Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Biol Chem ; 281(47): 35812-25, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17008315

RESUMO

Signal transducer and activator of transcription 3 (STAT3) can be stimulated by several G(s)-coupled receptors, but the precise mechanism of action has not yet been elucidated. We therefore examined the ability of Galpha(s)Q226L (Galpha(s)QL), a constitutively active mutant of Galpha(s), to stimulate STAT3 Tyr705 and Ser727 phosphorylations in human embryonic kidney 293 cells. Apart from Galpha(s)QL, the stimulation of Galpha(s) by cholera toxin or beta2-adrenergic receptor and the activation of adenylyl cyclase by forskolin, (Sp)-cAMP, or dibutyryl-cAMP all promoted both STAT3 Tyr705 and Ser727 phosphorylations. Moreover, the removal of Galpha(s) by RNA interference significantly reduced the beta2-adrenergic receptor-mediated STAT3 phosphorylations, denoting its capacity to regulate STAT3 activation by a G protein-coupled receptor. The possible downstream signaling molecules involved were assessed by using specific inhibitors and dominant negative mutants. Induction of STAT3 Tyr705 and Ser727 phosphorylations by Galpha(s)QL was suppressed by inhibition of protein kinase A, Janus kinase 2/3, Rac1, c-Jun N-terminal kinase (JNK), or phosphatidylinositol 3-kinase, and a similar profile was observed in response to beta2-adrenergic receptor stimulation. In contrast to the Galpha16-mediated regulation of STAT3 in HEK 293 cells (Lo, R. K., Cheung, H., and Wong, Y. H. (2003) J. Biol. Chem. 278, 52154-52165), the Galpha(s)-mediated responses, including STAT3-driven luciferase activation, were resistant to inhibition of phospholipase Cbeta. Surprisingly, Galpha(s)-mediated phosphorylation at Tyr705, but not at Ser727, was resistant to inhibition of c-Src, Raf-1, and MEK1/2 as well as to the expression of dominant negative Ras. Therefore, as with other Galpha-mediated activations of STAT3, the stimulatory signal arising from Galpha(s) is transduced via multiple signaling pathways. However, unlike the mechanisms employed by Galpha(i) and Galpha(14/16), Galpha(s) distinctively requires protein kinase A, JNK, and phosphatidylinositol 3-kinase for STAT3 activation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , MAP Quinase Quinase 4/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fator de Transcrição STAT3/fisiologia , Linhagem Celular , Colforsina/farmacologia , Genes Dominantes , Humanos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional
2.
Biochem J ; 380(Pt 2): 361-9, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15005654

RESUMO

A broad repertory of G-protein-coupled receptors shows effective coupling with the haematopoietic G16 protein. In the present study, individual residues along the C-terminal alpha5 helix of Galpha16 were examined for their contributions in defining receptor-coupling specificity. Residues that are relatively conserved within, but diverse between, the subfamilies of cloned Galpha subunits were mutated into the corresponding Galpha(z) residues. Six G(i)-linked receptors with different coupling efficiencies to Galpha16 were examined for their ability to utilize the various Galpha16 mutants to mediate agonist-induced inositol phosphate accumulation and Ca2+ mobilization. Co-operative enhancements of receptor coupling were observed with chimaeras harbouring multiple mutations at Glu350, Lys357 and Leu364 of Galpha16. Mutation of Leu364 into isoleucine appeared to be more efficient in enhancing receptor recognition compared with mutations at the other two sites. Mutation of a stretch of six consecutive residues (362-367) lying towards the end of the alpha5 helix was found to broaden significantly the receptor-coupling profile of Galpha16, and the effect was mediated partly through interactions with the beta2-beta3 loop. These results suggested that a stretch of six distinctive residues at the alpha5 helix of Galpha16 is particularly important, whereas other discrete residues spreading along the alpha5 helix function co-operatively for determining the specificity of receptor recognition.


Assuntos
Aminoácidos/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/química , Peptídeos/fisiologia , Receptores Acoplados a Proteínas G/química , Sequência de Aminoácidos/genética , Sequência de Aminoácidos/fisiologia , Aminoácidos/genética , Animais , Células COS , Chlorocebus aethiops , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/genética , Mutagênese Sítio-Dirigida/fisiologia , Peptídeos/química , Peptídeos/genética , Ligação Proteica/fisiologia , Receptores Acoplados a Proteínas G/genética , Alinhamento de Sequência
3.
Cell Signal ; 16(1): 51-62, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14607275

RESUMO

G(16) can couple indiscriminately to a large number of G protein-coupled receptors (GPCRs), making it a prime candidate as a universal adaptor for GPCRs. In order to increase the promiscuity of Galpha(16), three chimeras incorporating increasing lengths of G(s)-specific residues (25, 44 or 81 residues) into the C-terminus of Galpha(16) were constructed and named 16s25, 16s44 and 16s81, respectively. The chimeras were examined for their ability to mediate receptor-induced stimulation of phospholipase C (PLC) and Ca(2+) mobilization. 16s25 was more effective than 16s44 and 16s81 at coupling to G(s)-linked receptors. 16s25 coupled productively to 10 different G(s)-coupled receptors examined and, for 50% of these receptors, 16s25-mediated PLC activities were higher than those mediated via Galpha(16). Similar results were observed for agonist-induced Ca(2+) mobilizations. These results show that incorporating the alpha5 helix of Galpha(s) into Galpha(16) can increase the promiscuity of 16s25 towards G(s)-coupled receptors.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Sequência de Bases/genética , Células COS , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Estrutura Secundária de Proteína/genética , Estrutura Terciária de Proteína/genética , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/genética , Fosfolipases Tipo C/metabolismo
4.
Eur J Pharmacol ; 473(2-3): 105-15, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12892827

RESUMO

Replacement of beta6/alpha5 region at the C-terminus on Galpha16 with Galphaz-specific residues has been shown to broaden the promiscuity of Galpha16. Here, we substituted the last 44 residues of Galpha16 with the corresponding region from either Galphai2 or GalphaoA (16i44 and 16o44). 16i44 and 16o44 chimeras were more effective than Galpha16 at coupling to Gi-linked delta-opioid, mu-opioid, and Xenopus melatonin MT1c receptors when coexpressed in green monkey fibroblast (COS-7) cells. 16i44, but not 16o44, also enhanced the formyl peptide-induced stimulation of phospholipase C activity. Both chimeras were resistant to pertussis toxin-catalyzed [32P]ADP-ribosylation, despite the fact that pertussis toxin partially inhibited the chimera-mediated stimulation of phospholipase Cbeta. The use of Galphat1 as a Gbetagamma scavenger revealed that the pertussis toxin-sensitivity can be attributed to endogenous Gbetagamma subunits released from G(i/o). Although incorporation of a Galphai-like beta6/alpha5 region into the C-terminus of Galpha16 increases its promiscuity, this region is not sufficient to support recognition by pertussis toxin.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Toxina Pertussis/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Subunidade alfa Gi2 de Proteína de Ligação ao GTP , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/genética , Humanos , Fosfatos de Inositol/metabolismo , Isoenzimas/metabolismo , Camundongos , Dados de Sequência Molecular , Fosfolipase C beta , Subunidades Proteicas , Proteínas Proto-Oncogênicas/genética , Ratos , Receptores de Melatonina/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Fosfolipases Tipo C/metabolismo , Xenopus
5.
J Biomol Screen ; 8(1): 39-49, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12854997

RESUMO

G protein-coupled receptors (GPCRs) represent a class of important therapeutic targets for drug discovery. The integration of GPCRs into contemporary high-throughput functional assays is critically dependent on the presence of appropriate G proteins. Given that different GPCRs can discriminate against distinct G proteins, a universal G protein adapter is extremely desirable. In this report, the authors evaluated two highly promiscuous Galpha(16/z) chimeras, 16z25 and 16z44, for their ability to translate GPCR activation into Ca(2+) mobilization using the fluorescence imaging plate reader (FLIPR) and aequorin. A panel of 24 G(s)- or G(i)-coupled receptors was examined for their functional association with the Galpha(16/z) chimeras. Although most of the GPCRs tested were incapable of inducing Ca(2+) mobilization upon their activation by specific agonists, the introduction of 16z25 or 16z44 allowed all of these GPCRs to mediate agonist-induced Ca(2+) mobilization. In contrast, only 16 of the GPCRs tested were capable of using Galpha(16) to mobilize intracellular Ca(2+). Analysis of dose-response curves obtained with the delta-opioid, dopamine D(1), and Xenopus melatonin Mel1c receptors revealed that the Galpha(16/z) chimeras possess better sensitivity than Galpha(16) in both the FLIPR and aequorin assays. Collectively, these studies help to validate the promiscuity of the Galpha(16/z) chimeras as well as their application in contemporary drug-screening assays that are based on ligand-induced Ca(2+) mobilization.


Assuntos
Cálcio/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/genética , Animais , Células COS , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA