Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Genome Res ; 21(3): 477-86, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21173034

RESUMO

We have streamlined the process of transferring plasmids into any yeast strain library by developing a novel mating-based, high-throughput method called selective ploidy ablation (SPA). SPA uses a universal plasmid donor strain that contains conditional centromeres on every chromosome. The plasmid-bearing donor is mated to a recipient, followed by removal of all donor-strain chromosomes, producing a haploid strain containing the transferred plasmid. As proof of principle, we used SPA to transfer plasmids containing wild-type and mutant alleles of DNA topoisomerase I (TOP1) into the haploid yeast gene-disruption library. Overexpression of Top1 identified only one sensitive mutation, rpa34, while overexpression of top1-T(722)A allele, a camptothecin mimetic, identified 190 sensitive gene-disruption strains along with rpa34. In addition to known camptothecin-sensitive strains, this set contained mutations in genes involved in the Rpd3 histone deacetylase complex, the kinetochore, and vesicle trafficking. We further show that mutations in several ESCRT vesicle trafficking components increase Top1 levels, which is dependent on SUMO modification. These findings demonstrate the utility of the SPA technique to introduce plasmids into the haploid gene-disruption library to discover new interacting pathways.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , Redes Reguladoras de Genes , Ensaios de Triagem em Larga Escala/métodos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alelos , Camptotecina/farmacologia , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , DNA Topoisomerases Tipo I/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Expressão Gênica , Biblioteca Genômica , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Mutação , Plasmídeos/genética , Ploidias , Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transformação Genética
2.
Blood ; 116(16): 2915-20, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20606166

RESUMO

Fanconi anemia (FA) is an inherited chromosomal instability syndrome characterized by bone marrow failure, myelodysplasia (MDS), and acute myeloid leukemia (AML). Eight FA proteins associate in a nuclear core complex to monoubiquitinate FANCD2/FANCI in response to DNA damage. Additional functions have been described for some of the core complex proteins; however, in vivo genetic proof has been lacking. Here we show that double-mutant Fancc(-/-);Fancg(-/-) mice develop spontaneous hematologic sequelae including bone marrow failure, AML, MDS and complex random chromosomal abnormalities that the single-mutant mice do not. This genetic model provides evidence for unique core complex protein function independent of their ability to monoubiquitinate FANCD2/FANCI. Importantly, this model closely recapitulates the phenotypes found in FA patients and may be useful as a preclinical platform to evaluate the molecular pathogenesis of spontaneous bone marrow failure, MDS and AML in FA.


Assuntos
Medula Óssea/fisiopatologia , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Leucemia Mieloide Aguda/genética , Mutação , Síndromes Mielodisplásicas/genética , Animais , Aberrações Cromossômicas , Camundongos , Camundongos Endogâmicos C57BL
3.
Genetics ; 180(4): 1799-808, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18832360

RESUMO

We have created a resource to rapidly map genetic traits to specific chromosomes in yeast. This mapping is done using a set of 16 yeast strains each containing a different chromosome with a conditionally functional centromere. Conditional centromere function is achieved by integration of a GAL1 promoter in cis to centromere sequences. We show that the 16 yeast chromosomes can be individually lost in diploid strains, which become hemizygous for the destabilized chromosome. Interestingly, most 2n - 1 strains endoduplicate and become 2n. We also demonstrate how chromosome loss in this set of strains can be used to map both recessive and dominant markers to specific chromosomes. In addition, we show that this method can be used to rapidly validate gene assignments from screens of strain libraries such as the yeast gene disruption collection.


Assuntos
Cromossomos Fúngicos/genética , Saccharomyces cerevisiae/genética , Mapeamento Cromossômico , Diploide , Perda de Heterozigosidade , Meiose , Modelos Genéticos , Fenótipo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
4.
Blood ; 112(12): 4458-65, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18684868

RESUMO

Fanconi anemia (FA) is a complex genetic disorder characterized by congenital abnormalities, bone marrow failure, and myeloid malignancies. Identification of 13 FA genes has been instrumental to explore gene transfer technologies aimed at correction of autologous FA-deficient stem cells. To date, 3 human FA stem cell gene therapy trials with standard 4-day transduction protocols using gammaretroviral vectors failed to provide clinical benefit. In addition, 2- to 4 day ex vivo manipulation of bone marrow from mice containing a disruption of the homologue of human FANCC (Fancc) results in a time-dependent increase in apoptosis and a risk for malignant transformation of hematopoietic cells. Here, we show that a 14-hour transduction period allows a foamyviral vector construct expressing the human FANCC cDNA to efficiently transduce murine FA stem cells with 1 to 2 proviral integrations per genome. Functionally, the repopulating activity of Fancc(-/-) stem cells from reconstituted mice expressing the recombinant FANCC transgene was comparable with wild-type controls. Collectively, these data provide evidence that short-term transduction of c-kit(+) cells with a foamyviral vector is sufficient for functional correction of a stem cell phenotype in a murine FA model. These data could have implications for future gene therapy trials for FA patients.


Assuntos
Proliferação de Células , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Células-Tronco Hematopoéticas/fisiologia , Spumavirus/genética , Transdução Genética/métodos , Animais , Células Cultivadas , Ritmo Circadiano , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Anemia de Fanconi/veterinária , Proteína do Grupo de Complementação C da Anemia de Fanconi/metabolismo , Terapia Genética , Vetores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/genética , Fatores de Tempo
5.
Exp Hematol ; 36(9): 1084-90, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18495331

RESUMO

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.


Assuntos
Transplante de Medula Óssea , Anemia de Fanconi/cirurgia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/farmacologia , Transplante de Células-Tronco de Sangue Periférico , Animais , Benzilaminas , Células Cultivadas/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Ciclamos , Sinergismo Farmacológico , Quimioterapia Combinada , Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quimera por Radiação , Transplante Homólogo
6.
Blood ; 108(13): 4283-7, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16946306

RESUMO

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


Assuntos
Antivirais/farmacologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Terapia Genética , Interferon gama/farmacologia , Células Progenitoras Mieloides/transplante , Animais , Anemia de Fanconi/genética , Terapia Genética/métodos , Facilitação Imunológica de Enxerto/métodos , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Camundongos , Camundongos Knockout , Transdução Genética/métodos , Transplante Autólogo , Transplante Isogênico
7.
Carcinogenesis ; 27(3): 446-53, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16258176

RESUMO

Mitomycin C (MMC) induces various types of DNA damages that cause significant cytotoxicity to cells. Accordingly, repair of MMC-induced damages involves multiple repair pathways such as nucleotide excision repair, homologous recombination repair and translesion bypass repair pathways. Nonetheless, repair of the MMC-induced DNA damages in mammals have not been fully delineated. In this study, we investigated potential roles for Xeroderma pigmentosum (XP) proteins in the repair of MMC-induced DNA damages using an assay that detects the ssDNA patches generated following treatment with MMC or 8'-methoxy-psoralen (8-MOP) + UVA (ultraviolet light A). Human wild-type cells formed distinctive ssDNA foci following treatment with MMC or 8-MOP + UVA, but not with those inducing alkylation damage, oxidative damage or strand-break damage, suggesting that the foci represent ssDNA patches formed during the crosslink repair. In contrast to wild-type cells, mutant defective in XPE orXPG did not form the ssDNA foci following MMC treatment, while XPF mutant cells showed a significantly delayed response in forming the foci. A positive role for XPG in the repair of MMC-induced DNA damages was further supported by observations that cells treated with MMC induced a tight association of XPG with chromatin, and a targeted inhibition of XPG abolished MMC-induced ssDNA foci formation, rendering cells hypersensitive to MMC. Together, our results suggest that XPG along with XPE and XPF play unique role(s) in the repair of MMC-induced DNA damages.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Dano ao DNA , Reparo do DNA , Mitomicina/farmacologia , Técnicas de Cultura de Células , Cromatina/metabolismo , Proteínas de Ligação a DNA/fisiologia , Endonucleases/fisiologia , Fibroblastos , Humanos , Proteínas Nucleares/fisiologia , Estresse Oxidativo , Fatores de Transcrição/fisiologia
8.
Blood ; 105(9): 3465-71, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15644418

RESUMO

Current strategies for genetic therapy using Moloney retroviruses require ex vivo manipulation of hematopoietic cells to facilitate stable integration of the transgene. While many studies have evaluated the impact of ex vivo culture on normal murine and human stem/progenitor cells, the cellular consequences of ex vivo manipulation of stem cells with intrinsic defects in genome stability are incompletely understood. Here we show that ex vivo culture of Fancc(-/-) bone marrow cells results in a time-dependent increase in apoptosis of primitive Fancc(-/-) progenitor cells in conditions that promote the proliferation of wild-type stem/progenitor cells. Further, recipients reconstituted with the surviving Fancc(-/-) cells have a high incidence of cytogenetic abnormalities and myeloid malignancies that are associated with an acquired resistance to tumor necrosis factor alpha (TNF-alpha). Collectively, these data indicate that the intrinsic defects in the genomic stability of Fancc(-/-) stem/progenitor cells provide a selective pressure for cells that are resistant to apoptosis and have a propensity for the evolution to clonal hematopoiesis and malignancy. These studies could have implications for the design of genetic therapies for treatment of Fanconi anemia and potentially other genetic diseases with intrinsic defects in genome stability.


Assuntos
Apoptose , Aberrações Cromossômicas , Proteínas de Ligação a DNA/deficiência , Neoplasias Hematológicas/etiologia , Células-Tronco Hematopoéticas/patologia , Proteínas Nucleares/deficiência , Animais , Células da Medula Óssea/patologia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Células Clonais , Proteínas de Ligação a DNA/genética , Anemia de Fanconi/patologia , Anemia de Fanconi/terapia , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Risco , Fator de Necrose Tumoral alfa/farmacologia
9.
J Biol Chem ; 279(49): 50986-93, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15377654

RESUMO

Fanconi anemia (FA) is a complex, heterogeneous genetic disorder composed of at least 11 complementation groups. The FA proteins have recently been found to functionally interact with the cell cycle regulatory proteins ATM and BRCA1; however, the function of the FA proteins in cell cycle control remains incompletely understood. Here we show that the Fanconi anemia complementation group C protein (Fancc) is necessary for proper function of the DNA damage-induced G2/M checkpoint in vitro and in vivo. Despite apparently normal induction of the G2/M checkpoint after ionizing radiation, murine and human cells lacking functional FANCC did not maintain the G2 checkpoint as compared with wild-type cells. The increased rate of mitotic entry seen in Fancc-/-mouse embryo fibroblasts correlated with decreased inhibitory phosphorylation of cdc2 kinase on tyrosine 15. An increased inability to maintain the DNA damage-induced G2 checkpoint was observed in Fancc -/-; Trp53 -/-cells compared with Fancc -/-cells, indicating that Fancc and p53 cooperated to maintain the G2 checkpoint. In contrast, genetic disruption of both Fancc and Atm did not cooperate in the G2 checkpoint. These data indicate that Fancc and p53 in separate pathways converge to regulate the G2 checkpoint. Finally, fibroblasts lacking FANCD2 were found to have a G2 checkpoint phenotype similar to FANCC-deficient cells, indicating that FANCD2, which is activated by the FA complex, was also required to maintain the G2 checkpoint. Because a proper checkpoint function is critical for the maintenance of genomic stability and is intricately related to the function and integrity of the DNA repair process, these data have implications in understanding both the function of FA proteins and the mechanism of genomic instability in FA.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Dano ao DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Alelos , Animais , Bromodesoxiuridina/farmacologia , Proteína Quinase CDC2/metabolismo , Divisão Celular , Linhagem Celular , Células Cultivadas , Corantes/farmacologia , DNA/metabolismo , Reparo do DNA , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Fase G2 , Histonas/química , Humanos , Immunoblotting , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mitose , Mutação , Fosforilação , Ligação Proteica , Radiação Ionizante , Fatores de Tempo , Transgenes , Tirosina/química
10.
J Biol Chem ; 279(29): 30053-9, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15138265

RESUMO

Fanconi anemia (FANC) is a heterogeneous genetic disorder characterized by a hypersensitivity to DNA-damaging agents, chromosomal instability, and defective DNA repair. Eight FANC genes have been identified so far, and five of them (FANCA, -C, -E, -F, and -G) assemble in a multinuclear complex and function at least in part in a complex to activate FANCD2 by monoubiquitination. Here we show that FANCA and FANCG are redox-sensitive proteins that are multimerized and/or form a nuclear complex in response to oxidative stress/damage. Both FANCA and FANCG proteins exist as monomers under non-oxidizing conditions, whereas they become multimers following H2O2 treatment. Treatment of cells with oxidizing agent not only triggers the multimeric complex of FANCA and FANCG in vivo but also induces the interaction between FANCA and FANCG. N-Ethylmaleimide treatment abolishes multimerization and interaction of FANCA and FANCG in vitro. Taken together, our results lead us to conclude that FANCA and FANCG uniquely respond to oxidative damage by forming complex(es) via intermolecular disulfide linkage(s), which may be crucial in forming such complexes and in determining their function.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Anemia de Fanconi/metabolismo , Estresse Oxidativo , Proteínas/fisiologia , Animais , Western Blotting , Células COS , Clonagem Molecular , Dano ao DNA , Reparo do DNA , DNA Complementar/metabolismo , Dimerização , Dissulfetos , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Etilmaleimida/farmacologia , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/farmacologia , Mitomicina/farmacologia , Modelos Biológicos , Oxidantes/farmacologia , Oxirredução , Oxigênio/metabolismo , Testes de Precipitina , Estrutura Terciária de Proteína
11.
J Biol Chem ; 279(7): 6046-55, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14617623

RESUMO

Ku70-Ku80 complex is the regulatory subunit of DNA-dependent protein kinase (DNA-PK) and plays an essential role in double-strand break repair following ionizing radiation (IR). It preferentially interacts with chromosomal breaks and protects DNA ends from nuclease attack. Here we show evidence that cells defective in Ku80 exhibit a significantly slow S phase progression following DNA damage. IR-induced retardation in S phase progression in Ku80-/- cells was not due to the lack of DNA-PK kinase activity because both wild-type cells and DNA-PKcs-deficient cells showed no such symptom. Instead, proliferating cell nuclear antigen (PCNA) dissociated from chromosomes following IR in Ku80-deficient cells but not in wild-type or DNA-PKcs-deficient cells. Treatment of HeLa cells with IR induced colocalization of the Ku complex with PCNA on chromosomes. Together, these results suggest that binding of the Ku complex at chromosomal breaks may be necessary to maintain the sliding clamps (PCNA) on chromatin, which would allow cells to resume DNA replication without a major delay following IR.


Assuntos
Antígenos Nucleares/fisiologia , DNA Helicases , Replicação do DNA , Proteínas de Ligação a DNA/fisiologia , Animais , Antígenos Nucleares/biossíntese , Bromodesoxiuridina/farmacologia , Ciclo Celular , Núcleo Celular/metabolismo , Cromatina/química , Cromatina/metabolismo , Citosol/metabolismo , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/biossíntese , Dimerização , Relação Dose-Resposta à Radiação , Células HeLa , Humanos , Autoantígeno Ku , Camundongos , Microscopia Confocal , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Radiação Ionizante , Fatores de Tempo
12.
Blood ; 102(12): 4146-52, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12855557

RESUMO

Fanconi anemia (FA) is a recessive genomic instability syndrome characterized by developmental defects, progressive bone marrow failure, and cancer. FA is genetically heterogeneous, however; the proteins encoded by different FA loci interact functionally with each other and with the BRCA1, BRCA2, and ATM gene products. Although patients with FA are highly predisposed to the development of myeloid leukemia and solid tumors, the alterations in biochemical pathways responsible for the progression of tumorigenesis in these patients remain unknown. FA cells are hypersensitive to a range of genotoxic and cellular stresses that activate signaling pathways mediating apoptosis. Here we show that ionizing radiation (IR) induces modestly elevated levels of p53 in cells from FA type C (Fancc) mutant mice and that inactivation of Trp53 rescues tumor necrosis factor alpha-induced apoptosis in myeloid cells from Fancc-/- mice. Further, whereas Fancc-/- mice failed to form hematopoietic or solid malignancies, mice mutant at both Fancc and Trp53 developed tumors more rapidly than mice mutant at Trp53 alone. This shortened latency was associated with the appearance of tumor types that are found in patients with FA but not in mice mutant at Trp53 only. Collectively, these data demonstrate that p53 and Fancc interact functionally to regulate apoptosis and tumorigenesis in Fancc-deficient cells.


Assuntos
Apoptose , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Anemia de Fanconi/patologia , Neoplasias/etiologia , Proteínas Nucleares , Proteínas/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Embrião de Mamíferos/citologia , Anemia de Fanconi/complicações , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Fibroblastos/patologia , Predisposição Genética para Doença , Genótipo , Padrões de Herança , Camundongos , Camundongos Knockout , Camundongos Mutantes , Neoplasias/genética , Neoplasias/patologia , Proteínas/genética , Radiação Ionizante , Fator de Necrose Tumoral alfa/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/efeitos da radiação
13.
Blood ; 101(4): 1299-307, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393504

RESUMO

Fanconi anemia (FA) is a chromosomal instability disorder characterized by a progressive bone marrow (BM) failure and an increased incidence of myeloid leukemias. Children with FA are currently being enrolled in clinical trials to evaluate the safety of retroviral-mediated gene transfer. Previously, we used Fancc(-/-) mice to show that Fancc(-/-) hematopoietic stem cells (HSCs) have a profound defect in repopulating ability. Here, we examined whether retroviral-mediated gene transfer of recombinant Fancc (rFancc) would restore the repopulating ability of Fancc(-/-) HSC to wild-type levels. Fancc(-/-) HSCs transduced with a retrovirus encoding rFancc exhibited a repopulating ability that approached wild-type levels. Interestingly, approximately 30% of primary recipients (7 of 22) transplanted with uncorrected Fancc(-/-) cells developed a range of hematopoietic abnormalities including pancytopenia and BM hypoplasia similar to individuals with FA. Hematopoietic abnormalities were detected in only 1 of 22 mice transplanted with Fancc(-/-) cells transduced with a retrovirus encoding rFancc. Moreover, several mice with hematopoietic defects had progenitors that displayed a marked resistance to IFN-gamma, TNF-alpha, and MIP-1alpha compared to both Fancc(-/-) progenitors, which are uniquely hypersensitive to these cytokines, and wild-type progenitors. These data are analogous to studies using progenitors from patients with myelodysplasia and provide functional support for clonal evolution in these mice. Collectively, these data show that gene transfer can enhance HSC repopulating ability and suppresses the tendency for clonal evolution. These studies also reveal potential detrimental effects of ex vivo manipulation for untransduced Fancc(-/-) HSCs.


Assuntos
Proteínas de Ciclo Celular , Células Clonais , Proteínas de Ligação a DNA , Expressão Gênica , Células-Tronco Hematopoéticas/química , Células-Tronco Hematopoéticas/citologia , Proteínas Nucleares , Proteínas/genética , Retroviridae/genética , Transfecção , Animais , Western Blotting , Medula Óssea/patologia , Divisão Celular , Modelos Animais de Doenças , Anemia de Fanconi , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Hematopoese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Mielofibrose Primária/patologia , Proteínas Recombinantes , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA