Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Parasite Immunol ; 33(4): 217-25, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21204848

RESUMO

Enterocytozoon bieneusi is clinically the most significant microsporidian parasite associated with persistent diarrhoea, wasting and cholangitis in 30-50% of individuals with HIV/AIDS, as well as in malnutritional children and in the recipients of immunosuppressive therapy. However, the host immune responses to E. bieneusi have not been investigated until recently because of lack of sources of spores, cell culture system and animal models. In this study, we purified spores from heavily infected human or monkey faeces by serial salt-Percoll-sucrose-iodixanol centrifugation, and the purity of spores was confirmed by FACS and scanning electron microscopy. Exposure of dendritic cells to E. bieneusi spores induced the upregulation of the surface markers and production of pro-inflammatory cytokines. The cytokine production was independent of toll-like receptor 4, but MyD88 dependent, because dendritic cells from MyD88 knockout mice failed to secrete these pro-inflammatory cytokines, whereas dendritic cells from C3H/HeJ (a toll-like receptor 4 mutant) were activated by E. bieneusi and secreted these cytokines. Furthermore, MyD88-deficient mice were susceptible to E. bieneusi infection, in contrast to wild-type mice that resisted the infection. Collectively, the data demonstrate innate recognition of E. bieneusi by dendritic cells and the importance of MyD88-dependent signalling in resisting infection in a murine challenge model.


Assuntos
Citocinas/biossíntese , Enterocytozoon/imunologia , Imunidade Inata , Microsporidiose/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Enterocytozoon/isolamento & purificação , Fezes/microbiologia , Haplorrinos , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética
2.
Infect Immun ; 74(1): 549-56, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16369011

RESUMO

Cryptosporidium spp. cause diarrheal disease worldwide. Innate immune responses mediating resistance to this parasite are not completely understood. To determine whether MyD88-dependent pathways play a role in resistance to Cryptosporidium parvum, we compared the course of infection in MyD88(-/-) mice to that in their wild-type (WT) littermate controls. Three- to 4-week-old mice were infected with C. parvum, and infection was monitored by quantifying fecal oocyst shedding. Twelve days postinfection, the histology of the intestines was examined to quantify intestinal parasite burden and to determine if there were any pathological changes. Fecal oocyst shedding and intestinal parasite burden were significantly greater in MyD88(-/-) mice than in littermate controls. Nonetheless, both WT and MyD88(-/-) mice cleared the infection within 3 weeks. These results indicate that MyD88-dependent pathways are involved in mediating initial resistance to C. parvum. Since gamma interferon (IFN-gamma) is known to mediate resistance to C. parvum, we also studied infection in MyD88(-/-) mice and WT controls in which this cytokine was temporarily neutralized. Fecal oocyst shedding, as well as intestinal parasite burden, intestinal inflammation, and mortality, was significantly greater in MyD88(-/-) mice in which IFN-gamma was neutralized than in IFN-gamma-neutralized WT mice or in MyD88(-/-) mice in which this cytokine was active. These results suggest that MyD88 and IFN-gamma had an additive effect in conferring protection from C. parvum infection. While this study confirms the importance of IFN-gamma in conferring resistance to infection with C. parvum, it suggests that MyD88-mediated pathways also play a role in innate immunity to this parasite.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Antígenos de Diferenciação/fisiologia , Criptosporidiose/imunologia , Cryptosporidium parvum/imunologia , Receptores Imunológicos/fisiologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos de Diferenciação/genética , Criptosporidiose/metabolismo , Criptosporidiose/mortalidade , Enterocolite/imunologia , Enterocolite/metabolismo , Enterocolite/mortalidade , Enterocolite/parasitologia , Feminino , Imunidade Inata/genética , Interferon gama/antagonistas & inibidores , Interferon gama/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Transdução de Sinais/genética
3.
J Endotoxin Res ; 7(6): 447-50, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11753215

RESUMO

In mice, defense against an intraperitoneal Salmonella infection depends on a vigorous innate immune response. Mutations which lead to an inadequate early response to the pathogen thus identify genes involved in innate immunity. The best studied host resistance factor, NRAMP-1, is an endosomal membrane protein whose loss leads to an inability of the animals to hold the infection in check. However, innate defense against Salmonella is not restricted to mechanisms which directly attack the pathogen within macrophages. Here we have examined the contribution of the LBP, CD14 and TLR4 gene products to innate defense against Salmonella. To this end, we have generated mice which carry a wild-type allele of NRAMP-1, but which are deficient for the LBP, CD14 or TLR4 genes. Loss of any of these genes leads to a susceptibility to Salmonella as dramatic as that seen in animals lacking functional NRAMP-1 protein. This indicates that LBP, CD14 and TLR4 are all critical elements required in the proper induction of this innate defense system.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Drosophila , Receptores de Lipopolissacarídeos/fisiologia , Glicoproteínas de Membrana/fisiologia , Peritônio/microbiologia , Receptores de Superfície Celular/fisiologia , Salmonelose Animal/imunologia , Proteínas de Fase Aguda/fisiologia , Alelos , Animais , Pareamento Incorreto de Bases , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Peritônio/patologia , Peritonite/genética , Peritonite/imunologia , Peritonite/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Salmonelose Animal/genética , Salmonelose Animal/patologia , Salmonella typhimurium/patogenicidade , Receptor 4 Toll-Like , Receptores Toll-Like
4.
J Immunol ; 167(12): 7069-76, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11739528

RESUMO

Group B streptococcus (GBS) imposes a major health threat to newborn infants. Little is known about the molecular basis of GBS-induced sepsis. Both heat-inactivated whole GBS bacteria and a heat-labile soluble factor released by GBS during growth (GBS-F) induce nuclear translocation of NF-kappaB, the secretion of TNF-alpha, and the formation of NO in mouse macrophages. Macrophages from mice with a targeted disruption of MyD88 failed to secrete TNF-alpha in response to both heat-inactivated whole bacteria and GBS-F, suggesting that Toll-like receptors (TLRs) are involved in different aspects of GBS recognition. Immune cell activation by whole bacteria differed profoundly from that by secreted GBS-F. Whole GBS activated macrophages independently of TLR2 and TLR6, whereas a response to the secreted GBS-F was not observed in macrophages from TLR2-deficient animals. In addition to TLR2, TLR6 and CD14 expression were essential for GBS-F responses, whereas TLR1 and TLR4 or MD-2 did not appear to be involved. Heat lability distinguished GBS-F from peptidoglycan and lipoproteins. GBS mutants deficient in capsular polysaccharide or beta-hemolysin had GBS-F activity comparable to that of wild-type streptococci. We suggest that CD14 and TLR2 and TLR6 function as coreceptors for secreted microbial products derived from GBS and that cell wall components of GBS are recognized by TLRs distinct from TLR1, 2, 4, or 6.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-1 , Streptococcus agalactiae/fisiologia , Animais , Antígenos de Superfície/fisiologia , Fatores Biológicos/metabolismo , Células CHO , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Células Cultivadas , Cricetinae , Humanos , Mediadores da Inflamação/metabolismo , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Sepse/imunologia , Infecções Estreptocócicas/imunologia , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptor 6 Toll-Like , Receptores Toll-Like , Transfecção , Fator de Necrose Tumoral alfa/biossíntese
6.
J Immunol ; 167(4): 2257-67, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11490013

RESUMO

Down-regulation of cell surface expression of Toll-like receptor (TLR) 4 following LPS stimulation has been suggested to underlie endotoxin tolerance. In this study, we examined whether overexpression of TLR2 or TLR4 would affect the ability of cells to become tolerant to LPS or the mycobacterial components, arabinose-capped lipoarabinomannan (LAM) and soluble tuberculosis factor (STF). To this end, Chinese hamster ovary/CD14 cells stably transfected with a NF-kappaB-dependent reporter construct, endothelial leukocyte adhesion molecule CD25 (the 3E10 clone), were engineered to overexpress either human TLR2 or TLR4. Transfected TLRs exhibited proper signaling functions, as evidenced by increased LPS responsiveness of 3E10/TLR4 cells and acquisition of sensitivity to TLR2-specific ligands upon transfection of TLR2 into TLR2-negative 3E10 cells. Pretreatment of cells with LPS, LAM, or STF did not modulate TLR2 or TLR4 cell surface expression. Following LPS exposure, 3E10, 3E10/TLR2, and 3E10/TLR4 cells exhibited comparable decreases in LPS-mediated NF-kappaB activation and mitogen-activated protein (MAP) kinase phosphorylation. Likewise, LPS pretreatment profoundly inhibited LPS-induced NF-kappaB translocation in Chinese hamster ovary cells that concomitantly overexpressed human TLR4 and myeloid differentiation protein-2 (MD-2), but failed to modulate TLR4 or MD-2 cell surface expression. Pretreatment of 3E10/TLR2 cells with LAM or STF decreased their NF-kappaB responses induced by subsequent stimulation with these substances or LPS. Conversely, prior exposure of 3E10/TLR2 cells to LPS led to hyporesponsiveness to LPS, LAM, and STF, indicating that LPS and mycobacterial products induce cross-tolerance. Thus, tolerance to LPS and mycobacterial components cannot be attributed solely to a decrease in TLR/MD-2 expression levels, suggesting inhibition of expression or function of other signaling intermediates.


Assuntos
Antígenos de Bactérias/imunologia , Células CHO/imunologia , Proteínas de Drosophila , Tolerância Imunológica , Receptores de Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/imunologia , Glicoproteínas de Membrana/biossíntese , Mycobacterium tuberculosis/imunologia , Receptores de Superfície Celular/biossíntese , Animais , Antígenos de Superfície/biossíntese , Antígenos de Superfície/genética , Células CHO/metabolismo , Linhagem Celular , Células Clonais , Cricetinae , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Humanos , Tolerância Imunológica/genética , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/antagonistas & inibidores , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Receptores de Superfície Celular/genética , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção
7.
Circ Res ; 89(3): 244-50, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11485974

RESUMO

An early component of atherogenesis is abnormal vascular smooth muscle cell (VSMC) proliferation. The presence of Chlamydia pneumoniae in many atherosclerotic lesions raises the possibility that this organism plays a causal role in atherogenesis. In this study, C pneumoniae elementary bodies (EBs) rapidly activated p44/p42 mitogen-activated protein kinases (MAPKs) and stimulated proliferation of VSMCs in vitro. Exposure of VSMCs derived from human saphenous vein to C pneumoniae EBs (3x10(7) inclusion forming units/mL) enhanced bromodeoxyuridine (BrdU) incorporation 12+/-3-fold. UV- and heat-inactivated C pneumoniae EBs also stimulated VSMC proliferation, indicating a role of direct stimulation by chlamydial antigens. However, the mitogenic activity of C pneumoniae was heat-labile, thus excluding a role of lipopolysaccharide. Chlamydial hsp60 (25 microg/mL) replicated the effect of C pneumoniae, stimulating BrdU incorporation 7+/-3-fold. Exposure to C pneumoniae or chlamydial hsp60 rapidly activated p44/p42 MAPK, within 5 to 10 minutes of exposure. In addition, PD98059 and U0126, which are two distinct inhibitors of upstream MAPK kinase 1/2 (MEK1/2), abolished the mitogenic effect of C pneumoniae and chlamydial hsp60. Toll-like receptors (TLRs) act as sensors for microbial antigens and can signal via the p44/p42 MAPK pathway. Human VSMCs were shown to express TLR4 mRNA and protein, and a TLR4 antagonist abolished chlamydial hsp60-induced VSMC proliferation and attenuated C pneumoniae-induced MAPK activation and VSMC proliferation. Together these results indicate that C pneumoniae and chlamydial hsp60 are potent inducers of human VSMC proliferation and that these effects are mediated, at least in part, by rapid TLR4-mediated activation of p44/p42 MAPK.


Assuntos
Chaperonina 60/metabolismo , Proteínas de Drosophila , Glicoproteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Bromodesoxiuridina , Butadienos/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Chaperonina 60/antagonistas & inibidores , Chaperonina 60/farmacologia , Infecções por Chlamydophila/metabolismo , Chlamydophila pneumoniae/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Temperatura Alta , Humanos , Lipídeo A/análogos & derivados , Lipídeo A/farmacologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteína Quinase 3 Ativada por Mitógeno , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/microbiologia , Nitrilas/farmacologia , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veia Safena , Receptor 4 Toll-Like , Receptores Toll-Like
8.
J Immunol ; 167(2): 910-8, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11441098

RESUMO

Mycobacterium tuberculosis (MTB) induces vigorous immune responses, yet persists inside macrophages, evading host immunity. MTB bacilli or lysate was found to inhibit macrophage expression of class II MHC (MHC-II) molecules and MHC-II Ag processing. This report characterizes and identifies a specific component of MTB that mediates these inhibitory effects. The inhibitor was extracted from MTB lysate with Triton X-114, isolated by gel electroelution, and identified with Abs to be MTB 19-kDa lipoprotein. Electroelution- or immunoaffinity-purified MTB 19-kDa lipoprotein inhibited MHC-II expression and processing of both soluble Ags and Ag 85B from intact MTB bacilli. Inhibition of MHC-II Ag processing by either MTB bacilli or purified MTB 19-kDa lipoprotein was dependent on Toll-like receptor (TLR) 2 and independent of TLR 4. Synthetic analogs of lipopeptides from Treponema pallidum also inhibited Ag processing. Despite the ability of MTB 19-kDa lipoprotein to activate microbicidal and innate immune functions early in infection, TLR 2-dependent inhibition of MHC-II expression and Ag processing by MTB 19-kDa lipoprotein during later phases of macrophage infection may prevent presentation of MTB Ags and decrease recognition by T cells. This mechanism may allow intracellular MTB to evade immune surveillance and maintain chronic infection.


Assuntos
Aciltransferases , Apresentação de Antígeno/imunologia , Antígenos de Bactérias , Proteínas de Bactérias/farmacologia , Proteínas de Drosophila , Antígenos de Histocompatibilidade Classe II/biossíntese , Imunossupressores/farmacologia , Lipoproteínas/farmacologia , Macrófagos/imunologia , Glicoproteínas de Membrana/fisiologia , Mycobacterium tuberculosis/imunologia , Receptores de Superfície Celular/fisiologia , Animais , Apresentação de Antígeno/genética , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Detergentes , Epitopos/metabolismo , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Octoxinol , Polietilenoglicóis , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like
9.
J Exp Med ; 194(1): 79-88, 2001 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-11435474

RESUMO

Somatic cell mutagenesis is a powerful tool for characterizing receptor systems. We reported previously two complementation groups of mutant cell lines derived from CD14-transfected Chinese hamster ovary--K1 fibroblasts defective in responses to bacterial endotoxin. Both classes of mutants expressed a normal gene product for Toll-like receptor (TLR)4, and fully responded to stimulation by tumor necrosis factor (TNF)-alpha or interleukin (IL)-1 beta. We identified the lesion in one of the complementation groups in the gene for MD-2, a putative TLR4 coreceptor. The nonresponder phenotype of this mutant was reversed by transfection with MD-2. Cloning of MD-2 from the nonresponder cell line revealed a point mutation in a highly conserved region resulting in a C95Y amino acid exchange. Both forms of MD-2 colocalized with TLR4 on the cell surface after transfection, but only the wild-type cDNA reverted the lipopolysaccharide (LPS) nonresponder phenotype. Furthermore, soluble MD-2, but not soluble MD-2(C95Y), functioned to enable LPS responses in cells that expressed TLR4. Thus, MD-2 is a required component of the LPS signaling complex and can function as a soluble receptor for cells that do not otherwise express it. We hypothesize that MD-2 conformationally affects the extracellular domain of TLR4, perhaps resulting in a change in affinity for LPS or functioning as a portion of the true ligand for TLR4.


Assuntos
Antígenos de Superfície/genética , Proteínas de Drosophila , Endotoxinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Antígenos de Superfície/metabolismo , Células CHO/efeitos dos fármacos , Linhagem Celular , Células Clonais , Clonagem Molecular , Cricetinae , Análise Mutacional de DNA , Teste de Complementação Genética , Humanos , Interleucina-1/farmacologia , Interleucina-6/metabolismo , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/toxicidade , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/genética , Mutação , Receptores de Superfície Celular/genética , Transdução de Sinais , Receptor 4 Toll-Like , Receptores Toll-Like , Fator de Necrose Tumoral alfa/farmacologia
10.
J Immunol ; 167(1): 416-23, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11418678

RESUMO

Glycosylphosphatidylinositol (GPI) anchors and glycoinositolphospholipids (GIPLs) from parasitic protozoa have been shown to exert a wide variety of effects on cells of the host innate immune system. However, the receptor(s) that are triggered by these protozoan glycolipids has not been identified. Here we present evidence that Trypanosoma cruzi-derived GPI anchors and GIPLs trigger CD25 expression on Chinese hamster ovary-K1 cells transfected with CD14 and Toll-like receptor-2 (TLR-2), but not wild-type (TLR-2-deficient) Chinese hamster ovary cells. The protozoan-derived GPI anchors and GIPLs containing alkylacylglycerol and saturated fatty acid chains or ceramide were found to be active in a concentration range of 100 nM to 1 microM. More importantly, the GPI anchors purified from T. cruzi trypomastigotes, which contain a longer glycan core and unsaturated fatty acids in the sn-2 position of the alkylacylglycerolipid component, triggered TLR-2 at subnanomolar concentrations. We performed experiments with macrophages from TLR-2 knockout and TLR-4 knockout mice, and found that TLR-2 expression appears to be essential for induction of IL-12, TNF-alpha, and NO by GPI anchors derived from T. cruzi trypomastigotes. Thus, highly purified GPI anchors from T. cruzi parasites are potent activators of TLR-2 from both mouse and human origin. The activation of TLR-2 may initiate host innate defense mechanisms and inflammatory response during protozoan infection, and may provide new strategies for immune intervention during protozoan infections.


Assuntos
Proteínas de Drosophila , Glicosilfosfatidilinositóis/fisiologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Trypanosoma cruzi/imunologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Relação Dose-Resposta Imunológica , Glicolipídeos/fisiologia , Glicosilfosfatidilinositóis/isolamento & purificação , Inflamação/imunologia , Inflamação/parasitologia , Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , NF-kappa B/fisiologia , Fosfolipídeos/fisiologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores de Interleucina-2/biossíntese , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção , Trypanosoma cruzi/química , Trypanosoma cruzi/crescimento & desenvolvimento
11.
Infect Immun ; 69(4): 2230-6, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11254578

RESUMO

The recent isolation of a lipopolysaccharide (LPS)-deficient mutant of Neisseria meningitidis has allowed us to explore the roles of other gram-negative cell wall components in the host response to infection. The experiments in this study were designed to examine the ability of this mutant strain to activate cells. Although it was clearly less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in monocytic cells, inducing a response similar to that seen with Staphylococcus aureus. Cellular activation by the LPS mutant was related to expression of CD14, a high-affinity receptor for LPS and other microbial products, as well as Toll-like receptor 2, a member of the Toll family of receptors recently implicated in host responses to gram-positive bacteria. In contrast to the parental strain, the synthetic LPS antagonist E5564 did not inhibit the LPS-deficient mutant. We conclude that even in the absence of LPS, the gram-negative cell wall remains a potent inflammatory stimulant, utilizing signaling pathways independent of those involved in LPS signaling.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Drosophila , Lipídeo A/análogos & derivados , Lipopolissacarídeos/toxicidade , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Neisseria meningitidis/imunologia , Receptores de Superfície Celular/fisiologia , Animais , Células CHO , Parede Celular/fisiologia , Cricetinae , Humanos , Lipídeo A/farmacologia , Receptores de Lipopolissacarídeos/fisiologia , Mutação , Receptor 2 Toll-Like , Receptores Toll-Like , Fator de Necrose Tumoral alfa/metabolismo
12.
J Immunol ; 166(7): 4620-6, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11254720

RESUMO

Toll-like receptors (TLR) 2 and 4 are cell surface receptors that in association with CD14 enable phagocytic inflammatory responses to a variety of microbial products. Activation via these receptors triggers signaling cascades, resulting in nuclear translocation of NF-kappa B and a proinflammatory response including TNF-alpha production. We investigated whether TLRs participate in the host response to Cryptococcus neoformans glucuronoxylomannan (GXM), the major capsular polysaccharide of this fungus. Chinese hamster ovary fibroblasts transfected with human TLR2, TLR4, and/or CD14 bound fluorescently labeled GXM. The transfected Chinese hamster ovary cells were challenged with GXM, and activation of an NF-kappa B-dependent reporter construct was evaluated. Activation was observed in cells transfected with both CD14 and TLR4. GXM also stimulated nuclear NF-kappa B translocation in PBMC and RAW 264.7 cells. However, stimulation of these cells with GXM resulted in neither TNF-alpha secretion nor activation of the extracellular signal-regulated kinase 1/2, p38, and stress-activated protein kinase/c-Jun N-terminal kinase mitogen-activated protein kinase pathways. These findings suggest that TLRs, in conjunction with CD14, function as pattern recognition receptors for GXM. Furthermore, whereas GXM stimulates cells to translocate NF-kappa B to the nucleus, it does not induce activation of mitogen-activated protein kinase pathways or release of TNF-alpha. Taken together, these observations suggest a novel scenario whereby GXM stimulates cells via CD14 and TLR4, resulting in an incomplete activation of pathways necessary for TNF-alpha production.


Assuntos
Cryptococcus neoformans/imunologia , Proteínas de Drosophila , Glicoproteínas de Membrana/fisiologia , Polissacarídeos/imunologia , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Transporte Ativo do Núcleo Celular/imunologia , Animais , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Células CHO , Linhagem Celular , Cricetinae , Regulação da Expressão Gênica/imunologia , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Leucócitos Mononucleares/enzimologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Receptores de Lipopolissacarídeos/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Glicoproteínas de Membrana/genética , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Polissacarídeos/metabolismo , Polissacarídeos/fisiologia , Receptores de Superfície Celular/genética , Transdução de Sinais/genética , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
14.
J Immunol ; 166(6): 4074-82, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238656

RESUMO

We previously showed that viable Mycobacterium tuberculosis (Mtb) bacilli contain distinct ligands that activate cells via the mammalian Toll-like receptor (TLR) proteins TLR2 and TLR4. We now demonstrate that expression of a dominant negative TLR2 or TLR4 proteins in RAW 264.7 macrophages partially blocked Mtb-induced NF-kappa B activation. Coexpression of both dominant negative proteins blocked virtually all Mtb-induced NF-kappa B activation. The role of the TLR4 coreceptor MD-2 was also examined. Unlike LPS, Mtb-induced macrophage activation was not augmented by overexpression of ectopic MD-2. Moreover, cells expressing an LPS-unresponsive MD-2 mutant responded normally to Mtb. We also observed that the lipid A-like antagonist E5531 specifically inhibited TLR4-dependent Mtb-induced cellular responses. E5531 could substantially block LPS- and Mtb-induced TNF-alpha production in both RAW 264.7 cells and primary human alveolar macrophages (AM phi). E5531 inhibited Mtb-induced AM phi apoptosis in vitro, an effect that was a consequence of the inhibition of TNF-alpha production by E5531. In contrast, E5531 did not inhibit Mtb-induced NO production in RAW 264.7 cells and AM phi. Mtb-stimulated peritoneal macrophages from TLR2- and TLR4-deficient animals produced similar amounts of NO compared with control animals, demonstrating that these TLR proteins are not required for Mtb-induced NO production. Lastly, we demonstrated that a dominant negative MyD88 mutant could block Mtb-induced activation of the TNF-alpha promoter, but not the inducible NO synthase promoter, in murine macrophages. Together, these data suggest that Mtb-induced TNF-alpha production is largely dependent on TLR signaling. In contrast, Mtb-induced NO production may be either TLR independent or mediated by TLR proteins in a MyD88-independent manner.


Assuntos
Proteínas de Drosophila , Lipídeo A/farmacologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/antagonistas & inibidores , Mycobacterium tuberculosis/fisiologia , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Antígenos de Superfície/biossíntese , Antígenos de Superfície/fisiologia , Antituberculosos/farmacologia , Apoptose/efeitos dos fármacos , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Feminino , Regulação da Expressão Gênica , Lipídeo A/análogos & derivados , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Antígeno 96 de Linfócito , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/citologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacologia , Glicoproteínas de Membrana/fisiologia , Mesocricetus , Camundongos , Camundongos Endogâmicos C3H , Mutação , Mycobacterium tuberculosis/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Regiões Promotoras Genéticas/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Tuberculose/mortalidade , Tuberculose/prevenção & controle , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
15.
J Biol Chem ; 276(3): 1873-80, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11032843

RESUMO

ER-112022 is a novel acyclic synthetic lipid A analog that contains six symmetrically organized fatty acids on a noncarbohydrate backbone. Chinese hamster ovary (CHO)-K1 fibroblasts and U373 human astrocytoma cells do not respond to lipopolysaccharide (LPS) in the absence of CD14. In contrast, exposure to ER-112022 effectively induced activation of CHO and U373 cells under serum-free conditions. Expression of CD14 was not necessary for cells to respond to ER-112022, although the presence of soluble CD14 enhanced the sensitivity of the response. Several lines of evidence suggested that ER-112022 stimulates cells via the LPS signal transduction pathway. First, the diglucosamine-based LPS antagonists E5564 and E5531 blocked ER-112022-induced stimulation of CHO-K1, U373, and RAW264.7 cells. Second, ER-112022 was unable to activate C3H/HeJ mouse peritoneal macrophages, containing a mutation in Toll-like receptor (TLR) 4, as well as HEK293 cells, an epithelial cell line that does not express TLR4. Third, ER-112022 activated NF-kappaB in HEK293 cells transfected with TLR4/MD-2. Finally, tumor necrosis factor release from primary human monocytes exposed to ER-112022 was blocked by TLR4 antibodies but not by TLR2 antibodies. Our results suggest that ER-112022 and the family of lipid A-like LPS antagonists can functionally associate with TLR4 in the absence of CD14. Synthetic molecules like ER-112022 may prove to be valuable tools to characterize elements in the LPS receptor complex, as well as to activate or inhibit the TLR4 signaling pathway for therapeutic purposes.


Assuntos
Proteínas de Drosophila , Lipídeo A/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Animais , Configuração de Carboidratos , Linhagem Celular , Lipídeo A/química , Lipopolissacarídeos/química , Fosfolipídeos/farmacologia , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção
16.
Infect Immun ; 68(12): 6770-6, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11083794

RESUMO

Lipopolysaccharide (LPS) and related bacterial products can be recognized by host inflammatory cells in a particulate, bacterium-bound form, as well as in various soluble, released forms. In the present study we have compared the mechanisms used by LPS, detoxified LPS (DLPS), and mannuronic acid polymers (M-polymers), in solution or covalently linked to particles, in stimulating monocytes to tumor necrosis factor (TNF) production. The addition of recombinant LPS binding protein (LBP) and/or soluble CD14 (sCD14) enhanced the production of TNF from monocytes stimulated with soluble LPS, DLPS, or M-polymer, but did not affect the response to M-polymer or DLPS attached to particles. Treatment of monocytes with antibody to CD14, CD18, or CD11b showed that CD14, but not CR3 (CD11b/CD18), mediated monocyte TNF production in response to the soluble antigens. In contrast, anti-CD14, anti-CD11b and anti-CD18 monoclonal antibodies all inhibited the response to the particulate stimuli. On the other hand, B975, a synthetic analog of Rhodobacter capsulatus lipid A, completely abrogated the monocyte TNF response induced by LPS but did not affect the TNF induction by DLPS or M-polymer, either in soluble or particulate forms. These data demonstrate that the engagement of immune receptors by bacterial products such as LPS, DLPS, and M-polymer is dependent upon the presentation form of their constituent carbohydrates, and that factors such as aggregation state, acylation, carbohydrate chain length, and solid versus liquid phase of bacterial ligands influence the mechanisms used by cells in mediating proinflammatory responses.


Assuntos
Proteínas de Fase Aguda , Alginatos/farmacologia , Antígenos CD18/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana , Animais , Células CHO , Proteínas de Transporte/fisiologia , Cricetinae , Ácido Glucurônico , Ácidos Hexurônicos , Antígeno de Macrófago 1/fisiologia , Monócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese
17.
J Immunol ; 165(8): 4272-80, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11035061

RESUMO

Gram-negative bacteria and the LPS constituent of their outer membranes stimulate the release of inflammatory mediators believed to be responsible for the clinical manifestations of septic shock. The GPI-linked membrane protein, CD14, initiates the signaling cascade responsible for the induction of this inflammatory response by LPS. In this paper, we report the generation and characterization of CD14-null mice in which the entire coding region of CD14 was deleted. As expected, LPS failed to elicit TNF-alpha and IL-6 production in macrophages taken from these animals, and this loss in responsiveness is associated with impaired activation of both the NF-kappaB and the c-Jun N-terminal mitogen-activated protein kinase pathways. The binding and uptake of heat-killed Escherichia coli, measured by FACS analysis, did not differ between CD14-null and wild-type macrophages. However, in contrast to the findings with LPS, whole E. coli stimulated similar levels of TNF-alpha release from CD14-null and wild-type macrophages at a dose of 10 bioparticles per cell. This effect was dose dependent, and at lower bacterial concentrations CD14-deficient macrophages produced significantly less TNF-alpha than wild type. Approximately half of this CD14-independent response appeared to be mediated by CD11b/CD18, as demonstrated by receptor blockade using neutrophil inhibitory factor. An inhibitor of phagocytosis, cytochalasin B, abrogated the induction of TNF-alpha in CD14-deficient macrophages by E. coli. These data indicate that CD14 is essential for macrophage responses to free LPS, whereas other receptors, including CD11b/CD18, can compensate for the loss of CD14 in response to whole bacteria.


Assuntos
Escherichia coli/imunologia , Deleção de Genes , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Animais , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Antígenos CD18/fisiologia , Linhagem Celular , Citocinas/biossíntese , Escherichia coli/fisiologia , Feminino , Receptores de Lipopolissacarídeos/sangue , Ativação de Macrófagos/genética , Antígeno de Macrófago 1/fisiologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Insercional , Fagocitose/imunologia , Transdução de Sinais/imunologia
18.
Cytokine Growth Factor Rev ; 11(3): 219-32, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10817965

RESUMO

In 1997, a human homologue of the Drosophila Toll protein was described, a protein later to be designated Toll-like receptor 4 (TLR4). Since that time, additional human and murine TLR proteins have been identified. Mammalian TLR proteins appear to represent a conserved family of innate immune recognition receptors. These receptors are coupled to a signaling pathway that is conserved in mammals, insects, and plants, resulting in the activation of genes that mediate innate immune defenses. Numerous studies have now identified a wide variety of chemically-diverse bacterial products that serve as putative ligands for TLR proteins. More recent studies have identified the first endogenous protein ligands for TLR proteins. TLR signaling represents a key feature of innate immune response to pathogen invasion.


Assuntos
Proteínas de Drosophila , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Drosophila/genética , Drosophila/metabolismo , Humanos , Proteínas de Insetos/fisiologia , Ligantes , Lipopolissacarídeos/química , Mamíferos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Receptores de Superfície Celular/genética , Homologia de Sequência de Aminoácidos , Receptor 4 Toll-Like , Receptor 5 Toll-Like , Receptor 6 Toll-Like , Receptores Toll-Like
19.
J Immunol ; 164(4): 2064-9, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10657659

RESUMO

Human Toll like receptor (TLR) 2 has been implicated as a signaling receptor for LPS from Gram-negative bacteria and cell wall components from Gram-positive organisms. In this study, we investigated whether TLR2 can signal cell activation by the heat-killed group B streptococci type III (GBS) and Listeria monocytogenes (HKLM). HKLM, but not GBS, showed a time- and dose-dependent activation of Chinese hamster ovary cells transfected with human TLR2, as measured by translocation of NF-kappaB and induction of IL-6 production. A mAb recognizing a TLR2-associated epitope (TL2.1) was generated that inhibited IL-6 production from Chinese hamster ovary-TLR2 cells stimulated with HKLM or LPS. The TL2.1 mAb reduced HKLM-induced TNF production from human monocytes by 60%, whereas a CD14 mAb (3C10) reduced the TNF production by 30%. However, coadministrating TL2.1 and 3C10 inhibited the TNF response by 80%. In contrast to this, anti-CD14 blocked LPS-induced TNF production from monocytes, whereas anti-TLR2 showed no inhibition. Neither TL2.1 nor 3C10 affected GBS-induced TNF production. These results show that TLR2 can function as a signaling receptor for HKLM, possibly together with CD14, but that TLR2 is unlikely to be involved in cell activation by GBS. Furthermore, although LPS can activate transfected cell lines through TLR2, this receptor does not seem to be the main transducer of LPS activation of human monocytes. Thus, our data demonstrate the ability of TLR2 to distinguish between different pathogens.


Assuntos
Proteínas de Drosophila , Lipopolissacarídeos/imunologia , Listeria monocytogenes/imunologia , Glicoproteínas de Membrana/fisiologia , Monócitos/imunologia , Monócitos/microbiologia , Receptores de Superfície Celular/fisiologia , Streptococcus agalactiae/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Reações Antígeno-Anticorpo , Células CHO , Cricetinae , Relação Dose-Resposta Imunológica , Epitopos/imunologia , Temperatura Alta , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/biossíntese , Receptores de Lipopolissacarídeos/fisiologia , Lipopolissacarídeos/antagonistas & inibidores , Ativação de Macrófagos , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Receptores de Superfície Celular/imunologia , Receptor 2 Toll-Like , Receptores Toll-Like , Transfecção
20.
J Clin Invest ; 105(4): 497-504, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10683379

RESUMO

Lipopolysaccharide (LPS) is the main inducer of shock and death in Gram-negative sepsis. Recent evidence suggests that LPS-induced signal transduction begins with CD14-mediated activation of 1 or more Toll-like receptors (TLRs). The lipid A analogues lipid IVa and Rhodobacter sphaeroides lipid A (RSLA) exhibit an uncommon species-specific pharmacology. Both compounds inhibit the effects of LPS in human cells but display LPS-mimetic activity in hamster cells. We transfected human TLR4 or human TLR2 into hamster fibroblasts to determine if either of these LPS signal transducers is responsible for the species-specific pharmacology. RSLA and lipid IVa strongly induced NF-kappaB activity and IL-6 release in Chinese hamster ovary fibroblasts expressing CD14 (CHO/CD14), but these compounds antagonized LPS antagonists in CHO/CD14 fibroblasts that overexpressed human TLR4. No such antagonism occurred in cells overexpressing human TLR2. We cloned TLR4 from hamster macrophages and found that human THP-1 cells expressing the hamster TLR4 responded to lipid IVa as an LPS mimetic, as if they were hamster in origin. Hence, cells heterologously overexpressing TLR4 from different species acquired a pharmacological phenotype with respect to recognition of lipid A substructures that corresponded to the species from which the TLR4 transgene originated. These data suggest that TLR4 is the central lipid A-recognition protein in the LPS receptor complex.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Células CHO , Cricetinae , Glicolipídeos/metabolismo , Humanos , Ligantes , Lipídeo A/análogos & derivados , Lipídeo A/metabolismo , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/citologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Mimetismo Molecular , Receptores de Superfície Celular/genética , Proteínas Recombinantes/metabolismo , Rhodobacter sphaeroides , Transdução de Sinais , Especificidade da Espécie , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...