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.
Retrovirology ; 9: 77, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-23009398

RESUMO

BACKGROUND: The Tax protein encoded by Human T-lymphotropic virus type 1 (HTLV-1) is a powerful activator of the NF-κB pathway, a property critical for HTLV-1-induced immortalization of CD4⁺ T lymphocytes. Tax permanently stimulates this pathway at a cytoplasmic level by activating the IκB kinase (IKK) complex and at a nuclear level by enhancing the binding of the NF-κB factor RelA to its cognate promoters and by forming nuclear bodies, believed to represent transcriptionally active structures. In previous studies, we reported that Tax ubiquitination and SUMOylation play a critical role in Tax localization and NF-κB activation. Indeed, analysis of lysine Tax mutants fused or not to ubiquitin or SUMO led us to propose a two-step model in which Tax ubiquitination first intervenes to activate IKK while Tax SUMOylation is subsequently required for promoter activation within Tax nuclear bodies. However, recent studies showing that ubiquitin or SUMO can modulate Tax activities in either the nucleus or the cytoplasm and that SUMOylated Tax can serve as substrate for ubiquitination suggested that Tax ubiquitination and SUMOylation may mediate redundant rather than successive functions. RESULTS: In this study, we analyzed the properties of a new Tax mutant that is properly ubiquitinated, but defective for both nuclear body formation and SUMOylation. We report that reducing Tax SUMOylation and nuclear body formation do not alter the ability of Tax to activate IKK, induce RelA nuclear translocation, and trigger gene expression from a NF-κB promoter. Importantly, potent NF-κB promoter activation by Tax despite low SUMOylation and nuclear body formation is also observed in T cells, including CD4⁺ primary T lymphocytes. Moreover, we show that Tax nuclear bodies are hardly observed in HTLV-1-infected T cells. Finally, we provide direct evidence that the degree of NF-κB activation by Tax correlates with the level of Tax ubiquitination, but not SUMOylation. CONCLUSIONS: These data reveal that the formation of Tax nuclear bodies, previously associated to transcriptional activities in Tax-transfected cells, is dispensable for NF-κB promoter activation, notably in CD4⁺ T cells. They also provide the first evidence that Tax SUMOylation is not a key determinant for Tax-induced NF-κB activation.


Assuntos
Produtos do Gene tax/metabolismo , Espaço Intranuclear/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Ativação Transcricional , Substituição de Aminoácidos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Produtos do Gene tax/genética , Genes Reporter , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Quinase I-kappa B/metabolismo , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Microscopia Confocal , NF-kappa B/fisiologia , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteína SUMO-1/metabolismo , Transdução de Sinais , Sumoilação , Transcrição Gênica , Ubiquitinação
2.
J Cell Sci ; 125(Pt 4): 993-1002, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22399799

RESUMO

Gfi-1B is a transcriptional repressor essential for the regulation of erythropoiesis and megakaryopoiesis. Here we identify Gfi-1B p32, a Gfi-1B isoform, as essential for erythroid differentiation. Gfi-1B p32 is generated by alternative splicing and lacks the two first zinc finger domains of the protein. Selective knock down of Gfi-1B p32 compromises erythroid differentiation, whereas its ectopic expression induces erythropoiesis in the absence of erythropoietin. Gfi-1B p32 isoform binds to Gfi-1B target gene promoters and associates with the LSD1-CoREST repressor complex more efficiently than the major Gfi-1B p37 isoform. Furthermore, we show that Gfi-1B includes a KSKK motif in its SNAG domain, which recruits the repressor complex only when dimethylated on lysine 8. Mutation of lysine 8 prevents Gfi-1B p32-induced erythroid development. Our results thus highlight a key role for the alternatively spliced Gfi-1B p32 isoform in erythroid development.


Assuntos
Eritropoese , Histona Desmetilases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Processamento Alternativo , Motivos de Aminoácidos , Linhagem Celular , Proteínas Correpressoras , Eritropoese/genética , Eritropoetina , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lisina/metabolismo , Metilação , Peso Molecular , Regiões Promotoras Genéticas/genética , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Dedos de Zinco
3.
Blood ; 115(14): 2784-95, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20124515

RESUMO

Growth factor independence-1B (Gfi-1B) is a transcriptional repressor essential for erythropoiesis and megakaryopoiesis. Targeted gene disruption of GFI1B in mice leads to embryonic lethality resulting from failure to produce definitive erythrocytes, hindering the study of Gfi-1B function in adult hematopoiesis. We here show that, in humans, Gfi-1B controls the development of erythrocytes and megakaryocytes by regulating the proliferation and differentiation of bipotent erythro-megakaryocytic progenitors. We further identify in this cell population the type III transforming growth factor-beta receptor gene, TGFBR3, as a direct target of Gfi-1B. Knockdown of Gfi-1B results in altered transforming growth factor-beta (TGF-beta) signaling as shown by the increase in Smad2 phosphorylation and its inability to associate to the transcription intermediary factor 1-gamma (TIF1-gamma). Because the Smad2/TIF1-gamma complex is known to specifically regulate erythroid differentiation, we propose that, by repressing TGF-beta type III receptor (TbetaRIotaII) expression, Gfi-1B favors the Smad2/TIF1-gamma interaction downstream of TGF-beta signaling, allowing immature progenitors to differentiate toward the erythroid lineage.


Assuntos
Diferenciação Celular/fisiologia , Eritrócitos/metabolismo , Megacariócitos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Adulto , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células , Eritrócitos/citologia , Eritropoese/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Células K562 , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Megacariócitos/citologia , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação/fisiologia , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Trombopoese/fisiologia
4.
Blood ; 115(3): 687-95, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19965638

RESUMO

Gfi-1B is a transcriptional repressor that is crucial for erythroid differentiation: inactivation of the GFI1B gene in mice leads to embryonic death due to failure to produce differentiated red cells. Accordingly, GFI1B expression is tightly regulated during erythropoiesis, but the mechanisms involved in such regulation remain partially understood. We here identify HMGB2, a high-mobility group HMG protein, as a key regulator of GFI1B transcription. HMGB2 binds to the GFI1B promoter in vivo and up-regulates its trans-activation most likely by enhancing the binding of Oct-1 and, to a lesser extent, of GATA-1 and NF-Y to the GFI1B promoter. HMGB2 expression increases during erythroid differentiation concomitantly to the increase of GfI1B transcription. Importantly, knockdown of HMGB2 in immature hematopoietic progenitor cells leads to decreased Gfi-1B expression and impairs their erythroid differentiation. We propose that HMGB2 potentiates GATA-1-dependent transcription of GFI1B by Oct-1 and thereby controls erythroid differentiation.


Assuntos
Diferenciação Celular/genética , Eritrócitos/fisiologia , Proteína HMGB2/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA1/fisiologia , Proteína HMGB2/antagonistas & inibidores , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Fator 1 de Transcrição de Octâmero/metabolismo , Fator 1 de Transcrição de Octâmero/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/metabolismo , Ativação Transcricional/efeitos dos fármacos
5.
Stem Cells ; 27(9): 2153-62, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19522008

RESUMO

Growth Factor Independent-1B (Gfi-1B) is a transcriptional repressor that plays critical roles in the control of erythropoiesis and megakaryopoiesis. Gfi-1B expression was described to be repressed by an autoregulatory feedback control loop. Here, we show that Gfi-1 transcription is positively regulated early after induction of erythroid differentiation and remains highly active to late erythroblasts. Using chromatin immunoprecipitation assays in CD34+ cells from human cord blood, we found that Gfi-1 and GATA-2 in immature progenitors and then Gfi-1B and GATA-1 in erythroblasts are bound to the Gfi-1B promoter as well as to the promoter of c-myc, a known Gfi-1B target gene. Surprisingly, this Gfi-1/GATA-2-Gfi-1B/GATA-1 switch observed at erythroblast stages is associated to an increase in the Gfi-1B transcription whereas it triggers repression of c-myc transcription. Accordingly, analysis of chromatin modification patterns shows that HDAC, CoREST, and LSD1 are recruited to the c-myc promoter leading to appearance of repressive chromatin marks. In contrast, the Gfi-1B promoter remains associated with a transcriptionally active chromatin configuration as highlighted by an increase in histone H3 acetylation and concomitant release of the LSD1 and CoREST corepressors. The repressive function of Gfi-1B therefore depends on the nature of the proteins recruited to the target gene promoters and on chromatin modifications. We conclude that Gfi-1B behaves as a lineage-affiliated gene with an open chromatin configuration in multipotent progenitors and sustained activation as cells progress throughout erythroid differentiation.


Assuntos
Cromatina/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Western Blotting , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas Correpressoras , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Vetores Genéticos , Histona Desmetilases/metabolismo , Humanos , Lentivirus , Proteínas do Tecido Nervoso/metabolismo , Reação em Cadeia da Polimerase , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA