FANCONI ANEMIA (FA) is a rare autosomal recessive disease characterized by multiple congenital abnormalities, bone marrow (BM) failure, and cancer susceptibility. The mean age of onset of anemia is 8 years, and the mean survival is 16 years. Death usually results from complications of BM failure. Considerable progress in the field of FA research has resulted from the recent cloning of two of the five known FA genes. The purpose of the current review is to describe the structure and function of the FA genes and to outline novel approaches to FA diagnosis and therapy, based on the availability of these genes.

The congenital abnormalities and clinical course of FA have been extensively reviewed.1-9 Patients with FA have growth retardation and abnormalities of the skin (generalized hyperpigmentation and/or café au lait spots), upper extremities (frequently with defects in the thumbs or forearms), kidneys, and gastrointestinal system. Male FA patients have underdeveloped gonads and defective spermatogenesis. The large range of organ systems affected in FA implicates the FA genes in a general developmental process required during normal human embryogenesis.

The hematologic complications of FA have also been extensively reviewed.1,4 10 FA patients develop macrocytosis and pancytopenia, typically during the first decade of life. Deficiencies in platelets or red blood cells usually precede white blood cell abnormalities. The patients have “fetal-like” erythropoiesis, with increased i antigen and hemoglobin F, and generally have high serum erythropoietin levels. The progression to pancytopenia is highly varied among FA patients.

At least 20% of patients with FA develop cancers.1 Because many FA patients die of other causes before they might have developed cancer, the actuarial risk of cancer is even higher.11 FA patients primarily develop acute myeloblastic leukemia; however, cancers of several organ systems including the skin, gastrointestinal, and gynecological systems have been described. The skin and gastrointestinal tumors are usually squamous cell carcinomas.12-14 In addition, FA patients receiving androgen therapy for BM failure are prone to liver tumors. Cancer tends to be a disease of older FA patients. The average age of patients who develop cancers is 15 years for leukemia, 16 years for liver tumors, and 23 years for other tumors. There is a slightly higher risk of cancer for female FA patients, irrespective of the increased risk of gynecological cancers.15 

The diagnosis of FA exploits the sensitivity of FA cells to bifunctional alkylating agents. FA cells have increased spontaneous chromosomal breakage that is amplified by the addition of the cross-linking agents, diepoxybutane (DEB) or mitomycin C (MMC). Similar spontaneous, but not DEB-induced, chromosomal changes are observed in Bloom's syndrome and ataxia telangiectasia.16,17 The DEB test is a highly sensitive and specific test for FA.18 Although useful in the diagnosis of FA patients, the DEB test does not distinguish FA carriers from the general population. Also, the DEB test can give false-negative results, particularly in patients with cellular mosacism.19 The DEB test has been used successfully in prenatal diagnosis of FA. Prenatal diagnosis has been performed by chorionic villus sampling (CVS) at 9 to 12 weeks of gestation or by amniocentesis at 16 weeks in many cases of suspected FA.20 21 

The diagnosis of FA is complicated by the wide variability in FA patient phenotype.2 The differential diagnosis of FA includes other genetic syndromes including neurofibromatosis, VACTERL association, and TAR (thrombocytopenia with absence of radii). Although the DEB test is highly effective in discriminating FA from these syndromes, the test remains underutilized.18 Further confounding diagnosis, approximately 33% of patients with FA have no obvious congenital abnormalities.6 These FA patients may first present with BM failure or cancer.22 Despite this range in phenotypic variation, there exists no obvious correlation between the severity of the disease and the extent of cellular sensitivity to DEB.

The treatment of FA is similar, but not identical, to the treatment of other forms of acquired aplastic anemia. Patients are treated with supportive care (ie, blood transfusions) for their BM failure. FA patients respond transiently to therapy with androgens and the cytokines, granulocyte-macrophage colony-stimulating factor (GM-CSF )23 and G-CSF.24 The treatment of choice for FA is allogenic BM transplant with a histocompatible sibling donor. In one recent study,25 18 patients with FA had allogeneic BM transplants from matched sibling donors (MSD). Seventeen patients had sustained engraftment and transfusion independence. Still, FA patients have severe toxicity from graft-versus-host disease (GVHD), perhaps resulting from an increased cellular sensitivity to endogenous cytokines released during GVHD. T-cell depletion has reduced GVHD for BM transplantation from unrelated donors.26 Umbilical cord blood also offers a potential source of hematopoietic stem cells for FA patients without sibling matches.27-31 

Leukemia in FA patients differs significantly from leukemia in the general pediatric and adolescent populations.1,22,32 Treatment of leukemia in FA patients is largely ineffectual, with many patients dying within 1 to 3 months of diagnosis. Many of the FA patients who develop leukemia have pre-existing myelodysplastic syndrome, or preleukemia. Many of these patients have cytogenetic abnormalities in chromosomes 1 and 733,34 resulting from complete or partial chromosomal deletion or translocation. Leukemic cells from FA patients usually do not have abnormalities in the p53 gene.35 In one case, when a leukemic FA patient did achieve a remission, the marrow remained myelodysplastic.22 

Because of the cellular sensitivity to cross-linking agents, FA is often compared with other syndromes of drug sensitivity and genomic instability, including ataxia telangiectasia (AT), Xeroderma pigmentosum (XP), Cockayne syndrome (CS),36,37 Bloom's syndrome, and HNPCC38 (Table 1). The eight genes for XP encode proteins that comprise a DNA excision repair complex.39,40 ATM, the gene mutated in the inherited human disease ataxia-telangiectasia (AT), encodes a protein with homology to PI-3 kinase,41 suggesting that the ATM protein plays a role in sensing DNA damage and signaling the induction of p53.42 More recent evidence shows that the ATM protein product associates with meiotically pairing chromosomes.43,44 The gene for Bloom's syndrome encodes a protein related to a known yeast protein that regulates the cell cycle.45 46 Therefore, the Bloom's syndrome protein may regulate a G2/M checkpoint in the normal mammalian cell cycle. The genomic instability of these syndromes may result from a cellular defect in one of several processes, including DNA repair, cell-cycle regulation, or DNA replication.

In addition to cross-linking agent sensitivity, FA cells have several other phenotypic abnormalities3,10 47-68 (outlined in Table 2). A detailed description of these studies is beyond the scope of this review. Many of these cellular assays have been performed on FA cells from multiple complementation groups. Accordingly, it remains unclear whether these cellular abnormalities correspond to all FA complementation groups or to only a subset. Most of the abnormalities described for FA cells are probably epiphenomena and do not relate directly to the primary cellular defect in each FA complementation group. Therefore, a true understanding of the primary defect in FA, such as DNA repair, cell-cycle regulation, or prevention of apoptosis, can only come from a molecular understanding of the newly cloned FA proteins.

The complementation analysis of FA cells, using somatic cell fusion studies, has allowed the identification of at least five complementation groups.69-73 At least three of the five groups (A, C, and D) map to discrete chromosomal loci72,74-76 (Table 3). Therefore, FA is a genetically heterogenous disorder, unlike the syndromes, ataxia-telangiectasia (AT)41 and Bloom's syndrome,45 which arise from mutations in single genes.

Fig. 1.

Mutations of the FAC polypeptide found in patients with FA. The wild-type FAC protein (558 amino acids) is shown schematically at the top. The indicated FAC alleles are also shown. The IVS4+4 A < T mutant allele encodes a protein with an in-frame deletion of 37 amino acids.80,91 The 322delG has a frame shift mutation in exon 1.77 This allele encodes an amino terminal truncated FAC protein with translation reinitiation from methionine 55.81 The IVS4+4 A < T mutation and 322delG account for approximately 75% of FAC mutations. Less common mutant alleles of FAC are also shown. At least three of these mutations fall in exon 14, encoding the carboxy terminus of the FAC protein. An allele with a C to T nucleotide substitution at base 808 (exon 6) is predicted to encode a protein with premature termination at amino acid 185.83 The 1806insA mutation is predicted to encode a truncated protein of 526 amino acids.86 A nucleotide substitution of T to C, at base 1913, encodes a protein with an (L554P) mutation.77 Recent studies suggest that this mutant protein is expressed in FAC cell lines91 and has dominant negative activity when overexpressed.94 This allele is the only known missense mutant and suggests that the carboxy terminus of FAC has functional relevance (see text). The relative frequency of these various FAC alleles has recently been described.82 

Fig. 1.

Mutations of the FAC polypeptide found in patients with FA. The wild-type FAC protein (558 amino acids) is shown schematically at the top. The indicated FAC alleles are also shown. The IVS4+4 A < T mutant allele encodes a protein with an in-frame deletion of 37 amino acids.80,91 The 322delG has a frame shift mutation in exon 1.77 This allele encodes an amino terminal truncated FAC protein with translation reinitiation from methionine 55.81 The IVS4+4 A < T mutation and 322delG account for approximately 75% of FAC mutations. Less common mutant alleles of FAC are also shown. At least three of these mutations fall in exon 14, encoding the carboxy terminus of the FAC protein. An allele with a C to T nucleotide substitution at base 808 (exon 6) is predicted to encode a protein with premature termination at amino acid 185.83 The 1806insA mutation is predicted to encode a truncated protein of 526 amino acids.86 A nucleotide substitution of T to C, at base 1913, encodes a protein with an (L554P) mutation.77 Recent studies suggest that this mutant protein is expressed in FAC cell lines91 and has dominant negative activity when overexpressed.94 This allele is the only known missense mutant and suggests that the carboxy terminus of FAC has functional relevance (see text). The relative frequency of these various FAC alleles has recently been described.82 

Close modal

FAC was cloned by functional complementation of an Epstein-Barr virus (EBV)-immortalized type C FA cell line, HSC536.77 As predicted by the complementation test, the FAC cDNA corrects the MMC sensitivity and DEB sensitivity of FA(C) cell lines, but does not correct the MMC sensitivity of FA cells derived from type A, B, D, or E patients. Cells derived from patients with FA(C) have mutations in both alleles of the FAC gene, consistent with the autosomal recessive inheritance of the FA syndrome. The FAC cDNA encodes a 558–amino acid polypeptide (63 kD) with no homology to other proteins in GenBank. Moreover, the FAC gene is composed of 14 exons,78 spans approximately 80 kb, and maps to human chromosome 9q22.3.78 The murine FAC gene has been isolated, but no FAC homologues from other species have been reported. Murine FAC is only 66% identical to the human protein, but is able to functionally complement human cells from FA(C) patients.79 

Mutational analysis of the FAC gene has shown a relatively small number of characteristic mutations, represented in specific ancestral backgrounds (Fig 1). The IVS4+4 A > T mutation is found in patients of Ashkenazi-Jewish ancestry and accounts for greater than 80% of FA in this population.80 Patients homozygous for this mutation have more severe FA, with multiple congenital abnormalities and early onset of hematological disease.81,82 The 322delG mutation is found in patients of Northern European ancestry, particularly from Holland. Patients homozygous for this mutation have a comparatively mild FA, with fewer congenital abnormalities and later onset of hematological disease.82 Additional mutant alleles of FAC have been identified with mutations in exon 1,82 exon 6,83 and exon 14.77 84-86 A total of seven pathogenic mutations have now been identified, of which three are located in exon 14. Most of these mutations result in either truncation or internal deletion of the FAC protein. Only one pathogenic missense mutation (L554P) has been described. An additional change (D195V) is a polymorphism82; the FAC polypeptide containing this amino acid substitution has normal activity when expressed in an FA(C) indicator cell line (A. D'Andrea, unpublished observation, January 1997).

Analysis of the FAC mRNA and protein have provided some insight into the cellular function of the FAC gene. The FAC mRNA is expressed in multiple cell types and organ systems, consistent with a general function of FAC in organismal development. Increased expression of the FAC mRNA has been observed in the skeletal system, suggesting a more specialized function of FAC in bone development.87 The FAC mRNA exists as three different sizes of 2.3, 3.2, and 4.6 kb.77 The existence of multiple mRNA transcripts appears to result, at least in part, from variable lengths of the 3′ untranslated region. In addition, there is evidence that the FAC mRNA undergoes alternative splicing and exon skipping.83 For instance, a naturally occurring mRNA splice variant, lacking exon 13 sequences, has been detected by reverse transcriptase-polymerase chain reaction (RT-PCR) in normal and FA cell lines. The functional significance of these mRNA splice variants and encoded proteins remains unknown.

The FAC gene contains at least two alternative transcription initiation sites, suggesting the possibility of alternative mechanisms of transcriptional regulation.88 Still, there is little evidence for variable expression of the FAC mRNA under different physiological conditions. The FAC mRNA is not induced by MMC and is not differentially expressed during cell-cycle progression. Recent evidence shows some cell variation of FAC protein expression,89 suggesting possible posttranslational mechanisms of FAC regulation.

The FAC protein is a soluble cytoplasmic protein,90,91 suggesting that it does not play a direct role in DNA repair. The protein remains in the cytoplasm, irrespective of cell-cycle stage or cellular treatment with cross-linking agents. If the FAC protein does interact with DNA directly, it may do so during a narrow window of time in the cell cycle, when the nuclear membrane is degraded. Recent evidence suggests that FAC binds to a complex of cytoplasmic proteins of either 60, 50, or 34 kD.92 The level of FAC protein varies slightly during cell-cycle progression, reaching peak expression during the G2/M transition of the cell cycle. Other evidence shows that the FAC protein binds to the mitotic cyclin-dependent kinase cdc2, suggesting that it has a cellular function that is regulated during the G2/M transition.89 The interaction of FAC with cdc2 suggests a molecular mechanism for the prolonged G2 phase of FA cells.49,51 For instance, the FAC protein may be required for normal cdc2 activity during G2/M progression. Alternatively, the FAC protein may be part of a DNA repair pathway downstream of the cdc2 kinase. In addition, the FAC protein appears to bind to GRP94.93 GRP94 is a 94-kD member of the molecular chaperone family and is expressed in the endoplasmic reticulum where it plays a role in protein transport. The precise cellular function of the FAC protein or the importance of these molecular interactions remains unclear.

The mutant FAC proteins expressed by FA(C) patients have allowed the identification of critical functional domains of FAC. All FAC cell lines analyzed to date express at least some isoform of the FAC protein.81 No patient-derived cell lines with true null mutations in the FAC gene have been identified, suggesting that at least partial FAC protein function may be required for viability. Patients homozygous for the IVS4+4 A > T mutation [severe FA(C)] express a truncated 55-kD isoform of the FAC protein. This protein is an “in-frame” deletion of the FAC protein, missing 37 amino acids encoded by exon 4. In contrast, patients homozygous for the 322delG mutation (mild FAC) express a truncated 50-kD isoform of the FAC protein. This protein results from translation reinitiation at an internal methionine residue, downstream of the exon 1 mutation. This 50-kD form of FAC is also expressed in normal cells, suggesting that the internal reinitiation product may have a normal cellular function.81,91 The 55-kD and 50-kD isoforms of FAC, although expressed at normal levels in FA cells, fail to confer MMC resistance. These results suggest that these N-terminal–deleted domains of the FAC protein are required for normal FAC function in vivo. In addition, the HSC536 cells (described above) express the abnormal FAC(L554P) protein. This mutant protein does not confer normal MMC resistance85,91 and may have dominant inhibitory activity when overexpressed with the normal FAC polypeptide.94 This mutation, and several other patient-derived mutations in exon 14 (Fig 1), suggest that the C-terminus of the FAC protein is critical for its in vivo function. Further deletion analysis of the C-terminus of the FAC protein confirms the functional importance of the C-terminus. How these critical functional domains of the FAC protein relate to its intracellular function remains unknown. For instance, these regions of the FAC protein may be required for its ability to bind to other critical regulatory proteins in the cell. Recent evidence shows that the C-terminus of FAC binds to cdc289 while a central region binds to GRP94.93 

The cellular function of the FAC polypeptide has been analyzed by comparing isogenic cell lines, differing only by the expression of the normal FAC protein.60,77 95 Expression of the wild-type FAC protein in FA(C) cells corrects several phenotypic abnormalities of the parental cell line, including its MMC sensitivity, its G2 accumulation, and its chromosomal instability. Although overexpression of FAC corrects the MMC sensitivity of FA(C) cells to a normal (wild-type) level, it does not render cells superresistant to MMC. Accordingly, it is unlikely that the FAC protein performs an enzymatic role in the cell, such as efflux of MMC or scavenging of MMC or oxygen radical byproducts.

Interestingly, MMC-sensitive cells acquire resistance to MMC by multiple mechanisms, any one of which may be related to FAC. First, overexpression of the P-glycoprotein, p120, confers cellular resistance to MMC.96 Second, mitomycin C requires bioreduction to exert its cytoxic effect.97 Several cellular enzymes, including NAD(P)H:quinone oxidoreductase, are required for the activation of MMC to its ultimate DNA damaging state. Cellular inactivation of NAD(P)H:quinone oxidoreductase results in decreased MMC-generated DNA damage and increased cellular resistance to MMC.98 Third, several investigators have found that overexpression of ribosomal protein S3 can partially rescue the phenotype of FA cells99 (M. Kelley, personal communication, November 1996). Whether these mechanisms of cellular resistance to MMC bear any relevance to the FA gene pathway remains unknown.

Fig. 2.

Phenotypes of FAC knockout mice. Panels (A) (wild-type control) and (B) (FAC knockout mouse) show the results of chromosome breakage analysis. Cells were exposed to 20 ng/mL MMC for 48 hours. Multiple radial formation (arrows), gaps, and breaks can be seen in the mutant, but not control cells. Testicular histology of age-matched wild-type control (C) and FAC knockout mouse (D) at the same magnification. The normal control shows the cross-section of a seminiferous tubule containing all stages of spermatogenesis. In the mutant animal, two small abnormal seminiferous tubules can be seen. Spermatogenesis is absent and only Sertoli cells remain. Hypertrophy of interstitial tissue is also evident.

Fig. 2.

Phenotypes of FAC knockout mice. Panels (A) (wild-type control) and (B) (FAC knockout mouse) show the results of chromosome breakage analysis. Cells were exposed to 20 ng/mL MMC for 48 hours. Multiple radial formation (arrows), gaps, and breaks can be seen in the mutant, but not control cells. Testicular histology of age-matched wild-type control (C) and FAC knockout mouse (D) at the same magnification. The normal control shows the cross-section of a seminiferous tubule containing all stages of spermatogenesis. In the mutant animal, two small abnormal seminiferous tubules can be seen. Spermatogenesis is absent and only Sertoli cells remain. Hypertrophy of interstitial tissue is also evident.

Close modal

Conflicting studies have analyzed the possible interaction of FAC with the p53 pathway. p53 is a major cellular determinant of genomic stability.100 Some studies61 suggest that FA cells, like AT cells,42 are defective in the induction of p53. Other studies suggest that FA cells are functional for p53 induction.60,95 In more recent studies, overexpression of normal FAC protein in FA(C) cells corrected a cellular defect in apoptosis.64,65 These results implicate the FAC polypeptide in a general function in the prevention of cellular apoptosis. Consistent with these studies, anti-sense oligonucleotides to FAC inhibit hematopoietic cell growth, suggesting that FAC plays a role in maintaining blood cell viability.66 The molecular mechanism of enhanced cellular survival by FAC expression remains unknown.

Two murine models of FA(C) were developed using targeted recombination in embryonic stem cells.63,101 In one knockout mouse, exon 8 of the FAC gene was deleted101 and replaced by a neomycin resistance gene; in the other mouse model, exon 9 was deleted.63 These mutations are likely to represent null alleles. The phenotype of homozygous FAC mutant animals was similar in the two strains. FAC mutants are viable and show no obvious birth defects of the skeletal system or urinary system. Cells derived from these animals show the classic hypersensitivity to bifunctional cross-linking agents. Similar chromosomal abnormalities and G2 cell-cycle abnormalities were also observed (Fig 2A). Nonetheless, pancytopenia did not develop during the first year of life. Similarly, no leukemia or increased cancer susceptibility was observed during this time span. Although no peripheral blood abnormalities were detectable, mice with deleted exon 9 had an age-dependent decrease in burst-forming units-erythroid (BFU-E) and colony-forming unit GM (CFU-GM) progenitor assays. In addition, hematopoietic progenitor cells showed a distinct hypersensitivity to interferon-γ (IFN-γ). Other mitotic inhibitors had no differential effect. Increased cell susceptibility to IFN-γ is mediated by fas-induced apoptosis.63 102 Cells derived from the FAC knockout mouse showed increased levels of fas expression at lower levels of IFN-γ. It is possible that IFN-γ hypersensitivity is the major pathogenetic mechanism in the development of progressive anemia in FA patients. The relationship between this phenotype and the cellular response to DNA crosslinks is currently unclear.

Fig. 3.

Mutations of the FAA polypeptide. The wild-type FAA protein (1455 amino acids), predicted from the cloned FAA cDNA,108 109 is shown schematically at the top. The protein contains a putative bipartite nuclear localization signal at the N-terminus and an incomplete leucine zipper. The indicated FAA alleles are shown at the left. The mutant FAA polypeptides, predicted by the mutant alleles, are shown schematically. A mutant protein with an in-frame deletion is predicted by the 1514(del)156 allele. Detection of these predicted mutant proteins in FA(A) cells awaits the generation of an FAA specific antiserum.

Fig. 3.

Mutations of the FAA polypeptide. The wild-type FAA protein (1455 amino acids), predicted from the cloned FAA cDNA,108 109 is shown schematically at the top. The protein contains a putative bipartite nuclear localization signal at the N-terminus and an incomplete leucine zipper. The indicated FAA alleles are shown at the left. The mutant FAA polypeptides, predicted by the mutant alleles, are shown schematically. A mutant protein with an in-frame deletion is predicted by the 1514(del)156 allele. Detection of these predicted mutant proteins in FA(A) cells awaits the generation of an FAA specific antiserum.

Close modal

The major phenotypic abnormality in both mutant mouse strains was a reduction in the number of male and female germ cells (Fig 2B). No stage-specific defect of spermatogenesis was observed, making a meiotic defect very unlikely. This is in contrast to the aberrations in meiosis that have been observed in AT and mismatch repair mice.103-105 The reduction in germ cells in FAC knockout mice was present at birth, indicating a developmental defect. With respect to this germ cell phenotype, FAC mice are similar to w and steel mice, which have defects in the c-kit receptor106 and its ligand, stem cell factor,107 respectively.

The unexpected germ-cell and IFN-γ hypersensitivity phenotypes observed in FAC knockout mice have added another level of complexity to the understanding of FA. Models for the function of the FA pathway must ultimately explain these observations, along with several other cellular abnormalities (Table 2). The availability of the two murine knockouts will aid in the development and refinement of these models and will provide an experimental system for studying FA therapies.

The cloning of FAA was reported only very recently108,109 and therefore much less is known about the FAA protein. The major FAA mRNA transcript is 5.5 kb long and found in low abundance in all adult tissues tested. In addition to this major mRNA species, multiple transcripts, both larger and smaller, can be seen on Northern blot, suggesting a complex pattern of mRNA splicing of the gene. The 5.5-kb mRNA contains an open reading frame of 4,368 nucleotides (nt), predicting a 1,455–amino acid protein with a relative molecular mass of 163 kD. Functional complementation has been achieved with this protein, but the 5′ end of the mRNA has not yet been definitively mapped. Because no STOP codons have been found in the cDNA, 5′ to the putative translation START site, an additional 5′ protein coding sequence may exist. Similar to FAC, the predicted FAA protein has no significant homologies to other proteins in sequence databases. Also, no common motifs are shared between the two FA proteins. The predicted FAA polypeptide has two overlapping bipartite nuclear localization signals, located at amino acids 18-34 and 19-35. In addition, a partial leucine zipper consensus sequence is found at position 1069-1090. These homologies are consistent with a function of FAA in the nucleus, perhaps as a DNA binding protein. The chromosomal location of FAA at 16q23.4, between markers D16S3121 and D16S303,75 was confirmed by direct methods. The gene spans approximately 80 kb and consists of at least 43 exons.109a A locus involved in loss of heterozygosity in breast cancer has been mapped to the same region110 111 but is unlikely to be identical to the FAA gene.

Several FAA gene mutations have already been described, but no predominant alleles are apparent. However, patients of South African descent were not examined in the two cloning reports.108,109 Because of the known linkage dysequilibrium in this population75 which has the highest known incidence of FA,112 it appears likely that an Afrikaner founder mutation will be identified. In addition, there exists a high prevalence of complementation group A among patients from Italy,113 suggesting a possible predominant allele in these patients. Interestingly, six of the first eight reported FAA mutations caused deletions of the FAA cDNA. The cDNA deletions may result from splice site mutations or genomic deletions. Two additional alleles were splice mutations, while no missense alleles were reported. The predicted effects of these changes on the FAA protein are depicted in Fig 3. Four of the eight mutations are predicted to result in severely truncated proteins. One interesting allele (1514del156) predicts an in-frame deletion of 52 amino acids, indicating the potential functional importance of these residues. The high frequency of independent deletions is unusual for an autosomal recessive disorder and suggests the possibility of repeat sequences which predispose to genomic rearrangements. If additional analysis confirms the high incidence of mutations leading to amino acid deletions, the protein truncation test114 may prove to be useful in the study of FAA patients.

At least three and possibly more FA genes remain to be cloned.69,72 The two FA genes which have been isolated to date were originally identified by functional complementation of MMC hypersensitivity,77,109 underscoring the power of this experimental approach. However, positional cloning techniques have also been successful.108 Functional complementation by cDNA libraries is a technically difficult endeavor, which tends to yield many false-positive cDNA clones. Prior knowledge of the chromosomal location of an FA gene can greatly aid in rapidly narrowing down the number of candidate cDNAs. Because FA is a genetically heterogeneous disorder, mapping by linkage analysis requires that a large number of families first be categorized into a complementation group by somatic cell hybrid experiments. This was accomplished for the FAA locus, because the vast majority of FA patients belong to this group.75 Other complementation groups are more rare, making linkage analysis difficult for other FA genes. This limitation can be overcome by the use of microcell-mediated chromosome transfer to map FA genes. The feasibility of this approach has been shown for FAD, a rare complementation group for which only three families have been identified.76 

As more FA genes are cloned, it becomes likely that novel members of the pathway will be identified as proteins that interact with already known FA proteins. Intense efforts to isolate additional FA genes are ongoing in several laboratories and are likely to be successful in the near future.

Based on the striking clinical similarities among FA patients from all complementation groups, the proteins encoded by the FA genes probably interact within the cell. The FA proteins may interact by a direct physical association or by a functional interaction. Several models are possible. First, the FA proteins may physically associate in a protein complex. Such a complex may serve as a defense mechanism, protecting the cell from DNA or protein cross-link damage. Second, the proteins may function sequentially in an enzymatic cascade, resulting in the repair or removal of cross-linked cellular macromolecules. Third, the FA proteins may interact in a signal transduction cascade. Such a pathway may include several components, including a “sensor” of crosslinking damage in the cytoplasm, a “transducer” for transmitting signals to the nucleus, and an “inducer” of new gene transcription, new DNA repair processes, or cell cycle arrest. Consistent with this latter model, the FAC protein may perform the sensor function in the cytoplasm and the FAA protein may translocate to the nucleus upon cellular exposure to cross-linking agents. Analysis of the FAC and FAA proteins with specific antisera, following cellular exposure to cross-linking agents, may help to distinguish among these possible models. Recent evidence suggests that FAA and FAC proteins bind and form a protein complex (G. Kupfer and A. D'Andrea, unpublished observation, January 1997). Interestingly, mutant FAC polypeptide derived from FA(C) patients fails to bind FAA.

The cloning of the FA genes has already had a considerable impact on the diagnosis and treatment of patients with FA. Although the DEB test is highly sensitive and specific for the diagnosis of FA,18 it fails to distinguish among FA patients in different complementation groups and fails to identify heterozygote carriers of mutant FA genes. The availability of the FAA and FAC genes will allow the rapid and accurate diagnosis of most FA patients and will form the basis of a prenatal diagnostic test for known families with FA. The identification of “common” mutant alleles of the FAA and FAC gene will also aid in rapid diagnosis. As discussed above, two specific mutant alleles of the FAC gene (IVS4+4 A > T and 322delG) account for 75% of all FAC mutations.80,82 115 Exon 14 mutations account for most of the remaining FA(C) patients. Specific FAA mutations may also be associated with specific ancestral backgrounds, although the relative prevalence of these FAA mutations among FA(A) patients remains to be tested.

The phenotypic variation of FA patients makes the diagnosis of specific FA genotypes critical. For instance, in one complementation group, FA(C), patients with the IVS4+4 A > T mutation have more severe disease, with severe congenital abnormalities and early onset of hematopoietic disease, while patients with the 322delG mutation have a relatively mild disease, with few congenital abnormalities and later onset of hematopoietic disease.82 Similar genotype/phenotype analysis is now possible for FAA patients. Such correlations are important because they guide FA patient management. For instance, if a more severe phenotype correlates with a particular FAA genotype, these FAA patients may be treated more aggressively, with the early use of BM transplantation or gene therapy protocols.

Molecular diagnosis is not entirely dependent on the knowledge of both mutant alleles. Once a family has been assigned to a complementation group by a functional assay or by the detection of a specific mutant allele, chromosome linkage analysis will be convenient and useful. Highly polymorphic microsatellite markers are located in close proximity to both FAA and FAC genes and can be used for the detection of carriers and for prenatal diagnosis.

The availability of the FA genes will profoundly influence the treatment of FA. Regardless of complementation group, most FA patients die from complications of their BM failure.1 Since most patients do not have a histocompatible donor, an alternative is retroviral transduction of an FA gene. For several reasons, retroviral transfer of an FA gene into the hematopoietic stem cells of an FA patient is a plausible option. For example, the FAA gene can effectively correct the cross-linking agent sensitivity and chromosomal instability of FA-A cells.109 Also, retroviral transduction of the FAC cDNA into FA(C) primary cells improves their survival and longevity, compared with uninfected cells.116 The survival advantage of corrected FA cells in vitro may translate into a survival advantage of the cells for in vivo studies. Gene transfer studies with the FA genes will be limited by the small number of hematopoietic stem cells that can be isolated from FA patients. For this reason, many centers have initiated programs to collect and cryospreserve hematopoietic stem cells from FA patients before the onset of anemia. Although useful, gene transfer studies, like BM transplant, will not improve the developmental abnormalities or cancer risk in nonhematopoietic tissues of FA patients.

Finally, the cloning of the FA genes will provide molecular insight into their role in other clinical features of FA, such as susceptibility to cancer. Some genotypic variants of FAA may correlate with a higher incidence of cancer. There exists no clear evidence for an increase in cancer incidence among heterozygote carriers of FA.117-120 Still, somatic mutation or loss of FA genes could theoretically contribute to oncogenesis in some heterozygote carriers or in some individuals who do not harbor germline mutations in FA genes. Accordingly, a systematic survey of the FA genes in primary tumors and tumor cell lines may reveal FA mutations.

The increased incidence of AML in FA suggests that FA genes may play a specific role in AML in the general (non-FA) patient population. Several reports of interstitial deletions of the long arm of chromosome 9 (9q-) associated with AML have been reported.121 122 In primary leukemic cells from some AML patients, these interstitial deletions overlap the FAC locus (G. Kupfer and A. D'Andrea, unpublished observation, December 1996). Taken together, these results suggest that the somatic loss of one or both FA genes may predispose to leukemic transformation in non-FA patients or heterozygous carriers of FA.

The phenomenon of somatic reversion of disease has recently been described in hereditary disorders of blood and liver.123 If a genetic disease causes cells to grow poorly, then spontaneously corrected cells, resulting from new mutations that revert mutant genes, may have a substantial growth advantage. Patients who improved clinically after somatic reversion and selection have previously been reported for both adenosine deaminase deficiency124 and Bloom's syndrome.45 46 Recently the same phenomenon has been reported in FA. Approximately 15% of FA patients have somatic mosaicism in their peripheral blood (Hans Joenje, personal communication, May 1997). In these patients, a mixture of MMC-sensitive and -resistant cells are found. In one patient, mitotic recombination was documented as the molecular mechanism of somatic reversion. Somatic mosaicism may make the diagnosis of FA difficult because of inconclusive chromosome breakage studies. If FA is strongly suspected in a patient despite normal breakage studies in peripheral blood, a definitive test can be performed on primary skin fibroblasts.

The high percentage of patients with somatic mosaicism indicates a strong selective advantage for cells that have lost the FA phenotype. This raises the possibility that gene therapy in FA may be aided by in vivo selection. At this time it is unclear whether any special physiologic circumstances are required for this selection to occur. Somatic reversion also suggests that some mutant FA genes have frame-shift mutations. These mutations may be “corrected” by new mutations that correct the open reading frame of the FA gene.

Somatic mosaicism may also cause some complications for FA patients. A high incidence of graft rejection has been noted in the BM transplantation of FA patients. Because of their increased sensitivity to bifunctional alkylating agents, FA patients typically receive a much less aggressive ablative treatment before BMT. If the recipient FA patient has somatic mosacism (particularly in T cells), some endogenous cells may be resistant to the ablative regimen and cause graft rejection. Further studies will be required to confirm this hypothesis.

The cloning of two FA genes (FAC and FAA) has provided an unprecedented opportunity to understand the molecular basis of FA. With these genes, new approaches can be taken to explore the cellular role of FAC and FAA. First, the mammalian genes may be used to identify FAC or FAA homologues in yeast or Drosophila. A genetic dissection of the FA gene pathway in one of these organisms may implicate the pathway in a basic cellular function such as cell growth, cell cycle, or genomic stabilization. Second, the FA genes may be used for the generation of additional mouse models of FA. As described above, cells derived from mice deficient in the FAC gene product exhibit genomic instability, similar to that observed in cell lines derived from FA patients. Similarly, knockouts of the FAA gene are now possible. Genetic interactions of FAC and FAA could be further analyzed by crossing different knockout strains. Third, antibodies may be generated against the FAC and FAA proteins. These antibodies can be used to identify a (possible) interaction between FAC and FAA and can be used to identify other binding partners of these proteins.

Also, as described, the availability of the FAA and FAC genes will directly impact the diagnosis and therapy of FA. Mutations in FA genes can be directly screened as an adjunct diagnostic procedure to the DEB test. Retroviruses that transduce the FA genes can be used for rapid diagnosis and complementation analysis of primary cells from suspected FA patients. Gene therapy studies with autologous peripheral blood CD34+ stem cells can be initiated for patients who do not have sibling-matched histocompatible donors.116 The observation of somatic reversion in FA provides some reason for optimism regarding the efficacy of gene therapy. Finally, once the molecular basis of the disease is established, more rational therapies or preventative measures may be devised for patients and families with FA.

The authors thank H. Joenje, A. Savoia, and M. Kelley for providing unpublished data.

Address reprint requests to Alan D. D'Andrea, MD, Dana-Farber Cancer Institute, Pediatric Oncology, 44 Binney St, Boston, MA 02115.

1
Alter BP, Young NS: The bone marrow failure syndromes, in Nathan DG, Oski FA (eds): Hematology of Infancy and Childhood, vol 1 (ed 4). Philadelphia, PA Saunders, 1993, p 216
2
Alter
BP
Fanconi's anaemia and its variability.
Br J Haematol
85
1993
9
3
Auerbach
AD
Rogatko
A
Schroeder-Kurth
TM
International Fanconi anemia registry: Relation of clinical symptoms to diepoxybutane sensitivity.
Blood
73
1989
391
4
Liu
J
Buchwald
M
Walsh
CE
Young
NS
Fanconi anemia and novel strategies for therapy.
Blood
84
1994
3995
5
Giampietro
PF
Adler-Brecher
B
Verlander
PC
Pavlakis
SG
Davis
JG
Auerbach
AD
The need for more accurate and timely diagnosis in Fanconi anemia. A report from the international Fanconi Anemia Registry.
Pediatrics
91
1993
1116
6
Giampietro
PF
Verlander
PC
Davis
JG
Auerbach
AD
Diagnosis of Fanconi anemia in patients without congenital malformations: An International Fanconi Anemia Registry study.
Am J Med Genet
68
1997
58
7
Young NS, Alter BP: Clinical features of Fanconi's anemia, in Young NS, Alter BP (eds): Aplastic Anemia, Acquired and Inherited. Philadelphia, PA, Saunders, 1994
8
dos Santos CC
Gavish
H
Buchwald
M
Fanconi anemia revisited: Old ideas and new advances.
Stem Cells
12
1994
142
9
Auerbach
AD
Fanconi anemia.
Dermatol Clin
13
1995
41
10
Alter
BP
Knobloch
ME
Weinberg
RS
Erythropoiesis in Fanconi's anemia.
Blood
78
1991
602
11
Butturini
A
Gale
RP
Verlander
PC
Adler-Brecher
B
Gillio
A
Auerbach
AD
Hematologic abnormalities in Fanconi anemia. An International Fanconi Anemia Registry study.
Blood
84
1994
1650
12
Swift
M
Zimmerman
D
McDonough
ER
Squamous cell carcinomas in Fanconi's anemia.
JAMA
216
1971
325
13
Somers
GR
Tabrizi
SN
Tiedemann
K
Chow
CW
Garland
SM
Venter
DJ
Squamous cell carcinoma of the tongue in a child with Fanconi anemia: A case report and review of the literature.
Pediatr Pathol Lab Med
15
1995
597
14
Nara
N
Miyamoto
T
et al
Two siblings with Fanconi's anemia developing squamous cell carcinomas.
Rinsho Ketsueki
21
1980
1944
15
Alter
BP
Fanconi's anemia and malignancies.
Am J Hematol
53
1996
99
16
Hojo
ET
van DP
Darroudi F
Natarajan
AT
Spontaneous chromosomal aberrations in Fanconi anaemia, ataxia telangiectasia fibroblast and Bloom's syndrome lymphoblastoid cell lines as detected by conventional cytogenetic analysis and fluorescence in situ hybridisation (FISH) technique [published erratum appears in Mutat Res 334:268, 1995].
Mutat Res
334
1995
59
17
Meyn
MS
High spontaneous intrachromosomal recombination rates in ataxia-telangiectasia.
Science
260
1993
1327
18
Auerbach
AD
Fanconi anemia diagnosis and the diepoxybutane (DEB) test.
Exp Hematol
21
1993
731
19
Dokal
I
Chase
A
Morgan
NV
Coulthard
S
Hall
G
Mathew
CG
Roberts
I
Positive diepoxybutane test in only one of two brothers found to be compound heterozygotes for Fanconi's anaemia complementation group C mutations.
Br J Haematol
93
1996
813
20
Shipley
J
Rodeck
CH
Garrett
C
Galbraith
J
Giannelli
F
Mitomycin C-induced chromsome damage in fetal blood cultures and prenatal diagnosis of Fanconi's anaemia.
Prenat Diagn
4
1984
217
21
Auerbach AD, Alter BP: Prenatal and postnatal diagnosis of aplastic anemia., in Alter BPe (ed): Methods in Hematology: Perinatal Hematology. Edinburgh, UK, Churchill Livingstone, 1989, p 225
22
Auerbach
AD
Weiner
MA
Warburton
K
Yeboa
LL
Broxmeyer
HE
Acute myeloid leukemia as the first hematologic manifestation of Fanconi anemia.
Am J Hematol
12
1982
289
23
Guinan
EC
Lopez
KD
Huhn
RD
Felser
JM
Nathan
DG
Evaluation of granulocyte-macrophage colony-stimulating factor for treatment of pancytopenia in children with Fanconi anemia.
J Pediatr
124
1994
144
24
Rackoff
WR
Orazi
A
Robinson
CA
Cooper
RJ
Alter
BP
Freedman
MH
Harris
RE
Williams
DA
Prolonged administration of granulocyte colony-stimulating factor (Filgrastim) to patients with Fanconi anemia: A pilot study.
Blood
88
1996
1588
25
Kohli-Kumar
M
Morris
C
Delaat
C
Sambrano
J
Masterson
M
Mueller
R
Shahidi
NT
Yanik
G
Desantes
K
Friedman
DJ
Auerbach
AD
Harris
RE
Bone marrow transplantation in Fanconi anemia using matched sibling donors.
Blood
84
1994
2050
26
Davies
SM
Khan
S
Wagner
JE
Arthur
DC
Auerbach
AD
Ramsay
NKC
Weisdorf
DJ
Unrelated donor bone marrow transplantation for Fanconi anemia.
Bone Marrow Transplant
17
1996
43
27
Broxmeyer
HE
Douglas
GW
Hangoc
G
Cooper
S
Bard
J
English
D
Arny
M
Thomas
L
Boyse
EA
Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells.
Proc Natl Acad Sci USA
86
1989
3828
28
Broxmeyer
HE
Kurtzberg
J
Gluckman
E
Auerbach
AD
Douglas
G
Coopers
S
Fakenburg
JH
Bard
J
Boyse
EA
Umbilical cord blood hematopoietic stem and repopulating cells in human clinical transplantation.
Blood
17
1991
313
29
Kurtzberg
J
Umbilical cord blood: A novel alternative source of hematopoietic stem cells for bone marrow transplantation [editorial].
J Hematother
5
1996
95
30
Kurtzberg
J
Laughlin
M
Graham
ML
Smith
C
Olson
JF
Halperin
E
Ciocci
G
Carrier
C
Stevens
CE
Rubinstein
P
Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients.
N Engl J Med
335
1996
157
31
Wagner
JE
Rosenthal
J
Sweetman
R
Shu
XO
Davies
SM
Ramsay
NKC
McGlave
PB
Sender
L
Cairo
MS
Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: Analysis of engraftment and acute graft-versus-host disease.
Blood
88
1996
795
32
Auerbach
AD
Allen
RG
Leukemia and preleukemia in Fanconi anemia patients.
Cancer Genet Cytogenet
51
1991
1
33
Maarek
O
Jonveaux
P
Le Coniat
M
Derre
J
Berger
R
Fanconi anemia and bone marrow clonal chromosome abnormalities.
Leukemia
10
1996
1700
34
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
64
1984
173
35
Venkatraj
VS
Gaidano
G
Auerbach
AD
Clonality studies and N-ras and p53 mutation analysis of hematopoietic cells in Fanconi anemia.
Leukemia
8
1994
1354
36
Friedberg
E
Cockayne syndrome — A primary defect in DNA repair, transcription, both, or neither.
Bioessays
18
1996
731
37
Friedberg
E
Relationships between DNA repair and transcription.
Annu Rev Biochem
65
1996
15
38
Eshleman
J
Markowitz
S
Mismatch repair defects in human carcinogenesis.
Hum Mol Genet
5
1996
1489
39
Legerski
R
Peterson
C
Expression cloning of a human DNA repair gene involved in xeroderma pigmentosum group C.
Nature
359
1992
70
40
van Vuuren
AJ
Appeldoorn
E
Odijk
H
Yasui
A
Jaspers
NGJ
Bootsma
D
Hoeijmakers
JHJ
Evidence for a repair enzyme complex involving ERCC1 and complementing activities of ERCC4, ERCC11 and xeroderma pigmentosum group F.
EMBO J
12
1993
3693
41
Savitsky
K
Bar-Shira
A
Gilad
S
Rotman
G
Ziv
Y
Vanagaite
L
Tagle
DA
Smith
S
Uziel
T
Sfez
S
Ashkenazi
M
Pecker
I
Frydman
M
Harnik
R
Patanjali
SR
Simmons
A
Clines
GA
Sartiel
A
Gatti
RA
Chessa
L
Sanal
O
Lavin
MF
Jaspers
NGJ
Taylor
AMR
Arlett
CF
Miki
T
Weissman
SM
Lovett
M
Collins
FS
Shiloh
Y
A single ataxia telangiectasia gene with a product similar to PI-3 kinase.
Science
268
1995
1749
42
Kastan
MB
et al
A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasia.
Cell
71
1992
587
43
Hawley
RS
Friend
SH
Strange bedfellows in even stranger places: The role of ATM in meiotic cells, lymphocytes, tumors, and its functional links to p53.
Genes Dev
10
1996
2383
44
Keegan
KS
Holtzman
DA
Plug
AW
Christenson
ER
Brainerd
EE
Flaggs
G
Bently
NJ
Taylor
EM
Meyn
MS
Moss
SB
Carr
AM
Ashley
T
Hoekstra
MF
The Atr and Atm protein kinases associate with different sites along meiotically pairing chromosomes.
Genes Dev
10
1996
2423
45
Ellis
NA
Groden
J
Ye
T-Z
Straughen
J
Lennon
DJ
Ciocci
S
Proytcheva
M
German
J
The Bloom's syndrome gene product is homologous to RecQ helicases.
Cell
83
1995
655
46
Ellis
NA
Lennon
DJ
Proytcheva
M
Alhadeff
B
Henderson
EE
German
J
Somatic intragenic recombination within the mutated locus BLM can correct the high sister-chromatid exchange phenotype of Bloom syndrome cells.
Am J Hum Genet
57
1995
1019
47
Schroeder
T
Anschutz
F
Knopp
A
Spontane chromosomenaberrationen bei familiarer panmyelopathie.
Humangenetik
1
1964
194
48
German
J
Puglatta
C
Chromosomal breakage in diploid cell lines from Bloom's syndrome and Fanconi's anemia.
Ann Genet
9
1966
143
49
Kaiser
TN
Lojewski
A
Dougherty
C
Juergens
L
Sahar
E
Latt
SA
Flow cytometric characterization of the response of Fanconi's anemia cells to mitomycin C treatment.
Cytometry
2
1982
291
50
Korkina
LG
Samochatova
EV
Maschan
AA
Suslova
TB
Cheremisina
ZP
Afanaslev
IB
Release of active oxygen radicals by leukocytes of Fanconi Anemia patients.
J Leukoc Biol
52
1992
357
51
Kubbies
M
Schindler
D
Hoehn
H
Schinzel
A
Rabinovich
PS
Endogenous blockage and delay of the chromosome cycle despite normal recruitment and growth phase explain poor proliferation and frequent edomitosis in Fanconi anemia cells.
Am J Hum Genet
37
1985
1022
52
Schlindler
D
Hoehn
H
Fanconi anemia mutation causes cellular susceptibility to ambient oxygen.
Am J Hum Genet
43
1988
429
53
Gille
JJP
Wortelboer
HM
Joenje
H
Antioxidant status of Fanconi anemia fibroblasts.
Hum Genet
77
1987
28
54
Bigelow
SB
Rary
JM
Bender
MA
G2 chromosomal radiosenitivity in Fanconi's anamia.
Mutat Res
63
1979
189
55
Rosselli
F
Sanceau
J
Wietzerbin
J
Moustacchi
E
Abnormal lymphokine production: A novel feature of the genetic disease Fanconi anemia.
Hum Genet
89
1992
42
56
Takeuchi
T
Morimoto
K
Increased formation of 8-hydroxydeoxyguanosine in DNA damage, in lymphobasts from Fanconi's anemia patients due to possible catalase deficiency.
Carcinogen
14
1993
1115
57
Fujiwara
Y
Tatsumi
M
Sasaki
MS
Cross-link repair in human cells and its possible defect in Fanconi's anaemia cells.
Biochim Biophys Acta
113
1977
635
58
Papadopoulo
D
Guillouf
C
Mohnrenweiser
H
Moustacchi
E
Hypomutability in Fanconi anemia cells is associated with increased deletion frequency at the HPRT locus.
Proc Natl Acad Sci USA
87
1990
8383
59
Willingale-Theune
J
Schweiger
M
Hirsch-Kauffmann
M
Meek
AE
Paulin-Levasseur
M
Traub
P
Ultrastructure of Fanconi anemia fibroblasts.
J Cell Sci
93
1989
651
60
Kupfer
GM
D'Andrea
AD
The effect of the Fanconi anemia polypeptide, FAC, upon p53 induction and G2 checkpoint regulation.
Blood
88
1996
1019
61
Rosselli
F
Ridet
A
Soussi
T
Duchaud
E
Alapetite
C
Moustacchi
E
p53-dependent pathway of radio-induced apoptosis is altered in Fanconi anemia.
Oncogene
10
1995
9
62
Daneshbod-Skibba
G
Martin
J
Shahidi
NT
Myeloid and erythroid colony growth in non-anaemic patients with Fanconi's anaemia.
Br J Haematol
44
1980
33
63
Whitney
MA
Royle
G
Low
MJ
Kelly
MA
Axthelm
MK
Reifsteck
C
Olson
S
Braun
RE
Heinrich
MC
Rathbun
RK
Bagby
GC
Grompe
M
Germ cell defects and hematopoietic hypersensitivity to gamma-interferon in mice with a targeted disruption of the Fanconi anemia C gene.
Blood
88
1996
49
64
Wang J, Youssoufian H, Lo Ten Foe JR, Devetten M, Cumming RC, Buchwald M, Liu JM: Overexpression of the Fanconi anemia group C gene (FAC) protects hematopoietic progenitors from FAS-mediated apoptosis. Blood 88:548a, 1996 (abstr, suppl 1)
65
Cumming
RC
Liu
JM
Youssoufian
H
Buchwald
M
Suppression of apoptosis in hematopoietic factor-dependent progenitor cell lines by expression of the FAC gene.
Blood
88
1996
4558
66
Segal
G
Magenis
R
Brown
M
Keeble
W
Smith
T
Heinrich
M
Bagby
G
Repression of Fanconi anemia gene (FACC) expression inhibits growth of hematopoietic progenitor cells.
J Clin Invest
94
1994
846
67
Joenje
H
Frants
RR
Arwert
F
de Bruin
GJ
Kostense
PJ
van de Kamp
JJ
de Koning
J
Eriksson
AW
Erythrocyte superoxide dismutase deficiency in Fanconi's anaemia established by two independent methods of assay.
Scand J Clin Lab Invest
39
1979
759
68
Mavelli
I
Ciriolo
MR
Rotilio
G
De Sole
P
Castorino
M
Stabile
A
Superoxide dismutase, glutathione peroxidase and catalase in oxidative hemolysis. A study of Fanconi's anemia erythrocytes.
Biochem Biophys Res Commun
106
1982
1982
69
Buchwald
M
Complementation groups: One or more per gene?
Nat Genet
11
1995
228
70
Duckworth-Rysiecki
G
Cornish
K
Clarke
CA
Buchwald
M
Identification of two complementation groups in Fanconi anemia.
Somat Cell Mol Genet
11
1985
35
71
Joenje
H
Fanconi anemia complementation groups in Germany and The Netherlands.
Hum Genet
97
1996
280
72
Joenje
H
LoTen
F
Oostra
A
Berkel
CV
Rooimans
M
Schroeder
S
Kurth
T
Wegner
R
Gille
J
Buchwald
M
Arwert
F
Classification of Fanconi anemia patients by complementation analysis: Evidence for a fifth genetic subtype.
Blood
86
1995
2156
73
Strathdee
CA
Duncan
AMV
Buchwald
M
Evidence for at least four Fanconi Anemia genes including FACC on chromosome 9.
Nat Genet
1
1992
196
74
Strathdee CA, Duncan AM, Buchwald M: Nat Genet 1:196, 1992
75
Pronk
JC
Gibson
RA
Savoia
A
Wijker
M
Morgan
NS
Melchionda
S
Ford
D
Temtamy
S
Ortega
JJ
Jansen
S
Harenga
C
Cohn
RJ
de Ravel
TJ
Roberts
I
Westerveld
A
Easton
DJ
Joenje
H
Mathew
CG
Arwert
F
Localization of the Fanconi anemia complementation group A gene to chromosome 16q24.3.
Nat Genet
11
1995
338
76
Whitney
M
Thayer
M
Reifsteck
C
Olson
S
Smith
L
Jakobs
PM
Leach
R
Naylor
S
Joenje
H
Grompe
M
Microcell mediated chromosome transfer maps the Fanconi anemia group D gene to chromosome 3p.
Nat Genet
11
1995
341
77
Strathdee
CA
Gavish
H
Shannon
WR
Buchwald
M
Cloning of cDNAs for Fanconi's anaemia by functional complementation.
Nature
356
1992
763
78
Gibson
RA
Buchwald
M
Roberts
RG
Mathew
CG
Characterisation of the exon structure of the Fanconi anaemia group C gene by vectorette PCR.
Hum Mol Genet
2
1993
35
79
Wevrick
R
Clarke
CA
Buchwald
M
Cloning and analysis of the murine Fanconi anemia group C cDNA.
Hum Mol Genet
2
1993
655
80
Whitney
MA
Saito
H
Jakobs
PM
Gibson
RA
Moses
RE
Grompe
M
A common mutation in the FACC gene causes Fanconi anemia in Ashkenazi Jews.
Nat Genet
4
1993
202
81
Yamashita
T
Wu
N
Kupfer
G
Corless
C
Joenje
H
Grompe
M
D'Andrea
AD
The clinical variability of Fanconi anemia (type C) results from expression of an amino terminal truncated FAC polypeptide with partial activity.
Blood
87
1996
4424
82
Verlander
PC
Lin
JD
Udono
MU
Zhang
Q
Gibson
RA
Mathew
CG
Auerbach
AD
Mutation analysis of the Fanconi anemia gene FACC.
Am J Hum Genet
54
1994
595
83
Gibson
RA
Hajkanpour
A
Murer-Orlando
M
Buchwald
M
Mathew
CG
A nonsense mutation and exon skipping in the Fanconi Anemia group C gene.
Hum Mol Genet
2
1993
797
84
Gibson
RA
Morgan
NV
Goldstein
LH
Pearson
IC
Kesterton
IP
Foot
MJ
Jansen
S
Havenga
C
Pearson
T
de Ravel
TJ
Cohn
RJ
Marques
IM
Dokal
I
Roberts
I
Marsh
J
Ball
S
Milner
RD
Llerena
JC
Samochatova
E
Mohan
SP
Vasudevan
P
Birjandi
F
Hajianpour
A
Murer-Orlando
M
Mathew
CG
Novel mutations and polymorphisms in the Fanconi anaemia group C gene.
Hum Mutat
8
1996
140
85
Gavish
H
dos Santos CC
Buchwald
M
A Leu554-to-Pro substitution completely abolishes the functional complementing activity of the Fanconi anemia (FACC) protein.
Hum Mol Genet
2
1993
123
86
Lo
Ten Foe J
Rooimans
M
Joenje
H
Arwert
F
A novel frameshift mutation (1806insA) in exon 14 of the Fanconi anemia C gene, FAC.
Hum Mutat
7
1996
264
87
Krasnoshtein
F
Buchwald
M
Developmental expression of the Fac gene correlates with congenital defects in Fanconi anemia patients.
Hum Mol Genet
5
1996
85
88
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
4
1995
1321
89
Kupfer
G
Yamashita
T
Asano
Suliman A
D'Andrea
AD
The Fanconi anemia protein, FAC, binds to the cyclin-dependent kinase, cdc2.
Blood
90
1997
1047
90
Youssoufian
H
Localization of Fanconi anemia C protein to the cytoplasm of mammalian cells.
Proc Natl Acad Sci USA
91
1994
7975
91
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
91
1994
6712
92
Youssoufian
H
Auerbach
AD
Verlander
PC
Steimle
V
Mach
B
Identification of cytosolic proteins that bind to the Fanconi Anemia complementation group C polypeptide in vitro.
J Biol Chem
270
1995
9876
93
Hoshino T, Youssoufian H, Wang J, Devetten MP, Iwata N, Kajigaya S, Liu JM: The molecular chaperone GRP94 binds to a central domain within the group C Fanconi anemia protein. Blood 88:1734a, 1996 (abstr, suppl 1)
94
Youssoufian
H
Li
Y
Martin
ME
Buchwald
M
Induction of Fanconi anemia cellular phenotype in human 293 cells by overexpression of a mutant FAC allele.
J Clin Invest
97
1996
957
95
Kruyt
FAE
Dijkmans
LM
van den Berg
TK
Joenje
H
Fanconi Anemia genes act to suppress a cross-linker-inducible p53-independent apoptosis pathway in lymphoblastoid cell lines.
Blood
87
1996
938
96
Germann
UA
Pastan
I
Gottesman
MM
P-glycoproteins: Mediators of multidrug resistance.
Semin Cell Biol
4
1993
63
97
Hodnick WF, Sartorelli AC: Reductive activation of mitomycin C by NADH:cytochrome b5 reductase.1 Cancer Res 53:4907, 1993
98
Hu
L-T
Stamberg
J
Pan
S-S
The NAD(P)H:quinone oxidoreductase locus is human colon carcinoma HCT 116 cells resistant to mitomycin C.
Cancer Res
56
1996
5253
99
Allen C, Meyn MS: Isolation and charactrization of 3 cDNAs that complement the mutagen-sensitivity of Fanconi anemia group A fibroblasts. Am J Hum Genet 55:58, 1994 (abstr).
100
Vogelstein
B
Kinzler
KW
p53 function and dysfunction.
Cell
70
1992
523
101
Chen
M
Tomkins
DJ
Auerbach
W
McKerlie
C
Youssoufian
H
Liu
L
Gan
O
Carreau
M
Auerbach
A
Groves
T
Guidos
CJ
Freeman
MH
Cross
J
Percy
DH
Dick
JE
Joyner
AL
Buchwald
M
Inactivation of Fac in mice produces inducible chromosomal instability and reduced fertility reminiscent of Fanconi anaemia.
Nat Genet
12
1996
448
102
Heneveld M, Faulkner G, Royle G, Heinrich M, Keeble W, Kahn R, Grompe M, Rathbun K, Bagby GC: The pathogenesis of bone marrow failure in Fanconi anemia: The role of IFNγ-induced FAS expression. Blood 88:339a, 1996 (abstr, suppl 1)
103
Baker
SM
Bronner
CE
Zhang
L
Plug
AW
Robatzek
M
Warren
G
Elliott
EA
Yu
J
Ashley
T
Arnheim
N
Flavell
RA
Liskay
RM
Male mice defective in the DNA mismatch repair gene PMS2 exhibit abnormal chromosome synapsis in meiosis.
Cell
82
1995
309
104
Baker
SM
Plug
AW
Prolla
TA
Bronner
CE
Harris
AC
Yao
X
Christie
DM
Monell
C
Arnheim
N
Bradley
A
Ashley
T
Liskay
RM
Involvement of mouse MIh1 in DNA mismatch repair and meiotic crossing over.
Nat Genet
13
1996
336
105
Barlow
C
Hirotsune
S
Paylor
R
Liyanage
M
Eckhaus
M
Collins
F
Shiloh
Y
Crawley
JN
Ried
T
Tagle
D
Wynshaw-Boris
A
Atm-deficient mice: A paradigm of ataxia telangiectasia.
Cell
86
1996
159
106
Matthews
W
Jordan
CT
Wiegand
GW
Pardoll
D
Lemischka
IR
A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations.
Cell
65
1991
1143
107
Zsebo
KM
Williams
DA
Geissler
EN
Broudy
VC
Martin
FH
Atkins
HL
Hsu
RY
Birkett
NC
Okino
KH
Murdock
DC
Jacobsen
FW
Langely
KE
Smith
KA
Takeishi
T
Cattanach
BM
Galli
SJ
Suggs
SV
Stem cell factor is encoded at the S1 locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor.
Cell
63
1990
213
108
Apostolou
S
Whitmore
SA
Crawford
J
Alon
N
Wijker
M
Parker
L
Lightfoot
J
Carreau
M
Callen
DF
Savoia
A
Cheng
NC
van Berkel
CGM
Strunk
MHP
Gille
JJP
Pals
G
Kruyt
FAE
Pronk
JC
Arwert
F
Buchwald
M
Joenje
H
Positional cloning of the Fanconi Anaemia Group A gene.
Nat Genet
14
1996
324
109
Lo
Ten Foe JR
Rooimans
MA
Bosnoyan-Collins
L
Alan
N
Wijker
M
Parker
L
Lightfoot
J
Carreau
M
Callen
DF
Savoia
A
Cheng
NC
van Berkel
CG
Strunk
MH
Gille
JJ
Pals
G
Kruyt
FA
Pronk
JC
Arwert
F
Buchwald
M
Joenje
H
Expression cloning of a cDNA for the major Fanconi anemia gene, FAA.
Nature Genet
14
1996
320
109a
Ianzano L, D'Apolito M, Centra M, Savino M, Levran O, Auerbach AD, Cleton-Jansen A-M, Doggett NA, Pronk JC, Tipping AJ, Gibso RA, Mathew CG, Whitmore SA, Apostolou S, Callen DF, Zelante L, Savoia A: The genomic organization of the Fanconi Anemia Group A (FAA) gene. Genomics 41:309, 1997
110
Dorion-Bonnet
F
Mautalen
S
Hostein
I
Longy
M
Allelic imbalance study of 16q in human primary breast carcinomas using microsatellite markers.
Genes Chromosom Cancer
14
1995
171
111
Tsuda
H
Callen
DF
Fukutomi
T
Nakamura
Y
Hiroshashi
S
Allele loss on chromosome 16q24.2-qter occurs frequently in breast cancers irrespectively of differences in phenotype and extent of spread.
Cancer Res
54
1994
513
112
Rosendorff
J
Bernstein
R
Macdougall
L
Jenkins
T
Fanconi anemia; another disease of unusually high prevalence in the Afrikaans population of South Africa.
Am J Med Genet
2
1987
793
113
Savoia
A
Zatterale
A
Del Principe
D
Joenje
H
Fanconi anaemia in Italy: High prevalence of complementation group A in two geographic clusters.
Hum Genet
97
1996
599
114
Hogervorst
FB
Cornelis
RS
Bout
M
van-Vliet
M
Oosterwijk
JC
Olmer
R
Bakker
B
Klijn
JG
Vasen
HF
Meijers-Heijboer
H
Menko
FH
Cornelisse
CJ
den Dunnen
PD
van Ommen
G-JB
Rapid detection of BRCA1 mutations by the protein truncation test.
Nat Genet
10
1995
208
115
Lo
Ten Foe J
Barel
M
Tuss
P
Digweed
M
Arwert
F
Joenje
H
Sequence variations in the Fanconi anaemia gene, FAC: Pathogenicity of 1806insA and R548X and recognition of D195V as a polymorphic variant.
Hum Genet
98
1996
522
116
Walsh
CE
Grompe
M
Vanin
E
Buchwald
M
Young
NS
Nienhuis
AW
Liu
JM
A functionally active retrovirus vector for gene therapy in Fanconi anemia group C.
Blood
84
1994
453
117
Swift
M
Fanconi's anaemia in the genetics of neoplasia.
Nature
230
1971
370
118
Swift
MR
Hirschorn
K
Fanconi's anemia: Inherited susceptibility to chromosome breakage in various tissues.
Ann Intern Med
65
1966
486
119
Welshimer
K
Swift
M
Congenital malformations and developmental disabilities in ataxia-telangiectasia, Fanconi anemia, and xeroderma pigmentosum families.
Am J Hum Genet
34
1982
781
120
Potter
NU
Sarmousakis
C
Li
FP
Cancer in relatives of patients with aplastic anemia.
Cancer Genet Cytogenet
9
1983
61
121
Kwong
YL
Ha
SY
Liang
RHS
Wan
TSK
Chan
LC
Interstitial Deletion of 9q in a case of acute myeloid leukemia (FAB M6).
Cancer Genet Cytogenet
66
1993
79
122
Mecucci
C
Vermaelen
K
Kulling
G
Michaux
JL
Noens
L
Hove
WV
Tricot
G
Louwagie
A
Berghe
HVD
Interstitial 9q-deletions in hematologic malignancies.
Cancer Genet Cytogenet
12
1984
309
123
Kvittingen
EA
Rootwelt
H
Brandtzaeg
P
Bergan
A
Berger
R
Hereditary tyrosinemia type I. Self-induced correction of the fumarylacetoacetase defect.
J Clin Invest
91
1993
1816
124
Hirschhorn
R
Yang
DR
Puck
JM
Huie
ML
Jiang
CK
Kurlandsky
LE
Spontaneous in vivo reversion to normal of an inherited mutation in a patient with adenosine deaminase deficiency.
Nat Genet
13
1996
290
Sign in via your Institution