Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Microbiol ; 14(2): 155-67, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22044780

RESUMO

Bacterial infections cause substantial mortality and burden of disease globally. Induction of a strong innate inflammatory response is the first common host mechanism required for elimination of the invading pathogens. The host transcription factor, nuclear factor kappa B (NF-κB) is essential for immune activation. Conversely, bacterial pathogens have evolved strategies to interfere directly with host cell signalling by regulating or mimicking host proteins. Given the key role of NF-κB in the host inflammatory response, bacteria have expectedly developed virulence effectors interfering with NF-κB signalling pathways. In this review, we explore the bacterial mechanisms utilized to prevent effective NF-κB signalling, which in turn usurp the host inflammatory response.


Assuntos
Bactérias/imunologia , Bactérias/patogenicidade , Evasão da Resposta Imune , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Animais , Proteínas de Bactérias/metabolismo , Humanos , Modelos Biológicos , Fatores de Virulência/metabolismo
2.
Cell Microbiol ; 14(2): 168-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22050732

RESUMO

Viruses are the most abundant and diverse pathogens challenging the host immune system, and as such are a severe threat to human health. To this end, viruses have evolved multiple strategies to evade and subvert the host immune response. Host-pathogen interactions are usually initiated via recognition of pathogen-associated molecular patterns (PAMPs) by host sensors known as pattern recognition receptors (PRRs), which include, Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs) and DNA receptors. Effective sensing of PAMPs rapidly triggers host immune responses, via activation of complex signalling pathways that culminates in the induction of inflammatory responses and the eradication of pathogens. Activation of the nuclear factor-κB (NF-κB) transcription pathway is crucial for the immediate early step of immune activation. This review discusses the recent evidence describing a variety of viral effectors that have been shown to prevent NF-κB signalling. Most of these viral effectors can be broadly classified into three categories based on the site of inhibition within the NF-κB pathway, that is, at the (i) TLRs, (ii) IKK complex or (iii) the transcriptional level.


Assuntos
Evasão da Resposta Imune , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Vírus/imunologia , Vírus/patogenicidade , Animais , Humanos , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo
3.
Proc Natl Acad Sci U S A ; 106(10): 3935-40, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19223583

RESUMO

NLRP1 (NLR family, pyrin domain-containing 1) is a contributor to innate immunity involved in intracellular sensing of pathogens, as well as danger signals related to cell injury. NLRP1 is one of the core components of caspase-1-activating platforms termed "inflammasomes," which are involved in proteolytic processing of interleukin-1beta (IL-1beta) and in cell death. We previously discovered that anti-apoptotic proteins Bcl-2 and Bcl-X(L) bind to and inhibit NLRP1 in cells. Using an in vitro reconstituted system employing purified recombinant proteins, we studied the mechanism by which Bcl-2 and Bcl-X(L) inhibit NLRP1. Bcl-2 and Bcl-X(L) inhibited caspase-1 activation induced by NLRP1 in a concentration-dependent manner, with K(i) approximately 10 nM. Bcl-2 and Bcl-X(L) were also determined to inhibit ATP binding to NLRP1, which is required for oligomerization of NLRP1, and Bcl-X(L) was demonstrated to interfere with NLRP1 oligomerization. Deletion of the flexible loop regions of Bcl-2 and Bcl-X(L), which are located between the first and second alpha-helices of these anti-apoptotic proteins and which were previously shown to be required for binding NLRP1, abrogated ability to inhibit caspase-1 activation, ATP binding and oligomerization of NLRP1. Conversely, synthetic peptides corresponding to the loop region of Bcl-2 were sufficient to potently inhibit NLRP1. These findings thus demonstrate that the loop domain is necessary and sufficient to inhibit NLRP1, providing insights into the mechanism by which anti-apoptotic proteins Bcl-2 and Bcl-X(L) inhibit NLRP1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Inflamação/metabolismo , Proteína bcl-X/química , Proteína bcl-X/metabolismo , Caspase 1/metabolismo , Ativação Enzimática , Cinética , Peptídeos/química , Peptídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
4.
Proc Natl Acad Sci U S A ; 106(34): 14524-9, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19667203

RESUMO

NOD1 and NOD2 are members of the NOD-like receptor (NLR) protein family that are involved in sensing the presence of pathogens and are a component of the innate immune system. Upon activation by specific bacterial peptides derived from peptidoglycans, NODs interact via a CARD-CARD interaction with the receptor-interacting protein kinase RIP2, an inducer of NF-kappaB activation. In this report, we show that NOD signaling is dependent on XIAP, a member of the inhibitor of apoptosis protein (IAP) family. Cells deficient in XIAP exhibit a marked reduction in NF-kappaB activation induced by microbial NOD ligands and by over-expression of NOD1 or NOD2. Moreover, we show that XIAP interacts with RIP2 via its BIR2 domain, which could be disrupted by XIAP antagonists SMAC and SMAC-mimicking compounds. Both NOD1 and NOD2 associated with XIAP in a RIP2-dependent manner, providing evidence that XIAP associates with the NOD signalosome. Taken together, our data suggest a role for XIAP in regulating innate immune responses by interacting with NOD1 and NOD2 through interaction with RIP2.


Assuntos
Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Transdução de Sinais , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HCT116 , Humanos , Immunoblotting , Imunoprecipitação , Interleucina-8/metabolismo , Luciferases/genética , Luciferases/metabolismo , Mutação , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Ligação Proteica , RNA Interferente Pequeno/genética , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
5.
Cancer Immunol Immunother ; 58(5): 769-75, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18633610

RESUMO

Intravenously-applied bacteria tend to accumulate in tumors and can sporadically lead to tumor regression. Systemic administration of attenuated Salmonella typhimurium is safe and has shown no significant adverse effects in humans. The purpose of this study was to test the hypothesis that engineering S. typhimurium to express a chemokine, CCL21, would increase anti-tumor activity. We engineered an attenuated strain of S. typhimurium to produce the chemokine CCL21. Attenuated S. typhimurium expressing CCL21 significantly inhibited the growth of primary tumors and pulmonary metastases in preclinical models of multi-drug-resistant murine carcinomas, while control bacteria did not. Histological analysis of tumors showed marked inflammatory cell infiltrates in mice treated with CCL21-expressing but not control bacteria. Levels of cytokines and chemokines known to be induced by CCL21 [e.g., interferon-gamma (INFgamma), CXCL9, and CXCL10] were significantly elevated in tumors of mice treated with CCL21-expressing but not control S. typhimurium. The anti-tumor activity was found to be dependent on CD4- and CD8-expressing cells, based on antibody-mediated in vivo immuno-depletion experiments. Anti-tumor activity was achieved without evidence of toxicity. In summary, chemokine-expressing, attenuated bacteria may provide a novel approach to cancer immunotherapy for effective and well-tolerated in vivo delivery of immunomodulatory proteins.


Assuntos
Adenocarcinoma/terapia , Vacinas Bacterianas/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Quimiocina CCL21/fisiologia , Imunoterapia Ativa , Melanoma Experimental/terapia , Salmonella typhimurium/genética , Adenocarcinoma/química , Adenocarcinoma/imunologia , Adenocarcinoma/prevenção & controle , Adenocarcinoma/secundário , Animais , Neoplasias da Mama/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral/transplante , Quimiocina CCL21/genética , Quimiocinas/análise , Neoplasias do Colo/patologia , Citocinas/análise , Feminino , Neoplasias Pulmonares/química , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/química , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/fisiologia , Salmonella typhimurium/metabolismo , Vacinas Atenuadas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Microbiol ; 10(9): 1879-92, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18503636

RESUMO

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that causes various human diseases, including blindness caused by ocular infection and sexually transmitted diseases resulting from urogenital infection. After infecting host cells, Chlamydiae avoid alarming the host's immune system. Among the immune evasion mechanisms, Chlamydiae can inhibit NF-kappaB activation, a crucial pathway for host inflammatory responses. In this study, we show that ChlaDub1, a deubiquitinating and deNeddylating protease from C. trachomatis, is expressed in infected cells. In transfection experiments, ChlaDub1 suppresses NF-kappaB activation induced by several pro-inflammatory stimuli and binds the NF-kappaB inhibitory subunit IkappaBalpha, impairing its ubiquitination and degradation. Thus, we provide further insight into the mechanism by which C. trachomatis may evade the host inflammatory response by demonstrating that ChlaDub1, a protease produced by this microorganism, is capable of inhibiting IkappaBalpha degradation and blocking NF-kappaB activation.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/enzimologia , Endopeptidases/metabolismo , Proteínas I-kappa B/metabolismo , NF-kappa B/antagonistas & inibidores , Proteínas de Bactérias/genética , Linhagem Celular , Infecções por Chlamydia/microbiologia , Regulação para Baixo , Endopeptidases/genética , Humanos , Inibidor de NF-kappaB alfa , Ligação Proteica , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Transfecção , Proteases Específicas de Ubiquitina , Ubiquitinação
8.
Mol Immunol ; 46(6): 1163-70, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19121870

RESUMO

RIP2/RICK/CARDIAK is a member of the receptor interacting protein kinase (RIP) family. RIP2 promotes NF-kappaB activation as well as activation of the MAPKs JNK, ERK1/2 and p38 MAPK, thereby playing an emergent role in the innate immune response and NOD signaling. Moreover, RIP2 has been shown to interact with the CARD of caspase-1 and to induce IL-1beta maturation as well as in the induction of CD95-mediated programmed cell death by enhancing caspase-8 activity. Here, we report the identification and characterization of a novel alternative mRNA splice variant of RIP2, encoding a protein designated RIP2-beta, comprised of only a portion of the N-terminal kinase domain and lacking the intermediate region and C-terminal CARD. As revealed by gene transfer experiments, these structural changes in RIP2-beta are associated with a loss of activation with respect to NF-kappaB and MAPK activation, IL-1beta secretion, and caspase-8-mediated apoptosis. In conclusion, alternative mRNA splicing may be involved in the regulation of RIP2 actions, underlying the complexity of RIP2-dependent pathways regulating stress signaling and apoptosis.


Assuntos
Processamento Alternativo , RNA Mensageiro/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Sequência de Aminoácidos , Apoptose , Sequência de Bases , Caspase 8/metabolismo , Linhagem Celular , Humanos , Interleucina-1beta/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , NF-kappa B/metabolismo , RNA Mensageiro/genética , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Transdução de Sinais
9.
J Biol Chem ; 284(32): 21386-92, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19535337

RESUMO

Macrophages detect pathogen infection via the activation of their plasma membrane-bound Toll-like receptor proteins (TLRs). The heterotypic interaction between the Toll/interleukin-1 receptor (TIR) domains of TLRs and adaptor proteins, like Myeloid differentiation primary response gene 88 (MyD88), is the first intracellular step in the signaling pathway of the mammalian innate immune response. The hetero-oligomerization of the TIRs of the receptor and adaptor brings about the activation of the transcription factor NF-kappaB, which regulates the synthesis of pro-inflammatory cytokines. Here, we report the first crystal structure of a bacterial TIR domain solved at 2.5 A resolution. The three-dimensional fold of Paracoccus denitrificans TIR is identical to that observed for the TIR of human TLRs and MyD88 proteins. The structure shows a unique dimerization interface involving the DD-loop and EE-loop residues, whereas leaving the BB-loop highly exposed. Peptide amide hydrogen-deuterium exchange mass spectrometry also reveals that the same region is used for dimerization in solution and in the context of the full-length protein. These results, together with a functional interaction between P. denitrificans TIR and MyD88 visualized in a co-immunoprecipitation assay, further substantiate the model that bacterial TIR proteins adopt structural mimicry of the host active receptor TIR domains to interfere with the signaling of TLRs and their adaptors to decrease the inflammatory response.


Assuntos
Proteínas de Bactérias/química , Escherichia coli/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Paracoccus denitrificans/metabolismo , Cristalografia por Raios X/métodos , Citocinas/metabolismo , Dimerização , Humanos , Imunoprecipitação , Inflamação , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Fator 88 de Diferenciação Mieloide/química , Estrutura Terciária de Proteína , Receptores Toll-Like/química
10.
J Natl Cancer Inst ; 100(15): 1113-6, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18664657

RESUMO

Intravenous administration of bacteria leads to their accumulation in tumors and to sporadic tumor regression. We therefore explored the hypothesis that Salmonella typhimurium engineered to express the proapoptotic cytokine Fas ligand (FasL) would exhibit enhanced antitumor activity. Immunocompetent mice carrying tumors derived from syngeneic murine D2F2 breast carcinoma or CT-26 colon carcinoma cells were treated intravenously with FasL-expressing S. typhimurium or with phosphate-buffered saline (PBS; control). Treatment with FasL-expressing S. typhimurium inhibited growth of primary tumors by an average of 59% for D2F2 tumors and 82% for CT-26 tumors (eg, at 25 days after initial treatment, mean volume of PBS-treated CT-26 colon carcinomas = 1385 mm(3) and of S. typhimurium FasL-treated CT-26 tumors = 243 mm(3), difference = 1142 mm(3), 95% confidence interval = 800 mm(3) to 1484 mm(3), P < .001). Pulmonary D2F2 metastases (as measured by lung weight) were reduced by 34% in S. typhimurium FasL-treated mice compared with PBS-treated mice. FasL-expressing S. typhimurium had similar effects on growth of murine B16 melanoma tumors in wild-type mice but not in lpr/lpr mice, which lack Fas, or in mice with disrupted host inflammatory responses. Antitumor activity was achieved without overt toxicity. These preclinical results raise the possibility that using attenuated S. typhimurium to deliver FasL to tumors may be an effective and well-tolerated therapeutic strategy for some cancers.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/terapia , Proteína Ligante Fas/farmacologia , Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/terapia , Salmonella typhimurium , Animais , Proteína Ligante Fas/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica , Inflamação/induzido quimicamente , Injeções Intravenosas , Camundongos , Salmonella typhimurium/metabolismo , Transplante Isogênico
11.
J Immunol ; 180(7): 5045-56, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18354230

RESUMO

Salmonella enterica translocates virulent factors into host cells using type III secretion systems to promote host colonization, intracellular bacterial replication and survival, and disease pathogenesis. Among many effectors, the type III secretion system encoded in Salmonella pathogenicity island 2 translocates a Salmonella-specific protein, designated Salmonella secreted factor L (SseL), a putative virulence factor possessing deubiquitinase activity. In this study, we attempt to elucidate the mechanism and the function of SseL in vitro, in primary host macrophages and in vivo in infected mice. Expression of SseL in mammalian cells suppresses NF-kappaB activation downstream of IkappaBalpha kinases and impairs IkappaBalpha ubiquitination and degradation, but not IkappaBalpha phosphorylation. Disruption of the gene encoding SseL in S. enterica serovar typhimurium increases IkappaBalpha degradation and ubiquitination, as well as NF-kappaB activation in infected macrophages, compared with wild-type bacteria. Mice infected with SseL-deficient bacteria mount stronger inflammatory responses, associated with increased production of NF-kappaB-dependent cytokines. Thus, SseL represents one of the first bacterial deubiquitinases demonstrated to modulate the host inflammatory response in vivo.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Quinase I-kappa B/metabolismo , Imunidade Inata/imunologia , NF-kappa B/metabolismo , Salmonella typhimurium/enzimologia , Salmonella typhimurium/imunologia , Animais , Proteínas de Bactérias/genética , Células Cultivadas , Feminino , Deleção de Genes , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Infecções por Salmonella/genética , Infecções por Salmonella/imunologia , Infecções por Salmonella/metabolismo , Infecções por Salmonella/patologia , Ubiquitinação
12.
Proc Natl Acad Sci U S A ; 104(31): 12879-83, 2007 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-17652173

RESUMO

Intravenously administered bacteria reportedly accumulate in tumors. Furthermore, systemic administration of attenuated Salmonella typhimurium has little or no significant side-effects in humans. Consequently, we engineered such bacteria to improve their oncolytic activity by stably inserting a gene encoding LIGHT, a cytokine known to promote tumor rejection. Unlike control bacteria, attenuated S. typhimurium expressing LIGHT inhibited growth of primary tumors, as well as the dissemination of pulmonary metastases, in various mouse tumor models employing murine carcinoma cell lines in immunocompetent mice. Antitumor activity was achieved without significant toxicity and was associated with infiltration of inflammatory cells and dependent on the LIGHT receptors, herpes virus entry mediator (HVEM), and lymphotoxin-beta receptor (LTbetaR). These findings provide evidence that nonvirulent bacteria can be exploited as targeting vehicles for local generation of therapeutic proteins in tumors.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Salmonella typhimurium/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Terapia Genética , Humanos , Camundongos , Neoplasias/genética , Neoplasias/terapia , Engenharia de Proteínas , Salmonella typhimurium/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
13.
Infect Immun ; 72(10): 5733-40, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15385472

RESUMO

Cytotoxic necrotizing factor type 1 (CNF1) from Escherichia coli activates the small GTP-binding proteins of the Rho family (Rho, Rac, and Cdc42) by catalyzing their deamidation at a specific glutamine residue. Since RhoA, Rac, and Cdc42 play a pivotal role in cell migration during the early phase of wound repair, we investigated whether CNF1 was able to interfere with wound healing in intestinal epithelial monolayers (T84 cells). After mechanical injury, we found that CNF1 blocks epithelial wound repair within 48 h. This effect was characterized by cell elongation and filopodium formation on the leading edge, in association with permanent phosphorylation of the focal adhesion kinase (FAK) via Rho activation. Moreover, inhibition of Rho kinase with Y-27632 decreased CNF1-mediated permanent FAK phosphorylation, leading to complete restitution of wound repair within 24 h. In addition, we found that CNF1 induced upregulation of mitogen-activated protein kinases (MAPK) activation. Moreover, activation of Rac and MAPK by CNF1 increased matrix metalloproteinase 9 expression in wounded T84 monolayers. Taken together, these results provide evidence that CNF1 strongly impairs intestinal epithelial wound healing.


Assuntos
Toxinas Bacterianas/farmacologia , Citotoxinas/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/lesões , Proteínas de Escherichia coli , Escherichia coli , Intestinos/efeitos dos fármacos , Intestinos/lesões , Cicatrização/efeitos dos fármacos , Amidas/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Epitélio/fisiologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Intestinos/microbiologia , Intestinos/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Piridinas/farmacologia , Cicatrização/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA