Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Anemia ; 2012: 865170, 2012.
Article in English | MEDLINE | ID: mdl-22701786

ABSTRACT

Fanconi anemia (FA) is a recessive DNA instability disorder associated with developmental abnormalities, bone marrow failure, and a predisposition to cancer. Based on their sensitivity to DNA cross-linking agents, FA cells have been assigned to 15 complementation groups, and the associated genes have been identified. Founder mutations have been found in different FA genes in several populations. The majority of Dutch FA patients belongs to complementation group FA-C. Here, we report 15 patients of Dutch ancestry and a large Canadian Manitoba Mennonite kindred carrying the FANCC c.67delG mutation. Genealogical investigation into the ancestors of the Dutch patients shows that these ancestors lived in four distinct areas in The Netherlands. We also show that the Dutch and Manitoba Mennonite FANCC c.67delG patients share the same haplotype surrounding this mutation, indicating a common founder.

2.
Hum Mol Genet ; 21(1): 121-35, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21968513

ABSTRACT

Fanconi anemia (FA) is a human rare genetic disorder characterized by congenital defects, bone marrow (BM) failure and predisposition to leukemia. The progressive aplastic anemia suggests a defect in the ability of hematopoietic stem cells (HSC) to sustain hematopoieis. We have examined the role of the nuclear FA core complex gene Fancg in the functionality of HSC. In Fancg-/- mice, we observed a decay of long-term HSC and multipotent progenitors that account for the reduction in the LSK compartment containing primitive hematopoietic cells. Fancg-/- lymphoid and myeloid progenitor cells were also affected, and myeloid progenitors show compromised in vitro functionality. HSC from Fancg-/- mice failed to engraft and to reconstitute at short and long term the hematopoiesis in a competitive transplantation assay. Fancg-/- LSK cells showed a loss of quiescence, an impaired migration in vitro in response to the chemokine CXCL12 and a defective homing to the BM after transplantation. Finally, the expression of several key genes involved in self-renewal, quiescence and migration of HSC was dysregulated in Fancg-deficient LSK subset. Collectively, our data reveal that Fancg should play a role in the regulation of physiological functions of HSC.


Subject(s)
Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia/physiopathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow/metabolism , Cell Movement , Chemokine CXCL12/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group G Protein/genetics , Female , Hematopoiesis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
3.
J Pathol ; 226(1): 28-39, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21915857

ABSTRACT

Fanconi anaemia (FA) is a rare recessive disorder marked by developmental abnormalities, bone marrow failure, and a high risk for the development of leukaemia and solid tumours. The inactivation of FA genes, in particular FANCF, has also been documented in sporadic tumours in non-FA patients. To study whether there is a causal relationship between FA pathway defects and tumour development, we have generated a mouse model with a targeted disruption of the FA core complex gene Fancf. Fancf-deficient mouse embryonic fibroblasts displayed a phenotype typical for FA cells: they showed an aberrant response to DNA cross-linking agents as manifested by G(2) arrest, chromosomal aberrations, reduced survival, and an inability to monoubiquitinate FANCD2. Fancf homozygous mice were viable, born following a normal Mendelian distribution, and showed no growth retardation or developmental abnormalities. The gonads of Fancf mutant mice functioned abnormally, showing compromised follicle development and spermatogenesis as has been observed in other FA mouse models and in FA patients. In a cohort of Fancf-deficient mice, we observed decreased overall survival and increased tumour incidence. Notably, in seven female mice, six ovarian tumours developed: five granulosa cell tumours and one luteoma. One mouse had developed tumours in both ovaries. High-resolution array comparative genomic hybridization (aCGH) on these tumours suggests that the increased incidence of ovarian tumours correlates with the infertility in Fancf-deficient mice and the genomic instability characteristic of FA pathway deficiency.


Subject(s)
Fanconi Anemia Complementation Group F Protein/genetics , Granulosa Cell Tumor/genetics , Luteoma/genetics , Ovarian Neoplasms/genetics , Animals , Comparative Genomic Hybridization , Disease Models, Animal , Fanconi Anemia Complementation Group F Protein/deficiency , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
DNA Repair (Amst) ; 10(12): 1252-61, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-22036606

ABSTRACT

Fanconi anemia (FA) is a heritable disease characterized by bone marrow failure, congenital abnormalities, and cancer predisposition. The 15 identified FA genes operate in a molecular pathway to preserve genomic integrity. Within this pathway the FA core complex operates as an ubiquitin ligase that activates the complex of FANCD2 and FANCI to coordinate DNA repair. The FA core complex is formed by at least 12 proteins. However, only the FANCL subunit displays ubiquitin ligase activity. FANCA and FANCG are members of the FA core complex for which no other functions have been described than to participate in protein interactions. In this study we generated mice with combined null alleles for Fanca and Fancg to identify extended functions for these genes by characterizing the double mutant mice and cells. Double mutant a(-/-)/g(-/-) mice were born at near Mendelian frequencies without apparent developmental abnormalities. Histological analysis of a(-/-)/g(-/-) mice revealed a Leydig cell hyperplasia and frequent vacuolization of Sertoli cells in testes, while ovaries were depleted from developing follicles and displayed an interstitial cell hyperplasia. These gonadal aberrations were associated with a compromised fertility of a(-/-)/g(-/-) males and females. During the first year of life a(-/-)/g(-/-) did not develop malignancies or bone marrow failure. At the cellular level a(-/-)/g(-/-), Fanca(-/-), and Fancg(-/-) cells proved equally compromised in DNA crosslink and homology-directed repair. Overall the phenotype of a(-/-)/g(-/-) double knockout mice and cells appeared highly similar to the phenotype of Fanca or Fancg single knockouts. The lack of an augmented phenotype suggest that null mutations in Fanca or Fancg are fully epistatic, making additional important functions outside of the FA core complex highly unlikely.


Subject(s)
Epistasis, Genetic/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/genetics , Mutation/genetics , Active Transport, Cell Nucleus/drug effects , Animals , Bone Marrow Cells/cytology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromosome Breakage/drug effects , Embryo, Mammalian , Female , Fertility/genetics , Fibroblasts/cytology , Fluorobenzenes/pharmacology , Hematologic Tests , Humans , Male , Mice , Ovary/metabolism , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Stem Cells/drug effects , Stem Cells/metabolism , Testis/metabolism
5.
EMBO J ; 27(5): 770-81, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18239686

ABSTRACT

Although brain development abnormalities and brain cancer predisposition have been reported in some Fanconi patients, the possible role of Fanconi DNA repair pathway during neurogenesis is unclear. We thus addressed the role of fanca and fancg, which are involved in the activation of Fanconi pathway, in neural stem and progenitor cells during brain development and adult neurogenesis. Fanca(-/-) and fancg(-/-) mice presented with microcephalies and a decreased neuronal production in developing cortex and adult brain. Apoptosis of embryonic neural progenitors, but not that of postmitotic neurons, was increased in the neocortex of fanca(-/-) and fancg(-/-) mice and was correlated with chromosomal instability. In adult Fanconi mice, we showed a reduced proliferation of neural progenitor cells related to apoptosis and accentuated neural stem cells exhaustion with ageing. In addition, embryonic and adult Fanconi neural stem cells showed a reduced capacity to self-renew in vitro. Our study demonstrates a critical role for Fanconi pathway in neural stem and progenitor cells during developmental and adult neurogenesis.


Subject(s)
Brain/cytology , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group G Protein/deficiency , Neurons/cytology , Stem Cells/cytology , Animals , Apoptosis , Brain/embryology , Cell Proliferation , Chromosome Aberrations , DNA Repair , Embryonic Development , Fanconi Anemia , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Female , Mice , Mice, Knockout , Pregnancy
6.
Cell Oncol ; 29(3): 211-8, 2007.
Article in English | MEDLINE | ID: mdl-17452773

ABSTRACT

To identify the gene underlying Fanconi anemia (FA) complementation group I we studied informative FA-I families by a genome-wide linkage analysis, which resulted in 4 candidate regions together encompassing 351 genes. Candidates were selected via bioinformatics and data mining on the basis of their resemblance to other FA genes/proteins acting in the FA pathway, such as: degree of evolutionary conservation, presence of nuclear localization signals and pattern of tissue-dependent expression. We found a candidate, KIAA1794 on chromosome 15q25-26, to be mutated in 8 affected individuals previously assigned to complementation group I. Western blots of endogenous FANCI indicated that functionally active KIAA1794 protein is lacking in FA-I individuals. Knock-down of KIAA1794 expression by siRNA in HeLa cells caused excessive chromosomal breakage induced by mitomycin C, a hallmark of FA cells. Furthermore, phenotypic reversion of a patient-derived cell line was associated with a secondary genetic alteration at the KIAA1794 locus. These data add up to two conclusions. First, KIAA1794 is a FA gene. Second, this gene is identical to FANCI, since the patient cell lines found mutated in this study included the reference cell line for group I, EUFA592.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Adolescent , Adult , Base Sequence , Cell Line , Child , Chromosomal Instability/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Genome, Human/genetics , HeLa Cells , Humans , Male , Molecular Sequence Data , Mutation/genetics , Pedigree , Phenotype , Ubiquitin/metabolism
7.
Blood ; 108(13): 4283-7, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16946306

ABSTRACT

Fanconi anemia (FA) is a heterogeneous genetic disorder characterized by bone marrow (BM) failure and cancer susceptibility. Identification of the cDNAs of FA complementation types allows the potential of using gene transfer technology to introduce functional cDNAs as transgenes into autologous stem cells and provide a cure for the BM failure in FA patients. However, strategies to enhance the mobilization, transduction, and engraftment of exogenous stem cells are required to optimize efficacy prior to widespread clinical use. Hypersensitivity of Fancc-/- cells to interferon-gamma (IFN-gamma), a nongenotoxic immune-regulatory cytokine, enhances engraftment of syngeneic wild-type (WT) cells in Fancc-/- mice. However, whether this phenotype is of broad relevance in other FA complementation groups is unresolved. Here we show that primitive and mature myeloid progenitors in Fanca-/- and Fancg-/- mice are hypersensitive to IFN-gamma and that in vivo infusion of IFN-gamma at clinically relevant concentrations was sufficient to allow consistent long-term engraftment of isogenic WT repopulating stem cells. Given that FANCA, FANCC, and FANCG complementation groups account for more than 90% of all FA patients, these data provide evidence that IFN-gamma conditioning may be a useful nongenotoxic strategy for myelopreparation in FA patients.


Subject(s)
Antiviral Agents/pharmacology , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/therapy , Genetic Therapy , Interferon-gamma/pharmacology , Myeloid Progenitor Cells/transplantation , Animals , Fanconi Anemia/genetics , Genetic Therapy/methods , Graft Enhancement, Immunologic/methods , Graft Survival/drug effects , Graft Survival/genetics , Hematopoietic Stem Cell Mobilization/methods , Humans , Mice , Mice, Knockout , Transduction, Genetic/methods , Transplantation, Autologous , Transplantation, Isogeneic
8.
Mutat Res ; 601(1-2): 191-201, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16920162

ABSTRACT

Fanconi anemia (FA) is an inherited cancer-susceptibility disorder, characterized by genomic instability and hypersensitivity to DNA cross-linking agents. The discovery of biallelic BRCA2 mutations in the FA-D1 complementation group allows for the first time to study the characteristics of primary BRCA2-deficient human cells. FANCD1/BRCA2-deficient fibroblasts appeared hypersensitive to mitomycin C (MMC), slightly sensitive to methyl methane sulfonate (MMS), and like cells derived from other FA complementation groups, not sensitive to X-ray irradiation. However, unlike other FA cells, FA-D1 cells were slightly sensitive to UV irradiation. Despite the observed lack of X-ray sensitivity in cell survival, significant radioresistant DNA synthesis (RDS) was observed in the BRCA2-deficient fibroblasts but also in the FANCA-deficient fibroblasts, suggesting an impaired S-phase checkpoint. FA-D1/BRCA2 cells displayed greatly enhanced levels of spontaneous as well as MMC-induced chromosomal aberrations (CA), similar to cells deficient in homologous recombination (HR) and non-D1 FA cells. In contrast to Brca2-deficient rodent cells, FA-D1/BRCA2 cells showed normal sister chromatid exchange (SCE) levels, both spontaneous as well as after MMC treatment. Hence, these data indicate that human cells with biallelic BRCA2 mutations display typical features of both FA- and HR-deficient cells, which suggests that FANCD1/BRCA2 is part of the integrated FA/BRCA DNA damage response pathway but also controls other functions outside the FA pathway.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Fibroblasts/metabolism , Bleomycin/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , DNA Damage/genetics , DNA Repair/genetics , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Methyl Methanesulfonate/pharmacology , Mitomycin/pharmacology , Sister Chromatid Exchange/drug effects , Sister Chromatid Exchange/radiation effects
9.
Mutat Res ; 594(1-2): 39-48, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16154163

ABSTRACT

Fanconi anemia (FA) is a cancer susceptibility disorder characterized by chromosomal instability and hypersensitivity to DNA cross-linking agents. So far 11 complementation groups have been identified, from which only FA-D1/BRCA2 and FA-J are defective downstream of the central FANCD2 protein as cells from these groups are capable of monoubiquitinating FANCD2. In this study we show that cells derived from patients from the new complementation groups, FA-I, FA-J and FA-L are all proficient in DNA damage induced Rad51 foci formation, making the cells from FA-D1/BRCA2 patients that are defective in this process the sole exception. Although FA-B patient HSC230 was previously reported to also have biallelic BRCA2 mutations, we found normal Rad51 foci formation in cells from this patient, consistent with the recent identification of an X-linked gene being mutated in four unrelated FA-B patients. Thus, our data show that none of the FA proteins, except BRCA2, are required to sequester Rad51 into nuclear foci. Since cells from the FA-D1 and FA-J patient groups are both able to monoubiquitinate FANCD2, the "Rad51 foci phenotype" provides a convenient assay to distinguish between these two groups. Our results suggest that FANCJ and FANCD1/BRCA2 are part of the integrated FANC/BRCA DNA damage response pathway or, alternatively, that they represent sub-pathways in which only FANCD1/BRCA2 is directly connected to the process of homologous recombination.


Subject(s)
BRCA2 Protein/genetics , DNA Damage/genetics , Fanconi Anemia Complementation Group L Protein/genetics , Fanconi Anemia/genetics , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , BRCA2 Protein/metabolism , Cell Line, Transformed , Cells, Cultured , Fanconi Anemia/metabolism , Fibroblasts/metabolism , Humans , Recombination, Genetic
10.
Nat Genet ; 37(9): 934-5, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116423

ABSTRACT

The protein predicted to be defective in individuals with Fanconi anemia complementation group J (FA-J), FANCJ, is a missing component in the Fanconi anemia pathway of genome maintenance. Here we identify pathogenic mutations in eight individuals with FA-J in the gene encoding the DEAH-box DNA helicase BRIP1, also called FANCJ. This finding is compelling evidence that the Fanconi anemia pathway functions through a direct physical interaction with DNA.


Subject(s)
Chromosomes, Human, Pair 17 , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Mutation/genetics , RNA Helicases/deficiency , RNA Helicases/genetics , Fanconi Anemia Complementation Group Proteins , Genetic Complementation Test , Humans , Microsatellite Repeats , Molecular Sequence Data , Sequence Deletion
11.
Blood ; 104(13): 3927-35, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15319283

ABSTRACT

A number of DNA repair proteins also play roles in telomere metabolism. To investigate whether the accelerated telomere shortening reported in Fanconi anemia (FA) hematopoietic cells relates to a direct role of the FA pathway in telomere maintenance, we have analyzed telomere dynamics in Fancg-deficient mouse and human cells. We show here that both hematopoietic (stem and differentiated bone marrow cells, B and T lymphocytes) and nonhematopoietic (germ cells, mouse embryonic fibroblasts [MEFs]) Fancg(-/-) mouse cells display normal telomere length, normal telomerase activity, and normal chromosome end-capping, even in the presence of extensive clastogen-induced cytogenetic instability (mitomycin C [MMC], gamma-radiation). In addition, telomerase-deficient MEFs with humanlike telomere length and decreased Fancg expression (G5 Terc(-/-)/Fancg shRNA3 MEFs) display normal telomere maintenance. Finally, early-passage primary fibroblasts from patients with FA of complementation group G as well as primary human cells with reduced FANCG expression (FANCG shRNA IMR90 cells) show no signs of telomere dysfunction. Our observations indicate that accelerated telomere shortening in patients with FA is not due to a role of FANCG at telomeres but instead may be secondary to the disease. These findings suggest that telomerase-based therapies could be useful prophylactic agents in FA aplastic anemia by preserving their telomere reserve in the context of the disease.


Subject(s)
DNA-Binding Proteins/genetics , Telomere/genetics , Animals , Base Sequence , DNA Primers , DNA Repair/genetics , DNA-Binding Proteins/deficiency , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group G Protein , Fibroblasts/physiology , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA/genetics , RNA/metabolism , Spleen/cytology , Spleen/physiology , Telomerase/deficiency , Telomerase/genetics , Telomerase/metabolism
12.
J Biol Chem ; 279(38): 39421-30, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15262960

ABSTRACT

The Fanconi anemia (FA) protein FANCF is an essential component of a nuclear core complex that protects the genome against chromosomal instability, but the specific function of FANCF is still poorly understood. Based upon the homology between human and Xenopus laevis FANCF, we carried out an extensive mutagenesis study to examine which domains are functionally important and to gain more insight into the function of FANCF. In contrast to previous suggestions, we show that FANCF does not have a ROM-like function. We found that the C terminus of FANCF interacts directly with FANCG and allows the assembly of other FA proteins into a stable complex. The N terminus appears to stabilize the interaction with FANCA and FANCG and is essential for the binding of the FANCC/FANCE subcomplex. We identified several important amino acids in this N-terminal region but, surprisingly, many amino acid changes failed to affect the function of the FANCF protein. Our data demonstrate that FANCF acts as a flexible adaptor protein that plays a key role in the proper assembly of the FA core complex.


Subject(s)
Cell Nucleus/physiology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Fanconi Anemia Complementation Group F Protein , Humans , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , Sequence Homology, Amino Acid , Structure-Activity Relationship , Xenopus
13.
DNA Repair (Amst) ; 3(1): 77-84, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14697762

ABSTRACT

The genome protection pathway that is defective in patients with Fanconi anemia (FA) is controlled by at least eight genes, including BRCA2. A key step in the pathway involves the monoubiquitylation of FANCD2, which critically depends on a multi-subunit nuclear 'core complex' of at least six FANC proteins (FANCA, -C, -E, -F, -G, and -L). Except for FANCL, which has WD40 repeats and a RING finger domain, no significant domain structure has so far been recognized in any of the core complex proteins. By using a homology search strategy comparing the human FANCG protein sequence with its ortholog sequences in Oryzias latipes (Japanese rice fish) and Danio rerio (zebrafish) we identified at least seven tetratricopeptide repeat motifs (TPRs) covering a major part of this protein. TPRs are degenerate 34-amino acid repeat motifs which function as scaffolds mediating protein-protein interactions, often found in multiprotein complexes. In four out of five TPR motifs tested (TPR1, -2, -5, and -6), targeted missense mutagenesis disrupting the motifs at the critical position 8 of each TPR caused complete or partial loss of FANCG function. Loss of function was evident from failure of the mutant proteins to complement the cellular FA phenotype in FA-G lymphoblasts, which was correlated with loss of binding to FANCA. Although the TPR4 mutant fully complemented the cells, it showed a reduced interaction with FANCA, suggesting that this TPR may also be of functional importance. The recognition of FANCG as a typical TPR protein predicts this protein to play a key role in the assembly and/or stabilization of the nuclear FA protein core complex.


Subject(s)
DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Lymphocytes/metabolism , Mutation, Missense , Repetitive Sequences, Amino Acid , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Nucleus , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group G Protein , Humans , Lymphocytes/pathology , Molecular Sequence Data , Mutagenesis , Oryzias , Precipitin Tests , Sequence Homology, Amino Acid , Zebrafish
14.
Blood ; 103(7): 2498-503, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14630800

ABSTRACT

Fanconi anemia (FA) is an autosomal recessive syndrome featuring diverse symptoms including progressive bone marrow failure and early occurrence of acute myeloid leukemia. Nine genetic subtypes have been described for FA (A, B, C, D1, D2, E, F, G, and L), all of which have been connected to distinct disease genes, except B. Here we report on 8 unrelated FA patients who were excluded from the known subtypes on the basis of phenotypic correction or genetic data. Four of these cell lines failed to complement each other in somatic cell hybrids and therefore represent a new group, termed FA-I. The remaining cell lines complemented group FA-I but did not complement each other, thus representing a second new group, FA-J. Both FA-I and -J cell lines were capable of forming an FA multiprotein core complex. This complex is required for activation of the FANCD2 protein by mono-ubiquitination, a key downstream event in the FA pathway. In FA-I cells FANCD2 was not mono-ubiquitinated, indicating a defect upstream in the FA pathway, whereas in FA-J cells FANCD2 was mono-ubiquitinated, indicating a downstream defect. Our results suggest that the FA pathway of genome stabilization may be controlled by at least 11 different genes, including FANCI and FANCJ.


Subject(s)
Fanconi Anemia/classification , Fanconi Anemia/genetics , Polymorphism, Genetic , Cell Division , Cell Fusion , Cell Line , Child , Child, Preschool , Fanconi Anemia/pathology , Genes, Recessive , Genetic Complementation Test , Humans , Transfection
15.
Mol Ther ; 8(4): 600-10, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529833

ABSTRACT

Fanconi anemia (FA) is an autosomal recessive disease characterized by progressive bone marrow failure due to defective stem cell function. FA patients' cells are hypersensitive to DNA cross-linking agents such as mitomycin C (MMC), exposure to which results in cytogenetic aberrations and cell death. To date Moloney murine leukemia virus vectors have been used in clinical gene therapy. Recently, third-generation lentiviral vectors based on the HIV-1 genome have been developed for efficient gene transfer to hematopoietic stem cells. We generated a self-inactivating lentiviral vector expressing the FA group A cDNA driven by the murine stem cell virus U3 LTR promoter and used the vector to transduce side-population (SP) cells isolated from bone marrow of Fanconi anemia group A (Fanca) knockout mice. One thousand transduced SP cells reconstituted the bone marrow of sublethally irradiated Fanca recipient mice. Phenotype correction was demonstrated by stable hematopoiesis following MMC challenge. Using real-time PCR, one proviral vector DNA copy per cell was detected in all lineage-committed cells in the peripheral blood of both primary and secondary recipients. Our results suggest that the lentiviral vector transduces stem cells capable of self-renewal and long-term hematopoiesis in vivo and is potentially useful for clinical gene therapy of FA hematopoietic cells.


Subject(s)
DNA-Binding Proteins , Fanconi Anemia/drug therapy , Genetic Therapy , Genetic Vectors , Lentivirus , Proteins/genetics , Animals , Bone Marrow Transplantation , Fanconi Anemia Complementation Group A Protein , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Phenotype , Proteins/metabolism
16.
Acta Haematol ; 108(4): 231-6, 2002.
Article in English | MEDLINE | ID: mdl-12432219

ABSTRACT

Fanconi anemia (FA) is an autosomal recessively inherited disease with diverse clinical symptoms including developmental anomalies, predisposition to neoplasia, and a deficiency of hematopoietic stem cells resulting in progressive aplastic anemia. FA is genetically heterogeneous with at least 8 genes being implicated on the basis of functional complementation studies. To date, six FA genes are known: FANCA, FANCC, FANCD2, FANCE, FANCF and FANCG, all of which encode orphan proteins sharing no homology to each other nor to any other known protein. In addition, they do not appear to possess any domains with homology to currently known protein domains, which makes a prediction about their molecular action difficult. Studying the molecular evolution of FA genes and their products using sensitive database search methods such as PSI-BLAST may provide novel insight into the nature of the FA pathway and its relationship to hematopoiesis, embryonic development and the origin of malignancies. Preliminary results of such an approach show that at least one FA protein, FANCG, may contain a known domain, suggesting that this protein is a member of the family of tetratricopeptide repeat-containing proteins.


Subject(s)
Evolution, Molecular , Fanconi Anemia/genetics , Sequence Alignment , Algorithms , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Databases, Protein , Fanconi Anemia Complementation Group G Protein , Humans , Phylogeny , Protein Structure, Tertiary , Repetitive Sequences, Nucleic Acid
17.
Trends Mol Med ; 8(10): 458-60, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12383764

ABSTRACT

Surprisingly, biallelic mutations in the BRCA2 breast-cancer-susceptibility gene were found in Fanconi anemia (FA), a rare hereditary disorder characterized by chromosomal instability, hypersensitivity to DNA cross-linking agents, and cancer susceptibility. This suggests that a defect in the FA pathway might predispose to familial breast cancer. A previously reported molecular interaction between BRCA1 and the FA protein, FANCD2, supports the hypothesis that both breast-cancer-susceptibility genes are components of the FA pathway, functioning in DNA-damage response. However, an alternative hypothesis, that group FA-D1 with mutated BRCA2 represents a FA-like syndrome that is involved in a pathway distinct from the FA pathway, cannot be excluded. Similar syndromes would also be expected when recombination genes, such as Rad51 and its paralogs, are mutated.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia/genetics , Genes, BRCA2 , Breast Neoplasms/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein , Female , Genes, BRCA1 , Genetic Predisposition to Disease , Humans , Male , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Rad51 Recombinase
18.
Blood ; 100(6): 2032-9, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12200363

ABSTRACT

Fanconi anemia (FA) is a rare autosomal recessive disease, characterized by bone marrow failure and cancer predisposition. So far, 8 complementation groups have been identified, although mutations in FANCA account for the disease in the majority of FA patients. In this study we characterized the hematopoietic phenotype of a Fanca knockout mouse model and corrected the main phenotypic characteristics of the bone marrow (BM) progenitors using retroviral vectors. The hematopoiesis of these animals was characterized by a modest though significant thrombocytopenia, consistent with reduced numbers of BM megakaryocyte progenitors. As observed in other FA models, the hematopoietic progenitors from Fanca(-/-) mice were highly sensitive to mitomycin C (MMC). In addition, we observed for the first time in a FA mouse model a marked in vitro growth defect of Fanca(-/-) progenitors, either when total BM or when purified Lin(-)Sca-1(+) cells were subjected to in vitro stimulation. Liquid cultures of Fanca(-/-) BM that were stimulated with stem cell factor plus interleukin-11 produced low numbers of granulocyte macrophage colony-forming units, contained a high proportion of apoptotic cells, and generated a decreased proportion of granulocyte versus macrophage cells, compared to normal BM cultures. Aiming to correct the phenotype of Fanca(-/-) progenitors, purified Lin(-)Sca-1(+) cells were transduced with retroviral vectors encoding the enhanced green fluorescent protein (EGFP) gene and human FANCA genes. Lin(-)Sca-1(+) cells from Fanca(-/-) mice were transduced with an efficiency similar to that of samples from wild-type mice. More significantly, transductions with FANCA vectors corrected both the MMC hypersensitivity as well as the impaired ex vivo expansion ability that characterized the BM progenitors of Fanca(-/-) mice.


Subject(s)
DNA-Binding Proteins , Fanconi Anemia/pathology , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Animals , Apoptosis , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Culture Techniques , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group A Protein , Genetic Vectors/therapeutic use , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitomycin/pharmacology , Phenotype , Proteins/genetics , Retroviridae/genetics , Transduction, Genetic
19.
Genes Cells ; 7(3): 333-42, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11918676

ABSTRACT

BACKGROUND: Fanconi anaemia (FA) is an autosomal recessive chromosomal instability disorder. Six distinct FA disease genes have been identified, the products of which function in an integrated pathway that is thought to support a nuclear caretaker function. Comparison of FA gene characteristics in different species may help to unravel the molecular function of the FA pathway. RESULTS: We have cloned the murine homologue of the Fanconi anaemia complementation group G gene, FANCG/XRCC9. The murine Fancg protein shows an 83% similarity to the human protein sequence, and has a predicted molecular weight of 68.5 kDa. Expression of mouse Fancg in human FA-G lymphoblasts fully corrects their cross-linker hypersensitivity. At mRNA and protein levels we detected the co-expression of Fancg and Fanca in murine tissues. In addition, mouse Fancg and Fanca proteins co-purify by immunoprecipitation. Upon transfection into Fanca-deficient mouse embryonic fibroblasts EGFP-Fancg chimeric protein was detectable in the nucleus. CONCLUSIONS: We identified a murine cDNA, Fancg, which cross-complements the cellular defect of human FA-G cells and thus represents a true homologue of human FANCG. Spleen, thymus and testis showed the highest Fancg expression levels. Although Fancg and Fanca are able to form a complex, this interaction is not required for Fancg to accumulate in the nuclear compartment.


Subject(s)
DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group G Protein , Fibroblasts/metabolism , Green Fluorescent Proteins , Luminescent Proteins , Mice , Molecular Sequence Data , Proteins/metabolism , RNA, Messenger , Sequence Alignment
20.
Hum Mol Genet ; 11(3): 273-81, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11823446

ABSTRACT

Fanconi anemia (FA) is a heterogeneous autosomal recessive chromosomal instability syndrome associated with diverse developmental abnormalities, progressive bone marrow failure and a predisposition to cancer. Spontaneous chromosomal breakage and hypersensitivity to DNA cross-linking agents characterize the cellular FA phenotype. The gene affected in FA complementation group G patients was initially identified as XRCC9, for its ability to partially correct the cellular phenotype of the Chinese hamster ovary (CHO) cell mutant UV40. By targeted disruption we generated Fancg/Xrcc9 null mice. Fancg knock-out (KO) mice were born at expected Mendelian frequencies and showed normal viability. In mice, functional loss of Fancg did not result in developmental abnormalities or a pronounced incidence of malignancies. During a 1 year follow-up, blood cell parameters of Fancg KO mice remained within normal values, revealing no signs of anemia. Male and female mice deficient in Fancg showed hypogonadism and impaired fertility, consistent with the phenotype of FA patients. Mouse embryonic fibroblasts (MEFs) from the KO animals exhibited the FA characteristic cellular response in showing enhanced spontaneous chromosomal instability and a hyper-responsiveness to the clastogenic and antiproliferative effects of the cross-linking agent mitomycin C (MMC). The sensitivity to UV, X-rays and methyl methanesulfonate, reported for the CHO mutant cell line UV40, was not observed in Fancg(-/-) MEFs. Despite a lack of hematopoietic failure in the KO mice, clonogenic survival of bone marrow cells in vitro was strongly reduced in the presence of MMC. The characteristics of the Fancg(-/-) mice closely resemble those reported for Fancc and Fanca null mice, supporting a tight interdependence of the corresponding gene products in a common pathway.


Subject(s)
DNA-Binding Proteins/genetics , Mitomycin/pharmacology , Animals , DNA/drug effects , DNA Damage , Drug Hypersensitivity , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group G Protein , Female , Fibroblasts , Hematopoietic Stem Cells/drug effects , Infertility/genetics , Male , Mice , Mice, Knockout , Ovary/abnormalities , Testis/abnormalities
SELECTION OF CITATIONS
SEARCH DETAIL
...