Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters








Publication year range
1.
Stem Cell Reports ; 11(5): 1075-1091, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30449320

ABSTRACT

Fanconi anemia (FA) causes bone marrow failure early during childhood, and recent studies indicate that a hematopoietic defect could begin in utero. We performed a unique kinetics study of hematopoiesis in Fancg-/- mouse embryos, between the early embryonic day 11.5 (E11.5) to E12.5 developmental window (when the highest level of hematopoietic stem cells [HSC] amplification takes place) and E14.5. This study reveals a deep HSC defect with exhaustion of proliferative and self-renewal capacities very early during development, together with severe FA clinical and biological manifestations, which are mitigated at E14.5 due to compensatory mechanisms that help to ensure survival of Fancg-/- embryos. It also reports that a deep HSC defect is also observed during human FA development, and that human FA fetal liver (FL) HSCs present a transcriptome profile similar to that of mouse E12.5 Fancg-/- FL HSCs. Altogether, our results highlight that early mouse FL could represent a good alternative model for studying Fanconi pathology.


Subject(s)
Embryonic Development , Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Animals , Apoptosis , Cell Cycle , DNA Damage , Embryo, Mammalian/pathology , Erythrocytes/metabolism , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/metabolism , Female , Gene Ontology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Liver/embryology , Liver/metabolism , Mice, Inbred C57BL , Phenotype , Placenta/metabolism , Pregnancy , Transcriptome/genetics
2.
Nat Commun ; 9(1): 2280, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891926

ABSTRACT

Defects in DNA repair can cause various genetic diseases with severe pathological phenotypes. Fanconi anemia (FA) is a rare disease characterized by bone marrow failure, developmental abnormalities, and increased cancer risk that is caused by defective repair of DNA interstrand crosslinks (ICLs). Here, we identify the deubiquitylating enzyme USP48 as synthetic viable for FA-gene deficiencies by performing genome-wide loss-of-function screens across a panel of human haploid isogenic FA-defective cells (FANCA, FANCC, FANCG, FANCI, FANCD2). Thus, as compared to FA-defective cells alone, FA-deficient cells additionally lacking USP48 are less sensitive to genotoxic stress induced by ICL agents and display enhanced, BRCA1-dependent, clearance of DNA damage. Consequently, USP48 inactivation reduces chromosomal instability of FA-defective cells. Our results highlight a role for USP48 in controlling DNA repair and suggest it as a potential target that could be therapeutically exploited for FA.


Subject(s)
DNA Repair/genetics , DNA Repair/physiology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , BRCA1 Protein/metabolism , CRISPR-Cas Systems , Cell Line , Chromosomal Instability , DNA Damage , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Knockout Techniques , Genetic Therapy , Histones/metabolism , Humans , Mutation , Rad51 Recombinase/metabolism , Ubiquitin-Specific Proteases/deficiency , Ubiquitination
3.
Radiat Res ; 189(6): 560-578, 2018 06.
Article in English | MEDLINE | ID: mdl-29584588

ABSTRACT

Squamous cell carcinomas of the head and neck are appearing with increased frequency in both marrow transplanted and non-transplanted Fanconi anemia (FA) patients. FA patients commonly display radiosensitivity of epithelial tissues, complicating effective radiotherapy. Fancd2-/- mice (C57BL/6J and 129/Sv background) demonstrate epithelial tissue sensitivity to single-fraction or fractionated irradiation to the head and neck and distant marrow suppression (abscopal effect), both ameliorated by intraoral administration of the mitochondrial-targeted antioxidant, GS-nitroxide, JP4-039. We now report that mice of two other FA genotypes, Fancg-/- (B6) and the most prevalent human genotype Fanca-/- (129/Sv), also demonstrate: 1. reduced longevity of hematopoiesis in long-term bone marrow cultures; 2. radiosensitivity of bone marrow stromal cell lines; and 3. head and neck radiation-induced severe mucositis and abscopal suppression of distant marrow hematopoiesis. Intraoral administration of JP4-039/F15, but not non-mitochondrial-targeted 4-amino-Tempo/F15 or F15 alone, prior to each radiation treatment ameliorated both local and abscopal radiation effects. Head and neck irradiated TGF-ß-resistant SMAD3-/- (129/Sv) mice and double-knockout SMAD3-/- Fancd2-/- (129/Sv) mice treated daily with TGF-ß receptor antagonist, LY364947, still displayed abscopal bone marrow suppression, implicating a non-TGF-ß mechanism. Thus, amelioration of both local normal tissue radiosensitivity and distant marrow suppression by intraoral administration of JP4-039 in Fancg-/- and Fanca-/- mice supports a clinical trial of this locally administered normal tissue radioprotector and mitigator during head and neck irradiation in FA patients.


Subject(s)
Bone Marrow/drug effects , Head and Neck Neoplasms/radiotherapy , Mucositis/drug therapy , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Administration, Oral , Animals , Bone Marrow/pathology , Bone Marrow/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group G Protein/deficiency , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Interleukin-3/metabolism , Mice , Mitomycin/pharmacology , Mucositis/metabolism , Mucositis/pathology , Nitrogen Oxides/therapeutic use , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/therapeutic use , Signal Transduction/drug effects , Signal Transduction/radiation effects , Transforming Growth Factor beta/metabolism
4.
J Exp Med ; 211(6): 1011-8, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24799500

ABSTRACT

Fanconi anemia is a rare genetic disorder that can lead to bone marrow failure, congenital abnormalities, and increased risk for leukemia and cancer. Cells with loss-of-function mutations in the FANC pathway are characterized by chromosome fragility, altered mutability, and abnormal regulation of the nonhomologous end-joining (NHEJ) pathway. Somatic hypermutation (SHM) and immunoglobulin (Ig) class switch recombination (CSR) enable B cells to produce high-affinity antibodies of various isotypes. Both processes are initiated after the generation of dG:dU mismatches by activation-induced cytidine deaminase. Whereas SHM involves an error-prone repair process that introduces novel point mutations into the Ig gene, the mismatches generated during CSR are processed to create double-stranded breaks (DSBs) in DNA, which are then repaired by the NHEJ pathway. As several lines of evidence suggest a possible role for the FANC pathway in SHM and CSR, we analyzed both processes in B cells derived from Fanca(-/-) mice. Here we show that Fanca is required for the induction of transition mutations at A/T residues during SHM and that despite globally normal CSR function in splenic B cells, Fanca is required during CSR to stabilize duplexes between pairs of short microhomology regions, thereby impeding short-range recombination downstream of DSB formation.


Subject(s)
B-Lymphocytes/metabolism , Fanconi Anemia Complementation Group A Protein/metabolism , Immunoglobulin Class Switching/genetics , Point Mutation , Somatic Hypermutation, Immunoglobulin/genetics , Animals , B-Lymphocytes/immunology , Base Sequence , Blotting, Western , Cell Line , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group G Protein/metabolism , Humans , Immunoglobulin Switch Region/genetics , Mice, 129 Strain , Mice, Knockout , Molecular Sequence Data , MutS Homolog 2 Protein/metabolism , Polymerase Chain Reaction , Recombination, Genetic
5.
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
6.
Radiat Res ; 176(5): 603-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21939290

ABSTRACT

Fanconi anemia (FA) is an inherited disorder characterized by defective DNA repair and cellular sensitivity to DNA crosslinking agents. Clinically, FA is associated with high risk for marrow failure, leukemia and head and neck squamous cell carcinoma (HNSCC). Radiosensitivity in FA patients compromises the use of total-body irradiation for hematopoietic stem cell transplantation and radiation therapy for HNSCC. A radioprotector for the surrounding tissue would therefore be very valuable during radiotherapy for HNSCC. Clonogenic radiation survival curves were determined for pre- or postirradiation treatment with the parent nitroxide Tempol or JP4-039 in cells of four FA patient-derived cell lines and two transgene-corrected subclonal lines. FancG(-/-) (PD326) and FancD2(-/-) (PD20F) patient lines were more sensitive to the DNA crosslinking agent mitomycin C (MMC) than their transgene-restored subclonal cell lines (both P < 0.0001). FancD2(-/-) cells were more radiosensitive than the transgene restored subclonal cell line (ñ = 2.0 ± 0.7 and 4.7 ± 2.2, respectively, P = 0.03). In contrast, FancG(-/-) cells were radioresistant relative to the transgene-restored subclonal cell line (ñ = 9.4 ± 1.5 and 2.2 ± 05, respectively, P = 0.001). DNA strand breaks measured by the comet assay correlated with radiosensitivity. Cell lines from a Fanc-C and Fanc-A patients showed radiosensitivity similar to that of Fanc-D2(-/-) cells. A fluorophore-tagged JP4-039 (BODIPY-FL) analog targeted the mitochondria of the cell lines. Preirradiation or postirradiation treatment with JP4-039 at a lower concentration than Tempol significantly increased the radioresistance and stabilized the antioxidant stores of all cell lines. Tempol increased the toxicity of MMC in FancD2(-/-) cells. These data provide support for the potential clinical use of JP4-039 for normal tissue radioprotection during chemoradiotherapy in FA patients.


Subject(s)
Fanconi Anemia/pathology , Nitrogen Oxides/pharmacology , Radiation-Protective Agents/pharmacology , Biological Transport , Cell Line , Clone Cells , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group G Protein/deficiency , Glutathione/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Mitomycin/pharmacology , Nitrogen Oxides/metabolism , Radiation-Protective Agents/metabolism , Transgenes/genetics
7.
Cancer Res ; 70(23): 9703-10, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21118969

ABSTRACT

Fanconi anemia (FA) results from mutations in the FANC genes and is characterized by bone marrow failure, birth defects, and a high incidence of cancer. FANCG is a part of the FA core complex that is responsible for monoubiquitination of FANCD2 and FANCI. The precise role of the FA pathway is not well understood, although it may be involved in homologous recombination (HR), nonhomologous end joining, and translesion synthesis (TLS). Fancd2(-/-) mice have a more severe phenotype than Fancg(-/-), and other FA core complex-deficient mice, although both Fancg and Fancd2 belong to the same FA pathway. We hypothesized that Fancd2 deficiency results in a more severe phenotype because Fancd2 also has a FA pathway-independent function in the maintenance of genomic integrity. To test this hypothesis, we determined the level of DNA damage and genomic instability in Fancd2(-/-), Fancg(-/-), and wild-type controls. Fancd2(-/-) mice displayed a higher magnitude of chromosomal breakage and micronucleus formation than the wild-type or Fancg(-/-) mice. Also, DNA strand breaks were increased in Fancd2(-/-) but not in Fancg(-/-) mice. In addition, Fancd2(-/-) mice displayed an elevated frequency of DNA deletions, resulting from HR at the endogenous p(un) locus. In contrast, in Fancg(-/-) mice, the frequency of DNA deletions was decreased. Thus, Fancd2 but not Fancg deficiency results in elevated chromosomal/DNA breakage and permanent genome rearrangements. This provides evidence that Fancd2 plays an additional role in the maintenance of genomic stability than Fancg, which might explain the higher predisposition to cancer seen in the Fancd2(-/-) mice.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/genetics , Genomic Instability , Animals , Chromosome Breakage , Comet Assay , DNA Damage , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group G Protein/deficiency , Female , Gene Deletion , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Micronuclei, Chromosome-Defective , Micronucleus Tests , Recombination, Genetic , Retinal Pigment Epithelium/metabolism
8.
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
9.
J Clin Invest ; 117(5): 1440-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17431503

ABSTRACT

The Fanconi anemia (FA) pathway maintains genomic stability in replicating cells. Some sporadic breast, ovarian, pancreatic, and hematological tumors are deficient in FA pathway function, resulting in sensitivity to DNA-damaging agents. FA pathway dysfunction in these tumors may result in hyperdependence on alternative DNA repair pathways that could be targeted as a treatment strategy. We used a high-throughput siRNA screening approach that identified ataxia telangiectasia mutated (ATM) as a critical kinase for FA pathway-deficient human fibroblasts. Human fibroblasts and murine embryonic fibroblasts deficient for the FA pathway were observed to have constitutive ATM activation and Fancg(-/-)Atm(-/-) mice were found to be nonviable. Abrogation of ATM function in FA pathway-deficient cells resulted in DNA breakage, cell cycle arrest, and apoptotic cell death. Moreover, Fanconi anemia complementation group G- (FANCG-) and FANCC-deficient pancreatic tumor lines were more sensitive to the ATM inhibitor KU-55933 than isogenic corrected lines. These data suggest that ATM and FA genes function in parallel and compensatory roles to maintain genomic integrity and cell viability. Pharmaceutical inhibition of ATM may have a role in the treatment of FA pathway-deficient human cancers.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , DNA Damage , DNA-Binding Proteins/deficiency , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group G Protein/physiology , HeLa Cells , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Proteins/deficiency
10.
Clin Cancer Res ; 11(20): 7508-15, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16243825

ABSTRACT

PURPOSE: BRCA2, FANCC, and FANCG gene mutations are present in a subset of pancreatic cancer. Defects in these genes could lead to hypersensitivity to interstrand cross-linkers in vivo and a more optimal treatment of pancreatic cancer patients based on the genetic profile of the tumor. EXPERIMENTAL DESIGN: Two retrovirally complemented pancreatic cancer cell lines having defects in the Fanconi anemia pathway, PL11 (FANCC-mutated) and Hs766T (FANCG-mutated), as well as several parental pancreatic cancer cell lines with or without mutations in the Fanconi anemia/BRCA2 pathway, were assayed for in vitro and in vivo sensitivities to various chemotherapeutic agents. RESULTS: A distinct dichotomy of drug responses was observed. Fanconi anemia-defective cancer cells were hypersensitive to the cross-linking agents mitomycin C (MMC), cisplatin, chlorambucil, and melphalan but not to 5-fluorouracil, gemcitabine, doxorubicin, etoposide, vinblastine, or paclitaxel. Hypersensitivity to cross-linking agents was confirmed in vivo; FANCC-deficient xenografts of PL11 and BRCA2-deficient xenografts of CAPAN1 regressed on treatment with two different regimens of MMC whereas Fanconi anemia-proficient xenografts did not. The MMC response comprised cell cycle arrest, apoptosis, and necrosis. Xenografts of PL11 also regressed after a single dose of cyclophosphamide whereas xenografts of genetically complemented PL11(FANCC) did not. CONCLUSIONS: MMC or other cross-linking agents as a clinical therapy for pancreatic cancer patients with tumors harboring defects in the Fanconi anemia/BRCA2 pathway should be specifically investigated.


Subject(s)
Cross-Linking Reagents/pharmacology , Fanconi Anemia Complementation Group Proteins/genetics , Mitomycin/pharmacology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chlorambucil/pharmacology , Cisplatin/pharmacology , Cross-Linking Reagents/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Etoposide/pharmacology , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group Proteins/deficiency , Female , Fluorouracil/pharmacology , Humans , Inhibitory Concentration 50 , Melphalan/pharmacology , Mice , Mice, Nude , Mitomycin/therapeutic use , Mutation , Paclitaxel/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Time Factors , Vinblastine/pharmacology , Xenograft Model Antitumor Assays/methods , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL