Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Protein Expr Purif ; 178: 105782, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33122039

RESUMEN

Streptococcus pneumoniae is a gram-positive bacterial pathogen causing invasive pneumonia, meningitis, otitis media, and bacteremia. Owing to the current pitfalls of polysaccharide and polysaccharide-conjugate vaccines, protein vaccines are considered promising candidates against pneumonia. Pneumococcal surface protein A (PspA) and pneumococcal surface adhesin A (PsaA) are virulence proteins showing good immunogenicity and protective effects against S. pneumoniae strains in mice. In this study, we expressed the fusion protein PsaA-PspA, which consists of PsaA and the N-terminal region of PspA family 1 and 2, in Escherichia coli. We describe a novel and effective method to purify PsaA-PspA using hydroxyapatite and two-step chromatography. After determining the optimal induction conditions and a series of purification steps, we obtained PsaA-PspA fusion protein with over 95% purity at a final yield of 22.44% from the starting cell lysate. The molecular weight of PsaA-PspA was approximately 83.6 kDa and its secondary structure was evaluated by circular dichroism. Immunization with the purified protein induced high levels of IgG antibodies in mice. Collectively, these results demonstrate that our purification method can effectively produce high-purity PsaA-PspA fusion protein with biological activity and chemical integrity, which can be widely applied to the purification of other PspA subclass proteins.


Asunto(s)
Adhesinas Bacterianas , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión , Streptococcus pneumoniae/inmunología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/aislamiento & purificación , Adhesinas Bacterianas/farmacología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/farmacología , Escherichia coli , Femenino , Expresión Génica , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología
2.
J Periodontal Res ; 53(5): 762-776, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29777544

RESUMEN

BACKGROUND AND OBJECTIVE: The aim of this study was to explore the cellular mechanisms underlying gingipain-caused changes in cell morphology and apoptosis of osteoblasts. MATERIAL AND METHODS: Human calvarial osteoblasts and mouse osteoblasts MC3T3-E1 were treated with gingipain extracts from Porphyromonas gingivalis stain W83. Apoptosis was detected with annexin V and propidium iodide flow cytometry analysis or terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling staining. F-actin was determined by immunostaining. Western blotting was used to detect protein expression. Knocking down and overexpressing approaches were used to determine the role of integrin ß1. RESULTS: Osteoblasts exposed to gingipain extracts displayed increased apoptosis, accompanied by loss of F-actin integrity and cell shrinkage. The effects of gingipain extracts were abolished by the cysteine protease inhibitor N-tosyl-l-lysyl chloromethyl-ketone. Notably, gingipain extracts resulted in reduction of integrin ß1, accompanied by diminished active RhoA whereas without effect on the total RhoA. Knockdown of integrin ß1 resembled those seen in gingipain-treated osteoblasts. By contrast, the effects of gingipain extracts were abrogated by either overexpression of integrin ß1 or presence of RhoA agonist CN03. CONCLUSION: Gingipain-induced F-actin disruption and apoptosis are mediated by the degradation of integrin ß1 and inhibition of RhoA activity, which account for osteoblast apoptosis.


Asunto(s)
Actinas/metabolismo , Adhesinas Bacterianas/farmacología , Apoptosis/efectos de los fármacos , Cisteína Endopeptidasas/farmacología , Integrina beta1/metabolismo , Osteoblastos/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Citometría de Flujo , Cisteína-Endopeptidasas Gingipaínas , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Confocal , Osteoblastos/citología , Cráneo/citología , Transfección
3.
Eur J Oral Sci ; 126(5): 359-366, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30126013

RESUMEN

Bacterial metabolism during infection is related to bacterial persistence and virulence factors. Porphyromonas gingivalis is a key pathogen that contributes to chronic periodontitis. Our previous study showed that pckA, the gene encoding phosphoenolpyruvate carboxykinase, is a putative-specific pathogenic gene of virulent strains of P. gingivalis. Here, a pckA-deficient strain (ΔPG1676) was constructed in P. gingivalis W83. Virulence properties were compared between the mutant and wild-type strains. Specifically, hemagglutination activity was determined by the ability to agglutinate sheep erythrocytes. Gingipain activity was detected using synthetic-specific substrates. Gene expression levels were analyzed using RT-qPCR, and cell surface-associated polysaccharides were examined by silver staining and electron microscopy. Inactivation of the pckA gene did not affect bacterial growth and lipopolysaccharide formation but led to a reduction in hemagglutination activity and downregulation in expression of the hemagglutination-associated gene, rfa, when compared with the wild-type strain. Additionally, the ΔPG1676 mutant exhibited an alteration in the distribution of gingipain activity. Increased gingipain activity was detected on the cell surface, but a decrease in its activity in the culture supernatant was shown. Taken together, our results suggest that the pckA gene plays a role in modulating the virulence of P. gingivalis W83.


Asunto(s)
Adhesinas Bacterianas/farmacología , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/farmacología , Genes Bacterianos/genética , Hemaglutinación , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/metabolismo , Factores de Virulencia/genética , Animales , Errores Innatos del Metabolismo de los Carbohidratos/genética , Periodontitis Crónica/microbiología , Regulación hacia Abajo , Regulación Bacteriana de la Expresión Génica , Cisteína-Endopeptidasas Gingipaínas , Lipopolisacáridos/aislamiento & purificación , Hepatopatías/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/deficiencia , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Porphyromonas gingivalis/crecimiento & desarrollo , Porphyromonas gingivalis/patogenicidad , Eliminación de Secuencia , Ovinos , Especificidad por Sustrato , Transcriptoma , Virulencia/genética
4.
Mol Cell Probes ; 33: 51-56, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28302392

RESUMEN

OBJECTIVE: The present study aimed to explore the possible effects of osteopontin (OPN) in the proliferation of rat aortic smooth muscle cells (RASMCs) stimulated by gingipains. METHODS: The proliferation of RASMCs in response to active gingipains treatment was evaluated by CCK-8 assay. OPN siRNA was designed, constructed and transfected into RASMCs at different concentrations. The cell cycle of RASMCs was analyzed by flow cytometry. OPN, α-SMA and calponin expression were examined by real-time PCR and western blot analysis. RESULTS: Gingipains promoted the proliferation of RASMCs and OPN expression. With siRNA-mediated OPN expression knockdown, the cell cycle of RASMCs was blocked in the G0/G1 phase. Furthermore, the expression of specific differentiation markers, α-SMA and calponin, also decreased. CONCLUSIONS: These results demonstrate that OPN has an impact on the proliferation and differentiation of RASMCs stimulated by gingipains.


Asunto(s)
Adhesinas Bacterianas/farmacología , Proliferación Celular/efectos de los fármacos , Cisteína Endopeptidasas/farmacología , Osteopontina/genética , Animales , Aorta/efectos de los fármacos , Diferenciación Celular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Cisteína-Endopeptidasas Gingipaínas , Miocitos del Músculo Liso/efectos de los fármacos , Osteopontina/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Ratas
5.
J Cell Sci ; 127(Pt 20): 4420-8, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25146392

RESUMEN

Mesenchymal cell condensation is the initiating event in endochondral bone formation. Cell condensation is followed by differentiation into chondrocytes, which is accompanied by induction of chondrogenic gene expression. Gene mutations involved in chondrogenesis cause chondrodysplasias and other skeletal defects. Using mesenchymal stem cells (MSCs) in an in vitro chondrogenesis assay, we found that knockdown of the diastrophic dysplasia (DTD) sulfate transporter (DTDST, also known as SLC26A2), which is required for normal cartilage development, blocked cell condensation and caused a significant reduction in fibronectin matrix. Knockdown of fibronectin with small interfering RNAs (siRNAs) also blocked condensation. Fibrillar fibronectin matrix was detected prior to cell condensation, and its levels increased during and after condensation. Inhibition of fibronectin matrix assembly by use of the functional upstream domain (FUD) of adhesin F1 from Streptococcus pyogenes prevented cell condensation by MSCs and also by the chondrogenic cell line ATDC5. Our data show that cell condensation and induction of chondrogenesis depend on fibronectin matrix assembly and DTDST, and indicate that this transporter is required earlier in chondrogenesis than previously appreciated. They also raise the possibility that certain of the skeletal defects in DTD patients might derive from the link between DTDST, fibronectin matrix and condensation.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Condrocitos/fisiología , Condrogénesis , Fibronectinas/metabolismo , Células Madre Mesenquimatosas/fisiología , Adhesinas Bacterianas/farmacología , Proteínas de Transporte de Anión/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Enanismo/genética , Enanismo/metabolismo , Fibronectinas/antagonistas & inhibidores , Fibronectinas/genética , Humanos , ARN Interferente Pequeño/genética , Transportadores de Sulfato
6.
Exp Eye Res ; 143: 39-48, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26500187

RESUMEN

Dissecting the interactions between Pseudomonas aeruginosa and corneal cells is important to identify a novel target for prevention and treatment of Pseudomonas keratitis. The current study began with a peptide identified by phage display, and was to investigate the protective efficacy against P. aeruginosa infection in cornea. The original peptide Pc-E, with high homology to a hypothetical membrane protein (HmpA) in P. aeruginosa, and the derived peptide Pc-EP, with the same sequence as a region in HmpA, were synthesized. Peptide Pc-EP could directly bind to HCEC, stronger than Pc-E, and specifically activate toll-like receptor 5, and thereby significantly induce the production of pro-inflammatory factors, such as IL-1ß, IL-6, IFN-γ and IL-17. Moreover, Pc-EP could act as an antagonist to inhibit the adhesion of wild-type P. aeruginosa to HCEC and mouse corneas. No inhibitory effect was observed on the adhesion of the strain loss of HmpA. When compared to the wild-type strain, the adhesion of the hmpA mutant to corneal cells was significantly decreased. Treatment of infected mouse corneas with Pc-EP before infection significantly decreased the bacterial load in the cornea and attenuated the corneal pathology. These results indicate that Pc-EP can be a useful prophylactic agent for P. aeruginosa keratitis.


Asunto(s)
Adhesinas Bacterianas/farmacología , Úlcera de la Córnea/prevención & control , Infecciones Bacterianas del Ojo/prevención & control , Lectinas/farmacología , Péptidos/farmacología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/fisiología , Animales , Adhesión Bacteriana/efectos de los fármacos , Carga Bacteriana , Secuencia de Bases , Células Cultivadas , Recuento de Colonia Microbiana , Úlcera de la Córnea/microbiología , Ensayo de Inmunoadsorción Enzimática , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/metabolismo , Epitelio Corneal/microbiología , Infecciones Bacterianas del Ojo/microbiología , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Interleucina-17 , Lectinas/síntesis química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Péptidos/síntesis química , Infecciones por Pseudomonas/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 5/genética
7.
Eur J Immunol ; 44(11): 3403-12, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25130613

RESUMEN

Post-translational modifications of proteins significantly affect their structure and function. The carbamylation of positively charged lysine residues to form neutral homoitrulline occurs primarily under inflammatory conditions through myeloperoxidase-dependent cyanate (CNO-) formation. We analyzed the pattern of human IgG1 carbamylation under inflammatory conditions and the effects that this modification has on the ability of antibodies to trigger complement activation via the classical pathway. We found that the lysine residues of IgG1 are rapidly modified after brief exposure to CNO- . Interestingly, modifications were not random, but instead limited to only few lysines within the hinge area and the N-terminal fragment of the CH2 domain. A complement activation assay combined with mass spectrometry analysis revealed a highly significant inverse correlation between carbamylation of several key lysine residues within the hinge region and N-terminus of the CH2 domain and the proper binding of C1q to human IgG1 followed by subsequent complement activation. This severely hindered complement-dependent cytotoxicity of therapeutic IgG1 . The reaction can apparently occur in vivo, as we found carbamylated antibodies in synovial fluid from rheumatoid arthritis patients. Taken together, our data suggest that carbamylation has a profound impact on the complement-activating ability of IgG1 and reveals a pivotal role for previously uncharacterized lysine residues in this process.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Citrulina/análogos & derivados , Activación de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Inmunoglobulina G/inmunología , Adhesinas Bacterianas/farmacología , Anticuerpos Monoclonales de Origen Murino/farmacología , Artritis Reumatoide/inmunología , Citrulina/biosíntesis , Complemento C1q/inmunología , Cianatos/metabolismo , Cisteína Endopeptidasas/farmacología , Cisteína-Endopeptidasas Gingipaínas , Humanos , Inmunoglobulina G/metabolismo , Factores Inmunológicos/farmacología , Inflamación/inmunología , Lisina/química , Lisina/metabolismo , Espectrometría de Masas , Unión Proteica/inmunología , Procesamiento Proteico-Postraduccional , Rituximab
8.
EMBO J ; 27(20): 2669-80, 2008 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-18833195

RESUMEN

We identified a new bacterial transporter, the Pseudomonas aeruginosa CupB3 protein, which is an outer membrane usher involved in pili assembly. In CupB3, the usher domain has fused during evolution with a POTRA (polypeptide-transport-associated)-like domain found in TpsB transporters of two-partner secretion systems. In TpsBs, the POTRA captures the TpsA passenger, which is then transported across the outer membrane through the TpsB beta-barrel. We named CupB3 a 'P-usher' for POTRA-like domain-containing usher. We showed that CupB3 assembles CupB1 fimbrial subunits into pili and secretes CupB5, a TpsA-like protein. The CupB3 usher domain has the function of a TpsB beta-barrel in CupB5 translocation. We revealed that the POTRA-like domain is neither essential for CupB1 fimbriae assembly nor for cell surface exposition of CupB5, but is crucial to coordinate bona fide transport of CupB1 and CupB5 through the usher domain. The P-usher defines a novel transport pathway involving a molecular machine made with old spare parts.


Asunto(s)
Adhesinas Bacterianas/química , Fimbrias Bacterianas/metabolismo , Proteínas de Transporte de Membrana/fisiología , Pseudomonas aeruginosa/fisiología , Adhesinas Bacterianas/farmacología , Secuencia de Aminoácidos , Transporte Biológico , Membrana Celular/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Modelos Biológicos , Modelos Genéticos , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Pseudomonas aeruginosa/metabolismo , Homología de Secuencia de Aminoácido
9.
FASEB J ; 25(10): 3741-50, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21768393

RESUMEN

Degradation of immunoglobulins is an effective strategy of bacteria to evade the immune system. We have tested whether human IgG is a substrate for gingipain K of Porphyromonas gingivalis and found that the enzyme can hydrolyze subclass 1 and 3 of human IgG. The heavy chain of IgG(1) was cleaved at a single site within the hinge region, generating Fab and Fc fragments. IgG(3) was also cleaved within the heavy chain, but at several sites around the CH2 region. Investigation of the enzyme kinetics of IgG proteolysis by gingipain K, using FPLC- and isothermal titration calorimetry-based assays followed by Hill plots, revealed non-Michaelis-Menten kinetics involving a mechanism of positive cooperativity. In ex vivo studies, it was shown that gingipain K retained its IgG hydrolyzing activity in human plasma despite the high content of natural protease inhibitors; that IgG(1) cleavage products were detected in gingival crevicular fluid samples from patients with severe periodontitis; and that gingipain K treatment of serum samples from patients with high antibody titers against P. gingivalis significantly hindered opsonin-dependent phagocytosis of clinical isolates of P. gingivalis by neutrophils. Altogether, these findings underline a biological function of gingipain K as an IgG protease of pathophysiological importance.


Asunto(s)
Adhesinas Bacterianas/farmacología , Cisteína Endopeptidasas/farmacología , Inmunoglobulina G/metabolismo , Periodontitis/patología , Porphyromonas gingivalis/metabolismo , Adhesinas Bacterianas/metabolismo , Secuencia de Aminoácidos , Cisteína Endopeptidasas/metabolismo , Cisteína-Endopeptidasas Gingipaínas , Interacciones Huésped-Patógeno , Humanos , Datos de Secuencia Molecular , Periodontitis/microbiología
10.
J Dairy Sci ; 95(6): 3318-26, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22612965

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a major cause of intestinal disease and hemolytic uremic syndrome, a serious systemic complication that particularly affects children. Cattle are primary reservoirs for EHEC O157:H7 and the main source of infection for humans. Vaccination of cattle with different combinations of bacterial virulence factors has shown efficacy in decreasing EHEC O157:H7 shedding. It is, therefore, important to demonstrate whether vaccination of pregnant cows with EHEC O157:H7 induces high titers of transferable antibodies to avoid early colonization of calves by the bacteria. In this study we evaluated the ability of EspA, EspB, the C-terminal fragment of 280 amino acids of γ-intimin (γ-intimin C280) and inactivated Shiga toxin (Stx) 2 proteins to induce specific antibodies in colostrum and their passive transference to colostrum-fed calves. Friesian pregnant cows immunized by the intramuscular route mounted significantly high serum and colostrum IgG responses against EspB and γ-intimin C280 that were efficiently transferred to their calves. Antibodies to EspB and γ-intimin C280 were detected in milk samples of vaccinated cows at d 40 postparturition. Significant Stx2-neutralizing titers were also observed in colostrum from Stx2-vaccinated cows and sera from colostrum-fed calves. The results presented showed that bovine colostrum with increased levels of antibodies against EHEC O157:H7 may be obtained by systemic immunization of pregnant cows, and that these specific antibodies are efficiently transferred to newborn calves by feeding colostrum. Hyperimmune colostrum and milk may be an alternative to protect calves from early colonization by EHEC O157:H7 and a possible key source of antibodies to block colonization and toxic activity of this bacterium.


Asunto(s)
Adhesinas Bacterianas/farmacología , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/farmacología , Bovinos/inmunología , Calostro/inmunología , Escherichia coli O157/inmunología , Proteínas de Escherichia coli/farmacología , Inmunidad Materno-Adquirida/inmunología , Toxina Shiga II/farmacología , Vacunación/veterinaria , Adhesinas Bacterianas/inmunología , Animales , Animales Recién Nacidos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de Escherichia coli/inmunología , Femenino , Embarazo , Toxina Shiga II/inmunología
11.
Arch Oral Biol ; 140: 105453, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35580388

RESUMEN

OBJECTIVE: The aim was to determine the changes of inflammatory mediator expression in human macrophages stimulated with outer membrane vesicles purified from Porphyromonas gingivalis. DESIGN: outer membrane vesicles purified by ultracentrifugation from ATCC 33277 and W83 P. gingivalis strains were used for stimulating human macrophages and determine their inflammatory mediator expression changes. U937 monocyte cells line were differentiated into macrophages and stimulated with outer membrane vesicles for 30 min and six hours. In Independent experiments, the outer membrane vesicles and viable bacteria control were pre-treated with the gingipain inhibitors KYT-1 and KYT-36 (Arg-gingipain and Lys-gingipain, respectively) or Polymyxin-B to block the lipopolysaccharide activity to evaluate the secretion changes of immune mediators IL-1ß, IL-6, TNF-α, IL-8, MCP-1, MIP-1α and RANTES by flow cytometry. A factorial ANOVA was used to analyze the data. RESULTS: The outer membrane vesicles of P. gingivalis ATCC 33277 displayed higher Arg-gingipain activity than those obtained from the P. gingivalis W83 strain (0.6 U/µg vs. 0.46 U/µg). Although the outer membrane vesicles of P. gingivalis stimulated the production of cytokines and chemokines, specific Arg-gingipain and Lys-gingipain inhibition induced significant increases in IL-1ß, IL-6, IL-8, MCP-1, and RANTES levels, and this induction was significantly greater at 6 h compared to 30 min (*p < 0.05). On the contrary, TNF-α secretion decreased when gingipains were blocked. CONCLUSIONS: outer membrane vesicles may play a dual role during P. gingivalis infection based on their ability to induce changes in the immune responses of human macrophages, probably via gingipain-dependent events.


Asunto(s)
Quimiocina CCL5 , Porphyromonas gingivalis , Adhesinas Bacterianas/metabolismo , Adhesinas Bacterianas/farmacología , Quimiocina CCL5/metabolismo , Cisteína Endopeptidasas/metabolismo , Citocinas/metabolismo , Cisteína-Endopeptidasas Gingipaínas , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macrófagos/metabolismo , Porphyromonas gingivalis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
12.
Mol Microbiol ; 76(4): 861-73, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20233299

RESUMEN

Porphyromonas gingivalis is an obligately anaerobic bacterium recognized as an aetiological agent of adult periodontitis. P. gingivalis produces cysteine proteinases, the gingipains. The crystal structure of a domain within the haemagglutinin region of the lysine gingipain (Kgp) is reported here. The domain was named K2 as it is the second of three homologous structural modules in Kgp. The K2 domain structure is a 'jelly-roll' fold with two anti-parallel beta-sheets. This fold topology is shared with adhesive domains from functionally diverse receptors such as MAM domains, ephrin receptor ligand binding domains and a number of carbohydrate binding modules. Possible functions of K2 were investigated. K2 induced haemolysis of erythrocytes in a dose-dependent manner that was augmented by the blocking of anion transport. Further, cysteine-activated arginine gingipain RgpB, which degrades glycophorin A, sensitized erythrocytes to the haemolytic effect of K2. Cleaved K2, similar to that found in extracted Kgp, lacks the haemolytic activity indicating that autolysis of Kgp may be a staged process which is artificially enhanced by extraction of the protein. The data indicate a functional role for K2 in the integrated capacity conferred by Kgp to enable the porphyrin auxotroph P. gingivalis to capture essential haem from erythrocytes.


Asunto(s)
Adhesinas Bacterianas/química , Cisteína Endopeptidasas/química , Proteínas Hemolisinas/química , Hemólisis , Porphyromonas gingivalis/enzimología , Adhesinas Bacterianas/farmacología , Adhesinas Bacterianas/fisiología , Secuencia de Aminoácidos , Células Cultivadas , Cristalografía por Rayos X , Cisteína Endopeptidasas/farmacología , Cisteína Endopeptidasas/fisiología , Eritrocitos/efectos de los fármacos , Eritrocitos/patología , Cisteína-Endopeptidasas Gingipaínas , Proteínas Hemolisinas/farmacología , Proteínas Hemolisinas/fisiología , Humanos , Lisina/química , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
13.
Zhonghua Nei Ke Za Zhi ; 50(10): 863-7, 2011 Oct.
Artículo en Zh | MEDLINE | ID: mdl-22321330

RESUMEN

OBJECTIVE: To investigate the protection effect of bifidobacterial adhesin for intestine ischemia/reperfusion (I/R) injury on gut barrier function in rat. METHODS: Seventy-two male SD rats were randomly divided into sham operation group (n = 24), I/R model group (n = 24) and pretreatment group of bifidobacterial adhesin (pretreatment group, n = 24). Six rats were anatomized at 6 h, 1 d, 4 d and 7d after inducing I/R model in each group, respectively. The pathological changes of the terminal ilea and the blood levels of TNFα, IL-6, IL-10, diamine oxidase (DAO), and the activity and content of D-lactic acid were observed. RESULTS: The blood levels of TNFα, IL-6, DAO and D-lactic acid in I/R model group were significantly higher than sham operation group at all time points (P < 0.05), while the blood level of IL-10 was no significantly change. The activity of IL-6 and DAO in pretreatment group was significantly lower than I/R model group at all time points (P < 0.05), the blood level of TNFα in pretreatment group was significantly lower than I/R model group at 1 d, the blood level of D-lactic was significantly lower than I/R model group at 4 d and 7 d (P < 0.05). Intestinal pathological damages were obviously milder in pretreatment group than I/R model group at all time points (Chiu's pathological scores: 6 h, 3.22 ± 0.22 vs 3.57 ± 0.20; 1 d, 3.77 ± 0.13 vs 3.90 ± 0.12; 4 d, 2.93 ± 0.23 vs 3.07 ± 0.21; 7 d, 2.10 ± 0.30 vs 2.22 ± 0.17, all P < 0.05). CONCLUSION: The pretreatment of bifidobacterial adhesin could protect the intestinal mucosa from I/R injury, and alleviate intestinal ischemic reperfusion injury.


Asunto(s)
Adhesinas Bacterianas/farmacología , Bifidobacterium , Mucosa Intestinal/efectos de los fármacos , Intestinos/patología , Daño por Reperfusión/prevención & control , Animales , D-Aminoácido Oxidasa/sangre , Interleucina-10/sangre , Interleucina-6/sangre , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Precondicionamiento Isquémico/métodos , Ácido Láctico/sangre , Masculino , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/sangre
14.
J Periodontal Res ; 45(6): 772-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20682013

RESUMEN

BACKGROUND AND OBJECTIVE: Periodontitis is a chronic inflammatory disease of the periodontium that causes significant alveolar bone loss. Osteoclasts are bone-resorbing multinucleated cells. Osteoblasts regulate osteoclast differentiation by expression of RANKL and osteoprotegerin (OPG). Td92 is a surface-exposed outer membrane protein of Treponema denticola, a periodontopathogen. Although it has been demonstrated that Td92 acts as a stimulator of various proinflammatory mediators, the role of Td92 in alveolar bone resorption remains unclear. Therefore, in this study, we investigated the role of Td92 in bone resorption. MATERIAL AND METHODS: Mouse bone marrow cells were co-cultured with calvariae-derived osteoblasts in the presence or absence of Td92. Osteoclast formation was assessed by TRAP staining. Expressions of RANKL, osteoprotegerin (OPG) and prostaglandin E(2) (PGE(2) ) in osteoblasts were estimated by ELISA. RESULTS: Td92 induced osteoclast formation in the co-cultures. In the osteoblasts, RANKL and PGE(2) expressions were up-regulated, whereas OPG expression was down-regulated by Td92. The addition of OPG inhibited Td92-induced osteoclast formation. The prostaglandin synthesis inhibitors NS398 and indomethacin were also shown to inhibit Td92-induced osteoclast formation. The effects of Td92 on the expressions of RANKL, OPG and PGE(2) in osteoblasts were blocked by NS398 or indomethacin. CONCLUSION: These results suggest that Td92 promotes osteoclast formation through the regulation of RANKL and OPG production via a PGE(2) -dependent mechanism.


Asunto(s)
Adhesinas Bacterianas/fisiología , Pérdida de Hueso Alveolar/metabolismo , Dinoprostona/metabolismo , Osteoclastos/fisiología , Osteoprotegerina/biosíntesis , Ligando RANK/biosíntesis , Treponema denticola/química , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/farmacología , Pérdida de Hueso Alveolar/microbiología , Animales , Células de la Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos , Osteoblastos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoprotegerina/genética , Ligando RANK/genética , Proteínas Recombinantes/farmacología , Treponema denticola/fisiología
15.
Biochem J ; 419(1): 159-66, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19102726

RESUMEN

Porphyromonas gingivalis is one of the major pathogens of periodontitis, a condition characterized by excessive alveolar bone resorption by osteoclasts. The bacterium produces cysteine proteases called gingipains, which are classified according to their cleavage-site specificity into Kgps (lysine-specific gingipains) and Rgps (arginine-specific gingipains). In the present study we examined the effects of gingipains on osteoclast differentiation. In co-cultures of mouse bone-marrow cells and osteoblasts, formation of multinucleated osteoclasts induced by 1alpha,25(OH)(2)D(3) (1alpha,25-dihydroxyvitamin D(3)) was augmented by Kgp but not by RgpB. A physiological concentration (0.1 nM) of 1alpha,25(OH)(2)D(3) induced the osteoclast formation in the presence of 100 nM Kgp to an extent comparable with that induced by 10 nM 1alpha,25(OH)(2)D(3). Kgp also enhanced osteoclastogenesis induced by various microbial components, including lipopolysaccharide. Combined use of Kgp and 1alpha,25(OH)(2)D(3) or lipopolysaccharide also increased the number of resorption pits developed on dentin slices, indicating that the osteoclasts formed in the presence of Kgp possess bone-resorbing activity. The enhanced osteoclastogenesis by Kgp was correlated with a depletion of osteoprotegerin in co-culture medium and was proteolytic-activity-dependent, since benzyloxycarbonyl-L-phenylalanyl-L-lysylacycloxyketone, an inhibitor of Kgp, completely abolished osteoclastogenesis induced by Kgp. Kgp digested osteoprotegerin, since its recombinant protein was susceptible to degradation by Kgp in the presence of serum. As a result, Kgp did not augment osteoclastogenesis in co-cultures of osteoprotegerin-deficient osteoblasts and bone-marrow cells. In addition, enhanced osteoclastogenesis by Kgp was abolished by an excess amount of recombinant osteoprotegerin. These findings suggest that degradation of osteoprotegerin is one of the mechanisms underlying promotion of osteoclastogenesis by Kgp.


Asunto(s)
Adhesinas Bacterianas/farmacología , Colecalciferol/farmacología , Cisteína Endopeptidasas/farmacología , Lipopolisacáridos/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cisteína-Endopeptidasas Gingipaínas , Ratones , Ratones Mutantes , Osteoclastos/citología , Estabilidad Proteica/efectos de los fármacos
16.
Oral Microbiol Immunol ; 24(5): 396-400, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19702953

RESUMEN

INTRODUCTION: Human cells express membrane-bound complement regulatory proteins to prevent complement-mediated autologous tissue damage. In this study, we hypothesized that Porphyromonas gingivalis, the major etiological agent of chronic periodontitis, causes the shedding or proteolysis of the complement regulatory protein CD46 expressed by oral epithelial cells. METHODS: Oral epithelial cells were treated with a culture of P. gingivalis before measurement of membrane-bound and shed CD46 by enzyme-linked immunosorbent assay (ELISA). The effect of soluble recombinant CD46 on secretion of interleukin-8 (IL-8) by epithelial cells was evaluated by ELISA. The susceptibility of soluble recombinant CD46 to proteolytic degradation by cells and purified Lys-gingipain of P. gingivalis was investigated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis/western immunoblotting analysis. RESULTS: Oral epithelial cells treated with a culture of P. gingivalis showed a lower reactivity with antibodies directed to CD46. ELISA revealed that such a treatment resulted in increased amounts of CD46 in the conditioned media suggesting that P. gingivalis caused the shedding of membrane-anchored CD46. Stimulation of epithelial cells with soluble recombinant CD46 induced IL-8 secretion in a dose-dependent manner. Whole cells and purified Lys-gingipain of P. gingivalis degraded recombinant CD46 in a dose-dependent manner. CONCLUSION: This study showed the ability of P. gingivalis to induce the shedding/ proteolysis of CD46 from the surface of oral epithelial cells. This may render host cells susceptible to the complement system and contribute to tissue damage and the inflammatory process in periodontitis.


Asunto(s)
Proteína Cofactora de Membrana/metabolismo , Mucosa Bucal/inmunología , Porphyromonas gingivalis/fisiología , Adhesinas Bacterianas/farmacología , Anticuerpos Antibacterianos/análisis , Western Blotting , Línea Celular Tumoral , Membrana Celular/inmunología , Medios de Cultivo Condicionados , Cisteína Endopeptidasas/farmacología , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Cisteína-Endopeptidasas Gingipaínas , Hemaglutininas/farmacología , Humanos , Interleucina-8/metabolismo , Proteínas de la Membrana/inmunología , Mucosa Bucal/microbiología
17.
J Periodontal Res ; 44(4): 565-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18717779

RESUMEN

BACKGROUND AND OBJECTIVE: Porphyromonas gingivalis is a major aetiological agent in the development of periodontitis, the major clinical hallmark of which is bone resorption. The cysteine proteases (gingipains) produced by P. gingivalis have a critical role in the pathogenesis of the disease, and previous studies on whole bacteria have implicated these enzymes in osteoclastogenesis, a process which serves to upregulate bone resorption. The effects of the gingipains from P. gingivalis on osteoclast differentiation were investigated here to determine whether the enzymes directly contribute to osteoclastogenesis and thus to bone resorption. MATERIAL AND METHODS: The effects of the gingipains on osteoclast differentiation were investigated in primary mouse bone marrow cultures. The cultures harvested from C57BL6/J mice were incubated in the presence of parathyroid hormone, a known osteoclastogenic factor, or active/inactivated forms of three gingipains. Osteoclast differentiation was quantified by counting the number of multinucleated cells positive for tartrate-resistant acid phosphatase, an enzyme marker for these cells. RESULTS: After 10 days of culture, the gingipains, either active or inactive, failed to stimulate osteoclast differentiation in comparison to the parathyroid hormone. CONCLUSION: The data presented here demonstrate that the gingipains do not induce osteoclast differentiation in this system, indicating that the bacterium uses other mechanisms to induce bone loss.


Asunto(s)
Adhesinas Bacterianas/farmacología , Células de la Médula Ósea/efectos de los fármacos , Cisteína Endopeptidasas/farmacología , Hemaglutininas/farmacología , Osteoclastos/efectos de los fármacos , Porphyromonas gingivalis/fisiología , Fosfatasa Ácida/análisis , Animales , Resorción Ósea/patología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cisteína-Endopeptidasas Gingipaínas , Humanos , Isoenzimas/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Fosfatasa Ácida Tartratorresistente
18.
J Periodontol ; 80(9): 1511-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19722803

RESUMEN

BACKGROUND: The endothelial cell surface glycoprotein thrombomodulin (TM) inhibits vascular coagulation and inflammation via regulation of thrombin-mediated activation of protein C. Porphyromonas gingivalis is the major periodontopathic bacterium and has been found in vessel walls and atherosclerotic lesions in humans. P. gingivalis-derived cysteine proteases (gingipains) are known to enhance inflammatory and coagulant responses of vascular endothelial cells. However, it has not been elucidated whether gingipains affect vascular endothelial TM. METHODS: Purified arginine-specific gingipains (Rgps) and lysine-specific gingipain (Kgp) from P. gingivalis were used to investigate the effects of gingipains on recombinant human TM by immunoblot analyses. Flow cytometry and activated protein C assay were carried out to examine the effects of gingipains on vascular endothelial cell surface TM. Immunohistochemistry was performed to investigate TM expression in microvascular endothelia in gingival tissues taken from patients with periodontitis. RESULTS: Rgps and Kgp cleaved TM in vitro. Endothelial cell surface TM was also degraded by Rgps. Thrombin-mediated activation of protein C was reduced by Rgps through TM inactivation. Gingival microvascular endothelial TM was reduced in patients with periodontitis. CONCLUSIONS: P. gingivalis gingipains induced the degradation and inactivation of endothelial TM, which may promote vascular coagulation and inflammation. In addition, in vivo relevance was demonstrated by reduced expression of TM in gingival microvascular endothelia in patients with periodontitis, which may be involved in the pathogenesis of periodontitis.


Asunto(s)
Adhesinas Bacterianas/farmacología , Cisteína Endopeptidasas/farmacología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Hemaglutininas/farmacología , Porphyromonas gingivalis/enzimología , Trombomodulina/efectos de los fármacos , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Femenino , Citometría de Flujo , Cisteína-Endopeptidasas Gingipaínas , Encía/irrigación sanguínea , Gingivitis/patología , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Microvasos/patología , Persona de Mediana Edad , Pérdida de la Inserción Periodontal/patología , Bolsa Periodontal/patología , Periodontitis/patología , Proteína C/análisis , Trombomodulina/análisis
19.
Mol Membr Biol ; 25(6-7): 498-505, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18949626

RESUMEN

The electrophysiological technique of patch-clamp was used to characterize the pore properties of site-directed mutants in the Vibrio cholerae general diffusion porin OmpU. Changes in conductance and selectivity were observed, thus confirming the predicted pore location of these residues, based on homology with the Escherichia coli porins OmpF and OmpC. Some mutants acquire a weak selectivity for cations, which mirrors the properties of the homologous, deoxycholic acid sensitive, OmpT porin of V. cholerae. However, the mutants remain insensitive to deoxycholic acid, like wildtype OmpU. This result suggests that channel selectivity is not an important determinant in the sensitivity to this drug, and is in agreement with our finding that the neutral deoxycholic acid, and not deoxycholate, is the actual active form in channel block. Modifications in the kinetics of spontaneous closures were also noted, and are similar to those found for the E. coli channels. In addition, mutants at the D116 residue on the L3 loop display marked transitions to sub-conductance states. The results reported here are compared to a phenotypical characterization of the mutants in terms of permeability to maltodextrins and beta-lactam antibiotic sensitivity. No strict correlations are observed, suggesting that distinct, but somewhat overlapping, molecular determinants control electrophysiological properties and substrate permeability.


Asunto(s)
Adhesinas Bacterianas/genética , Adhesinas Bacterianas/farmacología , Mutación , Vibrio cholerae/química , Ácido Desoxicólico/farmacología , Electrofisiología , Cinética , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp , Permeabilidad/efectos de los fármacos , Polisacáridos/farmacología , Porosidad , Vibrio cholerae/patogenicidad , Resistencia betalactámica
20.
Infect Immun ; 76(1): 317-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18025101

RESUMEN

Porphyromonas gingivalis, a gram-negative anaerobe which is implicated in the etiology of active periodontitis, secretes degradative enzymes (gingipains) and sheds proinflammatory mediators (e.g., lipopolysaccharides [LPS]). LPS triggers the secretion of interleukin-8 (IL-8) from immune (72-amino-acid [aa] variant [IL-8(72aa)]) and nonimmune (IL-8(77aa)) cells. IL-8(77aa) has low chemotactic and respiratory burst-inducing activity but is susceptible to cleavage by gingipains. This study shows that both R- and K-gingipain treatments of IL-8(77aa) significantly enhance burst activation by fMLP and chemotactic activity (P < 0.05) but decrease burst activation and chemotactic activity of IL-8(72aa) toward neutrophil-like HL60 cells and primary neutrophils (P < 0.05). Using tandem mass spectrometry, we have demonstrated that R-gingipain cleaves 5- and 11-aa peptides from the N-terminal portion of IL-8(77aa) and the resultant peptides are biologically active, while K-gingipain removes an 8-aa N-terminal peptide yielding a 69-aa isoform of IL-8 that shows enhanced biological activity. During periodontitis, secreted gingipains may differentially affect neutrophil chemotaxis and activation in response to IL-8 according to the cellular source of the chemokine.


Asunto(s)
Adhesinas Bacterianas/farmacología , Quimiotaxis/fisiología , Cisteína Endopeptidasas/farmacología , Interleucina-8/genética , Interleucina-8/metabolismo , Porphyromonas gingivalis/metabolismo , Estallido Respiratorio/fisiología , Expresión Génica , Cisteína-Endopeptidasas Gingipaínas , Células HL-60 , Humanos , Activación Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA