Review Article Why does the bone marrow fail in Fanconi anemia?

Review Article
Why does the bone marrow fail in Fanconi anemia?
Juan I. Garaycoechea and K. J. Patel
Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
The inherited bone marrow failure (BMF)
syndromes are a rare and diverse group
of genetic disorders that ultimately result
in the loss of blood production. The molecular defects underlying many of these
conditions have been elucidated, and great
progress has been made toward understanding the normal function of these
gene products. This review will focus on
perhaps the most well-known and genetically heterogeneous BMF syndrome:
Fanconi anemia. More specifically, this
account will review the current state of
our knowledge on why the bone marrow
fails in this illness and what this might
tell us about the maintenance of bone
marrow function and hematopoiesis.
(Blood. 2014;123(1):26-34)
Historical timeline
It is now 85 years since the Swiss pediatrician Guido Fanconi first
described the case of 3 brothers with aplastic anemia associated with
developmental defects.1 It was later found that the condition was
inherited as a Mendelian recessive trait, thus setting the ground for
the eventual determination of the molecular defect driving “Fanconi
anemia” (FA). Over time, a single unifying feature of the disease
emerged: cells from afflicted individuals were extremely sensitive to
agents that cross-link the 2 strands of DNA together.2,3 Cross-linking
chemicals, such as mitomycin C (MMC) and diepoxybutane, are a
diverse class of molecules that are potently cytotoxic. Thus, the
pivotal observation that cells derived from FA children accumulate
broken chromosomes and die rapidly when exposed to crosslinking agents defined a cell autonomous molecular defect underpinning this condition. This linked FA to a failure to resolve a specific
class of genomic damage. Armed with cross-linker–induced chromosome breakage as a simple cellular test for FA, clinicians rapidly
identified many families throughout the world affected with this
illness.4 In 1992, the first FA gene (FANCC) was identified by
expression cloning and complementation of a patient-derived cell
line.5 However, the coding sequence of the FANCC gene revealed
nothing of how it may function to prevent FA; the FANCC
polypeptide is completely devoid of any domain signatures. In the
decades that followed, we came to realize that FA was genetically
heterogeneous, and a worldwide race ensued to identify the many
genes that constitute the 16 FA complementation groups.
At the same time as the complex molecular genetics was unraveling, studies were underway to understand the molecular function
of the FA gene products. The key questions to resolve were how the
known components prevented the cellular chromosome breakage
phenotype and, more challengingly, the clinical phenotype. Early
studies indicated that the FANCC protein localized to the cytoplasm.6,7
This unexpected result immediately suggested that this FA protein
might not exert a direct influence on DNA repair. It also brought into
question whether the chromosomal breakage defect in FA cells was
sufficient or indeed the primary reason for bone marrow failure
(BMF). Credence for this view came from a body of work indicating
that FA patient–derived cells were sensitive to certain cytokines.8,9
This unusual observation could not be easily reconciled with
defective DNA repair as the primary reason why the bone marrow
fails in FA. More recent studies indicated a more direct role
for certain FA proteins in modulating signaling responses10,11
(Figure 112-16).
On the other hand, the majority of the FA gene products were
shown to form a large nuclear complex, which also includes FANCC,
because it was later found that FANCE clearly promotes its import
into the nucleus.17 This multiprotein complex modified the key
downstream FA gene product FANCD2, by conjugation of a single
ubiquitin molecule (monoubiquitination). This critical modification
could be stimulated by DNA damage, resulting in the localization of
the FANCD2 protein to sites of nuclear DNA damage.18 Further
strong support for the view that the primary function of the FA gene
products was in the repair of DNA damage came from the eventual
identification of mutations in bona fide DNA repair genes in rare FA
patient complementation groups.19-21 Thus, 2 hypotheses (ie, defective DNA repair or abnormal cytokine response) emerged as rival
and contrasting explanations of why the bone marrow fails in FA.
However, a key limitation was that they both lacked evidence that
linked either of them directly to BMF. Subsequently, we set out the
merits of these 2 hypotheses in the origins of marrow failure in FA.
We review and assess the current state of knowledge and evidence to
support each of them in this context. Finally, we discuss recent work,
which has identified naturally derived aldehydes as drivers of BMF
in FA.
Submitted September 17, 2013; accepted October 28, 2013. Prepublished
online as Blood First Edition paper, November 7, 2013; DOI 10.1182/blood2013-09-427740.
© 2014 by The American Society of Hematology
26
FA gene products have a clear function in
DNA repair
All 16 FA gene products clearly function in a common process to
maintain genomic stability and to repair DNA interstrand cross-links
(ICLs) (Figure 1). The majority of the FA proteins (FANCA, B, C,
E, F, G, L, and M) assemble to form a large nuclear complex termed
the “FA core complex.”22 The core complex functions as a multisubunit E3 ubiquitin ligase with FANCL catalyzing the monoubiquitination of FANCD2 and FANCI after DNA damage18,23
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
WHY DOES THE BONE MARROW FAIL IN FANCONI ANEMIA?
27
Figure 1. Multifunctionality of the FA proteins. This figure summarizes the known FA proteins and their functions. All FA proteins cooperate in the FA pathway to repair
cross-linked DNA. Most of them have additional functions in other DNA repair transactions.12-14 Finally, some of them have been found to modulate cytokine responses or
interact with REDOX sensing proteins, independently of their role in DNA repair.15,16,82-84
(Figure 2). Loss of any one of the components of the core complex
leads to its destabilization, abrogation of E3 ligase activity, and
therefore, loss of robust FANCD2/FANCI ubiquitination in response
to DNA damage. Ubiquitinated FANCD2 and FANCI form a
heterodimer that is recruited to chromatin where it participates in
the DNA repair process. However, an ICL is a complex lesion,
requiring that multiple steps be removed. How then do the FA
proteins participate in this removal?
A comprehensive genetic analysis of DNA cross-link repair was
carried out in a tractable genetic system offered by chicken DT40
cells. These studies revealed that the FA proteins must work in
cooperation with 3 other key repair processes: excision repair,
translesion synthesis, and double-strand break repair through
homologous recombination (HR).24,25 The discovery that mutations
in bona fide HR genes (like BRCA2) and in components of the
excision nuclease machinery (SLX4 and, most recently, XPF) can
all result in FA confirmed the observations made in genetic
systems.19,20,26,27 However, the order and the regulation of these
various processes could not be further defined using such genetic
approaches, and the precise mechanism of how the FA proteins
participated in the repair of a DNA cross-link remained elusive.
The solution of the structure of FANCD2/FANCI provides some
insight into this question. This showed that both proteins form
a binding interface for both single- and double-stranded DNA; the
DNA structures that form when replicating DNA are stalled at
a cross-link.28
A key insight into how replication-coupled DNA cross-link
repair is achieved came from work using Xenopus egg extracts. In
this cell-free system, the replication of a plasmid bearing a single
cross-link can be followed with single nucleotide resolution in
vitro.29 First, replication stops before reaching the cross-linked
DNA. One fork advances up to the cross-link, and at this point, the
cross-link is excised. A translesion polymerase synthesizes across
the adducted DNA creating an intact duplex strand of DNA.29,30
This duplex is then used to repair the double-strand break using
HR.31 When FANCD2 is depleted from the nuclear extract or its
monoubiquitination is abrogated, then cross-link repair is impeded
because of a failure to initiate the nuclease incision step. This work
unequivocally showed that the Fanconi pathway is critical for
promoting endonucleolytic cleavage of cross-linked DNA, and
that its loss leads to a specific molecular defect in DNA repair.
However, our DNA is not normally exposed to nitrogen mustard,
the chemical used to create this cross-link, so the question remained as
to what the FA DNA repair pathway is repairing in a physiological
context.
BMF in FA is attributable to a stem cell defect
Ultimately, most FA patients develop BMF culminating in pancytopenia. Blood counts at birth are typically normal, but at the
median age of 7, hematologic complications arise. This initially
manifests as low platelet counts, thrombocytopenia, followed by
leukopenia before eventually developing into aplastic anemia.32,33 The fact that all blood lineages eventually become deficient
strongly implies hematopoietic stem cell (HSC) dysfunction. Indeed,
FA patients have very low numbers of CD341 cells, a bone marrow
fraction that is enriched for HSCs and is capable of producing all
blood components upon transplantation.34 More recent work has
shown significant depletion of the CD341 fraction in very young FA
patients, well before the onset of pancytopenia.35 The very low
number of CD341 cells in young FA patients points to a prenatal
origin of the stem cell defect. In fact, knockdown of FA genes in
human embryonic stem cells resulted in defective hematopoiesis.36
However, because of the limitations of studying the dynamics of
hematopoiesis in humans, numerous labs set out to establish murine
models of FA. Although BMF can be induced in these mice with the
use of the cross-linking agent MMC,37 these mouse knockouts do not
recapitulate the full severity of FA because they do not spontaneously
develop the hematologic phenotype.38 In a few instances, mice deficient
in the Fanconi pathway show hematologic dysfunction.39,40 However,
importantly, FA knockout mice do show quantifiable lower numbers
of HSCs coupled with a reduced ability to reconstitute blood
production in irradiated recipients after transplantation.41,42 Furthermore, Fancc2/2 embryos show reduced numbers of stem and progenitor pools with compromised repopulation activity.43 Collectively,
28
GARAYCOECHEA and PATEL
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
Is unresolved DNA damage the initiator
of BMF?
Despite the overwhelming genetic and biochemical evidence for the
role of FA gene products in repairing DNA cross-links, direct
evidence to show that this function is essential to preserve bone
marrow function has been lacking. Some circumstantial support
comes from the fact that FA patients are cancer prone and blood
malignancies from these patients often harbor gross chromosomal
changes. Both myelodysplastic syndrome (MDS) and acute myeloid
leukemia (AML) are thought to originate at the stem or progenitor cell level, as the longevity and proliferative capacity of
these cells renders them susceptible to the accumulation and
transmission of mutations. Such cytogenetic changes can only
occur as a consequence of misrepair of DNA damage, suggesting
that FA patients are prone to genetic instability within their HSC
and progenitor compartments.32,44-49
There are other aspects of the FA phenotype that might give us
a further clue as to which function of the FA gene products results in
the main feature of the human illness. FA is phenotypically heterogeneous; some patients present with a complex spectrum of congenital
abnormalities, whereas others only have a mild phenotype. The
hematologic features characteristic of the disease also vary; some
patients succumb to marrow failure as early as age 3, whereas others
might never develop it. With a few exceptions, most attempts at
correlating genotype-phenotype have proved unsuccessful so far.
However, what is clear is that children with severe developmental
abnormalities also present with early onset of BMF.50 This observation, together with the fact that the HSC defect starts in utero,
suggests that both developmental abnormalities and the contraction
of the stem cell pool could be triggered by the same cause.
One of the main cellular responses to DNA damage is the induction
of p53, which then initiates a response that results in cell cycle arrest
and, in certain instances, apoptosis. Genetic ablation or deficiency of
Figure 2. Current model of ICL repair by the FA pathway. (A) When DNA containing an ICL undergoes replication, the leading strands of 2 converging replication
forks stop at the lesion. The FA core complex is recruited to chromatin, where it
subsequently monoubiquitinates its 2 substrates, FANCD2 and FANCI. (B) Next, the 2
sister chromatids are uncoupled via dual incisions on either side of the ICL, possibly by
SLX4-XPF-ERCC1. (C) Subsequently, a translesion DNA polymerase (TLS pol)
extends the nascent strand beyond the ICL. (D) Finally, 2 fully repaired DNA duplexes
are generated through the action of nucleotide excision repair (NER) on the top duplex
and homologous recombination (HR) on the bottom duplex. The remaining FA
proteins (FANCD1/BRCA2, FANCO/RAD51C, FANCN/PALB2, and FANCJ/BRIP1)
are thought to be involved in these recombination transactions.
these data suggest that FA patients have an impaired HSC pool. This
defect begins in utero, worsens during childhood, and results in
spontaneous BMF early in life (Figure 3).
Figure 3. Progressive attrition of HSCs underlies BMF in FA. (A) The progressive
decline of HSC numbers in FA patients leads to BMF early in life. The HSC defect is
likely to start in utero. (B) Fanconi-deficient mice do not recapitulate the main feature of
FA. For this reason, their usefulness as disease models has been questioned.
However, FA-deficient mice do display a quantifiable defect in their HSC pool as well
as reduced ability to reconstitute blood production in irradiated recipients, which is also
present before birth. Even in the absence of BMF, it would be interesting to see if
Fanconi-deficient mice display age-dependent attrition in the quality of their HSC pool,
as has been observed for other DNA repair–deficient mice.
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
p53 enables cells to tolerate DNA damage, so it is no surprise that p53
deficiency can partially rescue DNA cross-linker sensitivity in FAdeficient cells. The lack of p53 also accelerates tumor formation in
Fancd22/2 and Fancc2/2 mice, suggesting that the FA DNA repair
pathway and p53 cooperate to suppress tumorigenesis.51,52 Most
recently, primary bone marrow cells from FA patients and Fancd22/2
and Fancg2/2 mice were shown to have elevated levels of p53, and
this was attributable to unresolved DNA damage. Finally, deletion or
knockdown of p53 also rescued the defects of FA human and mouse
hematopoietic progenitors.35 Although p53 inactivation allows
HSCs to survive DNA damage, a note of caution must be sounded:
this comes at the enormous cost of enhanced genomic instability and
tumor formation. Collectively, these data strongly indicate that
unresolved DNA damage in FA cells induces p53, which might then
lead to HSC depletion and hence progressive BMF in FA. Knockdown experiments and gene expression analysis initially suggested
that the p53-driven HSC elimination was mediated by p21 and cell
cycle arrest.35 However, genetic ablation of p21 does not rescue the
hematologic phenotype of Fancd22/2 mice, arguing against this
approach.53 Although it is possible that the p53 response in HSCs differs
between mice and humans, future genetic studies should address the
contribution of p53-dependent apoptosis to BMF in FA patients. In
support of this, a recent study has demonstrated an apoptotic response to
cytokinesis failure in FA-deficient HSCs. It remains to be established
whether Fanconi proteins have a distinct role during mitosis or the DNA
bridges in FA-deficient cells are a consequence of unresolved DNA
damage. However, this is one mechanism of HSC depletion that may
contribute to BMF in FA patients.54
The bone marrow is one of the most radiosensitive tissues in the
body, being the first organ system to fail following total body
irradiation. It is DNA damage to the HSCs that ultimately limits the
regeneration of hematopoiesis.55 Genetic evidence for the consequences of DNA damage on HSC function and bone marrow
homeostasis is provided by the severe consequences of disrupting
many different DNA repair pathways in mice.56-58 Mice bearing null
or hypomorphic alleles for genes involved in DNA damage response
(ATM2/2, conditional ATR2/2), nonhomologous end joining (Ku702/2 ,
Ku802/2 , DNA-PKc2/2 , DNA-PKcs3A/3A, LigIV Y288C/Y288C),
HR (Rad50S/S), nucleotide excision repair (XPDTTD), and mismatch repair (Msh22/2) show severe defects both in the quantity
and functional quality of their HSC pools. In some cases, this
resulted in spontaneous BMF (DNA-PKcs3A/3A, LigIV Y288C/Y288C,
Rad50S/S).
It is likely that the main driver of BMF in FA is DNA damage to
the HSC pool. Genetic instability within FA-deficient HSCs results
in the progressive attrition of this vital cell population or the genesis
of neoplastic clones. This notion is supported by the observation that
a strong p53 response mediates the FA hematologic phenotype and
by the fact that different DNA repair deficiencies, unrelated to FA,
also have profound consequences on HSC function.
What is the cause of DNA damage in HSCs?
Should unresolved DNA damage lead to HSC attrition in FA, then
a key question is how such damage arises in the physiological setting.
Without resolution of this key question, a DNA damage hypothesis
for the cause of BMF would be based solely on the circumstantial
evidence presented previously. Genomic DNA is intrinsically unstable, being subject to both spontaneous and enzymatic degradation.
Moreover, metabolism can generate a battery of reactive molecules
WHY DOES THE BONE MARROW FAIL IN FANCONI ANEMIA?
29
that are capable of attacking DNA, the most ubiquitous being reactive
oxygen species (ROS).
The first clue that such molecules might contribute to the endogenous burden of DNA damage driving the FA phenotype was the
hypersensitivity of patient-derived cell lines. ROS arise in cells as
a consequence of normal cellular metabolism, namely, the electron
transport chain and lipid peroxidation, but they also have important
roles in cell signaling. ROS can damage DNA, RNA, and proteins,
and they may also contribute to the aging process. It was demonstrated
long ago that the frequency of chromosomal aberrations in FA
lymphocyte cultures depends on oxygen tension.59 Since then, many
groups have shown that FA-deficient cells grow better under hypoxic
conditions than in ambient oxygen concentration.60 More recently,
the use of low oxygen tension allowed the generation of FA-deficient
iPS lines.61 Superoxide dismutases are a set of enzymes that
metabolize superoxide radicals (·O2-) to molecular oxygen and
hydrogen peroxide (H2O2), thus providing a defense against
oxygen toxicity. Genetic evidence linking oxidative stress to
bone marrow dysfunction in FA comes from Sod12/2 Fancc2/2
double mutant mice.62 These double knockout mice showed bone
marrow hypocellularity and decreased numbers of colony-forming
units. However, these mice showed normal numbers of HSCs as seen
by flow cytometry and did not display developmental defects or
chromosomal aberrations typical of FA.
In search of a source of endogenous DNA damage, our laboratory
has focused on simple aldehydes. Small aldehydes such as formaldehyde and acetaldehyde are ubiquitously found in the environment and are also by-products of cellular metabolism. They are also
highly reactive, being able to form DNA adducts in vitro and
in vivo.63,64 Moreover, FA-deficient cells have been shown to be
hypersensitive to both of these compounds and to accumulate
double-strand breaks and chromosomal aberrations.65-68 Finally,
acetaldehyde treatment induces the activation of the FA pathway
in wild-type cells.69
To genetically test if endogenous aldehydes are a source of DNA
damage, our laboratory generated mice harboring disruptions of the
key FA gene Fancd2 in combination with aldehyde dehydrogenase 2
(Aldh2). Aldh2 oxidizes acetaldehyde to acetate, thereby preventing
the accumulation of this genotoxic agent. Therefore, if metabolically
produced aldehydes are indeed DNA-damaging agents normally
counteracted by the FA pathway, then the simultaneous disruption of
both Aldh2 and Fancd2 should have a synergistic effect on the
phenotype of these mice.65 Most strikingly, the presence of aldehyde
catabolism in the mother was found to be essential for the development of Aldh22/2Fancd22/2 embryos. Surprisingly, if the mother
expressed Aldh2 (Aldh21/2), then double mutant mice could be born,
suggesting that the mother was capable of breaking down
acetaldehyde and compensating for the Aldh2 deficiency in the fetus.
When born, the Aldh22/2Fancd22/2 mice had increased prevalence
of developmental abnormalities such as kinked tails or eye defects.
Most Aldh22/2Fancd22/2 mice succumbed to acute lymphoblastic
leukemia within the first 6 months of life. In an attempt to link
acetaldehyde to the phenotype of double knockout mice, these
animals were challenged with ethanol, a precursor of acetaldehyde. The treatment not only potentiated the developmental
abnormalities but also precipitated myelotoxicity in adult double
mutant mice.
Analysis of the hematopoietic compartment of double knockout
mice revealed a profound defect in the stem and progenitor cell pool,
with a more than 600-fold reduction in their HSC pool.70 It has been
known for a long time that HSCs possess high levels of aldehyde
dehydrogenase activity, and this was shown, in the case of mouse
30
GARAYCOECHEA and PATEL
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
Figure 4. A 2-tier protection mechanism preserves
HSC function. (A) HSCs display high Aldh2 activity
that protects their genome against the toxic effects of
reactive aldehydes. If aldehydes should evade this
protection mechanism, then DNA damage is dealt with
by the FA DNA repair pathway. (B) In Aldh22/2Fancd22/2
mice, the HSC pool is exposed to a greater burden of
reactive aldehydes, and in the absence of DNA repair,
DNA damage persists and leads to neoplastic transformation or HSC loss and spontaneous BMF.
HSCs, to be attributable to Aldh2. When this protection was taken
away, the HSCs were particularly compromised, as Aldh22/2
Fancd22/2 HSCs seemed to be far more sensitive to acetaldehyde
than more mature progenitors. This increased sensitivity also
correlated with the accumulation of DNA damage, as measured by
the induction of g-H2AX. Most strikingly, a small group of Aldh22/2
Fancd22/2 mice that did not get leukemia developed spontaneous
BMF. All of this data put together led us to the conclusion that BMF in
FA patients could result from endogenous aldehyde-induced toxicity,
which then leads to the depletion of HSCs (Figure 4). Despite this
recent discovery, the question remains as to why single FA knockout
mice so poorly replicate the human hematologic phenotype. It is only
upon exposure to the exogenous cross-linking agent MMC or by
taking away aldehyde catabolism in Fancd22/2 mice that the key
features of this human illness can be recapitulated. We provide some
speculations on this paradox.
First, FA patients show an age-dependent decrease in the frequency
of CD341 cells, which correlates with the worsening of clinical signs
and results in the onset of hematologic abnormalities at the median age
of 7.32 It is possible that the 2-year life span of mice does not allow for
the complete exhaustion of the stem cell pool. It would be interesting to
analyze Fanc-deficient mice to ask if there are age-related changes in
the quantity/quality of the HSC pool (Figure 3). In support of this, it was
shown that mice deficient in different genomic maintenance pathways
(NER, nonhomologous end joining [NHEJ], and telomere maintenance) show a severe decrease in the functional capacity of their HSCs
with age.71 Additionally, Fancc2/2 HSCs were found to perform
poorly at serial transplantation.43
Second, laboratory mice spend their lives in a highly controlled
environment with the same food source throughout their lives. It is
possible that this limits their exposure to exogenous DNA-damaging
agents. Finally, the murine metabolism could produce less endogenous genotoxins, or, alternatively, the protection against them
could be higher in mice than in humans. The metabolic rate of mice
is much greater than that of larger mammals.72 This could result in
the more rapid and efficient detoxification of toxic metabolites. It is
possible that it is necessary to take this protection away in order to
reveal the FA phenotype in mice.
An important prediction is that mutations in ALDH2 will lead to
more severe clinical features in FA patients. In humans, ALDH2 is
mutated in ;1 billion people. This dominant-negative polymorphism
(E487K) causes a dramatic decrease in ALDH2 activity, and, because it is most common in Southeast Asia, it is known as the
Asian flushing syndrome.73 Hira and colleagues have recently determined the ALDH2 genotype of a group of Japanese FA patients.
They found that ALDH2 deficiency dramatically accelerates BMF
and increases the frequency of malformation in some tissues. Most
strikingly, those patients entirely deficient for ALDH2 developed
BMF within the first 7 months of life. These results unequivocally
confirm that reactive aldehydes play an important role in the pathogenesis of FA.74
Although Aldh2 commonly oxidizes acetaldehyde, it has broad
substrate specificity, being able to oxidize other aldehydes including
4-hydroxynonenal, acrolein, propionaldehyde, and butyraldehyde.75
It remains to be clarified if endogenous acetaldehyde is solely
responsible for the phenotype observed in Aldh22/2Fancd22/2
mice. Conversely, Aldh2 is 1 member of a family of 18 enzymes,
any of which could also be offering protection against various
aldehydes.76 Finally, there is a synthetic lethal interaction between
FANCD2 and alcohol dehydrogenase 5 (ADH5) in the chicken B-cell
line DT40.68 Adh5 is responsible for the detoxification of formaldehyde, the simplest and most toxic aldehyde, which can be produced
close to DNA by means of DNA and histone demethylation. It will be
interesting to see what the consequences of Adh5 disruption in
Fanconi-deficient mice are.
Do the FA gene products modulate
cytokine responses?
So far, we have discussed the very strong biochemical and genetic
evidence for a function of the FA genes in DNA repair and how
this function relates to BMF. However, we have already alluded to
the unusual sensitivity that FA cells display to certain inflammatory
cytokines.8,9 Three crucial observations underpin this discovery:
First, FA cells are prone to apoptosis when exposed in vitro to
tumor necrosis factor a (TNF-a) and interferon g (IFN-g).9,77,78
Second, exposing FA-deficient mice to these cytokines results in
bone marrow dysfunction.79,80 Third, elevated levels of these
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
WHY DOES THE BONE MARROW FAIL IN FANCONI ANEMIA?
31
Figure 5. Models for BMF in FA. (A) The accumulation of DNA damage in the HSC pool leads to p53dependent HSC depletion by diminishing the ability
of HSCs to proliferate and self-renew. Endogenously
generated aldehydes are an important source of such
damage. (B) The molecular basis for cytokine overproduction and hypersensitivity by FA cells remains
unclear. However, some FA proteins were proposed to
have a direct role in hematopoiesis and to regulate the
response to inflammatory signals, independent of DNA
repair. (C) Reconciliation of the roles of FA proteins in
hematopoiesis. A defect in DNA repair could be the
initiating event that would lead to cell death, tissue
injury, and the production of inflammatory cytokines.
These could then contribute to bone marrow dysfunction in a number of ways. TNF-a and IFN-g can cause
apoptosis in stem cells and progenitors, mediated, for
example, by the Fas receptor.78 Alternatively, TNF-a
could produce additional DNA damage by the induction
of reactive molecules, like ROS.85 Finally, inflammatory
cytokines like TNF-a and IFN-g have classically been
considered to inhibit HSC and limit stem cell function.
However, recent studies are challenging this view and
propose a role for inflammatory signals in the direct
regulation of HSCs.86 It is conceivable that these
signals could lead to the differentiation of HSCs, either
directly or indirectly, causing them to encounter new
DNA damage in the context of replication.
and other cytokines have been reported in FA patients and are
overproduced by FA-deficient cells in vitro.13,81 These observations
underpin a general hypothesis that the hematopoietic phenotype of
FA is attributable to the overproduction of precisely the cytokines to
which FA cells are hypersensitive.
Most work on understanding the hypersensitivity of FA cells to
cytokine overproduction has been conducted with the FANCC gene
product, as well as cells derived from this complementation group
and Fancc2/2 knockout mice. This is probably because of the fact
that FANCC was the first FA gene to be cloned and its product was
initially shown to localize to the cytoplasm. FANCC has been
proposed to have a completely separate role in hematopoiesis and to
modulate cytokine responses by suppressing protein kinase R activation and its interactions with the chaperone HSP70 and STAT1,
a key effector of IFN-g signaling82-84 (Figure 5B85,86). It has been
suggested that understanding all potential functions of FA proteins
outside of DNA repair is a prerequisite for defining the pathogenesis
of marrow failure in FA. On the other hand, BMF is a universal
feature of FA, and this will be explained not by the independent
functions of some FA proteins, but by understanding the function
common to all 16 FA proteins (Figure 1).
Alanine scanning mutation of FANCC identified a putative conserved motif that mediates these interactions. Mutation of this motif
renders cells resistant to cross-linkers but sensitive to inflammatory
cytokines, suggesting that the cytokine sensitivity and DNA repair
function of FANCC can be decoupled. Cells bearing a naturally
occurring mutation of FANCC (c.67delG, previously c.322delG)
carry a hypomorphic 50-kDa FANCC polypeptide reinitiated at
methionine 55 (FP-50 or M55).87 The M55 polypeptide preserves
the conserved motif required for normal STAT1 activation and
HSP70 interaction but does not rescue the hypersensitivity to
MMC. However, patients with the c.67delG (c.322delG) mutation
have milder congenital malformations but share the hematologic
phenotype of other FA patients.88 This critical genetic observation
32
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
GARAYCOECHEA and PATEL
does not support a role for the conserved STAT1/HSP70 binding
motif in BMF in FA. Additionally, it remains to be explained how
the aberrant signaling by FANCC might contribute to cytokine
hypersensitivity and BMF in patients from other complementation
groups.8,80 Although it is clear that FA cells are sensitive to
inflammatory cytokines, it has not been emphatically established
that this is independent of defective DNA repair or, indeed, that
this points to a distinct role for the FA proteins in limiting the
production and/or in resistance to these molecules. Hopefully, in the
future, genetic dissection of the various components (cytokine, cytokine receptors, and downstream effector molecules), in combination
with FA knockout mice, should clarify these potentially important
questions.
The molecular basis for the overproduction of inflammatory
cytokines in FA is unclear. A key question in this respect is whether
this is a primary defect attributable to the function of the FA proteins
or if this is a secondary phenomenon. In fact, high levels of cytokines can be induced by cytotoxic agents.89 Recent work from
Matsui and colleagues has, for the first time, looked at cytokine
production by bone marrow cells from patients with different inherited BMF syndromes. This work contradicts previous studies
because no evidence for the overproduction of TNF-a or IFN-g by
unstimulated Fanconi-deficient T cells could be found. Cells from
FA patients tended toward higher cytokine levels following lipopolysaccharide treatment, but this was not specific to FA cells and
was seen in bone marrow samples from all syndromes.90 It is
therefore plausible that raised cytokines levels are a generic
response to bone marrow dysfunction, chronic inflammation, and
tissue injury (Figure 5C).
Future directions
Over the past decade, much evidence for the role of FA gene
products in DNA repair has been established. However, until
recently, we have had little insight into what damages the DNA in
the first place. The identification of reactive aldehydes as potent
genotoxins in FA-deficient HSCs strengthens the link between
DNA repair and the onset of BMF in FA. It will be crucial to define
the sources of these reactive molecules and how they can be
neutralized. However, it is not yet clear if these chemicals are the
only or indeed main drivers of endogenous DNA damage in HSCs.
Similar genetic approaches may reveal a role for other reactive
molecules as endogenous genotoxins.
At present, we do not completely understand the precise nature
of the DNA damage caused by aldehydes that is repaired by the FA
pathway, and whether it is direct or indirect. For instance, is the
lesion a DNA-DNA cross-link, DNA-protein cross-link, or simply
an adducted base? At a more mechanistic and fundamental level,
much work needs to be done to determine the biochemical and
structural basis of how endogenous DNA damage is repaired by the
FA proteins. All these potentially fertile avenues of research look
set to keep this fascinating rare bone failure syndrome at the center
of basic and translational biomedical research.
Acknowledgments
The authors would like to thank the members of the Patel laboratory
for critically reading the manuscript.
Authorship
Contribution: J.I.G. and K.J.P. wrote the paper and had final
approval of the submitted manuscript.
Conflict-of-interest disclosure: The authors declare no competing financial interests.
Correspondence: Juan I. Garaycoechea, MRC Laboratory of
Molecular Biology, Francis Crick Ave, Cambridge Biomedical
Campus, Cambridge CB2 0QH, United Kingdom; e-mail: juang@
mrc-lmb.cam.ac.uk; and K. J. Patel, MRC Laboratory of Molecular Biology, Francis Crick Ave, Cambridge Biomedical
Campus, Cambridge CB2 0QH, United Kingdom; e-mail: kjp@
mrc-lmb.cam.ac.uk.
References
1. Lobitz S, Velleuer E. Guido Fanconi (1892-1979):
a jack of all trades. Nat Rev Cancer. 2006;6(11):
893-898.
2. Auerbach AD, Wolman SR. Susceptibility of
Fanconi’s anaemia fibroblasts to chromosome
damage by carcinogens. Nature. 1976;261(5560):
494-496.
3. Sasaki MS, Tonomura A. A high susceptibility of
Fanconi’s anemia to chromosome breakage by
DNA cross-linking agents. Cancer Res. 1973;
33(8):1829-1836.
4. Auerbach AD. Fanconi anemia and its diagnosis.
Mutat Res. 2009;668(1-2):4-10.
5. Strathdee CA, Gavish H, Shannon WR, Buchwald
M. Cloning of cDNAs for Fanconi’s anaemia by
functional complementation. Nature. 1992;
356(6372):763-767.
8. Dufour C, Corcione A, Svahn J, et al. TNF-alpha
and IFN-gamma are overexpressed in the bone
marrow of Fanconi anemia patients and TNFalpha suppresses erythropoiesis in vitro. Blood.
2003;102(6):2053-2059.
9. Haneline LS, Broxmeyer HE, Cooper S, et al.
Multiple inhibitory cytokines induce deregulated
progenitor growth and apoptosis in hematopoietic
cells from Fac-/- mice. Blood. 1998;91(11):
4092-4098.
10. Anur P, Yates J, Garbati MR, et al. p38 MAPK
inhibition suppresses the TLR-hypersensitive
phenotype in FANCC- and FANCA-deficient
mononuclear phagocytes. Blood. 2012;119(9):
1992-2002.
11. Vanderwerf SM, Svahn J, Olson S, et al. TLR8dependent TNF-(alpha) overexpression in
Fanconi anemia group C cells. Blood. 2009;
114(26):5290-5298.
6. Yamashita T, Barber DL, Zhu Y, Wu N, D’Andrea
AD. The Fanconi anemia polypeptide FACC is
localized to the cytoplasm. Proc Natl Acad Sci
USA. 1994;91(14):6712-6716.
12. Wilson JB, Yamamoto K, Marriott AS, et al.
FANCG promotes formation of a newly identified
protein complex containing BRCA2, FANCD2 and
XRCC3. Oncogene. 2008;27(26):3641-3652.
7. Youssoufian H. Localization of Fanconi anemia C
protein to the cytoplasm of mammalian cells. Proc
Natl Acad Sci USA. 1994;91(17):7975-7979.
13. Sarkies P, Murat P, Phillips LG, Patel KJ,
Balasubramanian S, Sale JE. FANCJ coordinates
two pathways that maintain epigenetic stability at
G-quadruplex DNA. Nucleic Acids Res. 2012;
40(4):1485-1498.
14. Wilson JS, Tejera AM, Castor D, Toth R, Blasco
MA, Rouse J. Localization-dependent and
-independent roles of SLX4 in regulating
telomeres. Cell Rep. 2013;4(5):853-860.
15. Dao KH, Rotelli MD, Petersen CL, et al. FANCL
ubiquitinates b-catenin and enhances its nuclear
function. Blood. 2012;120(2):323-334.
16. Du W, Adam Z, Rani R, Zhang X, Pang Q.
Oxidative stress in Fanconi anemia
hematopoiesis and disease progression. Antioxid
Redox Signal. 2008;10(11):1909-1921.
17. Pace P, Johnson M, Tan WM, et al. FANCE: the
link between Fanconi anaemia complex assembly
and activity. EMBO J. 2002;21(13):3414-3423.
18. Garcia-Higuera I, Taniguchi T, Ganesan S, et al.
Interaction of the Fanconi anemia proteins and
BRCA1 in a common pathway. Mol Cell. 2001;
7(2):249-262.
19. Howlett NG, Taniguchi T, Olson S, et al. Biallelic
inactivation of BRCA2 in Fanconi anemia.
Science. 2002;297(5581):606-609.
20. Kim Y, Lach FP, Desetty R, Hanenberg H,
Auerbach AD, Smogorzewska A. Mutations of the
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
SLX4 gene in Fanconi anemia. Nat Genet. 2011;
43(2):142-146.
21. Meetei AR, Medhurst AL, Ling C, et al. A human
ortholog of archaeal DNA repair protein Hef is
defective in Fanconi anemia complementation
group M. Nat Genet. 2005;37(9):958-963.
22. Ling C, Ishiai M, Ali AM, et al. FAAP100 is
essential for activation of the Fanconi anemiaassociated DNA damage response pathway.
EMBO J. 2007;26(8):2104-2114.
23. Alpi AF, Pace PE, Babu MM, Patel KJ.
Mechanistic insight into site-restricted
monoubiquitination of FANCD2 by Ube2t, FANCL,
and FANCI. Mol Cell. 2008;32(6):767-777.
24. Hirano S, Yamamoto K, Ishiai M, et al. Functional
relationships of FANCC to homologous
recombination, translesion synthesis, and BLM.
EMBO J. 2005;24(2):418-427.
25. Niedzwiedz W, Mosedale G, Johnson M, Ong CY,
Pace P, Patel KJ. The Fanconi anaemia gene
FANCC promotes homologous recombination and
error-prone DNA repair. Mol Cell. 2004;15(4):
607-620.
26. Kashiyama K, Nakazawa Y, Pilz DT, et al.
Malfunction of nuclease ERCC1-XPF results
in diverse clinical manifestations and causes
Cockayne syndrome, xeroderma pigmentosum,
and Fanconi anemia. Am J Hum Genet. 2013;
92(5):807-819.
27. Bogliolo M, Schuster B, Stoepker C, et al.
Mutations in ERCC4, encoding the DNA-repair
endonuclease XPF, cause Fanconi anemia. Am J
Hum Genet. 2013;92(5):800-806.
28. Joo W, Xu G, Persky NS, et al. Structure of the
FANCI-FANCD2 complex: insights into the
Fanconi anemia DNA repair pathway. Science.
2011;333(6040):312-316.
29. Raschle
¨
M, Knipscheer P, Enoiu M, et al.
Mechanism of replication-coupled DNA
interstrand crosslink repair [published correction
appears in Cell. 2009;137(5):972]. Cell. 2008;
134(6):969-980.
30. Knipscheer P, Raschle
¨
M, Smogorzewska A,
et al. The Fanconi anemia pathway promotes
replication-dependent DNA interstrand cross-link
repair. Science. 2009;326(5960):1698-1701.
31. Long DT, Raschle
¨
M, Joukov V, Walter JC.
Mechanism of RAD51-dependent DNA
interstrand cross-link repair. Science. 2011;
333(6038):84-87.
32. Butturini A, Gale RP, Verlander PC, AdlerBrecher B, Gillio AP, Auerbach AD. Hematologic
abnormalities in Fanconi anemia: an International
Fanconi Anemia Registry study. Blood. 1994;
84(5):1650-1655.
33. Kutler DI, Singh B, Satagopan J, et al. A 20-year
perspective on the International Fanconi Anemia
Registry (IFAR). Blood. 2003;101(4):1249-1256.
34. Kelly PF, Radtke S, von Kalle C, et al. Stem cell
collection and gene transfer in Fanconi anemia.
Mol Ther. 2007;15(1):211-219.
35. Ceccaldi R, Parmar K, Mouly E, et al. Bone
marrow failure in Fanconi anemia is triggered by
an exacerbated p53/p21 DNA damage response
that impairs hematopoietic stem and progenitor
cells. Cell Stem Cell. 2012;11(1):36-49.
36. Tulpule A, Lensch MW, Miller JD, et al.
Knockdown of Fanconi anemia genes in human
embryonic stem cells reveals early developmental
defects in the hematopoietic lineage. Blood. 2010;
115(17):3453-3462.
37. Carreau M, Gan OI, Liu L, et al. Bone marrow
failure in the Fanconi anemia group C mouse
model after DNA damage. Blood. 1998;91(8):
2737-2744.
38. Bakker ST, de Winter JP, te Riele H. Learning
from a paradox: recent insights into Fanconi
WHY DOES THE BONE MARROW FAIL IN FANCONI ANEMIA?
anaemia through studying mouse models. Dis
Model Mech. 2013;6(1):40-47.
39. Crossan GP, van der Weyden L, Rosado IV, et al;
Sanger Mouse Genetics Project. Disruption of
mouse Slx4, a regulator of structure-specific
nucleases, phenocopies Fanconi anemia. Nat
Genet. 2011;43(2):147-152.
40. Pulliam-Leath AC, Ciccone SL, Nalepa G, et al.
Genetic disruption of both Fancc and Fancg in
mice recapitulates the hematopoietic
manifestations of Fanconi anemia. Blood. 2010;
116(16):2915-2920.
41. Haneline LS, Gobbett TA, Ramani R, et al. Loss of
FancC function results in decreased
hematopoietic stem cell repopulating ability.
Blood. 1999;94(1):1-8.
33
57. Zhang S, Yajima H, Huynh H, et al. Congenital
bone marrow failure in DNA-PKcs mutant mice
associated with deficiencies in DNA repair. J Cell
Biol. 2011;193(2):295-305.
58. Ruzankina Y, Pinzon-Guzman C, Asare A, et al.
Deletion of the developmentally essential gene
ATR in adult mice leads to age-related
phenotypes and stem cell loss. Cell Stem Cell.
2007;1(1):113-126.
59. Joenje H, Arwert F, Eriksson AW, de Koning H,
Oostra AB. Oxygen-dependence of chromosomal
aberrations in Fanconi’s anaemia. Nature. 1981;
290(5802):142-143.
60. Schindler D, Hoehn H. Fanconi anemia mutation
causes cellular susceptibility to ambient oxygen.
Am J Hum Genet. 1988;43(4):429-435.
42. Zhang QS, Marquez-Loza L, Eaton L, et al.
Fancd2-/- mice have hematopoietic defects that
can be partially corrected by resveratrol. Blood.
2010;116(24):5140-5148.
61. Muller
¨
LU, Milsom MD, Harris CE, et al.
Overcoming reprogramming resistance of
Fanconi anemia cells. Blood. 2012;119(23):
5449-5457.
43. Kamimae-Lanning AN, Goloviznina NA, Kurre P.
Fetal origins of hematopoietic failure in a murine
model of Fanconi anemia. Blood. 2013;121(11):
2008-2012.
62. Hadjur S, Ung K, Wadsworth L, et al. Defective
hematopoiesis and hepatic steatosis in mice with
combined deficiencies of the genes encoding
Fancc and Cu/Zn superoxide dismutase. Blood.
2001;98(4):1003-1011.
44. 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(2):125-131.
45. Bonnet D, Dick JE. Human acute myeloid
leukemia is organized as a hierarchy that
originates from a primitive hematopoietic cell. Nat
Med. 1997;3(7):730-737.
46. Nilsson L, Astrand-Grundstr¨om I, Arvidsson I,
et al. Isolation and characterization of
hematopoietic progenitor/stem cells in 5q-deleted
myelodysplastic syndromes: evidence for
involvement at the hematopoietic stem cell level.
Blood. 2000;96(6):2012-2021.
63. Garcia CC, Angeli JP, Freitas FP, et al. [13C2]Acetaldehyde promotes unequivocal formation of
1,N2-propano-29-deoxyguanosine in human cells.
J Am Chem Soc. 2011;133(24):9140-9143.
64. Wang M, McIntee EJ, Cheng G, Shi Y, Villalta
PW, Hecht SS. Identification of DNA adducts of
acetaldehyde. Chem Res Toxicol. 2000;13(11):
1149-1157.
65. Langevin F, Crossan GP, Rosado IV, Arends MJ,
Patel KJ. Fancd2 counteracts the toxic effects of
naturally produced aldehydes in mice. Nature.
2011;475(7354):53-58.
47. Nilsson L, Eden
´ P, Olsson E, et al. The molecular
signature of MDS stem cells supports a stem-cell
origin of 5q myelodysplastic syndromes. Blood.
2007;110(8):3005-3014.
66. Mechilli M, Schinoppi A, Kobos K, Natarajan AT,
Palitti F. DNA repair deficiency and acetaldehydeinduced chromosomal alterations in CHO cells.
Mutagenesis. 2008;23(1):51-56.
48. Welch JS, Ley TJ, Link DC, et al. The origin and
evolution of mutations in acute myeloid leukemia.
Cell. 2012;150(2):264-278.
67. Ridpath JR, Nakamura A, Tano K, et al. Cells
deficient in the FANC/BRCA pathway are
hypersensitive to plasma levels of formaldehyde.
Cancer Res. 2007;67(23):11117-11122.
49. Chen W, Kumar AR, Hudson WA, et al. Malignant
transformation initiated by Mll-AF9: gene dosage
and critical target cells. Cancer Cell. 2008;13(5):
432-440.
50. Rosenberg PS, Huang Y, Alter BP. Individualized
risks of first adverse events in patients with
Fanconi anemia. Blood. 2004;104(2):350-355.
51. Freie B, Li X, Ciccone SL, et al. Fanconi anemia
type C and p53 cooperate in apoptosis and
tumorigenesis. Blood. 2003;102(12):4146-4152.
52. Houghtaling S, Granville L, Akkari Y, et al.
Heterozygosity for p53 (Trp531/-) accelerates
epithelial tumor formation in Fanconi anemia
complementation group D2 (Fancd2) knockout
mice. Cancer Res. 2005;65(1):85-91.
53. Zhang QS, Watanabe-Smith K, Schubert K, et al.
Fancd2 and p21 function independently in
maintaining the size of hematopoietic stem and
progenitor cell pool in mice. Stem Cell Res
(Amst). 2013;11(2):687-692.
54. Vinciguerra P, Godinho SA, Parmar K, Pellman D,
D’Andrea AD. Cytokinesis failure occurs in
Fanconi anemia pathway-deficient murine and
human bone marrow hematopoietic cells. J Clin
Invest. 2010;120(11):3834-3842.
55. Milyavsky M, Gan OI, Trottier M, et al. A
distinctive DNA damage response in human
hematopoietic stem cells reveals an apoptosisindependent role for p53 in self-renewal. Cell
Stem Cell. 2010;7(2):186-197.
56. Niedernhofer LJ. DNA repair is crucial for
maintaining hematopoietic stem cell function.
DNA Repair (Amst). 2008;7(3):523-529.
68. Rosado IV, Langevin F, Crossan GP, Takata M,
Patel KJ. Formaldehyde catabolism is essential in
cells deficient for the Fanconi anemia DNA-repair
pathway. Nat Struct Mol Biol. 2011;18(12):
1432-1434.
69. Marietta C, Thompson LH, Lamerdin JE,
Brooks PJ. Acetaldehyde stimulates FANCD2
monoubiquitination, H2AX phosphorylation, and
BRCA1 phosphorylation in human cells in vitro:
implications for alcohol-related carcinogenesis.
Mutat Res. 2009;664(1-2):77-83.
70. Garaycoechea JI, Crossan GP, Langevin F,
Daly M, Arends MJ, Patel KJ. Genotoxic
consequences of endogenous aldehydes on
mouse haematopoietic stem cell function. Nature.
2012;489(7417):571-575.
71. Rossi DJ, Bryder D, Seita J, Nussenzweig A,
Hoeijmakers J, Weissman IL. Deficiencies in DNA
damage repair limit the function of haematopoietic
stem cells with age. Nature. 2007;447(7145):
725-729.
72. Hayssen V, Lacy RC. Basal metabolic rates in
mammals: taxonomic differences in the allometry
of BMR and body mass. Comp Biochem Physiol
A. 1985;81(4):741-754.
73. Larson HN, Weiner H, Hurley TD. Disruption of
the coenzyme binding site and dimer interface
revealed in the crystal structure of mitochondrial
aldehyde dehydrogenase “Asian” variant. J Biol
Chem. 2005;280(34):30550-30556.
74. Hira A, Yabe H, Yoshida K, et al. Variant ALDH2
is associated with accelerated progression of
34
GARAYCOECHEA and PATEL
bone marrow failure in Japanese Fanconi anemia
patients. Blood. 2013;122(18):3206-3209.
75. Klyosov AA. Kinetics and specificity of human
liver aldehyde dehydrogenases toward aliphatic,
aromatic, and fused polycyclic aldehydes.
Biochemistry. 1996;35(14):4457-4467.
BLOOD, 2 JANUARY 2014 x VOLUME 123, NUMBER 1
80. Si Y, Ciccone S, Yang FC, et al. Continuous in
vivo infusion of interferon-gamma (IFN-gamma)
enhances engraftment of syngeneic wild-type
cells in Fanca-/- and Fancg-/- mice. Blood. 2006;
108(13):4283-4287.
76. Marchitti SA, Brocker C, Stagos D, Vasiliou V.
Non-P450 aldehyde oxidizing enzymes: the
aldehyde dehydrogenase superfamily. Expert
Opin Drug Metab Toxicol. 2008;4(6):697-720.
81. Rosselli F, Sanceau J, Gluckman E, Wietzerbin J,
Moustacchi E. Abnormal lymphokine production:
a novel feature of the genetic disease Fanconi
anemia. II. In vitro and in vivo spontaneous
overproduction of tumor necrosis factor alpha.
Blood. 1994;83(5):1216-1225.
77. Rathbun RK, Faulkner GR, Ostroski MH, et al.
Inactivation of the Fanconi anemia group C gene
augments interferon-gamma-induced apoptotic
responses in hematopoietic cells. Blood. 1997;
90(3):974-985.
82. Pang Q, Fagerlie S, Christianson TA, et al. The
Fanconi anemia protein FANCC binds to and
facilitates the activation of STAT1 by gamma
interferon and hematopoietic growth factors. Mol
Cell Biol. 2000;20(13):4724-4735.
78. Wang J, Otsuki T, Youssoufian H, et al.
Overexpression of the Fanconi anemia group C
gene (FAC) protects hematopoietic progenitors
from death induced by Fas-mediated apoptosis.
Cancer Res. 1998;58(16):3538-3541.
83. Pang Q, Keeble W, Christianson TA, Faulkner
GR, Bagby GC. FANCC interacts with Hsp70 to
protect hematopoietic cells from IFN-gamma/
TNF-alpha-mediated cytotoxicity. EMBO J. 2001;
20(16):4478-4489.
79. Li X, Yang Y, Yuan J, et al. Continuous in vivo
infusion of interferon-gamma (IFN-gamma)
preferentially reduces myeloid progenitor
numbers and enhances engraftment of syngeneic
wild-type cells in Fancc-/- mice. Blood. 2004;
104(4):1204-1209.
84. Pang Q, Keeble W, Diaz J, et al. Role of doublestranded RNA-dependent protein kinase in
mediating hypersensitivity of Fanconi anemia
complementation group C cells to interferon
gamma, tumor necrosis factor-alpha, and doublestranded RNA. Blood. 2001;97(6):1644-1652.
85. Sejas DP, Rani R, Qiu Y, et al. Inflammatory
reactive oxygen species-mediated hemopoietic
suppression in Fancc-deficient mice. J Immunol.
2007;178(8):5277-5287.
86. King KY, Goodell MA. Inflammatory modulation of
HSCs: viewing the HSC as a foundation for the
immune response. Nat Rev Immunol. 2011;
11(10):685-692.
87. Yamashita T, Wu N, Kupfer G, et al. Clinical
variability of Fanconi anemia (type C) results from
expression of an amino terminal truncated
Fanconi anemia complementation group C
polypeptide with partial activity. Blood. 1996;
87(10):4424-4432.
88. 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(1):105-110.
89. Cachac¸o AS, Carvalho T, Santos AC, et al. TNFalpha regulates the effects of irradiation in the
mouse bone marrow microenvironment. PLoS
ONE. 2010;5(2):e8980.
90. Matsui K, Giri N, Alter BP, Pinto LA. Cytokine
production by bone marrow mononuclear cells in
inherited bone marrow failure syndromes. Br J
Haematol. 2013;163(1):81-92.