Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 835, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149709

RESUMO

The majority of high-grade serous ovarian cancers (HGSCs) are deficient in homologous recombination (HR) DNA repair, most commonly due to mutations or hypermethylation of the BRCA1/2 genes. We aimed to discover how BRCA1/2 mutations shape the cellular phenotypes and spatial interactions of the tumor microenvironment. Using a highly multiplex immunofluorescence and image analysis we generate spatial proteomic data for 21 markers in 124,623 single cells from 112 tumor cores originating from 31 tumors with BRCA1/2 mutation (BRCA1/2mut), and from 13 tumors without alterations in HR genes. We identify a phenotypically distinct tumor microenvironment in the BRCA1/2mut tumors with evidence of increased immunosurveillance. Importantly, we report a prognostic role of a proliferative tumor-cell subpopulation, which associates with enhanced spatial tumor-immune interactions by CD8+ and CD4 + T-cells in the BRCA1/2mut tumors. The single-cell spatial landscapes indicate distinct patterns of spatial immunosurveillance with the potential to improve immunotherapeutic strategies and patient stratification in HGSC.


Assuntos
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Carcinoma Epitelial do Ovário/imunologia , Cistadenocarcinoma Seroso/imunologia , Neoplasias Ovarianas/imunologia , Microambiente Tumoral/imunologia , Proteína BRCA1/genética , Proteína BRCA2/genética , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Carcinoma Epitelial do Ovário/genética , Cistadenocarcinoma Seroso/genética , Feminino , Genes BRCA1 , Genes BRCA2 , Genótipo , Recombinação Homóloga , Humanos , Mutação , Neoplasias Ovarianas/genética , Prognóstico , Proteômica
2.
Ann Oncol ; 31(5): 590-598, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32245699

RESUMO

BACKGROUND: Little is known about mechanisms of resistance to poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPi) and platinum chemotherapy in patients with metastatic breast cancer and BRCA1/2 mutations. Further investigation of resistance in clinical cohorts may point to strategies to prevent or overcome treatment failure. PATIENTS AND METHODS: We obtained tumor biopsies from metastatic breast cancer patients with BRCA1/2 deficiency before and after acquired resistance to PARPi or platinum chemotherapy. Whole exome sequencing was carried out on each tumor, germline DNA, and circulating tumor DNA. Tumors underwent RNA sequencing, and immunohistochemical staining for RAD51 foci on tumor sections was carried out for functional assessment of intact homologous recombination (HR). RESULTS: Pre- and post-resistance tumor samples were sequenced from eight patients (four with BRCA1 and four with BRCA2 mutation; four treated with PARPi and four with platinum). Following disease progression on DNA-damaging therapy, four patients (50%) acquired at least one somatic reversion alteration likely to result in functional BRCA1/2 protein detected by tumor or circulating tumor DNA sequencing. Two patients with germline BRCA1 deficiency acquired genomic alterations anticipated to restore HR through increased DNA end resection: loss of TP53BP1 in one patient and amplification of MRE11A in another. RAD51 foci were acquired post-resistance in all patients with genomic reversion, consistent with reconstitution of HR. All patients whose tumors demonstrated RAD51 foci post-resistance were intrinsically resistant to subsequent lines of DNA-damaging therapy. CONCLUSIONS: Genomic reversion in BRCA1/2 was the most commonly observed mechanism of resistance, occurring in four of eight patients. Novel sequence alterations leading to increased DNA end resection were seen in two patients, and may be targetable for therapeutic benefit. The presence of RAD51 foci by immunohistochemistry was consistent with BRCA1/2 protein functional status from genomic data and predicted response to later DNA-damaging therapy, supporting RAD51 focus formation as a clinically useful biomarker.


Assuntos
Neoplasias da Mama , Neoplasias Ovarianas , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Platina/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico
4.
Carcinogenesis ; 22(12): 1939-46, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11751423

RESUMO

Fanconi anemia (FA) is a human autosomal disorder characterized by cancer susceptibility and cellular sensitivity to DNA crosslinking agents such as mitomycin C and diepoxybutane. Six FA genes have been cloned including a gene designated XRCC9 (for X-ray Repair Cross Complementing), isolated using a mitomycin C-hypersensitive Chinese hamster cell mutant termed UV40, and subsequently found to be identical to FANCG. A nuclear complex containing the FANCA, FANCC, FANCE, FANCF and FANCG proteins is needed for the activation of a sixth FA protein FANCD2. When monoubiquitinated, the FANCD2 protein co-localizes with the breast cancer susceptibility protein BRCA1 in DNA damage induced foci. In this study, we have assigned NM3, a nitrogen mustard-hypersensitive Chinese hamster mutant to the same genetic complementation group as UV40. NM3, like human FA cell lines (but unlike UV40) exhibits a normal spontaneous level of sister chromatid exchange. We show that both NM3 and UV40 are also hypersensitive to other DNA crosslinking agents (including diepoxybutane and chlorambucil) and to non-crosslinking DNA damaging agents (including bleomycin, streptonigrin and EMS), and that all these sensitivities are all corrected upon transfection of the human FANCG/XRCC9 cDNA. Using immunoblotting, NM3 and UV40 were found not to express the active monoubiquitinated isoform of the FANCD2 protein, although expression of the FANCD-L isoform was restored in the FANCG cDNA transformants, correlating with the correction of mutagen-sensitivity. These data indicate that cellular resistance to these DNA damaging agents requires FANCG and that the FA gene pathway, via its activation of FANCD2 and that protein's subsequent interaction with BRCA1, is involved in maintaining genomic stability in response not only to DNA interstrand crosslinks but also a range of other DNA damages including DNA strand breaks. NM3 and other "FA-like" Chinese hamster mutants should provide an important resource for the study of these processes in mammalian cells.


Assuntos
Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Mutagênicos/farmacologia , Mutação/genética , Proteínas Nucleares/metabolismo , Troca de Cromátide Irmã/genética , Ubiquitina/metabolismo , Animais , Bleomicina/farmacologia , Células CHO , Linhagem Celular , Cricetinae , Dano ao DNA/genética , Compostos de Epóxi/farmacologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Raios gama , Teste de Complementação Genética , Humanos , Células Híbridas , Mecloretamina/farmacologia , Mitomicina/farmacologia , Proteínas Nucleares/química , Troca de Cromátide Irmã/efeitos dos fármacos , Raios Ultravioleta
5.
Mol Genet Metab ; 74(4): 403-12, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11749045

RESUMO

Fanconi anemia (FA) is a human genetic disorder characterized by hypersensitivity to DNA crosslinking agents. Its cellular phenotypes include increased chromosome breakage and a marked cell-cycle delay with 4N DNA content after introduction of interstrand DNA crosslinks (ICL). To further understand the nature of this delay previously described as a G2/M arrest, we introduced ICL specifically during G2 and monitored the cells for passage into mitosis. Our results showed that, even at the highest doses, postreplication ICL produced neither G2/M arrest nor chromosome breakage in FA-A or FA-C cells. This suggests that, similar to wild-type cells, DNA replication is required to trigger both responses. Therefore, the 4N cell DNA content observed in FA cells after ICL treatment also represents incomplete DNA replication and arrest in late S phase. FA fibroblasts from complementation groups A and C were able to recover from the ICL-induced cell-cycle arrest, but took approximately 3 times longer than controls. These results indicate that the FA pathway is required for the efficient resolution of ICL-induced S-phase arrest.


Assuntos
Anemia de Fanconi/fisiopatologia , Fase S , Trioxsaleno/análogos & derivados , Linhagem Celular , Quebra Cromossômica , Reagentes de Ligações Cruzadas/farmacologia , DNA , Reparo do DNA , Anemia de Fanconi/genética , Fibroblastos , Quadruplex G , Fase G2/efeitos dos fármacos , Humanos , Mitose/efeitos dos fármacos , Fase S/efeitos dos fármacos , Trioxsaleno/farmacologia , Raios Ultravioleta
6.
Exp Hematol ; 29(12): 1448-55, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11750104

RESUMO

OBJECTIVE: Fanconi anemia (FA) is a human autosomal-recessive cancer susceptibility disorder characterized by multiple congenital abnormalities, progressive bone marrow failure, and cellular sensitivity to mitomycin C (MMC). FA has at least eight complementation groups (A, B, C, D1, D2, E, F, G), and six of the FA genes have been cloned. Several FA proteins, including FANCA, FANCC, FANCF, and FANCG, interact in a nuclear complex, and this complex is required for the activation (monoubiquitination) of the downstream FANCD2 protein. Activation of FANCD2 results in the assembly of FANCD2/BRCA1 foci. The aim of this study was to analyze the FA pathway in several FA patient-derived cell lines. MATERIALS AND METHODS: We generated an antibody to FANCF and analyzed FANCF expression in human lymphoblasts corresponding to all known FA subtypes. We systematically analyzed the FA pathway (FANCD2 monoubiquitination and assembly of FANCD2 nuclear foci) in patient-derived FA-F and FA-D1 cell lines. RESULTS: FANCF protein expression is normal in cells derived from all FA complementation groups except FA-F and does not vary during cell cycle progression. FANCF, but not FANCD2, is a component of the nuclear FA protein complex and appears to stabilize other subunits of the complex. FANCF is required for the monoubiquitination of the FANCD2 protein following ionizing radiation. FANCD2 is monoubiquitinated in FA-D1 cells, even though these cells are highly sensitive to MMC. CONCLUSIONS: The recently cloned FANCF protein is required for FANCD2 activation, and the yet uncloned FANCD1 protein functions further downstream or independently of the FA pathway.


Assuntos
Anemia de Fanconi/genética , Proteínas Nucleares/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Primers do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Anemia de Fanconi/fisiopatologia , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Proteína do Grupo de Complementação F da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Teste de Complementação Genética , Vetores Genéticos/genética , Humanos , Mitomicina/toxicidade , Proteínas Nucleares/fisiologia , Proteínas/genética , Proteínas/fisiologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Valores de Referência , Transdução de Sinais/fisiologia
7.
Blood ; 98(13): 3650-7, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11739169

RESUMO

Fanconi anemia (FA) is an autosomal recessive disease with congenital anomalies, bone marrow failure, and susceptibility to leukemia. Patient cells show chromosome instability and hypersensitivity to DNA cross-linking agents. At least 8 complementation groups (A-G) have been identified and 6 FA genes (for subtypes A, C, D2, E, F, and G) have been cloned. Increasing evidence indicates that a protein complex assembly of multiple FA proteins, including FANCA and FANCG, plays a crucial role in the FA pathway. Previously, it was reported that FANCA was phosphorylated in lymphoblasts from normal controls, whereas the phosphorylation was defective in those derived from patients with FA of multiple complementation groups. The present study examined phosphorylation of FANCA ectopically expressed in FANCA(-) cells. Several patient-derived mutations abrogated in vivo phosphorylation of FANCA in this system, suggesting that FANCA phosphorylation is associated with its function. In vitro phosphorylation studies indicated that a physiologic protein kinase for FANCA (FANCA-PK) forms a complex with the substrate. Furthermore, at least a part of FANCA-PK as well as phosphorylated FANCA were included in the FANCA/FANCG complex. Thus, FANCA-PK appears to be another component of the FA protein complex and may regulate function of FANCA. FANCA-PK was characterized as a cytoplasmic serine kinase sensitive to wortmannin. Identification of the protein kinase is expected to elucidate regulatory mechanisms that control the FA pathway.


Assuntos
Citoplasma/enzimologia , Proteínas de Ligação a DNA , Anemia de Fanconi/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Androstadienos/farmacologia , Western Blotting , Linhagem Celular Transformada , Inibidores Enzimáticos/farmacologia , Proteína do Grupo de Complementação A da Anemia de Fanconi , Fibroblastos/metabolismo , Granulócitos/metabolismo , Células HeLa/metabolismo , Humanos , Técnicas de Imunoadsorção , Células Jurkat/metabolismo , Linfócitos/metabolismo , Fosforilação , Transfecção , Wortmanina
8.
Blood ; 98(12): 3435-40, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11719385

RESUMO

Fanconi anemia (FA) is a human autosomal recessive cancer susceptibility disorder characterized by cellular sensitivity to mitomycin C and ionizing radiation. Six FA genes (corresponding to subtypes A, C, D2, E, F, and G) have been cloned, and the encoded FA proteins interact in a common cellular pathway. To further understand the in vivo role of one of these human genes (FANCG), we generated a targeted disruption of murine Fancg and bred mice homozygous for the targeted allele. Similar to the phenotype of the previously described Fancc(-/-) and Fanca(-/-) mice, the Fancg(-/-) mice had normal viability and no gross developmental abnormalities. Primary splenic lymphocytes, bone marrow progenitor cells, and murine embryo fibroblasts from the Fancg(-/-) mice demonstrated spontaneous chromosome breakage and increased sensitivity to mitomycin C and, to a lesser extent, ionizing radiation. Fancg(-/-) lymphocytes had a defect in the FA pathway, based on their failure to activate the monoubiquitination of the downstream Fancd2 protein in response to IR. Finally, Fancg(-/-) mice had decreased fertility and abnormal gonadal histology. In conclusion, disruption of the Fancg gene confirms the role of Fancg in the FA pathway. The Fancg(-/-) mouse may be useful as an animal model for future gene therapy and cancer susceptibility studies.


Assuntos
Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Anemia de Fanconi/genética , Alelos , Animais , Quebra Cromossômica , Modelos Animais de Doenças , Proteína do Grupo de Complementação G da Anemia de Fanconi , Células-Tronco Hematopoéticas/ultraestrutura , Homozigoto , Humanos , Immunoblotting , Infertilidade/genética , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Mutagênese , Fenótipo , Baço/ultraestrutura
9.
Exp Hematol ; 29(7): 842-9, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11438206

RESUMO

OBJECTIVE: Fanconi anemia (FA) is an autosomal-recessive cancer susceptibility syndrome with seven complementation groups. Six of the FA genes have been cloned (corresponding to subtypes A, C, D2, E, F, and G) and the encoded proteins interact in a common pathway. Patient-derived mutations in FA genes have been helpful in delineating functional domains of FA proteins. The purpose of this work was to subtype FA patient-derived cell lines in our repository and to identify FA gene mutations. METHODS: We subtyped 62 FA patients as type A, G, C, or non-ACG by using a combination of retroviral gene transfer and immunoblot analysis. Among these FA patients, we identified six FA-G patients for further analysis. We used a strategy involving amplification of FANCG/XRCC9 exons and direct sequencing to identify novel FANCG mutations in cell lines derived from these FA-G patients. We functionally analyzed FANCG mutant alleles by transducing the corresponding cDNAs into a known FA-G indicator cell line and scoring correction of MMC sensitivity. RESULTS: Our results demonstrate a wide range of mutations in the FANCG gene (splice, nonsense, and missense mutations). Based on this mutational screen, a carboxy terminal functional domain of the FANCG protein appears to be required for complementation of FA-G cells and for normal assembly of the FANCA/FANCG/FANCC protein complex. CONCLUSION: The identification of patient-derived mutant alleles of FA genes can provide important insights to the function of FA proteins. FA subtyping is also a necessary precondition for gene therapy.


Assuntos
Proteínas de Ligação a DNA/genética , Anemia de Fanconi/genética , Adolescente , Alelos , Criança , Pré-Escolar , DNA Complementar/análise , DNA Complementar/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi , Feminino , Heterozigoto , Humanos , Masculino , Mutação
10.
Blood ; 98(3): 636-42, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11468161

RESUMO

Protein ubiquitination is an important regulator of cytokine-activated signal transduction pathways and hematopoietic cell growth. Protein ubiquitination is controlled by the coordinate action of ubiquitin-conjugating enzymes and deubiquitinating enzymes. Recently a novel family of genes encoding growth-regulatory deubiquitinating enzymes (DUB-1 and DUB-2) has been identified. DUBs are immediate-early genes and are induced rapidly and transiently in response to cytokine stimuli. By means of polymerase chain reaction amplification with degenerate primers for the DUB-2 complementary DNA, 3 murine bacterial artificial chromosome (BAC) clones that contain DUB gene sequences were isolated. One BAC contained a novel DUB gene (DUB-2A) with extensive homology to DUB-2. Like DUB-1 and DUB-2, the DUB-2A gene consists of 2 exons. The predicted DUB-2A protein is highly related to other DUBs throughout the primary amino acid sequence, with a hypervariable region at its C-terminus. In vitro, DUB-2A had functional deubiquitinating activity; mutation of its conserved amino acid residues abolished this activity. The 5' flanking sequence of the DUB-2A gene has a hematopoietic-specific functional enhancer sequence. It is proposed that there are at least 3 members of the DUB subfamily (DUB-1, DUB-2, and DUB-2A) and that different hematopoietic cytokines induce specific DUB genes, thereby initiating a cytokine-specific growth response. (Blood. 2001;98:636-642)


Assuntos
Endopeptidases/genética , Proteínas Imediatamente Precoces/genética , Animais , Sequência de Bases , Domínio Catalítico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Clonagem Molecular , Citocinas/farmacologia , DNA Complementar , Endopeptidases/química , Endopeptidases/farmacologia , Elementos Facilitadores Genéticos/efeitos dos fármacos , Regulação da Expressão Gênica , Substâncias de Crescimento/química , Substâncias de Crescimento/genética , Substâncias de Crescimento/farmacologia , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/farmacologia , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Transfecção , Ubiquitinas/metabolismo
11.
Blood ; 97(8): 2230-7, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11290583

RESUMO

Erythropoietin (EPO) specifically activates the Janus kinase JAK2 and the transcription factor signal transducer and activator of transcription-5 (STAT5). All members of the STAT family are tyrosine phosphorylated in response to cytokine stimulation at a conserved carboxy-terminal tyrosine, Y694, in the case of STAT5. To determine structural features important for STAT signaling, we generated an activation-specific STAT5 antibody using a phosphopeptide containing amino acids 687 to 698 of STAT5 as antigen. This antibody specifically recognizes tyrosine- phosphorylated STAT5 but not nonphosphorylated STAT5. In immunoprecipitation reactions from cell lines and primary erythroblasts, 2 distinct polyclonal activation-specific STAT5 antibodies selectively immunoprecipitate the tyrosine phosphorylated EPO receptor (EPO-R) in addition to STAT5 under native and denaturing conditions. We propose that the activation-specific STAT5 antibody recognizes the 2 substrates to which the STAT5 SH2 domain interacts, namely, the tyrosine- phosphorylated EPO-R and STAT5 itself. Several studies have implicated EPO-R Y343, Y401, Y431, and Y479 in the recruitment of STAT5. Using a series of EPO-R tyrosine mutants expressed in Ba/F3 cells, we have shown that the activation-specific STAT5 antibody immunoprecipitates an EPO-R containing only 2 tyrosines at positions 343 and 401, confirming the importance of these tyrosines in STAT5 recruitment. These data uncover a novel aspect of STAT SH2 domain recognition and demonstrate the utility of activation-specific antibodies for examining the specificity of STAT-cytokine receptor interactions.


Assuntos
Proteínas de Ligação a DNA/imunologia , Epitopos/imunologia , Eritropoetina/farmacologia , Proteínas do Leite , Receptores da Eritropoetina/imunologia , Transativadores/imunologia , Animais , Especificidade de Anticorpos , Western Blotting , Células Cultivadas , Proteínas de Ligação a DNA/química , Epitopos/química , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fragmentos de Peptídeos/imunologia , Fosforilação , Fosfotirosina/química , Testes de Precipitina , Desnaturação Proteica , Processamento de Proteína Pós-Traducional , Coelhos , Receptores da Eritropoetina/química , Fator de Transcrição STAT5 , Ovinos , Transativadores/química , Domínios de Homologia de src
12.
Mol Cell ; 7(2): 241-8, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11239453

RESUMO

Fanconi anemia (FA) is a genetic disease with birth defects, bone marrow failure, and cancer susceptibility. To date, genes for five of the seven known complementation groups have been cloned. Complementation group D is heterogeneous, consisting of two distinct genes, FANCD1 and FANCD2. Here we report the positional cloning of FANCD2. The gene consists of 44 exons, encodes a novel 1451 amino acid nuclear protein, and has two protein isoforms. Similar to other FA proteins, the FANCD2 protein has no known functional domains, but unlike other known FA genes, FANCD2 is highly conserved in A. thaliana, C. elegans, and Drosophila. Retroviral transduction of the cloned FANCD2 cDNA into FA-D2 cells resulted in functional complementation of MMC sensitivity.


Assuntos
Anemia de Fanconi/genética , Proteínas Nucleares/genética , Alelos , Sequência de Aminoácidos , Sequência de Bases , Western Blotting , Linhagem Celular , Quebra Cromossômica/genética , Clonagem Molecular , Análise Mutacional de DNA , Proteínas de Drosophila , Compostos de Epóxi/farmacologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Feminino , Perfilação da Expressão Gênica , Teste de Complementação Genética , Humanos , Masculino , Mitomicina/farmacologia , Dados de Sequência Molecular , Mutação/genética , Proteínas Nucleares/química , Linhagem , Fenótipo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Alinhamento de Sequência , Transdução Genética
13.
Mol Cell ; 7(2): 249-62, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11239454

RESUMO

Fanconi anemia (FA) is a human autosomal recessive cancer susceptibility disorder characterized by cellular sensitivity to mitomycin C and ionizing radiation. Although six FA genes (for subtypes A, C, D2, E, F, and G) have been cloned, their relationship to DNA repair remains unknown. In the current study, we show that a nuclear complex containing the FANCA, FANCC, FANCF, and FANCG proteins is required for the activation of the FANCD2 protein to a monoubiquitinated isoform. In normal (non-FA) cells, FANCD2 is monoubiquitinated in response to DNA damage and is targeted to nuclear foci (dots). Activated FANCD2 protein colocalizes with the breast cancer susceptibility protein, BRCA1, in ionizing radiation-induced foci and in synaptonemal complexes of meiotic chromosomes. The FANCD2 protein, therefore, provides the missing link between the FA protein complex and the cellular BRCA1 repair machinery. Disruption of this pathway results in the cellular and clinical phenotype common to all FA subtypes.


Assuntos
Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Anemia de Fanconi/genética , Proteínas/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos da radiação , Animais , Linhagem Celular , Sobrevivência Celular , Dano ao DNA/genética , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Imunofluorescência , Teste de Complementação Genética , Humanos , Substâncias Macromoleculares , Masculino , Meiose/genética , Camundongos , Mitomicina/farmacologia , Proteínas Nucleares/metabolismo , Ligação Proteica , Radiação Ionizante , Espermatócitos/citologia , Espermatócitos/metabolismo , Complexo Sinaptonêmico/metabolismo , Ubiquitinas/metabolismo , Raios Ultravioleta
14.
Mol Ther ; 3(1): 14-23, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11162306

RESUMO

Fanconi anemia (FA) is an autosomal recessive disorder characterized by birth defects, increased incidence of malignancy, progressive bone marrow failure, and cellular hypersensitivity to DNA cross-linking agents. Bone marrow transplantation is therapeutic and therefore FA is a candidate disease for hematopoietic gene therapy. We have previously used mitomycin C (MMC) to achieve in vivo selection of wild-type hematopoietic stem cells (HSC) transplanted into FANCC knockout mice. However, clinical application of MMC in human FA gene therapy is unlikely because of its unknown toxicity profile in human FA patients. In contrast, cyclophosphamide (CPA) and gamma-irradiation (IR) are already in use with human FA patients and we therefore tested these regimens for their ability to achieve selection of genetically corrected HSCs in vivo. We found that nonmyeloablative doses of CPA or IR or combinations of CPA + IR were highly efficient at achieving in vivo selection of transplanted wild-type HSC. Furthermore, this nontoxic regimen also selected FANCC-mutant HSC corrected by ex vivo retroviral gene therapy. We suggest those nontoxic doses of CPA and/or IR could also be used to enhance gene therapy in human FA patients.


Assuntos
Anemia de Fanconi/terapia , Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Células 3T3 , Animais , Southern Blotting , Células da Medula Óssea/metabolismo , Células Cultivadas , Terapia Combinada , Ciclofosfamida/farmacologia , Relação Dose-Resposta a Droga , Anemia de Fanconi/genética , Feminino , Raios gama , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunossupressores/farmacologia , Masculino , Camundongos , Camundongos Mutantes , Fenótipo , Radiação Ionizante , Retroviridae/genética , Fatores de Tempo
15.
Eur J Hum Genet ; 8(11): 861-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11093276

RESUMO

FANCG was the third Faconi anaemia gene identified and proved to be identical to the previously cloned XRCC9 gene. We present the pathogenic mutations and sequence variants we have so far identified in a panel of FA-G patients. Mutation screening was performed by PCR, single strand conformational polymorphism analysis and protein truncation tests. Altogether 18 mutations have been determined in 20 families - 97% of all expected mutant alleles. All mutation types have been found, with the exception of large deletions, the large majority is predicted to lead to shortened proteins. One stop codon mutation, E105X, has been found in several German patients and this founder mutation accounts for 44% of the mutant FANCG alleles in German FA-G patients. Comparison of clinical phenotypes shows that patients homozygous for this mutation have an earlier onset of the haematological disorder than most other FA-G patients. The mouse Fancg sequence was established in order to evaluate missense mutations. A putative missense mutation, L71P, in a possible leucine zipper motif may affect FANCG binding of FANCA and seems to be associated with a milder clinical phenotype.


Assuntos
Proteínas de Ligação a DNA/genética , Anemia de Fanconi/genética , Mutação , Sequência de Aminoácidos , Sequência de Bases , DNA/química , DNA/genética , Análise Mutacional de DNA , Proteína do Grupo de Complementação G da Anemia de Fanconi , Humanos , Dados de Sequência Molecular , Polimorfismo Conformacional de Fita Simples , Homologia de Sequência de Aminoácidos
17.
Blood ; 96(9): 3224-30, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11050007

RESUMO

Fanconi anemia (FA) is an autosomal recessive cancer susceptibility syndrome with 8 complementation groups. Four of the FA genes have been cloned, and at least 3 of the encoded proteins, FANCA, FANCC, and FANCG/XRCC9, interact in a multisubunit protein complex. The FANCG protein binds directly to the amino terminal nuclear localization sequence (NLS) of FANCA, suggesting that FANCG plays a role in regulating FANCA nuclear accumulation. In the current study the functional consequences of FANCG/FANCA binding were examined. Correction of an FA-G cell line with the FANCG complementary DNA (cDNA) resulted in FANCA/FANCG binding, prolongation of the cellular half-life of FANCA, and an increase in the nuclear accumulation of the FA protein complex. Similar results were obtained upon correction of an FA-A cell line, with a reciprocal increase in the half-life of FANCG. Patient-derived mutant forms of FANCA, containing an intact NLS sequence but point mutations in the carboxy-terminal leucine zipper region, bound FANCG in the cytoplasm. The mutant forms failed to translocate to the nucleus of transduced cells, thereby suggesting a model of coordinated binding and nuclear translocation. These results demonstrate that the FANCA/FANCG interaction is required to maintain the cellular levels of both proteins. Moreover, at least one function of FANCG and FANCA is to regulate the nuclear accumulation of the FA protein complex. Failure to accumulate the nuclear FA protein complex results in the characteristic spectrum of clinical and cellular abnormalities observed in FA.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Anemia de Fanconi/metabolismo , Proteínas/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Humanos , Cinética , Ligação Proteica , Subunidades Proteicas , Proteínas/química , Proteínas/isolamento & purificação , Proteínas Recombinantes/metabolismo
18.
Blood ; 96(5): 1625-32, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10961856

RESUMO

Fanconi anemia (FA) is an autosomal recessive cancer susceptibility syndrome with eight complementation groups. Four of the FA genes have been cloned, and at least three of the encoded proteins, FANCA, FANCC, and FANCG/XRCC9, interact in a nuclear complex, required for the maintenance of normal chromosome stability. In the current study, mutant forms of the FANCA and FANCG proteins have been generated and analyzed with respect to protein complex formation, nuclear translocation, and functional activity. The results demonstrate that the amino terminal two-thirds of FANCG (FANCG amino acids 1-428) binds to the amino terminal nuclear localization signal (NLS) of the FANCA protein. On the basis of 2-hybrid analysis, the FANCA/FANCG binding is a direct protein-protein interaction. Interestingly, a truncated mutant form of the FANCG protein, lacking the carboxy terminus, binds in a complex with FANCA and translocates to the nucleus; however, this mutant protein fails to bind to FANCC and fails to correct the mitomycin C sensitivity of an FA-G cell line. Taken together, these results demonstrate that binding of FANCG to the amino terminal FANCA NLS sequence is necessary but not sufficient for the functional activity of FANCG. Additional amino acid sequences at the carboxy terminus of FANCG are required for the binding of FANCC in the complex. (Blood. 2000;96:1625-1632)


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares , Animais , Sítios de Ligação/genética , Ligação Competitiva , Transporte Biológico , Células COS , Linhagem Celular Transformada , Núcleo Celular/metabolismo , DNA Recombinante/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Teste de Complementação Genética , Humanos , Mutação , Sinais de Localização Nuclear/genética , Oligopeptídeos/metabolismo , Plasmídeos , Ligação Proteica , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Técnicas do Sistema de Duplo-Híbrido
19.
Biochim Biophys Acta ; 1446(3): 308-16, 1999 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-10524205

RESUMO

The murine DUB-1 gene is a hematopoietic-specific, immediate-early gene that encodes a growth-regulatory deubiquitinating enzyme. DUB-1 contains an IL-3-inducible enhancer element that is activated in a JAK2-dependent, STAT5-independent manner. In this study, we have further characterized this novel IL-3 response element. Transcriptional reporter assays in Ba/F3 cells revealed that two AP-1 sites, a GATA motif, and an Ets site are required for induction of DUB-1 enhancer activity. Gel shift assays indicated that IL-3 activates the binding of an AP-1 complex containing JunD to the AP-1 sites and the binding of another protein complex to the Ets motif. The latter complex was not detectable in Ba/F3 cells stably transfected with a dominant-negative mutant of JAK2. As previously shown, these cells do not express DUB-1 mRNA or protein. Furthermore, we demonstrated that GATA-1 constitutively binds to the DUB-1 enhancer element. The involvement of GATA-1 may be important for the hematopoietic-restricted expression pattern of DUB-1. This combination of inducible and constitutive elements of the DUB-1 enhancer appears to account for the unique STAT-independent expression characteristics of DUB-1.


Assuntos
Endopeptidases , Elementos Facilitadores Genéticos/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Interleucina-3/farmacologia , Proteínas Proto-Oncogênicas , Transativadores , Animais , Células Cultivadas , Sequência Consenso , Regulação da Expressão Gênica/efeitos dos fármacos , Janus Quinase 2 , Camundongos , Ligação Proteica , Proteínas Tirosina Quinases/farmacologia , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...