Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Tissue Antigens ; 68(1): 1-12, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16774534

RESUMO

Cytokine signaling is essential for intercellular communication and affects cell proliferation, differentiation, and survival. In the immune system, cytokines coordinate the activities of many cell types ultimately leading to both innate and adaptive immune responses. Dysregulation of these processes can result in a wide spectrum of diseases ranging from defective host responses to invading pathogens to autoimmunity. Most cytokines signal through the Janus kinase-signal transducer and activator of transcription (Jak-STAT) pathway initiated by the cytokine binding to its cell surface receptor, which leads to the activation of STAT proteins, their binding to response elements near target promoters ultimately changing the transcription of STAT-responsive genes. STAT proteins do not exist in isolation but act in concert with other transcription factors and cofactors which can either stimulate or inhibit their activity. One such factor is a ligand-dependent transcriptional regulator termed the glucocorticoid (GC) receptor (GR), which transduces the information conveyed by GC hormones and their synthetic analogs. GR is known for its anti-inflammatory and immunosuppressive properties; GC-like molecules have been used as drugs for inflammatory, autoimmune and lymphoproliferative diseases since the 1950s. In contrast, cytokines frequently promote activation of the immune system. In last several years, functional interactions have been described between virtually every member of the STAT family and GR or its cofactors. This review focuses on the recent literature on the modes and levels of interactions between these seemingly unrelated regulators and potential biological implications of STAT : GR cross-talk.


Assuntos
Citocinas/metabolismo , Glucocorticoides/farmacologia , Transdução de Sinais , Animais , Humanos , Janus Quinase 1 , Proteínas Tirosina Quinases/fisiologia , Receptores de Glucocorticoides/efeitos dos fármacos , Fatores de Transcrição STAT/fisiologia , Ativação Transcricional/fisiologia
2.
EMBO J ; 20(21): 6071-83, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11689447

RESUMO

To investigate determinants of specific transcriptional regulation, we measured factor occupancy and function at a response element, col3A, associated with the collagenase-3 gene in human U2OS osteosarcoma cells; col3A confers activation by phorbol esters, and repression by glucocorticoid and thyroid hormones. The subunit composition and activity of AP-1, which binds col3A, paralleled the intracellular level of cFos, which is modulated by phorbol esters and glucocorticoids. In contrast, a similar AP-1 site at the collagenase-1 gene, not inducible in U2OS cells, was not bound by AP-1. The glucocorticoid receptor (GR) associated with col3A through protein-protein interactions with AP-1, regardless of AP-1 subunit composition, and repressed transcription. TIF2/GRIP1, reportedly a coactivator for GR and the thyroid hormone receptor (TR), was recruited to col3A and potentiated GR-mediated repression in the presence of a GR agonist but not antagonist. GRIP1 mutants deficient in GR binding and coactivator functions were also defective for corepression, and a GRIP1 fragment containing the GR-interacting region functioned as a dominant-negative for repression. In contrast, repression by TR was unaffected by GRIP1. Thus, the composition of regulatory complexes, and the biological activities of the bound factors, are dynamic and dependent on cell and response element contexts. Cofactors such as GRIP1 probably contain distinct surfaces for activation and repression that function in a context-dependent manner.


Assuntos
Colagenases/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Osteossarcoma/metabolismo , Elementos de Resposta/fisiologia , Fatores de Transcrição/metabolismo , Animais , Colagenases/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Hormônios/farmacologia , Humanos , Metaloproteinase 13 da Matriz , Camundongos , Mifepristona/farmacologia , Coativador 2 de Receptor Nuclear , Ésteres de Forbol/farmacologia , Ligação Proteica/fisiologia , Ratos , Receptores de Glucocorticoides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Proteínas Repressoras/metabolismo , Elementos de Resposta/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas
3.
J Biol Chem ; 275(23): 17771-7, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10748103

RESUMO

Steroid hormones regulate the transcription of numerous genes via high affinity receptors that act in concert with chromatin remodeling complexes, coactivators and corepressors. We have compared the activities of a variety of glucocorticoid receptor (GR) antagonists in breast cancer and osteosarcoma cell lines engineered to stably maintain the mouse mammary tumor virus promoter. In both cell types, GR activation by dexamethasone occurs via the disruption of mouse mammary tumor virus chromatin structure and the recruitment of receptor coactivator proteins. However, when challenged with a variety of antagonists the GR displays differential ability to activate transcription within the two cell types. For the breast cancer cells, the antagonists fail to activate the promoter and do not promote the association of the GR with either remodeling or coactivator proteins. In contrast, in osteosarcoma cells, the antiglucocorticoids, RU486 and RU43044, exhibit partial agonist activity. The capacity of these antagonists to stimulate transcription in the osteosarcoma cells is reflected in the ability of the RU486-bound receptor to remodel chromatin and associate with chromatin-remodeling proteins. Similarly, the observation that the RU486-bound receptor does not fully activate transcription is consistent with its inability to recruit receptor coactivator proteins.


Assuntos
Dexametasona/farmacologia , Regulação Neoplásica da Expressão Gênica , Glucocorticoides/farmacologia , Antagonistas de Hormônios/farmacologia , Hidroxicorticosteroides , Vírus do Tumor Mamário do Camundongo/genética , Mifepristona/farmacologia , Receptores de Glucocorticoides/metabolismo , Animais , Neoplasias da Mama , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Osteossarcoma , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
4.
J Biol Chem ; 274(32): 22296-302, 1999 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-10428798

RESUMO

Both estradiol binding and phosphorylation regulate transcriptional activation by the human estrogen receptor alpha (ER). We have previously shown that activation of the cyclin A-CDK2 complex by overexpression of cyclin A leads to enhanced ER-dependent transcriptional activation and that the cyclin A-CDK2 complex phosphorylates the ER N-terminal activation function-1 (AF-1) between residues 82 and 121. Within ER AF-1, serines 104, 106, and 118 represent potential CDK phosphorylation sites, and in this current study, we ascertain their importance in mediating cyclin A-CDK2-dependent enhancement of ER transcriptional activity. Cyclin A overexpression does not enhance transcriptional activation by an ER derivative bearing serine-to-alanine changes at residues 104, 106, and 118. Likewise, the cyclin A-CDK2 complex does not phosphorylate this triple-mutated derivative in vitro. Individual serine-to-alanine mutations at residues 104 and 106, but not 118, decrease ER-dependent transcriptional enhancement in response to cyclin A. The same relationship holds for ER phosphorylation by cyclin A-CDK2 in vitro. Finally, enhancement of ER transcriptional activation by cyclin A is evident in the absence and presence of estradiol, as well as in the presence of tamoxifen, suggesting that the effect of the cyclin A-CDK2 on ER transcriptional activation is AF-2-independent. These results indicate that the enhancement of ER transcriptional activation by the cyclin A-CDK2 complex is mediated via the AF-1 domain by phosphorylation of serines 104 and 106. We propose that these residues control ER AF-1 activity in response to signals that affect cyclin A-CDK2 function.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclina A/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Estrogênio/metabolismo , Serina/metabolismo , Ativação Transcricional , Ciclina A/genética , Quinase 2 Dependente de Ciclina , Estradiol/metabolismo , Receptor alfa de Estrogênio , Humanos , Modelos Genéticos , Mutagênese Sítio-Dirigida , Fosforilação , Receptores de Estrogênio/genética , Proteínas Recombinantes/metabolismo , Serina/genética
5.
Mol Cell Biol ; 19(7): 5036-49, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10373553

RESUMO

Glucocorticoids act through the glucocorticoid receptor (GR), which can function as a transcriptional activator or repressor, to elicit cytostatic and cytotoxic effects in a variety of cells. The molecular mechanisms regulating these events and the target genes affected by the activated receptor remain largely undefined. Using cultured human osteosarcoma cells as a model for the GR antiproliferative effect, we demonstrate that in U20S cells, GR activation leads to irreversible growth inhibition, apoptosis, and repression of Bcl2. This cytotoxic effect is mediated by GR's transcriptional repression function, since transactivation-deficient mutants and ligands still bring about apoptosis and Bcl2 down-regulation. In contrast, the antiproliferative effect of GR in SAOS2 cells is reversible, does not result in apoptosis or repression of Bcl2, and is a function of the receptor's ability to stimulate transcription. Thus, the cytotoxic versus cytostatic outcome of glucocorticoid treatment is cell context dependent. Interestingly, the cytostatic effect of glucocorticoids in SAOS2 cells involves multiple GR activation surfaces. GR mutants and ligands that disrupt individual transcriptional activation functions (activation function 1 [AF-1] and AF-2) or receptor dimerization fail to fully inhibit cellular proliferation and, remarkably, discriminate between the targets of GR's cytostatic action, the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip1). Induction of p21(Cip1) is agonist dependent and requires AF-2 but not AF-1 or GR dimerization. In contrast, induction of p27(Kip1) is agonist independent, does not require AF-2 or AF-1, but depends on GR dimerization. Our findings indicate that multiple GR transcriptional regulatory mechanisms that employ distinct receptor surfaces are used to evoke either the cytostatic or cytotoxic response to glucocorticoids.


Assuntos
Antineoplásicos/metabolismo , Apoptose , Proteínas de Ciclo Celular , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas Supressoras de Tumor , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/metabolismo , Citotoxicidade Imunológica , Dimerização , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Mifepristona/farmacologia , Mutagênese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética , Ativação Transcricional , Células Tumorais Cultivadas
6.
J Biol Chem ; 273(23): 14315-21, 1998 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-9603939

RESUMO

Transcriptional activation by the glucocorticoid receptor (GR) is regulated by both glucocorticoid binding and phosphorylation. The rat GR N-terminal transcriptional regulatory domain contains four major phosphorylation sites: threonine 171 (Thr171), serine 224 (Ser224), serine 232 (Ser232), and serine 246 (Ser246). We have previously demonstrated that Ser224 and Ser232 are phosphorylated by cyclin-dependent kinases, while Ser246 is phosphorylated by the c-Jun N-terminal kinase. We report here that the remaining GR phosphorylation site, Thr171, is a target for glycogen synthase kinase-3 (GSK-3) in vitro and in cultured mammalian cells. Increasing GSK-3 activity through its overexpression in cultured cells inhibits GR transcriptional enhancement, an effect dependent upon Thr171. Correspondingly, overexpression of a constitutively active form of the GSK-3 inhibitor, protein kinase B/Akt, increases GR transcriptional enhancement. Overexpression of GSK-3 had no effect on GR-mediated transcriptional repression of AP1-dependent gene expression. Importantly, transcriptional activation by the human GR (hGR), which contains an alanine (Ala150) at the position equivalent to Thr171 in rat GR, is not affected by GSK-3 overexpression. Introduction of a threonine residue at this position (A150T) establishes GSK-3-mediated inhibition of hGR transcriptional activation. These findings demonstrate species-specific differences in GR signaling, as revealed through GSK-3 phosphorylation, which suggests that GR function in rodents may not fully recapitulate receptor action in humans and that hGR is capable of adopting the GSK-3 signaling pathway through a somatic mutation.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Serina-Treonina Quinases , Receptores de Glucocorticoides/metabolismo , Ativação Transcricional/genética , Animais , Regulação da Expressão Gênica/genética , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Humanos , Mutagênese Sítio-Dirigida , Fosfopeptídeos/química , Fosforilação , Fosfotreonina/análise , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Proteínas Recombinantes/metabolismo , Transfecção/genética , Células Tumorais Cultivadas
7.
Proc Natl Acad Sci U S A ; 95(5): 2050-5, 1998 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-9482836

RESUMO

The mitogen-activated protein kinases ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), and p38 phosphorylate and activate transcription factors that promote proliferative and inflammatory responses, whereas glucocorticoid receptor (GR) activation inhibits cell growth and inflammation. We demonstrate that JNK and ERK but not p38 phosphorylate GR in vitro primarily at Ser-246. Selective activation of either ERK or JNK in vivo inhibits GR-mediated transcriptional activation, which depends on receptor phosphorylation at Ser-246 by JNK but not ERK. Thus, JNK inhibits GR transcriptional activation by direct receptor phosphorylation, whereas ERK does so indirectly. We propose that phosphorylation of GR by JNK or of a GR cofactor by ERK provides mechanisms to ensure the rapid inhibition of GR-dependent gene expression when it conflicts with mitogenic or proinflammatory signals.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Dexametasona/farmacologia , Células HeLa , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Osteossarcoma , Mapeamento de Peptídeos , Fosfopeptídeos , Fosforilação , Ratos , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/química , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Serina , Transcrição Gênica/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
8.
Proc Natl Acad Sci U S A ; 94(19): 10132-7, 1997 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-9294175

RESUMO

We have found that ectopic expression of cyclin A increases hormone-dependent and hormone-independent transcriptional activation by the estrogen receptor in vivo in a number of cell lines, including HeLa cells, U-2 OS osteosarcoma cells and Hs 578Bst breast epithelial cells. This effect can be further enhanced in HeLa cells by the concurrent expression of the cyclin-dependent kinase activator, cyclin H, and cdk7, and abolished by expression of the cdk inhibitor, p27(KIP1), or by the expression of a dominant negative catalytically inactive cdk2 mutant. ER is phosphorylated between amino acids 82 and 121 in vitro by the cyclin A/cdk2 complex and incorporation of phosphate into ER is stimulated by ectopic expression of cyclin A in vivo. Together, these results strongly suggest a direct role for the cyclin A/cdk2 complex in phosphorylating ER and regulating its transcriptional activity.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Regulação da Expressão Gênica , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Estrogênio/genética , Transcrição Gênica , Linhagem Celular , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/antagonistas & inibidores , Ativação Enzimática , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Células Tumorais Cultivadas
9.
Mol Cell Biol ; 17(7): 3947-54, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199329

RESUMO

Cyclin-dependent kinase (CDK) and mitogen-activated protein kinase (MAPK) phosphorylate the rat glucocorticoid receptor in vitro at distinct sites that together correspond to the major phosphorylated receptor residues observed in vivo; MAPK phosphorylates receptor residues threonine 171 and serine 246, whereas multiple CDK complexes modify serines 224 and 232. Mutations in these kinases have opposite effects on receptor transcriptional activity in vivo. Receptor-dependent transcriptional enhancement is reduced in yeast strains deficient in the catalytic (p34CDC28) or certain regulatory (cyclin) subunits of CDK complexes and is increased in a strain devoid of the mammalian MAPK homologs FUS3 and KSS1. These findings indicate that the glucocorticoid receptor is a target for multiple kinases in vivo, which either positively or negatively regulate receptor transcriptional enhancement. The control of receptor transcriptional activity via phosphorylation provides an increased array of regulatory inputs that, in addition to steroid hormones, can influence receptor function.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Ciclinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Receptores de Glucocorticoides/fisiologia , Transcrição Gênica , Animais , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Mapeamento de Peptídeos , Fosfopeptídeos/análise , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Proteínas Recombinantes , Transdução de Sinais
10.
Mol Cell Biol ; 17(6): 3181-93, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9154817

RESUMO

Glucocorticoids inhibit proliferation of many cell types, but the events leading from the activated glucocorticoid receptor (GR) to growth arrest are not understood. Ectopic expression and activation of GR in human osteosarcoma cell lines U2OS and SAOS2, which lack endogenous receptors, result in a G1 cell cycle arrest. GR activation in U2OS cells represses expression of the cyclin-dependent kinases (CDKs) CDK4 and CDK6 as well as their regulatory partner, cyclin D3, leading to hypophosphorylation of the retinoblastoma protein (Rb). We also demonstrate a ligand-dependent reduction in the expression of E2F-1 and c-Myc, transcription factors involved in the G1-to-S-phase transition. Mitogen-activated protein kinase, CDK2, cyclin E, and the CDK inhibitors (CDIs) p27 and p21 are unaffected by receptor activation in U2OS cells. The receptor's N-terminal transcriptional activation domain is not required for growth arrest in U2OS cells. In Rb-deficient SAOS2 cells, however, the expression of p27 and p21 is induced upon receptor activation. Remarkably, in SAOS2 cells that express a GR deletion derivative lacking the N-terminal transcriptional activation domain, induction of CDI expression is abolished and the cells fail to undergo ligand-dependent cell cycle arrest. Similarly, murine S49 lymphoma cells, which, like SAOS2 cells, lack Rb, require the N-terminal activation domain for growth arrest and induce CDI expression upon GR activation. These cell-type-specific differences in receptor domains and cellular targets linking GR activation to cell cycle machinery suggest two distinct regulatory mechanisms of GR-mediated cell cycle arrest: one involving transcriptional repression of G1 cyclins and CDKs and the other involving enhanced transcription of CDIs by the activated receptor.


Assuntos
Proteínas de Ciclo Celular , Ciclo Celular/genética , Proteínas Proto-Oncogênicas , Receptores de Glucocorticoides/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor , Animais , Sítios de Ligação , Divisão Celular , Ciclina D3 , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA