Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 10(7): e1004207, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24992093

RESUMO

Delivery of microbial products into the mammalian cell cytosol by bacterial secretion systems is a strong stimulus for triggering pro-inflammatory host responses. Here we show that Salmonella enterica serovar Typhi (S. Typhi), the causative agent of typhoid fever, tightly regulates expression of the invasion-associated type III secretion system (T3SS-1) and thus fails to activate these innate immune signaling pathways. The S. Typhi regulatory protein TviA rapidly repressed T3SS-1 expression, thereby preventing RAC1-dependent, RIP2-dependent activation of NF-κB in epithelial cells. Heterologous expression of TviA in S. enterica serovar Typhimurium (S. Typhimurium) suppressed T3SS-1-dependent inflammatory responses generated early after infection in animal models of gastroenteritis. These results suggest that S. Typhi reduces intestinal inflammation by limiting the induction of pathogen-induced processes through regulation of virulence gene expression.


Assuntos
Sistemas de Secreção Bacterianos/imunologia , Gastroenterite/imunologia , Imunidade Inata , Salmonella typhi/imunologia , Febre Tifoide/imunologia , Fatores de Virulência/imunologia , Animais , Sistemas de Secreção Bacterianos/genética , Bovinos , Modelos Animais de Doenças , Gastroenterite/genética , Gastroenterite/patologia , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/imunologia , Células HeLa , Humanos , Camundongos , Salmonella typhi/genética , Salmonella typhi/patogenicidade , Febre Tifoide/genética , Febre Tifoide/patologia , Fatores de Virulência/genética
2.
Trends Immunol ; 35(3): 123-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24268520

RESUMO

Nucleotide-binding oligomerization domain protein (NOD)1 and NOD2 participate in signaling pathways that detect pathogen-induced processes, such as the presence of peptidoglycan fragments in the host cell cytosol, as danger signals. Recent work suggests that peptidoglycan fragments activate NOD1 indirectly, through activation of the small Rho GTPase Ras-related C3 botulinum toxin substrate 1 (RAC1). Excessive activation of small Rho GTPases by virulence factors of enteric pathogens also triggers the NOD1 signaling pathway. Many enteric pathogens use virulence factors that alter the activation state of small Rho GTPases, thereby manipulating the host cell cytoskeleton of intestinal epithelial cells to promote bacterial attachment or entry. These data suggest that the NOD1 signaling pathway in intestinal epithelial cells provides an important sentinel function for detecting 'breaking and entering' by enteric pathogens.


Assuntos
Imunidade nas Mucosas/imunologia , Intestinos/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Transdução de Sinais/imunologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/imunologia , Humanos , Intestinos/microbiologia , Virulência
3.
Cell Host Microbe ; 14(2): 159-70, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23954155

RESUMO

Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.


Assuntos
Brucella abortus/fisiologia , Glucose/metabolismo , Ativação de Macrófagos , Macrófagos/microbiologia , Viabilidade Microbiana , PPAR gama/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/imunologia , Brucella abortus/metabolismo , Macrófagos/imunologia , Camundongos
4.
Dev Comp Immunol ; 41(3): 316-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23628643

RESUMO

Toll-like receptors (TLRs) are a major class of innate immune pattern recognition receptors that have a key role in immune homeostasis and the defense against infections. The research explosion that followed the discovery of TLRs more than a decade ago has boosted fundamental knowledge on the function of the immune system and the resistance against disease, providing a rational for clinical modulation of the immune response. In addition, the conserved nature of the ancient TLR system throughout the animal kingdom has enabled a comparative biology approach to understand the evolution, structural architecture, and function of TLRs. In the present review we focus on TLR biology in the avian species, and, especially, on the unique functional properties of the chicken TLR repertoire.


Assuntos
Galinhas/imunologia , Imunidade Inata , Receptores Toll-Like/imunologia , Animais , Evolução Biológica , Citocinas/genética , Citocinas/imunologia , Flagelina/imunologia , Flagelina/metabolismo , Regulação da Expressão Gênica , Humanos , Ligantes , Mamíferos/imunologia , Especificidade de Órgãos , Transdução de Sinais , Receptores Toll-Like/classificação , Receptores Toll-Like/genética
5.
Nature ; 496(7444): 233-7, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23542589

RESUMO

Our innate immune system distinguishes microbes from self by detecting conserved pathogen-associated molecular patterns. However, these are produced by all microbes, regardless of their pathogenic potential. To distinguish virulent microbes from those with lower disease-causing potential the innate immune system detects conserved pathogen-induced processes, such as the presence of microbial products in the host cytosol, by mechanisms that are not fully resolved. Here we show that NOD1 senses cytosolic microbial products by monitoring the activation state of small Rho GTPases. Activation of RAC1 and CDC42 by bacterial delivery or ectopic expression of SopE, a virulence factor of the enteric pathogen Salmonella, triggered the NOD1 signalling pathway, with consequent RIP2 (also known as RIPK2)-mediated induction of NF-κB-dependent inflammatory responses. Similarly, activation of the NOD1 signalling pathway by peptidoglycan required RAC1 activity. Furthermore, constitutively active forms of RAC1, CDC42 and RHOA activated the NOD1 signalling pathway. Our data identify the activation of small Rho GTPases as a pathogen-induced process sensed through the NOD1 signalling pathway.


Assuntos
Proteína Adaptadora de Sinalização NOD1/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Citosol/metabolismo , Feminino , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Peptidoglicano/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Salmonella typhimurium/genética , Transdução de Sinais , Fatores de Virulência/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
6.
Science ; 339(6120): 708-11, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23393266

RESUMO

Changes in the microbial community structure are observed in individuals with intestinal inflammatory disorders. These changes are often characterized by a depletion of obligate anaerobic bacteria, whereas the relative abundance of facultative anaerobic Enterobacteriaceae increases. The mechanisms by which the host response shapes the microbial community structure, however, remain unknown. We show that nitrate generated as a by-product of the inflammatory response conferred a growth advantage to the commensal bacterium Escherichia coli in the large intestine of mice. Mice deficient in inducible nitric oxide synthase did not support the growth of E. coli by nitrate respiration, suggesting that the nitrate generated during inflammation was host-derived. Thus, the inflammatory host response selectively enhances the growth of commensal Enterobacteriaceae by generating electron acceptors for anaerobic respiration.


Assuntos
Colite/metabolismo , Colite/microbiologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Intestino Grosso/microbiologia , Nitratos/metabolismo , Anaerobiose , Animais , Bovinos , Escherichia coli/genética , Íleo/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/metabolismo
7.
PLoS Pathog ; 8(9): e1002918, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23028318

RESUMO

Intestinal inflammation changes the luminal habitat for microbes through mechanisms that have not been fully resolved. We noticed that the FepE regulator of very long O-antigen chain assembly in the enteric pathogen Salmonella enterica serotype Typhimurium (S. Typhimurium) conferred a luminal fitness advantage in the mouse colitis model. However, a fepE mutant was not defective for survival in tissue, resistance to complement or resistance to polymyxin B. We performed metabolite profiling to identify changes in the luminal habitat that accompany S. Typhimurium-induced colitis. This analysis suggested that S. Typhimurium-induced colitis increased the luminal concentrations of total bile acids. A mutation in fepE significantly reduced the minimal inhibitory concentration (MIC) of S. Typhimurium for bile acids in vitro. Oral administration of the bile acid sequestrant cholestyramine resin lowered the concentrations of total bile acids in colon contents during S. Typhimurium infection and significantly reduced the luminal fitness advantage conferred by the fepE gene in the mouse colitis model. Collectively, these data suggested that very long O-antigen chains function in bile acid resistance of S. Typhimurium, a property conferring a fitness advantage during luminal growth in the inflamed intestine.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colite/microbiologia , Antígenos O/genética , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Animais , Resina de Colestiramina/administração & dosagem , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Mutação , Antígenos O/química , Antígenos O/metabolismo , Polimixina B , Salmonelose Animal/imunologia , Salmonella typhimurium/enzimologia , Salmonella typhimurium/genética , Salmonella typhimurium/crescimento & desenvolvimento
8.
mBio ; 2(6)2011.
Artigo em Inglês | MEDLINE | ID: mdl-22186610

RESUMO

The invasion-associated type III secretion system (T3SS-1) of Salmonella enterica serotype Typhimurium (S. Typhimurium) activates the transcription factor NF-κB in tissue culture cells and induces inflammatory responses in animal models through unknown mechanisms. Here we show that bacterial delivery or ectopic expression of SipA, a T3SS-1-translocated protein, led to the activation of the NOD1/NOD2 signaling pathway and consequent RIP2-mediated induction of NF-κB-dependent inflammatory responses. SipA-mediated activation of NOD1/NOD2 signaling was independent of bacterial invasion in vitro but required an intact T3SS-1. In the mouse colitis model, SipA triggered mucosal inflammation in wild-type mice but not in NOD1/NOD2-deficient mice. These findings implicate SipA-driven activation of the NOD1/NOD2 signaling pathway as a mechanism by which the T3SS-1 induces inflammatory responses in vitro and in vivo.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Infecções por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Transdução de Sinais , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Infecções por Salmonella/genética , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética , Fatores de Virulência/genética
9.
Cell Microbiol ; 13(11): 1639-47, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21740501

RESUMO

Non-typhoidal Salmonella (NTS) serotypes cause a localized gastroenteritis in immunocompetent individuals. In contrast, primary immunodeficiencies that impair interleukin-23 (IL-23)-dependent pathways are associated in humans with disseminated NTS bloodstream infections (bacteraemia). The recent use of animal models has helped to define the role the IL-23 axis plays during NTS gastroenteritis, but additional work is needed to elucidate how this host defence pathway prevents NTS bacteraemia.


Assuntos
Interleucina-23/imunologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/patologia , Salmonella/imunologia , Salmonella/patogenicidade , Animais , Modelos Animais de Doenças , Gastroenterite/imunologia , Gastroenterite/microbiologia , Gastroenterite/patologia , Humanos , Interleucina-23/deficiência , Sepse/imunologia , Sepse/microbiologia , Sepse/patologia
10.
Infect Immun ; 79(8): 3131-40, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21576324

RESUMO

The development of T helper 17 (T(H)17) cells is a well-established adaptive mechanism for the production of interleukin-17A (IL-17A), a cytokine involved in neutrophil recruitment. However, pathways contributing to mucosal expression of IL-17A during the initial phase of a bacterial infection have received less attention. Here we used the mouse colitis model of Salmonella enterica serotype Typhimurium infection to investigate the contribution of myeloid differentiation primary response protein 88 (MyD88) to inflammation and mucosal IL-17A expression. Expression of IL-23 in the cecal mucosa during S. Typhimurium colitis was dependent on the presence of MyD88. Furthermore, initial expression of IL-17A at 24 h after S. Typhimurium infection was dependent on MyD88 and the receptor for IL-1ß. IL-23 and IL-1ß synergized in inducing expression of IL-17A in splenic T cells in vitro. In the intestinal mucosa, IL-17A was produced by three distinct T cell populations, including δγ T cells, T(H)17 cells, and CD4(-)CD8(-) T cells. The absence of IL-1ß signaling or IL-17 signaling reduced CXC chemokine expression but did not alter the overall severity of pathological lesions in the cecal mucosa. In contrast, cecal pathology and neutrophil recruitment were markedly reduced in Myd88-deficient mice during the initial phases of S. Typhimurium infection. Collectively, these data demonstrate that MyD88-dependent mechanisms, including an initial expression of IL-17A, are important for orchestrating early inflammatory responses during S. Typhimurium colitis.


Assuntos
Colite/veterinária , Interleucina-17/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Salmonelose Animal/imunologia , Salmonelose Animal/patologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Animais , Ceco/imunologia , Ceco/microbiologia , Linhagem Celular , Colite/imunologia , Colite/microbiologia , Colite/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Histocitoquímica , Humanos , Interleucina-17/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia , Fator 88 de Diferenciação Mieloide/imunologia , Doenças dos Roedores/imunologia , Doenças dos Roedores/microbiologia , Doenças dos Roedores/patologia , Salmonelose Animal/microbiologia , Subpopulações de Linfócitos T/imunologia
11.
Cell Host Microbe ; 9(3): 167-168, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21402352

RESUMO

Salmonella survives in macrophages by using a molecular syringe to deliver proteins into the host-cell cytosol where they manipulate phagocyte physiology. Arpaia and colleagues (Arpaia et al., 2011) show that deployment of this virulence factor is triggered by the very responses that are intended to confer host resistance.

12.
Proc Natl Acad Sci U S A ; 108(12): 4968-73, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21383168

RESUMO

Toll-like receptors (TLRs) are innate receptors that show high conservation throughout the animal kingdom. Most TLRs can be clustered into phylogenetic groups that respond to similar types of ligands. One exception is avian TLR15. This receptor does not categorize into one of the existing groups of TLRs and its ligand is still unknown. Here we report that TLR15 is a sensor for secreted virulence-associated fungal and bacterial proteases. Activation of TLR15 involves proteolytic cleavage of the receptor ectodomain and stimulation of NF-κB-dependent gene transcription. Receptor activation can be mimicked by the expression of a truncated TLR15 of which the entire ectodomain is removed, suggesting that receptor cleavage alleviates receptor inhibition by the leucine-rich repeat domain. Our results indicate TLR15 as a unique type of innate immune receptor that combines TLR characteristics with an activation mechanism typical for the evolutionary distinct protease-activated receptors.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas Fúngicas/imunologia , Imunidade Inata/fisiologia , Peptídeo Hidrolases/imunologia , Receptores Toll-Like/imunologia , Animais , Proteínas de Bactérias/metabolismo , Células COS , Chlorocebus aethiops , Proteínas Fúngicas/metabolismo , Células HEK293 , Células HeLa , Humanos , NF-kappa B/imunologia , NF-kappa B/metabolismo , Peptídeo Hidrolases/metabolismo , Estrutura Terciária de Proteína , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Transcrição Gênica/genética , Transcrição Gênica/imunologia
13.
Cell Host Microbe ; 8(1): 36-43, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20638640

RESUMO

The intestinal immune system has to strike a delicate balance between initiating inflammatory responses against invading bacterial pathogens and avoiding their induction against microbiota colonizing the lumen. Adequate inflammatory responses against bacterial invasion result in the lumenal secretion of antimicrobial peptides, as well as the release of cytokines in tissue that recruit and activate phagocytes. However, pathogens have evolved to utilize these environmental changes in the inflamed intestine to promote colonization. This review focuses on the costs and benefits of intestinal inflammation and the fine interplay between the host, its microbiota, and enteric pathogens.


Assuntos
Bactérias/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Animais , Bactérias/imunologia , Humanos
14.
Cell Microbiol ; 12(10): 1495-505, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20497180

RESUMO

Responses to host amyloids and curli amyloid fibrils of Escherichia coli and Salmonella enterica serotype Typhimurium are mediated through Toll-like receptor (TLR) 2. Here we show that TLR2 alone was not sufficient for mediating responses to curli. Instead, transfection experiments with human cervical cancer (HeLa) cells and antibody-mediated inhibition of TLR signalling in human macrophage-like (THP-1) cells suggested that TLR2 interacts with TLR1 to recognize curli amyloid fibrils. TLR1/TLR2 also serves as a receptor for tri-acylated lipoproteins, which are produced by E. coli and other Gram-negative bacteria. Despite the presence of multiple TLR1/TLR2 ligands on intact bacterial cells, an inability to produce curli amyloid fibrils markedly reduced the ability of E. coli to induce TLR2-dependent responses in vitro and in vivo. Collectively, our data suggest that curli amyloid fibrils from enterobacterial biofilms significantly contribute to TLR1/TLR2-mediated host responses against intact bacterial cells.


Assuntos
Amiloide/imunologia , Proteínas de Bactérias/imunologia , Escherichia coli/imunologia , Salmonella typhimurium/imunologia , Receptor 1 Toll-Like/imunologia , Receptor 2 Toll-Like/imunologia , Amiloide/metabolismo , Proteínas de Bactérias/metabolismo , Biofilmes , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Escherichia coli/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Salmonella typhimurium/metabolismo
15.
J Immunol ; 185(1): 460-7, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20498358

RESUMO

TLRs comprise a family of evolutionary conserved sensory receptors that respond to distinct classes of ligands. For one major evolutionary branch of TLRs, the ligands are still largely unknown. Here we report the cloning and function of one member of this group, chicken TLR21 (chTLR21). This TLR is absent in the human species but has homologs in fish and frog and displays similarity with mouse TLR13. Expression of chTLR21 in HEK293 cells resulted in activation of NF-kappaB in response to unmethylated CpG DNA, typically recognized by mammalian TLR9. Silencing of chTLR21 (but not chTLR4) in chicken macrophages inhibited the response to CpG-DNA (but not to LPS), indicating similar functionality of the endogenous receptor. ChTLR21 responded to human- and murine-specific TLR9 ligands, as well as to bacterial genomic DNA isolated from Salmonella enterica serovar Enteritidis. Confocal microscopy located chTLR21 in the same intracellular compartments as human TLR9. Inhibition of the chTLR21 response by the endosomal maturation inhibitor chloroquine suggested that the receptor is functional in endolysosomes, as known for TLR9. The analogous localization and function of the phylogenetically only distantly related chTLR21 and mammalian TLR9 suggest that during evolution different classes of TLRs have emerged that recognize the same type of ligands.


Assuntos
Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Aviárias/deficiência , Proteínas Aviárias/isolamento & purificação , Células COS , Linhagem Celular , Galinhas , Chlorocebus aethiops , Clonagem Molecular , Ilhas de CpG/imunologia , Proteínas de Peixes/metabolismo , Células HeLa , Humanos , Imunidade Inata/genética , Ligantes , Camundongos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos/química , Ligação Proteica/imunologia , Homologia de Sequência de Aminoácidos , Receptor Toll-Like 9/genética , Receptores Toll-Like/deficiência , Receptores Toll-Like/isolamento & purificação , Proteínas de Xenopus/metabolismo
16.
J Biol Chem ; 285(16): 12149-58, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20164175

RESUMO

Bacterial flagellin is important for intestinal immune homeostasis. Flagellins from most species activate Toll-like receptor 5 (TLR5). The principal bacterial food-borne pathogen Campylobacter jejuni escapes TLR5 recognition, probably due to an alternate flagellin subunit structure. We investigated the molecular basis of TLR5 evasion by aiming to reconstitute TLR5 stimulating activity in live C. jejuni. Both native glycosylated C. jejuni flagellins (FlaA and FlaB) and recombinant proteins purified from Escherichia coli failed to activate NF-kappaB in HEK293 cells expressing TLR5. Introduction of multiple defined regions from Salmonella flagellin into C. jejuni FlaA via a recombinatorial approach revealed three regions critical for the activation of human and mouse TLR5, including a beta-hairpin structure not previously implicated in TLR5 recognition. Surprisingly, this domain was not required for the activation of chicken TLR5, indicating a selective requirement for the beta-hairpin in the recognition of mammalian TLR5. Expression of the active chimeric protein in C. jejuni resulted in secreted glycosylated flagellin that induced a potent TLR5 response. Overall, our results reveal a novel structural requirement for TLR5 recognition of bacterial flagellin and exclude flagellin glycosylation as an additional mechanism of bacterial evasion of the TLR5 response.


Assuntos
Campylobacter jejuni/metabolismo , Flagelina/química , Flagelina/metabolismo , Receptor 5 Toll-Like/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Campylobacter jejuni/genética , Campylobacter jejuni/patogenicidade , Linhagem Celular , Galinhas , Primers do DNA/genética , Flagelina/genética , Glicosilação , Células HT29 , Células HeLa , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Subunidades Proteicas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Salmonella enteritidis/genética , Salmonella enteritidis/metabolismo , Homologia de Sequência de Aminoácidos , Receptor 5 Toll-Like/genética
17.
Infect Immun ; 78(3): 1229-38, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20038539

RESUMO

Campylobacter infection in humans is accompanied by severe inflammation of the intestinal mucosa, in contrast to colonization of chicken. The basis for the differential host response is unknown. Toll-like receptors (TLRs) sense and respond to microbes in the body and participate in the induction of an inflammatory response. Thus far, the interaction of Campylobacter with chicken TLRs has not been studied. Here, we investigated the potential of four Campylobacter strains to activate human TLR1/2/6, TLR4, TLR5, and TLR9 and chicken TLR2t2/16, TLR4, TLR5, and TLR21. Live bacteria showed no or very limited potential to activate TLR2, TLR4, and TLR5 of both the human and chicken species, with minor but significant differences between Campylobacter strains. In contrast, lysed bacteria induced strong NF-kappaB activation through human TLR1/2/6 and TLR4 and chicken TLR2t2/16 and TLR4 but not via TLR5 of either species. Interestingly, C. jejuni induced TLR4-mediated beta interferon in human but not chicken cells. Furthermore, isolated chromosomal Campylobacter DNA was unable to activate human TLR9 in our system, whereas chicken TLR21 was activated by DNA from all of the campylobacters tested. Our data are the first comparison of TLR-induced immune responses in humans and chickens. The results suggest that differences in bacterial cell wall integrity and in TLR responses to Campylobacter LOS and/or DNA may contribute to the distinct clinical manifestation between the species.


Assuntos
Infecções por Campylobacter/imunologia , Infecções por Campylobacter/veterinária , Campylobacter/imunologia , Portador Sadio/imunologia , Portador Sadio/veterinária , Galinhas/imunologia , Receptores Toll-Like/imunologia , Animais , Bactérias , Campylobacter jejuni , DNA Bacteriano/imunologia , Humanos , Lipopolissacarídeos/imunologia
18.
J Immunol ; 181(6): 4354-62, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768894

RESUMO

During evolution, mammals have evolved a powerful innate immune response to LPS. Chickens are much more resistant to LPS-induced septic shock. Herein we report that chickens sense LPS via orthologs of mammalian TLR4 and myeloid differentiation protein-2 (MD-2) rather than the previously implicated chicken TLR2 isoform type 2 (chTLR2t2) receptor. Cloning and expression of recombinant chTLR4 and chMD-2 in HeLa 57A cells activated NF-kappaB at concentrations of LPS as low as 100 pg/ml. Differential pairing of chicken and mammalian TLR4 and MD-2 indicated that the protein interaction was species-specific in contrast to the formation of functional human and murine chimeric complexes. The chicken LPS receptor responded to a wide variety of LPS derivatives and to the synthetic lipid A compounds 406 and 506. The LPS specificity resembled the functionality of the murine rather than the human TLR4/MD-2 complex. Polymorphism in chTLR4 (Tyr(383)His and Gln(611)Arg) did not influence the LPS response. Interestingly, LPS consistently failed to activate the MyD88-independent induction of IFN-beta in chicken cells, in contrast to the TLR3 agonist poly(I:C) that yielded a potent IFN-beta response. These results suggest that chicken lack a functional LPS-specific TRAM-TRIF (TRIF-related adapter molecule/TIR-domain-containing adapter-inducing IFN-beta) signaling pathway, which may explain their aberrant response to LPS compared with the mammalian species.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Aviárias/química , Lipopolissacarídeos/imunologia , Antígeno 96 de Linfócito/química , Fator 88 de Diferenciação Mieloide/fisiologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/química , Receptores Toll-Like/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/fisiologia , Linhagem Celular , Galinhas , Células HeLa , Humanos , Antígeno 96 de Linfócito/genética , Antígeno 96 de Linfócito/fisiologia , Camundongos , Dados de Sequência Molecular , Especificidade da Espécie , Receptor 2 Toll-Like/química , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/fisiologia , Receptores Toll-Like/genética , Receptores Toll-Like/fisiologia
19.
Infect Immun ; 76(8): 3801-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18490457

RESUMO

Neisseria meningitidis LpxL1 lipopolysaccharide (LPS) bearing penta-acylated lipid A is considered a promising adjuvant candidate for inclusion in future N. meningitidis vaccines, as it elicits a markedly reduced endotoxic response in human macrophages relative to that in wild-type (hexa-acylated) LPS, while it is an equally effective adjuvant in mice. As dendritic cells (DC) and Toll-like receptors (TLR) are regarded as central mediators in the initiation of an immune response, here we evaluated the ability of LpxL1 LPS to mature and to activate human DC and examined its TLR4-/MD-2-activating properties. Unexpectedly, purified LpxL1 LPS displayed minimal human DC-stimulating properties compared to wild-type LPS. Although whole bacteria induced DC maturation and activation irrespective of their type of LPS, the LpxL1 mutant failed to activate the human recombinant TLR4/MD-2 complex expressed in HeLa cells. Similarly, purified LpxL1 LPS was unable to activate human TLR4/MD-2 and it even acted as an antagonist of wild-type LPS. Both wild-type and LpxL1 LPSs activated the murine TLR4/MD-2 complex, consistent with their abilities to induce maturation and activation of murine DC. Assays with cells transfected with different combinations of human and murine TLR4 and MD-2 indicated that TLR4 was a more-major determinant of the LPS response than MD-2. The species-specific activation of the TLR4/MD-2 complex by LpxL1 LPS may have an impact on the use of LpxL1 LPS as an adjuvant and the use of murine immunization models in human meningococcal vaccine development.


Assuntos
Lipídeo A/imunologia , Neisseria meningitidis/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Células Dendríticas/imunologia , Células HeLa , Humanos , Antígeno 96 de Linfócito/imunologia , Camundongos
20.
Mol Immunol ; 45(5): 1298-307, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17964652

RESUMO

Mammalian Toll-like receptor 5 (TLR5) senses flagellin of several bacterial species and activates the innate immune system. The avian TLR repertoire exhibits considerable functional diversity compared to mammalian TLRs and evidence of a functional TLR5 in the avian species is lacking. In the present study we cloned and successfully expressed chicken TLR5 (chTLR5) in HeLa cells, as indicated by laser confocal microscopy. Infection of chTLR5 transfected cells with Salmonella enterica serovar Enteritidis activated NF-kappaB in a dose- and flagellin-dependent fashion. Similar NF-kappaB activation was observed with recombinant bacterial flagellin. Targeted mutagenesis of the proline residue at position 737 in the chTLR5-TIR domain was detrimental to chTLR5 function, confirming that the observed effects were conferred via chTLR5 and the MyD88 signaling pathway. Comparison of human, mouse and chicken TLR5 activation by flagellin of S. enterica serovar Typhimurium revealed that chTLR5 consistently yielded stronger responses than human but not mouse TLR5. This species-specific reactivity was not observed with flagellin of serovar Enteritidis. The species-specific TLR5 response was nullified after targeted mutagenesis of a single amino acid (Q89A) in serovar Typhimurium flagellin, while L415A and N100A substitutions had no effect. These results show that chickens express a functional TLR5 albeit with different flagellin sensing qualities compared to human TLR5. The finding that single amino acid substitutions in bacterial flagellin can alter the species-specific TLR5 response may influence the host range and susceptibility of infection.


Assuntos
Flagelina/metabolismo , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/metabolismo , Animais , Sequência de Bases , Galinhas , Clonagem Molecular , Predisposição Genética para Doença , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , NF-kappa B/metabolismo , Ligação Proteica/genética , Salmonella enteritidis/imunologia , Especificidade da Espécie , Receptor 5 Toll-Like/imunologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...