Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Hematol ; 36(5): 545-558, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18439488

RESUMO

OBJECTIVE: Homeostasis of the hematopoietic compartment is challenged and maintained during conditions of stress by mechanisms that are poorly defined. To understand how the bone marrow (BM) microenvironment influences hematopoiesis, we explored the role of Notch signaling and BM endothelial cells in providing microenvironmental cues to hematopoietic cells in the presence of inflammatory stimuli. MATERIALS AND METHODS: The human BM endothelial cell line (BMEC) and primary human BM endothelial cells were analyzed for expression of Notch ligands and the ability to expand hematopoietic progenitors in an in vitro coculture system. In vivo experiments were carried out to identify modulation of Notch signaling in BM endothelial and hematopoietic cells in mice challenged with tumor necrosis factor-alpha (TNF-alpha) or lipopolysaccharide (LPS), or in Tie2-tmTNF-alpha transgenic mice characterized by constitutive TNF-alpha activation. RESULTS: BM endothelial cells were found to express Jagged ligands and to greatly support progenitor's colony-forming ability. This effect was markedly decreased by Notch antagonists and augmented by increasing levels of Jagged2. Physiologic upregulation of Jagged2 expression on BMEC was observed upon TNF-alpha activation. Injection of TNF-alpha or LPS upregulated three- to fourfold Jagged2 expression on murine BM endothelial cells in vivo and resulted in increased Notch activation on murine hematopoietic stem/progenitor cells. Similarly, constitutive activation of endothelial cells in Tie2-tmTNF-alpha mice was characterized by increased expression of Jagged2 and by augmented Notch activation on hematopoietic stem/progenitor cells. CONCLUSIONS: Our results provide the first evidence that BM endothelial cells promote expansion of hematopoietic progenitor cells by a Notch-dependent mechanism and that TNF-alpha and LPS can modulate the levels of Notch ligand expression and Notch activation in the BM microenvironment in vivo.


Assuntos
Medula Óssea/imunologia , Células Endoteliais/imunologia , Inflamação/imunologia , Receptores Notch/metabolismo , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Medula Óssea/irrigação sanguínea , Medula Óssea/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-2 , Ligantes , Lipopolissacarídeos/farmacologia , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Receptores Notch/efeitos dos fármacos , Receptores Notch/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
2.
J Exp Med ; 202(1): 157-68, 2005 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-15998794

RESUMO

Cyclin-dependent kinase inhibitors (CKIs) and Notch receptor activation have been shown to influence adult stem cells and progenitors by altering stem cell self-renewal and proliferation. Yet, no interaction between these molecular pathways has been defined. Here we show that ligand-independent and ligand-dependent activation of Notch1 induces transcription of the S phase kinase-associated protein 2 (SKP2), the F-box subunit of the ubiquitin-ligase complex SCF(SKP2) that targets proteins for degradation. Up-regulation of SKP2 by Notch signaling enhances proteasome-mediated degradation of the CKIs, p27 Kip1 and p21 Cip1, and causes premature entry into S phase. Silencing of SKP2 by RNA interference in G1 stabilizes p27 Kip1 and p21 Cip1 and abolishes Notch effect on G1-S progression. Thus, SKP2 serves to link Notch1 activation with the cell cycle machinery. This novel pathway involving Notch/SKP2/CKIs connects a cell surface receptor with proximate mediators of cell cycle activity, and suggests a mechanism by which a known physiologic mediator of cell fate determination interfaces with cell cycle control.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fase G1/fisiologia , Receptores de Superfície Celular/metabolismo , Fase S/fisiologia , Proteínas Quinases Associadas a Fase S/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Células 3T3 , Animais , Sequência de Bases , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , DNA/genética , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Interferência de RNA , Receptor Notch1 , Transcrição Gênica , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...