Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Pathog ; 19(4): e1011284, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37023213

RESUMO

Porphyromonas gingivalis is a Gram-negative anaerobic bacterium that thrives in the inflamed environment of the gingival crevice, and is strongly associated with periodontal disease. The host response to P. gingivalis requires TLR2, however P. gingivalis benefits from TLR2-driven signaling via activation of PI3K. We studied TLR2 protein-protein interactions induced in response to P. gingivalis, and identified an interaction between TLR2 and the cytoskeletal protein vinculin (VCL), confirmed using a split-ubiquitin system. Computational modeling predicted critical TLR2 residues governing the physical association with VCL, and mutagenesis of interface residues W684 and F719, abrogated the TLR2-VCL interaction. In macrophages, VCL knock-down led to increased cytokine production, and enhanced PI3K signaling in response to P. gingivalis infection, effects that correlated with increased intracellular bacterial survival. Mechanistically, VCL suppressed TLR2 activation of PI3K by associating with its substrate PIP2. P. gingivalis induction of TLR2-VCL led to PIP2 release from VCL, enabling PI3K activation via TLR2. These results highlight the complexity of TLR signaling, and the importance of discovering protein-protein interactions that contribute to the outcome of infection.


Assuntos
Porphyromonas gingivalis , Receptor 2 Toll-Like , Porphyromonas gingivalis/genética , Receptor 2 Toll-Like/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Evasão da Resposta Imune , Vinculina/metabolismo , Composição de Bases , Filogenia , RNA Ribossômico 16S , Análise de Sequência de DNA
2.
Gut ; 73(5): 770-786, 2024 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-38233197

RESUMO

OBJECTIVE: Epidemiological studies highlight an association between pancreatic ductal adenocarcinoma (PDAC) and oral carriage of the anaerobic bacterium Porphyromonas gingivalis, a species highly linked to periodontal disease. We analysed the potential for P. gingivalis to promote pancreatic cancer development in an animal model and probed underlying mechanisms. DESIGN: We tracked P. gingivalis bacterial translocation from the oral cavity to the pancreas following administration to mice. To dissect the role of P. gingivalis in PDAC development, we administered bacteria to a genetically engineered mouse PDAC model consisting of inducible acinar cell expression of mutant Kras (Kras +/LSL-G12D; Ptf1a-CreER, iKC mice). These mice were used to study the cooperative effects of Kras mutation and P. gingivalis on the progression of pancreatic intraepithelial neoplasia (PanIN) to PDAC. The direct effects of P. gingivalis on acinar cells and PDAC cell lines were studied in vitro. RESULTS: P. gingivalis migrated from the oral cavity to the pancreas in mice and can be detected in human PanIN lesions. Repetitive P. gingivalis administration to wild-type mice induced pancreatic acinar-to-ductal metaplasia (ADM), and altered the composition of the intrapancreatic microbiome. In iKC mice, P. gingivalis accelerated PanIN to PDAC progression. In vitro, P. gingivalis infection induced acinar cell ADM markers SOX9 and CK19, and intracellular bacteria protected PDAC cells from reactive oxygen species-mediated cell death resulting from nutrient stress. CONCLUSION: Taken together, our findings demonstrate a causal role for P. gingivalis in pancreatic cancer development in mice.


Assuntos
Carcinoma in Situ , Carcinoma Ductal Pancreático , Microbiota , Neoplasias Pancreáticas , Lesões Pré-Cancerosas , Camundongos , Humanos , Animais , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Composição de Bases , Lesões Pré-Cancerosas/patologia , Filogenia , RNA Ribossômico 16S/metabolismo , Análise de Sequência de DNA , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Carcinoma in Situ/genética , Células Acinares/patologia , Bactérias/genética
3.
J Periodontal Res ; 56(3): 535-546, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33559894

RESUMO

OBJECTIVE: Periodontitis is one the most common chronic inflammatory conditions, resulting in destruction of tooth-supporting tissues and leading to tooth loss. Porphyromonas gingivalis activates host macrophages to secrete pro-inflammatory cytokines and elicit tissue damage, in part by inducing NF-kappa-B transactivation. Since NFκB transactivation is negatively regulated by the Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase enzyme Sirt1, we sought to assess if RAW264.7 macrophages exposed to P. gingivalis demonstrate impaired Sirt1 activity, to ultimately induce a pro-inflammatory response. METHODS: RAW264.7 macrophages were incubated with heat- killed P. gingivalis for 2, 4, 8, and 24 h. Stimulated RAW264.7 were assessed for TNFα expression via PCR, ELISA, and ChIP analysis. Following the activation of RAW264.7 macrophages, immunoblot analysis was executed to detect modifications in Sirt1 and the NFκB subunit RelA that is essential for NFκB transcriptional activity. RESULTS: TNFα expression was elevated 4 h after exposure to P. gingivalis. ChIP confirmed that RelA was enriched in the mouse TNFα promoter 4 h following stimulation, which correlated with the increased TNFα mRNA levels. Preceding TNFα expression, we detected Phosphoserine 536 and acetylated lysine 310 of RelA after 2 hours exposure with P. gingivalis. Moreover, reduced Sirt1 activity was associated with its cleavage in RAW264.7 protein extracts, after 2 hours of P. gingivalis exposure. Blocking TLR2/4 signaling prevented Sirt1 cleavage, loss of deacetylase activity, and TNFα secretion, while co-administering CA074Me (a cathepsin B inhibitor) with P. gingivalis reduced RelA promoter enrichment, resulting in impaired TNFα expression. CONCLUSIONS: Together, the results suggest that P. gingivalis induces TNFα expression, at least in part, by enhancing cleavage of Sirt1 via a TLR-dependent signaling circuit.


Assuntos
Periodontite , Porphyromonas gingivalis , Animais , Lipopolissacarídeos/farmacologia , Macrófagos , Camundongos , NF-kappa B , Sirtuína 1 , Fator de Necrose Tumoral alfa
4.
J Periodontal Res ; 54(4): 396-404, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30793777

RESUMO

AIM: To explore the role of keratinocyte myeloid differentiation primary response 88 (MyD88) expression in the adhesion of Porphyromonas gingivalis to the cells and its subsequent invasion and intracellular survival. MATERIALS AND METHODS: Primary mouse keratinocytes from wild-type (WT) or Myd88-/- mice were infected with P gingivalis alone or co-infected with Fusobacterium nucleatum. Bacterial adhesion and invasion were measured using fluorescent microscopy and flow cytometry, and intracellular survival in keratinocytes was quantified by an antibiotic protection assay. Keratinocyte expression of antimicrobial peptides was measured by real-time PCR. RESULTS: In the absence of MyD88, P gingivalis adherence, invasion, and intracellular survival were enhanced compared with WT keratinocytes. The presence of F nucleatum during infection increased the adhesion of P gingivalis to WT keratinocytes but reduced the adhesion to Myd88-/- keratinocytes. Fusobacterium nucleatum improved mildly the invasion and survival of P gingivalis in both cell types. Baseline expression of beta-defensin 2, 3, 4 and RegIII-γ was elevated in Myd88-/- keratinocytes compared to WT cells; however, following infection beta-defensin expression was strongly induced in WT cells but decreased dramatically in the MyD88 deficient cells. CONCLUSION: In the absence of MyD88 expression, P gingivalis adhesion to keratinocytes is improved, and invasion and intracellular survival are increased. Furthermore, keratinocyte infection by P gingivalis induces antimicrobial peptide expression in a MyD88-dependent manner. Thus, MyD88 plays a key role in the interaction between P gingivalis and keratinocytes.


Assuntos
Infecções por Bacteroidaceae/imunologia , Queratinócitos/microbiologia , Fator 88 de Diferenciação Mieloide/imunologia , Porphyromonas gingivalis , Animais , Peptídeos Catiônicos Antimicrobianos/imunologia , Aderência Bacteriana , Fusobacterium nucleatum , Queratinócitos/imunologia , Camundongos , Camundongos Knockout
5.
J Clin Periodontol ; 44(7): 739-748, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28453225

RESUMO

AIM: Peri-implantitis is a major health concern, with unclear pathogenesis, and with no accessible animal models. Our aim was to establish a mouse model for peri-implantitis and to investigate mediators of inflammation. MATERIALS AND METHODS: Mice were divided into implanted versus non-implanted groups. Implants were inserted immediately following the extraction of the upper first molar. Four weeks following implantation, implanted and non-implanted mice were challenged with either Porphyromonas gingivalis or vehicle (eight mice in each subgroup, 32 mice in total). Alveolar bone loss and expression of inflammatory mediators in the soft tissue were assessed 42 days following infection. RESULTS: Porphyromonas gingivalis infection induced greater bone loss around implants than around teeth. In non-infected animals, the presence of the implant correlated with elevated expression of Il-10, Foxp3 and Rankl/Opg ratio, while Tnf-α levels were decreased relative to tissue around teeth. Six weeks following infection, Tnf-α increased significantly while the expression of Foxp3 decreased in the tissue around the implants. No significant differences in anti- or pro-inflammatory mediators were found around teeth of infected, relative to non-infected mice. CONCLUSIONS: Oral infection with P. gingivalis of mice with implants induced bone loss and a shift in gingival cytokine expression. This mouse model enables exploration of the pathogenesis of peri-implantitis and testing of novel treatments.


Assuntos
Perda do Osso Alveolar/microbiologia , Peri-Implantite/microbiologia , Porphyromonas gingivalis/patogenicidade , Animais , Implantes Dentários , Planejamento de Prótese Dentária , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Propriedades de Superfície , Titânio , Fator de Necrose Tumoral alfa/metabolismo
6.
J Infect Dis ; 213(9): 1505-15, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26704610

RESUMO

Porphyromonas gingivalis,an anaerobic bacterium strongly linked to infection-driven inflammatory bone erosion, thrives within a highly inflamed milieu and disseminates to distant sites, such as atherosclerotic plaque. We examined the role of monocyte/macrophages in determining the outcome of infection with P. gingivalis. Surprisingly, transient monocyte/macrophage depletion led to greatly improved clearance of P. gingivalis. The chemokine receptors CCR2 and CX3CR1 play a major role in monocyte recruitment and differentiation to Ly6C(hi) vs CX3CR1(hi) subsets, respectively. To determine the contribution of particular monocyte/macrophage subsets to bacterial survival, we challenged chemokine receptor knockout mice and found that P. gingivalis clearance is significantly improved in the absence of CX3CR1. CX3CR1(hi) monocyte/macrophages promote P. gingivalis survival by downregulating neutrophil phagocytosis. Furthermore, CX3CR1 knockout mice resist bone resorption in the oral cavity following challenge with P. gingivalis Our findings provide an explanation for bacterial coexistence alongside an activate neutrophil infiltrate.


Assuntos
Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/microbiologia , Macrófagos/imunologia , Monócitos/imunologia , Porphyromonas gingivalis , Receptores CCR2/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Receptor 1 de Quimiocina CX3C , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Periodontite/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/patogenicidade , Receptores CCR2/genética , Receptores de Quimiocinas/genética
7.
J Immunol ; 184(3): 1455-62, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20042569

RESUMO

Porphyromonas gingivalis is a gram-negative anaerobe considered to be a major periodontal pathogen. TLR2 plays a central role in the response to P. gingivalis infection in vivo. In its absence there is a weak inflammatory response; however, bacteria are cleared rapidly compared with wild-type mice. We examined the role of the TLR adaptor proteins MyD88 and TLR/IL-1R-domain-containing adaptor-inducing IFN-beta in the inflammatory response to P. gingivalis in vivo and in the ability to clear the bacterial infection. Proinflammatory cytokine production in response to P. gingivalis infection depends on TLR2, but it does not require MyD88 or TLR/IL-1R-domain-containing adaptor-inducing IFN-beta. In contrast, the generation of intracellular toxic oxygen species and the ultimate clearance of P. gingivalis infection depend critically on MyD88, independent of TLR2. Thus, robust cytokine production and bacterial clearance are independent events mediated by distinct signaling pathways following infection with P. gingivalis.


Assuntos
Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Mediadores da Inflamação/fisiologia , Fator 88 de Diferenciação Mieloide/fisiologia , Porphyromonas gingivalis/imunologia , Receptor 2 Toll-Like/fisiologia , Animais , Infecções por Bacteroidaceae/patologia , Citocinas/biossíntese , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/patogenicidade , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética
8.
J Clin Periodontol ; 38 Suppl 11: 49-59, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21323704

RESUMO

BACKGROUND: Neutrophils are the predominant cells responsible for host defence against bacterial infection. Loss of neutrophil defence, due either to deficient number or function, strongly predisposes to bacterial infections such as periodontitis. Yet, the neutrophil oxidative and proteolytic arsenal has also been implicated in perpetrating periodontal tissue damage in periodontitis. AIM: In this review, we focus on recent developments that shed light on these two aspects of neutrophil function in periodontitis. METHODS: Primary search: using PubMed search for "neutophil", "periodontal", and "periodontitis". Secondary search: using references from the articles found in the first stage. RESULTS: Early histological studies showed that infiltrating neutrophils form a wall of cells abutting the junctional epithelium in periodontal inflammatory lesions. The chronic standoff between these neutrophils and the bacterial community suggests that bacterial evasion of neutrophil clearance is a major characteristic of periodontitis. Indeed, not all functional neutrophil deficiencies increase the risk of periodontitis, an observation that points the way towards identification of particular anti-bacterial pathways essential for protection against periodontal pathogens. The net result in the majority of periodontitis patients who exhibit normal neutrophil number and function, is that neutrophils accumulate in the periodontal tissue where they are available to participate in tissue destruction. Diminished neutrophil clearance further contributes to the persistence of activated neutrophils in the periodontal tissue. CONCLUSIONS: Data on the role of neutrophils in the pathogenesis of periodontitis are mixed. Neutrophils are a critical arm of the defence against periodontitis, but bacterial evasion of the neutrophil microbicidal machinery coupled with delayed neutrophil apoptosis may transform the neutrophil from defender to perpetrator. At this stage of knowledge, attempts to induce host modulation through neutrophil suppression or activation are premature.


Assuntos
Infecções Bacterianas/imunologia , Neutrófilos/imunologia , Periodontite/microbiologia , Suscetibilidade a Doenças/imunologia , Inserção Epitelial/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/fisiologia , Periodontite/imunologia , Periodontite/terapia , Fagocitose/imunologia
9.
Hum Vaccin Immunother ; 16(12): 3146-3154, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401698

RESUMO

Traditional non-living vaccines are often least effective in the populations that need them most, such as neonates and elderly adults. Vaccine adjuvants are one approach to boost the immunogenicity of antigens in populations with reduced immunity. Ideally, vaccine adjuvants will increase the seroconversion rates across the population, lead to stronger immune responses, and enable the administration of fewer vaccine doses. We previously demonstrated that a cationic liposomal formulation of the commercial influenza split virus vaccine (CCS/C-HA) enhanced cellular and humoral immunity to the virus, increased seroconversion rates, and improved survival after live virus challenge in a preclinical model, as compared to the commercial vaccine as is (F-HA). We now evaluated vaccine efficacy in different strains and sexes of mice and determined the role of innate immunity in the mechanism of action of the CCS/C adjuvant by testing the response of mice deficient in Toll-like receptors or the TLR/IL-1 adaptor protein MyD88 following immunization with CCS/C-HA vs. F-HA. Although TLR2- and TLR4-deficient mice responded to F-HA immunization, F-HA immunization failed to engender a significant immune response in the absence of MyD88. In contrast, immunization with the CCS/C-HA vaccine overcame the requirement for MyD88 in the response to the commercial vaccine and improved the immune responses and seroconversion rates in all strains of mice tested, including those deficient in TLR2 and TLR4.


Assuntos
Vacinas contra Influenza , Influenza Humana , Fator 88 de Diferenciação Mieloide , Infecções por Orthomyxoviridae , Adjuvantes Imunológicos , Animais , Anticorpos Antivirais , Humanos , Influenza Humana/prevenção & controle , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Fator 88 de Diferenciação Mieloide/genética , Infecções por Orthomyxoviridae/prevenção & controle
10.
Cancers (Basel) ; 12(8)2020 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-32824786

RESUMO

Porphyromonas gingivalis is a member of the dysbiotic oral microbiome associated with oral inflammation and periodontal disease. Intriguingly, epidemiological studies link P. gingivalis to an increased risk of pancreatic cancer. Given that oral bacteria are detected in human pancreatic cancer, and both mouse and human pancreata harbor microbiota, we explored the involvement of P. gingivalis in pancreatic tumorigenesis using cell lines and a xenograft model. Live P. gingivalis induced proliferation of pancreatic cancer cells; however, surprisingly, this effect was independent of Toll-like receptor 2, the innate immune receptor that is engaged in response to P. gingivalis on other cancer and immune cells, and is required for P. gingivalis to induce alveolar bone resorption. Instead, we found that P. gingivalis survives inside pancreatic cancer cells, a trait that can be enhanced in vitro and is increased by hypoxia, a central characteristic of pancreatic cancer. Increased tumor cell proliferation was related to the degree of intracellular persistence, and infection of tumor cells with P. gingivalis led to enhanced growth in vivo. To the best of our knowledge, this study is the first to demonstrate the direct effect of exposure to P. gingivalis on the tumorigenic behavior of pancreatic cancer cell lines. Our findings shed light on potential mechanisms underlying the pancreatic cancer-periodontitis link.

11.
Infect Immun ; 77(9): 3939-47, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19596768

RESUMO

Treponema denticola is considered an important oral pathogen in the development and progression of periodontal diseases. In the present study, the mechanisms of recognition and activation of murine macrophages by T. denticola and its major outer sheath protein (MSP) and lipooligosaccharide (LOS or glycolipid) were investigated. T. denticola cells and the MSP induced innate immune responses through TLR2-MyD88, whereas LOS induced a macrophage response through TLR4-MyD88. The presence of gamma interferon (IFN-gamma), or of high numbers of T. denticola, circumvented the requirement for TLR2 for the macrophage response to T. denticola, although the response was still dependent on MyD88. In contrast, synergy with IFN-gamma did not alter the TLR dependence of the response to the T. denticola surface components LOS and MSP, despite enhanced sensitivity. These data suggest that although there is flexibility in the requirements for recognition of T. denticola cells (TLR2 dependent or independent), MyD88 is a requirement for the downstream signaling events that lead to inflammation. We also demonstrate that both outer sheath molecules LOS and MSP induce macrophage tolerance to further stimulation with enterobacterial lipopolysaccharide. Tolerance induced by T. denticola components during mixed infections may represent a general mechanism through which bacteria evade clearance.


Assuntos
Proteínas de Bactérias/imunologia , Lipopolissacarídeos/imunologia , Porinas/imunologia , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Treponema denticola/imunologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Imunidade Inata , Interferon gama/fisiologia , Lipopolissacarídeos/toxicidade , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide/fisiologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-28848717

RESUMO

Porphyromonas gingivalis is a gram-negative anaerobic periodontal pathogen that persists in dysbiotic mixed-species biofilms alongside a dense inflammatory infiltrate of neutrophils and other leukocytes in the subgingival areas of the periodontium. Toll-like receptor 2 (TLR2) mediates the inflammatory response to P. gingivalis and TLR2-deficient mice resist alveolar bone resorption following oral challenge with this organism. Although, MyD88 is an adaptor protein considered necessary for TLR2-induced inflammation, we now report for the first time that oral challenge with P. gingivalis leads to alveolar bone resorption in the absence of MyD88. Indeed, in contrast to prototypical TLR2 agonists, such as the lipopeptide Pam3CSK4 that activates TLR2 in a strictly MyD88-dependent manner, P. gingivalis strikingly induced TLR2 signaling in neutrophils and macrophages regardless of the presence or absence of MyD88. Moreover, genetic or antibody-mediated inactivation of TLR2 completely reduced cytokine production in P. gingivalis-stimulated neutrophils or macrophages, suggesting that TLR2 plays a non-redundant role in the host response to P. gingivalis. In the absence of MyD88, inflammatory TLR2 signaling in P. gingivalis-stimulated neutrophils or macrophages depended upon PI3K. Intriguingly, TLR2-PI3K signaling was also critical to P. gingivalis evasion of killing by macrophages, since their ability to phagocytose this pathogen was reduced in a TLR2 and PI3K-dependent manner. Moreover, within those cells that did phagocytose bacteria, TLR2-PI3K signaling blocked phago-lysosomal maturation, thereby revealing a novel mechanism whereby P. gingivalis can enhance its intracellular survival. Therefore, P. gingivalis uncouples inflammation from bactericidal activity by substituting TLR2-PI3K in place of TLR2-MyD88 signaling. These findings further support the role of P. gingivalis as a keystone pathogen, which manipulates the host inflammatory response in a way that promotes bone loss but not bacterial clearance. Modulation of these host response factors may lead to novel therapeutic approaches to improve outcomes in disease conditions associated with P. gingivalis.


Assuntos
Perda do Osso Alveolar/microbiologia , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/metabolismo , Animais , Citocinas/análise , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fosfatidilinositol 3-Quinases/genética , Porphyromonas gingivalis/genética , Células RAW 264.7 , Receptor 2 Toll-Like/genética
13.
Sci Rep ; 7(1): 11516, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28912533

RESUMO

Tissue damage in chronic periodontal disease is driven by the host response to a dysbiotic microbiota, and not by bacteria directly. Among chronic inflammatory diseases of the oral cavity, inflammation and tissue damage around dental implants (peri-implantitis) is emerging as a major clinical challenge, since it is more severe and less responsive to treatment compared to inflammation around natural teeth. We tested whether oral fibroblasts from the periodontal ligament (PDLF), which are present around natural teeth but not around dental implants, actively regulate inflammatory responses to bacterial stimulation. We show that human PDLF down-regulate TNF-α post-transcriptionally in macrophages stimulated with the oral pathogen Porphyromonas gingivalis. Cell contact and secretion of IL-6 and IL-10 contribute to the modulation of inflammatory cytokine production. Although fibroblasts decreased TNF-α secretion, they enhanced the ability of macrophages to phagocytose bacteria. Surprisingly, donor matched oral fibroblasts from gingival tissues, or fibroblasts from peri-implant inflamed tissues were at least as active as PDLF in regulating macrophage responses to bacteria. In addition, priming fibroblasts with inflammatory mediators enhanced PDLF regulatory activity. A further understanding of the spectrum of fibroblast activities in inflammatory lesions is important in order to design ways to control inflammatory tissue damage.


Assuntos
Comunicação Celular , Fibroblastos/fisiologia , Macrófagos/imunologia , Porphyromonas gingivalis/imunologia , Linhagem Celular , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Fagocitose , Fator de Necrose Tumoral alfa/metabolismo
14.
Cell Rep ; 18(2): 419-431, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28076786

RESUMO

Whereas type I interferons (IFNs-I) were proposed to be elevated in human periodontitis, their role in the disease remains elusive. Using a bacterial-induced model of murine periodontitis, we revealed a prolonged elevation in IFN-I expression. This was due to the downregulation of TAM signaling, a major negative regulator of IFN-I. Further examination revealed that the expression of certain TAM components was reduced as a result of prolonged degradation of MYD88 by the infection. As a result of such prolonged IFN-I production, innate immunological functions of the gingiva were disrupted, and CD4+ T cells were constitutively primed by dendritic cells, leading to elevated RANKL expression and, subsequently, alveolar bone loss (ABL). Blocking IFN-I signaling restored proper immunological function and prevented ABL. Importantly, a loss of negative regulation on IFN-I expression by TAM signaling was also evident in periodontitis patients. These findings thus suggest a role for IFN-I in the pathogenesis of periodontitis.


Assuntos
Interferon Tipo I/biossíntese , Fator 88 de Diferenciação Mieloide/metabolismo , Porphyromonas gingivalis/fisiologia , Proteólise , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Perda do Osso Alveolar/complicações , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/patologia , Animais , Infecções por Bacteroidaceae/complicações , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Reabsorção Óssea/complicações , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Células Dendríticas/imunologia , Gengiva/microbiologia , Gengiva/patologia , Humanos , Interferon Tipo I/metabolismo , Leucócitos/patologia , Linfonodos/patologia , Camundongos , Mucosa Bucal/microbiologia , Mucosa Bucal/patologia , Periodontite/imunologia , Periodontite/microbiologia , Periodontite/patologia
15.
J Innate Immun ; 7(2): 127-135, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25228314

RESUMO

Cysteine proteases (gingipains) from Porphyromonas gingivalis are key virulence factors in chronic periodontitis. Innate immune receptors CD14, Toll-like receptor (TLR) 2 and TLR4 are important in P. gingivalis recognition. We examined the ability of gingipains to cleave CD14, TLR2 and TLR4, and the consequences for the cellular response to bacterial challenge. Macrophages were exposed to Arg (RgpA and RgpB)- and Lys (Kgp)-gingipains, and residual expression of TLR2, TLR4 and CD14 was determined by flow cytometry. The cellular response to live bacteria following exposure to purified gingipains was evaluated by TNFα production and bacterial phagocytosis. RgpA and Kgp decreased CD14 detection in a concentration (p = 0.0000002)- and time (p = 0.03)-dependent manner, whereas RgpB had no significant effect. TLR2 and TLR4 expression were unaffected. Reduction in CD14 expression was more efficient with Lys-gingipain than with Arg-gingipain. A reduced CD14 surface level correlated with decreased TNFα secretion and bacterial phagocytosis following challenge with live P. gingivalis, but the response to heat-killed bacteria was unaffected. Therefore, gingipains reduce CD14 expression without affecting expression of the bacterial-sensing TLRs. Reduced CD14 expression depends on the gingipain hemagglutinin/adhesion site and results in macrophage hyporesponsiveness to bacterial challenge. Further studies are needed to determine if reduced CD14 expression is linked to periodontitis induced by P. gingivalis.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Bacteroidaceae/imunologia , Periodontite Crônica/imunologia , Cisteína Proteases/metabolismo , Macrófagos/imunologia , Porphyromonas gingivalis/imunologia , Fatores de Virulência/metabolismo , Animais , Aderência Bacteriana , Infecções por Bacteroidaceae/complicações , Células Cultivadas , Periodontite Crônica/etiologia , Regulação para Baixo , Feminino , Humanos , Imunidade Celular , Imunidade Inata , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
16.
Oncotarget ; 6(26): 22613-23, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26158901

RESUMO

Oral squamous cell carcinoma (OSCC) is a lethal disease whose incidence is increasing. Epidemiologic studies demonstrate an association between periodontitis and oral cancer, and periodontal pathogens are implicated in the pathogenesis of numerous disorders, including rheumatoid arthritis, cardiovascular diseases, diabetes and gastrointestinal malignancies. Nevertheless, a causal role for periodontal pathogens in OSCC has not been shown, partly due to the lack of an appropriate animal model. Here, utilizing a newly-established murine model of periodontitis-associated oral tumorigenesis, we report that chronic bacterial infection promotes OSCC, and that augmented signaling along the IL-6-STAT3 axis underlies this effect. Our results indicate that periodontal pathogens P. gingivalis and F. nucleatum stimulate tumorigenesis via direct interaction with oral epithelial cells through Toll-like receptors. Furthermore, oral pathogens stimulate human OSCC proliferation and induce expression of key molecules implicated in tumorigenesis. To the best of our knowledge, these findings represent the first demonstration of a mechanistic role for oral bacteria in chemically induced OSCC tumorigenesis. These results are highly relevant for the design of effective prevention and treatment strategies for OSCC.


Assuntos
Carcinoma de Células Escamosas/microbiologia , Fusobacterium nucleatum/isolamento & purificação , Neoplasias de Cabeça e Pescoço/microbiologia , Neoplasias Bucais/microbiologia , Periodontite/patologia , Porphyromonas gingivalis/isolamento & purificação , Animais , Carcinogênese , Carcinoma de Células Escamosas/patologia , Modelos Animais de Doenças , Progressão da Doença , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Neoplasias Bucais/patologia , Periodontite/microbiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço
17.
Cell Host Microbe ; 15(6): 768-78, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24922578

RESUMO

Certain low-abundance bacterial species, such as the periodontitis-associated oral bacterium Porphyromonas gingivalis, can subvert host immunity to remodel a normally symbiotic microbiota into a dysbiotic, disease-provoking state. However, such pathogens also exploit inflammation to thrive in dysbiotic conditions. How these bacteria evade immunity while maintaining inflammation is unclear. As previously reported, P. gingivalis remodels the oral microbiota into a dysbiotic state by exploiting complement. Now we show that in neutrophils P. gingivalis disarms a host-protective TLR2-MyD88 pathway via proteasomal degradation of MyD88, whereas it activates an alternate TLR2-Mal-PI3K pathway. This alternate TLR2-Mal-PI3K pathway blocks phagocytosis, provides "bystander" protection to otherwise susceptible bacteria, and promotes dysbiotic inflammation in vivo. This mechanism to disengage bacterial clearance from inflammation required an intimate crosstalk between TLR2 and the complement receptor C5aR and can contribute to the persistence of microbial communities that drive dysbiotic diseases.


Assuntos
Disbiose/microbiologia , Periodontite/microbiologia , Porphyromonas gingivalis/patogenicidade , Receptor da Anafilatoxina C5a/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Infecções por Bacteroidaceae/imunologia , Proteínas do Sistema Complemento/imunologia , Disbiose/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/imunologia , Neutrófilos/microbiologia , Periodontite/imunologia , Fagocitose , Fosfatidilinositol 3-Quinases/metabolismo , Porphyromonas gingivalis/imunologia , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/imunologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia
18.
J Immunol ; 177(12): 8296-300, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17142724

RESUMO

Periodontitis is a chronic inflammatory disease that leads to destruction of the attachment apparatus of the teeth. The presence of particular oral bacteria and the host inflammatory response contribute to disease progression. Porphyromonas gingivalis is a Gram-negative anaerobe considered to be a major periodontal pathogen. Isolated Ags from P. gingivalis activate innate immune cells through TLR2 or TLR4. We challenged TLR2- and TLR4-deficient mice with live P. gingivalis and studied the inflammatory response and bacterial survival. Wild-type and TLR4-deficient mice produced high levels of cytokines in response to P. gingivalis challenge, whereas cytokine levels were nearly absent or delayed in TLR2-deficient mice. Surprisingly, P. gingivalis was cleared far more rapidly in TLR2-deficient mice. In addition, TLR2-deficient mice resisted bone loss following oral infection with P. gingivalis.


Assuntos
Perda do Osso Alveolar/etiologia , Imunidade Inata , Viabilidade Microbiana/imunologia , Porphyromonas gingivalis/imunologia , Receptor 2 Toll-Like/imunologia , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/microbiologia , Animais , Infecções por Bacteroidaceae , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodontite , Receptor 2 Toll-Like/deficiência , Receptor 4 Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA