Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 105(9): 3538-41, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15644420

RESUMO

In vitro studies suggest that Ras activation is necessary for erythroid cell development. However, genetic inactivation of the Ras isoforms H-Ras, N-Ras, and K-Ras in mice reportedly did not affect adult or fetal erythropoiesis, though K-Ras(-/-) embryos were anemic. Given these discrepancies, we performed a more detailed analysis of fetal erythropoiesis in K-Ras(-/-) embryos. Day-13.5 K-Ras(-/-) embryos were pale with a marked reduction of mature erythrocytes in their fetal livers. The frequency and number of both early (erythroid burst-forming unit [BFU-E]) and late erythroid progenitors (erythroid colony-forming unit [CFU-E]) were reduced in K-Ras(-/-) fetal livers compared with wild-type controls and displayed a delay in terminal erythroid cell maturation. Further, K-Ras(-/-) hematopoietic progenitors had reduced proliferation in response to erythropoietin and Kit ligand compared with control cells. Thus, these studies identify K-Ras as a unique Ras isoform that is essential for regulating fetal erythropoiesis in vivo.


Assuntos
Eritropoese , Fígado/embriologia , Proteínas ras/fisiologia , Anemia/etiologia , Animais , Proliferação de Células , Eritrócitos/citologia , Feto/citologia , Células-Tronco Hematopoéticas , Fígado/enzimologia , Fígado/fisiologia , Camundongos , Camundongos Knockout , Isoformas de Proteínas , Proteínas ras/genética
2.
J Clin Invest ; 112(12): 1851-61, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14679180

RESUMO

The NF1 tumor suppressor gene encodes a GTPase-activating protein called neurofibromin that negatively regulates Ras signaling. Mutations in NF1 cause neurofibromatosis type 1 (NF1). The development of neurofibromas, which are complex tumors composed of multiple cell types, is a hallmark of NF1. Somatic inactivation of murine Nf1 in Schwann cells is necessary, but not sufficient, to initiate neurofibroma formation. Neurofibromas occur with high penetrance in mice in which Nf1 is ablated in Schwann cells in the context of a heterozygous mutant (Nf1+/-) microenvironment. Mast cells infiltrate neurofibromas, where they secrete proteins that can remodel the ECM and initiate angiogenesis. Thus, identification of mechanisms responsible for mast cell migration to tumor microenvironments is important for understanding tumorigenesis and for designing potential therapies. Here, we show that homozygous Nf1 mutant (Nf1-/-) Schwann cells secrete Kit ligand (KitL), which stimulates mast cell migration, and that Nf1+/- mast cells are hypermotile in response to KitL. Furthermore, we link hyperactivation of the Ras-class IA-PI3K-Rac2 pathway to increased Nf1+/- mast cell migration. Thus, these studies identify a novel interaction between Nf1-/- Schwann cells and Nf1+/- mast cells that is likely to be important in neurofibroma formation.


Assuntos
Neurofibromina 1/genética , Neurofibromina 1/fisiologia , Células de Schwann/metabolismo , Animais , Células da Medula Óssea/citologia , Movimento Celular , Meios de Cultura/farmacologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Genes da Neurofibromatose 1 , Heterozigoto , Homozigoto , Mastócitos/metabolismo , Camundongos , Mutação , Neurofibroma/metabolismo , Plasmídeos/metabolismo , Retroviridae/genética , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Fatores de Tempo
3.
Blood ; 102(1): 142-5, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12623844

RESUMO

In vitro studies suggest that activation of class IA phosphatidylinositol 3 (PI-3) kinase is necessary for normal erythroid cell development. However, when class IA PI-3 kinase-deficient mice were generated by a targeted deletion of the p85alpha regulatory subunit, fetal erythropoiesis was reportedly unaffected. Given the discrepancies between these studies, we performed a more detailed in vivo analysis of class IA PI-3 kinase-deficient embryos. Day-14.5 p85alpha-/- embryos are pale with a marked reduction of mature erythrocytes in their peripheral blood. Further, the absolute number and frequency of both early (erythroid burst-forming unit [BFU-E]) and late erythroid progenitors (erythroid colony-forming unit [CFU-E]) are reduced in p85alpha-/- fetal livers compared with wild-type controls, which is associated with reduced proliferation. Taken together, these data establish an important role for p85alpha and class IA PI-3 kinase in regulating the development of both early and late erythroid progenitors in fetal liver.


Assuntos
Eritropoese/genética , Genes/fisiologia , Fosfatidilinositol 3-Quinases/genética , Animais , Apoptose , Contagem de Células , Divisão Celular , Células Precursoras Eritroides/citologia , Feto , Fígado , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/fisiologia
4.
Blood ; 101(5): 1984-6, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393498

RESUMO

The development of molecularly targeted treatments of adult leukemias warrants investigation of these targets in similar pediatric leukemias. The NF1 tumor suppressor gene, which encodes a GTPase activating protein for p21(ras), is frequently inactivated in juvenile myelomonocytic leukemia (JMML). Other patients with JMML acquire activating RAS gene mutations. Recipient mice reconstituted with Nf1-/- fetal hematopoietic cells develop a myeloproliferative disease (MPD) that models the human disease. JMML arises from clonal expansion of a hematopoietic stem cell, and JMML cells and murine Nf1-/- hematopoietic cells are hypersensitive to granulocyte macrophage-colony stimulating factor and KitL, the ligand for c-kit. We generated embryos doubly mutant for the Wv allele of c-kit and Nf1 to ask if reduction of c-kit activity would delay or prevent the development of MPD. Despite a reduction in c-kit activity to approximately 10% of wild-type levels, Nf1-/-;Wv/Wv cells induced MPD in recipient mice.


Assuntos
Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Genes da Neurofibromatose 1 , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Leucemia Mielomonocítica Aguda/genética , Transtornos Mieloproliferativos/etiologia , Neurofibromina 1/deficiência , Proteínas Proto-Oncogênicas c-kit/fisiologia , Alelos , Animais , Ensaio de Unidades Formadoras de Colônias , Cruzamentos Genéticos , Marcação de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-1/farmacologia , Interleucina-3/farmacologia , Fígado/citologia , Fígado/embriologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Proto-Oncogênicas c-kit/genética , Quimera por Radiação , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
5.
Blood ; 100(10): 3656-62, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393709

RESUMO

Ras plays an essential role in lymphocyte development and function. However, in vivo consequence(s) of regulation of Ras activity by guanosine triphosphatase (GTPase)-activating proteins (GAPs) on lymphocyte development and function are not known. In this study we demonstrate that neurofibromin, the protein encoded by the NF1 tumor suppressor gene functions as a GAP for Ras in T cells. Loss of Nf1 in T cells results in enhanced Ras activation, which is associated with thymic and splenic hyperplasia, and an increase in the absolute number of immature and mature T-cell subsets compared with control mice. Interestingly, in spite of a profound T-cell expansion and higher thymidine incorporation in unstimulated Nf1-deficient T cells, T-cell receptor and interleukin-2 receptor-mediated proliferation of thymocytes and mature T cells was substantially reduced compared with control mice. Collectively, these results identify neurofibromin as a GAP for Ras in T cells for maintaining immune homeostasis in vivo.


Assuntos
Transtornos Linfoproliferativos/metabolismo , Neurofibromina 1/fisiologia , Animais , Ativação Linfocitária , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/imunologia , Camundongos , Camundongos Mutantes , Neurofibromina 1/deficiência , Neurofibromina 1/imunologia , Baço/citologia , Subpopulações de Linfócitos T , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...