Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Parasite Immunol ; 33(4): 217-25, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21204848

RESUMEN

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.


Asunto(s)
Citocinas/biosíntesis , Enterocytozoon/inmunología , Inmunidad Innata , Microsporidiosis/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Enterocytozoon/aislamiento & purificación , Heces/microbiología , Haplorrinos , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética
2.
J Immunol ; 167(12): 7069-76, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11739528

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Interleucina-1 , Streptococcus agalactiae/fisiología , Animales , Antígenos de Superficie/fisiología , Factores Biológicos/metabolismo , Células CHO , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Células Cultivadas , Cricetinae , Humanos , Mediadores de Inflamación/metabolismo , Antígeno 96 de los Linfocitos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Modelos Inmunológicos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Sepsis/inmunología , Infecciones Estreptocócicas/inmunología , Receptor Toll-Like 1 , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptor Toll-Like 6 , Receptores Toll-Like , Transfección , Factor de Necrosis Tumoral alfa/biosíntesis
4.
J Immunol ; 167(4): 2257-67, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11490013

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/inmunología , Células CHO/inmunología , Proteínas de Drosophila , Tolerancia Inmunológica , Receptores de Lipopolisacáridos/biosíntesis , Lipopolisacáridos/inmunología , Glicoproteínas de Membrana/biosíntesis , Mycobacterium tuberculosis/inmunología , Receptores de Superficie Celular/biosíntesis , Animales , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Células CHO/metabolismo , Línea Celular , Células Clonales , Cricetinae , Activación Enzimática/genética , Activación Enzimática/inmunología , Humanos , Tolerancia Inmunológica/genética , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/antagonistas & inhibidores , Antígeno 96 de los Linfocitos , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptores de Superficie Celular/genética , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Transfección
5.
Circ Res ; 89(3): 244-50, 2001 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-11485974

RESUMEN

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.


Asunto(s)
Chaperonina 60/metabolismo , Proteínas de Drosophila , Glicoproteínas de Membrana/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Bromodesoxiuridina , Butadienos/farmacología , División Celular/efectos de los fármacos , Células Cultivadas , Chaperonina 60/antagonistas & inhibidores , Chaperonina 60/farmacología , Infecciones por Chlamydophila/metabolismo , Chlamydophila pneumoniae/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Calor , Humanos , Lípido A/análogos & derivados , Lípido A/farmacología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteína Quinasa 3 Activada por Mitógenos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/microbiología , Nitrilos/farmacología , ARN Mensajero/biosíntesis , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vena Safena , Receptor Toll-Like 4 , Receptores Toll-Like
6.
J Immunol ; 167(2): 910-8, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11441098

RESUMEN

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.


Asunto(s)
Aciltransferasas , Presentación de Antígeno/inmunología , Antígenos Bacterianos , Proteínas Bacterianas/farmacología , Proteínas de Drosophila , Antígenos de Histocompatibilidad Clase II/biosíntesis , Inmunosupresores/farmacología , Lipoproteínas/farmacología , Macrófagos/inmunología , Glicoproteínas de Membrana/fisiología , Mycobacterium tuberculosis/inmunología , Receptores de Superficie Celular/fisiología , Animales , Presentación de Antígeno/genética , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Detergentes , Epítopos/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Octoxinol , Polietilenglicoles , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like
7.
J Exp Med ; 194(1): 79-88, 2001 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-11435474

RESUMEN

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.


Asunto(s)
Antígenos de Superficie/genética , Proteínas de Drosophila , Endotoxinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Antígenos de Superficie/metabolismo , Células CHO/efectos de los fármacos , Línea Celular , Células Clonales , Clonación Molecular , Cricetinae , Análisis Mutacional de ADN , Prueba de Complementación Genética , Humanos , Interleucina-1/farmacología , Interleucina-6/metabolismo , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/toxicidad , Antígeno 96 de los Linfocitos , Glicoproteínas de Membrana/genética , Mutación , Receptores de Superficie Celular/genética , Transducción de Señal , Receptor Toll-Like 4 , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/farmacología
8.
J Immunol ; 167(1): 416-23, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11418678

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Glicosilfosfatidilinositoles/fisiología , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Trypanosoma cruzi/inmunología , Animales , Células CHO , Línea Celular , Cricetinae , Relación Dosis-Respuesta Inmunológica , Glucolípidos/fisiología , Glicosilfosfatidilinositoles/aislamiento & purificación , Inflamación/inmunología , Inflamación/parasitología , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , FN-kappa B/fisiología , Fosfolípidos/fisiología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores de Interleucina-2/biosíntesis , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Transfección , Trypanosoma cruzi/química , Trypanosoma cruzi/crecimiento & desarrollo
9.
Infect Immun ; 69(4): 2230-6, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11254578

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas de Drosophila , Lípido A/análogos & derivados , Lipopolisacáridos/toxicidad , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Neisseria meningitidis/inmunología , Receptores de Superficie Celular/fisiología , Animales , Células CHO , Pared Celular/fisiología , Cricetinae , Humanos , Lípido A/farmacología , Receptores de Lipopolisacáridos/fisiología , Mutación , Receptor Toll-Like 2 , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Immunol ; 166(7): 4620-6, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11254720

RESUMEN

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.


Asunto(s)
Cryptococcus neoformans/inmunología , Proteínas de Drosophila , Glicoproteínas de Membrana/fisiología , Polisacáridos/inmunología , Receptores de Superficie Celular/fisiología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Transporte Activo de Núcleo Celular/inmunología , Animales , Sitios de Unión/genética , Sitios de Unión/inmunología , Células CHO , Línea Celular , Cricetinae , Regulación de la Expresión Génica/inmunología , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Leucocitos Mononucleares/enzimología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Receptores de Lipopolisacáridos/fisiología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Glicoproteínas de Membrana/genética , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Polisacáridos/metabolismo , Polisacáridos/fisiología , Receptores de Superficie Celular/genética , Transducción de Señal/genética , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Transfección , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
12.
J Immunol ; 166(6): 4074-82, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238656

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Lípido A/farmacología , Macrófagos/microbiología , Glicoproteínas de Membrana/antagonistas & inhibidores , Mycobacterium tuberculosis/fisiología , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/fisiología , Antituberculosos/farmacología , Apoptosis/efectos de los fármacos , Células CHO , Línea Celular , Cricetinae , Cricetulus , Femenino , Regulación de la Expresión Génica , Lípido A/análogos & derivados , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/microbiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Glicoproteínas de Membrana/fisiología , Mesocricetus , Ratones , Ratones Endogámicos C3H , Mutación , Mycobacterium tuberculosis/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Regiones Promotoras Genéticas/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Tuberculosis/mortalidad , Tuberculosis/prevención & control , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
13.
J Biol Chem ; 276(3): 1873-80, 2001 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-11032843

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Lípido A/metabolismo , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Animales , Conformación de Carbohidratos , Línea Celular , Lípido A/química , Lipopolisacáridos/química , Fosfolípidos/farmacología , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Transfección
14.
Infect Immun ; 68(12): 6770-6, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11083794

RESUMEN

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.


Asunto(s)
Proteínas de Fase Aguda , Alginatos/farmacología , Antígenos CD18/fisiología , Receptores de Lipopolisacáridos/fisiología , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana , Animales , Células CHO , Proteínas Portadoras/fisiología , Cricetinae , Ácido Glucurónico , Ácidos Hexurónicos , Antígeno de Macrófago-1/fisiología , Monocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis
15.
J Immunol ; 165(8): 4272-80, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11035061

RESUMEN

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.


Asunto(s)
Escherichia coli/inmunología , Eliminación de Gen , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas de la Membrana Bacteriana Externa/fisiología , Antígenos CD18/fisiología , Línea Celular , Citocinas/biosíntesis , Escherichia coli/fisiología , Femenino , Receptores de Lipopolisacáridos/sangre , Activación de Macrófagos/genética , Antígeno de Macrófago-1/fisiología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Insercional , Fagocitosis/inmunología , Transducción de Señal/inmunología
16.
Cytokine Growth Factor Rev ; 11(3): 219-32, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10817965

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Drosophila/genética , Drosophila/metabolismo , Humanos , Proteínas de Insectos/fisiología , Ligandos , Lipopolisacáridos/química , Mamíferos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Receptores de Superficie Celular/genética , Homología de Secuencia de Aminoácido , Receptor Toll-Like 4 , Receptor Toll-Like 5 , Receptor Toll-Like 6 , Receptores Toll-Like
17.
J Immunol ; 164(4): 2064-9, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10657659

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Lipopolisacáridos/inmunología , Listeria monocytogenes/inmunología , Glicoproteínas de Membrana/fisiología , Monocitos/inmunología , Monocitos/microbiología , Receptores de Superficie Celular/fisiología , Streptococcus agalactiae/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Reacciones Antígeno-Anticuerpo , Células CHO , Cricetinae , Relación Dosis-Respuesta Inmunológica , Epítopos/inmunología , Calor , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Receptores de Lipopolisacáridos/fisiología , Lipopolisacáridos/antagonistas & inhibidores , Activación de Macrófagos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Receptores de Superficie Celular/inmunología , Receptor Toll-Like 2 , Receptores Toll-Like , Transfección
18.
J Clin Invest ; 105(4): 497-504, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10683379

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Células CHO , Cricetinae , Glucolípidos/metabolismo , Humanos , Ligandos , Lípido A/análogos & derivados , Lípido A/metabolismo , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/antagonistas & inhibidores , Macrófagos/citología , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Imitación Molecular , Receptores de Superficie Celular/genética , Proteínas Recombinantes/metabolismo , Rhodobacter sphaeroides , Transducción de Señal , Especificidad de la Especie , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like
19.
Life Sci ; 68(3): 241-58, 2000 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-11191641

RESUMEN

Beginning in 1997 with the identification of the first human homologue of the Drosophila protein Toll, a family of related molecules have been identified in both humans and other mammals. These Toll-like receptor (TLR) proteins appear to represent a conserved family of innate immune recognition receptors. TLR proteins share extended homology with receptors for the cytokines interleukin 1 (IL-1) and interleukin 18 (IL-18). These receptors are coupled to a signaling pathway that is conserved in mammals, insects, and plants, resulting in cellular activation, thereby stimulating innate immune defenses. A variety of bacterial and fungal products have been identified that serve as TLR ligands, and more recent studies have identified the first endogenous protein ligands for TLR proteins. While TLR signaling is likely to be a key feature of innate immune responses, these proteins may also regulate homeostasis via interaction with endogenous protein ligands.


Asunto(s)
Proteínas de Drosophila , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiología , Receptores de Superficie Celular/química , Receptores de Superficie Celular/fisiología , Secuencia de Aminoácidos , Animales , Drosophila , Humanos , Subunidad alfa del Receptor de Interleucina-18 , Ratones , Datos de Secuencia Molecular , Receptores de Interleucina/química , Receptores de Interleucina/fisiología , Receptores de Interleucina-1/química , Receptores de Interleucina-1/fisiología , Receptores de Interleucina-18 , Homología de Secuencia de Aminoácido , Transducción de Señal , Receptor Toll-Like 5 , Receptores Toll-Like
20.
J Endotoxin Res ; 6(5): 411-5, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11521065

RESUMEN

The inflammatory response to bacterial infections plays an important role in the detection and elimination of invading micro-organisms. Various components of the bacterial cell wall are capable of activating this pro-inflammatory response. In the case of Gram-negative bacteria, lipopolysaccharide (LPS) is the dominant trigger, although other bacterial factors are also capable of activating this systemic inflammatory response. Recently, Toll-like receptors (TLRs) have been implicated in host responses to bacterial pathogens. Specifically, TLR4 mediates LPS responses while TLR2 plays a broader role in the recognition of a variety of bacteria and bacterial antigens. The experiments in this study were designed to examine the role of Gram-negative cell wall components, other than LPS, and their cellular receptors in the host response to infection using an LPS-deficient mutant of Neisseria meningitidis. Although less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in a variety of cell types. Moreover, cellular activation by this mutant required expression of CD14 and TLR2.


Asunto(s)
Proteínas de Drosophila , Lipopolisacáridos/inmunología , Glicoproteínas de Membrana/inmunología , Neisseria meningitidis/inmunología , Receptores de Superficie Celular/inmunología , Animales , Células CHO , Pared Celular/inmunología , Cricetinae , Humanos , Técnicas In Vitro , Mediadores de Inflamación/inmunología , Leucocitos Mononucleares/inmunología , Receptores de Lipopolisacáridos/genética , Glicoproteínas de Membrana/genética , Mutación , Neisseria meningitidis/genética , Receptores de Superficie Celular/genética , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA