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










Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 157(5): 1323-1337, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31401143

RESUMO

BACKGROUND & AIMS: Epithelial tight junctions are compromised in gastrointestinal disease. Processes that contribute to the resulting barrier loss include endocytic occludin removal from the tight junction and reduced occludin expression. Nevertheless, the relatively-normal basal phenotype of occludin knockout (KO) mice has been taken as evidence that occludin does not contribute to gastrointestinal barrier function. We asked whether stress could unmask occludin functions within intestinal epithelia. METHODS: Wildtype (WT), universal and intestinal epithelial-specific occludin KO, and villin-EGFP-occludin transgenic mice as well as WT and occludin knockdown (KD) Caco-2BBe cell monolayers were challenged with DSS, TNBS, staurosporine, 5-FU, or TNF. Occludin and caspase-3 expression were assessed in patient biopsies. RESULTS: Intestinal epithelial occludin loss limited severity of DSS- and TNBS-induced colitis due to epithelial resistance to apoptosis; activation of both intrinsic and extrinsic apoptotic pathways was blocked in occludin KO epithelia. Promoter analysis revealed that occludin enhances CASP3 transcription and, conversely, that occludin downregulation reduces caspase-3 expression. Analysis of biopsies from Crohn's disease and ulcerative colitis patients and normal controls demonstrated that disease-associated occludin downregulation was accompanied by and correlated with reduced caspase-3 expression. In vitro, cytokine-induced occludin downregulation resulted in reduced caspase-3 expression and resistance to intrinsic and extrinsic pathway apoptosis, demonstrating an overall protective effect of inflammation-induced occludin loss. CONCLUSIONS: The tight junction protein occludin regulates apoptosis by enhancing caspase-3 transcription. These data suggest that reduced epithelial caspase-3 expression downstream of occludin downregulation is a previously-unappreciated anti-apoptotic process that contributes to mucosal homeostasis in inflammatory conditions.


Assuntos
Apoptose , Caspase 3/metabolismo , Colite/enzimologia , Colo/enzimologia , Células Epiteliais/enzimologia , Mucosa Intestinal/enzimologia , Ocludina/metabolismo , Animais , Células CACO-2 , Estudos de Casos e Controles , Caspase 3/deficiência , Caspase 3/genética , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colite Ulcerativa/enzimologia , Colite Ulcerativa/patologia , Colo/patologia , Doença de Crohn/enzimologia , Doença de Crohn/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Células Epiteliais/patologia , Humanos , Mucosa Intestinal/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ocludina/deficiência , Ocludina/genética , Transdução de Sinais , Ácido Trinitrobenzenossulfônico , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
2.
Nat Med ; 25(4): 690-700, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30936544

RESUMO

Epithelial barrier loss is a driver of intestinal and systemic diseases. Myosin light chain kinase (MLCK) is a key effector of barrier dysfunction and a potential therapeutic target, but enzymatic inhibition has unacceptable toxicity. Here, we show that a unique domain within the MLCK splice variant MLCK1 directs perijunctional actomyosin ring (PAMR) recruitment. Using the domain structure and multiple screens, we identify a domain-binding small molecule (divertin) that blocks MLCK1 recruitment without inhibiting enzymatic function. Divertin blocks acute, tumor necrosis factor (TNF)-induced MLCK1 recruitment as well as downstream myosin light chain (MLC) phosphorylation, barrier loss, and diarrhea in vitro and in vivo. Divertin corrects barrier dysfunction and prevents disease development and progression in experimental inflammatory bowel disease. Beyond applications of divertin in gastrointestinal disease, this general approach to enzymatic inhibition by preventing access to specific subcellular sites provides a new paradigm for safely and precisely targeting individual properties of enzymes with multiple functions.


Assuntos
Homeostase , Mucosa Intestinal/metabolismo , Espaço Intracelular/enzimologia , Quinase de Cadeia Leve de Miosina/metabolismo , Actomiosina/metabolismo , Animais , Células CACO-2 , Doença Crônica , Homeostase/efeitos dos fármacos , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/efeitos dos fármacos , Jejuno/efeitos dos fármacos , Jejuno/metabolismo , Jejuno/patologia , Camundongos , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/química , Fosforilação/efeitos dos fármacos , Domínios Proteicos , Bibliotecas de Moléculas Pequenas/farmacologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
3.
J Clin Invest ; 129(2): 902-914, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30667372

RESUMO

Graft-versus-host disease (GVHD) is a complication of hematopoietic stem cell transplantation (HSCT) that affects multiple organs. GVHD-associated intestinal damage can be separated into two distinct phases, initiation and propagation, which correspond to conditioning-induced damage and effector T cell activation and infiltration, respectively. Substantial evidence indicates that intestinal damage induced by pretransplant conditioning is a key driver of GVHD initiation. Here, we aimed to determine the impact of dysregulated intestinal permeability on the subsequent GVHD propagation phase. The initiation phase of GVHD was unchanged in mice lacking long MLCK (MLCK210), an established regulator of epithelial tight junction permeability. However, MLCK210-deficient mice were protected from sustained barrier loss and exhibited limited GVHD propagation, as indicated by reduced histopathology, fewer CD8+ effector T cells in the gut, and improved overall survival. Consistent with these findings, intestinal epithelial MLCK210 expression and enzymatic activity were similarly increased in human and mouse GVHD biopsies. Intestinal epithelial barrier loss mediated by MLCK210 is therefore a key driver of the GVHD propagation. These data suggest that inhibition of MLCK210-dependent barrier regulation may be an effective approach to limiting GVHD progression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Mucosa Intestinal/imunologia , Quinase de Cadeia Leve de Miosina/imunologia , Junções Íntimas/imunologia , Aloenxertos , Animais , Linfócitos T CD8-Positivos/patologia , Feminino , Doença Enxerto-Hospedeiro/patologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos BALB C , Junções Íntimas/patologia
4.
Compr Physiol ; 7(4): 1497-1518, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28915334

RESUMO

Epithelial cells are essential to the survival and homeostasis of complex organisms. These cells cover the surfaces of all mucosae, the skin, and other compartmentalized structures essential to physiological function. In addition to maintenance of barriers that separate internal and external compartments, epithelia display a variety of organ-specific differentiated functions. Function is reflected in overall epithelial structure and organization, shape of individual cells, and proteins expressed by these cells. More than one epithelial cell type is often present within a single organ and, in many cases, individual cells differentiate to change their functional behaviors as part of normal development or in response to extracellular stimuli. This article discusses the diversity of epithelial structure and function in general terms and explores representative tissues in greater depth to highlight organ specific functions and their contributions to physiology and disease. © 2017 American Physiological Society. Compr Physiol 7:1497-1518, 2017.


Assuntos
Células Epiteliais/metabolismo , Absorção Intestinal , Mucosa Intestinal/citologia , Animais , Células Epiteliais/citologia , Humanos , Mucosa Intestinal/metabolismo , Néfrons/citologia , Néfrons/metabolismo , Especificidade de Órgãos , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Urotélio/citologia , Urotélio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...