Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 116(12): 2057-60, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20554974

RESUMO

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


Assuntos
Proteína do Grupo de Complementação C da Anemia de Fanconi/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Endotoxinas/farmacologia , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Humanos , Hipersensibilidade Imediata/induzido quimicamente , Camundongos , Camundongos Knockout , Transgenes
2.
Blood ; 96(13): 4204-11, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11110692

RESUMO

Hematopoietic progenitor cells (HPC) from mice nullizygous at the Fanconi anemia (FA) group C locus and children with Fanconi anemia group C (FA-C) are hypersensitive to interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha. This hypersensitivity results, in part, from the capacity of these cytokines to prime the fas pathway. Because fas-mediated programmed cell death in many cells involves sequential activation of specific caspases, we tested the hypothesis that programmed cell death in FA HPC involves the ordered activation of specific caspase molecules. Lysates from lymphoblasts treated with both agonistic anti-fas antibody and IFN-gamma contained activated caspase 3 family members (caspases 3, 6, and 7), as well as caspase 8, whereas activation of caspases 1, 2, 4, 9, and 10 was not detected. The apoptotic effects of fas agonists in IFN-gamma-treated human and murine FA-C cells were blocked when pretreated with inhibitors (ac-DEVD-cho, CP-DEVD-cho, Z-DEVD-FMK) of the caspase 3 protease. Inhibitors (ac-YVAD-cho, CP-YVAD-cho, Z-YVAD-FMK) of caspase 1 did not block apoptosis or caspase 3 activation. Treatment of FA cells with the fluoromethyl ketone tetrapeptide caspase 8 inhibitor (ac-IETD-FMK) did suppress caspase 3 activation. A 4-fold greater fraction of IFN-induced FA-C cells expressed caspase 3 than FA-C cells complemented by retroviral-mediated transfer of FANCC. Therefore fas-induced apoptosis in Fanconi anemia cells of the C type involves the activation of caspase 8, which controls activation of caspase 3 family members and one direct or indirect function of the FANCC protein is to suppress apoptotic responses to IFN-gamma upstream of caspase 3 activation. (Blood. 2000;96:4204-4211)


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Caspases/fisiologia , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Anemia de Fanconi/patologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interferon gama/farmacologia , Isoenzimas/metabolismo , Proteínas Nucleares , Proteínas/fisiologia , Animais , Medula Óssea/patologia , Caspase 3 , Caspase 8 , Caspase 9 , Inibidores de Caspase , Linhagem Celular Transformada , Células Cultivadas , Criança , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática/efeitos dos fármacos , Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Heterogeneidade Genética , Células-Tronco Hematopoéticas/enzimologia , Humanos , Isoenzimas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/genética , Proteínas Recombinantes de Fusão/fisiologia , Proteínas Recombinantes , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Transfecção , Receptor fas/fisiologia
3.
Mol Cell Biol ; 20(13): 4724-35, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10848598

RESUMO

Hematopoietic progenitor cells from Fanconi anemia (FA) group C (FA-C) patients display hypersensitivity to the apoptotic effects of gamma interferon (IFN-gamma) and constitutively express a variety of IFN-dependent genes. Paradoxically, however, STAT1 activation is suppressed in IFN-stimulated FA cells, an abnormality corrected by transduction of normal FANCC cDNA. We therefore sought to define the specific role of FANCC protein in signal transduction through receptors that activate STAT1. Expression and phosphorylation of IFN-gamma receptor alpha chain (IFN-gammaRalpha) and JAK1 and JAK2 tyrosine kinases were equivalent in both normal and FA-C cells. However, in coimmunoprecipitation experiments STAT1 did not dock at the IFN-gammaR of FA-C cells, an abnormality corrected by transduction of the FANCC gene. In addition, glutathione S-transferase fusion genes encoding normal FANCC but not a mutant FANCC bearing an inactivating point mutation (L554P) bound to STAT1 in lysates of IFN-gamma-stimulated B cells and IFN-, granulocyte-macrophage colony-stimulating factor- and stem cell factor-stimulated MO7e cells. Kinetic studies revealed that the initial binding of FANCC was to nonphosphorylated STAT1 but that subsequently the complex moved to the receptor docking site, at which point STAT1 became phosphorylated. The STAT1 phosphorylation defect in FA-C cells was functionally significant in that IFN induction of IFN response factor 1 was suppressed and STAT1-DNA complexes were not detected in nuclear extracts of FA-C cells. We also determined that the IFN-gamma hypersensitivity of FA-C hematopoietic progenitor cells does not derive from STAT1 activation defects because granulocyte-macrophage CFU and erythroid burst-forming units from STAT1(-/-) mice were resistant to IFN-gamma. However, BFU-E responses to SCF and erythropoietin were suppressed in STAT(-/-) mice. Consequently, because the FANCC protein is involved in the activation of STAT1 through receptors for at least three hematopoietic growth and survival factor molecules, we reason that FA-C hematopoietic cells are excessively apoptotic because of an imbalance between survival cues (owing to a failure of STAT1 activation in FA-C cells) and apoptotic and mitogenic inhibitory cues (constitutively activated in FA-C cells in a STAT1-independent fashion).


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Substâncias de Crescimento/metabolismo , Interferon gama/metabolismo , Proteínas Nucleares , Proteínas/metabolismo , Proteínas Proto-Oncogênicas , Transativadores/metabolismo , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linhagem Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Substâncias de Crescimento/farmacologia , Humanos , Fator Regulador 1 de Interferon , Interferon gama/farmacologia , Janus Quinase 1 , Janus Quinase 2 , Camundongos , Camundongos Knockout , Mutação , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Testes de Precipitina , Proteínas Tirosina Quinases/metabolismo , Proteínas/genética , Receptores de Interferon/efeitos dos fármacos , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1 , Transativadores/genética , Receptor de Interferon gama
4.
Leukemia ; 13(11): 1784-9, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10557053

RESUMO

Specific chromosomal deletions are commonly found in bone marrow cells of children with Fanconi anemia (FA) whose disease has evolved to myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Identical deletions are found in adults with MDS/AML with a history of exposure to alkylating agents (secondary MDS/AML). While deleted chromosomal regions likely harbor genes encoding proteins with tumor suppressor (TS) function, such genes have not been identified and the environmental forces by which these mutant clones are selected remain unclear. A consistent signaling abnormality in cells bearing mutations of the Fanconi anemia complementation group C (FA-C) gene (FANCC) has revealed a potential selective force. Hematopoietic progenitor cells from patients and mice with FANCC mutations are hypersensitive to the inhibitory effects of IFNgamma and TNFalpha. Consequently, clonal outgrowths in FA likely result from strong selective pressure for stem and/or progenitor cells resistant to these inhibitory cytokines. Additional mutations that inactivate signaling pathways for these inhibitors would create a cell with a profound proliferative advantage over its apoptosis-prone counterparts. Here, we present preliminary evidence supporting a selection-based model of leukemic evolution and argue that MDS in FA patients is a de facto model of secondary MDS in non-FA adults.


Assuntos
Células Clonais/metabolismo , Evolução Molecular , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Leucemia Mieloide/patologia , Seleção Genética , Doença Aguda , Animais , Apoptose , Aberrações Cromossômicas/genética , Células Clonais/patologia , Proteínas de Ligação a DNA/genética , Anemia de Fanconi/complicações , Humanos , Fator Regulador 1 de Interferon , Leucemia Mieloide/complicações , Leucemia Mieloide/genética , Modelos Genéticos , Síndromes Mielodisplásicas/complicações , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Fosfoproteínas/genética
5.
Blood ; 94(6): 2151-8, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10477746

RESUMO

Fanconi anemia (FA) is an autosomal recessive disorder characterized by birth defects, increased incidence of malignancy, and progressive bone marrow failure. Bone marrow transplantation is therapeutic and, therefore, FA is a candidate disease for hematopoietic gene therapy. The frequent finding of somatic mosaicism in blood of FA patients has raised the question of whether wild-type bone marrow may have a selective growth advantage. To test this hypothesis, a cohort radio-ablated wild-type mice were transplanted with a 1:1 mixture of FA group C knockout (FACKO) and wild-type bone marrow. Analysis of peripheral blood at 1 month posttransplantation showed only a moderate advantage for wild-type cells, but upon serial transplantation, clear selection was observed. Next, a cohort of FACKO mice received a transplant of wild-type marrow cells without prior radio-ablation. No wild-type cells were detected in peripheral blood after transplantation, but a single injection of mitomycin C (MMC) resulted in an increase to greater than 25% of wild-type DNA. Serial transplantation showed that the selection occurred at the level of hematopoietic stem cells. No systemic side effects were observed. Our results show that in vivo selection for wild-type hematopoietic stem cells occurs in FA and that it is enhanced by MMC administration.


Assuntos
Transplante de Medula Óssea , Anemia de Fanconi/terapia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/patologia , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Genótipo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Reação em Cadeia da Polimerase
6.
Exp Hematol ; 26(1): 19-26, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9430510

RESUMO

The Fanconi anemia group C gene (FAC) encodes a 63-kDa protein that plays a role in the growth and differentiation of hematopoietic progenitor cells and in cellular resistance to bifunctional cross-linking agents. The function of the gene product is unknown, as are the factors that govern expression of the gene itself. Seeking to associate a function of this protein with a general metabolic pathway, we attempted to identify factors that induce or repress expression of the gene encoding it. Using two plasmids from which mutant FAC mRNA molecules were transcribed in vitro to serve as competitor mRNAs in quantitative-competitive reverse transcriptase-polymerase chain reaction analysis and novel rabbit antisera raised to recombinant FAC proteins, we quantified gene expression in human hematopoietic cells. We determined that FAC is expressed constitutively in unstimulated normal peripheral blood mononuclear leukocytes, in Epstein-Barr virus (EBV)-transformed B lymphocytes, and in the factor-dependent human myeloid leukemic cell line MO7e at levels of approximately 2000, 200, and 200 FAC mRNA molecules/cell, respectively, and in CD34+ cells from normal human bone marrow at approximately 2000 FAC mRNA molecules/cell. Neither mRNA nor protein increased in any of the cells studied after exposure to mitomycin C, diepoxybutane, hydrogen peroxide, gamma radiation, heat, transforming growth factor-beta, or interferon-gamma. Using these sensitive methods, we confirmed that the FAC gene is constitutively expressed, even in the face of extracellular factors for which the gene product is a known effector of resistance. We conclude that the protective functions of the FAC gene product do not depend upon stressor-induced FAC gene expression.


Assuntos
Proteínas de Ciclo Celular , Reagentes de Ligações Cruzadas/farmacologia , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Hematopoese/genética , Monócitos/metabolismo , Proteínas Nucleares , Proteínas/genética , Animais , Células Cultivadas , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Transtornos de Estresse por Calor , Humanos , Mitose/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/patologia , Estresse Oxidativo , Coelhos , Transfecção
7.
Blood ; 90(3): 974-85, 1997 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9242526

RESUMO

Hematopoietic progenitor cells (HPC) from mice nullizygous at the Fanconi anemia (FA) group C locus (FAC -/-) are hypersensitive to the mitotic inhibitory effects of interferon (IFN-gamma). We tested the hypothesis that HPC from the bone marrow of Fanconi group C children are similarly hypersensitive and that the fas pathway is involved in affecting programmed cell death in response to low doses of IFN-gamma. In normal human and murine HPC, IFN-gamma primed the fas pathway and induced both fas and interferon response factor-1 (IRF-1) gene expression. These IFN-gamma-induced apoptotic responses in HPC from the marrow of a child with FA of the C group (FA-C) and in FAC -/- mice occurred at significantly lower IFN doses (by an order of magnitude) than did the apoptotic responses of normal HPC. Treatment of FA-C CD34+ cells with low doses of recombinant IFN-gamma, inhibited growth of colony forming unit granulocyte-macrophage and burst-forming unit erythroid, while treatment with blocking antibodies to fas augmented clonal growth and abrogated the clonal inhibitory effect of IFN-gamma. Transfer of the normal FAC gene into FA-C B-cell lines prevented mitomycin C-induced apoptosis, but did not suppress fas expression or inhibit the primed fas pathway. However, the kinetics of Stat1-phosphate decay in IFN-gamma-treated cells was prolonged in mutant cells and was normalized by transduction of the normal FAC gene. Therefore, the normal FAC protein serves, in part, to modulate IFN-gamma signals. HPC bearing inactivating mutations of FAC fail to normally modulate IFN-gamma signals and, as a result, undergo apoptosis executed through the fas pathway.


Assuntos
Apoptose/genética , Proteínas de Ciclo Celular , Anemia de Fanconi/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Nucleares , Proteínas/fisiologia , Anemia Aplástica/etiologia , Anemia Aplástica/fisiopatologia , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Células Cultivadas , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Anemia de Fanconi/complicações , Anemia de Fanconi/fisiopatologia , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Proteína Ligante Fas , Teste de Complementação Genética , Humanos , Fator Regulador 1 de Interferon , Interferon gama/farmacologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Proteínas/genética , Proteínas Recombinantes , Transdução de Sinais , Transfecção , Receptor fas/genética , Receptor fas/fisiologia
8.
Blood ; 88(1): 49-58, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8704201

RESUMO

Fanconi anemia (FA) is an autosomal recessive chromosome instability syndrome characterized by progressive bone marrow (BM) failure, skeletal defects, and increased susceptibility to malignancy. FA cells are hypersensitive to DNA cross-linking agents, oxygen and have cell cycle abnormalities. To develop an animal model of the disease we generated mice homozygous for a targeted deletion of exon 9 of the murine FA complementation group C gene (fac). Mutant mice had normal neonatal viability and gross morphology, but their cells had the expected chromosome breakage and DNA cross-linker sensitivity. Surprisingly, male and female mutant mice had reduced numbers of germ cells and females had markedly impaired fertility. No anemia was detectable in the peripheral blood during the first year of life, but the colony forming capacity of marrow progenitor cells was abnormal in vitro in mutant mice. Progenitor cells from fac knock-out mice were hypersensitive to interferon gamma. This previously unrecognized phenotype may form the basis for BM failure in human FA.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Anemia de Fanconi/genética , Células Germinativas/patologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Infertilidade Feminina/genética , Interferon gama/farmacologia , Proteínas Nucleares , Proteínas/genética , Animais , Sequência de Bases , Ciclo Celular , Células Cultivadas , Quimiocina CCL4 , Aberrações Cromossômicas , Ensaio de Unidades Formadoras de Colônias , Reagentes de Ligações Cruzadas/farmacologia , Dano ao DNA , Éxons/genética , Anemia de Fanconi/patologia , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Feminino , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Células-Tronco Hematopoéticas/patologia , Proteínas Inflamatórias de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Monocinas/farmacologia , Ovário/patologia , Proteínas/fisiologia , Proteínas Recombinantes/farmacologia , Método Simples-Cego , Testículo/patologia , Fator de Necrose Tumoral alfa/farmacologia
9.
Exp Hematol ; 14(8): 789-93, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3488917

RESUMO

Monocyte-derived recruiting activity (MRA) stimulates the release of granulopoietic colony-stimulating factors (CSF) by endothelial cells. We carried out studies designed to test the hypotheses that human placental conditioned medium (HPCM), a widely utilized source of CSA for in vitro studies, contains both MRA and CSA and that these molecules could be separated on the basis of their isoelectric points. In five separate studies, concentrated samples of HPCM were chromatofocused and fractions tested in bioassays for MRA and CSA. We found that HPCM contains both MRA and CSA, that the majority of the MRA eluted at a pH of from 7.6 to 8.2 and was 2000-fold purified. CSA eluted at pH 5.6 or below. No CSA was detectable in the major MRA peak. We conclude that HPCM contains heterogeneous granulopoietic activities, that the isoelectric points of MRA and CSA are substantially different and that chromatofocusing provides a rapid single-step method for separating these two distinct granulopoietic factors from complex conditioned media.


Assuntos
Fatores Estimuladores de Colônias/análise , Monócitos/fisiologia , Placenta/fisiologia , Proteínas/análise , Meios de Cultura , Humanos , Ponto Isoelétrico , Monocinas , Placenta/análise
10.
J Clin Invest ; 77(4): 1340-8, 1986 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3485660

RESUMO

The capsular polysaccharide of Hemophilus influenzae type b, polyribosyl ribitol phosphate (PRP), is released from growing organisms during human infection and can be found in body fluids. It binds to untreated erythrocytes. Many patients with invasive infections with this organism develop significant hemolysis, but the mechanism has been unclear. We have found that PRP binds to human erythrocytes in vivo. PRP-coated erythrocytes have a shortened circulation time in mice, but do not lyse spontaneously or fix complement. PRP-coated erythrocytes exposed to antiserum to H. influenzae type b are undamaged in the absence of complement, but are rapidly and effectively lysed in the presence of an intact complement system both in vitro and in vivo in mice. PRP-coated red cells are taken up by liver and spleen. Antiserum to PRP increases hepatic uptake of PRP-coated red cells more than splenic, and appears to induce intravascular, complement-mediated hemolysis, as well as extravascular hemolysis. Patients with invasive infection develop hemolysis when circulating PRP and antibody to PRP are present simultaneously. PRP can sometimes be detected on patient erythrocytes when free PRP is present in serum, but this is an inconsistent finding. The hemolytic anemia that occurs during human infection with H. influenzae type b may be due to absorption of PRP to red cells and immune destruction of sensitized erythrocytes. The process requires an intact complement system; both complement-mediated cell lysis and extravascular hemolysis contribute to red cell destruction.


Assuntos
Infecções por Haemophilus/fisiopatologia , Hemólise , Anemia Hemolítica/etiologia , Animais , Sobrevivência Celular , Cromo/metabolismo , Eritrócitos/citologia , Cobaias , Infecções por Haemophilus/complicações , Haemophilus influenzae , Testes de Hemaglutinação , Humanos , Fígado/metabolismo , Pulmão/metabolismo , Meningite/complicações , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Polissacarídeos/metabolismo , Sepse/complicações , Sepse/fisiopatologia , Baço/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...