Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Virol J ; 7: 230, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20843322

RESUMO

BACKGROUND: The prevalence of HIV/hepatitis co-infection in sub-Saharan Africa is not well documented, while both HIV and HBV are endemic in this area. OBJECTIVE: The aim of this study is to determine the seroprevalence of HBV and HCV virus in HIV-infected subjects in the Gambia. METHODS: Plasma samples from HIV infected patients (190 individuals with clinically defined AIDS and 382 individuals without AIDS) were tested retrospectively for the presence of HBV sero-markers and for serum HBV DNA, screened for HCV infection by testing for anti-HCV antibody and HCV RNA. RESULTS: HBsAg prevalence in HIV-positive individuals is 12.2%. HIV/HBV co-infected individuals with CD4 count of <200 cells µL⁻¹ have a higher HBV DNA viral load than patients with higher CD4 count (log 4.0 vs. log 2.0 DNA copies/ml, p < 0.05). Males (OR = 1.8, 95% CI: 1.0, 3.2) were more likely to be HBsAg positive than female. HCV seroprevalence was 0.9% in HIV-positive individuals. CONCLUSION: The prevalence of HBsAg carriage in HIV- infected Gambians is similar to that obtained in the general population. However co-infected individuals with reduced CD4 levels, indicative of AIDS had higher prevalence of HBeAg retention and elevated HBV DNA levels compared to non-AIDS patients with higher CD4 count.


Assuntos
Infecções por HIV/complicações , Hepatite B/epidemiologia , Hepatite C/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Contagem de Linfócito CD4 , Criança , Pré-Escolar , Comorbidade , DNA Viral/sangue , Feminino , Gâmbia/epidemiologia , Infecções por HIV/virologia , HIV-1/isolamento & purificação , HIV-2/isolamento & purificação , Antígenos de Superfície da Hepatite B/sangue , Anticorpos Anti-Hepatite C/sangue , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue , Estudos Retrospectivos , Estudos Soroepidemiológicos , Adulto Jovem
2.
Curr Biol ; 12(7): 594-8, 2002 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-11937030

RESUMO

Gam1 is an early gene product of the avian adenovirus CELO and is essential for viral replication. Gam1 has no homology to any known proteins; however, its early expression and nuclear localization suggest that the protein functions to influence transcription in the infected cell. A determinant of eukaryotic gene expression is the acetylation state of chromosomal histones and other nuclear proteins. We find that Gam1 expression increases the level of transcription from a variety of eukaryotic promoters, similar to the effect of treating cells with the histone deacetylase (HDAC) inhibitor trichostatin A (TSA ). We show that Gam1 can effectively inhibit histone deacetylation by HDAC1 and that Gam1 binds to HDAC1 both in vitro and in vivo. A CELO virus lacking Gam1 (CELOdG) is replication defective, but the defect can be overcome by either expressing an interfering HDAC1 mutant or by treating infected cells with TSA. The identification of a viral early gene product having the specific function of binding and inactivating HDAC suggests that deacetylase complexes play an important role in limiting early gene expression from invading viruses.


Assuntos
Aviadenovirus/metabolismo , Inibidores de Histona Desacetilases , Proteínas Imediatamente Precoces/metabolismo , Proteínas Repressoras , Proteínas Virais/metabolismo , Animais , Aviadenovirus/genética , Ativação Enzimática , Expressão Gênica , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Proteínas de Choque Térmico/genética , Histona Desacetilase 1 , Histona Desacetilase 2 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas Imediatamente Precoces/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética
3.
Cancer Res ; 65(2): 379-82, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695376

RESUMO

Targeted inhibition of protein kinases with small molecule drugs has evolved into a viable approach for anticancer therapy. However, the true selectivity of these therapeutic agents has remained unclear. Here, we used a proteomic method to profile the cellular targets of the clinical epidermal growth factor receptor kinase inhibitor gefitinib. Our data suggest alternative cellular modes of action for gefitinib and provide rationales for the development of related drugs.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Animais , Células COS , Chlorocebus aethiops , Receptores ErbB/antagonistas & inibidores , Gefitinibe , Células HeLa , Humanos , Proteômica/métodos , Relação Estrutura-Atividade
4.
Cancer Res ; 65(15): 6919-26, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061676

RESUMO

Knowledge about molecular drug action is critical for the development of protein kinase inhibitors for cancer therapy. Here, we establish a chemical proteomic approach to profile the anticancer drug SU6668, which was originally designed as a selective inhibitor of receptor tyrosine kinases involved in tumor vascularization. By employing immobilized SU6668 for the affinity capture of cellular drug targets in combination with mass spectrometry, we identified previously unknown targets of SU6668 including Aurora kinases and TANK-binding kinase 1. Importantly, a cell cycle block induced by SU6668 could be attributed to inhibition of Aurora kinase activity. Moreover, SU6668 potently suppressed antiviral and inflammatory responses by interfering with TANK-binding kinase 1-mediated signal transmission. These results show the potential of chemical proteomics to provide rationales for the development of potent kinase inhibitors, which combine rather unexpected biological modes of action by simultaneously targeting defined sets of both serine/threonine and tyrosine kinases involved in cancer progression.


Assuntos
Inibidores da Angiogênese/farmacologia , Indóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Animais , Aurora Quinases , Células COS , Divisão Celular/efeitos dos fármacos , Chlorocebus aethiops , Células HeLa , Humanos , Oxindóis , Propionatos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transfecção
5.
Nucleic Acids Res ; 31(21): e128, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14576328

RESUMO

Faced with the current wealth of genomic data, it is essential to have robust and reliable methods of converting DNA sequences into their functional gene products. We demonstrate here that when conditions are established that take advantage of the replication-associated virus amplification, the virus-induced shutdown of host protein synthesis as well as the activation of signalling pathways that normally occur during virus replication, adenovirus biology can be exploited to generate a potent kinase expression system. Residual virus in the protein production has always been a limitation for adenovirus systems and we describe a DNA intercalator/ultraviolet light treatment that eliminates residual adenovirus in protein preparations that has no deleterious effect on enzyme activity. The use of mammalian cells in combination with adenovirus generated a variety of active enzymes which could not be produced in Escherichia coli or baculovirus-infected insect cells. Thus, the utility of adenovirus-mediated enzyme expression as a versatile alternative to established protein production technologies is demonstrated.


Assuntos
Adenoviridae/fisiologia , Mamíferos , Proteínas Quinases/biossíntese , Proteínas Quinases/genética , Replicação Viral , Adenoviridae/efeitos dos fármacos , Adenoviridae/genética , Adenoviridae/efeitos da radiação , Animais , Baculoviridae/genética , Baculoviridae/fisiologia , Linhagem Celular , Cromatografia de Afinidade , Contaminação de Medicamentos , Ativação Enzimática , Engenharia Genética , Humanos , Substâncias Intercalantes/farmacologia , Mamíferos/virologia , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/isolamento & purificação , Complexos Multienzimáticos/metabolismo , Proteínas Quinases/isolamento & purificação , Proteínas Quinases/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transfecção , Raios Ultravioleta
6.
J Mol Biol ; 328(4): 779-90, 2003 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-12729754

RESUMO

Chicken embryo lethal orphan adenovirus (CELO) is used as a vector for expression of exogenous genes in mammalian cells. Here, we analyzed transcriptional alterations in mouse epithelial host cells following infection with CELO using cDNA microarray analysis. Sequence data characterization revealed that a major portion of CELO-induced genes contained short interspersed nuclear elements of the B2 subclass (B2 SINEs). In fact, we could identify SINEs and other repetitive sequences as contributing significantly to the cDNAs used for microarray construction. Moreover, we found that the CELO protein Gam1 was able to mediate transcriptional activation of these B2 SINE-containing RNAs. We hypothesize that upregulation of B2-SINE-containing RNAs could be a novel contribution of Gam1 to CELO host cell infection.


Assuntos
Aviadenovirus/genética , Células Epiteliais/virologia , Genoma Viral , Elementos Nucleotídeos Curtos e Dispersos/genética , Transcrição Gênica , Animais , Sequência de Bases , Embrião de Galinha , DNA Complementar/metabolismo , Bases de Dados como Assunto , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Proteoma , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/biossíntese
7.
J Biol Chem ; 283(18): 11964-71, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18310079

RESUMO

The activation of p38 MAPK by dual phosphorylation aggravates myocardial ischemic injury and depresses cardiac contractile function. SB203580, an ATP-competitive inhibitor of p38 MAPK and other kinases, prevents this dual phosphorylation during ischemia. Studies in non-cardiac tissue have shown receptor-interacting protein 2 (RIP2) lies upstream of p38 MAPK, is SB203580-sensitive and ischemia-responsive, and aggravates ischemic injury. We therefore examined the RIP2-p38 MAPK signaling axis in the heart. Adenovirus-driven expression of wild-type RIP2 in adult rat ventricular myocytes caused robust, SB203580-sensitive dual phosphorylation of p38 MAPK associated with activation of p38 MAPK kinases MKK3, MKK4, and MKK6. The effect of SB203580 was recapitulated by unrelated inhibitors of RIP2 or the downstream MAPK kinase kinase, TAK1. However, overexpression of wild-type, kinase-dead, caspase recruitment domain-deleted, or kinase-dead and caspase recruitment domain-deleted forms of RIP2 had no effect on the activating dual phosphorylation of p38 MAPK during simulated ischemia. Similarly, p38 MAPK activation and myocardial infarction size in response to true ischemia did not differ between hearts from wild-type and RIP2 null mice. However, both p38 MAPK activation and the contractile depression caused by the endotoxin component muramyl dipeptide were attenuated by SB203580 and in RIP2 null hearts. Although RIP2 can cause myocardial p38 MAPK dual phosphorylation in the heart under some circumstances, it is not responsible for the SB203580-sensitive pattern of activation during ischemia.


Assuntos
Miocárdio/enzimologia , Miocárdio/patologia , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Imidazóis/farmacologia , Técnicas In Vitro , Masculino , Camundongos , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Estrutura Terciária de Proteína , Piridinas/farmacologia , Pirimidinas/farmacologia , Ratos , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/antagonistas & inibidores , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
8.
J Gen Virol ; 86(Pt 2): 285-295, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15659747

RESUMO

The activation of NF-kappaB has long been considered a positive factor for human cytomegalovirus (HCMV) replication. The HCMV immediate-early promoter, the initial transcriptional element in the HCMV replication cycle, is activated by the transcription factor NF-kappaB, and several HCMV gene products have been demonstrated to activate this transcription factor. However, the role of NF-kappaB in the full replication cycle of the virus has not been carefully examined. A series of experiments that demonstrate an important inhibitory role of NF-kappaB for HCMV replication in fibroblasts is presented here. Using both genetic and pharmaceutical methods, it was shown that blocking NF-kappaB activation in cell culture does not inhibit HCMV replication, but rather leads to a modest increase in replication. Two cytokines inhibitory for HCMV, tumour necrosis factor-alpha and interferon-gamma, no longer inhibit HCMV when NF-kappaB activation is blocked. Furthermore, forced expression of the NF-kappaB activating IkappaB kinase beta (IKKbeta), but not a kinase inactive mutant, also inhibits HCMV replication. In addition, it was shown that NF-kappaB signalling is essential for the production of an anti-viral factor in the supernatant of HCMV-infected fibroblasts, and identified interferon-beta as this factor. Thus, the role of NF-kappaB in fibroblasts is to activate a host defence against HCMV.


Assuntos
Citomegalovirus/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais , Animais , Células Cultivadas , Citomegalovirus/imunologia , Fibroblastos , Humanos , Quinase I-kappa B , Interferon beta/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Replicação Viral
9.
J Gene Med ; 6(4): 464-70, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15079821

RESUMO

BACKGROUND: Dendritic cells (DCs) are the most potent antigen-presenting cells in initiating primary immune responses. Given the unique properties of DCs, gene-modified DCs represent a particularly attractive approach for immunotherapy of diseases such as cancer. METHODS: Gene-modified DCs were obtained by a receptor-mediated gene delivery system using adenovirus (Ad) particles as ligand and RNA or DNA condensed by polyethylenimine (PEI). In vitro transcribed polyadenylated or non-polyadenylated RNA was used. RNA-transduced DCs were generated expressing chicken ovalbumin (OVA) or chimeric constructs thereof, and compared with DNA-transduced DCs. RESULTS: Ad/PEI transfection complexes efficiently delivered RNA into DCs. Such RNA-transduced DCs induced OVA-specific T cell responses more effectively than DNA-transduced DCs. Furthermore, DCs transduced with polyadenylated RNA were more potent in stimulating CD4(+) and CD8(+) T cell responses than DCs transduced with non-polyadenylated RNA and this was particularly important for CD4(+) T cell responses. CONCLUSIONS: Ad/PEI/RNA transfection is an efficient means for generating RNA-transduced DCs and for stimulating antigen-specific T cell responses. Polyadenylation of RNA enhances CD8(+) T cell responses and is essential for CD4(+) T cell responses.


Assuntos
Adenoviridae/genética , Células Dendríticas/fisiologia , RNA/administração & dosagem , Linfócitos T/imunologia , Linfócitos T/fisiologia , Transdução Genética/métodos , Animais , Antígenos/genética , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Células Cultivadas , DNA/administração & dosagem , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária/genética , Substâncias Macromoleculares , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/genética , Poliadenilação , Polietilenoimina , RNA/química , Transfecção/métodos
10.
J Gen Virol ; 82(Pt 6): 1465-1472, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11369892

RESUMO

The coxsackievirus and adenovirus receptor (CAR) is a high affinity receptor used by adenoviruses, including adenovirus type 5 (Ad5). The adenovirus fibre molecule bears the high affinity cell binding domain of Ad5, allowing virions to attach to CAR. The avian adenovirus CELO displays two fibre molecules on its capsid and it was logical to expect that the cell binding functions of CELO might also reside in one or both of these fibres. We had previously shown that the cell binding properties of CELO resemble Ad5, suggesting that the two viruses use similar receptors. Experiments with CAR-deficient CHO cells and CHO cells modified to express CAR demonstrated that CELO has CAR-dependent transduction behaviour like Ad5. Mutations were introduced into the CELO genome to disrupt either the long fibre 1 or the short fibre 2. A CELO genome with fibre 2 disrupted did not generate virus, demonstrating that fibre 2 is essential for some stage in virus growth, assembly or spread. However, a CELO genome with disrupted fibre 1 gene produced virus (CELOdF1) that was capable of entering chicken cells, but had lost both the ability to efficiently transduce human cells and the CAR-specific transduction displayed by wild-type CELO. The ability of CELOdF1 to transduce chicken cells suggests that CELOdF1 may still bind, probably via fibre 2, to a receptor expressed on avian but not mammalian cells. CELOdF1 replication was dramatically impaired in chicken embryos, demonstrating that fibre 1 is important for the in vivo biology of CELO.


Assuntos
Aviadenovirus/metabolismo , Proteínas do Capsídeo , Capsídeo/metabolismo , Deleção de Genes , Receptores Virais/metabolismo , Animais , Aviadenovirus/genética , Aviadenovirus/crescimento & desenvolvimento , Western Blotting , Células CHO , Capsídeo/genética , Embrião de Galinha , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Cricetinae , Fibroblastos , Genes Essenciais/genética , Genes Virais/genética , Hepatócitos , Humanos , Camundongos , Receptores Virais/genética , Especificidade da Espécie , Especificidade por Substrato , Transdução Genética , Células Tumorais Cultivadas , Montagem de Vírus , Replicação Viral
11.
Somat Cell Mol Genet ; 27(1-6): 65-74, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12774941

RESUMO

Dendritic cells are professional antigen presenting cells and are unique in their ability to prime naïve T cells. Gene modification of dendritic cells is of particular interest for immunotherapy of diseases where the immune system has failed or is aberrantly regulated, such as in cancer or autoimmune disease, respectively. Dendritic cells abundantly express mannose receptor and mannose receptor-related receptors, and receptor-mediated gene transfer via mannose receptor offers a versatile tool for targeted gene delivery into these cells. Accordingly, mannose polyethylenimine DNA transfer complexes were generated and used for gene delivery into dendritic cells. Mannose receptor belongs to the group of scavenger receptors that allow dendritic cells to take up pathogenic material, which is directed for degradation and MHC class II presentation. Therefore, a limiting step of transgene expression by mannose receptor-mediated gene delivery is endosomal degradation of DNA. Several strategies have been explored to overcome this limitation including the addition of endosomolytic components to DNA transfer complexes like adenovirus particles and influenza peptides. Here, we review the current understanding of mannose receptor-mediated gene delivery into dendritic cells and discuss strategies to identify appropriate endosomolytic agents to improve DNA transfer efficacy.


Assuntos
Células Apresentadoras de Antígenos/efeitos dos fármacos , Antígenos/metabolismo , Células Dendríticas/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Técnicas de Transferência de Genes , Lectinas Tipo C , Lectinas de Ligação a Manose , Receptores de Superfície Celular/metabolismo , Adenoviridae , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos/efeitos dos fármacos , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/farmacologia , DNA/administração & dosagem , DNA/química , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Humanos , Substâncias Macromoleculares , Receptor de Manose , Polietilenoimina/metabolismo , Polietilenoimina/farmacologia
12.
J Virol ; 76(16): 8124-37, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12134018

RESUMO

Phosphorylation of hepatitis B virus (HBV) core protein has recently been shown to be a prerequisite for pregenomic RNA encapsidation into viral capsids, but the host cell kinases mediating this essential step of the HBV replication cycle have not been identified. We detected two kinases of 95 and 115 kDa in HuH-7 total cell lysates which interacted specifically with the HBV core protein and phosphorylated its arginine-rich C-terminal domain. The 95-kDa kinase was purified and characterized as SR protein-specific kinase 1 (SRPK1) by mass spectrometry. Based on this finding, the 115-kDa kinase could be identified as the related kinase SRPK2 by immunoblot analysis. In vitro, both SRPKs phosphorylated HBV core protein on the same serine residues which are found to be phosphorylated in vivo. Moreover, the major cellular HBV core kinase activity detected in the total cell lysate showed biochemical properties identical to those of SRPK1 and SRPK2, as examined by measuring binding to a panel of chromatography media. We also clearly demonstrate that neither the cyclin-dependent kinases Cdc2 and Cdk2 nor protein kinase C, previously implicated in HBV core protein phosphorylation, can account for the HBV core protein kinase activity. We conclude that both SRPK1 and SRPK2 are most likely the cellular protein kinases mediating HBV core protein phosphorylation during viral infection and therefore represent important host cell targets for therapeutic intervention in HBV infection.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Antígenos do Núcleo do Vírus da Hepatite B/metabolismo , Vírus da Hepatite B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas do Core Viral/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Células COS , Linhagem Celular , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Hepatite B/tratamento farmacológico , Hepatite B/enzimologia , Hepatite B/virologia , Antígenos do Núcleo do Vírus da Hepatite B/genética , Vírus da Hepatite B/genética , Vírus da Hepatite B/fisiologia , Humanos , Técnicas In Vitro , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas do Core Viral/genética , Replicação Viral
13.
J Biol Chem ; 279(10): 9642-52, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-14670961

RESUMO

The adapter kinase receptor interacting protein-like interacting caspase-like apoptosis regulatory protein kinase (RICK, also called RIP2 and CARDIAK) was found to be elevated at both the protein and RNA levels during human cytomegalovirus (HCMV) replication, suggesting either that the virus may require RICK for replication or that RICK is part of an unsuccessful host attempt to inhibit HCMV replication. It is demonstrated here that forced expression of RICK in either a kinase active or inactive form activates nuclear factor (NF)-kappaB by means of its intermediate domain and potently blocks HCMV replication in human fibroblasts. Importantly, NF-kappaB activation, which exerted a modestly positive effect on the early phase of infection, clearly had a strongly negative impact during later viral steps. A stable inhibitor of NF-kappaB (IkappaB) reverses the RICK inhibitory effect, and activation of NF-kappaB by IkappaB kinase beta expression is inhibitory to HCMV, demonstrating that NF-kappaB activation is part of a potent anti-HCMV response. Supernatant transfer experiments identified interferon-beta as a downstream component of the RICK inhibitory pathway. RICK expression was found to synergize with HCMV infection in the induction of interferon-beta expression. This study identifies an endogenous RICK-activated, NF-kappaB- and interferon-beta-dependent antiviral pathway that is either inhibited or faulty under normal HCMV replication conditions; efforts to bolster this pathway may lead to novel anti-viral approaches.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , NF-kappa B/metabolismo , Proteínas Quinases/metabolismo , Linhagem Celular , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Replicação Viral
14.
Mol Cell Proteomics ; 3(12): 1181-93, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15475568

RESUMO

Small molecule inhibitors belonging to the pyrido[2,3-d]pyrimidine class of compounds were developed as antagonists of protein tyrosine kinases implicated in cancer progression. Derivatives from this compound class are effective against most of the imatinib mesylate-resistant BCR-ABL mutants isolated from advanced chronic myeloid leukemia patients. Here, we established an efficient proteomics method employing an immobilized pyrido[2,3-d]pyrimidine ligand as an affinity probe and identified more than 30 human protein kinases affected by this class of compounds. Remarkably, in vitro kinase assays revealed that the serine/threonine kinases Rip-like interacting caspase-like apoptosis-regulatory protein kinase (RICK) and p38alpha were among the most potently inhibited kinase targets. Thus, pyrido[2,3-d]pyrimidines did not discriminate between tyrosine and serine/threonine kinases. Instead, we found that these inhibitors are quite selective for protein kinases possessing a conserved small amino acid residue such as threonine at a critical site of the ATP binding pocket. We further demonstrated inhibition of both p38 and RICK kinase activities in intact cells upon pyrido[2,3-d]pyrimidine inhibitor treatment. Moreover, the established functions of these two kinases as signal transducers of inflammatory responses could be correlated with a potent in vivo inhibition of cytokine production by a pyrido[2,3-d]pyrimidine compound. Thus, our data demonstrate the utility of proteomic methods employing immobilized kinase inhibitors for identifying new targets linked to previously unrecognized therapeutic applications.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteômica/métodos , Piridinas/farmacologia , Pirimidinas/farmacologia , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Apoptose , Sítios de Ligação , Células COS , Linhagem Celular Tumoral , Células Cultivadas , Cromatografia Líquida , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/química , Células HeLa , Humanos , Inflamação , Concentração Inibidora 50 , Ligantes , Lipopolissacarídeos/química , Espectrometria de Massas , Modelos Químicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmídeos/metabolismo , Polietilenoglicóis/química , Inibidores de Proteínas Quinases/química , Piridinas/química , Pirimidinas/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo
15.
Proc Natl Acad Sci U S A ; 100(26): 15434-9, 2003 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-14668439

RESUMO

Small molecule inhibitors of protein kinases are widely used in signal transduction research and are emerging as a major class of drugs. Although interpretation of biological results obtained with these reagents critically depends on their selectivity, efficient methods for proteome-wide assessment of kinase inhibitor selectivity have not yet been reported. Here, we address this important issue and describe a method for identifying targets of the widely used p38 kinase inhibitor SB 203580. Immobilization of a suitable SB 203580 analogue and thoroughly optimized biochemical conditions for affinity chromatography permitted the dramatic enrichment and identification of several previously unknown protein kinase targets of SB 203580. In vitro kinase assays showed that cyclin G-associated kinase (GAK) and CK1 were almost as potently inhibited as p38alpha whereas RICK [Rip-like interacting caspase-like apoptosis-regulatory protein (CLARP) kinase/Rip2/CARDIAK] was even more sensitive to inhibition by SB 203580. The cellular kinase activity of RICK, a known signal transducer of inflammatory responses, was already inhibited by submicromolar concentrations of SB 203580 in intact cells. Therefore, our results warrant a reevaluation of the vast amount of data obtained with SB 203580 and might have significant implications on the development of p38 inhibitors as antiinflammatory drugs. Based on the procedures described here, efficient affinity purification techniques can be developed for other protein kinase inhibitors, providing crucial information about their cellular modes of action.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/química , Inibidores de Proteínas Quinases , Proteômica , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Espectrometria de Massas , Proteínas Quinases Ativadas por Mitógeno/isolamento & purificação , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
16.
J Gen Virol ; 84(Pt 3): 555-560, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12604805

RESUMO

The hepatitis C virus (HCV) NS5A protein is highly phosphorylated by cellular protein kinases. To study how NS5A might be integrated in cellular kinase signalling, we isolated phosphoproteins from HuH-7 hepatoma cells that specifically interacted with recombinant NS5A protein. Subsequent mass spectrometry identified the adaptor protein amphiphysin II as a novel interaction partner of NS5A. Mutational analysis revealed that complex formation is primarily mediated by a proline-rich region in the C-terminal part of NS5A, which interacts with the amphiphysin II Src homology 3 domain. Importantly, we could further demonstrate specific co-precipitation and cellular co-localization of endogenous amphiphysin II with NS5A in HuH-7 cells carrying a persistently replicating subgenomic HCV replicon. Although the NS5A-amphiphysin II interaction appeared to be dispensable for replication of these HCV RNAs in cell culture, our results indicate that NS5A-amphiphysin II complex formation might be of physiological relevance for the HCV life cycle.


Assuntos
Hepacivirus/fisiologia , Proteínas do Tecido Nervoso/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/metabolismo , Citoplasma/química , Técnica Indireta de Fluorescência para Anticorpo , Hepacivirus/química , Hepacivirus/genética , Humanos , Immunoblotting , Zíper de Leucina , MAP Quinase Quinase Quinases , Mutação , Proteínas do Tecido Nervoso/química , Prolina , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Proteínas Recombinantes/metabolismo , Replicon , Células Tumorais Cultivadas , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
17.
EMBO J ; 21(17): 4621-31, 2002 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12198164

RESUMO

The mammalian SIN3 complex consists of histone deacetylases (HDAC1, HDAC2), several known proteins (SAP30, N-CoR) and as yet unidentified proteins. Here we show that the mouse tetradecanoyl phorbol acetate induced sequence 7 (TIS7) protein is a novel transcriptional co-repressor that can associate with the SIN3 complex. We have identified tis7 as a gene that is up-regulated upon loss of polarity in a mouse mammary gland epithelial cell line expressing an estrogen-inducible c-JunER fusion protein. In unpolarized cells, TIS7 protein levels increase and TIS7 translocates into the nucleus. Overexpression of tis7 causes loss of polarity and represses a set of genes, as revealed by cDNA microarray analysis. We have shown that TIS7 protein interacts with several proteins of the SIN3 complex (mSin3B, HDAC1, N-CoR and SAP30) by yeast two-hybrid screening and co-immunoprecipitations. TIS7 co-immunoprecipitated HDAC complex is enzymatically active and represses a GAL4-dependent reporter transcription. The transcriptional repression of endogenous genes by tis7 overexpression is HDAC dependent. Thus, we propose TIS7 as a transcriptional co-repressor affecting the expression of specific genes in a HDAC activity-dependent manner during cell fate decisions, e.g. scattering.


Assuntos
Células Epiteliais/metabolismo , Histona Desacetilases/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Polaridade Celular , DNA Complementar/genética , Estradiol/farmacologia , Feminino , Perfilação da Expressão Gênica , Células HeLa , Histona Desacetilase 1 , Histona Desacetilase 2 , Humanos , Glândulas Mamárias Animais/citologia , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , Análise de Sequência com Séries de Oligonucleotídeos , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-jun/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Complexo Correpressor Histona Desacetilase e Sin3 , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA