Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 11(12)2020 12 21.
Article in English | MEDLINE | ID: mdl-33371494

ABSTRACT

Fanconi anemia (FA), a chromosomal instability syndrome, is caused by inherited pathogenic variants in any of 22 FANC genes, which cooperate in the FA/BRCA pathway. This pathway regulates the repair of DNA interstrand crosslinks (ICLs) through homologous recombination. In FA proper repair of ICLs is impaired and accumulation of toxic DNA double strand breaks occurs. To repair this type of DNA damage, FA cells activate alternative error-prone DNA repair pathways, which may lead to the formation of gross structural chromosome aberrations of which radial figures are the hallmark of FA, and their segregation during cell division are the origin of subsequent aberrations such as translocations, dicentrics and acentric fragments. The deficiency in DNA repair has pleiotropic consequences in the phenotype of patients with FA, including developmental alterations, bone marrow failure and an extreme risk to develop cancer. The mechanisms leading to the physical abnormalities during embryonic development have not been clearly elucidated, however FA has features of premature aging with chronic inflammation mediated by pro-inflammatory cytokines, which results in tissue attrition, selection of malignant clones and cancer onset. Moreover, chromosomal instability and cell death are not exclusive of the somatic compartment, they also affect germinal cells, as evidenced by the infertility observed in patients with FA.


Subject(s)
Chromosomal Instability , DNA Repair , Fanconi Anemia/genetics , Aging/genetics , BRCA1 Protein/physiology , BRCA2 Protein/physiology , Bone Marrow Failure Disorders/etiology , Cell Cycle , Chromatids/ultrastructure , Chromosome Aberrations , Chromosomes, Human/ultrastructure , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Fanconi Anemia/complications , Fanconi Anemia/diagnosis , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Humans , Infertility/genetics , Neoplastic Syndromes, Hereditary/genetics , Phenotype , Protein Processing, Post-Translational , Ubiquitination
2.
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
3.
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
4.
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
5.
Exp Hematol ; 44(5): 352-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26860989

ABSTRACT

Fanconi anemia (FA) is an inherited disorder of genomic instability associated with high risk of myelodysplasia and acute myeloid leukemia (AML). Young mice deficient in FA core complex genes do not naturally develop cancer, hampering preclinical studies on malignant hematopoiesis in FA. Here we describe that aging Fancc(-/-) mice are prone to genomically unstable AML and other hematologic neoplasms. We report that aneuploidy precedes malignant transformation during Fancc(-/-) hematopoiesis. Our observations reveal that Fancc(-/-) mice develop hematopoietic chromosomal instability followed by leukemia in an age-dependent manner, recapitulating the clinical phenotype of human FA and providing a proof of concept for future development of preclinical models of FA-associated leukemogenesis.


Subject(s)
Aging/genetics , Chromosomal Instability , Fanconi Anemia Complementation Group C Protein/genetics , Leukemia, Myeloid/genetics , Acute Disease , Age Factors , Aneuploidy , Animals , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Hematopoiesis/genetics , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid/metabolism , Mice, Inbred C57BL , Mice, Knockout
6.
J Immunol ; 196(7): 2986-94, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895835

ABSTRACT

Fanconi anemia (FA) is characterized by a progressive bone marrow failure and an increased incidence of cancer. FA patients have high susceptibility to immune-related complications such as infection and posttransplant graft-versus-host disease. In this study, we investigated the effect of FA deficiency in B cell function using the Fancc mouse model. Fancc(-/-) B cells show a specific defect in IgG2a switch and impaired Ab-secreting cell (ASC) differentiation. Global transcriptome analysis of naive B cells by mRNA sequencing demonstrates that FA deficiency deregulates a network of genes involved in immune function. Significantly, many genes implicated in Wnt signaling were aberrantly expressed in Fancc(-/-) B cells. Consistently, Fancc(-/-) B cells accumulate high levels of ß-catenin under both resting and stimulated conditions, suggesting hyperactive Wnt signaling. Using an in vivo Wnt GFP reporter assay, we verified the upregulation of Wnt signaling as a potential mechanism responsible for the impaired Fancc(-/-) B cell differentiation. Furthermore, we showed that Wnt signaling inhibits ASC differentiation possibly through repression of Blimp1 and that Fancc(-/-) B cells are hypersensitive to Wnt activation during ASC differentiation. Our findings identify Wnt signaling as a physiological regulator of ASC differentiation and establish a role for the Wnt pathway in normal B cell function and FA immune deficiency.


Subject(s)
Antibody-Producing Cells/cytology , Antibody-Producing Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Fanconi Anemia Complementation Group C Protein/deficiency , Wnt Signaling Pathway , Animals , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cluster Analysis , Gene Expression Profiling , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Knockout , Transcriptome , Wnt Proteins/metabolism
7.
Nucleic Acids Res ; 42(9): 5605-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24589582

ABSTRACT

Accumulating evidence suggests that dormant DNA replication origins play an important role in the recovery of stalled forks. However, their functional interactions with other fork recovery mechanisms have not been tested. We previously reported intrinsic activation of the Fanconi anemia (FA) pathway in a tumor-prone mouse model (Mcm4chaos3) with a 60% loss of dormant origins. To understand this further, we introduced a null allele of Fancc (Fancc-), encoding a member of the FA core complex, into the Mcm4chaos3 background. Primary embryonic fibroblasts double homozygous for Mcm4chaos3 and Fancc- (Mcm4chaos3/chaos3;Fancc-/-) showed significantly increased levels of markers of stalled/collapsed forks compared to either single homozygote. Interestingly, a loss of dormant origins also increased the number of sites in which replication was delayed until prophase, regardless of FA pathway activation. These replication defects coincided with substantially elevated levels of genome instability in Mcm4chaos3/chaos3;Fancc-/- cells, resulting in a high rate of perinatal lethality of Mcm4chaos3/chaos3;Fancc-/- mice and the accelerated tumorigenesis of surviving mice. Together, these findings uncover a specialized role of dormant origins in replication completion while also identifying important functional overlaps between dormant origins and the FA pathway in maintaining fork progression, genome stability, normal development and tumor suppression.


Subject(s)
DNA Replication , Fanconi Anemia Complementation Group C Protein/genetics , Genomic Instability , Animals , Cell Nucleus/genetics , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Micronucleus Tests , Minichromosome Maintenance Complex Component 4/genetics , Minichromosome Maintenance Complex Component 4/metabolism , S Phase Cell Cycle Checkpoints , Signal Transduction , Tumor Suppressor p53-Binding Protein 1
8.
Antioxid Redox Signal ; 21(12): 1675-92, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-24483844

ABSTRACT

AIMS: This study seeks at investigating the cause of hydrocephalus, and at identifying therapeutic targets for the prevention of hydrocephalus. RESULTS: In this study, we show that inactivation of the Foxo3a gene in two mouse models of Fanconi anemia (FA) leads to the development of hydrocephalus in late embryonic stage and after birth. More than 50% of Foxo3a(-/-) Fancc(-/-) or Foxo3a(-/-) Fancd2(-/-) mice die during embryonic development or within 6 months of life as a result of hydrocephalus characterized by cranial distortion, dilation of the ventricular system, reduced thickness of the cerebral cortex, and disorganization of the ependymal cilia and subcommissural organ. Combined deficiency of Foxo3a and Fancc or Fancd2 not only impairs the self-renewal capacity but also markedly increases the apoptosis of neural stem and progenitor cells (NSPCs), leading to defective neurogenesis. Increased accumulation of reactive oxygen species (ROS) and subsequently de-regulated mitosis and ultimately apoptosis in the neural stem or progenitor cells is identified as one of the potential mechanisms of congenital obstructive hydrocephalus. INNOVATION: The work unravels a two-tier protective mechanism for preventing oxidative stress-induced hydrocephalus. CONCLUSION: The deletion of Foxo3a in FA mice increased the accumulation of ROS and subsequently de-regulated mitosis and ultimately apoptosis in the NSPCs, leading to hydrocephalus development.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Forkhead Transcription Factors/metabolism , Hydrocephalus/metabolism , Oxidative Stress , Animals , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/deficiency , Forkhead Box Protein O3 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Hydrocephalus/genetics , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism
9.
Biochem J ; 448(1): 153-63, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22873408

ABSTRACT

FA (Fanconi anaemia) is a rare hereditary disorder characterized by congenital malformations, progressive bone marrow failure and an extraordinary predisposition to develop cancer. At present, 15 genes have been related to this condition and mutations of them have also been found in different types of cancer. Bone marrow failure threatens the life of FA patients during the first decade of their life, but the mechanisms underlying this process are not completely understood. In the present study we investigate a possible imbalance between the expression of pro- and anti-apoptotic proteins as a cause for the hypersensitivity of FANCC (FA, complementation group C)-deficient cells to genotoxic stress. We found a BIK (Bcl-2 interacting killer) over-expression in lymphoblastoid cell lines derived from FA-C patients when compared with their phenotypically corrected counterparts. This overexpression has a transcriptional basis since the regulatory region of the gene shows higher activity in FANCC-deficient cells. We demonstrate the involvement of BIK in the sensitivity of FA-C lymphoblasts to interstrand DNA cross-linking agents as it is induced by these drugs and interference of its expression in these cells preserves their viability and reduces apoptosis. We investigate the mechanism of BIK overexpression in FANCC-deficient cells by analysing the activity of many different signalling pathways in these cells. Finally, we provide evidence of a previously undescribed indirect epigenetic regulation of BIK in FA-C lymphoblasts mediated by ΔNp73, an isoform of p73 lacking its transactivation domain that activates BIK through a proximal element in its promoter.


Subject(s)
Apoptosis Regulatory Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Fanconi Anemia/pathology , Membrane Proteins/physiology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , 5' Untranslated Regions , Apoptosis/physiology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cell Line , Cell Line, Transformed , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA/drug effects , DNA Methylation , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Genes, Reporter , Humans , Lymphocytes/metabolism , Membrane Proteins/genetics , Mitochondrial Proteins , Mitomycin/pharmacology , Promoter Regions, Genetic , Protein Structure, Tertiary , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Transcriptional Activation , Tumor Protein p73 , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
10.
Cell Stem Cell ; 11(1): 36-49, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22683204

ABSTRACT

Fanconi anemia (FA) is an inherited DNA repair deficiency syndrome. FA patients undergo progressive bone marrow failure (BMF) during childhood, which frequently requires allogeneic hematopoietic stem cell transplantation. The pathogenesis of this BMF has been elusive to date. Here we found that FA patients exhibit a profound defect in hematopoietic stem and progenitor cells (HSPCs) that is present before the onset of clinical BMF. In response to replicative stress and unresolved DNA damage, p53 is hyperactivated in FA cells and triggers a late p21(Cdkn1a)-dependent G0/G1 cell-cycle arrest. Knockdown of p53 rescued the HSPC defects observed in several in vitro and in vivo models, including human FA or FA-like cells. Taken together, our results identify an exacerbated p53/p21 "physiological" response to cellular stress and DNA damage accumulation as a central mechanism for progressive HSPC elimination in FA patients, and have implications for clinical care.


Subject(s)
Bone Marrow/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Aging/pathology , Animals , Bone Marrow/metabolism , Child , Child, Preschool , Disease Models, Animal , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , G1 Phase Cell Cycle Checkpoints , G2 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , Gene Silencing , Hematopoietic Stem Cells/metabolism , Humans , Infant , Mice , Middle Aged , S Phase
11.
Blood ; 120(2): 323-34, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22653977

ABSTRACT

Bone marrow failure is a nearly universal complication of Fanconi anemia. The proteins encoded by FANC genes are involved in DNA damage responses through the formation of a multisubunit nuclear complex that facilitates the E3 ubiquitin ligase activity of FANCL. However, it is not known whether loss of E3 ubiquitin ligase activity accounts for the hematopoietic stem cell defects characteristic of Fanconi anemia. Here we provide evidence that FANCL increases the activity and expression of ß-catenin, a key pluripotency factor in hematopoietic stem cells. We show that FANCL ubiquitinates ß-catenin with atypical ubiquitin chain extension known to have nonproteolytic functions. Specifically, ß-catenin modified with lysine-11 ubiquitin chain extension efficiently activates a lymphocyte enhancer-binding factor-T cell factor reporter. We also show that FANCL-deficient cells display diminished capacity to activate ß-catenin leading to reduced transcription of Wnt-responsive targets c-Myc and Cyclin D1. Suppression of FANCL expression in normal human CD34(+) stem and progenitor cells results in fewer ß-catenin active cells and inhibits expansion of multilineage progenitors. Together, these results suggest that diminished Wnt/ß-catenin signaling may be an underlying molecular defect in FANCL-deficient hematopoietic stem cells leading to their accelerated loss.


Subject(s)
Fanconi Anemia Complementation Group L Protein/metabolism , beta Catenin/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cyclin D1/metabolism , Fanconi Anemia/etiology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , 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 L Protein/deficiency , Fanconi Anemia Complementation Group L Protein/genetics , Fetal Blood/cytology , Fetal Blood/metabolism , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Knockout , Models, Biological , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , TCF Transcription Factors/metabolism , Ubiquitination , beta Catenin/chemistry
12.
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
13.
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
14.
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
15.
J Leukoc Biol ; 91(2): 333-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22106009

ABSTRACT

FA is a genetic disorder characterized by BM failure, developmental defects, and cancer predisposition. Previous studies suggest that FA patients exhibit alterations in immunologic function. However, it is unclear whether the defects are immune cell-autonomous or secondary to leukopenia from evolving BM failure. Given the central role that macrophages have in the innate immune response, inflammation resolution, and antigen presentation for acquired immunity, we examined whether macrophages from Fancc-/- mice exhibit impaired function. Peritoneal inflammation induced by LPS or sodium periodate resulted in reduced monocyte/macrophage recruitment in Fancc-/- mice compared with WT controls. Fancc-/- mice also had decreased inflammatory monocytes mobilized into the peripheral blood after LPS treatment compared with controls. Furthermore, Fancc-/- peritoneal macrophages displayed cell-autonomous defects in function, including impaired adhesion to FN or endothelial cells, reduced chemoattractant-mediated migration, and decreased phagocytosis. Moreover, dysregulated F-actin rearrangement was detected in Fancc-/- macrophages after adhesion to FN, which was consistent with an observed reduction in RhoA-GTP levels. Importantly, these data suggest that impaired cytoskeletal rearrangements in Fancc-/- macrophages may be the common mechanism responsible for cell-autonomous defects detected in vitro, as well as altered monocyte/macrophage trafficking in vivo.


Subject(s)
Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia/pathology , Macrophages, Peritoneal/physiology , Actins/analysis , Animals , Cell Adhesion , Cells, Cultured/pathology , Chemotaxis/drug effects , Coculture Techniques , Cytoskeleton/chemistry , Cytoskeleton/ultrastructure , Endothelial Cells/cytology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Models, Animal , Phagocytosis/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Superoxides/metabolism , rho GTP-Binding Proteins/physiology , rhoA GTP-Binding Protein
16.
Blood ; 116(12): 2057-60, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20554974

ABSTRACT

Fancc suppresses cross-linker-induced genotoxicity, modulates growth-inhibitory cytokine responses, and modulates endotoxin responses. Although loss of the latter function is known to account for endotoxin-induced marrow failure in murine Fancc (mFancc)-deficient mice, some argue that cytokine and endotoxin hypersensitivities devolve simply from genomic instability. Seeking to resolve this question, we planned to ectopically express instructive human FANCC (hFANCC) mutants in murine Fancc-deficient hematopoietic stem cells. To first assure that hFANCC cDNA was competent in murine cells, we compared hFANCC and mFancc in complementation assays for cross-linking agent hypersensitivity and endotoxin hypersensitivity. We found that mFancc complemented murine Fancc-deficient cells in both assays, but that hFANCC fully suppressed only endotoxin hypersensitivity, not cross-linking agent hypersensitivity. These results support the notions that Fancc is multifunctional and that structural prerequisites for its genoprotective functions differ from those required to constrain endotoxin responses known to lead to marrow failure in Fancc-deficient mice.


Subject(s)
Fanconi Anemia Complementation Group C Protein/physiology , Hematopoietic Stem Cells/metabolism , Animals , Endotoxins/pharmacology , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Humans , Hypersensitivity, Immediate/chemically induced , Mice , Mice, Knockout , Transgenes
17.
Blood ; 114(26): 5290-8, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-19850743

ABSTRACT

Tumor necrosis factor alpha (TNF-alpha) production is abnormally high in Fanconi anemia (FA) cells and contributes to the hematopoietic defects seen in FA complementation group C-deficient (Fancc(-/-)) mice. Applying gene expression microarray and proteomic methods to studies on FANCC-deficient cells we found that genes encoding proteins directly involved in ubiquitinylation are overrepresented in the signature of FA bone marrow cells and that ubiquitinylation profiles of FA-C and complemented cells were substantially different. Finding that Toll-like receptor 8 (TLR8) was one of the proteins ubiquitinylated only in mutant cells, we confirmed that TLR8 (or a TLR8-associated protein) is ubiquitinylated in mutant FA-C cells and that TNF-alpha production in mutant cells depended upon TLR8 and the canonical downstream signaling intermediates interleukin 1 receptor-associated kinase (IRAK) and IkappaB kinase-alpha/beta. FANCC-deficient THP-1 cells and macrophages from Fancc(-/-) mice overexpressed TNF-alpha in response to TLR8 agonists but not other TLR agonists. Ectopically expressed FANCC point mutants were capable of fully complementing the mitomycin-C hypersensitivity phenotype of FA-C cells but did not suppress TNF-alpha overproduction. In conclusion, FANCC suppresses TNF-alpha production in mononuclear phagocytes by suppressing TLR8 activity and this particular function of FANCC is independent of its function in protecting the genome from cross-linking agents.


Subject(s)
Fanconi Anemia/metabolism , Signal Transduction/physiology , Toll-Like Receptor 8/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Gene Expression , Gene Expression Profiling , Humans , Immunoprecipitation , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitination/physiology , Up-Regulation
18.
Blood ; 113(21): 5111-20, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19270262

ABSTRACT

Ectopic delivery of HOXB4 elicits the expansion of engrafting hematopoietic stem cells (HSCs). We hypothesized that inhibition of tumor necrosis factor-alpha (TNF-alpha) signaling may be central to the self-renewal signature of HOXB4. Because HSCs derived from Fanconi anemia (FA) knockout mice are hypersensitive to TNF-alpha, we studied Fancc(-/-) HSCs to determine the physiologic effects of HOXB4 on TNF-alpha sensitivity and the relationship of these effects to the engraftment defect of FA HSCs. Overexpression of HOXB4 reversed the in vitro hypersensitivity to TNF-alpha of Fancc(-/-) HSCs and progenitors (P) and partially rescued the engraftment defect of these cells. Coexpression of HOXB4 and the correcting FA-C protein resulted in full correction compared with wild-type (WT) HSCs. Ectopic expression of HOXB4 resulted in a reduction in both apoptosis and reactive oxygen species in Fancc(-/-) but not WT HSC/P. HOXB4 overexpression was also associated with a significant reduction in surface expression of TNF-alpha receptors on Fancc(-/-) HSC/P. Finally, enhanced engraftment was seen even when HOXB4 was expressed in a time-limited fashion during in vivo reconstitution. Thus, the HOXB4 engraftment signature may be related to its effects on TNF-alpha signaling, and this pathway may be a molecular target for timed pharmacologic manipulation of HSC during reconstitution.


Subject(s)
Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/pharmacology , Transcription Factors/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Fanconi Anemia Complementation Group C Protein/deficiency , Graft Survival , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/administration & dosage , Mice , Mice, Knockout , Reactive Oxygen Species , Receptors, Tumor Necrosis Factor/analysis , Transcription Factors/administration & dosage
19.
Blood ; 113(12): 2655-60, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19168785

ABSTRACT

The underlying molecular mechanisms that promote bone marrow failure in Fanconi anemia are incompletely understood. Evidence suggests that enhanced apoptosis of hematopoietic precursors is a major contributing factor. Previously, enhanced apoptosis of Fanconi anemia type C-deficient (Fancc(-/-)) progenitors was shown to involve aberrant p38 MAPK activation. Given the importance of c-Jun N-terminal kinase (JNK) in the stress response, we tested whether enhanced apoptosis of Fancc(-/-) cells also involved altered JNK activation. In Fancc(-/-) murine embryonic fibroblasts, tumor necrosis factor alpha (TNF-alpha) induced elevated JNK activity. In addition, JNK inhibition protected Fancc(-/-) murine embryonic fibroblasts and c-kit(+) bone marrow cells from TNF-alpha-induced apoptosis. Importantly, hematopoietic progenitor assays demonstrated that JNK inhibition enhanced Fancc(-/-) colony formation in the presence of TNF-alpha. Competitive repopulation assays showed that Fancc(-/-) donor cells cultured with the JNK inhibitor had equivalent levels of donor chimerism compared with Fancc(-/-) donor cells cultured with vehicle control. In contrast, culturing Fancc(-/-) cells with a p38 MAPK inhibitor significantly increased repopulating ability, supporting an integral role of p38 MAPK in maintaining Fancc(-/-) hematopoietic stem cell function. Taken together, these data suggest that p38 MAPK, but not JNK, has a critical role in maintaining the engraftment of Fancc(-/-)-reconstituting cells under conditions of stress.


Subject(s)
Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia/enzymology , Mitogen-Activated Protein Kinase 8/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis/drug effects , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cells, Cultured/transplantation , Colony-Forming Units Assay , Enzyme Activation , Fanconi Anemia/pathology , Fibroblasts/drug effects , Fibroblasts/enzymology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Models, Animal , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , RNA Interference , RNA, Small Interfering/pharmacology , Radiation Chimera , Stress, Physiological , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
20.
Blood ; 112(5): 2062-70, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18550849

ABSTRACT

Fanconi anemia (FA) proteins are thought to play a role in chromosome stability and repair of DNA cross-links; however, these functions may not fully explain the developmental abnormalities and bone marrow failure that are characteristic of FA individuals. Here we associate the FA proteins with the Notch1 developmental pathway through a direct protein-protein interaction between the FA core complex and the hairy enhancer of split 1 (HES1). HES1 interaction with FA core complex members is dependent on a functional FA pathway. Cells depleted of HES1 exhibit an FA-like phenotype that includes cellular hypersensitivity to mitomycin C (MMC) and lack of FANCD2 monoubiquitination and foci formation. HES1 is also required for proper nuclear localization or stability of some members of the core complex. Our results suggest that HES1 is a novel interacting protein of the FA core complex.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Homeodomain Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell Line, Transformed , Drug Resistance/genetics , Drug Resistance/physiology , Fanconi Anemia/genetics , Fanconi Anemia/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 Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , HeLa Cells , Homeodomain Proteins/genetics , Humans , Mice , Mice, Knockout , Mitomycin/pharmacology , Multiprotein Complexes , Protein Binding , RNA, Small Interfering/genetics , Receptor, Notch1/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transcription Factor HES-1 , Two-Hybrid System Techniques , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...