Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 10(2): e1004647, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25679217

RESUMO

Signaling via pattern recognition receptors (PRRs) expressed on professional antigen presenting cells, such as dendritic cells (DCs), is crucial to the fate of engulfed microbes. Among the many PRRs expressed by DCs are Toll-like receptors (TLRs) and C-type lectins such as DC-SIGN. DC-SIGN is targeted by several major human pathogens for immune-evasion, although its role in intracellular routing of pathogens to autophagosomes is poorly understood. Here we examined the role of DC-SIGN and TLRs in evasion of autophagy and survival of Porphyromonas gingivalis in human monocyte-derived DCs (MoDCs). We employed a panel of P. gingivalis isogenic fimbriae deficient strains with defined defects in Mfa-1 fimbriae, a DC-SIGN ligand, and FimA fimbriae, a TLR2 agonist. Our results show that DC-SIGN dependent uptake of Mfa1+P. gingivalis strains by MoDCs resulted in lower intracellular killing and higher intracellular content of P. gingivalis. Moreover, Mfa1+P. gingivalis was mostly contained within single membrane vesicles, where it survived intracellularly. Survival was decreased by activation of TLR2 and/or autophagy. Mfa1+P. gingivalis strain did not induce significant levels of Rab5, LC3-II, and LAMP1. In contrast, P. gingivalis uptake through a DC-SIGN independent manner was associated with early endosomal routing through Rab5, increased LC3-II and LAMP-1, as well as the formation of double membrane intracellular phagophores, a characteristic feature of autophagy. These results suggest that selective engagement of DC-SIGN by Mfa-1+P. gingivalis promotes evasion of antibacterial autophagy and lysosome fusion, resulting in intracellular persistence in myeloid DCs; however TLR2 activation can overcome autophagy evasion and pathogen persistence in DCs.


Assuntos
Autofagia/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Células Mieloides/metabolismo , Porphyromonas gingivalis/metabolismo , Receptores de Superfície Celular/metabolismo , Receptor 2 Toll-Like/metabolismo , Dendritos/ultraestrutura , Células Dendríticas/imunologia , Células Dendríticas/ultraestrutura , Fímbrias Bacterianas , Humanos , Espaço Intracelular/imunologia , Espaço Intracelular/metabolismo , Monócitos/imunologia , Monócitos/ultraestrutura , Células Mieloides/imunologia , Receptor 2 Toll-Like/imunologia
2.
BMC Genomics ; 15: 1176, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25540039

RESUMO

BACKGROUND: Atherosclerosis is a progressive disease characterized by inflammation and accumulation of lipids in vascular tissue. Porphyromonas gingivalis (Pg) and Chlamydia pneumoniae (Cp) are associated with inflammatory atherosclerosis in humans. Similar to endogenous mediators arising from excessive dietary lipids, these Gram-negative pathogens are pro-atherogenic in animal models, although the specific inflammatory/atherogenic pathways induced by these stimuli are not well defined. In this study, we identified gene expression profiles that characterize P. gingivalis, C. pneumoniae, and Western diet (WD) at acute and chronic time points in aortas of Apolipoprotein E (ApoE-/-) mice. RESULTS: At the chronic time point, we observed that P. gingivalis was associated with a high number of unique differentially expressed genes compared to C. pneumoniae or WD. For the top 500 differentially expressed genes unique to each group, we observed a high percentage (76%) that exhibited decreased expression in P. gingivalis-treated mice in contrast to a high percentage (96%) that exhibited increased expression in WD mice. C. pneumoniae treatment resulted in approximately equal numbers of genes that exhibited increased and decreased expression. Gene Set Enrichment Analysis (GSEA) revealed distinct stimuli-associated phenotypes, including decreased expression of mitochondrion, glucose metabolism, and PPAR pathways in response to P. gingivalis but increased expression of mitochondrion, lipid metabolism, carbohydrate and amino acid metabolism, and PPAR pathways in response to C. pneumoniae; WD was associated with increased expression of immune and inflammatory pathways. DAVID analysis of gene clusters identified by two-way ANOVA at acute and chronic time points revealed a set of core genes that exhibited altered expression during the natural progression of atherosclerosis in ApoE-/- mice; these changes were enhanced in P. gingivalis-treated mice but attenuated in C. pneumoniae-treated mice. Notable differences in the expression of genes associated with unstable plaques were also observed among the three pro-atherogenic stimuli. CONCLUSIONS: Despite the common outcome of P. gingivalis, C. pneumoniae, and WD on the induction of vascular inflammation and atherosclerosis, distinct gene signatures and pathways unique to each pro-atherogenic stimulus were identified. Our results suggest that pathogen exposure results in dysregulated cellular responses that may impact plaque progression and regression pathways.


Assuntos
Aorta/metabolismo , Apolipoproteínas E/deficiência , Chlamydophila pneumoniae/fisiologia , Dieta Ocidental/efeitos adversos , Perfilação da Expressão Gênica , Porphyromonas gingivalis/fisiologia , Animais , Aorta/patologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Família Multigênica/genética , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/microbiologia , Placa Aterosclerótica/patologia
3.
J Vis Exp ; (90): e51556, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25146644

RESUMO

Chronic inflammation is a major driver of pathological tissue damage and a unifying characteristic of many chronic diseases in humans including neoplastic, autoimmune, and chronic inflammatory diseases. Emerging evidence implicates pathogen-induced chronic inflammation in the development and progression of chronic diseases with a wide variety of clinical manifestations. Due to the complex and multifactorial etiology of chronic disease, designing experiments for proof of causality and the establishment of mechanistic links is nearly impossible in humans. An advantage of using animal models is that both genetic and environmental factors that may influence the course of a particular disease can be controlled. Thus, designing relevant animal models of infection represents a key step in identifying host and pathogen specific mechanisms that contribute to chronic inflammation. Here we describe a mouse model of pathogen-induced chronic inflammation at local and systemic sites following infection with the oral pathogen Porphyromonas gingivalis, a bacterium closely associated with human periodontal disease. Oral infection of specific-pathogen free mice induces a local inflammatory response resulting in destruction of tooth supporting alveolar bone, a hallmark of periodontal disease. In an established mouse model of atherosclerosis, infection with P. gingivalis accelerates inflammatory plaque deposition within the aortic sinus and innominate artery, accompanied by activation of the vascular endothelium, an increased immune cell infiltrate, and elevated expression of inflammatory mediators within lesions. We detail methodologies for the assessment of inflammation at local and systemic sites. The use of transgenic mice and defined bacterial mutants makes this model particularly suitable for identifying both host and microbial factors involved in the initiation, progression, and outcome of disease. Additionally, the model can be used to screen for novel therapeutic strategies, including vaccination and pharmacological intervention.


Assuntos
Infecções por Bacteroidaceae/microbiologia , Modelos Animais de Doenças , Inflamação/microbiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Animais , Masculino , Camundongos , Camundongos Transgênicos , Boca/microbiologia , Porphyromonas gingivalis/genética
4.
PLoS Pathog ; 10(7): e1004215, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25010102

RESUMO

Several successful pathogens have evolved mechanisms to evade host defense, resulting in the establishment of persistent and chronic infections. One such pathogen, Porphyromonas gingivalis, induces chronic low-grade inflammation associated with local inflammatory bone loss and systemic inflammation manifested as atherosclerosis. P. gingivalis expresses an atypical lipopolysaccharide (LPS) structure containing heterogeneous lipid A species, that exhibit Toll-like receptor-4 (TLR4) agonist or antagonist activity, or are non-activating at TLR4. In this study, we utilized a series of P. gingivalis lipid A mutants to demonstrate that antagonistic lipid A structures enable the pathogen to evade TLR4-mediated bactericidal activity in macrophages resulting in systemic inflammation. Production of antagonistic lipid A was associated with the induction of low levels of TLR4-dependent proinflammatory mediators, failed activation of the inflammasome and increased bacterial survival in macrophages. Oral infection of ApoE(-/-) mice with the P. gingivalis strain expressing antagonistic lipid A resulted in vascular inflammation, macrophage accumulation and atherosclerosis progression. In contrast, a P. gingivalis strain producing exclusively agonistic lipid A augmented levels of proinflammatory mediators and activated the inflammasome in a caspase-11-dependent manner, resulting in host cell lysis and decreased bacterial survival. ApoE(-/-) mice infected with this strain exhibited diminished vascular inflammation, macrophage accumulation, and atherosclerosis progression. Notably, the ability of P. gingivalis to induce local inflammatory bone loss was independent of lipid A expression, indicative of distinct mechanisms for induction of local versus systemic inflammation by this pathogen. Collectively, our results point to a pivotal role for activation of the non-canonical inflammasome in P. gingivalis infection and demonstrate that P. gingivalis evades immune detection at TLR4 facilitating chronic inflammation in the vasculature. These studies support the emerging concept that pathogen-mediated chronic inflammatory disorders result from specific pathogen-mediated evasion strategies resulting in low-grade chronic inflammation.


Assuntos
Infecções por Bacteroidaceae/imunologia , Lipídeo A/imunologia , Porphyromonas gingivalis/imunologia , Vasculite/imunologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/imunologia , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/microbiologia , Aterosclerose/patologia , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/patologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Osteoporose/genética , Osteoporose/imunologia , Osteoporose/microbiologia , Osteoporose/patologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Vasculite/genética , Vasculite/microbiologia , Vasculite/patologia
5.
Bioorg Med Chem Lett ; 20(24): 7516-20, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21036041

RESUMO

The synthesis and in vitro binding affinity for a novel series of potent androgen receptor modulators is described. One of the more potent compounds (17, RAD35010) was further characterized in vivo where it restored levator ani weight in castrated male rats to near sham level while having no significant effect on prostate weight.


Assuntos
Carbazóis/química , Receptores Androgênicos/química , Administração Oral , Animais , Carbazóis/síntese química , Carbazóis/farmacologia , Masculino , Próstata/efeitos dos fármacos , Ratos , Receptores Androgênicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...