Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
J Biol Chem ; 296: 100460, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33639171

RESUMO

Bacterial survival during lethal heat stress relies on the cellular ability to reactivate aggregated proteins. This activity is typically executed by the canonical 70-kDa heat shock protein (Hsp70)-ClpB bichaperone disaggregase, which is most widespread in bacteria. The ClpB disaggregase is a member of the ATPase associated with diverse cellular activities protein family and exhibits an ATP-driven threading activity. Substrate binding and stimulation of ATP hydrolysis depends on the Hsp70 partner, which initiates the disaggregation reaction. Recently elevated heat resistance in gamma-proteobacterial species was shown to be mediated by the ATPase associated with diverse cellular activities protein ClpG as an alternative disaggregase. Pseudomonas aeruginosa ClpG functions autonomously and does not cooperate with Hsp70 for substrate binding, enhanced ATPase activity, and disaggregation. With the underlying molecular basis largely unknown, the fundamental differences in ClpG- and ClpB-dependent disaggregation are reflected by the presence of sequence alterations and additional ClpG-specific domains. By analyzing the effects of mutants lacking ClpG-specific domains and harboring mutations in conserved motifs implicated in ATP hydrolysis and substrate threading, we show that the N-terminal, ClpG-specific N1 domain generally mediates protein aggregate binding as the molecular basis of autonomous disaggregation activity. Peptide substrate binding strongly stimulates ClpG ATPase activity by overriding repression by the N-terminal N1 and N2 domains. High ATPase activity requires two functional nucleotide binding domains and drives substrate threading which ultimately extracts polypeptides from the aggregate. ClpG ATPase and disaggregation activity is thereby directly controlled by substrate availability.


Assuntos
Antígenos de Bactérias/metabolismo , Endopeptidase Clp/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Choque Térmico/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Antígenos de Bactérias/fisiologia , Endopeptidase Clp/fisiologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/fisiologia , Agregados Proteicos , Ligação Proteica , Domínios Proteicos/genética
2.
Cells ; 10(1)2020 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-33375694

RESUMO

Gastric cancer constitutes one of the most prevalent malignancies in both sexes; it is currently the fourth major cause of cancer-related deaths worldwide. The pathogenesis of gastric cancer is associated with the interaction between genetic and environmental factors, among which infection by Helicobacter pylori (H. pylori) is of major importance. The invasion, survival, colonization, and stimulation of further inflammation within the gastric mucosa are possible due to several evasive mechanisms induced by the virulence factors that are expressed by the bacterium. The knowledge concerning the mechanisms of H. pylori pathogenicity is crucial to ameliorate eradication strategies preventing the possible induction of carcinogenesis. This review highlights the current state of knowledge and the most recent findings regarding H. pylori virulence factors and their relationship with gastric premalignant lesions and further carcinogenesis.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Fatores de Virulência/fisiologia , Animais , Carcinogênese/patologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Humanos
3.
J Immunol ; 203(5): 1218-1229, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31375544

RESUMO

Mycobacterium tuberculosis employs defense mechanisms to protect itself from reactive oxygen species (ROS)-mediated cytotoxicity inside macrophages. In the current study, we found that a secretory protein of M. tuberculosis PPE2 disrupted the assembly of NADPH oxidase complex. PPE2 inhibited NADPH oxidase-mediated ROS generation in RAW 264.7 macrophages and peritoneal macrophages from BALB/c mice. PPE2 interacted with the cytosolic subunit of NADPH oxidase, p67phox, and prevented translocation of p67phox and p47phox to the membrane, resulting in decreased NADPH oxidase activity. Trp236 residue present in the SH3-like domain of PPE2 was found to be critical for its interaction with p67phox Trp236Ala mutant of PPE2 did not interact with p67phox and thereby did not affect ROS generation. M. tuberculosis expressing PPE2 and PPE2-null mutants complemented with PPE2 survived better than PPE2-null mutants in infected RAW 264.7 macrophages. Altogether, this study suggests that PPE2 inhibits NADPH oxidase-mediated ROS production to favor M. tuberculosis survival in macrophages. The findings that M. tuberculosis PPE2 protein is involved in the modulation of oxidative response in macrophages will help us in improving our knowledge of host-pathogen interactions and the application of better therapeutics against tuberculosis.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Fosfoproteínas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Antígenos de Bactérias/química , Proteínas de Bactérias/química , Interações Hospedeiro-Patógeno , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Acetato de Tetradecanoilforbol/farmacologia , Domínios de Homologia de src
4.
FASEB J ; 33(10): 10808-10818, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31262188

RESUMO

Colonization of the oropharynx is the initial step in Group A Streptococcus (GAS) pharyngeal infection. We have previously reported that the highly virulent M1T1 GAS clone attaches to oral epithelial cells via M1 protein interaction with blood group antigen carbohydrate structures. Here, we have identified that colonization of human oral epithelial cells by GAS serotypes M3 and M12 is mediated by human blood group antigens [ABO(H)] and Lewis (Le) antigen expression. Removal of linkage-specific fucose, galactose, N-acetylgalactosamine, and sialic acid modulated GAS colonization, dependent on host ABO(H) blood group and Le expression profile. Furthermore, N-linked glycans from human salivary glycoproteins, when released and purified, were potent inhibitors of M1, M3, and M12 GAS colonization ex vivo. These data highlight the important role played by human protein glycosylation patterns in GAS attachment to oral epithelial cell surfaces.-De Oliveira, D. M. P., Everest-Dass, A., Hartley-Tassell, L., Day, C. J., Indraratna, A., Brouwer, S., Cleary, A., Kautto, L., Gorman, J., Packer, N. H., Jennings, M. P., Walker, M. J., Sanderson-Smith, M. L. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells.


Assuntos
Interações entre Hospedeiro e Microrganismos/fisiologia , Polissacarídeos/metabolismo , Streptococcus pyogenes/patogenicidade , Antígenos de Bactérias/fisiologia , Aderência Bacteriana/imunologia , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Antígenos de Grupos Sanguíneos/química , Proteínas de Transporte/fisiologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Glicosilação , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Técnicas In Vitro , Polissacarídeos/química , Polissacarídeos/imunologia , Ligação Proteica , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/metabolismo , Infecções Estreptocócicas/etiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/fisiologia , Virulência/fisiologia
5.
Cell Mol Immunol ; 16(4): 380-391, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29572547

RESUMO

The intracellular pathogen Mycobacterium tuberculosis (Mtb) can survive in the host and cause disease by interfering with a variety of cellular functions. The mammalian cell entry 2 (mce2) operon of Mtb has been shown to contribute to tuberculosis pathogenicity. However, little is known about the regulatory roles of Mtb Mce2 family proteins towards host cellular functions. Here we show that the Mce2 family protein Mce2E suppressed the macrophage innate immune response and promoted epithelial cell proliferation. Mce2E inhibited activation of the extracellular signal-regulated kinase (ERK) and Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) signaling pathways in a non-canonical D motif (a MAPK-docking motif)-dependent manner, leading to reduced expression of TNF and IL-6 in macrophages. Furthermore, Mce2E promoted proliferation of human lung epithelium-derived lung adenoma A549 cells by inhibiting K48-linked polyubiquitination of eEF1A1 in a ß strand region-dependent manner. In summary, Mce2E is a novel multifunctional Mtb virulence factor that regulates host cellular functions in a niche-dependent manner. Our data suggest a potential novel target for TB therapy.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Células Epiteliais/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Motivos de Aminoácidos/genética , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Citocinas/metabolismo , Feminino , Humanos , Imunidade Inata , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Mycobacterium tuberculosis/patogenicidade , Fator 1 de Elongação de Peptídeos/metabolismo , Transplante Heterólogo , Ubiquitinação/genética , Ubiquitinação/imunologia , Fatores de Virulência/metabolismo
6.
EMBO J ; 37(13)2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29724755

RESUMO

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Assuntos
Antígenos de Bactérias/fisiologia , Antígenos CD/fisiologia , Proteínas de Bactérias/fisiologia , Moléculas de Adesão Celular/fisiologia , Helicobacter pylori/fisiologia , Proteínas Oncogênicas/fisiologia , Antígenos CD/química , Proteínas de Bactérias/química , Moléculas de Adesão Celular/química , Células HEK293 , Humanos , Mutagênese , Multimerização Proteica , Transporte Proteico
7.
J Immunol ; 198(12): 4772-4780, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28507027

RESUMO

Tuberculosis caused by Mycobacterium tuberculosis continues to pose a serious global health threat. The attenuated Mycobacterium bovis bacillus Calmette-Guérin, as the only licensed vaccine, has limited protective efficacy against TB. The development of more effective antituberculosis vaccines is urgent and demands for further identification and understanding of M. tuberculosis Ags. MPT83 (Rv2873), a secreted mycobacterial lipoprotein, has been applied into subunit vaccine development and shown protective effects against M. tuberculosis infection in animals; however, the understanding of the underlying mechanism is limited. In present study, we systematically studied the effect of MPT83 on macrophage apoptosis by constructing Mycobacterium smegmatis strain overexpressing MPT83 (MS_MPT83) and purifying rMPT83 protein. We found that MPT83 induced apoptosis in both human and mouse macrophages. MPT83 induced cyclooxygenase-2 (COX-2) expression at both the transcriptional and protein levels in macrophages, whereas silencing or inhibiting COX-2 blocked rMPT83-induced apoptosis or the enhanced apoptotic response to MS_MPT83 in comparison with M. smegmatis transfected with pMV261 vector (MS_Vec), indicating that COX-2 is required for MPT83-induced apoptosis. Additionally, tlr2 deficiency led to significant reduction of COX-2 expression, accompanied by less apoptosis in macrophages stimulated with rMPT83 or infected with MS_MPT83. Moreover, the activation of p38 accounted for MPT83-induced COX-2 expression. Finally, lower bacteria burdens in the lungs and spleens and enhanced survival were observed in mice i.v. infected with MS_MPT83 compared with MS_Vec. Taken together, our results established a proapoptotic effect of MPT83 and identified the TLR2/p38/COX-2 axis in MPT83-induced macrophage apoptosis.


Assuntos
Antígenos de Bactérias/fisiologia , Apoptose , Proteínas de Bactérias/fisiologia , Ciclo-Oxigenase 2/metabolismo , Macrófagos/microbiologia , Macrófagos/fisiologia , Proteínas de Membrana/fisiologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/isolamento & purificação , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Ciclo-Oxigenase 2/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Mycobacterium smegmatis/imunologia , Fosforilação , Receptor 2 Toll-Like/imunologia
9.
Apoptosis ; 22(4): 502-509, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27987050

RESUMO

Mycobacterium tuberculosis PE/PPE family proteins, named after the presence of conserved PE (Pro-Glu) and PPE (Pro-Pro-Glu) domains at N-terminal, are prevalent in M. tuberculosis genome. The function of most PE/PPE family proteins remains elusive. To characterize the function of PE_PGRS18, the encoding gene was heterologously expressed in M. smegmatis, a nonpathogenic mycobacterium. The recombinant PE_PGRS18 is cell wall associated. M. smegmatis PE_PGRS18 recombinant showed differential response to stresses and altered the production of host cytokines IL-6, IL-1ß, IL-12p40 and IL-10, as well as enhanced survival within macrophages largely via attenuating the apoptosis of macrophages. In summary, the study firstly unveiled the role of PE_PGRS18 in physiology and pathogenesis of mycobacterium.


Assuntos
Antígenos de Bactérias/fisiologia , Apoptose/fisiologia , Proteínas de Bactérias/fisiologia , Citocinas/biossíntese , Macrófagos/microbiologia , Proteínas de Membrana/fisiologia , Mycobacterium smegmatis/fisiologia , Mycobacterium tuberculosis/fisiologia , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Genes Bacterianos , Humanos , Macrófagos/metabolismo , Proteínas de Membrana/genética , Camundongos , Mycobacterium smegmatis/citologia , Mycobacterium tuberculosis/citologia , Mycobacterium tuberculosis/genética , Óxido Nítrico/metabolismo , Domínios Proteicos , Proteínas Recombinantes de Fusão/metabolismo , Células THP-1 , Regulação para Cima
10.
Oncol Rep ; 36(6): 3087-3094, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27748858

RESUMO

Infection with Helicobacter pylori is the strongest risk factor for the development of chronic gastritis, gastric ulcer and gastric carcinoma. The majority of the H. pylori-infected population remains asymptomatic, and only 1% of individuals may progress to gastric cancer. The clinical outcomes caused by H. pylori infection are considered to be associated with bacterial virulence, genetic polymorphism of hosts as well as environmental factors. Most H. pylori strains possess a cytotoxin-associated gene (cag) pathogenicity island (cagPAI), encoding a 120-140 kDa CagA protein, which is the most important bacterial oncoprotein. CagA is translocated into host cells via T4SS system and affects the expression of signaling proteins in a phosphorylation-dependent and independent manner. Thus, this review summarizes the results of relevant studies, discusses the pathogenesis of CagA-mediated gastric cancer.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Infecções por Helicobacter/complicações , Helicobacter pylori/fisiologia , Neoplasias Gástricas/microbiologia , Animais , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Transporte Proteico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
12.
Oncotarget ; 7(34): 55290-55301, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27421133

RESUMO

Early detection of gastric cancers saves lives, but remains a diagnostic challenge. In this study, we aimed to identify cell-surface biomarkers of early gastric cancer. We hypothesized that a subset of plasma membrane proteins induced by the Helicobacter pylori oncoprotein CagA will be retained in early gastric cancers through non-oncogene addiction. An inducible system for expression of CagA was used to identify differentially upregulated membrane protein transcripts in vitro. The top hits were then analyzed in gene expression datasets comparing transcriptome of gastric cancer with normal tissue, to focus on markers retained in cancer. Among the transcripts enriched upon CagA induction in vitro, a significant elevation of CEACAM6 was noted in gene expression datasets of gastric cancer. We used quantitative digital immunohistochemistry to measure CEACAM6 protein levels in tissue microarrays of gastric cancer. We demonstrate an increase in CEACAM6 in early gastric cancers, when compared to matched normal tissue, with an AUC of 0.83 for diagnostic validity. Finally, we show that a fluorescently conjugated CEACAM6 antibody binds avidly to freshly resected gastric cancer xenograft samples and can be detected by endoscopy in real time. Together, these results suggest that CEACAM6 upregulation is a cell surface response to H. pylori CagA, and is retained in early gastric cancers. They highlight a novel link between CEACAM6 expression and CagA in gastric cancer, and suggest CEACAM6 to be a promising biomarker to aid with the fluorescent endoscopic diagnosis of early neoplastic lesions in the stomach.


Assuntos
Antígenos de Bactérias/fisiologia , Antígenos CD/análise , Proteínas de Bactérias/fisiologia , Biomarcadores Tumorais/análise , Moléculas de Adesão Celular/análise , Neoplasias Gástricas/diagnóstico , Animais , Imunofluorescência , Proteínas Ligadas por GPI/análise , Infecções por Helicobacter/metabolismo , Humanos , Camundongos , Regulação para Cima
13.
Cell Microbiol ; 18(10): 1471-85, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27062290

RESUMO

The ability of macrophages to eradicate intracellular pathogens is normally greatly enhanced by IFNγ, a cytokine produced mainly after onset of adaptive immunity. However, adaptive immunity is unable to provide sterilizing immunity against mycobacteria, suggesting that mycobacteria have evolved virulence strategies to inhibit the bactericidal effect of IFNγ-signalling in macrophages. Still, the host-pathogen interactions and cellular mechanisms responsible for this feature have remained elusive. We demonstrate that the ESX-1 type VII secretion systems of Mycobacterium tuberculosis and Mycobacterium marinum exploit type I IFN-signalling to promote an IL-12(low) /IL-10(high) regulatory macrophage phenotype characterized by secretion of IL-10, IL-27 and IL-6. This mechanism had no impact on intracellular growth in the absence of IFNγ but suppressed IFNγ-mediated autophagy and growth restriction, indicating that the regulatory phenotype extends to function. The IFNγ-refractory phenotype was partly mediated by IL-27-signalling, establishing functional relevance for this downstream cytokine. These findings identify a novel macrophage-modulating function for the ESX-1 secretion system that may contribute to suppress the efficacy of adaptive immunity and provide mechanistic insight into the antagonistic cross talk between type I IFNs and IFNγ in mycobacterial infection.


Assuntos
Antígenos de Bactérias/fisiologia , Autofagia/imunologia , Proteínas de Bactérias/fisiologia , Macrófagos/metabolismo , Mycobacterium tuberculosis/fisiologia , Tuberculose/microbiologia , Animais , Sistemas de Secreção Bacterianos , Células Cultivadas , Interações Hospedeiro-Patógeno , Imunidade Inata , Interferon Tipo I/fisiologia , Interferon gama/fisiologia , Interleucinas/metabolismo , Interleucinas/normas , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais , Tuberculose/imunologia
14.
FEBS J ; 282(21): 4114-29, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26260636

RESUMO

Early secreted antigenic target 6 kDa (ESAT-6) and culture filtrate protein 10 kDa (CFP-10) are complex proteins secreted by Mycobacterium tuberculosis that play a major role in the pathogenesis of tuberculosis. However, studies focusing on the biological functions of ESAT-6 led to discordant results and the role of ESAT-6 remains controversial. In the present study, we aim to address a potential explanation for this discrepancy and to highlight the physiological impact of two conformational states of ESAT-6. Analysis of a recombinant form of ESAT-6 by native gel electrophoresis, size exclusion chromatography and CD spectroscopy revealed that ESAT-6 forms dimers/multimers with higher molecular weight, which disappeared under the action of the detergent amidosulfobetaine-14 (ASB), giving rise to another conformational state of the protein. NMR has further indicated that ASB-treated versus nontreated ESAT-6 adopted distinct structural forms but with no well defined tertiary structure. However, protein-protein docking analysis favored a dimeric state of ESAT-6. Interestingly, the two preparations presented opposing effects on mycobacterial infectivity, as well as macrophage survival, interferon-γ secretion and membrane pore formation. Thereafter, we generated a recombinant form of the physiological heterodimer ESAT-6/CFP-10 that ASB was also able to dissociate and which showed functions similar to those of ESAT-6 dimers/multimers. Our data suggest that, in the absence of CFP-10, the hydrophobic regions of the ESAT-6 can form dimers/multimers, mimicking the ESAT-6/CFP-10 heterodimer, whereas their dissociation generates a protein presenting entirely different activities. Overall, the present study clarifies the intriguing divergences between reports that could be attributed to the ESAT-6 oligomeric state and sheds light on its importance for a better comprehension of the physiopathology of tuberculosis.


Assuntos
Antígenos de Bactérias/química , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Mycobacterium tuberculosis/patogenicidade , Betaína/análogos & derivados , Morte Celular , Detergentes , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/biossíntese , Modelos Moleculares , Mycobacterium tuberculosis/fisiologia , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Tuberculose/etiologia , Virulência/fisiologia , Fatores de Virulência/química , Fatores de Virulência/fisiologia
15.
PLoS One ; 10(3): e0121463, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822999

RESUMO

Pathogenicity of the human pathogen Helicobacter pylori relies on its capacity to adapt to a hostile environment and to escape the host response. Although there have been great advances in our understanding of the bacterial cytoskeleton, major gaps remain in our knowledge of its contribution to virulence. In this study we have explored the influence of coiled coil rich proteins (Ccrp) cytoskeletal elements on pathogenicity factors of H. pylori. Deletion of any of the ccrp resulted in a strongly decreased activity of the main pathogenicity factor urease. We further investigated their role using in vitro co-culture experiments with the human gastric adenocarcinoma cell line AGS modeling H. pylori - host cell interactions. Intriguingly, host cell showed only a weak "scattering/hummingbird" phenotype, in which host cells are transformed from a uniform polygonal shape into a severely elongated state characterized by the formation of needle-like projections, after co-incubation with any ccrp deletion mutant. Furthermore, co-incubation with the ccrp59 mutant resulted in reduced type IV secretion system associated activities, e.g. IL-8 production and CagA translocation/phosphorylation. Thus, in addition to their role in maintaining the helical cell shape of H. pylori Ccrp proteins influence many cellular processes and are thereby crucial for the virulence of this human pathogen.


Assuntos
Proteínas de Bactérias/fisiologia , Helicobacter pylori/patogenicidade , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/genética , Linhagem Celular , Genes Bacterianos , Infecções por Helicobacter/etiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Interleucina-8/biossíntese , Mutação , Fenótipo , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/fisiologia , Urease/metabolismo , Virulência/genética , Virulência/fisiologia , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
16.
Gut ; 64(7): 1040-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25080447

RESUMO

OBJECTIVE: Infection with Helicobacter pylori is the strongest known risk factor for adenocarcinoma of the stomach. Tumorigenic transformation of gastric epithelium induced by H. pylori is a highly complex process driven by an active interplay between bacterial virulence and host factors, many aspects of which remain obscure. In this work, we investigated the degradation of p53 tumour suppressor induced by H. pylori. DESIGN: Expression of p53 protein in gastric biopsies was assessed by immunohistochemistry. Gastric cells were co-cultured with H. pylori strains isolated from high-gastric risk and low-gastric risk areas and assessed for expression of p53, p14ARF and cytotoxin-associated gene A (CagA) by immunoblotting. siRNA was used to inhibit activities of ARF-BP1 and Human Double Minute 2 (HDM2) proteins. RESULTS: Our analysis demonstrated that H. pylori strains expressing high levels of CagA virulence factor and associated with a higher gastric cancer risk more strongly suppress p53 compared with low-risk strains in vivo and in vitro. We found that degradation of p53 induced by bacterial CagA protein is mediated by host HDM2 and ARF-BP1 E3 ubiquitin ligases, while the p14ARF protein counteracts H. pylori-induced signalling. CONCLUSIONS: Our results provide novel evidence that tumorigenicity associated with H. pylori infection is linked to inhibition of p53 protein by CagA. We propose a model in which CagA-induced degradation of p53 protein is determined by a relative level of p14ARF. In cells in which p14ARF levels were decreased due to hypermethylation or deletion of the p14ARF gene, H. pylori efficiently degraded p53, whereas p53 is protected in cells expressing high levels of p14ARF.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Neoplasias Gástricas/microbiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Antígenos de Bactérias/classificação , Proteínas de Bactérias/classificação , Linhagem Celular Tumoral , Epitélio/metabolismo , Mucosa Gástrica/microbiologia , Humanos , Imuno-Histoquímica , Neoplasias Gástricas/fisiopatologia
17.
Nat Commun ; 5: 4423, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25055241

RESUMO

Cytotoxin-associated gene A (CagA) is an oncoprotein and a major virulence factor of H. pylori. CagA is delivered into gastric epithelial cells via a type IV secretion system and causes cellular transformation. The loss of epithelial adhesion that accompanies the epithelial-mesenchymal transition (EMT) is a hallmark of gastric cancer. Although CagA is a causal factor in gastric cancer, the link between CagA and the associated EMT has not been elucidated. Here, we show that CagA induces the EMT by stabilizing Snail, a transcriptional repressor of E-cadherin expression. Mechanistically we show that CagA binds GSK-3 in a manner similar to Axin and causes it to shift to an insoluble fraction, resulting in reduced GSK-3 activity. We also find that the level of Snail protein is increased in H. pylori infected epithelium in clinical samples. These results suggest that H. pylori CagA acts as a pathogenic scaffold protein that induces a Snail-mediated EMT via the depletion of GSK-3.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Regulação para Baixo/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Helicobacter pylori/fisiologia , Fatores de Transcrição/fisiologia , Biópsia , Carcinogênese/metabolismo , Carcinogênese/patologia , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mucosa Gástrica/metabolismo , Gastrite/metabolismo , Gastrite/patologia , Humanos , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Estômago/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
18.
Mem Inst Oswaldo Cruz ; 109(4): 414-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25075780

RESUMO

While the influence of water in Helicobacter pylori culturability and membrane integrity has been extensively studied, there are little data concerning the effect of this environment on virulence properties. Therefore, we studied the culturability of water-exposed H. pylori and determined whether there was any relation with the bacterium's ability to adhere, produce functional components of pathogenicity and induce inflammation and alterations in apoptosis in an experimental model of human gastric epithelial cells. H. pylori partially retained the ability to adhere to epithelial cells even after complete loss of culturability. However, the microorganism is no longer effective in eliciting in vitro host cell inflammation and apoptosis, possibly due to the non-functionality of the cag type IV secretion system. These H. pylori-induced host cell responses, which are lost along with culturability, are known to increase epithelial cell turnover and, consequently, could have a deleterious effect on the initial H. pylori colonisation process. The fact that adhesion is maintained by H. pylori to the detriment of other factors involved in later infection stages appears to point to a modulation of the physiology of the pathogen after water exposure and might provide the microorganism with the necessary means to, at least transiently, colonise the human stomach.


Assuntos
Aderência Bacteriana/fisiologia , Células Epiteliais/microbiologia , Helicobacter pylori/patogenicidade , Água , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Sistemas de Secreção Bacterianos , Mucosa Gástrica/citologia , Helicobacter pylori/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Humanos , Virulência/fisiologia
19.
Mem. Inst. Oswaldo Cruz ; 109(4): 414-419, 03/07/2014. graf
Artigo em Inglês | LILACS | ID: lil-716305

RESUMO

While the influence of water in Helicobacter pylori culturability and membrane integrity has been extensively studied, there are little data concerning the effect of this environment on virulence properties. Therefore, we studied the culturability of water-exposed H. pylori and determined whether there was any relation with the bacterium’s ability to adhere, produce functional components of pathogenicity and induce inflammation and alterations in apoptosis in an experimental model of human gastric epithelial cells. H. pylori partially retained the ability to adhere to epithelial cells even after complete loss of culturability. However, the microorganism is no longer effective in eliciting in vitro host cell inflammation and apoptosis, possibly due to the non-functionality of the cag type IV secretion system. These H. pylori-induced host cell responses, which are lost along with culturability, are known to increase epithelial cell turnover and, consequently, could have a deleterious effect on the initial H. pylori colonisation process. The fact that adhesion is maintained by H. pylori to the detriment of other factors involved in later infection stages appears to point to a modulation of the physiology of the pathogen after water exposure and might provide the microorganism with the necessary means to, at least transiently, colonise the human stomach.


Assuntos
Humanos , Aderência Bacteriana/fisiologia , Células Epiteliais/microbiologia , Helicobacter pylori/patogenicidade , Água , Antígenos de Bactérias/fisiologia , Sistemas de Secreção Bacterianos , Proteínas de Bactérias/fisiologia , Mucosa Gástrica/citologia , Interações Hospedeiro-Patógeno , Helicobacter pylori/crescimento & desenvolvimento , Virulência/fisiologia
20.
PLoS One ; 9(5): e96427, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24797940

RESUMO

Our previous work has demonstrated the roles played by L-type Voltage Gated Calcium Channels (VGCC) in regulating Mycobacterium tuberculosis (M. tb) survival and pathogenesis. Here we decipher mechanisms and pathways engaged by the pathogen to regulate VGCC expression in macrophages. We show that M. tb and its antigen Rv3416 use phospho-CREB (pCREB), Reactive Oxygen Species (ROS), Protein Kinase C (PKC) and Mitogen Activated Protein Kinase (MAPK) to modulate VGCC expression in macrophages. siRNA mediated knockdown of MyD88, IRAK1, IRAK2 or TRAF6 significantly inhibited antigen mediated VGCC expression. Inhibiting Protein Kinase C (PKC) or MEK-ERK1/2 further increased VGCC expression. Interestingly, inhibiting intracellular calcium release upregulated antigen mediated VGCC expression, while inhibiting extracellular calcium influx had no significant effect. siRNA mediated knockdown of transcription factors c-Jun, SOX5 and CREB significantly inhibited Rv3416 mediated VGCC expression. A dynamic reciprocal cross-regulation between ROS and pCREB was observed that in turn governed VGCC expression with ROS playing a limiting role in the process. Further dissection of the mechanisms such as the interplay between ROS and pCREB would improve our understanding of the regulation of VGCC expression during M. tb infection.


Assuntos
Canais de Cálcio/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Mycobacterium tuberculosis/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Antígenos de Bactérias/fisiologia , Linhagem Celular , Humanos , Macrófagos/metabolismo , Redes e Vias Metabólicas/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Mycobacterium tuberculosis/patogenicidade , Proteína Quinase C/metabolismo , Proteína Quinase C/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA