Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Stem Cell Res ; 40: 101550, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31472450

RESUMO

Members of the Fanconi anemia (FA) protein family are involved in multiple cellular processes including response to DNA damage and oxidative stress. Here we show that a major FA protein, Fancd2, plays a role in mitochondrial biosynthesis through regulation of mitochondrial translation. Fancd2 interacts with Atad3 and Tufm, which are among the most frequently identified components of the mitochondrial nucleoid complex essential for mitochondrion biosynthesis. Deletion of Fancd2 in mouse hematopoietic stem and progenitor cells (HSPCs) leads to increase in mitochondrial number, and enzyme activity of mitochondrion-encoded respiratory complexes. Fancd2 deficiency increases mitochondrial protein synthesis and induces mitonuclear protein imbalance. Furthermore, Fancd2-deficient HSPCs show increased mitochondrial respiration and mitochondrial reactive oxygen species. By using a cell-free assay with mitochondria isolated from WT and Fancd2-KO HSPCs, we demonstrate that the increased mitochondrial protein synthesis observed in Fancd2-KO HSPCs was directly linked to augmented mitochondrial translation. Finally, Fancd2-deficient HSPCs are selectively sensitive to mitochondrial translation inhibition and depend on augmented mitochondrial translation for survival and proliferation. Collectively, these results suggest that Fancd2 restricts mitochondrial activity through regulation of mitochondrial translation, and that augmented mitochondrial translation and mitochondrial respiration may contribute to HSC defect and bone marrow failure in FA.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Anemia de Fanconi/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Mitocôndrias/metabolismo , Biossíntese de Proteínas , ATPases Associadas a Diversas Atividades Celulares/genética , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Anemia de Fanconi/genética , Anemia de Fanconi/fisiopatologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Técnicas de Inativação de Genes , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fator Tu de Elongação de Peptídeos/genética , Fator Tu de Elongação de Peptídeos/metabolismo , Ligação Proteica
2.
Int J Stem Cells ; 12(3): 457-462, 2019 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-31474030

RESUMO

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure and high risk of cancer particularly leukemia. Here we show that inactivation of the non-homologous end-joining (NHEJ) activity of DNA-PKcs prevented DNA damage-induced expansion of FA pre-leukemic hematopoietic stem cells (HSCs). Furthermore, we performed serial BM transplantation to demonstrate that the DNA damage-induced expanded FA HSC compartment contained pre-leukemic stem cells that required the NHEJ activity of DNA-PKcs to induce leukemia in the secondary recipients. These results suggest that NHEJ may collaborate with FA deficiency to promote DNA damage-induced expansion of pre-leukemic HSCs.

3.
Stem Cell Res Ther ; 10(1): 114, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30925933

RESUMO

BACKGROUND: Recent studies have shown that deficiency in the Fanconi anemia (FA) DNA repair pathway enhances the error-prone non-homologous end-joining (NHEJ) repair, leading to increased genomic instability, and that genetic or pharmacological inhibition of the NHEJ pathway could rescue the FA phenotype. METHODS: First, we exposed LSK cells from WT and Fanca-/- mice to DNA-PKcs inhibitor NU7026 or Ku70 knockdown to examine whether inhibition of NHEJ sensitizes Fanca-/- HSPCs to PARP inhibitor (PARPi)- or interstrand crosslinking (ICL)-induced cell death and genomic instability. We then generated DNA-PKcs3A/3AFanca-/- mice to investigate the effect of specific inactivation of NHEJ on fetal HSCs. Lastly, we used two p53 mutant models to test whether specific inactivation of the p53 function in apoptosis is sufficient to rescue embryonic lethality and fetal HSC depletion in Fanca-/- DNA-PKcs3A/3A mice. RESULTS: Inhibition of NHEJ sensitizes HSPCs from Fanca-/- mice to PARP inhibition- and ICL-induced cell death and genomic instability and further decreases Fanca-/- HSPC proliferation and hematopoietic repopulation in irradiated transplant recipients. Specific inactivation of NHEJ activity by the knockin DNA-PKcs3A/3A mutation in two FA mouse models, Fanca-/- and Fancc-/-, leads to embryonic lethality. DNA-PKcs3A/3A causes fetal HSC depletion in developing Fanca-/- embryos due to increased HSC apoptosis and cycling. Both p53-/- and a knockin p53515C mutation, which selectively impairs the p53 function in apoptosis, can rescue embryonic lethality and fetal HSC depletion in Fanca-/- DNA-PKcs3A/3A mice. CONCLUSION: These results demonstrate that the NHEJ pathway functions to maintain Fanconi anemia fetal HSCs.


Assuntos
Apoptose , Reparo do DNA por Junção de Extremidades , Anemia de Fanconi , Feto , Instabilidade Genômica , Células-Tronco Hematopoéticas , Animais , Modelos Animais de Doenças , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Feto/metabolismo , Feto/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Knockout
4.
Leukemia ; 33(3): 749-761, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30254339

RESUMO

Mobilization of hematopoietic stem cells (HSCs) from bone marrow (BM) to peripheral blood (PB) by cytokine granulocyte colony-stimulating factor (G-CSF) or the chemical antagonist of CXCR4, AMD3100, is important in the treatment of blood diseases. Due to clinical conditions of each application, there is a need for continued improvement of HSC mobilization regimens. Previous studies have shown that genetic ablation of the Rho GTPase Cdc42 in HSCs results in their mobilization without affecting survival. Here we rationally identified a Cdc42 activity-specific inhibitor (CASIN) that can bind to Cdc42 with submicromolar affinity and competitively interfere with guanine nucleotide exchange activity. CASIN inhibits intracellular Cdc42 activity specifically and transiently to induce murine hematopoietic stem/progenitor cell egress from the BM by suppressing actin polymerization, adhesion, and directional migration of stem/progenitor cells, conferring Cdc42 knockout phenotypes. We further show that, although, CASIN administration to mice mobilizes similar number of phenotypic HSCs as AMD3100, it produces HSCs with better long-term reconstitution potential than that by AMD3100. Our work validates a specific small molecule inhibitor for Cdc42, and demonstrates that signaling molecules downstream of cytokines and chemokines, such as Cdc42, constitute a useful target for long-term stem cell mobilization.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Animais , Benzilaminas , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Movimento Celular/efeitos dos fármacos , Ciclamos , Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
6.
Stem Cell Res Ther ; 9(1): 145, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29784053

RESUMO

Although p53 mutations are common in solid tumors, such mutations are found at a lower frequency in hematologic malignancies. In the genetic disorder Fanconi anemia (FA), p53 has been proposed as an important pathophysiological factor for two important hematologic hallmarks of the disease: bone marrow failure and leukemogenesis. Here we show that low levels of the p53 protein enhance the capacity of leukemic stem cells from FA patients to repopulate immunodeficient mice. Furthermore, boosting p53 protein levels with the use of the small molecule Nutlin-3 reduced leukemia burden in recipient mice. These results demonstrate that the level of p53 protein plays a crucial role in FA leukemogenesis.


Assuntos
Anemia de Fanconi/genética , Proteína Supressora de Tumor p53/genética , Proliferação de Células , Humanos
7.
Stem Cell Reports ; 10(2): 339-346, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29307578

RESUMO

Overactive p53 has been proposed as an important pathophysiological factor for bone marrow failure syndromes, including Fanconi anemia (FA). Here, we report a p53-dependent effect on hematopoietic stem and progenitor cell (HSPC) proliferation in mice deficient for the FA gene Fanca. Deletion of p53 in Fanca-/- mice leads to replicative exhaustion of the hematopoietic stem cell (HSC) in transplant recipients. Using Fanca-/- HSCs expressing the separation-of-function mutant p53515C transgene, which selectively impairs the p53 function in apoptosis but keeps its cell-cycle checkpoint activities intact, we show that the p53 cell-cycle function is specifically required for the regulation of Fanca-/- HSC proliferation. Our results demonstrate that p53 plays a compensatory role in preventing FA HSCs from replicative exhaustion and suggest a cautious approach to manipulating p53 signaling as a therapeutic utility in FA.


Assuntos
Anemia Aplástica/genética , Doenças da Medula Óssea/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Hemoglobinúria Paroxística/genética , Proteína Supressora de Tumor p53/genética , Anemia Aplástica/patologia , Animais , Doenças da Medula Óssea/patologia , Transtornos da Insuficiência da Medula Óssea , Pontos de Checagem do Ciclo Celular/genética , Proliferação de Células/genética , Anemia de Fanconi/patologia , Regulação da Expressão Gênica no Desenvolvimento , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/patologia , Humanos , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Transgenes/genética
8.
Bio Protoc ; 8(9): e2824, 2018 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-34286035

RESUMO

Bone Marrow Hematopoietic Stem Cells (HSCs) require bone marrow microenvironment for their maintenance and proliferation. Culture of Bone Marrow Mesenchymal Stromal Cells (MSCs) provides appropriate environmental signals for HSCs survival in vitro. Here, we provide a detailed protocol that describes culture conditions for MSCs, flow cytometric isolation of HSCs from mouse bone marrow, and perform co-culture of MSCs and HSCs known as Cobblestone area-forming cell (CAFC) assay. Altogether, CAFC assays can be used as a high-throughput in vitro screening model where efforts are made to understand and develop therapies for complex bone marrow diseases. This protocol needs 3 to 4 weeks starting from culturing MSCs, isolating LSK cells (HSCs), and to performing limited dilution CAFC assay.

9.
Sci Rep ; 7: 45626, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28378742

RESUMO

Fancd2 is a component of the Fanconi anemia (FA) DNA repair pathway, which is frequently found defective in human cancers. The full repertoire of Fancd2 functions in normal development and tumorigenesis remains to be determined. Here we developed a Flag- and hemagglutinin-tagged Fancd2 knock-in mouse strain that allowed a high throughput mass spectrometry approach to search for Fancd2-binding proteins in different mouse organs. In addition to DNA repair partners, we observed that many Fancd2-interacting proteins are mitochondrion-specific. Fancd2 localizes in the mitochondrion and associates with the nucleoid complex components Atad3 and Tufm. The Atad3-Tufm complex is disrupted in Fancd2-/- mice and those deficient for the FA core component Fanca. Fancd2 mitochondrial localization requires Atad3. Collectively, these findings provide evidence for Fancd2 as a crucial regulator of mitochondrion biosynthesis, and of a molecular link between FA and mitochondrial homeostasis.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Fator Tu de Elongação de Peptídeos/metabolismo , Mapas de Interação de Proteínas , Animais , Técnicas de Introdução de Genes , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica
10.
Stem Cell Reports ; 8(5): 1242-1255, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28416286

RESUMO

Hematopoietic stem cell (HSC) defects can cause repopulating impairment leading to hematologic diseases. To target HSC deficiency in a disease setting, we exploited the repopulating defect of Fanconi anemia (FA) HSCs to conduct an in vivo short hairpin RNA (shRNA) screen. We exposed Fancd2-/- HSCs to a lentiviral shRNA library targeting 947 genes. We found enrichment of shRNAs targeting genes involved in the PPARγ pathway that has not been linked to HSC homeostasis. PPARγ inhibition by shRNA or chemical compounds significantly improves the repopulating ability of Fancd2-/- HSCs. Conversely, activation of PPARγ in wild-type HSCs impaired hematopoietic repopulation. In mouse HSCs and patient-derived lymphoblasts, PPARγ activation is manifested in upregulating the p53 target p21. PPARγ and co-activators are upregulated in total bone marrow and stem/progenitor cells from FA patients. Collectively, this work illustrates the utility of RNAi technology coupled with HSC transplantation for the discovery of novel genes and pathways involved in stress hematopoiesis.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Células-Tronco Hematopoéticas/metabolismo , Homeostase , PPAR gama/metabolismo , Animais , Benzamidas/farmacologia , Células Cultivadas , Cromanos/farmacologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Hematopoese , Humanos , Camundongos , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Piridinas/farmacologia , RNA Interferente Pequeno/genética , Tiazolidinedionas/farmacologia , Troglitazona , Proteína Supressora de Tumor p53/metabolismo
12.
Cell Rep ; 17(4): 1141-1157, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760317

RESUMO

Precise epigenetic regulation of the sex chromosomes is vital for the male germline. Here, we analyze meiosis in eight mouse models deficient for various DNA damage response (DDR) factors, including Fanconi anemia (FA) proteins. We reveal a network of FA and DDR proteins in which FA core factors FANCA, FANCB, and FANCC are essential for FANCD2 foci formation, whereas BRCA1 (FANCS), MDC1, and RNF8 are required for BRCA2 (FANCD1) and SLX4 (FANCP) accumulation on the sex chromosomes during meiosis. In addition, FA proteins modulate distinct histone marks on the sex chromosomes: FA core proteins and FANCD2 regulate H3K9 methylation, while FANCD2 and RNF8 function together to regulate H3K4 methylation independently of FA core proteins. Our data suggest that RNF8 integrates the FA-BRCA pathway. Taken together, our study reveals distinct functions for FA proteins and illuminates the male sex chromosomes as a model to dissect the function of the FA-BRCA pathway.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Histonas/metabolismo , Meiose , Processamento de Proteína Pós-Traducional , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular , Quebras de DNA de Cadeia Dupla , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisina/metabolismo , Masculino , Metilação , Camundongos , Rad51 Recombinase/metabolismo , Recombinação Genética/genética , Cromossomos Sexuais/metabolismo , Fatores de Tempo
13.
Oncotarget ; 7(37): 60005-60020, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27507053

RESUMO

The Fanconi anemia (FA) pathway is involved in DNA damage and other cellular stress responses. We have investigated the role of the FA pathway in oncogenic stress response by employing an in vivo stress-response model expressing the Gadd45ß-luciferase transgene. Using two inducible models of oncogenic activation (LSL-K-rasG12D and MycER), we show that hematopoietic stem and progenitor cells (HSPCs) from mice deficient for the FA core complex components Fanca or Fancc exhibit aberrant short-lived response to oncogenic insults. Mechanistic studies reveal that FA deficiency in HSPCs impairs oncogenic stress-induced G1 cell-cycle checkpoint, resulting from a compromised K-rasG12D-induced arginine methylation of p53 mediated by the protein arginine methyltransferase 5 (PRMT5). Furthermore, forced expression of PRMT5 in HSPCs from LSL-K-rasG12D/CreER-Fanca-/- mice prolongs oncogenic response and delays leukemia development in recipient mice. Our study defines an arginine methylation-dependent FA-p53 interplay that controls oncogenic stress response.


Assuntos
Antígenos de Diferenciação/metabolismo , Anemia de Fanconi/genética , Células-Tronco Hematopoéticas/fisiologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Antígenos de Diferenciação/genética , Arginina/metabolismo , Carcinogênese/genética , Ciclo Celular/genética , Dano ao DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Humanos , Luciferases/genética , Metilação , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
14.
Sci Rep ; 6: 22167, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26916217

RESUMO

The prominent role of Fanconi anemia (FA) proteins involves homologous recombination (HR) repair. Poly[ADP-ribose] polymerase1 (PARP1) functions in multiple cellular processes including DNA repair and PARP inhibition is an emerging targeted therapy for cancer patients deficient in HR. Here we show that PARP1 activation in hematopoietic stem and progenitor cells (HSPCs) in response to genotoxic or oxidative stress attenuates HSPC exhaustion. Mechanistically, PARP1 controls the balance between HR and non-homologous end joining (NHEJ) in double strand break (DSB) repair by preventing excessive NHEJ. Disruption of the FA core complex skews PARP1 function in DSB repair and led to hyper-active NHEJ in Fanca(-/-) or Fancc(-/-) HSPCs. Re-expression of PARP1 rescues the hyper-active NHEJ phenotype in Brca1(-/-)Parp1(-/-) but less effective in Fanca(-/-)Parp1(-/-) cells. Inhibition of NHEJ prevents myeloid/erythroid pathologies associated with synthetic lethality. Our results suggest that hyper-active NHEJ may select for "synthetic lethality" resistant and pathological HSPCs.


Assuntos
Reparo do DNA por Junção de Extremidades/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células-Tronco Hematopoéticas/citologia , Recombinação Homóloga/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Mutações Sintéticas Letais/genética , Animais , Proteína BRCA1 , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Proteínas Supressoras de Tumor/genética
15.
J Immunol ; 196(7): 2986-94, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26895835

RESUMO

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.


Assuntos
Células Produtoras de Anticorpos/citologia , Células Produtoras de Anticorpos/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Via de Sinalização Wnt , Animais , Células Produtoras de Anticorpos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica , Switching de Imunoglobulina/genética , Switching de Imunoglobulina/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Camundongos Knockout , Transcriptoma , Proteínas Wnt/metabolismo
16.
Stem Cells ; 34(4): 960-71, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26676373

RESUMO

Fanconi anemia (FA) is an inherited bone marrow (BM) failure syndrome, presumably resulting from defects in hematopoietic stem cells (HSCs). Normal HSCs depend more on glycolysis than on oxidative phosphorylation (OXPHOS) for energy production. Here, we show that FA HSCs are more sensitive to the respiration inhibitor NaN3 treatment than to glycolytic inhibitor 2-deoxy-d-glucose (2-DG), indicating more dependence on OXPHOS. FA HSCs undergo glycolysis-to-OXPHOS switch in response to oxidative stress through a p53-dependent mechanism. Metabolic stresses induce upregulation of p53 metabolic targets in FA HSCs. Inactivation of p53 in FA HSCs prevents glycolysis-to-OXPHOS switch. Furthermore, p53-deficient FA HSCs are more sensitive to 2-DG-mediated metabolic stress. Finally, oxidative stress-induced glycolysis-to-OXPHOS switch is mediated by synthesis of cytochrome c oxidase 2 (SCO2). These findings demonstrate p53-mediated OXPHOS function as a compensatory alteration in FA HSCs to ensure a functional but mildly impaired energy metabolism and suggest a cautious approach to manipulating p53 signaling in FA.


Assuntos
Proteínas de Transporte/biossíntese , Anemia de Fanconi/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Mitocondriais/biossíntese , Proteína Supressora de Tumor p53/genética , Apoptose/efeitos dos fármacos , Proteínas de Transporte/genética , Desoxiglucose/administração & dosagem , Metabolismo Energético , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Glicólise/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Humanos , Proteínas Mitocondriais/genética , Chaperonas Moleculares , Fosforilação Oxidativa/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Azida Sódica/administração & dosagem , Proteína Supressora de Tumor p53/metabolismo
17.
Sci Rep ; 5: 18127, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26658157

RESUMO

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure, variable congenital malformations and a predisposition to malignancies. FANCB (also known as FAAP95), is the only X-linked FA gene discovered thus far. In the present study, we investigated hematopoiesis in adult Fancb deficient (Fancb(-/y)) mice and found that Fancb(-/y) mice have decreased hematopoietic stem cell (HSC) quiescence accompanied by reduced progenitor activity in vitro and reduced repopulating capacity in vivo. Like other FA mouse models previously reported, the hematopoietic system of Fancb(-/y) mice is hypersensitive to DNA cross-linking agent mitomycin C (MMC), which induces bone marrow failure in Fancb(-/y) mice. Furthermore, Fancb(-/y) BM exhibits slower recovery kinetics and less tolerance to myelotoxic stress induced by 5-fluorouracil than wild-type littermates. RNA-seq analysis reveals altered expression of genes involved in HSC function and cell cycle regulation in Fancb(-/y) HSC and progenitor cells. Thus, this Fancb(-/y) mouse model provides a novel approach for studying the critical role of the FA pathway not only in germ cell development but also in the maintenance of HSC function.


Assuntos
Modelos Animais de Doenças , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Antineoplásicos/farmacologia , Contagem de Células Sanguíneas , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Anemia de Fanconi/sangue , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Fluoruracila/farmacologia , Perfilação da Expressão Gênica/métodos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitomicina/farmacologia
18.
Hum Mol Genet ; 24(18): 5234-49, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26123487

RESUMO

Fanconi anemia (FA) is a recessive X-linked and autosomal genetic disease associated with bone marrow failure and increased cancer, as well as severe germline defects such as hypogonadism and germ cell depletion. Although deficiencies in FA factors are commonly associated with germ cell defects, it remains unknown whether the FA pathway is involved in unique epigenetic events in germ cells. In this study, we generated Fancb mutant mice, the first mouse model of X-linked FA, and identified a novel function of the FA pathway in epigenetic regulation during mammalian gametogenesis. Fancb mutant mice were infertile and exhibited primordial germ cell (PGC) defects during embryogenesis. Further, Fancb mutation resulted in the reduction of undifferentiated spermatogonia in spermatogenesis, suggesting that FANCB regulates the maintenance of undifferentiated spermatogonia. Additionally, based on functional studies, we dissected the pathway in which FANCB functions during meiosis. The localization of FANCB on sex chromosomes is dependent on MDC1, a binding partner of H2AX phosphorylated at serine 139 (γH2AX), which initiates chromosome-wide silencing. Also, FANCB is required for FANCD2 localization during meiosis, suggesting that the role of FANCB in the activation of the FA pathway is common to both meiosis and somatic DNA damage responses. H3K9me2, a silent epigenetic mark, was decreased on sex chromosomes, whereas H3K9me3 was increased on sex chromosomes in Fancb mutant spermatocytes. Taken together, these results indicate that FANCB functions at critical stages of germ cell development and reveal a novel function of the FA pathway in the regulation of H3K9 methylation in the germline.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Genes Essenciais , Células Germinativas/metabolismo , Histonas/metabolismo , Meiose/genética , Cromossomos Sexuais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Epigênese Genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Loci Gênicos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Metilação , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , Transporte Proteico , Recombinação Genética , Espermatogônias/citologia , Espermatogônias/metabolismo
19.
Stem Cells ; 33(11): 3382-96, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26212365

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Glicerofosfolipídeos/biossíntese , Células-Tronco Hematopoéticas/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia
20.
Genom Data ; 4: 148-149, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25984447

RESUMO

Functional maintenance of hematopoietic stem cells (HSCs) is constantly challenged by stresses like DNA damage and oxidative stress. Foxo factors, particularly Foxo3a, function to regulate the self-renewal of HSCs and contribute to the maintenance of the HSC pool during aging by providing resistance to oxidative stress. Fancd2-deficient mice had multiple hematopoietic defects, including HSC loss in early development and in response to cellular stresses including oxidative stress. The cellular mechanisms underlying HSC loss in Fancd2-deficient mice include abnormal cell cycle status, loss of quiescence, and compromised hematopoietic repopulating capacity of HSCs. To address on a genome wide level the genes and pathways that are impacted by deletion of the Fancd2 and Foxo3a, we performed microarray analysis on phenotypic HSCs (Lin-ckit+Sca-1+CD150+CD48-) from Fancd2 single knockout, Foxo3a single knockout and Fancd2-/-Foxo3a-/- double-knockout (dKO) mice. Here we provide detailed methods and analysis on these microarray data which has been deposited in Gene Expression Omnibus (GEO): GSE64215.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA