Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int Immunopharmacol ; 7(7): 937-44, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17499196

RESUMO

Adapter molecule Grb2-associated binder-like protein 2 (Gab2) plays a critical role in FcepsilonRI-induced mast cell degranulation and activation. The present study aimed to investigate the pharmacological effects of an antisense oligonucleotide (ASO) targeted at Gab2 on the immune responses of rat basophilic leukemic (RBL)-2H3 cells. Gab2 ASOs were rationally designed and transfected into RBL-2H3 cells. Gab2 mRNA and protein knockdown was confirmed by real-time RT-PCR and immunoblotting, respectively. Effects of Gab2 ASO on FcepsilonRI-induced release of histamine and beta-hexosaminidase was measured by EIA and an enzymatic assay, respectively; signaling events by immunoblotting; and cytokine mRNA expression by RT-PCR. Effects of Gab2 ASO on cell adhesion and migration were performed on fibronectin-coated 96-well plate and transwells cell culture chambers, respectively. We have characterized a phosphorothioate-modified ASO targeted at Gab2 mRNA that was able to knockdown Gab2 mRNA and protein in RBL-2H3 cells. Gab2 ASO significantly blocked IgE-mediated mast cell release of beta-hexosaminidase and histamine; phosphorylation of Akt, p38 mitogen-activated protein kinase and PKCdelta; and up-regulation of cytokine mRNA levels (e.g. IL-4, -6, -9 and -13, and TNF-alpha). In addition, Gab2 ASO markedly prevented mast cell adhesion to fibronectin-coated plates and restrained random migration of RBL-2H3 cells in cell culture chambers. Our findings show that Gab2 knockdown in RBL-2H3 cells by ASO strategy can suppress many aspects of the mast cell functions and, therefore, a selective Gab2 ASO may have therapeutic potential for mast cell-dependent allergic disorders.


Assuntos
Basófilos/imunologia , Leucemia Basofílica Aguda/tratamento farmacológico , Leucemia Basofílica Aguda/imunologia , Oligonucleotídeos Antissenso/farmacologia , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Adesão Celular/genética , Movimento Celular/genética , Citocinas/genética , Citocinas/metabolismo , Fibronectinas/metabolismo , Marcação de Genes , Liberação de Histamina/genética , Liberação de Histamina/imunologia , Mastócitos/imunologia , Fosfoproteínas/biossíntese , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/genética , Células Tumorais Cultivadas , beta-N-Acetil-Hexosaminidases/imunologia
2.
Clin Exp Pharmacol Physiol ; 33(5-6): 533-40, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16700890

RESUMO

1. An antisense oligonucleotide (ASO) is a short strand of deoxyribonucleotide analogue that hybridizes with the complementary mRNA in a sequence-specific manner via Watson-Crick base pairing. Formation of the ASO-mRNA heteroduplex either triggers RNase H activity, leading to mRNA degradation, induces translational arrest by steric hindrance of ribosomal activity, interferes with mRNA maturation by inhibiting splicing or destabilizes pre-mRNA in the nucleus, resulting in downregulation of target protein expression. 2. The ASO is not only a useful experimental tool in protein target identification and validation, but also a highly selective therapeutic strategy for diseases with dysregulated protein expression. 3. In the present review, we discuss various theoretical approaches to rational design of ASO, chemical modifications of ASO, ASO delivery systems and ASO-related toxicology. Finally, we survey ASO drugs in various current clinical studies.


Assuntos
Antineoplásicos/metabolismo , Inativação Gênica , Neoplasias/metabolismo , Oligonucleotídeos Antissenso/metabolismo , RNA Mensageiro/metabolismo , Animais , Clusterina/genética , Clusterina/metabolismo , Marcação de Genes/tendências , Humanos , Neoplasias/genética , Conformação de Ácido Nucleico , Oligodesoxirribonucleotídeos , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/química , RNA Mensageiro/genética , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Tionucleotídeos/síntese química , Tionucleotídeos/genética , Tionucleotídeos/metabolismo
3.
Am J Respir Crit Care Med ; 171(6): 571-8, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15557129

RESUMO

The p38 mitogen-activated protein kinase (MAPK) plays a critical role in the activation of inflammatory cells. Therefore, we investigated the antiinflammatory effects of a respirable p38alpha MAPK antisense oligonucleotide (p38alpha-ASO) in a mouse asthma model. A potent and selective p38alpha-ASO was characterized in vitro. Inhalation of aerosolized p38alpha-ASO using an aerosol chamber dosing system produced measurable lung deposition of ASO and significant reduction of ovalbumin (OVA-)-induced increases in total cells, eosinophils, and interleukin 4 (IL-4), IL-5, and IL-13 levels in bronchoalveolar lavage fluid, and dose-dependent inhibition of airway hyperresponsiveness in allergen-challenged mice. Furthermore, inhaled p38alpha-ASO markedly inhibited OVA-induced lung tissue eosinophilia and airway mucus hypersecretion. Quantitative polymerase chain reaction analysis of bronchoalveolar lavage fluid cells and peribronchial lymph node cells showed that p38alpha-ASO significantly reduced p38alpha MAPK mRNA expression. Nose-only aerosol exposure of mice verified the p38alpha-ASO-induced inhibition of OVA-induced pulmonary eosinophilia, mucus hypersecretion, and airway hyperresponsiveness. None of the effects of the p38alpha-ASO were produced by a six-base mismatched control oligonucleotide. These findings demonstrate antisense pharmacodynamic activity in the airways after aerosol delivery and suggest that a p38alpha MAPK ASO approach may have therapeutic potential for asthma and other inflammatory lung diseases.


Assuntos
Asma/prevenção & controle , Proteína Quinase 14 Ativada por Mitógeno/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Administração por Inalação , Aerossóis , Animais , Asma/fisiopatologia , Hiper-Reatividade Brônquica , Líquido da Lavagem Broncoalveolar , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Muco/metabolismo , Ovalbumina/imunologia , Reação em Cadeia da Polimerase , Eosinofilia Pulmonar/prevenção & controle
4.
J Immunol ; 172(11): 7053-9, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15153527

RESUMO

Mitogen-activated protein kinase (MAPK) signaling cascade plays a pivotal role in the activation of inflammatory cells. Recent findings revealed that the activity of p42/44 MAPK (also known as extracellular signal-regulated kinase (ERK)) in the lungs was significantly higher in asthmatic mice than in normal controls. We hypothesized that inhibition of ERK activity may have anti-inflammatory effects in allergic asthma. BALB/c mice were sensitized with OVA and, upon OVA aerosol challenge, developed airway eosinophilia, mucus hypersecretion, elevation in cytokine and chemokine levels, up-regulation of VCAM-1 expression, and airway hyperresponsiveness. Intraperitoneal administration of U0126, a specific MAPK/ERK kinase inhibitor, significantly (p < 0.05) inhibited OVA-induced increases in total cell counts, eosinophil counts, and IL-4, IL-5, IL-13, and eotaxin levels recovered in bronchoalveolar lavage fluid in a dose-dependent manner. U0126 also substantially (p < 0.05) reduced the serum levels of total IgE and OVA-specific IgE and IgG1. Histological studies show that U0126 dramatically inhibited OVA-induced lung tissue eosinophilia, airway mucus production, and expression of VCAM-1 in lung tissues. In addition, U0126 significantly (p < 0.05) suppressed OVA-induced airway hyperresponsiveness to inhaled methacholine in a dose-dependent manner. Western blot analysis of whole lung lysates shows that U0126 markedly attenuated OVA-induced tyrosine phosphorylation of ERK1/2. Taken together, our findings implicate that inhibition of ERK signaling pathway may have therapeutic potential for the treatment of allergic airway inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Asma/tratamento farmacológico , Butadienos/farmacologia , Inibidores Enzimáticos/farmacologia , Nitrilas/farmacologia , Animais , Hiper-Reatividade Brônquica/etiologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/análise , Modelos Animais de Doenças , Eosinófilos/efeitos dos fármacos , Eosinófilos/fisiologia , Imunoglobulina G/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Muco/fisiologia , Ovalbumina/imunologia , Fosforilação , Molécula 1 de Adesão de Célula Vascular/análise
5.
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
6.
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
7.
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
8.
Biochem Biophys Res Commun ; 304(2): 417-24, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12711332

RESUMO

CC chemokine receptor 1 (CCR1) has been implicated in inflammation. The present study examined the signaling mechanisms that mediate GM-CSF/IL-10-induced synergistic CCR1 protein expression in monocytic U937 cells. GM-CSF alone markedly increased both the mRNA and protein expression of CCR1. IL-10 augmented GM-CSF-induced CCR1 protein expression with no effect on mRNA expression. PD098059 and U0126 (two MEK inhibitors), and LY294002 (a PI3K inhibitor) inhibited GM-CSF/IL-10-induced CCR1 gene and protein expression. PD098059, U0126, and LY294002 also attenuated chemotaxis of GM-CSF/IL-10-primed U937 cells in response to MIP-1alpha. Immunoblotting studies show that GM-CSF alone induced ERK2 phosphorylation; whereas, IL-10 alone induced p70(S6k) phosphorylation in U937 cells. Neither cytokine when used alone induced PKB/Akt phosphorylation. Combined GM-CSF/IL-10 treatment of U937 cells induced phosphorylation of ERK2, p70(S6k), and PKB/Akt. PD098059 and U0126 completely abrogated ERK2 phosphorylation; whereas, LY294002 completely blocked PKB/Akt and p70(S6k) phosphorylation. Our findings indicate that IL-10 may potentiate GM-CSF-induced CCR1 protein expression in U937 cells via activation of PKB/Akt and p70(S6k).


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-10/farmacologia , Monócitos/imunologia , Proteínas Serina-Treonina Quinases , Receptores de Quimiocinas/biossíntese , Quimiocina CCL3 , Quimiocina CCL4 , Quimiotaxia de Leucócito , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Humanos , Proteínas Inflamatórias de Macrófagos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/biossíntese , Receptores CCR1 , Receptores de Quimiocinas/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA