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

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hepatology ; 58(2): 589-602, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23322710

RESUMEN

UNLABELLED: Nonalcoholic steatohepatitis (NASH) is the most common etiology of chronic liver dysfunction in the United States and can progress to cirrhosis and liver failure. Inflammatory insult resulting from fatty infiltration of the liver is central to disease pathogenesis. Dendritic cells (DCs) are antigen-presenting cells with an emerging role in hepatic inflammation. We postulated that DCs are important in the progression of NASH. We found that intrahepatic DCs expand and mature in NASH liver and assume an activated immune phenotype. However, rather than mitigating the severity of NASH, DC depletion markedly exacerbated intrahepatic fibroinflammation. Our mechanistic studies support a regulatory role for DCs in NASH by limiting sterile inflammation through their role in the clearance of apoptotic cells and necrotic debris. We found that DCs limit CD8(+) T-cell expansion and restrict Toll-like receptor expression and cytokine production in innate immune effector cells in NASH, including Kupffer cells, neutrophils, and inflammatory monocytes. Consistent with their regulatory role in NASH, during the recovery phase of disease, ablation of DC populations results in delayed resolution of intrahepatic inflammation and fibroplasia. CONCLUSION: Our findings support a role for DCs in modulating NASH. Targeting DC functional properties may hold promise for therapeutic intervention in NASH.


Asunto(s)
Comunicación Celular/fisiología , Células Dendríticas/fisiología , Progresión de la Enfermedad , Hígado Graso/fisiopatología , Hígado/fisiopatología , Animales , Apoptosis/fisiología , Linfocitos T CD8-positivos/patología , Células Cultivadas , Células Dendríticas/patología , Modelos Animales de Enfermedad , Hígado Graso/patología , Macrófagos del Hígado/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Necrosis/fisiopatología , Neutrófilos/patología , Receptores Toll-Like/fisiología
2.
Gastroenterology ; 143(4): 1061-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705178

RESUMEN

BACKGROUND & AIMS: Immune cells of the liver must be able to recognize and react to pathogens yet remain tolerant to food molecules and other nonpathogens. Dendritic cells (DCs) are believed to contribute to hepatic tolerance. Lipids have been implicated in dysfunction of DCs in cancer. Therefore, we investigated whether high lipid content in liver DCs affects induction of tolerance. METHODS: Mouse and human hepatic nonparenchymal cells were isolated by mechanical and enzymatic digestion. DCs were purified by fluorescence-activated cell sorting or with immunomagnetic beads. DC lipid content was assessed by flow cytometry, immune fluorescence, and electron microscopy and by measuring intracellular component lipids. DC activation was determined from surface phenotype and cytokine profile. DC function was assessed in T-cell, natural killer (NK) cell, and NKT cell coculture assays as well as in vivo. RESULTS: We observed 2 distinct populations of hepatic DCs in mice and humans based on their lipid content and expression of markers associated with adipogenesis and lipid metabolism. This lipid-based dichotomy in DCs was unique to the liver and specific to DCs compared with other hepatic immune cells. However, rather than mediate tolerance, the liver DC population with high concentrations of lipid was immunogenic in multiple models; they activated T cells, NK cells, and NKT cells. Conversely, liver DCs with low levels of lipid induced regulatory T cells, anergy to cancer, and oral tolerance. The immunogenicity of lipid-rich liver DCs required their secretion of tumor necrosis factor α and was directly related to their high lipid content; blocking DC synthesis of fatty acids or inhibiting adipogenesis (by reducing endoplasmic reticular stress) reduced DC immunogenicity. CONCLUSIONS: Human and mouse hepatic DCs are composed of distinct populations that contain different concentrations of lipid, which regulates immunogenic versus tolerogenic responses in the liver.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Lípidos/análisis , Hígado/inmunología , Hígado/metabolismo , Adipogénesis , Animales , Antígenos CD1d/metabolismo , Apoptosis , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígeno CD11b/metabolismo , Antígenos CD40/metabolismo , Células Cultivadas , Células Dendríticas/química , Humanos , Tolerancia Inmunológica , Molécula 1 de Adhesión Intercelular/metabolismo , Células Asesinas Naturales/fisiología , Antígenos Comunes de Leucocito/metabolismo , Metabolismo de los Lípidos , Hígado/química , Activación de Linfocitos , Ratones , Células T Asesinas Naturales/fisiología , Fenotipo , Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Exp Med ; 209(9): 1671-87, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22908323

RESUMEN

The transition of chronic pancreatic fibroinflammatory disease to neoplasia is a primary example of the paradigm linking inflammation to carcinogenesis. However, the cellular and molecular mediators bridging these entities are not well understood. Because TLR4 ligation can exacerbate pancreatic inflammation, we postulated that TLR4 activation drives pancreatic carcinogenesis. In this study, we show that lipopolysaccharide accelerates pancreatic tumorigenesis, whereas TLR4 inhibition is protective. Furthermore, blockade of the MyD88-independent TRIF pathway is protective against pancreatic cancer, whereas blockade of the MyD88-dependent pathway surprisingly exacerbates pancreatic inflammation and malignant progression. The protumorigenic and fibroinflammatory effects of MyD88 inhibition are mediated by dendritic cells (DCs), which induce pancreatic antigen-restricted Th2-deviated CD4(+) T cells and promote the transition from pancreatitis to carcinoma. Our data implicate a primary role for DCs in pancreatic carcinogenesis and illustrate divergent pathways in which blockade of TLR4 signaling via TRIF is protective against pancreatic cancer and, conversely, MyD88 inhibition exacerbates pancreatic inflammation and neoplastic transformation by augmenting the DC-Th2 axis.


Asunto(s)
Adenocarcinoma/patología , Células Dendríticas/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias Pancreáticas/patología , Células Th2/patología , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adenocarcinoma/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Células Dendríticas/inmunología , Humanos , Masculino , Ratones , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/genética , Neoplasias Pancreáticas/metabolismo , Pancreatitis Crónica/metabolismo , Pancreatitis Crónica/patología , Células Th2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA