Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Cell Microbiol ; 22(8): e13209, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32329166

RESUMEN

The host-microbe relationship is pivotal for oral health as well as for peri-implant diseases. Peri-implant mucosa and commensal biofilm play important roles in the maintenance of host-microbe homeostasis, but little is known about how they interact. We have therefore investigated the early host-microbe interaction between commensal multispecies biofilm (Streptococcus oralis, Actinomyces naeslundii, Veillonella dispar, Porphyromonas gingivalis) and organotypic peri-implant mucosa using our three-dimensional model. After 24 hr, biofilms induced weak inflammatory reaction in the peri-implant mucosa by upregulation of five genes related to immune response and increased secretion of IL-6 and CCL20. Biofilm volume was reduced which might be explained by secretion of ß-Defensins-1, -2, and CCL20. The specific tissue reaction without intrinsic overreaction might contribute to intact mucosa. Thus, a relationship similar to homeostasis and oral health was established within the first 24 hr. In contrast, the mucosa was damaged and the bacterial distribution was altered after 48 hr. These were accompanied by an enhanced immune response with upregulation of additional inflammatory-related genes and increased cytokine secretion. Thus, the homeostasis-like relationship was disrupted. Such profound knowledge of the host-microbe interaction at the peri-implant site may provide the basis to improve strategies for prevention and therapy of peri-implant diseases.


Asunto(s)
Biopelículas , Fibroblastos/microbiología , Interacciones Microbiota-Huesped , Modelos Anatómicos , Mucosa Bucal/microbiología , Actinomyces/fisiología , Citocinas/inmunología , Fibroblastos/inmunología , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Mucosa Bucal/inmunología , Porphyromonas gingivalis/inmunología , Porphyromonas gingivalis/fisiología , Veillonella/inmunología , Veillonella/fisiología
2.
Cell Microbiol ; 21(10): e13078, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31270923

RESUMEN

The impact of oral commensal and pathogenic bacteria on peri-implant mucosa is not well understood, despite the high prevalence of peri-implant infections. Hence, we investigated responses of the peri-implant mucosa to Streptococcus oralis or Aggregatibacter actinomycetemcomitans biofilms using a novel in vitro peri-implant mucosa-biofilm model. Our 3D model combined three components, organotypic oral mucosa, implant material, and oral biofilm, with structural assembly close to native situation. S. oralis induced a protective stress response in the peri-implant mucosa through upregulation of heat shock protein (HSP70) genes. Attenuated inflammatory response was indicated by reduced cytokine levels of interleukin-6 (IL-6), interleukin-8 (CXCL8), and monocyte chemoattractant protein-1 (CCL2). The inflammatory balance was preserved through increased levels of tumor necrosis factor-alpha (TNF-α). A. actinomycetemcomitans induced downregulation of genes important for cell survival and host inflammatory response. The reduced cytokine levels of chemokine ligand 1 (CXCL1), CXCL8, and CCL2 also indicated a diminished inflammatory response. The induced immune balance by S. oralis may support oral health, whereas the reduced inflammatory response to A. actinomycetemcomitans may provide colonisation advantage and facilitate later tissue invasion. The comprehensive characterisation of peri-implant mucosa-biofilm interactions using our 3D model can provide new knowledge to improve strategies for prevention and therapy of peri-implant disease.


Asunto(s)
Aggregatibacter actinomycetemcomitans/fisiología , Biopelículas/crecimiento & desarrollo , Modelos Inmunológicos , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Periimplantitis/inmunología , Streptococcus oralis/fisiología , Aggregatibacter actinomycetemcomitans/patogenicidad , Células Cultivadas , Quimiocina CCL2/metabolismo , Implantes Dentales/efectos adversos , Implantes Dentales/microbiología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Periimplantitis/microbiología , Periimplantitis/patología , Infecciones Relacionadas con Prótesis/inmunología , Titanio/química , Factor de Necrosis Tumoral alfa/metabolismo
3.
Biomedicines ; 10(2)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35203495

RESUMEN

Cytokine profiles are often perturbed after infections of medical implants. With a non-invasive in vivo imaging system, we report in a mouse model that interferon expression after infection of subcutaneous implants with Streptococcus oralis, Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, and Treponema denticola (alone or as a combination) was species-specific, persisted longer in the presence of implants, and notably decreased upon dual species infections. This type I interferon expression disappeared within two weeks; however, histology of implant-tissue interface indicated high recruitment of immune cells even after three weeks. This was suggestive that biomaterial-associated infections could have prolonged effects, including the systemic stimulation of inflammatory cytokines. The present study investigated the systemic impact of this chronic peri-implant inflammation on the systemic expression of inflammatory cytokines (23) using a multiplex assay. Initially, the cytokine measurement in murine fibroblasts exposed to periodontal pathogens remained limited to the expression of five cytokines, namely, IL-6, G-CSF, CXCL-1/KC, MCP-1 (MCAF), and IL-12 (p40). The systemic determination of cytokines in mice increased to 19 cytokines (IL-1α, IL-2, IL-3, IL-5, IL-6, IL-9, IL-12 (p40), IL-12 (p70), IL-13, IL-17A, CCL-11/Eotaxin, G-CSF, IFN-γ, CXCL1/KC, MCP-1 (MCAF), MIP-1α/CCL3, MIP-1ß/CCL4, CCL5/RANTES, and TNF-α). Systemic induction of cytokines was species-specific in the mouse model. The cytokine induction from infected implants differed significantly from sole tissue infections and sterile implants. Notably, systemic cytokine induction decreased after infections with dual species compared to single species infections. These findings describe the systemic effect of chronic peri-implant inflammation on the systemic induction of inflammatory cytokines, and this effect was strongly correlated to the type and composition of initial infection. Systemic modulations in cytokine expression upon dual species infections exhibit an exciting pattern that might explain the complications associated with biomaterial-related infection in patients. Moreover, these findings validate the requirement of multispecies infections for pre-clinical studies involving animal models.

4.
Cells ; 9(5)2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32429151

RESUMEN

Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.


Asunto(s)
Biopelículas , Células Epiteliales/microbiología , Fibroblastos/microbiología , Prótesis e Implantes/microbiología , Streptococcus oralis/fisiología , Adhesión Celular , Forma de la Célula , Supervivencia Celular , Citocinas/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Encía/patología , Humanos , Mediadores de Inflamación/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA