Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biosci Rep ; 39(3)2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30842338

RESUMEN

There is a strong association between periodontal disease and atherosclerotic cardiovascular disorders. A key event in the development of atherosclerosis is accumulation of modified lipoproteins within the arterial wall. We hypothesise that patients with periodontitis have an altered lipoprotein profile towards an atherogenic form. Therefore, the present study aims at identifying modifications of plasma lipoproteins in periodontitis. Lipoproteins from ten female patients with periodontitis and gender- and age-matched healthy controls were isolated by density-gradient ultracentrifugation. Proteins were separated by 2D gel-electrophoresis and identified by map-matching or by nano-LC followed by MS. Apolipoprotein (Apo) A-I (ApoA-I) methionine oxidation, Oxyblot, total antioxidant capacity and a multiplex of 71 inflammation-related plasma proteins were assessed. Reduced levels of apoJ, phospholipid transfer protein, apoF, complement C3, paraoxonase 3 and increased levels of α-1-antichymotrypsin, apoA-II, apoC-III were found in high-density lipoprotein (HDL) from the patients. In low-density lipoprotein (LDL)/very LDL (VLDL), the levels of apoL-1 and platelet-activating factor acetylhydrolase (PAF-AH) as well as apo-B fragments were increased. Methionine oxidation of apoA-I was increased in HDL and showed a relationship with periodontal parameters. α-1 antitrypsin and α-2-HS glycoprotein were oxidised in LDL/VLDL and antioxidant capacity was increased in the patient group. A total of 17 inflammation-related proteins were important for group separation with the highest discriminating proteins identified as IL-21, Fractalkine, IL-17F, IL-7, IL-1RA and IL-2. Patients with periodontitis have an altered plasma lipoprotein profile, defined by altered protein levels as well as post-translational and other structural modifications towards an atherogenic form, which supports a role of modified plasma lipoproteins as central in the link between periodontal and cardiovascular disease (CVD).


Asunto(s)
Aterosclerosis/sangre , Enfermedades Cardiovasculares/sangre , Lipoproteínas/sangre , Periodontitis/sangre , Apolipoproteína A-I , Apolipoproteínas B/sangre , Citocinas/sangre , Electroforesis en Gel Bidimensional , Femenino , Humanos , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Lipoproteínas VLDL/sangre , Espectrometría de Masas/métodos , Persona de Mediana Edad
2.
Microbiologyopen ; 7(6): e00606, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29536668

RESUMEN

Antimicrobial resistance needs to be tackled from new angles, and antimicrobial peptides could be future candidates for combating bacterial infections. This study aims to investigate in vitro the bactericidal effects of the lantibiotic gallidermin on Staphylococcus epidermidis and Staphylococcus aureus, possible cytotoxic effects and its impact on host-microbe interactions. Minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of gallidermin were determined, and cytotoxicity and proinflammatory effects of gallidermin on fibroblasts, red blood cells (RBCs) and in whole blood were investigated. Both MIC and MBC for all four tested strains of S. epidermidis was 6.25 µg/ml. Both MIC and MBC for methicillin-sensitive S. aureus was 12.5 µg/ml and for methicillin-resistant S. aureus (MRSA) 1.56 µg/ml. Gallidermin displayed no cytotoxic effects on fibroblasts, only a high dose of gallidermin induced low levels of CXCL8 and interleukin-6. Gallidermin hemolyzed less than 1% of human RBCs, and did not induce reactive oxygen species production or cell aggregation in whole blood. In cell culture, gallidermin inhibited the cytotoxic effects of the bacteria and totally suppressed the bacteria-induced release of CXCL8 and interleukin-6 from fibroblasts. We demonstrate that gallidermin, expressing low cell cytotoxicity, is a promising candidate for treating bacterial infections caused by S. epidermidis and S. aureus, especially MRSA.


Asunto(s)
Antibacterianos/farmacología , Bacteriocinas/farmacología , Dermis/microbiología , Fibroblastos/inmunología , Péptidos/farmacología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus epidermidis/efectos de los fármacos , Adulto , Células Cultivadas , Dermis/inmunología , Fibroblastos/microbiología , Humanos , Interleucina-6/inmunología , Interleucina-8/inmunología , Masculino , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/fisiología
3.
Biomed Mater ; 13(2): 025014, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29047451

RESUMEN

Wound dressings based on bacterial cellulose (BC) can form a soft and conformable protective layer that can stimulate wound healing while preventing bacteria from entering the wound. Bacteria already present in the wound can, however, thrive in the moist environment created by the BC dressing which can aggravate the healing process. Possibilities to render the BC antimicrobial without affecting the beneficial structural and mechanical properties of the material would hence be highly attractive. Here we present methods for functionalization of BC with ε-poly-L-Lysine (ε-PLL), a non-toxic biopolymer with broad-spectrum antimicrobial activity. Low molecular weight ε-PLL was cross-linked in pristine BC membranes and to carboxymethyl cellulose functionalized BC using carbodiimide chemistry. The functionalization of BC with ε-PLL inhibited growth of S. epidermidis on the membranes but did not affect the cytocompatibility to cultured human fibroblasts as compared to native BC. The functionalization had no significant effects on the nanofibrous structure and mechanical properties of the BC. The possibility to functionalize BC with ε-PLL is a promising, green and versatile approach to improve the performance of BC in wound care and other biomedical applications.


Asunto(s)
Antibacterianos/química , Vendajes , Celulosa/química , Fibroblastos/efectos de los fármacos , Polilisina/química , Cicatrización de Heridas/efectos de los fármacos , Adsorción , Aminas/química , Biopolímeros/química , Fibroblastos/metabolismo , Humanos , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Nanofibras/química , Reproducibilidad de los Resultados , Reología , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Staphylococcus epidermidis/efectos de los fármacos , Estrés Mecánico
4.
APMIS ; 125(2): 157-169, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28120492

RESUMEN

Porphyromonas gingivalis, which is considered a keystone agent in periodontitis, has evolved elaborate mechanisms to grow and survive in a hostile milieu. The gingival fibroblast is the major cell type in the gingiva and is considered to be important in the periodontitis-associated inflammation. As a part of the innate immune response, they produce cytokines such as CXCL8 and interleukin (IL)-6 which are believed to contribute to the destruction of the tooth-supporting tissues. This study investigates how the expression of protease-activated receptors (PAR1, PAR2) and toll-like receptors (TLR2, TLR4) changes with P. gingivalis exposure and how silencing of one receptor affects the expression of the other receptors. The importance of protein kinase C (PKC) and p38 in the regulation of CXCL8 and IL-6 was also examined. Receptors were knockdown with small-interfering RNA. PKC or p38 was blocked prior to stimulation with P. gingivalis. Fibroblasts were able to compensate for PAR1 knockdown with increased expression of PAR2. PKC and p38 were involved in the regulation of P. gingivalis-induced CXCL8 and IL-6. Our results indicate that PAR1 and PAR2 could be implicated in periodontitis and that PKC and P38 play a role in the inflammatory response in P. gingivalis-infected gingival fibroblasts.


Asunto(s)
Fibroblastos/inmunología , Fibroblastos/microbiología , Porphyromonas gingivalis/inmunología , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Western Blotting , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteína Quinasa C/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Cytokine ; 76(2): 424-432, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26318255

RESUMEN

Porphyromonas gingivalis is a periodontitis-associated pathogen and interactions between the bacterium and gingival fibroblasts play an important role in development and progression of periodontitis, an inflammatory disease leading to degeneration of tooth-supporting structures. Gingival fibroblasts, which expresses protease activated receptors (PARs) as well as toll-like receptors (TLRs), produces inflammatory mediators upon bacterial challenges. In this study, we elucidated the importance of PAR1, PAR2, TLR2 and TLR4 for the expression and secretion of CXCL8, interleukin-6 (IL-6), transforming growth factor-ß1 (TGF-ß1) and secretory leukocyte inhibitor (SLPI). Human gingival fibroblasts were transfected with small-interfering RNA against the target genes, and then stimulated with P. gingivalis wild-type W50 and W50-derived double rgp mutant E8 and kgp mutant K1A. TLR2-silencing reduced P. gingivalis-induced CXCL8 and IL-6. IL-6 was also reduced after PAR1-silencing. No effects were observed for TGF-ß1. SLPI was suppressed by P. gingivalis and silencing of PAR1 as well as TLR2, gave additional suppression at the mRNA level. TLR4 was not involved in the regulation of the investigated mediators. CXCL8 and IL-6 are important for progression and development of periodontitis, leading to a chronic inflammation that may contribute to the tissue destruction that follows an exacerbated host response. Therefore, regulating the expression of TLR2 and subsequent release of CXCL8 and IL-6 in periodontitis could attenuate the tissue destruction seen in periodontitis.


Asunto(s)
Citocinas/metabolismo , Encía/metabolismo , Porphyromonas gingivalis/fisiología , Receptores Proteinasa-Activados/fisiología , Receptores Toll-Like/fisiología , Células Cultivadas , Fibroblastos/metabolismo , Encía/citología , Encía/microbiología , Humanos
6.
J Periodontol ; 85(1): 113-22, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23594192

RESUMEN

BACKGROUND: High levels of hepatocyte growth factor (HGF), a healing factor with regenerative and cytoprotective effects, are associated with inflammatory diseases, including periodontitis. HGF biologic activity requires binding to its receptors, the proto-oncogene c-Met and heparan sulfate proteoglycan (HSPG). This study investigates HGF expression and its relationship to subgingival microbiota in medically healthy individuals with and without periodontitis. METHODS: Saliva, gingival crevicular fluid (GCF), and blood samples from 30 patients with severe periodontitis and 30 healthy controls were analyzed for HGF concentration using enzyme-linked immunosorbent assay and binding affinity for HSPG and c-Met using surface plasmon resonance. The regenerative effects of saliva from three patients and controls were analyzed in an in vitro model of cell injury. Subgingival plaques were analyzed for the presence of 18 bacterial species. RESULTS: Patients with periodontitis showed higher HGF concentrations in saliva, GCF, and serum (P <0.001); however, the binding affinities for HSPG and c-Met were reduced in GCF and saliva (P <0.002). In contrast to the controls, saliva from patients showed no significant regenerative effect over time on gingival epithelial cells. Compared with controls, patients had a higher prevalence of periodontally related bacteria. CONCLUSIONS: Higher circulatory HGF levels indicate a systemic effect of periodontitis. However, the HGF biologic activity at local inflammation sites was reduced, and this effect was associated with the amount of periodontal bacteria. Loss of function of healing factors may be an important mechanism in degenerative processes in periodontally susceptible individuals.


Asunto(s)
Factor de Crecimiento de Hepatocito/análisis , Periodontitis/metabolismo , Adulto , Anciano , Bacterias/clasificación , Carga Bacteriana , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Movimiento Celular/efectos de los fármacos , Placa Dental/microbiología , Células Epiteliales/efectos de los fármacos , Femenino , Encía/microbiología , Líquido del Surco Gingival/química , Proteoglicanos de Heparán Sulfato/metabolismo , Factor de Crecimiento de Hepatocito/sangre , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Índice Periodontal , Bolsa Periodontal/clasificación , Periodontitis/sangre , Periodontitis/microbiología , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/metabolismo , Saliva/química , Resonancia por Plasmón de Superficie
7.
BMC Microbiol ; 13: 155, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23841502

RESUMEN

BACKGROUND: Porphyromonas gingivalis is a key pathogen in periodontitis, an inflammatory disease leading to destruction of bone and tooth-supporting tissue. P. gingivalis possesses a number of pathogenic properties to enhance growth and survival, including proteolytic gingipains. Accumulating data shows that gingipains are involved in the regulation of host inflammatory responses. The aim of this study was to determine if P. gingivalis infection modulates the inflammatory response of fibroblasts, including the release of chemokines and cytokines. Human gingival fibroblasts or primary dermal fibroblasts were pre-stimulated with tumor-necrosis factor-α (TNF- α) and cocultured with P. gingivalis. Gingipain inhibitors were used to explore the effect of gingipains. CXCL8 levels were determined with ELISA and the relative levels of various inflammatory mediators were determined by a cytokine assay. RESULTS: TNF-α-triggered CXCL8 levels were completely abolished by viable P. gingivalis, whereas heat-killed P. gingivalis did not suppress CXCL8. Accumulation of CXCL8 was partially restored by an arginine-gingipain inhibitor. Furthermore, fibroblasts produced several inflammatory mediators, notably chemokines, all of which were suppressed by viable P. gingivalis. CONCLUSION: These findings provide evidence that fibroblast-derived inflammatory signals are modulated by heat-instable gingipains, whereby the bacteria can escape killing by the host immune system and promote its own growth and establishment. In addition, we show that fibroblasts are important mediators of inflammation in response to infection and thereby play a crucial role in determining the nature and magnitude of the invasion of immune cells.


Asunto(s)
Fibroblastos/inmunología , Fibroblastos/microbiología , Evasión Inmune , Porphyromonas gingivalis/inmunología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Interleucina-8/inmunología , Viabilidad Microbiana , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...