Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 288(33): 23814-22, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23814052

RESUMO

The mechanisms of generation of the antineoplastic effects of interferons (IFNs) in malignant hematopoietic cells remain to be precisely defined. We examined the activation of type I IFN-dependent signaling pathways in malignant cells transformed by Jak2V617F, a critical pathogenic mutation in myeloproliferative neoplasms (MPNs). Our studies demonstrate that during engagement of the type I IFN receptor (IFNAR), there is activation of Jak-Stat pathways and also engagement of Mnk kinases. Activation of Mnk kinases is regulated by the Mek/Erk pathway and is required for the generation of IFN-induced growth inhibitory responses, but Mnk kinase activation does not modulate IFN-regulated Jak-Stat signals. We demonstrate that for type I IFNs to exert suppressive effects in malignant hematopoietic progenitors from patients with polycythemia vera, induction of Mnk kinase activity is required, as evidenced by studies involving pharmacological inhibition of Mnk or siRNA-mediated Mnk knockdown. Altogether, these findings provide evidence for key and essential roles of the Mnk kinase pathway in the generation of the antineoplastic effects of type I IFNs in Jak2V617F-dependent MPNs.


Assuntos
Neoplasias da Medula Óssea/metabolismo , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transtornos Mieloproliferativos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Neoplasias da Medula Óssea/patologia , Diferenciação Celular , Linhagem Celular Transformada , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Células Eritroides/patologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Camundongos , Mutação/genética , Transtornos Mieloproliferativos/patologia
2.
J Biol Chem ; 287(50): 42352-60, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23074222

RESUMO

Interferons (IFNs) have important antiviral and antineoplastic properties, but the precise mechanisms required for generation of these responses remain to be defined. We provide evidence that during engagement of the Type I IFN receptor (IFNR), there is up-regulation of expression of Sprouty (Spry) proteins 1, 2, and 4. Our studies demonstrate that IFN-inducible up-regulation of Spry proteins is Mnk kinase-dependent and results in suppressive effects on the IFN-activated p38 MAP kinase (MAPK), the function of which is required for transcription of interferon-stimulated genes (ISGs). Our data establish that ISG15 mRNA expression and IFN-dependent antiviral responses are enhanced in Spry1,2,4 triple knock-out mouse embryonic fibroblasts, consistent with negative feedback regulatory roles for Spry proteins in IFN-mediated signaling. In other studies, we found that siRNA-mediated knockdown of Spry1, Spry2, or Spry4 promotes IFN-inducible antileukemic effects in vitro and results in enhanced suppressive effects on malignant hematopoietic progenitors from patients with polycythemia vera. Altogether, our findings demonstrate that Spry proteins are potent regulators of Type I IFN signaling and negatively control induction of Type I IFN-mediated biological responses.


Assuntos
Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Interferon Tipo I/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Fosfoproteínas/genética , Policitemia Vera/genética , Policitemia Vera/metabolismo , Policitemia Vera/patologia , Proteínas Serina-Treonina Quinases , Receptor de Interferon alfa e beta/genética , Células U937 , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
J Biol Chem ; 286(8): 6017-26, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21149447

RESUMO

IFNγ exhibits potent antitumor effects and plays important roles in the innate immunity against cancer. However, the mechanisms accounting for the antiproliferative effects of IFNγ still remain to be elucidated. We examined the role of Mnk1 (MAPK-interacting protein kinase 1) in IFNγ signaling. Our data demonstrate that IFNγ treatment of sensitive cells results in engagement of Mnk1, activation of its kinase domain, and downstream phosphorylation of the cap-binding protein eIF4E on Ser-209. Such engagement of Mnk1 plays an important role in IFNγ-induced IRF-1 (IFN regulatory factor 1) gene mRNA translation/protein expression and is essential for generation of antiproliferative responses. In studies aimed to determine the role of Mnk1 in the induction of the suppressive effects of IFNs on primitive hematopoietic progenitors, we found that siRNA-mediated Mnk1/2 knockdown results in partial reversal of the suppressive effects of IFNγ on human CD34+-derived myeloid (CFU-GM) and erythroid (BFU-E) progenitors. These findings establish a key role for the Mnk/eIF4E pathway in the regulatory effects of IFNγ on normal hematopoiesis and identify Mnk kinases as important elements in the control of IFNγ-inducible ISG mRNA translation.


Assuntos
Células Precursoras Eritroides/enzimologia , Hematopoese/efeitos dos fármacos , Interferon gama/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Progenitoras Mieloides/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Transformada , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células Precursoras Eritroides/citologia , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Técnicas de Silenciamento de Genes , Hematopoese/fisiologia , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Células Progenitoras Mieloides/citologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/fisiologia
4.
Exp Hematol ; 33(5): 550-7, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15850832

RESUMO

OBJECTIVE: The mechanisms by which interferon alpha (IFN-alpha) induces antileukemic responses in chronic myelogenous leukemia (CML) cells are not known. We examined whether a member of the protein kinase C (PKC) family of proteins, PKC-delta, is activated during treatment of BCR-ABL cells with IFN-alpha and participates in the induction of interferon responses. METHODS: Immunoblots and immune complex kinase assays were performed to study the phosphorylation and activation of PKC-delta in response to IFN-alpha in CML-derived cell lines. The effects of pharmacological inhibition of PKC-delta on the suppressive effects of IFN-alpha on leukemic CFU-GM progenitors from CML patients were assessed by clonogenic assays in methylcellulose. RESULTS: IFN-alpha treatment of the sensitive CML-derived KT-1 cell line resulted in phosphorylation of PKC-delta and activation of its kinase domain. Such phosphorylation/activation of PKC-delta was required for phosphorylation of Stat1 on serine 727, as inhibition of PKC-delta activity blocked the IFN-alpha-dependent serine phosphorylation of Stat1 and IFN-alpha-inducible gene transcription. IFN-alpha treatment strongly inhibited leukemic CFU-GM progenitor colony formation from bone marrow or peripheral blood of patients with CML, and such inhibition was reversed by concomitant treatment of the cells with the PKC-delta pharmacologic inhibitor rottlerin. CONCLUSION: Taken altogether, our data demonstrate that PKC-delta plays a critical role in Type I IFN signaling in BCR-ABL expressing cells, acting as a serine kinase for Stat1, to regulate transcriptional activation of interferon-regulated genes and induction of antileukemic responses.


Assuntos
Interferon-alfa/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Proteína Quinase C/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Humanos , Imunoprecipitação , Interferon-alfa/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Fosforilação , Proteína Quinase C-delta , Fator de Transcrição STAT1 , Serina/metabolismo , Transativadores/metabolismo
5.
Blood ; 106(7): 2436-43, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15790787

RESUMO

The precise mechanisms by which imatinib mesylate (STI571) and interferon alpha (IFNalpha) exhibit antileukemic effects are not known. We examined the effects of IFNs or imatinib mesylate on signaling pathways regulating initiation of mRNA translation in BCR-ABL-expressing cells. Treatment of IFN-sensitive KT-1 cells with IFNalpha resulted in phosphorylation/activation of mammalian target of rapamycin (mTOR) and downstream activation of p70 S6 kinase. The IFN-activated p70 S6 kinase was found to regulate phosphorylation of S6 ribosomal protein, which regulates translation of mRNAs with oligopyrimidine tracts in the 5'-untranslated region. In addition, IFNalpha treatment resulted in an mTOR- and/or phosphatidyl-inositol 3'(PI 3') kinase-dependent phosphorylation of 4E-BP1 repressor of mRNA translation on sites that are required for its deactivation and dissociation from the eukaryotic initiation factor-4E (eIF4E) complex. In contrast to the effects of IFNs, imatinib mesylate suppressed p70 S6 kinase activity, consistent with inhibition of BCR-ABL-mediated activation of the mTOR/p70 S6 kinase pathway. Moreover, the mTOR inhibitor rapamycin enhanced the suppressive effects of imatinib mesylate on primary leukemic granulocyte macrophage-colony-forming unit (CFU-GM) progenitors from patients with chronic myelogenous leukemia (CML). Taken altogether, our data demonstrate that IFNs and imatinib mesylate differentially regulate PI 3' kinase/mTOR-dependent signaling cascades in BCR-ABL-transformed cells, consistent with distinct effects of these agents on pathways regulating mRNA translation. They also support the concept that combined use of imatinib mesylate with mTOR inhibitors may be an appropriate future therapeutic strategy for the treatment of CML.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interferon-alfa/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Proteínas Quinases S6 Ribossômicas 70-kDa/biossíntese , Regiões 5' não Traduzidas , Androstadienos/farmacologia , Benzamidas , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Fator de Iniciação 4E em Eucariotos/metabolismo , Regulação Enzimológica da Expressão Gênica , Granulócitos/citologia , Granulócitos/metabolismo , Humanos , Mesilato de Imatinib , Immunoblotting , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Piperazinas/farmacologia , Biossíntese de Proteínas , Proteínas Quinases/metabolismo , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Transdução de Sinais , Sirolimo/farmacologia , Células-Tronco , Serina-Treonina Quinases TOR , Fatores de Tempo , Wortmanina
6.
J Biol Chem ; 280(11): 10001-10, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15644321

RESUMO

There is accumulating evidence that the p38 MAP kinase pathway plays important roles in Type I interferon (IFN) signaling, but the mechanisms regulating p38 activation during engagement of the Type I IFN receptor remain to be defined. We sought to identify the events that lead to activation of the p38 MAP kinase in response to Type I IFNs. Our data demonstrate that treatment of sensitive cell lines with IFNalpha results in activation of both MAP kinase kinase 3 (MKK3) and MAP kinase kinase 6 (MKK6). Such IFN-inducible activation of MKK3 and MKK6 is essential for downstream phosphorylation and activation of the p38 MAP kinase, as shown by studies using mouse embryonic fibroblasts (MEFs) with targeted disruption of the Mkk3 and Mkk6 genes (MKK3-/- MKK6-/-). Similarly, IFN-dependent activation of the downstream effectors of p38, MAPKAPK-2 and MAPKAPK-3, is not detectable in cells lacking Mkk3 and Mkk6, demonstrating that the function of these MAP kinase kinases is required for full activation of the p38 pathway. To define the functional relevance of MKK3/6 engagement in Type I IFN signaling, IFN-inducible gene transcription was evaluated in the MKK3/MKK6 double knock-out cells. IFNalpha- and IFNbeta-dependent transcription via either interferon-stimulated response element or IFNgamma activated site elements was defective in MKK3 -/-/MKK6 -/- MEFs in luciferase reporter assays. In addition, IFN-dependent induction of two genes known to be of importance in the generation of IFN responses, Isg15 and Irf-9, was diminished in the absence of Mkk3 and Mkk6. The effects of Mkk3 and Mkk6 on IFN-dependent transcription were unrelated to any effects on the phosphorylation and activation of STAT proteins, indicating the presence of a STAT-independent mechanism. Altogether, our findings demonstrate that MKK3 and MKK6 are rapidly activated during engagement of the Type I IFN receptor and play important roles in Type I IFN signaling and the generation of IFN responses.


Assuntos
Interferon Tipo I/química , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase 6/metabolismo , Animais , Células Cultivadas , Citocinas/química , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Fibroblastos/metabolismo , Genes Reporter , Humanos , Immunoblotting , Imunoprecipitação , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Interferons/metabolismo , Luciferases/metabolismo , Camundongos , Fosforilação , Proteínas Recombinantes/química , Elementos de Resposta , Serina/química , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Tirosina/química , Ubiquitinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Biol Chem ; 279(29): 29911-20, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15150272

RESUMO

Protein kinase C-theta (PKC-theta) plays important roles in the activation and survival of lymphocytes and is the predominant PKC isoform expressed in T-cells. Interferons regulate T-cell function and activation, but the precise signaling mechanisms by which they mediate such effects have not been elucidated. We determined whether PKC-theta is engaged in interferon (INF) signaling in T-cells. Both Type I (alpha, beta) and Type II (gamma) IFNs induced phosphorylation of PKC-theta in human T-cell lines and primary human T-lymphocytes. Such phosphorylation of PKC-theta resulted in activation of its kinase domain, suggesting that this kinase plays a functional role in interferon signaling. Consistent with this, inhibition of PKC-theta protein expression using small interfering RNAs (siRNA) abrogated IFN-alpha- and IFN-gamma-dependent gene transcription via GAS elements. Similarly, blocking of PKC-theta kinase activity by overexpression of a dominant-negative PKC-theta mutant also blocked GAS-driven transcription, further demonstrating a requirement for PKC-theta in IFN-dependent transcriptional activation. The effects of PKC-theta on IFN-dependent gene transcription were not mediated by regulation of the IFN-activated STAT pathway, as siRNA-mediated PKC-theta knockdown had no effects on STAT1 phosphorylation and binding of STAT1-containing complexes to SIE/GAS elements. On the other hand, siRNA-mediated PKC-theta inhibition blocked phosphorylation/activation of MKK4, suggesting that interferon-dependent PKC-theta activation regulates downstream engagement of MAP kinase pathways. Altogether, these findings demonstrate that PKC-theta is an interferon-inducible kinase and strongly suggest that it plays an important role in the generation of interferon-responses in T-cells.


Assuntos
Interferons/metabolismo , Isoenzimas/metabolismo , MAP Quinase Quinase 4 , Proteína Quinase C/metabolismo , Linfócitos T/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Genes Dominantes , Humanos , Interferon Tipo I/metabolismo , Interferon gama/metabolismo , Células Jurkat , Luciferases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Isoformas de Proteínas , Proteína Quinase C-theta , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional , Transfecção
8.
J Biol Chem ; 279(24): 25345-52, 2004 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-15056660

RESUMO

Imatinib mesylate (STI571), a specific inhibitor of the BCR-ABL tyrosine kinase, exhibits potent antileukemic effects in vitro and in vivo. Despite the well established role of STI571 in the treatment of chronic myelogenous leukemia, the precise mechanisms by which inhibition of BCR-ABL tyrosine kinase activity results in generation of antileukemic responses remain unknown. In the present study we provide evidence that treatment of CML-derived BCR-ABL-expressing leukemia cells with STI571 results in activation of the p38 mitogen-activated protein (MAP) kinase signaling pathway. Our data indicate that STI571 induces phosphorylation of the p38 and activation of its kinase domain, in KT-1 cells and other BCR-ABL-expressing cell lines. We also identify the kinases MAP kinase-activated protein kinase-2 and Msk1 as two downstream effectors of p38, activated during inhibition of BCR-ABL activity by STI571. Importantly, pharmacological inhibition of p38 reverses the growth inhibitory effects of STI571 on primary leukemic colony-forming unit granulocyte/macrophage progenitors from patients with CML. Altogether, our data establish that activation of the p38 MAP kinase signaling cascade plays an important role in the generation of the effects of STI571 on BCR-ABL-expressing cells. They also suggest that, in addition to activation of mitogenic pathways, BCR-ABL promotes leukemogenesis by suppressing the function of growth inhibitory signaling cascades.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Benzamidas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Mesilato de Imatinib , Interferon-alfa/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno
9.
Exp Cell Res ; 295(1): 173-82, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15051500

RESUMO

The signals generated by the IFNgamma receptor to initiate mRNA translation and generation of protein products that mediate IFNgamma responses are largely unknown. In the present study, we provide evidence for the existence of an IFNgamma-dependent signaling cascade activated downstream of the phosphatidylinositol (PI) 3'-kinase, involving the mammalian target of rapamycin (mTOR) and the p70 S6 kinase. Our data demonstrate that p70 S6K is rapidly phosphorylated and activated during engagement of the IFNgamma receptor in sensitive cell lines. Such activation of p70 S6 kinase is blocked by pharmacological inhibitors of the PI 3' kinase and mTOR, and is abrogated in double-knockout mouse embryonic fibroblasts for the alpha and beta isoforms of the p85 regulatory subunit of the PI 3'-kinase. The IFNgamma-activated p70 S6 kinase subsequently phosphorylates the 40S S6 ribosomal protein on serines 235/236, to regulate IFNgamma-dependent mRNA translation. In addition to phosphorylation of 40S ribosomal protein, IFNgamma also induces phosphorylation of the 4E-BP1 repressor of mRNA translation on threonines 37/46, threonine 70, and serine 65, sites whose phosphorylation is required for the inactivation of 4E-BP1 and its dissociation from the eukaryotic initiation factor-4E (eIF4E) complex. Thus, engagement of the PI 3'-kinase and mTOR by the IFNgamma receptor results in the generation of two distinct signals that play roles in the initiation of mRNA translation, suggesting an important role for this pathway in IFNgamma signaling.


Assuntos
Interferon gama/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteína S6 Ribossômica/metabolismo , Neoplasias Ósseas , Linhagem Celular Tumoral , Cromonas/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Morfolinas/farmacologia , Osteossarcoma , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/genética
10.
J Biol Chem ; 279(2): 970-9, 2004 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-14578350

RESUMO

Multiple signaling pathways are activated during engagement of the Type I interferon (IFN) receptor to mediate biological responses, including the Jak-Stat and Rac1/p38 Map kinase signaling cascades. In the present study we sought to determine the functional relevance of the p38alpha isoform in IFN signaling, using cells from mouse embryos with targeted disruption of the p38alpha gene. Our data demonstrate that p38alpha activation is essential for Type I IFN-dependent transcriptional regulation via ISRE or GAS elements. On the other hand, the function of p38alpha is not required for IFN-dependent Ser727 or Tyr701 phosphorylation of Stat1 and does not impact on the formation of ISGF3 or SIF nuclear binding complexes. In efforts to identify downstream effectors of p38 that may mediate IFN-dependent transcriptional responses, we found that IFNalpha activates the kinase Msk1, a known regulator of histone phosphorylation and chromatin remodeling. In other studies, we demonstrate that Type I IFN-dependent activation of the kinases MapKapK-2 and MapKapK-3 is defective in the absence of p38alpha, while Type I IFN-dependent antiviral properties are decreased in cells with targeted disruption of the MapKapK-2 gene. Altogether, our data establish that the p38alpha Map kinase pathway regulates activation of downstream effectors that participate in the induction of IFN-dependent gene transcription, to mediate IFN-responses.


Assuntos
Interferon Tipo I/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Fibroblastos/metabolismo , Humanos , Immunoblotting , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Luciferases/metabolismo , Camundongos , Proteína Quinase 14 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osmose , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes/metabolismo , Serina/química , Estresse Fisiológico , Fatores de Tempo , Transcrição Gênica , Transfecção , Tirosina/química
11.
J Biol Chem ; 278(35): 32544-51, 2003 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-12805378

RESUMO

All-trans-retinoic acid (RA) is a potent inhibitor of leukemia cell proliferation and induces differentiation of acute promyelocytic leukemia cells in vitro and in vivo. For RA to induce its biological effects in target cells, binding to specific retinoic acid nuclear receptors is required. The resulting complexes bind to RA-responsive elements (RAREs) in the promoters of RA-inducible genes to initiate gene transcription and to generate protein products that mediate the biological effects of RA. In this report, we provide evidence that a member of the protein kinase C (PKC) family of proteins, PKC delta, is activated during RA treatment of the NB-4 and HL-60 acute myeloid leukemia cell lines as well as the MCF-7 breast cancer cell line. Such RA-dependent phosphorylation was also observed in primary acute promyelocytic leukemia cells and resulted in activation of the kinase domain of PKC delta. In studies aimed at understanding the functional relevance of PKC delta in the induction of RA responses, we found that pharmacological inhibition of PKC delta (but not of other PKC isoforms) diminished RA-dependent gene transcription via RAREs. On the other hand, overexpression of a constitutively active form of the kinase strongly enhanced RA-dependent gene transcription via RAREs. Gel shift assays and chromatin immunoprecipitation studies demonstrated that PKC delta associated with retinoic acid receptor-alpha and was present in an RA-inducible protein complex that bound to RAREs. Pharmacological inhibition of PKC delta activity abrogated the induction of cell differentiation and growth inhibition of NB-4 blast cells, demonstrating that its function is required for such effects. Altogether, our data provide strong evidence that PKC delta is activated in an RA-dependent manner and plays a critical role in the generation of the biological effects of RA in malignant cells.


Assuntos
Ativação Enzimática , Proteína Quinase C/metabolismo , Tretinoína/metabolismo , Diferenciação Celular , Divisão Celular , Cromatina/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Células HL-60 , Humanos , Immunoblotting , Interferon-alfa/metabolismo , Luciferases/metabolismo , Testes de Precipitina , Regiões Promotoras Genéticas , Isoformas de Proteínas , Proteína Quinase C-delta , Estrutura Terciária de Proteína , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Transcrição Gênica , Células Tumorais Cultivadas
12.
J Immunol ; 171(1): 267-73, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12817007

RESUMO

Engagement of the type II IFN (IFN-gamma) receptor results in activation of the Janus kinase-Stat pathway and induction of gene transcription via IFN-gamma-activated site (GAS) elements in the promoters of IFN-gamma-inducible genes. An important event in IFN-gamma-dependent gene transcription is phosphorylation of Stat1 on Ser(727), which is regulated by a kinase activated downstream of the phosphatidylinositol 3'-kinase. Here we provide evidence that a member of the protein kinase C (PKC) family of proteins is activated downstream of the phosphatidylinositol 3'-kinase and is engaged in IFN-gamma signaling. Our data demonstrate that PKCdelta is rapidly phosphorylated during engagement of the type II IFNR and its kinase domain is induced. Subsequently, the activated PKCdelta associates with a member of the Stat family of proteins, Stat1, which acts as a substrate for its kinase activity and undergoes phosphorylation on Ser(727). Inhibition of PKCdelta activity diminishes phosphorylation of Stat1 on Ser(727) and IFN-gamma-dependent transcriptional regulation via IFN-gamma-activated site elements, without affecting the phosphorylation of the protein on Tyr(701). Thus, PKCdelta is activated during engagement of the IFN-gamma receptor and plays an important role in IFN-gamma signaling by mediating serine phosphorylation of Stat1 and facilitating transcription of IFN-gamma-stimulated genes.


Assuntos
Interferon gama/fisiologia , Proteína Quinase C/metabolismo , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Carbazóis/farmacologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Interferon gama/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Mesilatos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/fisiologia , Proteína Quinase C-delta , Piridinas/farmacologia , Pirróis/farmacologia , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais/imunologia , Transativadores/metabolismo , Transativadores/fisiologia , Células Tumorais Cultivadas , Receptor de Interferon gama
13.
J Biol Chem ; 278(30): 27772-80, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12759354

RESUMO

The Type I IFN receptor-generated signals required for initiation of mRNA translation and, ultimately, induction of protein products that mediate IFN responses, remain unknown. We have previously shown that IFNalpha and IFNbeta induce phosphorylation of insulin receptor substrate proteins and downstream engagement of the phosphatidylinositol (PI) 3'-kinase pathway. In the present study we provide evidence for the existence of a Type I IFN-dependent signaling cascade activated downstream of PI 3'-kinase, involving p70 S6 kinase. Our data demonstrate that p70 S6K is rapidly phosphorylated on threonine 421 and serine 424 and is activated during treatment of cells with IFNalpha or IFNbeta. Such activation of p70 S6K is blocked by pharmacological inhibitors of the PI 3'-kinase or the FKBP 12-rapamycin-associated protein/mammalian target of rapamycin (FRAP/mTOR). Consistent with this, the Type I IFN-dependent phosphorylation/activation of p70 S6K is defective in embryonic fibroblasts from mice with targeted disruption of the p85alpha and p85beta subunits of the PI 3'-kinase (p85alpha-/-beta-/-). Treatment of sensitive cell lines with IFNalpha or IFNbeta also results in phosphorylation/inactivation of the 4E-BP-1 repressor of mRNA translation. Such 4E-BP1 phosphorylation is also PI3'-kinase-dependent and rapamycin-sensitive, indicating that the Type I IFN-inducible activation of PI3'-kinase and FRAP/mTOR results in dissociation of 4E-BP1 from the eukaryotic initiation factor-4E (eIF4E) complex. Altogether, our data establish that the Type I IFN receptor-activated PI 3'-kinase pathway mediates activation of the p70 S6 kinase and inactivation of 4E-BP1, to regulate mRNA translation and induction of Type I IFN responses.


Assuntos
Proteínas de Transporte/metabolismo , Interferon Tipo I/metabolismo , Fosfoproteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cromatografia , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Fatores de Iniciação em Eucariotos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Guanosina Difosfato/metabolismo , Humanos , Immunoblotting , Interferons/metabolismo , Luciferases/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Biossíntese de Proteínas , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Sefarose/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
14.
J Rheumatol ; 30(5): 934-40, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12734885

RESUMO

OBJECTIVE: To evaluate the therapeutic potential of interferon-a (IFN-a) in osteoarthritis (OA) and rheumatoid arthritis (RA) by examining regulation of cytokine antagonist expression. METHODS: Expression of interleukin 1 receptor antagonist (IL-1Ra) and soluble tumor necrosis factor receptor (sTNFR) was examined by ELISA in cells from freshly isolated synovial fluids (SF) and synovial tissues (ST) from patients with OA or RA, either left untreated or treated with IFN-a. Single (7) and paired (5) SF and ST cells from OA and RA patients were examined. As well, the ability of IFN-a to regulate gene expression levels for osteoprotegerin (OPG) and osteoprotegerin ligand (OPGL) was examined in freshly isolated SF cells from patients with RA, by reverse transcriptase polymerase chain reaction. RESULTS: IL-1Ra and sTNFR were found to be constitutively expressed in OA and RA SF and ST cells. IFN-a treatment resulted in an increase in both IL 1Ra and sTNFR production. Freshly isolated RA SF cells exhibited constitutive OPGL gene expression in both the non-T and T cell fractions of the SF. In contrast, OPG gene expression levels were undetectable or low. IFN-a treatment of RA SF cells resulted in upregulation of OPG gene expression in the T cell fraction of the RA SF cells, whereas OPGL gene expression remained unaffected. CONCLUSION: These in vitro data suggest a therapeutic role for IFN-a in the treatment of arthritis through upregulation of critical cytokine antagonists.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Fatores Imunológicos/farmacologia , Interferon-alfa/farmacologia , Osteoartrite/tratamento farmacológico , Adulto , Artrite Reumatoide/imunologia , Proteínas de Transporte/genética , Etanercepte , Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Imunoglobulina G/genética , Técnicas In Vitro , Proteína Antagonista do Receptor de Interleucina 1 , Glicoproteínas de Membrana/genética , Osteoartrite/imunologia , Osteoprotegerina , Projetos Piloto , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Fator de Necrose Tumoral/genética , Sialoglicoproteínas/genética , Líquido Sinovial/citologia , Membrana Sinovial/citologia , Regulação para Cima/efeitos dos fármacos
15.
Biochem Biophys Res Commun ; 291(4): 744-50, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11866427

RESUMO

We sought to determine the functional role of the CrkL adapter protein and downstream pathways in interferon signaling. In experiments using CrkL(--) mouse embryonic fibroblasts, we found that CrkL is required for IFN alpha-dependent gene transcription via GAS elements, apparently via the formation of DNA-binding complexes with Stat5. On the other hand, gene transcription via ISRE elements is intact in the absence of CrkL, indicating that the regulatory effects on gene transcription are mediated only via the formation of CrkL:Stat5 complexes. Our studies also indicate that activation of the small GTPase Rap1 by IFN alpha is defective in cells lacking CrkL, indicating that the protein plays a critical role in regulating activation of the growth inhibitory C3G/Rap1 pathway. The IFN alpha-inducible activation of the small GTPase Rap1 requires a functional N-terminus SH3 domain in the CrkL protein, while the C-terminus SH3 domain does not appear to play a role in such a CrkL-function. We also demonstrate that both the Tyk-2 and Jak-1 kinases are required for activation of the CrkL/Rap1 pathway, as the Type I IFN-dependent GTP-bound form of Rap1 is inhibited by overexpression of dominant-negative Tyk-2 or Jak-1 mutants and is defective in cells lacking Tyk-2 or Jak-1. Taken altogether, these findings indicate that CrkL provides an important link between Jak-kinases and downstream cascades that play critical roles in IFN-dependent transcriptional regulation and induction of growth inhibitory responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Interferon-alfa/farmacologia , Proteínas Nucleares/metabolismo , Ativação Transcricional , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Ativação Enzimática , Deleção de Genes , Genes Reporter , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Humanos , Janus Quinase 1 , Cinética , Camundongos , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Elementos de Resposta , Células Tumorais Cultivadas , Domínios de Homologia de src
16.
J Biol Chem ; 277(17): 14408-16, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-11839738

RESUMO

It is well established that engagement of the Type I interferon (IFN) receptor results in activation of JAKs (Janus kinases), which in turn regulate tyrosine phosphorylation of STAT proteins. Subsequently, the IFN-dependent tyrosine-phosphorylated/activated STATs translocate to the nucleus to regulate gene transcription. In addition to tyrosine phosphorylation, phosphorylation of Stat1 on serine 727 is essential for induction of its transcriptional activity, but the IFNalpha-dependent serine kinase that regulates such phosphorylation remains unknown. In the present study we provide evidence that PKC-delta, a member of the protein kinase C family of proteins, is activated during engagement of the Type I IFN receptor and associates with Stat1. Such an activation of PKC-delta appears to be critical for phosphorylation of Stat1 on serine 727, as inhibition of PKC-delta activation diminishes the IFNalpha- or IFNbeta-dependent serine phosphorylation of Stat1. In addition, treatment of cells with the PKC-delta inhibitor rottlerin or the expression of a dominant-negative PKC-delta mutant results in inhibition of IFNalpha- and IFNbeta-dependent gene transcription via ISRE or GAS elements. Interestingly, PKC-delta inhibition also blocks activation of the p38 MAP kinase, the function of which is required for IFNalpha-dependent transcriptional regulation, suggesting a dual mechanism by which this kinase participates in the generation of IFNalpha responses. Altogether, these findings indicate that PKC-delta functions as a serine kinase for Stat1 and an upstream regulator of the p38 MAP kinase and plays an important role in the induction of Type I IFN-biological responses.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interferon Tipo I/fisiologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Serina/metabolismo , Transativadores/metabolismo , Sequência de Bases , Linhagem Celular , Primers do DNA , Proteínas de Ligação a DNA/química , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Isoenzimas/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Fator de Transcrição STAT1 , Transativadores/química , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA