Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38892314

RESUMO

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Assuntos
Apolipoproteínas E , Aterosclerose , Doenças Periodontais , Porphyromonas gingivalis , Animais , Camundongos , Apolipoproteínas E/deficiência , Doenças Periodontais/microbiologia , Doenças Periodontais/prevenção & controle , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Aterosclerose/microbiologia , Telomerase/metabolismo , Fragmentos de Peptídeos/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/microbiologia , Periodontite/microbiologia , Periodontite/prevenção & controle , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/complicações , Infecções por Bacteroidaceae/prevenção & controle , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Masculino , Humanos
2.
J Nanobiotechnology ; 16(1): 69, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-30219060

RESUMO

BACKGROUND: Porphyromonas gingivalis adherence to oral streptococci is a key point in the pathogenesis of periodontal diseases (Honda in Cell Host Microbe 10:423-425, 2011). Previous work in our groups has shown that a region of the streptococcal antigen denoted BAR (SspB Adherence Region) inhibits P. gingivalis/S. gordonii interaction and biofilm formation both in vitro and in a mouse model of periodontitis (Daep et al. in Infect Immun 74:5756-5762, 2006; Daep et al. in Infect immun 76:3273-3280, 2008; Daep et al. in Infect Immun 79:67-74, 2011). However, high localized concentration and prolonged exposure are needed for BAR to be an effective therapeutic in the oral cavity. METHODS: To address these challenges, we fabricated poly(lactic-co-glycolic acid) (PLGA) and methoxy-polyethylene glycol PLGA (mPEG-PLGA) nanoparticles (NPs) that encapsulate BAR peptide, and assessed the potency of BAR-encapsulated NPs to inhibit and disrupt in vitro two-species biofilms. In addition, the kinetics of BAR-encapsulated NPs were compared after different durations of exposure in a two-species biofilm model, against previously evaluated BAR-modified NPs and free BAR. RESULTS: BAR-encapsulated PLGA and mPEG-PLGA NPs potently inhibited biofilm formation (IC50 = 0.7 µM) and also disrupted established biofilms (IC50 = 1.3 µM) in a dose-dependent manner. In addition, BAR released during the first 2 h of administration potently inhibits biofilm formation, while a longer duration of 3 h is required to disrupt pre-existing biofilms. CONCLUSIONS: These results suggest that BAR-encapsulated NPs provide a potent platform to inhibit (prevent) and disrupt (treat) P. gingivalis/S. gordonii biofilms, relative to free BAR.


Assuntos
Antibacterianos/farmacologia , Antígenos de Bactérias/farmacologia , Biofilmes/efeitos dos fármacos , Portadores de Fármacos/química , Nanopartículas/química , Porphyromonas gingivalis/efeitos dos fármacos , Streptococcus gordonii/efeitos dos fármacos , Antibacterianos/administração & dosagem , Antibacterianos/imunologia , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Aderência Bacteriana/efeitos dos fármacos , Infecções por Bacteroidaceae/prevenção & controle , Humanos , Ácido Láctico/química , Polietilenoglicóis/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Porphyromonas gingivalis/fisiologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus/imunologia , Streptococcus gordonii/fisiologia
3.
J Periodontal Res ; 52(2): 285-291, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27282938

RESUMO

OBJECTIVE: Following Porphyromonas gingivalis infection in mice, the efficacy of vaccination by recombinant and native RgpA in modulating the early local anti-inflammatory and immune responses and periodontal bone loss were examined. MATERIAL AND METHODS: Using the subcutaneous chamber model, exudates were analyzed for cytokines after treatment with native RgpA and adjuvant (test), or adjuvant and saline alone (controls). Mice were also immunized with recombinant RgpA after being orally infected with P. gingivalis. After 6 wk, serum was examined for anti-P. gingivalis IgG1 and IgG2a titers and for alveolar bone resorption. RESULTS: Immunization with native RgpA shifted the immune response toward an anti-inflammatory response as demonstrated by decreased proinflammatory cytokine IL-1ß production and greater anti-inflammatory cytokine IL-4 in chamber exudates. Systemically, immunization with recombinant RgpA peptide prevented alveolar bone loss by 50%, similar to immunization with heat-killed whole bacteria. Furthermore, recombinant RgpA shifted the humoral response toward high IgG1 and low IgG2a titers, representing an in vivo anti-inflammatory response. CONCLUSIONS: The present study demonstrates the potential of RgpA to shift the early local immune response toward an anti-inflammatory response while vaccination with recRgpA protected against P. gingivalis-induced periodontitis.


Assuntos
Adesinas Bacterianas/imunologia , Perda do Osso Alveolar/prevenção & controle , Vacinas Bacterianas/uso terapêutico , Infecções por Bacteroidaceae/prevenção & controle , Cisteína Endopeptidases/imunologia , Porphyromonas gingivalis , Perda do Osso Alveolar/microbiologia , Animais , Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/imunologia , Infecções por Bacteroidaceae/imunologia , Feminino , Cisteína Endopeptidases Gingipaínas , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Porphyromonas gingivalis/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
4.
Scand J Immunol ; 81(6): 476-82, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25689343

RESUMO

The outer membrane protein RagB is one of the major virulence factors of Porphyromonas gingivalis (P. gingivalis). To prevent periodontitis and associated systemic diseases induced by P. gingivalis, we built B cell antigen epitope vaccine characterized by pIRES-ragB'-mGITRL to induce a protective immune responses. The B cell antigen epitope and scrambled peptide of ragB were predicted, cloned into pIRES and constructed pIRES-ragB', pIRES-scrambled epitopes and pIRES-ragB'-mGITRL. pIRES-ragB'-mGITRL was transfected into COS-7 cells. Subsequently, the 6-week-old female BALB/c mice were challenged by P. gingivalis following three time immunization by pIRES, pIRES-ragB', pIRES-scrambled epitopes and pIRES-ragB'-mGITRL. The levels of RagB-specific antibody in the serum and Tfh cells in the spleen were measured by ELISA and flow cytometry, respectively. And higher levels of RagB-specific IgG were produced in the immunized mice with pIRES-ragB'-mGITRL. Additionally, the number of Tfh cells was also expanded and lesions were diminished in pIRES-ragB'-mGITRL mice comparing with control groups. Our results clearly demonstrated that P. gingivalis B cell antigen epitope vaccine, pIRES-ragB'-mGITRL, could induce protective immune responses. Furthermore, our data also indicated that pIRES-ragB'-mGITRL was a potential therapeutic vaccine against P. gingivalis.


Assuntos
Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Epitopos de Linfócito B/imunologia , Porphyromonas gingivalis/imunologia , Fatores de Necrose Tumoral/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/prevenção & controle , Infecções por Bacteroidaceae/virologia , Células COS , Proliferação de Células , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Imunização , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos Endogâmicos BALB C , Periodontite/imunologia , Periodontite/prevenção & controle , Periodontite/virologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/fisiologia , Receptores de Antígenos de Linfócitos B/imunologia , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Necrose Tumoral/genética , Vacinas de DNA/farmacologia
5.
Biosci Biotechnol Biochem ; 77(1): 80-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23291749

RESUMO

Arg-specific gingipain (Rgp) is a major pathogenic determinant of Porphyromonas gingivalis which is a major pathogen in periodontal disease. We prepared protein extracts with Rgp-inhibitory activity from polished rice (Oryza sativa) and evaluated the effects of these extracts on the growth and pathogenicity of P. gingivalis. The extracts inhibited the proteolytic degradation of human proteins by P. gingivalis proteinases, and repressed the growth and homotypic biofilm formation of P. gingivalis. The disruption of adhesion of epithelial cells by P. gingivalis was also restricted by the rice protein extracts. Our results suggested that the rice protein extracts suppressed the pathogenicity and growth of P. gingivalis by inhibiting the bacterial proteinase activities, implying that the Rgp-inhibitory proteins prepared from rice may be potentially valuable as nutraceutical agents for preventing periodontal diseases.


Assuntos
Adesinas Bacterianas/metabolismo , Biofilmes/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Oryza/química , Extratos Vegetais/farmacologia , Proteínas de Plantas/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Infecções por Bacteroidaceae/prevenção & controle , Biofilmes/crescimento & desenvolvimento , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/isolamento & purificação , Ensaios Enzimáticos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Cisteína Endopeptidases Gingipaínas , Humanos , Extratos Vegetais/isolamento & purificação , Proteínas de Plantas/isolamento & purificação , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Proteólise/efeitos dos fármacos
6.
J Periodontal Res ; 47(5): 578-83, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22448761

RESUMO

BACKGROUND AND OBJECTIVE: Porphyromonas gingivalis has been implicated as one of the major pathogens in chronic periodontitis, an infectious disease affecting the majority of the adult population. We have previously demonstrated that a surface protein, arginine deiminase (ArcA), of Streptococcus cristatus represses production of P. gingivalis long fimbriae and interrupts the formation of P. gingivalis biofilms in vitro. Our in vivo studies have also shown that the distribution of P. gingivalis and S. cristatus in human subgingival plaque is negatively correlated. The objective of this study was to determine if S. cristatus ArcA inhibits P. gingivalis colonization and attenuates its subsequent pathogenesis in alveolar bone loss in the murine oral cavity. MATERIAL AND METHODS: A wild-type strain of S. cristatus (CC5A) and its arcA knockout mutant (ArcAE) were used as initial colonizers in the oral cavity of BALB/cByJ mice. Colonization of P. gingivalis on the existing S. cristatus biofilms was assessed by quantitative PCR, and P. gingivalis-induced alveolar bone loss was measured 6 wk after P. gingivalis infection. RESULTS: The presence of S. cristatus CC5A, but not its arcA mutant, attenuated P. gingivalis colonization in the murine oral cavity. In addition, P. gingivalis-induced alveolar bone loss was significantly lower in mice initially infected with S. cristatus CC5A than in those infected with the arcA mutant. CONCLUSION: This study provides direct evidence that S. cristatus ArcA has an inhibitory effect on P. gingivalis colonization, which may in turn attenuate the pathogenicity of P. gingivalis.


Assuntos
Proteínas de Bactérias/fisiologia , Hidrolases/fisiologia , Boca/microbiologia , Porphyromonas gingivalis/patogenicidade , Streptococcus/enzimologia , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/prevenção & controle , Animais , Antibiose/fisiologia , Aderência Bacteriana/fisiologia , Carga Bacteriana , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/prevenção & controle , Biofilmes/crescimento & desenvolvimento , Placa Dentária/microbiologia , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Gengiva/microbiologia , Hidrolases/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Mutação/genética , Reação em Cadeia da Polimerase , Organismos Livres de Patógenos Específicos , Streptococcus/genética , Fatores de Tempo , Dente/microbiologia
7.
Front Immunol ; 13: 806825, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250977

RESUMO

Porphyromonas gingivalis is a Gram-negative pathogenic bacterium associated with chronic periodontitis. The development of a chimeric peptide-based vaccine targeting this pathogen could be highly beneficial in preventing oral bone loss as well as other severe gum diseases. We applied a computational framework to design a multi-epitope-based vaccine candidate against P. gingivalis. The vaccine comprises epitopes from subunit proteins prioritized from the P. gingivalis reference strain (P. gingivalis ATCC 33277) using several reported vaccine properties. Protein-based subunit vaccines were prioritized through genomics techniques. Epitope prediction was performed using immunoinformatic servers and tools. Molecular modeling approaches were used to build a putative three-dimensional structure of the vaccine to understand its interactions with host immune cells through biophysical techniques such as molecular docking simulation studies and binding free energy methods. Genome subtraction identified 18 vaccine targets: six outer-membrane, nine cytoplasmic membrane-, one periplasmic, and two extracellular proteins. These proteins passed different vaccine checks required for the successful development of a vaccine candidate. The shortlisted proteins were subjected to immunoinformatic analysis to map B-cell derived T-cell epitopes, and antigenic, water-soluble, non-toxic, and good binders of DRB1*0101 were selected. The epitopes were then modeled into a multi-epitope peptide vaccine construct (linked epitopes plus adjuvant) to enhance immunogenicity and effectively engage both innate and adaptive immunity. Further, the molecular docking approach was used to determine the binding conformation of the vaccine to TLR2 innate immune receptor. Molecular dynamics simulations and binding free energy calculations of the vaccine-TLR2 complex were performed to highlight key intermolecular binding energies. Findings of this study will be useful for vaccine developers to design an effective vaccine for chronic periodontitis pathogens, specifically P. gingivalis.


Assuntos
Vacinas Bacterianas , Infecções por Bacteroidaceae , Porphyromonas gingivalis , Infecções por Bacteroidaceae/prevenção & controle , Periodontite Crônica/prevenção & controle , Biologia Computacional , Epitopos de Linfócito T , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Porphyromonas gingivalis/imunologia , Receptor 2 Toll-Like , Vacinas de Subunidades Antigênicas
8.
Mol Oral Microbiol ; 37(2): 31-41, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34921516

RESUMO

It has been reported that GroEL, a heat shock protein (HSP) produced by the representative periodontopathogenic bacterium, Porphyromonas gingivalis, induces inflammation-induced osteoclastogenesis and promotes alveolar bone resorption. In this study, we demonstrated the efficacy of a mucosal vaccine targeting GroEL against bone resorption induced by P. gingivalis. Female BALB/c mice received sublingual CpG oligodeoxynucleotide as an adjuvant with recombinant GroEL (rGroEL) prior to P. gingivalis exposure. Animals were euthanized 30 days after P. gingivalis inoculation. Sublingual immunization (SLI) with rGroEL elicited significant rGroEL-specific serum immunoglobulin (Ig)G and salivary IgA antibody (Ab) responses, and these responses were sustained for approximately 1 year. Interestingly, 10-fold more GroEL-specific IgA Ab-producing cells were detected in the submandibular glands (SMGs) than in the spleen. Antigen (Ag)-specific cells isolated from the spleen and SMGs induced significantly higher levels of IFN-γ expression after Ag restimulation in vitro. Flow cytometry illustrated that the frequency of CD11b+ dendritic cells with enhanced expression of CD80, CD86, CD40, and major histocompatibility complex II molecules was significantly increased in the SMGs. Furthermore, SLI with rGroEL significantly suppressed P. gingivalis-induced alveolar bone resorption and P. gingivalis-stimulated tumor necrosis factor-α, interleukin-6, and HSP60 expression in the gingiva. These findings suggest that SLI with rGroEL and CpG oligodeoxynucleotide is a beneficial strategy for preventing periodontal disease, mainly by presenting Ags in the oral region and inducing antibody production in the mucosal and systemic systems.


Assuntos
Perda do Osso Alveolar , Infecções por Bacteroidaceae , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/prevenção & controle , Animais , Anticorpos Antibacterianos , Infecções por Bacteroidaceae/prevenção & controle , Feminino , Imunização , Imunoglobulina A Secretora/metabolismo , Imunoglobulina G , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/metabolismo , Porphyromonas gingivalis/metabolismo
9.
Biomed Res Int ; 2022: 9770899, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35028318

RESUMO

Periodontitis is a chronic inflammatory disease caused by periodontopathogenic bacteria that form biofilms in periodontal pockets. The gingival epithelium acts as the first physical barrier in fighting attacks by periodontopathogenic pathogens, such as the primary etiological agent Porphyromonas gingivalis, and various exogenous chemicals, as well as regulates the local innate immune responses. Therefore, the development of novel oral care products to inhibit inflammatory reactions caused by bacterial infection and protect the gingival epithelium is necessary. Juncus effusus L. has generally been used as an indigenous medicine, such as a diuretic, an antipyretic, and an analgesic, in ancient practice. In this study, we examined the effects of a water extract from J. effusus L. on the inhibition of the inflammatory reaction elicited by bacterial infection and protection of the oral epithelium by chemical irritation. Pretreatment of oral epithelial cells with the water extract from J. effusus L. significantly reduced P. gingivalis or its lipopolysaccharide- (LPS-) mediated production of chemokines (interleukin-8 and C-C-chemokine ligand20) in a concentration-dependent manner with comparable to or greater effects than epigallocatechin gallate and protected oral epithelial cells from injury by chemical irritants, cetylpyridinium chloride, and benzethonium chloride. Moreover, the water extract from J. effusus L. in the presence of antimicrobial agents or antifibrinolytics already used as ingredients in mouthwash could significantly reduce the production of chemokines from P. gingivalis LPS-stimulated oral epithelial cells in a concentration-dependent manner. These findings suggest that the water extract from J. effusus L. is potentially useful for oral care to prevent oral infections, such as periodontal infections, and maintain oral epithelial function.


Assuntos
Anti-Inflamatórios , Queratinócitos/metabolismo , Magnoliopsida/química , Mucosa Bucal/metabolismo , Extratos Vegetais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/prevenção & controle , Linhagem Celular Transformada , Humanos , Queratinócitos/patologia , Mucosa Bucal/patologia , Periodontite/metabolismo , Periodontite/patologia , Periodontite/prevenção & controle , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Porphyromonas gingivalis/metabolismo
10.
Infect Immun ; 79(2): 895-904, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21115722

RESUMO

We assessed the efficacy of a fusion protein consisting of the 25-kDa antigenic region of Porphyromonas gingivalis hemagglutinin A and the Escherichia coli maltose-binding protein (25k-hagA-MBP) as a nasal vaccine for the prevention of oral infection with P. gingivalis. Nasal immunization with 25k-hagA-MBP induced high levels of 25k-hagA-specific serum IgG, serum IgA, and salivary IgA antibodies in a Toll-like receptor 4 (TLR4)-dependent manner. These antibody responses were maintained for at least 1 year after immunization. Analysis of cytokine responses showed that nasal administration of 25k-hagA-MBP induced antigen-specific CD4(+) T cells producing interleukin 4 (IL-4) and IL-5, but not gamma interferon (IFN-γ), in the spleen and cervical lymph nodes (CLNs). Furthermore, increased numbers of CD11c(+) CD8α(+), but not CD11c(+) CD11b(+) or CD11c(+) B220(+), dendritic cells with upregulated expression of CD80, CD86, CD40, and major histocompatibility complex class II (MHC II) molecules were noted in the spleen, CLNs, and nasopharynx-associated lymphoreticular tissues (NALT). Interestingly, when 25k-hagA-MBP or cholera toxin (CT) was given intranasally to enable examination of their presence in neuronal tissues, the amounts of 25k-hagA-MBP were significantly lower than those of CT. Importantly, mice given 25k-hagA-MBP nasally showed a significant reduction in alveolar bone loss caused by oral infection with P. gingivalis, even 1 year after the immunization. These results suggest that 25k-hagA-MBP administered nasally would be an effective and safe mucosal vaccine against P. gingivalis infection and may be an important tool for the prevention of chronic periodontitis in humans.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Células Dendríticas/imunologia , Proteínas Ligantes de Maltose/imunologia , Porphyromonas gingivalis/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Perda do Osso Alveolar/prevenção & controle , Animais , Antígenos de Bactérias , Proteínas de Bactérias/química , Vacinas Bacterianas/administração & dosagem , Infecções por Bacteroidaceae/prevenção & controle , Antígeno CD11c/imunologia , Antígenos CD8/imunologia , Regulação da Expressão Gênica/imunologia , Lectinas/química , Lectinas/imunologia , Proteínas Ligantes de Maltose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Porphyromonas gingivalis/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
11.
Methods Mol Biol ; 2210: 157-166, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32815136

RESUMO

Bacteria release spherical nanobodies, known as membrane vesicles (MVs), during various growth phases. MVs have been gaining recognition as structurally stable vehicles in the last two decades because they deliver a wide range of antigens, virulence factors, and immunomodulators to the host. These functions suggest not only the possible contribution of MVs to pathogenicity but also the potential applicability of low-dose MVs for use as vaccines. Here, we describe a series of methods for isolating MVs of Porphyromonas gingivalis, which is an important species among periodontopathic bacteria. The present chapter also introduces a mouse model of intranasal immunization using MVs from P. gingivalis.


Assuntos
Membrana Externa Bacteriana/imunologia , Vacinas Bacterianas/uso terapêutico , Infecções por Bacteroidaceae/prevenção & controle , Porphyromonas gingivalis/imunologia , Administração Intranasal , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Infecções por Bacteroidaceae/imunologia , Centrifugação com Gradiente de Concentração/métodos , Modelos Animais de Doenças , Feminino , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Ultracentrifugação/métodos
12.
Front Immunol ; 12: 634923, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717178

RESUMO

Our previous studies showed that a combination of a DNA plasmid encoding Flt3 ligand (pFL) and CpG oligodeoxynucleotides 1826 (CpG ODN) (FL/CpG) as a nasal adjuvant provoked antigen-specific immune responses. In this study, we investigated the efficacy of a nasal vaccine consisting of FimA as the structural subunit of Porphyromonas gingivalis (P. gingivalis) fimbriae and FL/CpG for the induction of FimA-specific antibody (Ab) responses and their protective roles against nasal and lung infection by P. gingivalis, a keystone pathogen in the etiology of periodontal disease. C57BL/6 mice were nasally immunized with recombinant FimA (rFimA) plus FL/CpG three times at weekly intervals. As a control, mice were given nasal rFimA alone. Nasal washes (NWs) and bronchoalveolar lavage fluid (BALF) of mice given nasal rFimA plus FL/CpG resulted in increased levels of rFimA-specific secretory IgA (SIgA) and IgG Ab responses when compared with those in controls. Significantly increased numbers of CD8- or CD11b-expressing mature-type dendritic cells (DCs) were detected in the respiratory inductive and effector tissues of mice given rFimA plus FL/CpG. Additionally, significantly upregulated Th1/Th2-type cytokine responses by rFimA-stimulated CD4+ T cells were noted in the respiratory effector tissues. When mice were challenged with live P. gingivalis via the nasal route, mice immunized nasally with rFimA plus FL/CpG inhibited P. gingivalis colonization in the nasal cavities and lungs. In contrast, controls failed to show protection. Of interest, when IgA-deficient mice given nasal rFimA plus FL/CpG were challenged with nasal P. gingivalis, the inhibition of bacterial colonization in the respiratory tracts was not seen. Taken together, these results show that nasal FL/CpG effectively enhanced DCs and provided balanced Th1- and Th2-type cytokine response-mediated rFimA-specific IgA protective immunity in the respiratory tract against P. gingivalis. A nasal administration with rFimA and FL/CpG could be a candidate for potent mucosal vaccines for the elimination of inhaled P. gingivalis in periodontal patients.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antibacterianos/metabolismo , Vacinas Bacterianas/administração & dosagem , Infecções por Bacteroidaceae/prevenção & controle , Proteínas de Fímbrias/administração & dosagem , Imunogenicidade da Vacina , Imunoglobulina A Secretora/metabolismo , Porphyromonas gingivalis/imunologia , Sistema Respiratório/efeitos dos fármacos , Administração Intranasal , Animais , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Modelos Animais de Doenças , Feminino , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Esquemas de Imunização , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Porphyromonas gingivalis/patogenicidade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo , Sistema Respiratório/microbiologia , Fatores de Tempo , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
13.
J Immunol ; 181(6): 4141-9, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768871

RESUMO

The periodontal pathogen Porphyromonas gingivalis is implicated in certain systemic diseases including atherosclerosis and aspiration pneumonia. This organism induces innate responses predominantly through TLR2, which also mediates its ability to induce experimental periodontitis and accelerate atherosclerosis. Using a validated mouse model of intratracheal challenge, we investigated the role of TLR2 in the control of P. gingivalis acute pulmonary infection. TLR2-deficient mice elicited reduced proinflammatory or antimicrobial responses (KC, MIP-1alpha, TNF-alpha, IL-6, IL-12p70, and NO) in the lung and exhibited impaired clearance of P. gingivalis compared with normal controls. However, the influx of polymorphonuclear leukocytes into the lung and the numbers of resident alveolar macrophages (AM) were comparable between the two groups. TLR2 signaling was important for in vitro killing of P. gingivalis by polymorphonuclear leukocytes or AM and, moreover, the AM bactericidal activity required NO production. Strikingly, AM were more potent than peritoneal or splenic macrophages in P. gingivalis killing, attributed to diminished AM expression of complement receptor-3 (CR3), which is exploited by P. gingivalis to promote its survival. The selective expression of CR3 by tissue macrophages and the requirement of TLR2 inside-out signaling for CR3 exploitation by P. gingivalis suggest that the role of TLR2 in host protection may be contextual. Thus, although TLR2 may mediate destructive effects, as seen in models of experimental periodontitis and atherosclerosis, we have now shown that the same receptor confers protection against P. gingivalis in acute lung infection.


Assuntos
Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/metabolismo , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Porphyromonas gingivalis/imunologia , Receptor 2 Toll-Like/fisiologia , Doença Aguda , Animais , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/microbiologia , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/prevenção & controle , Células Cultivadas , Feminino , Imunidade Inata/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/microbiologia , Periodontite/imunologia , Periodontite/metabolismo , Periodontite/microbiologia , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/prevenção & controle , Baço/citologia , Baço/imunologia , Baço/microbiologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética
14.
J Clin Periodontol ; 37(9): 812-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20670341

RESUMO

AIM: To assess the potential of using vaccination with Porphyromonas gingivalis or Fusobacterium nucleatum, in modulating local subcutaneous inflammatory response and alveolar bone loss following coinfection with both bacteria. MATERIALS AND METHODS: Mice were immunized against either P. gingivalis or F. nucleatum. The cytokine response to mixed infection with P. gingivalis and F. nucleatum was evaluated using the subcutaneous chamber model. The alveolar bone loss induced by oral mixed infection was evaluated by micro-CT using the experimental periodontitis model. Serum levels of specific antibodies were determined by ELISA. RESULTS: Vaccination with either bacterium produced a specific humoral response before infection. Animals immunized against either bacteria following a mixed infection with P. gingivalis and F. nucleatum, showed decreased TNFalpha (but not IL-1beta) levels as compared with non-immunized animals. However, the vaccination did not change the level of mixed infection-induced alveolar bone loss when compared with non-immunized animals. Six weeks following the oral mixed infection, specific antibody titres remained high. Furthermore, specific antibodies against the non-immunized bacterium were present at high levels. CONCLUSIONS: While vaccination produced specific antibodies and suppressed the inflammatory response, it failed to prevent or reduce the progression of experimental periodontitis induced by mixed infection with P. gingivalis and F. nucleatum.


Assuntos
Vacinas Bacterianas , Infecções por Bacteroidaceae/imunologia , Infecções por Fusobacterium/imunologia , Fusobacterium nucleatum/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Vacinação , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Infecções por Bacteroidaceae/prevenção & controle , Progressão da Doença , Feminino , Infecções por Fusobacterium/prevenção & controle , Imunização , Injeções Subcutâneas , Interleucina-1beta/sangue , Camundongos , Camundongos Endogâmicos BALB C , Periodontite/prevenção & controle , Organismos Livres de Patógenos Específicos , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Vacinas de Produtos Inativados , Microtomografia por Raio-X
15.
Biochem Biophys Res Commun ; 390(3): 937-41, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19852927

RESUMO

In this study, we demonstrated that the 40-kDa outer membrane protein of Porphyromonas gingivalis (40k-OMP) sublingually administered with a cDNA vector plasmid encoding Flt3 ligand (pFL) elicited a protective immune response. Sublingual immunization of mice with 40k-OMP plus pFL induced significant serum IgG and IgA, as well as salivary IgA, antibody responses that were comparable to those induced by 40k-OMP plus cholera toxin as adjuvant. When the subclasses of 40k-OMP-specific IgG were evaluated, sublingual immunization with 40k-OMP plus pFL induced both IgG1 and IgG2a antibody responses. Sublingual delivery of pFL resulted in FL expression in submandibular glands, but not in other oral tissues. Furthermore, marked increases in FL protein occurred in saliva and serum, and the frequencies of both CD11c(+)CD11b(+) and CD11c(+)CD8alpha(+) dendritic cells with up-regulated expression of CD80, CD86 and CD40 molecules significantly increased in submandibular lymph nodes and spleen. Importantly, the mice given sublingual 40k-OMP plus pFL showed a significant reduction of alveolar bone loss caused by oral infection with P. gingivalis. These findings suggest that sublingual administration of 40k-OMP with pFL acts as an effective and safe mucosal vaccine against oral P. gingivalis infection, and may be a useful tool in the prevention of chronic periodontitis.


Assuntos
Proteínas da Membrana Bacteriana Externa/administração & dosagem , Infecções por Bacteroidaceae/prevenção & controle , Periodontite Crônica/prevenção & controle , Proteínas de Membrana/imunologia , Boca/imunologia , Porphyromonas gingivalis/imunologia , Vacinação , Administração Sublingual , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/prevenção & controle , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Periodontite Crônica/microbiologia , Feminino , Vetores Genéticos/administração & dosagem , Imunoglobulina G/imunologia , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Boca/microbiologia
16.
Med Hypotheses ; 72(1): 41-2, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18783892

RESUMO

Periodontitis are among the most frequent infectious diseases affecting children and adolescents. The primary etiology of periodontal diseases is the bacterial plaque. Studies have demonstrated the feasibility of inducing immune responses by DNA vaccines against Porphyromonas gingivalis, the major aetiological agent of chronic periodontitis, and the subsequent development of periodontitis in animal models. But until now, the poor immunogenicity, particularly in large animals, is still a major problem in DNA vaccination. Cytotoxic T lymphocyte-associated antigen 4 (CTLA4) is a membrane bound molecule mainly located on activated T cells. Its extracellular V-domain is considered to be involved in mediating the binding to the B7 molecule on antigen-presenting cells (APCs). By utilizing the interaction between CTLA4 and B7, specific antigens can be targeted to APCs by fusion to CTLA4 and DNA vaccines encoding CTLA4 and specific antigens greatly increased the systemic antibody responses following immunization in mice. We hypothesize that targeting antigens to APCs offers a potential strategy to enhance the immune responses generated by DNA vaccines against periodontitis.


Assuntos
Antígenos CD/imunologia , Infecções por Bacteroidaceae/imunologia , Periodontite/imunologia , Porphyromonas gingivalis , Adolescente , Animais , Antígenos CD/genética , Infecções por Bacteroidaceae/prevenção & controle , Antígeno CTLA-4 , Criança , Modelos Animais de Doenças , Humanos , Periodontite/microbiologia , Periodontite/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/uso terapêutico
17.
Immunopharmacol Immunotoxicol ; 31(3): 352-7, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19555215

RESUMO

Periodontal disease is a highly prevalent disorder affecting up to 90% of the global population. Recent epidemiological studies have shown that an association exists between periodontal disease and cardiovascular disease. Porphyromonas gingivalis, the causative agent of destructive chronic inflammation in the periodontium, can accelerate atheroma deposition in animal models. Emerging evidence suggests that vaccination against this pathogen's virulence factors may confer disease resistance. In this review, we focus on the role of inflammatory mechanisms in the formation and activation of atherosclerotic plaques accelerated by P. gingivalis in an apo E-deficient mouse model. Further, we examine whether a nasal vaccine-induced antigen-specific mucosal response can reduce P. gingivalis-accelerated atherosclerosis.


Assuntos
Aterosclerose/imunologia , Vacinas Bacterianas/farmacologia , Infecções por Bacteroidaceae/imunologia , Imunidade nas Mucosas/imunologia , Imunização , Doenças Periodontais/imunologia , Porphyromonas gingivalis/imunologia , Administração Intranasal , Animais , Aterosclerose/prevenção & controle , Vacinas Bacterianas/imunologia , Infecções por Bacteroidaceae/prevenção & controle , Humanos , Imunidade nas Mucosas/efeitos dos fármacos , Inflamação/imunologia , Inflamação/prevenção & controle , Camundongos , Camundongos Knockout , Doenças Periodontais/prevenção & controle
18.
Nutrients ; 11(9)2019 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-31527555

RESUMO

BACKGROUND: Coffee is a major dietary source of polyphenols. Previous research found that coffee had a protective effect on periodontal disease. In this study, we aimed to investigate whether coffee extract and its primary phenolic acid, chlorogenic acid, affect the growth and protease activity of a periodontopathogen Porphyromonas gingivalis (P. gingivalis). METHODS: Coffee extract and chlorogenic acid were prepared by a two-fold serial dilution. The turbid metric test and plate count method were used to examine the inhibitory effects of chlorogenic acid on P. gingivalis. The time-kill assay was used to measure changes in the viability of P. gingivalis after exposure to chlorogenic acid for 0-24 h. The protease activity of P. gingivalis was analyzed using the optical density of a chromogenic substrate. RESULTS: As a result, the minimum inhibitory concentration (MIC) of chlorogenic acid was 4 mg/mL, and the minimum bactericidal concentration was 16 mg/mL. Chlorogenic acid at concentrations above MIC resulted in a longer-lasting inhibitory effect on P. gingivalis viability and significantly reduced associated protease activity. The coffee extract showed antibacterial activity as observed by the disk diffusion test, whereas these inhibitory effects were not affected by different roast degrees of coffee. CONCLUSIONS: Collectively, our novel findings indicate that chlorogenic acid not only has antimicrobial activity but also reduced the protease activity of P. gingivalis. In addition, coffee extract inhibits the proliferation of P. gingivalis, which may partly be attributed to the effect of chlorogenic acid.


Assuntos
Antibacterianos/farmacologia , Infecções por Bacteroidaceae/prevenção & controle , Ácido Clorogênico/farmacologia , Coffea/química , Periodontite/tratamento farmacológico , Extratos Vegetais/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Antibacterianos/isolamento & purificação , Proteínas de Bactérias/metabolismo , Infecções por Bacteroidaceae/microbiologia , Ácido Clorogênico/isolamento & purificação , Testes de Sensibilidade a Antimicrobianos por Disco-Difusão , Viabilidade Microbiana/efeitos dos fármacos , Peptídeo Hidrolases/metabolismo , Periodontite/microbiologia , Extratos Vegetais/isolamento & purificação , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/patogenicidade , Sementes/química , Fatores de Tempo , Fatores de Virulência/metabolismo
19.
Cancer Immunol Res ; 7(9): 1440-1456, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31350278

RESUMO

Pathogens are capable of hijacking immune defense mechanisms, thereby creating a tolerogenic environment for hypermutated malignant cells that arise within the site of infection. Immune checkpoint-oriented immunotherapies have shown considerable promise. Equally important, the epigenetic reprogramming of an immune-evasive phenotype that activates the immune system in a synergistic manner can improve immunotherapy outcomes. These advances have led to combinations of epigenetic- and immune-based therapeutics. We previously demonstrated that Porphyromonas gingivalis isolated from esophageal squamous cell carcinoma (ESCC) lesions represents a major pathogen associated with this deadly disease. In this study, we examined the mechanisms associated with host immunity during P. gingivalis infection and demonstrated that experimentally infected ESCC responds by increasing the expression of B7-H4 and lysine demethylase 5B, which allowed subsequent in vivo analysis of the immunotherapeutic effects of anti-B7-H4 and histone demethylase inhibitors in models of chronic infection and immunity against xenografted human tumors. Using three different preclinical mouse models receiving combined therapy, we showed that mice mounted strong resistance against P. gingivalis infection and tumor challenge. This may have occurred via generation of a T cell-mediated response in the microenvironment and formation of immune memory. In ESCC subjects, coexpression of B7-H4 and KDM5B correlated more significantly with bacterial load than with the expression of either molecule alone. These results highlight the unique ability of P. gingivalis to evade immunity and define potential targets that can be exploited therapeutically to improve the control of P. gingivalis infection and the development of associated neoplasia.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Infecções por Bacteroidaceae/prevenção & controle , Carcinoma de Células Escamosas do Esôfago/imunologia , Imunidade/efeitos dos fármacos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Porphyromonas/imunologia , Proteínas Repressoras/antagonistas & inibidores , Inibidor 1 da Ativação de Células T com Domínio V-Set/antagonistas & inibidores , Animais , Infecções por Bacteroidaceae/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Epigênese Genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imuno-Histoquímica , Imunofenotipagem , Ativação Linfocitária , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Porphyromonas/genética , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
20.
Infect Immun ; 76(6): 2777-84, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18411288

RESUMO

In this study, we demonstrated that the 40-kDa outer membrane protein of Porphyromonas gingivalis (40-kDa OMP) nasally administered with a nontoxic chimeric adjuvant that combines the A subunit of mutant cholera toxin E112K with the pentameric B subunit of heat-labile enterotoxin from enterotoxigenic Escherichia coli (mCTA/LTB) elicited a long-term protective immune response. Immunization with the 40-kDa OMP and mCTA/LTB induced high levels of 40-kDa-OMP-specific immunoglobulin G (IgG) and IgA antibodies (Abs) in sera and elicited a significant IgA anti-40-kDa OMP Ab response in saliva. These Ab responses were maintained for at least 1 year after the immunization. Although using adjuvant mCTA/LTB gave Ab responses in the saliva comparable to those obtained using native cholera toxin (nCT) as the adjuvant, the levels of total IgE and 40-kDa-OMP-specific IgE Abs as well as interleukin-4 levels induced by the immunization with mCTA/LTB were lower than those induced by the immunization with nCT. Importantly, IgG Abs generated by nasal immunization with the 40-kDa OMP plus mCTA/LTB inhibited the coaggregation and hemagglutinin activities of P. gingivalis. Furthermore, the mice given nasal 40-kDa OMP plus mCTA/LTB showed a significant reduction of alveolar bone loss caused by oral infection with P. gingivalis even 1 year after the immunization compared to the loss in unimmunized mice. Because mCTA/LTB is nontoxic, nasally administered 40-kDa OMP together with mCTA/LTB should be an effective and safe mucosal vaccine against P. gingivalis infection in humans and may be an important tool for the prevention of chronic periodontitis.


Assuntos
Adjuvantes Imunológicos , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Enterotoxinas/imunologia , Imunoglobulina E/sangue , Porphyromonas gingivalis/imunologia , Administração Intranasal , Perda do Osso Alveolar/microbiologia , Perda do Osso Alveolar/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Infecções por Bacteroidaceae/prevenção & controle , Feminino , Imunidade nas Mucosas , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Periodontite/complicações , Periodontite/microbiologia , Periodontite/prevenção & controle , Proteínas Recombinantes de Fusão/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA