--actin, Abcam, Cambridge, MA) diluted in TBS-T. The detection was performed
Rio/Hanenberg 214
with the Western Breeze Immunodetection Kit (Invitrogen). The concentration of protein was
measured by the Bradford assay.
Results
siRNA2
Control
512
Control 128
siRNA-FITC
24h
Events
Events
transfection
Time after
M1 M1 48h
0 0
100 101 102 103 104 100 101 102 103 104
72h
a EGFP EGFP
b Anti-RAD51
Fig. 1. Transfection of HeLa cells with two different siRNAs against RAD51. (a)
HeLa cells were cotransfected with a fluorescent oligo (FITC) to analyze the efficiency of
transfection. (b) Western blot of HeLa cells transfected with siRNA1, 2, or both. Western blot
analyses were performed on cells harvested 24, 48 and 72 h after transfection. (c) Ability of
HeLa cells transfected with RAD51 siRNAs to form RAD51 foci after induction of DNA
damage with MMC. RAD51 foci appeared red due to staining with a Tx-Red labeled
secondary antibody.
Rio/Hanenberg 216
LV-THM
MluI 5612
5894 bp
ClaI 5628 Cloning sites
for shRNAs
SD 744 SA 1907
Number of cells with 5–10 foci Number of cells with ⬎10 foci
60 60
Number of cells with
foci/200 cells
40 40
30 30
20 20
10 10
0 0
b Control LV-THM shRNA5 siRNA6 siRNA7 c Control LV-THM shRNA5 shRNA6 shRNA7
Fig. 2. (a) Map of the lentiviral vector LV-THM expressing shRNA. The shRNAs were
cloned between the MluI and ClaI sites and expressed under control of the human H1 pro-
moter. The EF1␣ promoter was used to express the enhanced green fluorescent protein
(EGFP) as marker gene. (b, c) Number of RAD51 foci per 200 cells was counted in primary
fibroblasts after transduction with LV-THM vectors expressing the shRNA5, -6 and -7
against RAD51. As control, nontransfected cells and cells transduced with LV-THM vector
were utilized. (b) Number of cells with 5–10 foci per cell. (c) Number of cells with more than
10 foci per cell. Number of cells with foci after MMC treatment (purple column) and with-
out MMC (yellow).
cells containing more than 10 foci per cell being strongly reduced compared to
control cells. These results suggest that although formation of RAD51 foci is
not completely abolished, shRNA6 and -7 expressing cells experience at least
partial inhibition of RAD51 mRNA processing.
Events
Events
Events
HeLa 67.42% 67.42% 99.7%
M1 M1 M1
0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
EGFP EGFP EGFP
Events
Events
HeLa transduced 0% 0.59% 3.34%
LV-tTRKRAB M1 M1 M1
0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
EGFP EGFP EGFP
Rio/Hanenberg 218
HELA LV-THM:LV-tTRKRAB (1:1)
HELA control 0 g/ml DOX 2.5 g/ml DOX
1,023 512 512 512
Events
Events
Events
SSC
5.4% 96.4%
M1 M1 M1
0 0 0 0
0 1,023 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
FSC EGFP EGFP EGFP
Events
Events
Events
Events
0 0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
EGFP EGFP EGFP EGFP
HELA LV-THM:LV-tTRKRAB (1:1)
Fig. 4. Cells cotransduced with LV-THM and LV-tTRKRAB vector were incubated
with different concentrations of doxycycline (DOX). HeLa cells were analyzed by flow
cytometry five days after incubation with 0–12.5 g/ml DOX.
We also tested the time period after adding DOX to LV-tTRKRAB express-
ing cells that would be required to completely abrogate the negative effect of the
tTRKRAB protein on transcription from the H1 and EF1␣ promoters of the LV-
THM vector. To this end, the percentage of cells transduced with both LV-THM
and LV-tTRKRAB at a ratio of 1:1 was analyzed for the expression of EGFP in
the presence of 5 g/ml DOX. As shown in figure 5, flow cytometric analysis at
24 h intervals revealed that the tTRKRAB mediated inhibition of transcription
starts to decrease at 24 h after incubation with DOX. It nevertheless took 72 h
until the maximum EGFP expression occurred as determined by the percentage
of EGFP positive cells and the mean fluorescence intensity (MFI) of cells in
region M1 (data not shown).
46.74%
Events
Events
Events
SSC
4.55% M1
M1
M1
0 0 0 0
0 1,023 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
FSC EGFP EGFP EGFP
Events
Events
Events
Events
0 0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
EGFP EGFP EGFP EGFP
Fig. 5. Time for DOX to sequester tTRKRAB thereby inducing EGFP expression. Cells
cotransduced with LV-THM and LV-tTRKRAB were treated with 5 g/ml of DOX and the
expression of EGFP was analyzed by flow cytometry between day 0 and day 5 after treatment.
Rio/Hanenberg 220
shRNA6 RAD51
0 1 2 3 4
Anti-RAD51
Anti-GFP
Anti--actin
S11EGRAIN
4938 bp
Discussion
Rio/Hanenberg 222
transduced at a 1:1 ratio with shRNA and tTRKRAB expressing vectors. We
also showed that knock-down using a single shRNA construct still allowed
RAD51 foci formation in primary human cells albeit at reduced levels. We
finally demonstrated that overexpression of a 5⬘ EGFP tagged human RAD51
protein in combination with knockdown of endogenous RAD51 protein led
to unimpaired foci formation as detected in transduced cells through the
EGFP tag.
Previous results showed that a vector expressing RAD51 fused to GFP
was mainly expressed in the cytoplasm of cells. Translocation into the cell
nucleus occurred only in response to DNA damage [25]. Although we still
have to formally prove that our EGFP-RAD51 fusion protein functions nor-
mally and that its in vivo expression is stable over time, the normal distribu-
tion of EGFP positive RAD51 foci after DNA damage that we observed in
cells with knock-down of the endogenous RAD51 suggests that the combina-
tion of the vectors we describe here could be an important approach for study-
ing the role of RAD51 in cells of FA patients and normal controls. The
possibility to induce local DNA damage employing a UV laser and confocal
microscopy offers an exciting tool to describe the kinetics of RAD51 mobi-
lization in living cells and to visualize the movement of FA proteins tagged
with other fluorescent dyes to the site of damaged DNA. This approach might
also be useful to study how homologous recombination (HR) takes place in
cells of FA patients belonging to complementation group FA-D1, and how dif-
ferent mutations in BRCA2 resulting in truncated proteins might still allow
residual HR activity.
There is clear need for further improvement of our retroviral vectors as
the shRNA against the RAD51 ORF will also influence the EGFP-tagged
overexpressed protein. This problem can be addressed by introducing silent
mutations in the RAD51 cDNA thereby changing the particular sequences that
are used for the shRNA targeting. In addition, the mutated and fluorescent
tagged protein could be introduced into cells with the same lentiviral vector as
the shRNA. With this design, it would be possible to utilize one construct to do
both, overexpression of a tagged exogenous and knock-down of the endoge-
nous protein. In addition, EGFP as a marker to visualize transduced cells
should be replaced in the shRNA vector either by selectable genes such as
NEO or by surface expressed molecules that enable selection of transduced
cells by MACS or FACS. With these modifications, a multicolor approach
using the same regulatable shRNA vector construct to target the endogenous
protein and to overexpress a fluorescence-tagged version of the same protein
appears to provide a fascinating starting point to visualize and better under-
stand the complex mechanisms in FA cells undergoing HR following exposure
to crosslinking agents.
We would like to thank Didier Trono for providing his inducible lentiviral shRNA vec-
tor system. We are deeply indebted to Juan Bueren for his continuous support and helpful
discussions. This study was supported by the Fanconi Anemia Research Fund, (FARF), Inc.,
Portland, Oregon, U.S.A., and the Elterninitiative Kinderkrebsklinik e. V., Duesseldorf,
Germany.
References
Rio/Hanenberg 224
20 Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T: Duplexes of 21-nucleotide
RNAs mediate RNA interference in cultured mammalian cells. Nature 2001;411:494–498.
21 Wiznerowicz M, Szulc J, Trono D: Tuning silence: conditional systems for RNA interference. Nat
Methods 2006;3:682–688.
22 Raderschall E, Bazarov A, Cao J, Lurz R, Smith A, et al: Formation of higher-order nuclear Rad51
structures is functionally linked to p21 expression and protection from DNA damage-induced
apoptosis. J Cell Sci 2002;115:153–164.
23 Ohnishi T, Taki T, Hiraga S, Arita N, Morita T: In vitro and in vivo potentiation of radiosensitivity
of malignant gliomas by antisense inhibition of the RAD51 gene. Biochem Biophys Res Commun
1998;245:319–324.
24 Henning W, Stuerzbecher HW: Homologous recombination and cell cycle checkpoints: Rad51 in
tumour progression and therapy resistance. Toxicology 2003;193:91–109.
25 Kraakman-van der Zwet M, Overkamp WJ, van Lange RE, Essers J, van Duijn-Goedhart A, et al:
Brca2 (XRCC11) deficiency results in radioresistant DNA synthesis and a higher frequency of
spontaneous deletions. Mol Cell Biol 2002;22:669–679.
Bechtold, A. 131, 149 Herterich, S. 39, 131, 149 Schlegel, P.G. 173
Buwe, A. 183 Hoehn, H. 23, 110, 149 Schmid, M. 183
Huber, S. 95 Schmugge Liner, M. 131
Demuth, I. 200 Schroeder-Kurth, T. 1
Dietrich, R. 9 Kalb, R. 39, 59, 110, 131, Sperling, K. 23
Digweed, M. 23, 200 149 Sun, Y. 149
Kubbies, M. 110
Ebell, W. 79 Kühl, J.-S. 79 Takata, M. 183
Eyrich, M. 173 Tönnies, H. 79, 95
Linka, Y. 110
Friedl, R. 110, 131, 149
Velleuer, E. 9
Nanda, I. 183
Gavvovidis, I. 110 Volarikova, E. 95
Neitzel, H. 79, 95
Gerlach, A. 79, 95 Neveling, K. 39, 59, 110,
Gottwald, B. 131 Winkler, B. 173
131, 149
Gruhn, B. 149 Wizenman, A. 183
Rio, P. 211
Haaf, T. 183
Hanenberg, H. 110, 131, Schartl, M. 183
149, 211, Schindler, D. 39, 59, 110,
Heilmann, C. 131 131, 149
226
Subject Index
227
FA research 23 Neoplasia 61
milestones in 26 Nocadazol 115
FANCF silencing 71, 73 Non-FA tumor patients 66
Fibroblasts 112, 155 AML 65, 91
Flow cytometry 111, 117, 135, 214 breast cancer 69
Fluorescence in situ hybridization (FISH) ovarian cancer 69
84 pancreatic cancer 68
Founder mutations 50 squamous cell carcinoma 71
Non-homologous end-joining (NHEJ) 201
Gene conversion 34, 166
Genetic heterogeneity 27, 53 Ovarian cancer 69
Genotype-phenotype correlation 52
Genotyping in PGD Pancreatic cancer 68
direct 138 Peripheral blood cells (PBC) 95
indirect 137 Phenotype variability 10
Germline mutations 65 Phosphohistone H3 (H3P) 115
Gonad 192 Preimplantation genetic diagnosis (PGD)
Graft-versus-host disease (GvHD) 174 33, 144
Green fluorescent protein (GFP) 112, 202 Prenatal diagnosis of FA 131
G2-phase accumulation 112 functional testing 134
molecular testing 137
H3P staining 115 Protein-protein interaction 31
Hematopoietic stem cell transplantation
(HSCT) 9, 33, 64 RAD51
HLA-identical sibling 174 foci formation 203, 211
Hoechst 33258 117 functional knock-down 211
Homologous recombination 200 immunofluorescence 215
Homology directed repair (HDR) 201, 204 Reactive oxygen species 185
Reverse mutations 5
Immune reconstitution 177 Revertant mosaicism 149
Interphase FISH (I-FISH) 95 cell cycle testing 154
sensitivity 98 chromosome breakage test 152
Interstrand crosslinks (ICL) 40, 111, 200 clinical consequences 169
Intragenic crossover 165 hematopoietic precursor cells 160
lymphocytes 156
Leukemia 62 molecular confirmation 156
Liver tumors 64 skin fibroblast testing 155
Loss of heterozygosity (LOH) 68
Sex chromosomes 183
Megadose concept 175 sG2/GF ratio 122
Microsatellite markers 137 shRNA delivery system 211
Mitomycin C (MMC) Solid tumors 62
sensitivity to 133, 149 Somatic reversion 34, 53
treatment 112 cell lines 151
Monosomy 7 80, 87, 100 FANCD2
Multiplex ligation-dependent probe molecular mechanisms 165
amplification (MLPA) 46 multilineage 161, 169
Myelodysplastic syndrome (MDS) 61 target FA genes 163