Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Microbes Infect ; 26(3): 105274, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38081475

RESUMO

Leptospira interrogans are pathogenic bacteria responsible for leptospirosis, a worldwide zoonosis. All vertebrates can be infected, and some species like humans are susceptible to the disease whereas rodents such as mice are resistant and become asymptomatic renal carriers. Leptospires are stealth bacteria that are known to escape several immune recognition pathways and resist killing mechanisms. We recently published that leptospires may survive intracellularly in and exit macrophages, avoiding xenophagy, a pathogen-targeting form of autophagy. Interestingly, the latter is one of the antimicrobial mechanisms often highjacked by bacteria to evade the host immune response. In this study we explored whether leptospires subvert the key molecular players of autophagy to facilitate infection. We showed in macrophages that leptospires triggered a specific accumulation of autophagy-adaptor p62 in puncta-like structures, without altering autophagic flux. We demonstrated that Leptospira-induced p62 accumulation is a passive mechanism depending on the leptospiral virulence factor LPS signaling via TLR4/TLR2. p62 is a central pleiotropic protein, also mediating cell stress and death, via the translocation of transcription factors. We demonstrated that Leptospira-driven accumulation of p62 induced the translocation of transcription factor NRF2, a key player in the anti-oxidant response. However, NRF2 translocation upon Leptospira infection did not result as expected in antioxydant response, but dampened the production of inflammatory mediators such as iNOS/NO, TNF and IL6. Overall, these findings highlight a novel passive bacterial mechanism linked to LPS and p62/NRF2 signaling that decreases inflammation and contributes to the stealthiness of leptospires.


Assuntos
Leptospira , Leptospirose , Humanos , Camundongos , Animais , Lipopolissacarídeos , Fator 2 Relacionado a NF-E2/metabolismo , Regulação para Cima , Macrófagos/metabolismo , Inflamação , Autofagia
2.
J Immunol ; 210(4): 459-474, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36602965

RESUMO

Leptospira interrogans are bacteria that can infect all vertebrates and are responsible for leptospirosis, a neglected zoonosis. Some hosts, such as humans, are susceptible to the disease, whereas mice are resistant and get chronically colonized. Although leptospires escape recognition by some immune receptors, they activate the NOD-like receptor pyrin 3-inflammasome and trigger IL-1ß secretion. Classically, IL-1ß secretion is associated with lytic inflammatory cell death called pyroptosis, resulting from cytosolic LPS binding to inflammatory caspases, such as caspase 11. Interestingly, we showed that L. interrogans and Leptospira biflexa do not trigger cell death in either murine, human, hamster, or bovine macrophages, escaping both pyroptosis and apoptosis. We showed, in murine cells, that the mild IL-1ß secretion induced by leptospires occurred through nonlytic caspase 8-dependent gasdermin D pore formation and not through activation of caspase 11/noncanonical inflammasome. Strikingly, we demonstrated a potent antagonistic effect of pathogenic L. interrogans and their atypical LPS on spontaneous and Escherichia coli LPS-induced cell death. Indeed, LPS of L. interrogans efficiently prevents caspase 11 dimerization and subsequent massive gasdermin D cleavage. Finally, we showed that pyroptosis escape by leptospires prevents massive IL-1ß release, and we consistently found no major role of IL-1R in controlling experimental leptospirosis in vivo. Overall, to our knowledge, our findings described a novel mechanism by which leptospires dampen inflammation, thus potentially contributing to their stealthiness.


Assuntos
Leptospira interrogans , Leptospirose , Animais , Bovinos , Cricetinae , Humanos , Camundongos , Caspases/metabolismo , Gasderminas , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Leptospira interrogans/metabolismo , Leptospirose/metabolismo , Leptospirose/microbiologia , Lipopolissacarídeos , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Morte Celular
3.
Proc Natl Acad Sci U S A ; 117(49): 31398-31409, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229580

RESUMO

Toxin-antitoxin systems are found in many bacterial chromosomes and plasmids with roles ranging from plasmid stabilization to biofilm formation and persistence. In these systems, the expression/activity of the toxin is counteracted by an antitoxin, which, in type I systems, is an antisense RNA. While the regulatory mechanisms of these systems are mostly well defined, the toxins' biological activity and expression conditions are less understood. Here, these questions were investigated for a type I toxin-antitoxin system (AapA1-IsoA1) expressed from the chromosome of the human pathogen Helicobacter pylori We show that expression of the AapA1 toxin in H. pylori causes growth arrest associated with rapid morphological transformation from spiral-shaped bacteria to round coccoid cells. Coccoids are observed in patients and during in vitro growth as a response to different stress conditions. The AapA1 toxin, first molecular effector of coccoids to be identified, targets H. pylori inner membrane without disrupting it, as visualized by cryoelectron microscopy. The peptidoglycan composition of coccoids is modified with respect to spiral bacteria. No major changes in membrane potential or adenosine 5'-triphosphate (ATP) concentration result from AapA1 expression, suggesting coccoid viability. Single-cell live microscopy tracking the shape conversion suggests a possible association of this process with cell elongation/division interference. Oxidative stress induces coccoid formation and is associated with repression of the antitoxin promoter and enhanced processing of its transcript, leading to an imbalance in favor of AapA1 toxin expression. Our data support the hypothesis of viable coccoids with characteristics of dormant bacteria that might be important in H. pylori infections refractory to treatment.


Assuntos
Helicobacter pylori/citologia , Helicobacter pylori/efeitos dos fármacos , Peptídeos/farmacologia , Sistemas Toxina-Antitoxina , Trifosfato de Adenosina/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Helicobacter pylori/ultraestrutura , Peróxido de Hidrogênio/toxicidade , Espaço Intracelular/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Peptidoglicano/metabolismo
4.
Sci Rep ; 7(1): 9909, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855660

RESUMO

Helicobacter pullorum is an avian bacterium that causes gastroenteritis, intestinal bowel and hepatobiliary diseases in humans. Although H. pullorum has been shown to activate the mammalian innate immunity with release of nitric oxide (NO), the proteins that afford protection against NO and reactive nitrogen species (RNS) remain unknown. Here several protein candidates of H. pullorum, namely a truncated (TrHb) and a single domain haemoglobin (SdHb), and three peroxiredoxin-like proteins (Prx1, Prx2 and Prx3) were investigated. We report that the two haemoglobin genes are induced by RNS, and that SdHb confers resistance to nitrosative stress both in vitro and in macrophages. For peroxiredoxins, the prx2 and prx3 expression is enhanced by peroxynitrite and hydrogen peroxide, respectively. Mutation of prx1 does not alter the resistance to these stresses, while the single ∆prx2 and double ∆prx1∆prx2 mutants have decreased viability. To corroborate the physiological data, the biochemical analysis of the five recombinant enzymes was done, namely by stopped-flow spectrophotometry. It is shown that H. pullorum SdHb reacts with NO much more quickly than TrHb, and that the three Prxs react promptly with peroxynitrite, Prx3 displaying the highest reactivity. Altogether, the results unveil SdHb and Prx3 as major protective systems of H. pullorum against nitrosative stress.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter/patogenicidade , Estresse Nitrosativo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Helicobacter/genética , Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Humanos , Intestinos/microbiologia , Intestinos/patologia , Fígado/microbiologia , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Viabilidade Microbiana/genética , Mutação , Óxido Nítrico/metabolismo , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Virulência
5.
FEBS J ; 281(19): 4495-505, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25132107

RESUMO

Helicobacter pylori is a pathogen that infects the gastric mucosa of a large percentage of the human population worldwide, and predisposes to peptic ulceration and gastric cancer. Persistent colonization of humans by H. pylori triggers an inflammatory response that leads to the production of reactive nitrogen species. However, the mechanisms of H. pylori defence against nitrosative stress remain largely unknown. In this study, we show that the NADH-flavin oxidoreductase FrxA of H. pylori, besides metabolizing nitrofurans and metronidazole, has S-nitrosoglutathione reductase activity. In agreement with this, inactivation of the FrxA-encoding gene resulted in a strain that was more sensitive to S-nitrosoglutathione. FrxA was also shown to contribute to the proliferation of H. pylori in macrophages, which are key phagocytic cells of the mammalian innate immune system. Moreover, FrxA was shown to support the virulence of the pathogen upon mouse infection. Altogether, we provide evidence for a new function of FrxA that contributes to the successful chronic colonization ability that characterizes H. pylori.


Assuntos
Aldeído Oxirredutases/química , Proteínas de Bactérias/química , Infecções por Helicobacter/microbiologia , Helicobacter pylori/enzimologia , Aldeído Oxirredutases/fisiologia , Animais , Proteínas de Bactérias/fisiologia , Sequência de Bases , Indução Enzimática , Feminino , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/patogenicidade , Cinética , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana , Nitrocompostos/química , Oxirredução , S-Nitrosoglutationa/química , S-Nitrosoglutationa/farmacologia , Estresse Fisiológico , Ativação Transcricional
6.
Infect Immun ; 82(7): 2881-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24778119

RESUMO

Helicobacter pylori infection systematically causes chronic gastric inflammation that can persist asymptomatically or evolve toward more severe gastroduodenal pathologies, such as ulcer, mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric cancer. The cag pathogenicity island (cag PAI) of H. pylori allows translocation of the virulence protein CagA and fragments of peptidoglycan into host cells, thereby inducing production of chemokines, cytokines, and antimicrobial peptides. In order to characterize the inflammatory response to H. pylori, a new experimental protocol for isolating and culturing primary human gastric epithelial cells was established using pieces of stomach from patients who had undergone sleeve gastrectomy. Isolated cells expressed markers indicating that they were mucin-secreting epithelial cells. Challenge of primary epithelial cells with H. pylori B128 underscored early dose-dependent induction of expression of mRNAs of the inflammatory mediators CXCL1 to -3, CXCL5, CXCL8, CCL20, BD2, and tumor necrosis factor alpha (TNF-α). In AGS cells, significant expression of only CXCL5 and CXCL8 was observed following infection, suggesting that these cells were less reactive than primary epithelial cells. Infection of both cellular models with H. pylori B128ΔcagM, a cag PAI mutant, resulted in weak inflammatory-mediator mRNA induction. At 24 h after infection of primary epithelial cells with H. pylori, inflammatory-mediator production was largely due to cag PAI substrate-independent virulence factors. Thus, H. pylori cag PAI substrate appears to be involved in eliciting an epithelial response during the early phases of infection. Afterwards, other virulence factors of the bacterium take over in development of the inflammatory response. Using a relevant cellular model, this study provides new information on the modulation of inflammation during H. pylori infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Quimiocinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Helicobacter pylori/imunologia , Estômago/citologia , Antígenos de Bactérias/imunologia , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Bactérias/imunologia , Células Cultivadas , Quimiocinas/genética , Ilhas Genômicas , Helicobacter pylori/metabolismo , Humanos
7.
J Infect Dis ; 209(7): 1045-54, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24265438

RESUMO

Mycobacteria produce an unusual, glycolylated form of muramyl dipeptide (MDP) that is more potent and efficacious at inducing NOD2-mediated host responses. We tested the importance of this modified form of MDP in Mycobacterium tuberculosis by disrupting the gene, namH, responsible for this modification. In vitro, the namH mutant did not produce N-glycolylated muropeptides, but there was no alteration in colony morphology, growth kinetics, cellular morphology, or mycolic acid profile. Ex vivo, the namH mutant survived and replicated normally in murine and human macrophages, yet induced diminished production of tumor necrosis factor α. In vivo, namH disruption did not affect the bacterial burden during infection of C57BL/6 mice or cellular recruitment to the lungs but modestly prolonged survival after infection in Rag1(-/-) mice. These results indicate that the modified MDP is an important contributor to the unusual immunogenicity of mycobacteria but has a limited role in the pathogenesis of M. tuberculosis infection.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/imunologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Peptidoglicano/imunologia , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animais , Carga Bacteriana , Células Cultivadas , Modelos Animais de Doenças , Deleção de Genes , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/enzimologia , Peptidoglicano/química , Processamento de Proteína Pós-Traducional , Análise de Sobrevida , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Virulência
8.
J Bacteriol ; 195(23): 5250-61, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24056102

RESUMO

The intracellular bacterial pathogen Listeria monocytogenes activates a robust type I interferon response upon infection. This response is partially dependent on the multidrug resistance (MDR) transporter MdrM and relies on cyclic-di-AMP (c-di-AMP) secretion, yet the functions of MdrM and cyclic-di-AMP that lead to this response are unknown. Here we report that it is not MdrM alone but a cohort of MDR transporters that together contribute to type I interferon induction during infection. In a search for a physiological function of these transporters, we revealed that they play a role in cell wall stress responses. A mutant with deletion of four transporter genes (ΔmdrMTAC) was found to be sensitive to sublethal concentrations of vancomycin due to an inability to produce and shed peptidoglycan under this stress. Remarkably, c-di-AMP is involved in this phenotype, as overexpression of the c-di-AMP phosphodiesterase (PdeA) resulted in increased susceptibility of the ΔmdrMTAC mutant to vancomycin, whereas overexpression of the c-di-AMP diadenylate cyclase (DacA) reduced susceptibility to this drug. These observations suggest a physiological association between c-di-AMP and the MDR transporters and support the model that MDR transporters mediate c-di-AMP secretion to regulate peptidoglycan synthesis in response to cell wall stress.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Parede Celular/fisiologia , Fosfatos de Dinucleosídeos/metabolismo , Interferon Tipo I/metabolismo , Listeria monocytogenes/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Antibacterianos/farmacologia , Fosfatos de Dinucleosídeos/genética , Farmacorresistência Bacteriana Múltipla , Regulação Bacteriana da Expressão Gênica/fisiologia , Interferon Tipo I/genética , Interferon beta/genética , Interferon beta/metabolismo , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/genética , Estresse Fisiológico
9.
PLoS One ; 8(4): e60657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23577140

RESUMO

H. pylori colonizes half of the world's population leading to gastritis, ulcers and gastric cancer. H. pylori strains resistant to antibiotics are increasing which raises the need for alternative therapeutic approaches. Docosahexaenoic acid (DHA) has been shown to decrease H. pylori growth and its associated-inflammation through mechanisms poorly characterized. We aimed to explore DHA action on H. pylori-mediated inflammation and adhesion to gastric epithelial cells (AGS) and also to identify bacterial structures affected by DHA. H. pylori growth and metabolism was assessed in liquid cultures. Bacterial adhesion to AGS cells was visualized by transmission electron microscopy and quantified by an Enzyme Linked Immunosorbent Assay. Inflammatory proteins were assessed by immunoblotting in infected AGS cells, previously treated with DHA. Bacterial total and outer membrane protein composition was analyzed by 2-dimensional gel electrophoresis. Concentrations of 100 µM of DHA decreased H. pylori growth, whereas concentrations higher than 250 µM irreversibly inhibited bacteria survival. DHA reduced ATP production and adhesion to AGS cells. AGS cells infected with DHA pre-treated H. pylori showed a 3-fold reduction in Interleukin-8 (IL-8) production and a decrease of COX2 and iNOS. 2D electrophoresis analysis revealed that DHA changed the expression of H. pylori outer membrane proteins associated with stress response and metabolism and modified bacterial lipopolysaccharide phenotype. As conclusions our results show that DHA anti-H. pylori effects are associated with changes of bacteria morphology and metabolism, and with alteration of outer membrane proteins composition, that ultimately reduce the adhesion of bacteria and the burden of H. pylori-related inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Células Epiteliais/microbiologia , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/fisiologia , Estômago/citologia , Anti-Inflamatórios/uso terapêutico , Aderência Bacteriana/efeitos dos fármacos , Proteínas da Membrana Bacteriana Externa/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Helicobacter pylori/citologia , Helicobacter pylori/crescimento & desenvolvimento , Inflamação/tratamento farmacológico , Inflamação/microbiologia
10.
J Infect Dis ; 207(10): 1535-43, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23402825

RESUMO

BACKGROUND: Yersinia pestis (the plague bacillus) and its ancestor, Yersinia pseudotuberculosis (which causes self-limited bowel disease), encode putative homologues of the periplasmic lysozyme inhibitor Ivy and the membrane-bound lysozyme inhibitor MliC. The involvement of both inhibitors in virulence remains subject to debate. METHODS: Mutants lacking ivy and/or mliC were generated. We evaluated the mutants' ability to counter lysozyme, grow in serum, and/or counter leukocytes; to produce disease in wild-type, neutropenic, or lysozyme-deficient rodents; and to induce host inflammation. RESULTS: MliC was not required for lysozyme resistance and the development of plague. Deletion of ivy decreased Y. pestis' ability to counter lysozyme and polymorphonuclear neutrophils, but it did not affect the bacterium's ability to grow in serum or resist macrophages. Y. pestis lacking Ivy had attenuated virulence, unless animals were neutropenic or lysozyme deficient. The Ivy mutant induced inflammation to a degree similar to that of the parental strain. Last, Y. pseudotuberculosis did not require Ivy to counter lysozyme and for virulence. CONCLUSIONS: Ivy is required to counter lysozyme during infection, but its role as a virulence factor is species dependent. Our study also shows that a gene that is not necessary for the virulence of an ancestral bacterium may become essential in the emergence of a new pathogen.


Assuntos
Proteínas de Bactérias/genética , Imunidade Inata , Muramidase/antagonistas & inibidores , Fatores de Virulência/genética , Yersinia pestis/patogenicidade , Animais , Proteínas de Bactérias/metabolismo , Sangue/imunologia , Sangue/microbiologia , Bovinos , Linhagem Celular , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Evolução Molecular , Feminino , Deleção de Genes , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Muramidase/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Periplasma/química , Fagócitos/metabolismo , Fagócitos/microbiologia , Peste/imunologia , Peste/microbiologia , Peste/patologia , Ratos , Ratos Endogâmicos BN , Soroalbumina Bovina/química , Virulência , Fatores de Virulência/metabolismo , Yersinia pestis/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidade , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/microbiologia , Infecções por Yersinia pseudotuberculosis/patologia
11.
Immunity ; 37(6): 1076-90, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23219392

RESUMO

Ly6C(hi) monocytes seed the healthy intestinal lamina propria to give rise to resident CX(3)CR1(+) macrophages that contribute to the maintenance of gut homeostasis. Here we report on two alternative monocyte fates in the inflamed colon. We showed that CCR2 expression is essential to the recruitment of Ly6C(hi) monocytes to the inflamed gut to become the dominant mononuclear cell type in the lamina propria during settings of acute colitis. In the inflammatory microenvironment, monocytes upregulated TLR2 and NOD2, rendering them responsive to bacterial products to become proinflammatory effector cells. Ablation of Ly6C(hi) monocytes ameliorated acute gut inflammation. With time, monocytes differentiated into migratory antigen-presenting cells capable of priming naive T cells, thus acquiring hallmarks reminiscent of dendritic cells. Collectively, our results highlight cellular dynamics in the inflamed colon and the plasticity of Ly6C(hi) monocytes, marking them as potential targets for inflammatory bowel disease (IBD) therapy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos Ly/metabolismo , Movimento Celular/imunologia , Colite/imunologia , Monócitos/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Antígenos Ly/imunologia , Receptor 1 de Quimiocina CX3C , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Imunofenotipagem , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Proteína Adaptadora de Sinalização NOD2/imunologia , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptores CCR2/imunologia , Receptores CCR2/metabolismo , Receptores de Quimiocinas/metabolismo , Linfócitos T/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 2 Toll-Like/metabolismo
12.
Glycobiology ; 22(9): 1193-206, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22522599

RESUMO

Helicobacter pylori infects more than half of the world's population. Although most patients are asymptomatic, persistent infection may cause chronic gastritis and gastric cancer. Adhesion of the bacteria to the gastric mucosa is a necessary prerequisite for the pathogenesis of H. pylori-related diseases and is mediated by mucin O-glycans. In order to define which glycans may be implicated in the binding of the bacteria to the gastric mucosa in humans, we have characterized the exact pattern of glycosylation of gastric mucins. We have identified that the major component was always a core 2-based glycan carrying two blood group H antigens, whatever was the blood group of individuals. We have also demonstrated that around 80% of O-glycans carried blood group A, B or H antigens, suggesting that the variation of gastric mucin glycosylation between individuals is partly due to the blood group status. This study will help better understanding the role of O-glycans in the physiology and homeostasis of gastric mucosa. Overall, the results reported here give us the necessary background information to begin studies to determine whether individuals who express certain carbohydrate epitopes on specific mucins are predisposed to certain gastric diseases.


Assuntos
Sistema ABO de Grupos Sanguíneos/química , Mucinas Gástricas/química , Mucosa Gástrica/química , Helicobacter pylori/química , Antígenos do Grupo Sanguíneo de Lewis/química , Polissacarídeos/química , Sistema ABO de Grupos Sanguíneos/metabolismo , Adolescente , Adulto , Sítios de Ligação , Sequência de Carboidratos , Cromatografia Líquida de Alta Pressão , Suscetibilidade a Doenças , Feminino , Mucinas Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Glicosilação , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Dados de Sequência Molecular , Polissacarídeos/metabolismo , Ligação Proteica
13.
J Immunol ; 188(6): 2805-14, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22323544

RESUMO

Leptospira interrogans is responsible for a zoonotic disease known to induce severe kidney dysfunction and inflammation. In this work, we demonstrate that L. interrogans induces NLRP3 inflammasome-dependent secretion of IL-1ß through the alteration of potassium transport in bone marrow-derived macrophages. Lysosome destabilization also contributed to the IL-1ß production upon stimulation with live, but not dead, bacteria. Using bone marrow-derived macrophages from various TLRs and nucleotide-binding oligomerization domain-deficient mice, we further determined that IL-1ß production was dependent on TLR2 and TLR4, suggesting a participation of the leptospiral LPS to this process. Hypokaliemia in leptospirosis has been linked to the presence of glycolipoprotein, a cell wall component of L. interrogans that is known to inhibit the expression and functions of the Na/K-ATPase pump. We show in this study that glycolipoprotein activates the inflammasome and synergizes with leptospiral LPS to produce IL-1ß, mimicking the effect of whole bacteria. These results were confirmed in vivo, as wild-type mice expressed more IL-1ß in the kidney than TLR2/4-deficient mice 3 d postinfection with L. interrogans. Collectively, these findings provide the first characterization, to our knowledge, of bacteria-induced activation of the NLRP3 inflammasome through the downregulation of a specific host potassium transporter.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Leptospirose/metabolismo , Macrófagos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Proteínas de Bactérias/imunologia , Western Blotting , Proteínas de Transporte/imunologia , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Feminino , Inflamassomos/imunologia , Interleucina-1beta/biossíntese , Interleucina-1beta/imunologia , Leptospira/imunologia , Leptospira/metabolismo , Leptospirose/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Cell Mol Life Sci ; 68(22): 3661-73, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21984599

RESUMO

Pattern recognition receptors are somatically encoded and participate in the innate immune responses of a host to microbes. It is increasingly acknowledged that these receptors play a central role both in beneficial and pathogenic interactions with microbes. In particular, these receptors participate actively in shaping the gut environment to establish a fruitful life-long relationship between a host and its microbiota. Commensal bacteria engage Toll-like receptors (TLRs) and nucleotide oligomerization domain (NOD)-like receptors (NLRs) to induce specific responses by intestinal epithelial cells such as production of antimicrobial products or of a functional mucus layer. Furthermore, a complex crosstalk between intestinal epithelial cells and the immune system is initiated leading to a mature gut-associated lymphoid tissue to secrete IgA. Impairment in NLR and TLR functionality in epithelial cells is strongly associated with chronic inflammatory diseases such as Crohn's disease, cancer, and with control of the commensal microbiota creating a more favorable environment for the emergence of new infections.


Assuntos
Células Epiteliais/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Receptores de Reconhecimento de Padrão/imunologia , Receptores Toll-Like/imunologia , Animais , Autofagia/imunologia , Células Epiteliais/citologia , Homeostase/imunologia , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Metagenoma , Proteínas Adaptadoras de Sinalização NOD/genética , Proteínas Adaptadoras de Sinalização NOD/imunologia , Receptores de Reconhecimento de Padrão/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética
15.
J Infect Dis ; 204(5): 731-40, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21844299

RESUMO

Microbial pathogens have evolved mechanisms to overcome immune responses and successfully infect their host. Here, we studied how Listeria monocytogenes evades immune detection by peptidoglycan (PGN) modification. By analyzing L. monocytogenes muropeptides, we detected O-acetylated muramic acid residues. We identified an O-acetyltransferase gene, oatA, in the L. monocytogenes genome sequence. Comparison of PGN from parental and isogenic oatA mutant strains showed that the O-acetyltransferase OatA O-acetylates Listeria PGN. We also found that PGN O-acetylation confers resistance to different types of antimicrobial compounds targeting bacterial cell wall such as lysozyme, ß-lactam antibiotics, and bacteriocins and that O-acetylation is required for Listeria growth in macrophages. Moreover, oatA mutant virulence is drastically affected in mice following intravenous or oral inoculation. In addition, the oatA mutant induced early secretion of proinflammatory cytokines and chemokines in vivo. These results suggest an important role for OatA in limiting innate immune responses and promoting bacterial survival in the infected host.


Assuntos
Acetiltransferases/imunologia , Citocinas/metabolismo , Listeria monocytogenes/imunologia , Listeriose/imunologia , Peptidoglicano/imunologia , Fatores de Virulência/imunologia , Acetilação , Acetiltransferases/genética , Animais , Linhagem Celular , Feminino , Humanos , Imunidade Inata , Dose Letal Mediana , Listeria monocytogenes/genética , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Listeriose/genética , Fígado/metabolismo , Fígado/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Ácidos Murâmicos/metabolismo , Peptidoglicano/química , Baço/microbiologia , Células Th1/metabolismo , Células Th2/metabolismo , Fatores de Virulência/genética
16.
Cell Microbiol ; 12(5): 626-39, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20039881

RESUMO

The cytosolic innate immune molecule, NOD1, recognizes peptidoglycan (PG) delivered to epithelial cells via the Helicobacter pylori cag pathogenicity island (cagPAI), and has been implicated in host defence against cagPAI(+)H. pylori bacteria. To further clarify the role of NOD1 in host defence, we investigated NOD1-dependent regulation of human beta-defensins (DEFBs) in two epithelial cell lines. Our findings identify that NOD1 activation, via either cagPAI(+) bacteria or internalized PG, was required for DEFB4 and DEFB103 expression in HEK293 cells. To investigate cell type-specific induction of DEFB4 and DEFB103, we generated stable NOD1'knockdown' (KD) and control AGS cells. Reporter gene assay and RT-PCR analyses revealed that only DEFB4 was induced in an NOD1-/cagPAI-dependent fashion in AGS cells. Moreover, culture supernatants from AGS control, but not AGS NOD1 KD cells, stimulated with cagPAI(+)H. pylori, significantly reduced H. pylori bacterial numbers. siRNA studies confirmed that human beta-defensin 2 (hBD-2), but not hBD-3, contributes to the antimicrobial activity of AGS cell supernatants against H. pylori. This study demonstrates, for the first time, the involvement of NOD1 and hBD-2 in direct killing of H. pylori bacteria by epithelial cells and confirms the importance of NOD1 in host defence mechanisms against cagPAI(+)H. pylori infection.


Assuntos
Células Epiteliais/imunologia , Helicobacter pylori/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , beta-Defensinas/imunologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Viabilidade Microbiana , Proteína Adaptadora de Sinalização NOD1/genética , Peptidoglicano/imunologia , beta-Defensinas/biossíntese
17.
Proc Natl Acad Sci U S A ; 104(3): 997-1002, 2007 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-17215377

RESUMO

Listeria monocytogenes is a human intracellular pathogen that is able to survive in the gastrointestinal environment and replicate in macrophages, thus bypassing the early innate immune defenses. Peptidoglycan (PG) is an essential component of the bacterial cell wall readily exposed to the host and, thus, an important target for the innate immune system. Characterization of the PG from L. monocytogenes demonstrated deacetylation of N-acetylglucosamine residues. We identified a PG N-deacetylase gene, pgdA, in L. monocytogenes genome sequence. Inactivation of pgdA revealed the key role of this PG modification in bacterial virulence because the mutant was extremely sensitive to the bacteriolytic activity of lysozyme, and growth was severely impaired after oral and i.v. inoculations. Within macrophage vacuoles, the mutant was rapidly destroyed and induced a massive IFN-beta response in a TLR2 and Nod1-dependent manner. Together, these results reveal that PG N-deacetylation is a highly efficient mechanism used by Listeria to evade innate host defenses. The presence of deacetylase genes in other pathogenic bacteria indicates that PG N-deacetylation could be a general mechanism used by bacteria to evade the host innate immune system.


Assuntos
Infecções por Bactérias Gram-Positivas/imunologia , Sistema Imunitário/imunologia , Imunidade Inata/imunologia , Listeria/imunologia , Peptidoglicano/imunologia , Peptidoglicano/metabolismo , Acetilação , Amidoidrolases/genética , Amidoidrolases/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/patologia , Humanos , Interleucina-6/biossíntese , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Muramidase/metabolismo , Mutação/genética , Peptidoglicano/química , Peptidoglicano/classificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
18.
PLoS Pathog ; 2(9): e97, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17002496

RESUMO

The human gastric pathogen Helicobacter pylori is responsible for peptic ulcers and neoplasia. Both in vitro and in the human stomach it can be found in two forms, the bacillary and coccoid forms. The molecular mechanisms of the morphological transition between these two forms and the role of coccoids remain largely unknown. The peptidoglycan (PG) layer is a major determinant of bacterial cell shape, and therefore we studied H. pylori PG structure during the morphological transition. The transition correlated with an accumulation of the N-acetyl-D-glucosaminyl-beta(1,4)-N-acetylmuramyl-L-Ala-D-Glu (GM-dipeptide) motif. We investigated the molecular mechanisms responsible for the GM-dipeptide motif accumulation, and studied the role of various putative PG hydrolases in this process. Interestingly, a mutant strain with a mutation in the amiA gene, encoding a putative PG hydrolase, was impaired in accumulating the GM-dipeptide motif and transforming into coccoids. We investigated the role of the morphological transition and the PG modification in the biology of H. pylori. PG modification and transformation of H. pylori was accompanied by an escape from detection by human Nod1 and the absence of NF-kappaB activation in epithelial cells. Accordingly, coccoids were unable to induce IL-8 secretion by AGS gastric epithelial cells. amiA is, to our knowledge, the first genetic determinant discovered to be required for this morphological transition into the coccoid forms, and therefore contributes to modulation of the host response and participates in the chronicity of H. pylori infection.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Transporte/fisiologia , Helicobacter pylori/fisiologia , Lipoproteínas/fisiologia , Motivos de Aminoácidos , Amoxicilina/farmacologia , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Linhagem Celular , Forma Celular/fisiologia , Parede Celular/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Teste de Complementação Genética , Helicobacter pylori/citologia , Helicobacter pylori/genética , Helicobacter pylori/imunologia , Humanos , Lipoproteínas/genética , Mutação , Peptidoglicano/genética , Peptidoglicano/metabolismo
19.
J Biol Chem ; 280(44): 36714-8, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16150702

RESUMO

The mammalian innate immune system recognizes pathogen-associated molecular patterns through pathogen recognition receptors. Nod1 has been described recently as a cytosolic receptor that detects specifically diaminopimelate-containing muropeptides from Gram-negative bacteria peptidoglycan. In the present study we investigated the potential role of Nod1 in the innate immune response against the opportunistic pathogen Pseudomonas aeruginosa. We demonstrate that Nod1 detects the P. aeruginosa peptidoglycan leading to NF-kappaB activation and that this activity is diminished in epithelial cells expressing a dominant-negative Nod1 construct or in mouse embryonic fibroblasts from Nod1 knock-out mice infected with P. aeruginosa. Finally, we demonstrate that the cytokine secretion kinetics and bacterial killing are altered in Nod1-deficient cells infected with P. aeruginosa in the early stages of infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose , Peptidoglicano/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/patogenicidade , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Citocinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Genes Dominantes , Humanos , Imunidade Inata , Cinética , Camundongos , Camundongos Knockout/microbiologia , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1 , Infecções por Pseudomonas/metabolismo
20.
J Biol Chem ; 280(43): 35859-67, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16115863

RESUMO

NOD2/CARD15 is the first characterized susceptibility gene in Crohn disease. The Nod2 1007fs (Nod2fs) frameshift mutation is the most prevalent in Crohn disease patients. Muramyl dipeptide from bacterial peptidoglycan is the minimal motif detected by Nod2 but not by Nod2fs. Here we investigated the response of human peripheral blood mononuclear cells (PBMCs) from Crohn disease patients not only to muramyl dipeptide but also to several other muramyl peptides. Most unexpectedly, we observed that patients homozygous for the Nod2fs mutation were totally unresponsive to MurNAc-L-Ala-D-Glu-meso-diaminopimelic acid (DAP) (M-Tri(DAP)), the specific agonist of Nod1, and to Gram-negative bacterial peptidoglycan. In contrast, PBMCs from a patient homozygous for the Nod2 R702W mutation, also associated with Crohn disease, displayed normal response to Gram-negative bacterial peptidoglycan. In addition, the blockage of the Nod1/M-Tri(DAP) pathway could be partially overcome by co-stimulation with the Toll-like receptors agonists lipoteichoic acid or lipopolysaccharide. Investigation into the mechanism of this finding revealed that Nod2fs did not act as a dominant-negative molecule for the Nod1/M-Tri(DAP) pathway, implying that the blockage is dependent upon the expression or activity of other factors. We demonstrated that PBMCs from Nod2fs patients express high levels of the peptidoglycan recognition protein S, a secreted protein known to interact with muramyl peptides. We proposed that through a scavenger function, peptidoglycan recognition protein S may dampen M-Tri(DAP)-dependent responses in Nod2fs patients. Together, our results identified a cross-talk between the Nod1 and Nod2 pathways and suggested that down-regulation of Nod1/M-Tri(DAP) pathway may be associated with Crohn disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mutação da Fase de Leitura , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptidoglicano/química , Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos , Animais , Linhagem Celular , Doença de Crohn/genética , Regulação para Baixo , Genótipo , Homozigoto , Humanos , Hidrólise , Interleucina-10/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Leucócitos Mononucleares/citologia , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Químicos , Mutação , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1 , Proteína Adaptadora de Sinalização NOD2 , Peptídeos/química , Peptidoglicano/metabolismo , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácidos Teicoicos/metabolismo , Receptores Toll-Like/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA