Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Nat Commun ; 10(1): 632, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733432

RESUMO

To reveal how cells exit human pluripotency, we designed a CRISPR-Cas9 screen exploiting the metabolic and epigenetic differences between naïve and primed pluripotent cells. We identify the tumor suppressor, Folliculin(FLCN) as a critical gene required for the exit from human pluripotency. Here we show that FLCN Knock-out (KO) hESCs maintain the naïve pluripotent state but cannot exit the state since the critical transcription factor TFE3 remains active in the nucleus. TFE3 targets up-regulated in FLCN KO exit assay are members of Wnt pathway and ESRRB. Treatment of FLCN KO hESC with a Wnt inhibitor, but not ESRRB/FLCN double mutant, rescues the cells, allowing the exit from the naïve state. Using co-immunoprecipitation and mass spectrometry analysis we identify unique FLCN binding partners. The interactions of FLCN with components of the mTOR pathway (mTORC1 and mTORC2) reveal a mechanism of FLCN function during exit from naïve pluripotency.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Via de Sinalização Wnt/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiologia , Linhagem Celular , Estrona/genética , Estrona/metabolismo , Humanos , Imunoprecipitação , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Proteômica , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Via de Sinalização Wnt/genética
2.
Oncogene ; 34(46): 5739-48, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25728676

RESUMO

The resistance of melanoma to current treatment modalities represents a major obstacle for durable therapeutic response, and thus the elucidation of mechanisms of resistance is urgently needed. The crucial functions of activating transcription factor-2 (ATF2) in the development and therapeutic resistance of melanoma have been previously reported, although the precise underlying mechanisms remain unclear. Here, we report a protein kinase C-ɛ (PKCɛ)- and ATF2-mediated mechanism that facilitates resistance by transcriptionally repressing the expression of interferon-ß1 (IFNß1) and downstream type-I IFN signaling that is otherwise induced upon exposure to chemotherapy. Treatment of early-stage melanomas expressing low levels of PKCɛ with chemotherapies relieves ATF2-mediated transcriptional repression of IFNß1, resulting in impaired S-phase progression, a senescence-like phenotype and increased cell death. This response is lost in late-stage metastatic melanomas expressing high levels of PKCɛ. Notably, nuclear ATF2 and low expression of IFNß1 in melanoma tumor samples correlates with poor patient responsiveness to biochemotherapy or neoadjuvant IFN-α2a. Conversely, cytosolic ATF2 and induction of IFNß1 coincides with therapeutic responsiveness. Collectively, we identify an IFNß1-dependent, cell-autonomous mechanism that contributes to the therapeutic resistance of melanoma via the PKCɛ-ATF2 regulatory axis.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Resistencia a Medicamentos Antineoplásicos , Interferon beta/genética , Melanoma/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Regulação para Baixo , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melanoma/patologia , Regiões Promotoras Genéticas , Proteína Quinase C-épsilon/metabolismo , Transdução de Sinais , Transcrição Gênica
3.
Oncogene ; 31(1): 93-103, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21666715

RESUMO

Human breast tumors comprise a minor sub-population of tumor-initiating cells (TICs), commonly termed cancer stem cells. TICs are thought to sustain tumor growth and to confer resistance to current anticancer therapies. Hence, targeting TIC may be essential to achieving durable cancer cures. To identify molecular targets in breast TIC, we employed a transgenic mouse model of ERBB2 breast cancer; tumors arising in this model comprise a very high frequency of TIC, which is maintained in tumor cell populations propagated in vitro as non-adherent tumorspheres. The Notch pathway is dysregulated in human breast tumors and overexpression of constitutively active Notch proteins induces mammary tumors in mice. The Notch pathway has also been implicated in stem cell processes including those of mammary epithelial stem cells. Hence, we investigated the potential that the Notch pathway is required for TIC activity. We found that an antagonist of Notch signaling, a gamma (γ)-secretase inhibitor termed MRK-003, inhibited the survival of tumorsphere-derived cells in vitro and eliminated TIC as assessed by cell transplantation into syngeneic mice. Whereas MRK-003 also inhibited the self-renewal and/or proliferation of mammosphere-resident cells, this effect of the inhibitor was reversible thus suggesting that it did not compromise the survival of these cells. MRK-003 administration to tumor-bearing mice eliminated tumor-resident TIC and resulted in rapid and durable tumor regression. MRK-003 inhibited the proliferation of tumor cells, and induced their apoptosis and differentiation. These findings suggest that MRK-003 targets breast TIC and illustrate that eradicating these cells in breast tumors ensures long-term, recurrence-free survival.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Óxidos S-Cíclicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Genes erbB-2 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Tiadiazóis/uso terapêutico , Animais , Óxidos S-Cíclicos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Perfilação da Expressão Gênica , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Receptores Notch/fisiologia , Tiadiazóis/farmacologia
4.
Br J Pharmacol ; 158(5): 1183-95, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19775282

RESUMO

BACKGROUND AND PURPOSE: gamma-Secretase inhibitors (GSIs) block NOTCH receptor cleavage and pathway activation and have been under clinical evaluation for the treatment of malignancies such as T-cell acute lymphoblastic leukaemia (T-ALL). The ability of GSIs to decrease T-ALL cell viability in vitro is a slow process requiring >8 days, however, such treatment durations are not well tolerated in vivo. Here we study GSI's effect on tumour and normal cellular processes to optimize dosing regimens for anti-tumour efficacy. EXPERIMENTAL APPROACH: Inhibition of the Notch pathway in mouse intestinal epithelium was used to evaluate the effect of GSIs and guide the design of dosing regimens for xenograft models. Serum Abeta(40) and Notch target gene modulation in tumours were used to evaluate the degree and duration of target inhibition. Pharmacokinetic and pharmacodynamic correlations with biochemical, immunohistochemical and profiling data were used to demonstrate GSI mechanism of action in xenograft tumours. KEY RESULTS: Three days of >70% Notch pathway inhibition was sufficient to provide an anti-tumour effect and was well tolerated. GSI-induced conversion of mouse epithelial cells to a secretory lineage was time- and dose-dependent. Anti-tumour efficacy was associated with cell cycle arrest and apoptosis that was in part due to Notch-dependent regulation of mitochondrial homeostasis. CONCLUSIONS AND IMPLICATIONS: Intermittent but potent inhibition of Notch signalling is sufficient for anti-tumour efficacy in these T-ALL models. These findings provide support for the use of GSI in Notch-dependent malignancies and that clinical benefits may be derived from transient but potent inhibition of Notch.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Antineoplásicos/farmacologia , Óxidos S-Cíclicos/farmacologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Receptor Notch1/fisiologia , Tiadiazóis/farmacologia , Peptídeos beta-Amiloides/sangue , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Apoptose , Diferenciação Celular , Linhagem Celular Tumoral , Colo/citologia , Colo/efeitos dos fármacos , Óxidos S-Cíclicos/administração & dosagem , Óxidos S-Cíclicos/efeitos adversos , Regulação para Baixo , Esquema de Medicação , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Nus , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Transplante de Neoplasias , Fragmentos de Peptídeos/sangue , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Receptor Notch1/genética , Transdução de Sinais , Tiadiazóis/administração & dosagem , Tiadiazóis/efeitos adversos , Transplante Heterólogo
5.
Neuroscience ; 118(4): 881-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12732234

RESUMO

For the last 100 years secretin has been extensively studied for its hormonal effects on digestion. Recent observations that the deficits in social reciprocity skills seen in young (3-4-year-old) autistic children are improved after secretin infusions suggest an additional influence on neuronal activity. We show here that i.v. administration of secretin in rats induces Fos protein expression in the neurons of the central amygdala as well as the area postrema, bed nucleus of the stria terminalis, external lateral parabrachial nucleus and supraoptic nucleus. However, secretin infusion did not induce Fos expression in the solitary tract nucleus or paraventricular nucleus, regions normally activated by related peptides such as cholecystokinin. The peak blood levels of secretin that induce Fos protein expression in rat brain are similar to the peak blood levels observed during i.v. treatment with secretin in humans. The amygdala is known to be critical for developing reciprocal social interaction skills and abnormalities in this brain region have been demonstrated in autistic children.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Secretina/administração & dosagem , Sincalida/análogos & derivados , Tonsila do Cerebelo/metabolismo , Animais , Área Postrema/metabolismo , Área Sob a Curva , Humanos , Imuno-Histoquímica/métodos , Infusões Intravenosas , Masculino , Neurônios/metabolismo , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Peptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Ponte/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Secretina/farmacologia , Núcleos Septais/metabolismo , Sincalida/farmacologia , Núcleo Supraóptico/metabolismo , Fatores de Tempo , Peptídeo Intestinal Vasoativo/farmacologia
7.
J Immunol ; 167(11): 6330-7, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11714797

RESUMO

LIGHT, a member of the TNF family of cytokines (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for herpesvirus entry mediator, a receptor expressed on T cells), is induced on activated T cells and mediates costimulatory and antitumor activity in vitro. Relatively little information is available on the in vivo effects of LIGHT expression, particularly within the T cell compartment. In this work, we describe transgenic mice that express human LIGHT under the control of the CD2 promoter, resulting in constitutive transgene expression in cells of the T lymphocyte lineage. LIGHT-transgenic animals exhibit abnormalities in both lymphoid tissue architecture and the distribution of lymphocyte subsets. They also show signs of inflammation that are most severe in the intestine, along with tissue destruction of the reproductive organs. These LIGHT-mediated effects were recapitulated when immune-deficient mice were reconstituted with bone marrow from LIGHT-transgenic donor mice. T cells in the LIGHT-transgenic mice have an activated phenotype and mucosal T cells exhibit enhanced Th1 cytokine activity. The results indicate that LIGHT may function as an important regulator of T cell activation, and implicate LIGHT signaling pathways in inflammation focused on mucosal tissues.


Assuntos
Regulação da Expressão Gênica/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Tecido Linfoide/patologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Linhagem Celular , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Feminino , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hibridomas , Infertilidade Feminina/genética , Infertilidade Feminina/imunologia , Infertilidade Feminina/fisiopatologia , Inflamação/genética , Inflamação/imunologia , Inflamação/mortalidade , Inflamação/patologia , Tecido Linfoide/imunologia , Receptor beta de Linfotoxina , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Ligação Proteica/genética , Ligação Proteica/imunologia , Quimera por Radiação/genética , Quimera por Radiação/imunologia , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral , Receptores Virais/antagonistas & inibidores , Receptores Virais/metabolismo , Análise de Sobrevida , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
8.
Neuron ; 31(6): 913-27, 2001 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-11580893

RESUMO

Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant disorder caused by a CAG repeat expansion. To determine the mechanism of neurotoxicity, we produced transgenic mice and observed a cone-rod dystrophy. Nuclear inclusions were present, suggesting that the disease pathway involves the nucleus. When yeast two-hybrid assays indicated that cone-rod homeobox protein (CRX) interacts with ataxin-7, we performed further studies to assess this interaction. We found that ataxin-7 and CRX colocalize and coimmunoprecipitate. We observed that polyglutamine-expanded ataxin-7 can dramatically suppress CRX transactivation. In SCA7 transgenic mice, electrophoretic mobility shift assays indicated reduced CRX binding activity, while RT-PCR analysis detected reductions in CRX-regulated genes. Our results suggest that CRX transcription interference accounts for the retinal degeneration in SCA7 and thus may provide an explanation for how cell-type specificity is achieved in this polyglutamine repeat disease.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Peptídeos/química , Transativadores/antagonistas & inibidores , Repetições de Trinucleotídeos , Fatores Etários , Animais , Ataxina-7 , Linhagem Celular , Núcleo Celular/ultraestrutura , Modelos Animais de Doenças , Eletrorretinografia , Proteínas do Olho/química , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Perfilação da Expressão Gênica , Genes Sintéticos , Proteínas de Homeodomínio/fisiologia , Humanos , Substâncias Macromoleculares , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Príons/genética , Regiões Promotoras Genéticas , Ligação Proteica , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Transmissão Sináptica , Transativadores/fisiologia , Ativação Transcricional , Transfecção , Transgenes , Técnicas do Sistema de Duplo-Híbrido
9.
Virology ; 289(1): 1-5, 2001 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11601911

RESUMO

Herpesviruses appear to peacefully coexist with their natural hosts, with infection typically manifested as a benign, but lifelong process. However, coexistence depends on active resistance by innate and specific immune defenses as revealed in the striking virulence of herpesviruses when immunity fails. This pattern of infection is characteristic of a viral pathogen, such as cytomegalovirus, that has evolved efficient strategies targeted at host defense systems. Targeting members of the tumor necrosis factor (TNF)/lymphotoxin (LT) superfamily of cytokines is a strategy found in all herpesviruses, which suggests the existence of an intimate evolutionary link in their host-parasite relationship. Here we examine some of the strategies used by herpesvirus that target members of the TNF superfamily and discuss a recent study that revealed a novel mechanism that links LT-related ligands and interferons (IFN) to the establishment of coexistence between herpesvirus and its host cell.


Assuntos
Herpesviridae/fisiologia , Linfotoxina-alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Evolução Biológica , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Humanos
10.
J Immunol ; 167(9): 5122-8, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11673523

RESUMO

LIGHT is a member of the TNF cytokine superfamily that signals through the lymphotoxin (LT)beta receptor and the herpesvirus entry mediator. LIGHT may function as a costimulatory factor for the activation of lymphoid cells and as a deterrent to infection by herpesvirus, which may provide significant selective pressure shaping the evolution of LIGHT. Here, we define the molecular genetics of the human LIGHT locus, revealing its close linkage to the TNF superfamily members CD27 ligand and 4-1BB ligand, and the third complement protein (C3), which positions LIGHT within the MHC paralog on chromosome 19p13.3. An alternately spliced isoform of LIGHT mRNA that encodes a transmembrane-deleted form is detected in activated T cells and gives rise to a nonglycosylated protein that resides in the cytosol. Furthermore, membrane LIGHT is shed from the cell surface of human 293 T cells. These studies reveal new mechanisms involved in regulating the physical forms and cellular compartmentalization of LIGHT that may contribute to the regulation and biological function of this cytokine.


Assuntos
Processamento Alternativo , Antígenos CD , Mapeamento Cromossômico , Cromossomos Humanos Par 9 , Ligação Genética , Proteínas de Membrana/genética , Fator de Necrose Tumoral alfa/genética , Ligante 4-1BB , Sequência de Aminoácidos , Ligante CD27 , Humanos , Proteínas de Membrana/química , Dados de Sequência Molecular , Isoformas de Proteínas , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/química
11.
Immunity ; 15(4): 617-26, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11672543

RESUMO

Tumor necrosis factor (TNF)-related cytokines regulate cell death and survival and provide strong selective pressures for viruses, such as cytomegalovirus (CMV), to evolve counterstrategies in order to persist in immune-competent hosts. Signaling by the lymphotoxin (LT)-beta receptor or TNF receptor-1, but not Fas or TRAIL receptors, inhibits the cytopathicity and replication of human CMV by a nonapoptotic, reversible process that requires nuclear factor kappa B (NF-kappa B)-dependent induction of interferon-beta (IFN-beta). Efficient induction of IFN-beta requires virus infection and LT signaling, demonstrating the need for both host and viral factors in the curtailment of viral replication without cellular elimination. LT alpha-deficient mice and LT beta R-Fc transgenic mice were profoundly susceptible to murine CMV infection. Together, these results reveal an essential and conserved role for LTs in establishing host defense to CMV.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Citomegalovirus/fisiologia , Interações Hospedeiro-Parasita , Interferon beta/biossíntese , Linfotoxina-alfa/farmacologia , Proteínas de Membrana/farmacologia , Ativação Transcricional , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas de Transporte/fisiologia , Células Cultivadas , Citomegalovirus/crescimento & desenvolvimento , Citomegalovirus/patogenicidade , Proteína de Domínio de Morte Associada a Fas , Infecções por Herpesviridae/etiologia , Humanos , Interferon beta/genética , Interferon beta/fisiologia , Receptor beta de Linfotoxina , Linfotoxina-alfa/genética , Camundongos , Camundongos Transgênicos , Muromegalovirus , NF-kappa B/fisiologia , Proteínas/fisiologia , RNA Mensageiro/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Taxa de Sobrevida , Fator 3 Associado a Receptor de TNF , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Replicação Viral/efeitos dos fármacos
12.
Am J Pathol ; 159(4): 1397-404, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11583967

RESUMO

Activated macrophages (M(phi)) isolated from inflamed glomeruli or generated by interferon-gamma and lipopolysaccharide treatment in vitro induce glomerular mesangial cell apoptosis by hitherto incompletely understood mechanisms. In this report we demonstrate that nitric oxide-independent killing of co-cultured mesangial cells by interferon-gamma/lipopolysaccharide-activated M(phi) is suppressed by binding/ingestion of apoptotic cells and is mediated by tumor necrosis factor (TNF). Thus, soluble TNF receptor-1 significantly inhibited induction of mesangial cell apoptosis by 1) rodent M(phi) in the presence of nitric oxide synthase inhibitors or 2) human M(phi), both situations in which nitric oxide release was minimal. Furthermore, murine TNF knockout M(phi) were completely unable to induce mesangial cell apoptosis in the presence of nitric oxide synthase inhibitors. We conclude that TNF-restricted M(phi)-directed apoptosis of glomerular mesangial cells can be down-regulated by M(phi) binding/ingestion of apoptotic cells, suggesting a new mechanism for negative feedback regulation of M(phi) controls on resident cell number at inflamed sites.


Assuntos
Apoptose/fisiologia , Mesângio Glomerular/fisiologia , Macrófagos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antígenos CD , Técnicas de Cocultura , Mesângio Glomerular/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/fisiologia , Ratos , Ratos Wistar , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/genética
13.
J Immunol ; 167(5): 2511-21, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11509590

RESUMO

During embryogenesis, the Peyer's patch anlagen are induced by a cell population that produces lymphotoxin (LT) alpha(1)beta(2) following stimulation of IL-7Ralpha. In this study, we show that the LT-producing cell is localized within the IL-7Ralpha(+) and integrin alpha(4)beta(7) (alpha(4)beta(7))(+) population in the embryonic intestine. Lineage commitment to the LT producer phenotype in the fetal liver coincides with expression of alpha(4)beta(7). Before expression of alpha(4)beta(7), the potential of IL-7Ralpha(+) population to generate B cells is lost. However, the progenitors for T cells and LT producer cells reside in the IL-7Ralpha(+)alpha(4)beta(7)(+) cells, but during subsequent differentiation, the potential to give rise to T cells is lost. This IL-7Ralpha(+)alpha(4)beta(7)(+) population migrates to the intestine, where it induces the Peyer's patch anlagen. When stimulated with IL-15 or IL-3 and TNF, the intestinal IL-7Ralpha(+)alpha(4)beta(7)(+) population can differentiate into fully competent NK1.1(+) NK cells or CD11c(+) APCs. Expression of alpha(4)beta(7) is lost during differentiation of both lineages; IL-7Ralpha expression is lost during NK1.1(+) cells differentiation. A newly discovered lineage(-)IL-7Ralpha(+)c-Kit(+)alpha(4)beta(7)(+) population in the fetal liver is committed to T, NK, dendritic, and fetal intestinal LT producer lineage, the latter being an intermediate stage during differentiation of NK and dendritic cells.


Assuntos
Integrinas/metabolismo , Nódulos Linfáticos Agregados/embriologia , Nódulos Linfáticos Agregados/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia , Animais , Diferenciação Celular , Movimento Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Integrinas/genética , Interleucina-15/farmacologia , Interleucina-3/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Linfotoxina-alfa/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/citologia , Gravidez , Receptores de Interleucina-7/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia
14.
J Biol Chem ; 276(40): 37426-30, 2001 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-11448951

RESUMO

The bioactivity of tumor necrosis factor (TNF) is mediated by two TNF receptors (TNF-Rs), more particularly TNF-RI and TNF-RII. Although human TNF (hTNF) and murine TNF (mTNF) are very homologous, hTNF binds only to mTNF-RI. By measuring the binding of a panel of mTNF/hTNF chimeras to both mTNF-R, we pinpointed the TNF region that mediates the interaction with mTNF-RII. Using site-specific mutagenesis, we identified amino acids 71-73 and 89 as the main interacting residues. Mutein hTNF-S71D/T72Y/H73 Delta/T89E interacts with both types of mTNF-R and is active in CT6 cell proliferation assays mediated by mTNF-RII. Mutein mTNF-D71S/Y72T/Delta 73H/E89T binds to mTNF-RI only and is no longer active on CT6 cells. However, the L929s cytotoxicity of this mutein (an effect mediated by mTNF-RI triggering) was also 100-fold lower than that of wild-type mTNF due to enhanced dissociation during incubation at subnanomolar concentrations. The additional mutation of amino acid 102, resulting in the mutein mTNF-D71S/Y72T/Delta 73H/E89T/P102Q, restored the trimer stability, which led to an enhanced specific activity on L929s cells. Hence the specific activity of a TNF species is governed not only by its receptor binding characteristics but also by its trimer stability after incubation at subnanomolar concentrations. In conclusion, the mutation of TNF amino acids 71-73, 89, and 102 is sufficient to obtain a mTNF mutein selective for mTNF-RI and a hTNF mutein that, unlike wild-type hTNF, also acts on mTNF-RII.


Assuntos
Antígenos CD/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Cromatografia em Gel , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/genética
16.
Virology ; 283(2): 178-87, 2001 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11336543

RESUMO

Hepatitis C virus (HCV) core protein has been shown to interact with the death domain (DD) of tumor necrosis factor receptor-1 (TNFR1). In this study, we further examined the interaction of the core protein with the signaling molecules of TNFR1, including FADD, TRADD, and TRAF2, in a human embryonic kidney cell line, HEK-293, that overexpresses the HCV core protein. This core protein-expressing cell line exhibited enhanced sensitivity to TNF-induced apoptosis. By in vitro binding and in vivo coimmunoprecipitation assays, we showed that the HCV core protein interacted with the DD of FADD and enhanced apoptosis induced by FADD overexpression. This enhancement could be blocked by a dominant-negative mutant of FADD. In contrast, the core protein did not directly interact with the DD of TRADD, but could disrupt the binding of TRADD to TNFR1. TRAF2 recruitment to the TNFR1 signaling complex was also disrupted by the core protein. Correspondingly, TRAF2-dependent activation of the protein kinase JNK was suppressed in the core protein-expressing cells. However, NF kappa B activation by TNF was not significantly altered by the HCV core protein, suggesting the existence of TRAF2-independent pathways for NF kappa B activation. These results combined indicate that the HCV core protein sensitizes cells to TNF-induced apoptosis primarily by facilitating FADD recruitment to TNFR1. The inhibition of JNK activation by the HCV core protein may also contribute to the increased propensity of cells for apoptosis. These results, in comparison with other published studies, suggest that the effects of the HCV core protein and their underlying mechanisms vary significantly among cells of different origins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Hepacivirus , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral , Proteínas do Core Viral/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proteína de Domínio de Morte Associada a Fas , Regulação da Expressão Gênica , Humanos , MAP Quinase Quinase 4 , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas/genética , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Proteína de Domínio de Morte Associada a Receptor de TNF , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF , Transfecção , Proteínas do Core Viral/genética
17.
Oncogene ; 20(7): 812-8, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11314015

RESUMO

Radiation induces apoptosis of crypt intestinal epithelial cells (IEC) through a pathway that is largely dependent on p53. However, exactly how p53 mediates IEC apoptosis is unclear. Studies in vitro suggest that one mechanism by which p53 mediates apoptosis is through its ability to transactivate members of the TNF receptor family of 'Death Receptors'. Here, we examined the role of one of its member, TNF receptor type 1 (TNFR1), in an in vivo model of p53-dependent radiation-induced IEC apoptosis. We demonstrate that mice genetically engineered to be deficient in TNF receptor type 1 (TNFR1(-/-)) and mice injected with TNFR1-fusion chimeric protein (TNFR1-Fc; a competitive inhibitor of TNFR1) were partially protected (30-40%) from p53-dependent radiation-induced IEC apoptosis. However, we found no evidence to support the possibility p53 transcriptionally regulates the expression of TNFR1 nor increases the susceptibility of IEC to TNF-mediated apoptosis. Interestingly, we found that injection of TNF readily induced IEC apoptosis and that radiation induced a p53-dependent increase in the intestinal level of TNF. Furthermore, injection of a neutralizing anti-TNF mAb reduced p53-dependent radiation-induced IEC apoptosis by approximately 60%. Overall, these results suggest that p53-dependent radiation-induced IEC apoptosis is mediated in part through ability of p53 to regulate TNF, which subsequently induces IEC apoptosis through TNFR1.


Assuntos
Antígenos CD/metabolismo , Apoptose , Mucosa Intestinal/efeitos da radiação , Intestino Delgado/efeitos da radiação , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Relação Dose-Resposta a Droga , Raios gama , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores Tipo I de Fatores de Necrose Tumoral , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia , Proteína Supressora de Tumor p53/genética
18.
J Biol Chem ; 276(26): 24242-52, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11279055

RESUMO

We have identified three new tumor necrosis factor-receptor associated factor (TRAF) domain-containing proteins in humans using bioinformatics approaches, including: MUL, the product of the causative gene in Mulibrey Nanism syndrome; USP7 (HAUSP), an ubiquitin protease; and SPOP, a POZ domain-containing protein. Unlike classical TRAF family proteins involved in TNF family receptor (TNFR) signaling, the TRAF domains (TDs) of MUL, USP7, and SPOP are located near the NH(2) termini or central region of these proteins, rather than carboxyl end. MUL and USP7 are capable of binding in vitro via their TDs to all of the previously identified TRAF family proteins (TRAF1, TRAF2, TRAF3, TRAF4, TRAF5, and TRAF6), whereas the TD of SPOP interacts weakly with TRAF1 and TRAF6 only. The TD of MUL also interacted with itself, whereas the TDs of USP7 and SPOP did not self-associate. Analysis of various MUL and USP7 mutants by transient transfection assays indicated that the TDs of these proteins are necessary and sufficient for suppressing NF-kappaB induction by TRAF2 and TRAF6 as well as certain TRAF-binding TNF family receptors. In contrast, the TD of SPOP did not inhibit NF-kappaB induction. Immunofluorescence confocal microscopy indicated that MUL localizes to cytosolic bodies, with targeting to these structures mediated by a RBCC tripartite domain within the MUL protein. USP7 localized predominantly to the nucleus, in a TD-dependent manner. Data base searches revealed multiple proteins containing TDs homologous to those found in MUL, USP7, and SPOP throughout eukaryotes, including yeast, protists, plants, invertebrates, and mammals, suggesting that this branch of the TD family arose from an ancient gene. We propose the moniker TEFs (TD-encompassing factors) for this large family of proteins.


Assuntos
Endopeptidases/química , Proteínas Nucleares/química , Sequência de Aminoácidos , Animais , Biologia Computacional , Citosol/metabolismo , Endopeptidases/genética , Endopeptidases/fisiologia , Evolução Molecular , Humanos , Células Jurkat , Dados de Sequência Molecular , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Filogenia , Estrutura Terciária de Proteína , Proteínas/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Repressoras , Homologia de Sequência de Aminoácidos , Fator 1 Associado a Receptor de TNF , Proteínas com Motivo Tripartido , Ubiquitina Tiolesterase , Ubiquitina-Proteína Ligases , Peptidase 7 Específica de Ubiquitina
19.
J Biol Chem ; 276(5): 3270-8, 2001 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-11050095

RESUMO

Adenovirus encodes multiple gene products that regulate proapoptotic cellular responses to viral infection mediated by both the innate and adaptive immune systems. The E3-10.4K and 14.5K gene products are known to modulate the death receptor Fas. In this study, we demonstrate that an additional viral E3 protein, 6.7K, functions in the specific modulation of the two death receptors for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The 6.7K protein is expressed on the cell surface and forms a complex with the 10.4K and 14.5K proteins, and this complex is sufficient to induce down-modulation of TRAIL receptor-1 and -2 from the cell surface and reverse the sensitivity of infected cells to TRAIL-mediated apoptosis. Down-modulation of TRAIL-R2 by the E3 complex is dependent on the cytoplasmic tail of the receptor, but the death domain alone is not sufficient. These results identify a mechanism for viral modulation of TRAIL receptor-mediated apoptosis and suggest the E3 protein complex has evolved to regulate the signaling of selected cytokine receptors.


Assuntos
Proteínas E3 de Adenovirus/farmacologia , Apoptose , Proteínas de Membrana , Receptores do Fator de Necrose Tumoral/metabolismo , Adenoviridae/metabolismo , Proteínas E3 de Adenovirus/metabolismo , Regulação para Baixo , Proteína Ligante Fas , Células HT29/virologia , Humanos , Glicoproteínas de Membrana/metabolismo , Receptores de Citocinas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Transdução de Sinais , Frações Subcelulares , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA