Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 199(5): 1772-1782, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28733486

RESUMO

The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1ß secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1ß promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.


Assuntos
Toxinas Bacterianas/imunologia , Granulócitos/imunologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas Hemolisinas/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Fatores de Virulência/imunologia , Infecção dos Ferimentos/imunologia , Animais , Carga Bacteriana , Toxinas Bacterianas/genética , Diferenciação Celular , Proliferação de Células , Granulócitos/microbiologia , Proteínas Hemolisinas/genética , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fator 88 de Diferenciação Mieloide/genética , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Fatores de Virulência/genética
2.
Stem Cells ; 35(6): 1493-1504, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28276602

RESUMO

Mesenchymal stem cell therapies promote wound healing by manipulating the local environment to enhance the function of host cells. Aggregation of mesenchymal stem cells (MSCs) into three-dimensional spheroids increases cell survival and augments their anti-inflammatory and proangiogenic potential, yet there is no consensus on the preferred conditions for maximizing spheroid function in this application. The objective of this study was to optimize conditions for forming MSC spheroids that simultaneously enhance their anti-inflammatory and proangiogenic nature. We applied a design of experiments (DOE) approach to determine the interaction between three input variables (number of cells per spheroid, oxygen tension, and inflammatory stimulus) on MSC spheroids by quantifying secretion of prostaglandin E2 (PGE2 ) and vascular endothelial growth factor (VEGF), two potent molecules in the MSC secretome. DOE results revealed that MSC spheroids formed with 40,000 cells per spheroid in 1% oxygen with an inflammatory stimulus (Spheroid 1) would exhibit enhanced PGE2 and VEGF production versus those formed with 10,000 cells per spheroid in 21% oxygen with no inflammatory stimulus (Spheroid 2). Compared to Spheroid 2, Spheroid 1 produced fivefold more PGE2 and fourfold more VEGF, providing the opportunity to simultaneously upregulate the secretion of these factors from the same spheroid. The spheroids induced macrophage polarization, sprout formation with endothelial cells, and keratinocyte migration in a human skin equivalent model-demonstrating efficacy on three key cell types that are dysfunctional in chronic non-healing wounds. We conclude that DOE-based analysis effectively identifies optimal culture conditions to enhance the anti-inflammatory and proangiogenic potential of MSC spheroids. Stem Cells 2017;35:1493-1504.


Assuntos
Anti-Inflamatórios/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Esferoides Celulares/metabolismo , Polaridade Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Meios de Cultivo Condicionados/farmacologia , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , NF-kappa B/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Reprodutibilidade dos Testes , Projetos de Pesquisa , Transdução de Sinais/efeitos dos fármacos , Pele Artificial , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Cicatrização/efeitos dos fármacos
3.
Blood ; 122(10): 1770-8, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23869087

RESUMO

During bacterial infection, hematopoietic stem and progenitor cells (HSPCs) differentiate into polymorphonuclear leukocytes (PMNs) in the bone marrow. We reported that HSPCs recruited to Staphylococcus aureus-infected skin wounds in mice undergo granulopoiesis, whereas other authors have demonstrated their differentiation in vitro after Toll-like receptor 2 (TLR2)/MyD88 stimulation. Here, we examined this pathway in HSPC trafficking and granulopoiesis within S aureus-infected wounds. Lineage- HSPCs from TLR2- or MyD88-deficient mice injected into infected wounds of wild-type (WT) mice exhibited impaired granulopoiesis. However, HSPCs from WT mice produced similar numbers of PMNs whether transferred into wounds of TLR2-, MyD88-deficient, or WT mice. Prostaglandin E2 (PGE2), which stimulates HSPC survival and proliferation, was produced by HSPCs after TLR2 stimulation, suggesting that TLR2/MyD88 activation promotes granulopoiesis in part by production and autocrine activity of PGE2. Pretreatment of TLR2- or MyD88-deficient HSPCs with PGE2 rescued granulocytic differentiation in vivo. Finally, we demonstrate that bone marrow-derived lin-/Sca-1+/c-kit+ cells produced PGE2 and underwent granulopoiesis after TLR2 stimulation. lin-/Sca-1+/c-kit+ cells deficient in TLR2 or MyD88 produced PMNs after PGE2 treatment when transferred into uninfected wounds. We conclude that granulopoiesis in S aureus-infected wounds is induced by TLR2/MyD88 activation of HSPCs through a mechanism that involves autocrine production and activity of PGE2.


Assuntos
Dinoprostona/metabolismo , Células-Tronco Hematopoéticas/imunologia , Leucopoese , Fator 88 de Diferenciação Mieloide/metabolismo , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/metabolismo , Ferimentos e Lesões/microbiologia , Animais , Medula Óssea/patologia , Contagem de Células , Diferenciação Celular , Proliferação de Células , Separação Celular , Dinoprostona/biossíntese , Feminino , Granulócitos/imunologia , Granulócitos/patologia , Células-Tronco Hematopoéticas/microbiologia , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Transdução de Sinais/imunologia , Pele/imunologia , Pele/microbiologia , Pele/patologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Receptor 2 Toll-Like/deficiência , Ferimentos e Lesões/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...