Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 9: 3082, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30692992

RESUMO

The detection of microbial pathogens relies on the recognition of highly conserved microbial structures by the membrane sensor Toll-like receptors (TLRs) and cytosolic sensor NOD-like receptors (NLRs). Upon detection, these sensors trigger innate immune responses to eradicate the invaded microbial pathogens. However, it is unclear whether TLR and NOD signaling are both critical for innate immunity to initiate inflammatory and antimicrobial responses against microbial infection. Here we report that activation of both TLR and NOD signaling resulted in an augmented inflammatory response and the crosstalk between TLR and NOD led to an amplified downstream NF-κB activation with increased nuclear transactivation of p65 at both TNF-α and IL-6 promoters. Furthermore, co-stimulation of macrophages with TLR and NOD agonists maximized antimicrobial activity with accelerated phagosome maturation. Importantly, administration of both TLR and NOD agonists protected mice against polymicrobial sepsis-associated lethality with increased serum levels of inflammatory cytokines and accelerated clearance of bacteria from the circulation and visceral organs. These results demonstrate that activation of both TLR and NOD signaling synergizes to induce efficient inflammatory and antimicrobial responses, thus conferring protection against microbial infection.


Assuntos
Infecções Bacterianas/imunologia , Imunidade Inata , Macrófagos/imunologia , Proteínas NLR/metabolismo , Receptores Toll-Like/metabolismo , Animais , Bactérias/imunologia , Infecções Bacterianas/microbiologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Citosol/imunologia , Citosol/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas NLR/genética , Proteínas NLR/imunologia , Cultura Primária de Células , Receptor Cross-Talk/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
2.
PLoS One ; 12(1): e0171146, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28141831

RESUMO

The costimulatory protein B7-H3 has been shown to play a contributory role in the development and progression of experimental pneumococcal meningitis by augmentation of the innate immunity-associated inflammatory response via a TLR2-dependent manner. This study aimed to clarify the component(s) of TLR2-mediated signal transduction pathways responsible for B7-H3-augmented inflammatory response and subsequent brain damage during experimental pneumococcal meningitis. Administration of B7-H3 did not augment expression of TLR2 and other TLR2 upstream components, but led to an enhanced formation of MyD88-IRAK immunocomplex in the brain of S. pneumoniae-infected mice. Furthermore, B7-H3 substantially augmented S. pneumoniae-induced activation of TLR2 downstream NF-κB p65 and MAPK p38 pathways in the brain of S. pneumoniae-infected mice. Notably, blockage of NF-κB p65 and/or MAPK p38 with their specific inhibitors strongly attenuated B7-H3-amplified inflammatory response with significantly reduced proinflammatory cytokine and chemokine production, and markedly ameliorated B7-H3-exacerbated disruption of blood-brain barrier and severity of disease status in S. pneumoniae-infected mice. These results indicate that targeting NF-κB p65 and/or MAPK p38 may represent a promising therapeutic option for amelioration of overwhelming inflammatory response-associated brain injury frequently observed during pneumococcal meningitis.


Assuntos
Antígenos B7/metabolismo , Lesões Encefálicas/patologia , Progressão da Doença , Inflamação/patologia , Meningite Pneumocócica/metabolismo , Meningite Pneumocócica/microbiologia , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/microbiologia , Encéfalo/patologia , Ativação Enzimática , Inflamação/complicações , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Meningite Pneumocócica/patologia , Camundongos Endogâmicos BALB C , Fator 88 de Diferenciação Mieloide/metabolismo , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Infecções Estreptocócicas/enzimologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus pneumoniae/fisiologia , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 2 Toll-Like/metabolismo , Regulação para Cima
3.
Sci Rep ; 7: 40418, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28079153

RESUMO

Tolerance to bacterial components represents an essential regulatory mechanism during bacterial infection. Bacterial lipoprotein (BLP)-induced tolerance confers protection against microbial sepsis by attenuating inflammatory responses and augmenting antimicrobial activity in innate phagocytes. It has been well-documented that BLP tolerance-attenuated proinflammatory cytokine production is associated with suppressed TLR2 signalling pathway; however, the underlying mechanism(s) involved in BLP tolerance-enhanced antimicrobial activity is unclear. Here we report that BLP-tolerised macrophages exhibited accelerated phagosome maturation and enhanced bactericidal activity upon bacterial infection, with upregulated expression of membrane-trafficking regulators and lysosomal enzymes. Notably, bacterial challenge resulted in a strong activation of NF-κB pathway in BLP-tolerised macrophages. Importantly, activation of NF-κB pathway is critical for BLP tolerance-enhanced antimicrobial activity, as deactivation of NF-κB in BLP-tolerised macrophages impaired phagosome maturation and intracellular killing of the ingested bacteria. Finally, activation of NF-κB pathway in BLP-tolerised macrophages was dependent on NOD1 and NOD2 signalling, as knocking-down NOD1 and NOD2 substantially inhibited bacteria-induced activation of NF-κB and overexpression of Rab10 and Acp5, two membrane-trafficking regulators and lysosomal enzymes contributed to BLP tolerance-enhanced bactericidal activity. These results indicate that activation of NF-κB pathway is essential for BLP tolerance-augmented antimicrobial activity in innate phagocytes and depends primarily on both NOD1 and NOD2.


Assuntos
Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Tolerância Imunológica , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , NF-kappa B/metabolismo , Animais , Infecções Bacterianas/patologia , Membrana Celular/metabolismo , Inativação Gênica , Lisossomos/metabolismo , Camundongos Endogâmicos C57BL , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Fagossomos/metabolismo , Salmonella typhimurium/imunologia , Staphylococcus aureus/imunologia , Regulação para Cima , Proteínas rab de Ligação ao GTP/metabolismo
4.
Sci Rep ; 5: 13694, 2015 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-26329314

RESUMO

Myeloid-related protein 8 (Mrp8) is the active component of Mrp8/14 protein complex released by phagocytes at the site of infection and stimulates inflammatory responses. However, it is unclear whether Mrp8 could induce self-tolerance and cross-tolerance to bacterial infection. Here we report that Mrp8 triggered TNF-α and IL-6 release via a Toll-like receptor 4 (TLR4)-dependent manner. Pre-stimulation of murine macrophages and human monocytes with Mrp8 induced self-tolerance to Mrp8 re-stimulation and cross-tolerance to lipopolysaccharide (LPS), bacterial lipoprotein (BLP), gram-negative and gram-positive bacterial challenges, with substantially attenuated TNF-α and IL-6 release. Moreover, Mrp8 tolerisation significantly reduced serum TNF-α and IL-6, increased polymorphonuclear neutrophil (PMN) recruitment and accelerated bacterial clearance, thus protecting mice against LPS-induced lethality and cecal ligation and puncture (CLP)-induced polymicrobial sepsis. In addition to TLR4, TLR2 also contributed to Mrp8-induced inflammatory response and tolerance. Down-regulation of phosphorylated p38 by Mrp8 pre-stimulation was predominantly responsible for the intracellular mechanism of Mrp8-induced tolerance. Thus, our findings of Mrp8-induced self-tolerance and cross-tolerance may provide a potential strategy for attenuating an overwhelming proinflammatory cascade and enhancing antimicrobial responses during microbial sepsis.


Assuntos
Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Calgranulina A/metabolismo , Tolerância Imunológica , Proteínas/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Ceco/patologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Inflamação/patologia , Ligadura , Lipopolissacarídeos/farmacologia , Lipoproteínas/metabolismo , Camundongos , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Punções , Sepse/metabolismo , Sepse/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Eur J Immunol ; 43(5): 1322-32, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23404483

RESUMO

Neonates and infants, due to the immaturity in their adaptive immunity, are thought to depend largely on the innate immune system for protection against bacterial infection. However, the innate immunity-mediated antimicrobial response in neonates and infants is incompletely characterized. Here, we report that infant mice were more susceptible to microbial sepsis than adult mice, with significantly reduced bacterial clearance from the circulation and visceral organs. Infant PMNs exhibited less constitutive expression of the chemokine receptor CXCR2, and bacterial infection caused further reduction of PMN CXCR2 in infant mice compared with adult mice. This correlates with diminished in vitro chemotaxis of infant PMNs toward the chemoattractant CXCL2 and impaired in vivo recruitment of infant PMNs into the infectious site. Furthermore, consistent with the reduced antimicrobial response in vivo, infant macrophages displayed an impaired bactericidal activity with a defect in phagosome maturation after ingestion of either gram-positive or gram-negative bacteria. Thus, infant mice exhibit an increased vulnerability to microbial infection with delayed bacterial clearance, which is associated with the inefficiency in their innate phagocyte-associated antimicrobial functions characterized by defects in PMN recruitment and macrophage phagosome maturation during microbial sepsis.


Assuntos
Imunidade Inata , Macrófagos/imunologia , Neutrófilos/imunologia , Sepse/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas , Quimiocina CXCL2/farmacologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Suscetibilidade a Doenças , Expressão Gênica , Injeções Intraperitoneais , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Fagossomos/efeitos dos fármacos , Fagossomos/imunologia , Fagossomos/patologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/imunologia , Sepse/microbiologia , Sepse/patologia , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/imunologia
6.
J Immunol ; 189(1): 347-55, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22661093

RESUMO

In addition to a well-documented role in regulating T cell-mediated immune responses, B7-H3, a newly discovered member of the B7 superfamily, has been recently identified as a costimulator in the innate immunity-mediated inflammatory response. In this study, we further report that B7-H3 participates in the development of pneumococcal meningitis in a murine model. Exogenous administration of B7-H3 strongly amplified the inflammatory response, exacerbated blood-brain barrier disruption, and aggravated the clinical disease status in Streptococcus pneumoniae-infected C3H/HeN wild-type mice. Consistent with the in vivo findings, B7-H3 substantially augmented proinflammatory cytokine and chemokine production, upregulated NF-κB p65 and MAPK p38 phosphorylation, and enhanced the nuclear transactivation of NF-κB p65 at both TNF-α and IL-6 promoters in S. pneumoniae-stimulated primary murine microglia cells. These B7-H3-associated in vitro and in vivo effects appeared to be dependent on TLR2 signaling, as B7-H3 almost completely lost its amplifying actions in both TLR2-deficient microglial cells and TLR2-deficient mice. Furthermore, administration of the anti-B7-H3 mAb (MIH35) attenuated the inflammatory response and ameliorated blood-brain barrier disruption in S. pneumoniae-infected wild-type mice. Collectively, our results indicate that B7-H3 plays a contributory role in the development of S. pneumoniae infection-induced bacterial meningitis.


Assuntos
Antígenos B7/fisiologia , Mediadores da Inflamação/fisiologia , Meningite Pneumocócica/imunologia , Meningite Pneumocócica/patologia , Receptor 2 Toll-Like/fisiologia , Animais , Antígenos B7/administração & dosagem , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/patologia , Células Cultivadas , Mediadores da Inflamação/administração & dosagem , Meningite Pneumocócica/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Microglia/metabolismo , Microglia/microbiologia , Microglia/patologia , Distribuição Aleatória , Transdução de Sinais/imunologia , Streptococcus pneumoniae/imunologia , Receptor 2 Toll-Like/deficiência
7.
J Immunol ; 187(8): 4293-9, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21911606

RESUMO

ST2, a member of the Toll/IL-1R superfamily, negatively regulates both TLR2 and TLR4 signaling. In this study, we report that ST2-deficient mice were more susceptible to polymicrobial sepsis than their wild-type littermates, with increased production of proinflammatory cytokines. Bacterial clearance from the circulation and visceral organs following polymicrobial infection was markedly impaired in ST2-deficient mice. This was associated with substantially reduced uptake, phagocytosis, and intracellular killing of both Gram-positive and Gram-negative bacteria by ST2-deficient phagocytes. Consistent with a reduced antimicrobial response, phagocytes lacking ST2 displayed a defect in bactericidal activity in response to bacterial challenges with severely impaired phagosome maturation and NOX2 function. Thus, ST2-deficient mice exhibit an increased susceptibility to polymicrobial infection with impaired bacterial clearance, which is associated with defects in phagosome maturation and NOX2-derived production of reactive oxygen species characterized in ST2-deficient phagocytes.


Assuntos
Glicoproteínas de Membrana/imunologia , NADPH Oxidases/imunologia , Fagossomos/imunologia , Espécies Reativas de Oxigênio/imunologia , Receptores de Interleucina/imunologia , Sepse/imunologia , Animais , Infecções Bacterianas/complicações , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Separação Celular , Citometria de Fluxo , Imunofluorescência , Proteína 1 Semelhante a Receptor de Interleucina-1 , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Fagossomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Interleucina/metabolismo , Sepse/metabolismo , Sepse/patologia
8.
Surg Infect (Larchmt) ; 9(2): 183-94, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18426351

RESUMO

BACKGROUND: Death attributable to septic shock syndrome depends highly on the inflammatory response and cytokine production. Inhibition of inflammation is one of the many pleiotropic effects of statins. The aim of this study was to test the hypothesis that statins have a role in altering the host response to bacterial infections and thus would prove beneficial in the prevention of microbial sepsis. METHODS: Male C57BL/6 and C3H/HeN mice received cerivastatin (4 mg/kg) or phosphate-buffered saline (PBS) intraperitoneally (i.p.), 24 and 1 h before and 24, 48, and 72 h after bacterial challenges. Sepsis was induced by i.p. injection of lipopolysaccharide (LPS) (45 mg/kg), Staphylococcus aureus (5 x 10(7) colony-forming units [CFU]/mouse), or Salmonella typhimurium (2.5 x 10(6) CFU/mouse). RESULTS: Administration of cerivastatin improved significantly the survival rate of mice challenged with LPS (31% vs. 19% in the PBS group; p = 0.001), S. aureus (56% vs. 20% in PBS group; p = 0.01), or S. typhimurium (48% vs. 10% in PBS group; p = 0.03). Significantly reduced release of the pro-inflammatory cytokines tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 was evident in cerivastatin-treated mice after LPS challenge. Cerivastatin-treated mice showed insignificant reductions in serum TNF-alpha and IL-6 concentrations after bacterial challenge. However, significantly accelerated bacterial clearance was demonstrated in cerivastatin-treated mice 24 h after S. typhimurium infection and 48 h after S. aureus infection. CONCLUSIONS: Cerivastatin protects mice against LPS- and live bacteria-induced death, an effect associated with cerivastatin-attenuated pro-inflammatory cytokine production and enhanced bacterial clearance. Hence, application of statins in the clinical setting may prove beneficial in prevention of LPS or bacterial infection-related sepsis.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Piridinas/farmacologia , Choque Séptico/imunologia , Choque Séptico/prevenção & controle , Animais , Modelos Animais de Doenças , Escherichia coli/imunologia , Interleucina-6/imunologia , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Salmonella typhi/imunologia , Staphylococcus aureus/imunologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos
9.
J Immunol ; 170(2): 795-804, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12517943

RESUMO

Beta(1) integrins play a crucial role in supporting tumor cell attachment to and invasion into the extracellular matrix. Endotoxin/LPS introduced by surgery has been shown to enhance tumor metastasis in a murine model. Here we show the direct effect of LPS on tumor cell adhesion and invasion in extracellular matrix proteins through a beta(1) integrin-dependent pathway. The human colorectal tumor cell lines SW480 and SW620 constitutively expressed high levels of the beta(1) subunit, whereas various low levels of alpha(1), alpha(2), alpha(4), and alpha(6) expression were detected. SW480 and SW620 did not express membrane-bound CD14; however, LPS in the presence of soluble CD14 (sCD14) significantly up-regulated beta(1) integrin expression; enhanced tumor cell attachment to fibronectin, collagen I, and laminin; and strongly promoted tumor cell invasion through the Matrigel. Anti-beta(1) blocking mAbs (4B4 and 6S6) abrogated LPS- plus sCD14-induced tumor cell adhesion and invasion. Furthermore, LPS, when combined with sCD14, resulted in NF-kappaB activation in both SW480 and SW620 cells. Inhibition of the NF-kappaB pathway significantly attenuated LPS-induced up-regulation of beta(1) integrin expression and prevented tumor cell adhesion and invasion. These results provide direct evidence that although SW480 and SW620 cells do not express membrane-bound CD14, LPS in the presence of sCD14 can activate NF-kappaB, up-regulate beta(1) integrin expression, and subsequently promote tumor cell adhesion and invasion. Moreover, LPS-induced tumor cell attachment to and invasion through extracellular matrix proteins is beta(1) subunit-dependent.


Assuntos
Adjuvantes Imunológicos/farmacologia , Movimento Celular/imunologia , Endotoxinas/farmacologia , Integrina beta1/fisiologia , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Células Tumorais Cultivadas/patologia , Adesão Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Humanos , Proteínas I-kappa B/genética , Integrina alfaVbeta3/biossíntese , Integrina beta1/biossíntese , Receptores de Lipopolissacarídeos/biossíntese , Receptores de Lipopolissacarídeos/metabolismo , Receptores de Lipopolissacarídeos/farmacologia , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Peptídeos/farmacologia , Transdução de Sinais/imunologia , Solubilidade , Transfecção , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/metabolismo , Regulação para Cima/imunologia
10.
J Immunol ; 170(1): 14-8, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12496376

RESUMO

Tolerance to bacterial cell wall components is an adaptive host response. Endotoxin/LPS tolerance is characterized by a survival advantage against subsequent lethal LPS challenge. However, it is uncertain whether LPS tolerance can afford protection against other septic challenges. In this study, we show that tolerance induced by bacterial lipoprotein (BLP) protects mice against not only BLP-induced lethality, but also LPS-, live bacteria-, and polymicrobial sepsis-induced lethality. In contrast, LPS tolerance offers no survival benefit against the latter two challenges. Furthermore, induction of BLP tolerance results in overexpression of complement receptor type 3 and FcgammaIII/IIR on neutrophils (polymorphonuclear neutrophils) and peritoneal macrophages, with increased bacterial recognition and bactericidal activity, whereas LPS-tolerized mice exhibit an impaired ability to ingest and to kill bacteria. These results indicate that BLP tolerance is a novel adaptive host response associated with a unique protective effect during septic shock.


Assuntos
Proteínas da Membrana Bacteriana Externa/administração & dosagem , Tolerância Imunológica/imunologia , Lipopolissacarídeos/administração & dosagem , Lipoproteínas/administração & dosagem , Choque Séptico/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Atividade Bactericida do Sangue/imunologia , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Inflamação/imunologia , Inflamação/prevenção & controle , Injeções Intraperitoneais , Lipopolissacarídeos/imunologia , Lipoproteínas/imunologia , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Choque Séptico/mortalidade , Choque Séptico/prevenção & controle , Análise de Sobrevida
11.
J Biol Chem ; 277(39): 36068-75, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12133836

RESUMO

Tolerance to bacterial cell wall components including lipopolysaccharide (LPS) may represent an essential regulatory mechanism during bacterial infection. Two members of the Toll-like receptor (TLR) family, TLR2 and TLR4, recognize the specific pattern of bacterial cell wall components. TLR4 has been found to be responsible for LPS tolerance. However, the role of TLR2 in bacterial lipoprotein (BLP) tolerance and LPS tolerance is unclear. Pretreatment of human THP-1 monocytic cells with a synthetic bacterial lipopeptide induced tolerance to a second BLP challenge with diminished tumor necrosis factor-alpha and interleukin-6 production, termed BLP tolerance. Furthermore, BLP-tolerized THP-1 cells no longer responded to LPS stimulation, indicating a cross-tolerance to LPS. Induction of BLP tolerance was CD14-independent, as THP-1 cells that lack membrane-bound CD14 developed tolerance both in serum-free conditions and in the presence of a specific CD14 blocking monoclonal antibody (MEM-18). Pre-exposure of THP-1 cells to BLP suppressed mitogen-activated protein kinase phosphorylation and nuclear factor-kappaB activation in response to subsequent BLP and LPS stimulation, which is comparable with that found in LPS-tolerized cells, indicating that BLP tolerance and LPS tolerance may share similar intracellular pathways. However, BLP strongly enhanced TLR2 expression in non-tolerized THP-1 cells, whereas LPS stimulation had no effect. Furthermore, a specific TLR2 blocking monoclonal antibody (2392) attenuated BLP-induced, but not LPS-induced, tumor necrosis factor-alpha and interleukin-6 production, indicating BLP rather than LPS as a ligand for TLR2 engagement and activation. More importantly, pretreatment of THP-1 cells with BLP strongly inhibited TLR2 activation in response to subsequent BLP stimulation. In contrast, LPS tolerance did not prevent BLP-induced TLR2 overexpression. These results demonstrate that BLP tolerance develops through down-regulation of TLR2 expression.


Assuntos
Proteínas da Membrana Bacteriana Externa/farmacologia , Proteínas de Drosophila , Tolerância Imunológica , Lipopolissacarídeos/farmacologia , Lipoproteínas/metabolismo , Glicoproteínas de Membrana/biossíntese , Receptores de Superfície Celular/biossíntese , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Separação Celular , Meios de Cultura Livres de Soro/farmacologia , Citocinas/metabolismo , Regulação para Baixo , Citometria de Fluxo , Humanos , Interleucina-6/metabolismo , Receptores de Lipopolissacarídeos/biossíntese , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/metabolismo , Luciferases/metabolismo , Sistema de Sinalização das MAP Quinases , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/metabolismo , Fosforilação , Receptores de Superfície Celular/metabolismo , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...