Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(9): e1009874, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34473800

RESUMEN

Staphylococcus aureus is a major human pathogen, which can invade and survive in non-professional and professional phagocytes. Uptake by host cells is thought to contribute to pathogenicity and persistence of the bacterium. Upon internalization by epithelial cells, cytotoxic S. aureus strains can escape from the phagosome, replicate in the cytosol and induce host cell death. Here, we identified a staphylococcal cysteine protease to induce cell death after translocation of intracellular S. aureus into the host cell cytoplasm. We demonstrated that loss of staphopain A function leads to delayed onset of host cell death and prolonged intracellular replication of S. aureus in epithelial cells. Overexpression of staphopain A in a non-cytotoxic strain facilitated intracellular killing of the host cell even in the absence of detectable intracellular replication. Moreover, staphopain A contributed to efficient colonization of the lung in a mouse pneumonia model. In phagocytic cells, where intracellular S. aureus is exclusively localized in the phagosome, staphopain A did not contribute to cytotoxicity. Our study suggests that staphopain A is utilized by S. aureus to exit the epithelial host cell and thus contributes to tissue destruction and dissemination of infection.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Células Epiteliales/patología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Animales , Muerte Celular/fisiología , Células Epiteliales/microbiología , Humanos , Ratones , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo
2.
J Biol Chem ; 290(45): 27248-27260, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26385924

RESUMEN

Porphyromonas gingivalis is a peptide-fermenting asaccharolytic periodontal pathogen. Its genome contains several genes encoding cysteine peptidases other than gingipains. One of these genes (PG1055) encodes a protein called Tpr (thiol protease) that has sequence similarity to cysteine peptidases of the papain and calpain families. In this study we biochemically characterize Tpr. We found that the 55-kDa Tpr inactive zymogen proteolytically processes itself into active forms of 48, 37, and 33 kDa via sequential truncations at the N terminus. These processed molecular forms of Tpr are associated with the bacterial outer membrane where they are likely responsible for the generation of metabolic peptides required for survival of the pathogen. Both autoprocessing and activity were dependent on calcium concentrations >1 mm, consistent with the protein's activity within the intestinal and inflammatory milieus. Calcium also stabilized the Tpr structure and rendered the protein fully resistant to proteolytic degradation by gingipains. Together, our findings suggest that Tpr is an example of a bacterial calpain, a calcium-responsive peptidase that may generate substrates required for the peptide-fermenting metabolism of P. gingivalis. Aside from nutrient generation, Tpr may also be involved in evasion of host immune response through degradation of the antimicrobial peptide LL-37 and complement proteins C3, C4, and C5. Taken together, these results indicate that Tpr likely represents an important pathogenesis factor for P. gingivalis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Calcio/metabolismo , Endopeptidasas/metabolismo , Porphyromonas gingivalis/enzimología , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Calpaína/química , Calpaína/genética , Calpaína/metabolismo , Caseínas/metabolismo , Dominio Catalítico/genética , Bovinos , Endopeptidasas/química , Endopeptidasas/genética , Estabilidad de Enzimas , Fibrinógeno/metabolismo , Fibronectinas/metabolismo , Genes Bacterianos , Humanos , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidad , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Catelicidinas
3.
J Immunol ; 192(11): 5363-72, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24771854

RESUMEN

Cathelicidin LL-37 plays an essential role in innate immunity by killing invading microorganisms and regulating the inflammatory response. These activities depend on the cationic character of the peptide, which is conferred by arginine and lysine residues. At inflammatory foci in vivo, LL-37 is exposed to peptidyl arginine deiminase (PAD), an enzyme released by inflammatory cells. Therefore, we hypothesized that PAD-mediated citrullination of the arginine residues within LL-37 will abrogate its immunomodulatory functions. We found that, when citrullinated, LL-37 was at least 40 times less efficient at neutralizing the proinflammatory activity of LPS due to a marked decrease in its affinity for endotoxin. Also, the ability of citrullinated LL-37 to quench macrophage responses to lipoteichoic acid and poly(I:C) signaling via TLR2 and TLR3, respectively, was significantly reduced. Furthermore, in stark contrast to native LL-37, the modified peptide completely lost the ability to prevent morbidity and mortality in a mouse model of d-galactosamine-sensitized endotoxin shock. In fact, administration of citrullinated LL-37 plus endotoxin actually exacerbated sepsis due to the inability of LL-37 to neutralize LPS and the subsequent enhancement of systemic inflammation due to increased serum levels of IL-6. Importantly, serum from septic mice showed increased PAD activity, which strongly correlated with the level of citrullination, indicating that PAD-driven protein modification occurs in vivo. Because LL-37 is a potential treatment for sepsis, its administration should be preceded by a careful analysis to ensure that the citrullinated peptide is not generated in treated patients.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Citrulina/inmunología , Inmunidad Innata , Macrófagos/inmunología , Sepsis/inmunología , Sepsis/prevención & control , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/farmacología , Línea Celular , Citrulina/genética , Femenino , Humanos , Hidrolasas/genética , Hidrolasas/inmunología , Inductores de Interferón/farmacología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/inmunología , Macrófagos/patología , Masculino , Ratones , Poli I-C/farmacología , Sepsis/genética , Sepsis/patología , Ácidos Teicoicos/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Catelicidinas
4.
Clin Oral Investig ; 20(4): 675-83, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26264638

RESUMEN

OBJECTIVES: The aim of the present study was to assess human and bacterial peptidylarginine deiminase (PAD) activity in the gingival crevicular fluid (GCF) in the context of serum levels of antibodies against citrullinated epitopes in rheumatoid arthritis and periodontitis. MATERIALS AND METHODS: Human PAD and Porphyromonas gingivalis-derived enzyme (PPAD) activities were measured in the GCF of 52 rheumatoid arthritis (RA) patients (48 with periodontitis and 4 without) and 44 non-RA controls (28 with periodontitis and 16 without). Serum antibodies against citrullinated epitopes were measured by ELISA. Bacteria being associated with periodontitis were determined by nucleic-acid-based methods. RESULTS: Citrullination was present in 26 (50%) RA patients and 23 (48%) controls. PAD and PPAD activities were detected in 36 (69%) and 30 (58%) RA patients, respectively, and in 30 (68%) and 21 (50%) controls, respectively. PPAD activity was higher in RA and non-RA patients with periodontitis than in those without (p = 0.038; p = 0.004), and was detected in 35 of 59 P. gingivalis-positive samples, and in 16 of 37 P. gingivalis-negative samples in association with high antibody levels against that species. CONCLUSIONS: PAD and PPAD activities within the periodontium are elevated in RA and non-RA patients with periodontitis. PPAD secreted by P. gingivalis residing in epithelial cells may exert its citrullinating activity in distant regions of the periodontium or even distant tissues. CLINICAL RELEVANCE: In periodontitis, the citrullination of proteins/peptides by human and bacterial peptidylarginine deiminases may generate antibodies after breaching immunotolerance in susceptible individuals.


Asunto(s)
Artritis Reumatoide/complicaciones , Citrulinación , Periodontitis/complicaciones , Periodoncio/metabolismo , Humanos , Péptidos , Porphyromonas gingivalis
5.
PLoS Pathog ; 9(9): e1003627, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068934

RESUMEN

Rheumatoid arthritis and periodontitis are two prevalent chronic inflammatory diseases in humans and are associated with each other both clinically and epidemiologically. Recent findings suggest a causative link between periodontal infection and rheumatoid arthritis via bacteria-dependent induction of a pathogenic autoimmune response to citrullinated epitopes. Here we showed that infection with viable periodontal pathogen Porphyromonas gingivalis strain W83 exacerbated collagen-induced arthritis (CIA) in a mouse model, as manifested by earlier onset, accelerated progression and enhanced severity of the disease, including significantly increased bone and cartilage destruction. The ability of P. gingivalis to augment CIA was dependent on the expression of a unique P. gingivalis peptidylarginine deiminase (PPAD), which converts arginine residues in proteins to citrulline. Infection with wild type P. gingivalis was responsible for significantly increased levels of autoantibodies to collagen type II and citrullinated epitopes as a PPAD-null mutant did not elicit similar host response. High level of citrullinated proteins was also detected at the site of infection with wild-type P. gingivalis. Together, these results suggest bacterial PAD as the mechanistic link between P. gingivalis periodontal infection and rheumatoid arthritis.


Asunto(s)
Artritis/microbiología , Proteínas Bacterianas/metabolismo , Infecciones por Bacteroidaceae/microbiología , Modelos Animales de Enfermedad , Hidrolasas/metabolismo , Periodontitis/microbiología , Porphyromonas gingivalis/enzimología , Animales , Artritis/inmunología , Artritis/patología , Artritis/fisiopatología , Autoanticuerpos/análisis , Proteínas Bacterianas/genética , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/patología , Infecciones por Bacteroidaceae/fisiopatología , Resorción Ósea/etiología , Citrulina/metabolismo , Progresión de la Enfermedad , Eliminación de Gen , Hidrolasas/genética , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/microbiología , Articulaciones/patología , Masculino , Ratones Endogámicos DBA , Infiltración Neutrófila , Periodontitis/inmunología , Periodontitis/metabolismo , Periodontitis/patología , Porphyromonas gingivalis/inmunología , Porphyromonas gingivalis/aislamiento & purificación , Prevotella intermedia/enzimología , Prevotella intermedia/inmunología , Prevotella intermedia/aislamiento & purificación , Procesamiento Proteico-Postraduccional , Desiminasas de la Arginina Proteica , Índice de Severidad de la Enfermedad
6.
J Biol Chem ; 288(20): 14636-14646, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23546879

RESUMEN

The colonic epithelial surface is protected by an inner mucus layer that the commensal microflora cannot penetrate. We previously demonstrated that Entamoeba histolytica secretes a protease capable of dissolving this layer that is required for parasite penetration. Here, we asked whether there are bacteria that can secrete similar proteases. We screened bacterial culture supernatants for such activity using recombinant fragments of the MUC2 mucin, the major structural component, and the only gel-forming mucin in the colonic mucus. MUC2 has two central heavily O-glycosylated mucin domains that are protease-resistant and has cysteine-rich N and C termini responsible for polymerization. Culture supernatants of Porphyromonas gingivalis, a bacterium that secretes proteases responsible for periodontitis, cleaved the MUC2 C-terminal region, whereas the N-terminal region was unaffected. The active enzyme was isolated and identified as Arg-gingipain B (RgpB). Two cleavage sites were localized to IR↓TT and NR↓QA. IR↓TT cleavage will disrupt the MUC2 polymers. Because this site has two potential O-glycosylation sites, we tested whether recombinant GalNAc-transferases (GalNAc-Ts) could glycosylate a synthetic peptide covering the IRTT sequence. Only GalNAc-T3 was able to glycosylate the second Thr in IRTT, rendering the sequence resistant to cleavage by RgpB. Furthermore, when GalNAc-T3 was expressed in CHO cells expressing the MUC2 C terminus, the second threonine was glycosylated, and the protein became resistant to RgpB cleavage. These findings suggest that bacteria can produce proteases capable of dissolving the inner protective mucus layer by specific cleavages in the MUC2 mucin and that this cleavage can be modulated by site-specific O-glycosylation.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Mucina 2/metabolismo , Porphyromonas gingivalis/enzimología , Secuencia de Aminoácidos , Animales , Células CHO , Cromatografía , Colitis/microbiología , Colon/metabolismo , Cricetinae , Epitelio/metabolismo , Cisteína-Endopeptidasas Gingipaínas , Glicosilación , Humanos , Espectrometría de Masas , Datos de Secuencia Molecular
7.
Infect Immun ; 81(1): 55-64, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23090954

RESUMEN

Porphyromonas gingivalis is a Gram-negative bacterium associated with the development of periodontitis. The evolutionary success of this pathogen results directly from the presence of numerous virulence factors, including peptidylarginine deiminase (PPAD), an enzyme that converts arginine to citrulline in proteins and peptides. Such posttranslational modification is thought to affect the function of many different signaling molecules. Taking into account the importance of tissue remodeling and repair mechanisms for periodontal homeostasis, which are orchestrated by ligands of the epidermal growth factor receptor (EGFR), we investigated the ability of PPAD to distort cross talk between the epithelium and the epidermal growth factor (EGF) signaling pathway. We found that EGF preincubation with purified recombinant PPAD, or a wild-type strain of P. gingivalis, but not with a PPAD-deficient isogenic mutant, efficiently hindered the ability of the growth factor to stimulate epidermal cell proliferation and migration. In addition, PPAD abrogated EGFR-EGF interaction-dependent stimulation of expression of suppressor of cytokine signaling 3 and interferon regulatory factor 1. Biochemical analysis clearly showed that the PPAD-exerted effects on EGF activities were solely due to deimination of the C-terminal arginine. Interestingly, citrullination of two internal Arg residues with human endogenous peptidylarginine deiminases did not alter EFG function, arguing that the C-terminal arginine is essential for EGF biological activity. Cumulatively, these data suggest that the PPAD-activity-abrogating EGF function in gingival pockets may at least partially contribute to tissue damage and delayed healing within P. gingivalis-infected periodontia.


Asunto(s)
Infecciones por Bacteroidaceae/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Periodoncio/metabolismo , Periodoncio/microbiología , Porphyromonas gingivalis/metabolismo , Arginina/metabolismo , Infecciones por Bacteroidaceae/microbiología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Epitelio/metabolismo , Epitelio/microbiología , Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Hidrolasas/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Periodontitis/metabolismo , Periodontitis/microbiología , Desiminasas de la Arginina Proteica , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Cicatrización de Heridas
8.
Infect Immun ; 79(2): 797-805, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21098107

RESUMEN

Enhanced production of proinflammatory bradykinin-related peptides, the kinins, has been suggested to contribute to the pathogenesis of periodontitis, a common inflammatory disease of human gingival tissues. In this report, we describe a plausible mechanism of activation of the kinin-generating system, also known as the contact system or kininogen-kallikrein-kinin system, by the adsorption of its plasma-derived components such as high-molecular-mass kininogen (HK), prekallikrein (PK), and Hageman factor (FXII) to the cell surface of periodontal pathogen Porphyromonas gingivalis. The adsorption characteristics of mutant strains deficient in selected proteins of the cell envelope suggested that the surface-associated cysteine proteinases, gingipains, bearing hemagglutinin/adhesin domains (RgpA and Kgp) serve as the major platforms for HK and FXII adhesion. These interactions were confirmed by direct binding tests using microplate-immobilized gingipains and biotinylated contact factors. Other bacterial cell surface components such as fimbriae and lipopolysaccharide were also found to contribute to the binding of contact factors, particularly PK. Analysis of kinin release in plasma upon contact with P. gingivalis showed that the bacterial surface-dependent mechanism is complementary to the previously described kinin generation system dependent on HK and PK proteolytic activation by the gingipains. We also found that several P. gingivalis clinical isolates differed in the relative significance of these two mechanisms of kinin production. Taken together, these data show the importance of this specific type of bacterial surface-host homeostatic system interaction in periodontal infections.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Cininas/metabolismo , Proteínas de la Membrana/metabolismo , Porphyromonas gingivalis/metabolismo , Adhesinas Bacterianas/genética , Adsorción , Biotinilación , Membrana Celular , Cisteína Endopeptidasas/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Cisteína-Endopeptidasas Gingipaínas , Quininógenos/genética , Quininógenos/metabolismo , Cininas/genética , Proteínas de la Membrana/genética , Porphyromonas gingivalis/genética
9.
PLoS Pathog ; 5(5): e1000419, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19424422

RESUMEN

Infection, survival, and proliferation of pathogenic bacteria in humans depend on their capacity to impair host responses and acquire nutrients in a hostile environment. Among such nutrients is heme, a co-factor for oxygen storage, electron transport, photosynthesis, and redox biochemistry, which is indispensable for life. Porphyromonas gingivalis is the major human bacterial pathogen responsible for severe periodontitis. It recruits heme through HmuY, which sequesters heme from host carriers and delivers it to its cognate outer-membrane transporter, the TonB-dependent receptor HmuR. Here we report that heme binding does not significantly affect the secondary structure of HmuY. The crystal structure of heme-bound HmuY reveals a new all-beta fold mimicking a right hand. The thumb and fingers pinch heme iron through two apical histidine residues, giving rise to highly symmetric octahedral iron co-ordination. The tetrameric quaternary arrangement of the protein found in the crystal structure is consistent with experiments in solution. It shows that thumbs and fingertips, and, by extension, the bound heme groups, are shielded from competing heme-binding proteins from the host. This may also facilitate heme transport to HmuR for internalization. HmuY, both in its apo- and in its heme-bound forms, is resistant to proteolytic digestion by trypsin and the major secreted proteases of P. gingivalis, gingipains K and R. It is also stable against thermal and chemical denaturation. In conclusion, these studies reveal novel molecular properties of HmuY that are consistent with its role as a putative virulence factor during bacterial infection.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Portadoras/química , Hemoproteínas/química , Porphyromonas gingivalis/química , Factores de Virulencia/química , Proteínas Bacterianas/metabolismo , Infecciones por Bacteroidaceae , Proteínas Portadoras/metabolismo , Dicroismo Circular , Proteínas de Unión al Hemo , Hemoproteínas/metabolismo , Porphyromonas gingivalis/metabolismo , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Factores de Virulencia/metabolismo
10.
Anal Biochem ; 415(2): 158-67, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21569755

RESUMEN

Porphyromonas gingivalis is one of the major periodontitis-causing pathogens. P. gingivalis secretes a group of proteases termed gingipains, and in this study we have used the RgpB gingipain as a biomarker for P. gingivalis. We constructed a naive camel nanobody library and used phage display to select one nanobody toward RgpB with picomolar affinity. The nanobody was used in an inhibition assay for detection of RgpB in buffer as well as in saliva. The nanobody was highly specific for RgpB given that it did not bind to the homologous gingipain HRgpA. This indicated the presence of a binding epitope within the immunoglobulin-like domain of RgpB. A subtractive inhibition assay was used to demonstrate that the nanobody could bind native RgpB in the context of intact cells. The nanobody bound exclusively to the P. gingivalis membrane-bound RgpB isoform (mt-RgpB) and to secreted soluble RgpB. Further cross-reactivity studies with P. gingivalis gingipain deletion mutants showed that the nanobody could discriminate between native RgpB and native Kgp and RgpA in complex bacterial samples. This study demonstrates that RgpB can be used as a specific biomarker for P. gingivalis detection and that the presented nanobody-based assay could supplement existing methods for P. gingivalis detection.


Asunto(s)
Adhesinas Bacterianas/análisis , Anticuerpos Antibacterianos/inmunología , Infecciones por Bacteroidaceae/diagnóstico , Cisteína Endopeptidasas/análisis , Porphyromonas gingivalis/aislamiento & purificación , Anticuerpos de Cadena Única/inmunología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Secuencia de Aminoácidos , Infecciones por Bacteroidaceae/microbiología , Biomarcadores/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/inmunología , Cisteína-Endopeptidasas Gingipaínas , Humanos , Datos de Secuencia Molecular , Biblioteca de Péptidos , Porphyromonas gingivalis/enzimología , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saliva/microbiología
11.
Arthritis Rheum ; 62(9): 2662-72, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20506214

RESUMEN

OBJECTIVE: To investigate protein citrullination by the periodontal pathogen Porphyromonas gingivalis as a potential mechanism for breaking tolerance to citrullinated proteins in rheumatoid arthritis (RA). METHODS: The expression of endogenous citrullinated proteins was analyzed by immunoblotting of cell extracts from P gingivalis and 10 other oral bacteria. P gingivalis-knockout strains lacking the bacterial peptidylarginine deiminases (PADs) or gingipains were created to assess the role of these enzymes in citrullination. Citrullination of human fibrinogen and α-enolase by P gingivalis was studied by incubating live wild-type and knockout strains with the proteins and analyzing the products by immunoblotting and mass spectrometry. RESULTS: Endogenous protein citrullination was abundant in P gingivalis but lacking in the other oral bacteria. Deletion of the bacterial PAD gene resulted in complete abrogation of protein citrullination. Inactivation of arginine gingipains, but not lysine gingipains, led to decreased citrullination. Incubation of wild-type P gingivalis with fibrinogen or α-enolase caused degradation of the proteins and citrullination of the resulting peptides at carboxy-terminal arginine residues, which were identified by mass spectrometry. CONCLUSION: Our findings demonstrate that among the oral bacterial pathogens tested, P gingivalis is unique in its ability to citrullinate proteins. We further show that P gingivalis rapidly generates citrullinated host peptides by proteolytic cleavage at Arg-X peptide bonds by arginine gingipains, followed by citrullination of carboxy-terminal arginines by bacterial PAD. Our results suggest a novel model where P gingivalis-mediated citrullination of bacterial and host proteins provides a molecular mechanism for generating antigens that drive the autoimmune response in RA.


Asunto(s)
Artritis Reumatoide/microbiología , Biomarcadores de Tumor/metabolismo , Citrulina/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibrinógeno/metabolismo , Hidrolasas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Porphyromonas gingivalis/enzimología , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Artritis Reumatoide/inmunología , Autoinmunidad/inmunología , Cromatografía Líquida de Alta Presión , Citrulina/química , Fibrinógeno/química , Técnicas de Inactivación de Genes , Silenciador del Gen , Humanos , Hidrolasas/química , Datos de Secuencia Molecular , Organismos Modificados Genéticamente , Mapeo Peptídico , Periodontitis/inmunología , Periodontitis/microbiología , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidad , Desiminasas de la Arginina Proteica , Autotolerancia/genética , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
12.
J Immunol ; 183(6): 3700-11, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19687097

RESUMEN

Porphyromonas gingivalis, a Gram-negative bacterium that causes periodontitis, activates the kinin system via the cysteine protease R-gingipain. Using a model of buccal infection based on P. gingivalis inoculation in the anterior mandibular vestibule, we studied whether kinins released by gingipain may link mucosal inflammation to T cell-dependent immunity through the activation of bradykinin B(2) receptors (B(2)R). Our data show that P. gingivalis W83 (wild type), but not gingipain-deficient mutant or wild-type bacteria pretreated with gingipain inhibitors, elicited buccal edema and gingivitis in BALB/c or C57BL/6 mice. Studies in TLR2(-/-), B(2)R(-/-), and neutrophil-depleted C57BL/6 mice revealed that P. gingivalis induced edema through the sequential activation of TLR2/neutrophils, with the initial plasma leakage being amplified by gingipain-dependent release of vasoactive kinins from plasma-borne kininogens. We then used fimbriae (Fim) Ag as a readout to verify whether activation of the TLR2-->PMN-->B(2)R axis (where PMN is polymorphonuclear neutrophil) at early stages of mucosal infection had impact on adaptive immunity. Analyzes of T cell recall responses indicated that gingipain drives B(2)R-dependent generation of IFN-gamma-producing Fim T cells in submandibular draining lymph nodes of BALB/c and C57BL/6 mice, whereas IL-17-producing Fim T cells were generated only in BALB/c mice. In summary, our studies suggest that two virulence factors, LPS (an atypical TLR2 ligand) and gingipain, forge a trans-cellular cross-talk between TLR2 and B(2)R, thus forming an innate axis that guides the development of Fim-specific T cells in mice challenged intrabuccally by P. gingivalis. Ongoing research may clarify whether kinin-driven modulation of T cell responses may also influence the severity of chronic periodontitis.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Fimbrias Bacterianas/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Cininas/metabolismo , Porphyromonas gingivalis/inmunología , Receptor de Bradiquinina B2/metabolismo , Linfocitos T/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Cisteína-Endopeptidasas Gingipaínas , Inmunidad , Inflamación , Ratones , Mucosa Bucal/microbiología , Mucosa Bucal/patología , Péptido Hidrolasas , Transducción de Señal , Linfocitos T/inmunología
13.
Photodiagnosis Photodyn Ther ; 34: 102330, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33965605

RESUMEN

BACKGROUND: Porphyromonas gingivalis possess the ability to invade host cells which prevents this pathogen from eradication by conventional periodontal therapy. Recently, antimicrobial photodynamic therapy (aPDT) was introduced to periodontal treatment as a complementary antibacterial method. The aim of this study was to evaluate the effect of toluidine blue-O (TBO) mediated aPDT on the viability of P. gingivalis invading gingival fibroblasts and keratinocytes in an in vitro model of infection. METHODS: Primary human gingival fibroblasts (PHGF) and telomerase immortalized gingival keratinocytes (TIGK) were infected with Pg ATCC 33277. Two concentrations of TBO (0.01 mg/mL, TBO-c1 and 0.001 mg/mL, TBO-c2) and a non-laser red light source (λ = 630 nm) were applied to treat both cell-adherent/intracellular Pg (the adhesion/invasion model) or exclusively the intracellular bacteria (the intracellular infection model). RESULTS: The median viability of cell-adherent/intracellular Pg in infected keratinocytes declined from 1.88 × 105 cfu/mL in infected cells treated with TBO without irradiation to 40 cfu/mL upon irradiation for 10 s with TBO-c1. At higher light doses a complete photokilling of P. gingivalis was observed. Pg from exclusively intracellular infection model was also efficiently eradicated as the residual viability dropped from 1.44 × 105 cfu/mL in control samples to 160, 20 and 10 cfu/mL upon irradiation for 10, 20 and 30 s, respectively. In the infected fibroblasts irradiation significantly reduced bacterial viability but did not completely eradicate the intracellular pathogen. CONCLUSIONS: Antimicrobial PDT is effective in reducing the viability of intracellular periopathogens, however those residing within gingival fibroblasts seems to attenuate the photokilling effectiveness of this method.


Asunto(s)
Antiinfecciosos , Fotoquimioterapia , Antibacterianos , Fibroblastos , Humanos , Queratinocitos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Porphyromonas gingivalis
14.
Front Immunol ; 8: 353, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28424689

RESUMEN

Periodontal inflammation is one of the most common chronic inflammatory conditions in humans. Despite recent advances in identifying and characterizing oral microbiota dysbiosis in the pathogenesis of gum disease, just how host factors maintain a healthy homeostatic oral microbial community or prevent the development of a pathogenic oral microbiota remains poorly understood. An important determinant of microbiota fate is local antimicrobial proteins. Here, we report that chemoattractant protein chemerin, which we recently identified as a potent endogenous antimicrobial agent in body barriers such as the skin, is present in the oral cavity under homeostatic and inflammatory conditions. Chemerin and a chemerin-derived antimicrobial peptide are bactericidal against select bacteria strategically positioned in dental biofilm. Gingival crevicular samples from patients with gingivitis but not periodontitis contain abundant bioactive chemerin capable of inducing CMKLR1-dependent leukocyte migration. Gingipains secreted by the periodontopathogen P. gingivalis inactivate chemerin. Together, these data suggest that as an antimicrobial agent and leukocyte chemoattractant, chemerin likely contributes to antimicrobial immune defense in the oral cavity.

15.
Sci Rep ; 5: 11969, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26132828

RESUMEN

Citrullination is a post-translational modification of higher organisms that deiminates arginines in proteins and peptides. It occurs in physiological processes but also pathologies such as multiple sclerosis, fibrosis, Alzheimer's disease and rheumatoid arthritis (RA). The reaction is catalyzed by peptidylarginine deiminases (PADs), which are found in vertebrates but not in lower organisms. RA has been epidemiologically associated with periodontal disease, whose main infective agent is Porphyromonas gingivalis. Uniquely among microbes, P. gingivalis secretes a PAD, termed PPAD (Porphyromonas peptidylarginine deiminase), which is genetically unrelated to eukaryotic PADs. Here, we studied function of PPAD and its substrate-free, substrate-complex, and substrate-mimic-complex structures. It comprises a flat cylindrical catalytic domain with five-fold α/ß-propeller architecture and a C-terminal immunoglobulin-like domain. The PPAD active site is a funnel located on one of the cylinder bases. It accommodates arginines from peptide substrates after major rearrangement of a "Michaelis loop" that closes the cleft. The guanidinium and carboxylate groups of substrates are tightly bound, which explains activity of PPAD against arginines at C-termini but not within peptides. Catalysis is based on a cysteine-histidine-asparagine triad, which is shared with human PAD1-PAD4 and other guanidino-group modifying enzymes. We provide a working mechanism hypothesis based on 18 structure-derived point mutants.


Asunto(s)
Proteínas Bacterianas/química , Hidrolasas/química , Porphyromonas gingivalis/enzimología , Factores de Virulencia/química , Dominio Catalítico , Citrulina/química , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Desiminasas de la Arginina Proteica , Homología Estructural de Proteína
16.
Curr Protein Pept Sci ; 4(6): 397-407, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14683426

RESUMEN

Gingipains, extracellular cysteine proteinases of Porphyromonas gingivalis, constitute the major virulence factor of this periodontopathogenic bacterium. They are the product of three genes, two coding for an Arg-specific (RgpA and RgpB) and one for a Lys-specific proteinase (Kgp). Proteinase domains of RgpA and RgpB are virtually identical; however, the gene encoding the former enzyme is missing a large segment coding for hemaglutinin / adhesin (HA) domains. The latter domains are present also in Kgp. The tertiary structure of RgpB revealed that the proteinase domain of gingipains has a protein fold referred to as the caspase-hemoglobinase fold. On this basis, they are also evolutionary related to other highly specific proteinases including clostripain, caspases, legumains and separase (clan CD of cysteine peptidases). Gingipains are produced as large preproproteins and are subject to elaborate, not yet fully understood, secretion, glycosylation, activation, and maturation processes. How they traverse the outer membrane is unknown, although it can be hypothesized that they use an autotransporter pathway. Apparently during transport through the periplasm the LPS-like glycan moiety is added at the conserved C-terminal portion of progingipains. At the cell surface pro-gingipains fold into partially active, single-chain zymogens and undergo autocatalytic, intermolecular processing. Two sequential cleavages within the profragment domain enhance zymogen activity and in the case of RgpA and Kgp are followed by excision of the individual HA domains. These domains are further truncated at the C-terminus by concerted action of Kgp and carboxypeptidase and form a non-covalent multidomain, multifunctional complex anchored into the outer membrane by the glycated, C-terminal HA domain. This hypothetical scenario is a reasonable explanation for the occurrence of many forms of gingipains.


Asunto(s)
Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Hemaglutininas/química , Hemaglutininas/metabolismo , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/patogenicidad , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Adhesinas Bacterianas , Cisteína Endopeptidasas/genética , Activación Enzimática , Cisteína-Endopeptidasas Gingipaínas , Hemaglutininas/genética , Porphyromonas gingivalis/genética , Conformación Proteica , Procesamiento Proteico-Postraduccional , Factores de Virulencia/genética
17.
J Periodontol ; 82(7): 1051-60, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21235330

RESUMEN

BACKGROUND: The analysis of samplings from periodontal pockets is important in the diagnosis and therapy of periodontitis. In this study, three different sampling techniques were compared to determine whether one method yielded samples suitable for the reproducible and simultaneous determination of bacterial load, cytokines, neutrophil elastase, and arginine-specific gingipains (Rgps). Rgps are an important virulence factor of Porphyromonas gingivalis, the exact concentration of which in gingival crevicular fluid (GCF) has not been quantified. METHODS: GCF was sampled from four sites per patient (one sample per quadrant using two samples per method) in 36 patients with chronic periodontitis. One week later, the procedure was repeated with alternative methods. Variables determined were loads of Aggregatibacter actinomycetemcomitans (previously Actinobacillus actinomycetemcomitans) and P. gingivalis, levels of interleukin-6 and -8, activity of neutrophil elastase, and level of Rgps. RESULTS: The detected cytokine levels were higher using paper strips compared to paper points. Bacteria were found in similar loads from paper strips and paper points. Rgps were only detectable in high quantities by washing the periodontal pocket. The level of Rgps correlated with the load of P. gingivalis. CONCLUSIONS: The use of paper strips was suitable for the simultaneous determination of microbial and immunologic parameters. Obtaining GCF by washing can be useful for special purposes. The gingipain concentration in periodontal pockets was directly determined to be ≤1.5 µM. This value indicated that most of the substrates of these proteases by in vitro assays identified until now can be easily degraded in P. gingivalis-infected sites.


Asunto(s)
Periodontitis Crónica/metabolismo , Líquido del Surco Gingival/química , Manejo de Especímenes/métodos , Adhesinas Bacterianas/análisis , Adulto , Aggregatibacter actinomycetemcomitans/aislamiento & purificación , Carga Bacteriana , Cisteína Endopeptidasas/análisis , Femenino , Cisteína-Endopeptidasas Gingipaínas , Hemorragia Gingival/metabolismo , Hemaglutininas/análisis , Humanos , Interleucina-6/análisis , Interleucina-8/análisis , Elastasa de Leucocito/análisis , Masculino , Persona de Mediana Edad , Papel , Pérdida de la Inserción Periodontal/metabolismo , Bolsa Periodontal/metabolismo , Porphyromonas gingivalis/aislamiento & purificación , Porphyromonas gingivalis/patogenicidad , Manejo de Especímenes/instrumentación , Irrigación Terapéutica/métodos , Factores de Virulencia/análisis
18.
PLoS One ; 6(2): e17182, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21390208

RESUMEN

Haem (iron protoporphyrin IX) is both an essential growth factor and virulence regulator for the periodontal pathogen Porphyromonas gingivalis, which acquires it mainly from haemoglobin via the sequential actions of the R- and K-specific gingipain proteases. The haem-binding lipoprotein haemophore HmuY and its cognate receptor HmuR of P. gingivalis, are responsible for capture and internalisation of haem. This study examined the role of the HmuY in acquisition of haem from haemoglobin and the cooperation between HmuY and gingipain proteases in this process. Using UV-visible spectroscopy and polyacrylamide gel electrophoresis, HmuY was demonstrated to wrest haem from immobilised methaemoglobin and deoxyhaemoglobin. Haem extraction from oxyhaemoglobin was facilitated after oxidation to methaemoglobin by pre-treatment with the P. gingivalis R-gingipain A (HRgpA). HmuY was also capable of scavenging haem from oxyhaemoglobin pre-treated with the K-gingipain (Kgp). This is the first demonstration of a haemophore working in conjunction with proteases to acquire haem from haemoglobin. In addition, HmuY was able to extract haem from methaemalbumin, and could bind haem, either free in solution or from methaemoglobin, even in the presence of serum albumin.


Asunto(s)
Adhesinas Bacterianas/fisiología , Cisteína Endopeptidasas/fisiología , Hemo/metabolismo , Péptido Hidrolasas/metabolismo , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/metabolismo , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Electroforesis , Fenómenos Electrofisiológicos , Cisteína-Endopeptidasas Gingipaínas , Hemo/química , Hemoglobinas/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Oxihemoglobinas/metabolismo , Péptido Hidrolasas/química , Péptido Hidrolasas/fisiología , Porphyromonas gingivalis/química , Unión Proteica/efectos de los fármacos , Protoporfirinas/metabolismo , Albúmina Sérica/farmacología
19.
Microbiology (Reading) ; 155(Pt 2): 328-337, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19202082

RESUMEN

PorT is a membrane-associated protein shown to be essential for the maturation and secretion of a class of cysteine proteinases, the gingipains, from the periodontal pathogen Porphyromonas gingivalis. It was previously reported that PorT is located on the periplasmic surface of the inner membrane to function as a chaperone for the maturing proteinases. Our modelling suggested it to be an integral outer-membrane protein with eight anti-parallel, membrane-traversing beta-strands. In this report, the outer-membrane localization model was confirmed by the structural and functional tolerance of PorT to hexahistidine (6xHis) tag insertions at selected locations within the protein using site-directed mutagenesis. Interestingly, those PorT mutations adversely affecting gingipain secretion enhanced expression of the porT gene but at the same time suppressed the transcription of the gingipain rgpB gene. Further, PorT mutants deficient in gingipain activities produced significantly more di- and triaminopeptidase activities. PorT homologues have been found in restricted members of the Bacteroidetes phylum where there is potential for PorT to participate in the maturation and secretion of proteins with characteristic C-terminal domains (CTDs). Knowledge of the cellular localization of PorT will enable analysis of the role of this protein in a new secretory pathway for the export of gingipains and other CTD-class proteins.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Porphyromonas gingivalis/química , Porphyromonas gingivalis/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Mutagénesis Insercional , Mutación , Porphyromonas gingivalis/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas
20.
Arch Microbiol ; 189(3): 197-210, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17922109

RESUMEN

Porphyromonas gingivalis HmuY is a putative heme-binding lipoprotein associated with the outer membrane. It is part of an operon together with a gene encoding an outer-membrane hemin utilization receptor (HmuR) and four uncharacterized genes. A similar operon organization was found in Bacteroides fragilis and B. thetaiotaomicron, with the former containing an additional HmuY homologue encoded upstream of the hmuR-like gene. In P. gingivalis cultured under heme-limited conditions, a approximately 1-kb hmuY transcript was produced at high levels along with some approximately 3.5 and approximately 9-kb transcripts. Compared with the parental strain, mutants deficient in hmuY or hmuR or hmuY-hmuR gene function grew more slowly and bound lower amounts of hemin and hemoglobin. Significantly, they grew more slowly or were unable to grow when human serum was used as the sole iron/heme source. Analysis of the hmu promoter showed that it is regulated by iron. The HmuY protein normally occurs as a homodimer, but in the presence of hemin it may form tetramers. These results show that HmuY may be the first reported member of a new class of proteins in Porphyromonas and Bacteroides species involved in heme utilization, a function being exerted in conjunction with HmuR, an outer-membrane heme transporter.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemo/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Porphyromonas gingivalis/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bacteroides/genética , ADN Bacteriano/química , ADN Bacteriano/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Orden Génico , Hemina/metabolismo , Hemoglobinas , Humanos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Datos de Secuencia Molecular , Mutagénesis Insercional , Operón , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/crecimiento & desarrollo , Regiones Promotoras Genéticas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Suero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA