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











Publication year range
1.
Genes (Basel) ; 12(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33918752

ABSTRACT

Fanconi anemia (FA) patients have an exacerbated risk of head and neck squamous cell carcinoma (HNSCC). Treatment is challenging as FA patients display enhanced toxicity to standard treatments, including radio/chemotherapy. Therefore, better therapies as well as new disease models are urgently needed. We have used CRISPR/Cas9 editing tools in order to interrupt the human FANCA gene by the generation of insertions/deletions (indels) in exon 4 in two cancer cell lines from sporadic HNSCC having no mutation in FA-genes: CAL27 and CAL33 cells. Our approach allowed efficient editing, subsequent purification of single-cell clones, and Sanger sequencing validation at the edited locus. Clones having frameshift indels in homozygosis did not express FANCA protein and were selected for further analysis. When compared with parental CAL27 and CAL33, FANCA-mutant cell clones displayed a FA-phenotype as they (i) are highly sensitive to DNA interstrand crosslink (ICL) agents such as mitomycin C (MMC) or cisplatin, (ii) do not monoubiquitinate FANCD2 upon MMC treatment and therefore (iii) do not form FANCD2 nuclear foci, and (iv) they display increased chromosome fragility and G2 arrest after diepoxybutane (DEB) treatment. These FANCA-mutant clones display similar growth rates as their parental cells. Interestingly, mutant cells acquire phenotypes associated with more aggressive disease, such as increased migration in wound healing assays. Therefore, CAL27 and CAL33 cells with FANCA mutations are phenocopies of FA-HNSCC cells.


Subject(s)
Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia/pathology , Gene Editing , Head and Neck Neoplasms/pathology , Mutation , Phenotype , Squamous Cell Carcinoma of Head and Neck/pathology , Cell Movement , Cell Proliferation , DNA Damage , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Head and Neck Neoplasms/genetics , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Cells, Cultured , Wound Healing
2.
Mol Genet Genomics ; 296(1): 103-112, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33025164

ABSTRACT

Reduced fertility is a common clinical feature of the individuals with Fanconi anemia (FA), a rare autosomal recessive disorder due to deficiency in FA pathway during DNA repair. Our previous study reported that the heterozygous pathogenic variants in FANCA (Fanconi anemia complementation group A) induced premature ovarian insufficiency (POI). However, the genotype-phenotype correlation in POI caused by FANCA variants remains considerably uncertain. Herein, a heterozygous non-frameshift Fanca-mutated mouse strain (Fanca+/hypo) carrying a 9-bp deletion (c.3581del9, p.QEA1194-1196del) was generated. The mutant mice exhibited slightly decreased Fanca protein level in ovaries, suggesting the non-frameshift deletion mutant is hypomorphic. Female fertility test showed decreased number of litters, litter sizes and prolonged litter interval time in the female Fanca+/hypo mice compared to wild-type mice. Follicle counting revealed a consistent decreasing pattern of follicle numbers in Fanca+/hypo females compared to that in wild-type mice with aging. Furthermore, embryonic fibroblasts of Fanca+/hypo mice were hyper-responsive to Mitomycin C in vitro, demonstrating a partial loss of function of this hypomorphic Fanca mutant in DNA repair. Collectively, our experimental observations suggest that the hypomorphic Fanca allele is sufficient to reduce female fertility in mice, providing new insights into the genetic counseling of FANCA variants in subfertile women.


Subject(s)
Base Sequence , Fanconi Anemia Complementation Group A Protein/genetics , Infertility, Female/genetics , Infertility/genetics , Primary Ovarian Insufficiency/genetics , Sequence Deletion , Alkylating Agents/pharmacology , Animals , DNA Repair/drug effects , Disease Models, Animal , Embryo, Mammalian , Fanconi Anemia Complementation Group A Protein/deficiency , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Heterozygote , Humans , Infertility/metabolism , Infertility/pathology , Infertility, Female/metabolism , Infertility, Female/pathology , Litter Size , Mice , Mice, Knockout , Mitomycin/pharmacology , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Primary Cell Culture , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology
3.
JCI Insight ; 5(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32434992

ABSTRACT

NOD-like receptor 12 (NLRP12) is a member of the nucleotide-binding domain and leucine-rich repeat containing receptor inflammasome family that plays a central role in innate immunity. We previously showed that DNA damage upregulated NLRP12 in hematopoietic stem cells (HSCs) of mice deficient in the DNA repair gene Fanca. However, the role of NLRP12 in HSC maintenance is not known. Here, we show that persistent DNA damage-induced NLRP12 improves HSC function in both mouse and human models of DNA repair deficiency and aging. Specifically, treatment of Fanca-/- mice with the DNA cross-linker mitomycin C or ionizing radiation induces NLRP12 upregulation in phenotypic HSCs. NLRP12 expression is specifically induced by persistent DNA damage. Functionally, knockdown of NLRP12 exacerbates the repopulation defect of Fanca-/- HSCs. Persistent DNA damage-induced NLRP12 was also observed in the HSCs from aged mice, and depletion of NLRP12 in these aged HSCs compromised their self-renewal and hematopoietic recovery. Consistently, overexpression of NLRP12 substantially improved the long-term repopulating function of Fanca-/- and aged HSCs. Finally, persistent DNA damage-induced NLRP12 maintains the function of HSCs from patients with FA or aged donors. These results reveal a potentially novel role of NLRP12 in HSC maintenance and suggest that NLRP12 targeting has therapeutic potential in DNA repair disorders and aging.


Subject(s)
Aging/metabolism , DNA Damage , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Aging/genetics , Aging/pathology , Animals , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/metabolism , Hematopoietic Stem Cells/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout
4.
Stem Cells ; 37(7): 937-947, 2019 07.
Article in English | MEDLINE | ID: mdl-30977208

ABSTRACT

Emerging evidence has shown that resting quiescent hematopoietic stem cells (HSCs) prefer to utilize anaerobic glycolysis rather than mitochondrial respiration for energy production. Compelling evidence has also revealed that altered metabolic energetics in HSCs underlies the onset of certain blood diseases; however, the mechanisms responsible for energetic reprogramming remain elusive. We recently found that Fanconi anemia (FA) HSCs in their resting state are more dependent on mitochondrial respiration for energy metabolism than on glycolysis. In the present study, we investigated the role of deficient glycolysis in FA HSC maintenance. We observed significantly reduced glucose consumption, lactate production, and ATP production in HSCs but not in the less primitive multipotent progenitors or restricted hematopoietic progenitors of Fanca-/- and Fancc-/- mice compared with that of wild-type mice, which was associated with an overactivated p53 and TP53-induced glycolysis regulator, the TIGAR-mediated metabolic axis. We utilized Fanca-/- HSCs deficient for p53 to show that the p53-TIGAR axis suppressed glycolysis in FA HSCs, leading to enhanced pentose phosphate pathway and cellular antioxidant function and, consequently, reduced DNA damage and attenuated HSC exhaustion. Furthermore, by using Fanca-/- HSCs carrying the separation-of-function mutant p53R172P transgene that selectively impairs the p53 function in apoptosis but not cell-cycle control, we demonstrated that the cell-cycle function of p53 was not required for glycolytic suppression in FA HSCs. Finally, ectopic expression of the glycolytic rate-limiting enzyme PFKFB3 specifically antagonized p53-TIGAR-mediated metabolic reprogramming in FA HSCs. Together, our results suggest that p53-TIGAR metabolic axis-mediated glycolytic suppression may play a compensatory role in attenuating DNA damage and proliferative exhaustion in FA HSCs. Stem Cells 2019;37:937-947.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Fanconi Anemia/genetics , Genomic Instability , Hematopoietic Stem Cells/metabolism , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle/genetics , DNA Damage , Disease Models, Animal , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Gene Expression Regulation , Glycolysis/genetics , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Oxidative Phosphorylation , Pentose Phosphate Pathway/genetics , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
5.
J Bone Miner Res ; 33(11): 2007-2020, 2018 11.
Article in English | MEDLINE | ID: mdl-29989666

ABSTRACT

Fanconi anemia (FA) is a rare genetic disorder associated with a progressive decline in hematopoietic stem cells leading to bone marrow failure. FA is also characterized by a variety of developmental defects including short stature and skeletal malformations. More than half of children affected with FA have radial-ray abnormalities, and many patients have early onset osteopenia/osteoporosis. Although many Fanconi anemia genes have been identified and a molecular pathway defined, the underlying mechanism leading to bone defects remains elusive. To understand the role of FA genes in skeletal development and bone microarchitecture, we evaluated bone physiology during embryogenesis and in adult FancA- and FancC-deficient mice. We found that both FancA-/- and FancC-/- embryos have abnormal skeletal development shown by skeletal malformations, growth delay, and reduced bone mineralization. FancC-/- adult mice present altered bone morphology and microarchitecture with a significant decrease in cortical bone mineral density in a sex-specific manner. Mechanical testing revealed that male but not female FancC-/- mice show reduced bone strength compared with their wild-type littermates. Ex vivo cultures showed that FancA-/- and FancC-/- bone marrow-derived mesenchymal stem cells (BM MSC) have impaired differentiation capabilities together with altered gene expression profiles. Our results suggest that defective bone physiology in FA occurs in utero and possibly results from altered BM MSC function. These results provide valuable insights into the mechanism involved in FA skeletal defects. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/abnormalities , Bone and Bones/physiopathology , Calcification, Physiologic , Fanconi Anemia Complementation Group C Protein/genetics , Gene Deletion , Animals , Bone and Bones/pathology , Cell Differentiation , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Female , Gene Expression Regulation , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Skull/embryology , Spine/embryology
6.
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
7.
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
9.
Stem Cells ; 33(11): 3382-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26212365

ABSTRACT

Fanconi anemia (FA) patients develop bone marrow (BM) failure or leukemia. One standard care for these devastating complications is hematopoietic stem cell transplantation. We identified a group of mesenchymal stromal cells (MSCs)-derived metabolites, glycerophospholipids, and their endogenous inhibitor, 5-(tetradecyloxy)-2-furoic acid (TOFA), as regulators of donor hematopoietic stem and progenitor cells. We provided two pieces of evidence that TOFA could improve hematopoiesis-supporting function of FA MSCs: (a) limiting-dilution cobblestone area-forming cell assay revealed that TOFA significantly increased cobblestone colonies in Fanca-/- or Fancd2-/- cocultures compared to untreated cocultures. (b) Competitive repopulating assay using output cells collected from cocultures showed that TOFA greatly alleviated the abnormal expansion of the donor myeloid (CD45.2+Gr1+Mac1+) compartment in both peripheral blood and BM of recipient mice transplanted with cells from Fanca-/- or Fancd2-/- cocultures. Furthermore, mechanistic studies identified Tlr4 signaling as the responsible pathway mediating the effect of glycerophospholipids. Thus, targeting glycerophospholipid biosynthesis in FA MSCs could be a therapeutic strategy to improve hematopoiesis and stem cell transplantation.


Subject(s)
Cell Differentiation/physiology , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/deficiency , Glycerophospholipids/biosynthesis , Hematopoietic Stem Cells/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Coculture Techniques , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
10.
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
11.
Blood ; 123(12): 1938-47, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24501220

ABSTRACT

Fanconi anemia (FA) is a genetic disorder associated with bone marrow (BM) failure and leukemia. Recent studies demonstrate variable immune defects in FA. However, the cause for FA immunodeficiency is unknown. Here we report that deletion of Fanca or Fancd2 dysregulates the suppressive activity of regulatory T cells (Tregs), shown functionally as exacerbation of graft-vs-host disease (GVHD) in mice. Recipient mice of Fanca(-/-) or Fancd2(-/-) BM chimeras exhibited severe acute GVHD after allogeneic BM transplantation (BMT). T cells from Fanca(-/-) or Fancd2(-/-) mice induced higher GVHD lethality than those from wild-type (WT) littermates. FA Tregs possessed lower proliferative suppression potential compared with WT Tregs, as demonstrated by in vitro proliferation assay and BMT. Analysis of CD25(+)Foxp3(+) Tregs indicated that loss of Fanca or Fancd2 dysregulated Foxp3 target gene expression. Additionally, CD25(+)Foxp3(+) Tregs of Fanca(-/-) or Fancd2(-/-) mice were less efficient in suppressing the production of GVHD-associated inflammatory cytokines. Consistently, aberrant NF-κB activity was observed in infiltrated T cells from FA GVHD mice. Conditional deletion of p65 in FA Tregs decreased GVHD mortality. Our study uncovers an essential role for FA proteins in maintaining Treg homeostasis, possibly explaining, at least in part, the immune deficiency reported in some FA patients.


Subject(s)
Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/immunology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , Cytokines/biosynthesis , Fanconi Anemia/genetics , Fanconi Anemia/immunology , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Forkhead Transcription Factors/metabolism , Gene Expression , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Transplantation Chimera
12.
Haematologica ; 98(5): 705-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23585528

ABSTRACT

Progressive bone marrow failure starting in the first decade of life is one of the main characteristics of Fanconi anemia. Along with the bone marrow failure, this pathology is characterized by congenital malformations, endocrine dysfunction and an extraordinary predisposition to develop cancer. The fact that hematopoietic progenitor cells from subjects with Fanconi anemia are sensitive to both DNA-interstrand crosslinking agents and inflammatory cytokines, which are aberrantly overproduced in these patients, has led to different explanations for the causes of the bone marrow failure. We analyzed STAT1 expression in lymphoblastoid cell lines derived from patients with Fanconi anemia group A and correlated this with aspects of the Fanconi anemia phenotype such as sensitivity to genotoxic agents or to inhibitory cytokines. We provide evidence of overexpression of STAT1 in FANCA-deficient cells which has both transcriptional and post-translational components, and is related to the constitutive activation of ERK in Fanconi anemia group A cells, since it can be reverted by treatment with U0126. STAT1 phosphorylation was not defective in the lymphoblasts, so these cells accumulated higher levels of active STAT1 in response to interferon gamma, probably in relation to their greater sensitivity to this cytokine. On the other hand, inhibition of STAT1 by genetic or chemical means reverted the hypersensitivity of Fanconi anemia group A lymphoblasts to DNA interstrand crosslinking agents. Our data provide an explanation for the mixed sensitivity of Fanconi anemia group A cells to both genotoxic stress and inflammatory cytokines and indicate new targets for the treatment of bone marrow failure in these patients.


Subject(s)
Cross-Linking Reagents/toxicity , DNA Damage/drug effects , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Lymphoid Progenitor Cells/drug effects , Lymphoid Progenitor Cells/metabolism , STAT1 Transcription Factor/genetics , Cell Line , Drug Resistance/genetics , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Gene Expression , Humans , Interferon-gamma/pharmacology , Phosphorylation/drug effects , RNA Processing, Post-Transcriptional , STAT1 Transcription Factor/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
13.
Antioxid Redox Signal ; 17(8): 1083-98, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22482891

ABSTRACT

AIMS: Insulin resistance is a hallmark of obesity and type 2 diabetes. Reactive oxygen species (ROS) have been proposed to play a causal role in insulin resistance. However, evidence linking ROS to insulin resistance in disease settings has been scant. Since both oxidative stress and diabetes have been observed in patients with the Fanconi anemia (FA), we sought to investigate the link between ROS and insulin resistance in this unique disease model. RESULTS: Mice deficient for the Fanconi anemia complementation group A (Fanca) or Fanconi anemia complementation group C (Fancc) gene seem to be diabetes-prone, as manifested by significant hyperglycemia and hyperinsulinemia, and rapid weight gain when fed with a high-fat diet. These phenotypic features of insulin resistance are characterized by two critical events in insulin signaling: a reduction in tyrosine phosphorylation of the insulin receptor (IR) and an increase in inhibitory serine phosphorylation of the IR substrate-1 in the liver, muscle, and fat tissues from the insulin-challenged FA mice. High levels of ROS, spontaneously accumulated or generated by tumor necrosis factor alpha in these insulin-sensitive tissues of FA mice, were shown to underlie the FA insulin resistance. Treatment of FA mice with the natural anti-oxidant Quercetin restores IR signaling and ameliorates the diabetes- and obesity-prone phenotypes. Finally, pairwise screen identifies protein-tyrosine phosphatase (PTP)-α and stress kinase double-stranded RNA-dependent protein kinase (PKR) that mediate the ROS effect on FA insulin resistance. INNOVATION: These findings establish a pathogenic and mechanistic link between ROS and insulin resistance in a unique human disease setting. CONCLUSION: ROS accumulation contributes to the insulin resistance in FA deficiency by targeting both PTP-α and PKR.


Subject(s)
Fanconi Anemia/metabolism , Insulin Resistance , Obesity/metabolism , Reactive Oxygen Species/metabolism , 3T3-L1 Cells , Animals , Cells, Cultured , Disease Models, Animal , 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 , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Oxidative Stress , Quercetin/administration & dosage , Quercetin/pharmacology , Receptor, Insulin/metabolism , Signal Transduction/drug effects
14.
Blood ; 119(18): 4162-73, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22427203

ABSTRACT

Salidroside is a phenylpropanoid glycoside isolated from the medicinal plant Rhodiola rosea, which has potent antioxidant properties. Here we show that salidroside prevented the loss of hematopoietic stem cells (HSCs) in mice under oxidative stress. Quiescent HSCs were recruited into cell cycling on in vivo challenge with oxidative stress, which was blocked by salidroside. Surprisingly, salidroside does not prevent the production of reactive oxygen species but reduces hydrogen peroxide-induced DNA-strand breaks in bone marrow cells enriched for HSCs. We tested whether salidroside enhances oxidative DNA damage repair in mice deficient for 5 DNA repair pathways known to be involved in oxidative DNA damage repair; we found that salidroside activated poly(ADP-ribose)polymerase-1 (PARP-1), a component of the base excision repair pathway, in mouse bone marrow HSCs as well as primary fibroblasts and human lymphoblasts. PARP-1 activation by salidroside protects quiescent HSCs from oxidative stress-induced cycling in native animals and self-renewal defect in transplanted recipients, which was abrogated by genetic ablation or pharmacologic inhibition of PARP-1. Together, these findings suggest that activation of PARP-1 by salidroside could affect the homeostasis and function of HSCs and contribute to the antioxidant effects of salidroside.


Subject(s)
Antioxidants/pharmacology , DNA Repair/drug effects , Glucosides/pharmacology , Hematopoietic Stem Cells/drug effects , Oxidative Stress/drug effects , Phenols/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Cycle/drug effects , DNA/drug effects , DNA Breaks, Double-Stranded , Drug Evaluation, Preclinical , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group C Protein/deficiency , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hydrogen Peroxide/toxicity , Mice , Mice, Inbred C57BL , Oxidants/toxicity , Plants, Medicinal/chemistry , Poly (ADP-Ribose) Polymerase-1 , Reactive Oxygen Species , Rhodiola/chemistry
15.
Blood ; 119(9): 1992-2002, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22234699

ABSTRACT

Fanconi anemia, complementation group C (FANCC)-deficient hematopoietic stem and progenitor cells are hypersensitive to a variety of inhibitory cytokines, one of which, TNFα, can induce BM failure and clonal evolution in Fancc-deficient mice. FANCC-deficient macrophages are also hypersensitive to TLR activation and produce TNFα in an unrestrained fashion. Reasoning that suppression of inhibitory cytokine production might enhance hematopoiesis, we screened small molecules using TLR agonist-stimulated FANCC- and Fanconi anemia, complementation group A (FANCA)-deficient macrophages containing an NF-κB/AP-1-responsive reporter gene (SEAP). Of the 75 small molecules screened, the p38 MAPK inhibitor BIRB 796 and dasatinib potently suppressed TLR8-dependent expression of the reporter gene. Fanconi anemia (FA) macrophages were hypersensitive to the TLR7/8 activator R848, overproducing SEAP and TNFα in response to all doses of the agonist. Low doses (50nM) of both agents inhibited p38 MAPK-dependent activation of MAPKAPK2 (MK2) and suppressed MK2-dependent TNFα production without substantially influencing TNFα gene transcription. Overproduction of TNFα by primary FA cells was likewise suppressed by these agents and involved inhibition of MK2 activation. Because MK2 is also known to influence production and/or sensitivity to 2 other suppressive factors (MIP-1α and IFNγ) to which FA hematopoietic progenitor cells are uniquely vulnerable, targeting of p38 MAPK in FA hematopoietic cells is a rational objective for preclinical evaluation.


Subject(s)
Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group C Protein/deficiency , Phagocytes/metabolism , Protein Kinase Inhibitors/pharmacology , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Dasatinib , Disease Models, Animal , Enzyme Activation/drug effects , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Humans , Macrophages/drug effects , Macrophages/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Naphthalenes/pharmacology , Phagocytes/drug effects , Phagocytes/enzymology , Phenotype , Proto-Oncogene Proteins c-jun/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA Processing, Post-Transcriptional/drug effects , Small Molecule Libraries , Thiazoles/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , src-Family Kinases/antagonists & inhibitors
16.
Cancer Lett ; 292(1): 73-9, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20034732

ABSTRACT

Genetic or epigenetic inactivation of the pathway formed by the Fanconi Anemia (FA) proteins occurs in several cancer types, including head and neck squamous cell carcinomas (HNSCC), rendering the affected tumors potentially hypersensitive to DNA crosslinking agents. However, the cytotoxicity of other commonly used cancer therapeutics in cells with FA pathway defects remains to be defined. Here, we focused on the effects of cisplatin and oxaliplatin in a panel of HNSCC and fibroblast cell lines. We found that FANCC- and FANCD2-mutant cells were unexpectedly more sensitive to platinum drugs than FANCA-mutant cells, and mono-ubiquitination of FANCD2, which is mediated by the FANCA and FANCC containing FA core complex was not required for platinum resistance. Interestingly, platinum hypersensitivity could be dissociated from mitomycin C hypersensitivity suggesting different underlying mechanisms. FANCD2 or RAD51 subnuclear foci were not useful as biomarkers of platinum hypersensitivity of FANCC/FANCD2-mutant cells. Our data add to an emerging body of evidence indicating that the FA pathway is not linear and that several protein subcomplexes with different functions exist. It will be important to establish biomarkers that can predict the sensitivity of tumors with specific FA defects to chemotherapeutic agents.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cisplatin/pharmacology , Fanconi Anemia Complementation Group Proteins/metabolism , Head and Neck Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Fibroblasts/drug effects , Head and Neck Neoplasms/genetics , Humans , Mutation , Oxaliplatin
17.
Biochem J ; 422(1): 161-70, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19473116

ABSTRACT

FA (Fanconi anaemia) is a hereditary disease characterized by congenital malformations, progressive bone marrow failure and an extraordinary elevated predisposition to develop cancer. In the present manuscript we describe an anomalous high level of the proinflammatory cytokine IL-1beta (interleukin-1beta) present in the serum of FA patients. The elevated levels of IL-1beta were completely reverted by transduction of a wild-type copy of the FancA cDNA into FA-A (FA group A) lymphocytes. Although the transcription factor NF-kappaB (nuclear factor-kappaB) is a well established regulator of IL-1beta expression, our experiments did not show any proof of elevated NF-kappaB activity in FA-A cells. However, we found that the overexpression of IL-1beta in FA-A cells is related to a constitutively activated PI3K (phosphoinositide 3-kinase)-Akt pathway in these cells. We provide evidence that the effect of Akt on IL-1beta activation is mediated by the inhibition of GSK3beta (glycogen synthase kinase 3beta). Finally, our data indicate that the levels of IL-1beta produced by FA-A lymphoblasts are enough to promote an activation of the cell cycle in primary glioblastoma progenitor cells. Together, these results demonstrate that the constitutive activation of the PI3K-Akt pathway in FA cells upregulates the expression of IL-1beta through an NF-kappaB-independent mechanism and that this overproduction activates the proliferation of tumour cells.


Subject(s)
Fanconi Anemia/enzymology , Fanconi Anemia/pathology , Interleukin-1beta/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Fanconi Anemia Complementation Group A Protein/deficiency , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Mutation/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology
18.
Cancer Res ; 68(23): 9693-702, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047147

ABSTRACT

Members of the Fanconi anemia (FA) protein family are involved in repair of genetic damage caused by DNA cross-linkers. It is not clear whether the FA proteins function in oxidative DNA damage and oncogenic stress response. Here, we report that deficiency in the Fanca gene in mice elicits a p53-dependent growth arrest and DNA damage response to oxidative DNA damage and oncogenic stress. Using a Fanca-/-Trp53-/- double knockout model and a functionally switchable p53 retrovirus, we define the kinetics, dependence, and persistence of p53-mediated response to oxidative and oncogenic stresses in Fanca-/- cells. Notably, oxidative stress induces persistent p53 response in Fanca-/- cells, likely due to accumulation of unrepaired DNA damage. On the other hand, whereas wild-type cells exhibit prolonged response to oncogene activation, the p53-activating signals induced by oncogenic ras are short-lived in Fanca-/- cells, suggesting that Fanca may be required for the cell to engage p53 during constitutive ras activation. We propose that the FA proteins protect cells from stress-induced proliferative arrest and tumor evolution by acting as a modulator of the signaling pathways that link FA to p53.


Subject(s)
DNA Damage , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/genetics , Tumor Suppressor Protein p53/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Division/drug effects , Cell Division/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , G2 Phase/drug effects , G2 Phase/genetics , Humans , Hydrogen Peroxide/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/pharmacology , Oxidative Stress/genetics , Signal Transduction , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
19.
Exp Hematol ; 36(9): 1084-90, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495331

ABSTRACT

Fanconi anemia (FA) is a heterogeneous inherited disorder characterized by a progressive bone marrow (BM) failure and susceptibility to myeloid leukemia. Genetic correction using gene-transfer technology is one potential therapy. A major hurdle in applying this technology in FA patients is the inability of granulocyte colony-stimulating factor (G-CSF) to mobilize sufficient numbers of hematopoietic stem (HSC)/progenitor cells (HPC) from the BM to the peripheral blood. Whether the low number of CD34(+) cells is a result of BM hypoplasia or an inability of G-CSF to adequately mobilize FA HSC/HPC remains incompletely understood. Here we use competitive repopulation of lethally irradiated primary and secondary recipients to show that in two murine models of FA, AMD3100 synergizes with G-CSF resulting in a mobilization of HSC, whereas G-CSF alone fails to mobilize stem cells even in the absence of hypoplasia.


Subject(s)
Bone Marrow Transplantation , Fanconi Anemia/surgery , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Peripheral Blood Stem Cell Transplantation , Animals , Benzylamines , Cells, Cultured/drug effects , Colony-Forming Units Assay , Cyclams , Drug Synergism , Drug Therapy, Combination , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Graft Survival , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera , Transplantation, Homologous
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL