Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
2.
Liver Int ; 34(9): 1402-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24650018

RESUMEN

BACKGROUND & AIMS: Inflammation promotes the progression of non-alcoholic steatohepatitis (NASH). Toll-like receptor 4 (TLR4) and TLR9 activation through myeloid differentiation primary response gene 88 (MyD88) and production of mature interleukin-1ß (IL-1ß) via inflammasome activation contribute to steatohepatitis. Here, we investigated the inter-relationship between TLR signalling and inflammasome activation in dietary steatohepatitis. METHODS: Wild type (WT), TLR4- and MyD88-deficient (KO) mice received methionine-choline-deficient (MCD) or -supplemented (MCS) diets for 5 weeks and a subset was challenged with TLR9 ligand CpG-DNA. RESULTS: TLR4, TLR9, AIM2 (absent in melanoma 2) and NLRP3 (NLR family pyrin domain containing 3) inflammasome mRNA, and mature IL-1ß protein levels were increased in MCD diet-induced steatohepatitis compared to MCS controls. TLR9 stimulation resulted in greater up-regulation of the DNA-sensing AIM2 expression and IL-1ß production in livers of MCD compared to MCS diet-fed mice. High mobility group box 1 (HMGB1), a TLR9-activating danger molecule and phospho-HMGB1 protein levels were also increased in livers of MCD diet-fed mice. MyD88- but not TLR4-deficiency prevented up-regulation of AIM2, NLRP3 mRNA and IL-1ß protein production in dietary steatohepatitis. Selective MyD88 deficiency either in bone marrow (BM)-derived or non-BM-derived cells attenuated hepatic up-regulation of inflammasome mRNA, caspase-1 activation and IL-1ß protein production, but only BM-derived cell-specific MyD88-deficiency attenuated liver injury. CONCLUSIONS: Our data demonstrate that both bone marrow-derived and non-BM-derived cells contribute to inflammasome activation in a MyD88-dependent manner in dietary steatohepatitis. We show that AIM2 inflammasome expression and activation are further augmented by TLR9 ligands in dietary steatohepatitis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Inflamasomas/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Células de la Médula Ósea/metabolismo , Deficiencia de Colina , Dieta , Inmunoprecipitación , Metionina/deficiencia , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo
5.
Gastroenterology ; 140(2): 697-708.e4, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20727895

RESUMEN

BACKGROUND & AIMS: Liver inflammation and injury are mediated by the innate immune response, which is regulated by Toll-like receptors (TLR). Activation of TLR9 induces type I interferons (IFNs) via the interferon regulatory factor (IRF)-7. We investigated the roles of type I IFNs in TLR9-associated liver injury. METHODS: Wild-type (WT), IRF7-deficient, and IFN-α/ß receptor 1 (IFNAR1)-deficient mice were stimulated with TLR9 or TLR2 ligands. Findings from mice were verified in cultured hepatocytes and liver mononuclear cells (LMNCs) as well as in vivo experiments using recombinant type I IFN and interleukin-1 receptor antagonist (IL-1ra). RESULTS: Type I IFNs were up-regulated during TLR9-associated liver injury in WT mice. IRF7- and IFNAR1-deficient mice, which have disruptions in type I IFN production or signaling, respectively, had increased liver damage and inflammation, decreased recruitment of dendritic cells, and increased production of tumor necrosis factor α by LMNCs. These findings indicate that type I IFNs have anti-inflammatory activities in liver. IL-1ra, which is produced by LMNCs and hepatocytes, is an IFN-regulated antagonist of the proinflammatory cytokine IL-1ß; IRF7- and IFNAR1-deficient mice had decreased levels of IL-1ra compared with WT mice. IL-1ra protected cultured hepatocytes from IL-1ß-mediated sensitization to cytotoxicity from tumor necrosis factor α. In vivo exposure to type I IFN, which induced IL-1ra, or administration of IL-1ra reduced TLR9-associated liver injury; the protective effect of type I IFNs therefore appears to be mediated by IFN-dependent induction of IL-1ra. CONCLUSIONS: Type I IFNs have anti-inflammatory effects mediated by endogenous IL-1ra, which regulates the extent of TLR9-induced liver damage. Type I IFN signaling is therefore required for protection from immune-mediated liver injury.


Asunto(s)
Hepatitis/inmunología , Hepatitis/patología , Interferón Tipo I/inmunología , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Receptor Toll-Like 9/inmunología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Hepatocitos/inmunología , Factor 7 Regulador del Interferón/genética , Interferón Tipo I/biosíntesis , Interferón Tipo I/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/genética , Receptor Toll-Like 2/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/inmunología
6.
Hepatology ; 54(1): 133-44, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21488066

RESUMEN

UNLABELLED: The pathogenesis of nonalcoholic steatohepatitis (NASH) and inflammasome activation involves sequential hits. The inflammasome, which cleaves pro-interleukin-1ß (pro-IL-1ß) into secreted IL-1ß, is induced by endogenous and exogenous danger signals. Lipopolysaccharide (LPS), a toll-like receptor 4 ligand, plays a role in NASH and also activates the inflammasome. In this study, we hypothesized that the inflammasome is activated in NASH by multiple hits involving endogenous and exogenous danger signals. Using mouse models of methionine choline-deficient (MCD) diet-induced NASH and high-fat diet-induced NASH, we found up-regulation of the inflammasome [including NACHT, LRR, and PYD domains-containing protein 3 (NALP3; cryopyrin), apoptosis-associated speck-like CARD-domain containing protein, pannexin-1, and pro-caspase-1] at the messenger RNA (mRNA) level increased caspase-1 activity, and mature IL-1ß protein levels in mice with steatohepatitis in comparison with control livers. There was no inflammasome activation in mice with only steatosis. The MCD diet sensitized mice to LPS-induced increases in NALP3, pannexin-1, IL-1ß mRNA, and mature IL-1ß protein levels in the liver. We demonstrate for the first time that inflammasome activation occurs in isolated hepatocytes in steatohepatitis. Our novel data show that the saturated fatty acid (FA) palmitic acid (PA) activates the inflammasome and induces sensitization to LPS-induced IL-1ß release in hepatocytes. Furthermore, PA triggers the release of danger signals from hepatocytes in a caspase-dependent manner. These hepatocyte-derived danger signals, in turn, activate inflammasome, IL-1ß, and tumor necrosis factor α release in liver mononuclear cells. CONCLUSION: Our novel findings indicate that saturated FAs represent an endogenous danger in the form of a first hit, up-regulate the inflammasome in NASH, and induce sensitization to a second hit with LPS for IL-ß release in hepatocytes. Furthermore, hepatocytes exposed to saturated FAs release danger signals that trigger inflammasome activation in immune cells. Thus, hepatocytes play a key role in orchestrating tissue responses to danger signals in NASH.


Asunto(s)
Endotoxinas/farmacología , Ácidos Grasos/farmacología , Hígado Graso/etiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Sistema Inmunológico/fisiología , Inflamasomas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 1/metabolismo , Deficiencia de Colina/complicaciones , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Endotoxinas/efectos adversos , Ácidos Grasos/efectos adversos , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Femenino , Hepatocitos/patología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Factor de Necrosis Tumoral alfa/metabolismo
7.
Hepatology ; 53(6): 1917-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21425308

RESUMEN

UNLABELLED: Mitochondrial dysfunction is a pathogenic feature of nonalcoholic steatohepatitis (NASH). NASH complicates hepatotropic viral disease. The mitochondrial antiviral signaling protein (MAVS) is the adapter of helicase receptors involved in sensing double-stranded RNA (dsRNA). We hypothesized that impaired MAVS function may contribute to insufficient antiviral response and liver damage in steatohepatitis. We identified reduced MAVS protein levels and increased MAVS association with the proteasome subunit alpha type 7 (PSMA7) in livers from mice given a methionine-choline-deficient (MCD) diet. Decreased association of MAVS with mitochondria and increased cytosolic cytochrome c indicated mitochondrial damage in steatohepatitis. In vivo administration of the synthetic dsRNA polyinosinic:polycytidylic acid [poly(I:C)], but not lipopolysaccharide or cytidine-phosphate-guanosine-rich DNA, resulted in impaired induction of type I interferons (IFNs) and proinflammatory cytokines in steatohepatitis. Consistent with a defect in helicase receptor-induced signaling, there was loss of poly(I:C)-induced translocation of MAVS to the cytosol and decreased IFN regulatory factor 3 phosphorylation. Caspases 1 and 8, both of which cleave MAVS, were increased in MCD diet-fed mice. At baseline, steatohepatitis was associated with increased serum alanine aminotransferase (ALT), apoptosis and caspase 3 activation compared with controls. In contrast to apoptosis in controls, necrosis was induced by poly(I:C) stimulation in steatohepatitis. Hepatocyte necrosis was indicated by elevated serum high-mobility group box protein-1 and ALT and was correlated with increased expression of receptor-interacting protein 3 (RIP3), a master regulator of necrosis. Increased expression of MAVS, PSMA7, and RIP3 messenger RNA was also present in human NASH livers. CONCLUSION: Our novel findings suggest that mitochondrial damage in steatohepatitis extends to MAVS, an adapter of helicase receptors, resulting in inefficient type I IFN and inflammatory cytokine response but increased hepatocyte necrosis and RIP3 induction in response to a dsRNA viral challenge. These mechanisms may contribute to progressive liver damage and impaired viral clearance in NASH.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Hígado Graso/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Poli I-C/farmacología , ARN Bicatenario/farmacología , Animales , Apoptosis/efectos de los fármacos , Biopsia , Deficiencia de Colina/complicaciones , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/patología , Femenino , Humanos , Interferón Tipo I/metabolismo , Hígado/patología , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
8.
Hepatology ; 53(2): 649-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21274885

RESUMEN

UNLABELLED: Alcoholic liver disease (ALD) features increased hepatic exposure to bacterial lipopolysaccharide (LPS). Toll-like receptor-4 (TLR4) recognizes LPS and activates signaling pathways depending on MyD88 or TRIF adaptors. We previously showed that MyD88 is dispensable in ALD. TLR4 induces Type I interferons (IFNs) in an MyD88-independent manner that involves interferon regulatory factor-3 (IRF3). We fed alcohol or control diets to wild-type (WT) and IRF3 knock-out (KO) mice, and to mice with selective IRF3 deficiency in liver parenchymal and bone marrow-derived cells. Whole-body IRF3-KO mice were protected from alcohol-induced liver injury, steatosis, and inflammation. In contrast to WT or bone marrow-specific IRF3-KO mice, deficiency of IRF3 only in parenchymal cells aggravated alcohol-induced liver injury, associated with increased proinflammatory cytokines, lower antiinflammatory cytokine interleukin 10 (IL-10), and lower Type I IFNs compared to WT mice. Coculture of WT primary murine hepatocytes with liver mononuclear cells (LMNC) resulted in higher LPS-induced IL-10 and IFN-ß, and lower tumor necrosis factor alpha (TNF-α) levels compared to LMNC alone. Type I IFN was important because cocultures of hepatocytes with LMNC from Type I IFN receptor KO mice showed attenuated IL-10 levels compared to control cocultures from WT mice. We further identified that Type I IFNs potentiated LPS-induced IL-10 and inhibited inflammatory cytokine production in both murine macrophages and human leukocytes, indicating preserved cross-species effects. These findings suggest that liver parenchymal cells are the dominant source of Type I IFN in a TLR4/IRF3-dependent manner. Further, parenchymal cell-derived Type I IFNs increase antiinflammatory and suppress proinflammatory cytokines production by LMNC in paracrine manner. CONCLUSION: Our results indicate that IRF3 activation in parenchymal cells and resulting type I IFNs have protective effects in ALD by way of modulation of inflammatory functions in macrophages. These results suggest potential therapeutic targets in ALD.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Etanol/efectos adversos , Hepatocitos/patología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Células Mieloides/patología , Animales , Comunicación Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Femenino , Hepatocitos/metabolismo , Humanos , Factor 3 Regulador del Interferón/deficiencia , Factor 3 Regulador del Interferón/genética , Interleucina-10/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Células Mieloides/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo
9.
Liver Int ; 32(5): 732-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22292891

RESUMEN

BACKGROUND/AIMS: Liver diseases are common in the United States and often require liver transplantation; however, donated organs are limited and thus alternative sources for liver cells are in high demand. Embryonic stem cells (ESC) can provide a continuous and readily available source of liver cells. ESC differentiation to liver cells is yet to be fully understood and comprehensive differentiation protocols are yet to be defined. Here, we aimed to achieve human (h)ESC differentiation into mature hepatocytes using defined recombinant differentiation factors and metabolites. METHODS: Embryonic stem cell H1 line was sub-cultured on feeder layer. We induced hESCs into endodermal differentiation succeeded by early/late hepatic specification and finally into hepatocyte maturation using step combinations of Activin A and fibroblast growth factor (FGF)-2 for 7 days; followed by FGF-4 and bone morphogenic protein 2 (BMP2) for 7 days, succeeded by FGF-10 + hepatocyte growth factor 4 + epidermal growth factor for 14 days. Specific inhibitors/stimulators were added sequentially throughout differentiation. Cells were analysed by PCR, flow cytometry, microscopy or functional assays. RESULTS: Our hESC differentiation protocol resulted in viable cells with hepatocyte shape and morphology. We observed gradual changes in cell transcriptome, including up-regulation of differentiation-promoting GATA4, GATA6, POU5F1 and HNF4 transcription factors, steady levels of stemness-promoting SOX-2 and low levels of Nanog, as defined by PCR. The hESC-derived hepatocytes expressed alpha-antitrypsin, CD81, cytokeratin 8 and low density lipoprotein (LDL) receptor. The levels of alpha-fetoprotein and proliferation marker Ki-67 in hESC-derived hepatocytes remained elevated. Unlike stem cells, the hESC-derived hepatocytes performed LDL uptake, produced albumin and alanine aminotransferase and had functional alcohol dehydrogenase. CONCLUSION: We report a novel protocol for hESC differentiation into morphological and functional yet immature hepatocytes as an alternative method for hepatocyte generation.


Asunto(s)
Activinas/farmacología , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Factores de Crecimiento de Fibroblastos/farmacología , Hepatocitos/citología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Combinación de Medicamentos , Células Madre Embrionarias/metabolismo , Citometría de Flujo , Hepatocitos/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , Proyectos de Investigación , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Regulación hacia Arriba
11.
Alcohol Clin Exp Res ; 36(8): 1301-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22551004

RESUMEN

Alcohol is the most abused substance worldwide and a significant source of liver injury; the mechanisms of alcohol-induced liver disease are not fully understood. Significant cellular toxicity and impairment of protein synthesis and degradation occur in alcohol-exposed liver cells, along with changes in energy balance and modified responses to pathogens. Autophagy is the process of cellular catabolism through the lysosomal-dependent machinery, which maintains a balance among protein synthesis, degradation, and recycling of self. Autophagy is part of normal homeostasis and it can be triggered by multiple factors that threaten cell integrity, including starvation, toxins, or pathogens. Multiple factors regulate autophagy; survival and preservation of cellular integrity at the expense of inadequately folded proteins and damaged high-energy generating intracellular organelles are prominent targets of autophagy in pathological conditions. Coincidentally, inadequately folded proteins accumulate and high-energy generating intracellular organelles, such as mitochondria, are damaged by alcohol abuse; these alcohol-induced pathological findings prompted investigation of the role of autophagy in the pathogenesis of alcohol-induced liver damage. Our review summarizes the current knowledge about the role and implications of autophagy in alcohol-induced liver disease.


Asunto(s)
Autofagia/fisiología , Hepatopatías Alcohólicas/patología , Animales , Biomarcadores , Muerte Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/toxicidad , Modelos Animales de Enfermedad , Etanol/toxicidad , Humanos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Fagosomas/metabolismo , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Receptores Toll-Like/efectos de los fármacos
12.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G433-41, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21233280

RESUMEN

Toll-like receptor 4 (TLR4) and its coreceptor, myeloid differentiation factor-2 (MD-2), are key in recognition of lipopolysaccharide (LPS) and activation of proinflammatory pathways. Here we tested the hypothesis that TLR4 and its coreceptor MD-2 play a central role in nonalcoholic steatohepatitis (NASH) and liver fibrosis in nonalcoholic fatty liver disease. Mice of control genotypes and those deficient in MD-2 or TLR4 [knockout (KO)] received methionine choline-deficient (MCD) or methionine choline-supplemented (MCS) diet. In mice of control genotypes, MCD diet resulted in NASH, liver triglycerides accumulation, and increased thiobarbituric acid reactive substances, a marker of lipid peroxidation, compared with MCS diet. These features of NASH were significantly attenuated in MD-2 KO and TLR4 KO mice. Serum alanine aminotransferase, an indicator of liver injury, was increased in MCD diet-fed genotype controls but was attenuated in MD-2 KO and TLR4 KO mice. Inflammatory activation, indicated by serum TNF-α and nictoinamide adenine dinucleotide phosphate oxidase complex mRNA expression and activation, was significantly lower in MCD diet-fed MD-2 KO and TLR4 KO compared with corresponding genotype control mice. Markers of liver fibrosis [collagen by Sirius red and α-smooth muscle actin (SMA) staining, procollagen-I, transforming growth factor-ß1, α-SMA, matrix metalloproteinase-2, and tissue inhibitor of matrix metalloproteinase-1 mRNA] were attenuated in MD-2 and TLR4 KO compared with their control genotype counterparts. In conclusion, our results demonstrate a novel, critical role for LPS recognition complex, including MD-2 and TLR4, through NADPH activation in liver steatosis, and fibrosis in a NASH model in mice.


Asunto(s)
Cirrosis Hepática Experimental/prevención & control , Hígado/inmunología , Antígeno 96 de los Linfocitos/deficiencia , Receptor Toll-Like 4/deficiencia , Actinas/genética , Alanina Transaminasa/sangre , Animales , Deficiencia de Colina/complicaciones , Colágeno/genética , Hígado Graso/genética , Hígado Graso/inmunología , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Femenino , Genotipo , Peroxidación de Lípido , Hígado/metabolismo , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/inmunología , Cirrosis Hepática Experimental/metabolismo , Antígeno 96 de los Linfocitos/genética , Metaloproteinasa 2 de la Matriz/genética , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Estrés Oxidativo , Fenotipo , ARN Mensajero/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Receptor Toll-Like 4/genética , Factor de Crecimiento Transformador beta1/genética , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/sangre
13.
Hepatology ; 49(3): 989-97, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19115316

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) and its advanced stage, nonalcoholic steatohepatitis (NASH), are the most common causes of chronic liver disease in the United States. NASH features the metabolic syndrome, inflammation, and fibrosis. Probiotics exhibit immunoregulatory and anti-inflammatory activity. We tested the hypothesis that probiotic VSL#3 may ameliorate the methionine-choline-deficient (MCD) diet-induced mouse model of NASH. MCD diet resulted in NASH in C57BL/6 mice compared to methionine-choline-supplemented (MCS) diet feeding evidenced by liver steatosis, increased triglycerides, inflammatory cell accumulation, increased tumor necrosis factor alpha levels, and fibrosis. VSL#3 failed to prevent MCD-induced liver steatosis or inflammation. MCD diet, even in the presence of VSL#3, induced up-regulation of serum endotoxin and expression of the Toll-like receptor 4 signaling components, including CD14 and MD2, MyD88 adaptor, and nuclear factor kappaB activation. In contrast, VSL#3 treatment ameliorated MCD diet-induced liver fibrosis resulting in diminished accumulation of collagen and alpha-smooth muscle actin. We identified increased expression of liver peroxisome proliferator-activated receptors and decreased expression of procollagen and matrix metalloproteinases in mice fed MCD+VSL#3 compared to MCD diet alone. MCD diet triggered up-regulation of transforming growth factor beta (TGFbeta), a known profibrotic agent. In the presence of VSL#3, the MCD diet-induced expression of TGFbeta was maintained; however, the expression of Bambi, a TGFbeta pseudoreceptor with negative regulatory function, was increased. In summary, our data indicate that VSL#3 modulates liver fibrosis but does not protect from inflammation and steatosis in NASH. The mechanisms of VSL#3-mediated protection from MCD diet-induced liver fibrosis likely include modulation of collagen expression and impaired TGFbeta signaling.


Asunto(s)
Dieta/efectos adversos , Hígado Graso/inducido químicamente , Cirrosis Hepática/prevención & control , Probióticos/uso terapéutico , Actinas/metabolismo , Animales , Deficiencia de Colina/complicaciones , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Femenino , Cirrosis Hepática/metabolismo , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Probióticos/farmacología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
14.
Gastroenterology ; 135(6): 2119-27, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835391

RESUMEN

BACKGROUND & AIMS: Dendritic cells (DCs) initiate and sustain an efficient T-lymphocyte response. Chronic hepatitis C virus (HCV) infection is associated with inefficient T-cell functions that fail to eradicate the virus, so defects in DC function might be involved in HCV pathogenesis. This study analyzed the activities of myeloid DCs and distinct CD4(+) T-cell populations in samples collected from patients with HCV. METHODS: The abilities of primary BDCA1(+) or monocyte-derived DCs from HCV patients (HCV-DC) to stimulate CD4(+), CD4(+)CD25(-), or different ratios of CD4(+)CD25(+)/CD4(+)CD25(-) T cells were evaluated in mixed lymphocyte reactions. T-cell proliferation and phenotype were evaluated by flow cytometry; cytokine production was evaluated by enzyme-linked immunosorbent assay and marker expression by polymerase chain reaction analyses. RESULTS: HCV-DCs were poor activators of CD4(+) T cells; this defect was reversed by addition of interleukin-2, neutralization of interleukin-10, or elimination of CD4(+)CD25(+) T cells. HCV-DC stimulated proliferation of regulatory T cells (Tregs; CD4(+)CD25(+)FoxP3(+)), which limit proliferation of HCV-specific T lymphocytes. We observed an increased frequency of CD4(+)CD25(+) T cells in peripheral blood of HCV patients and that HCV-DC overexpressed a number of alternative costimulatory molecules, including PD-L1. Finally, HCV-DC stimulated expansion rather than de novo induction of FoxP3(+) Tregs. CONCLUSIONS: Our results indicate a role for myeloid DC in expansion of Tregs to promote chronic infection of patients with HCV.


Asunto(s)
Células Dendríticas/inmunología , Hepatitis C Crónica/inmunología , Células Mieloides/inmunología , Linfocitos T Reguladores/patología , Proliferación Celular , Citocinas/biosíntesis , Citocinas/genética , Células Dendríticas/patología , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/patología , Humanos , Células Mieloides/patología , ARN/genética , Linfocitos T Reguladores/inmunología
15.
Hepatology ; 48(4): 1342-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18798338

RESUMEN

Toll-like receptors (TLRs) expressed on both immune cells and hepatocytes recognize microbial danger signals and regulate immune responses. Previous studies showed that TLR9 and TLR2 mediate Propionibacterium acnes-induced sensitization to lipopolysaccharide-triggered acute liver injury in mice. Ligand-specific activation of TLR2 and TLR9 are dependent on the common TLR adaptor, myeloid differentiation factor 88 (MyD88). Here, we dissected the role of MyD88 in parenchymal and bone marrow (BM)-derived cells in liver sensitization. Using chimeric mice with green fluorescent protein-expressing BM cells, we identified that P. acnes-induced liver inflammatory foci are of BM origin. Chimeras with MyD88-deficient BM showed no inflammatory foci after P. acnes or TLR2+TLR9 challenge, suggesting that recruitment of inflammatory cells to the liver required MyD88 expression in BM-derived cells. Further, selective MyD88 deficiency in parenchymal cells in mice with wild-type BM failed to prevent inflammatory cell infiltration. These results demonstrate that MyD88 in immune cells rather than in liver parenchymal cells plays an important role in inflammatory cell recruitment and liver injury.


Asunto(s)
Células de la Médula Ósea/metabolismo , Granuloma/metabolismo , Hepatopatías/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Animales , Células de la Médula Ósea/citología , Quimera , Femenino , Granuloma/microbiología , Granuloma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Hepatopatías/microbiología , Hepatopatías/patología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Propionibacterium acnes , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 9/metabolismo
16.
Hepatology ; 48(4): 1224-31, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18792393

RESUMEN

UNLABELLED: The Toll-like receptor 4 (TLR4) that recognizes endotoxin, a trigger of inflammation in alcoholic liver disease (ALD), activates two signaling pathways utilizing different adapter molecules: the common TLR adapter, myeloid differentiation factor 88 (MyD88), or Toll/interleukin immune-response-domain-containing adaptor inducing interferon (IFN)-beta. The MyD88 pathway induces proinflammatory cytokine activation, a critical mediator of ALD. Here we evaluated the role of MyD88 in alcohol-induced liver injury in wild-type, TLR2-deficient, TLR4-deficient, or MyD88-deficient (knockout [KO]) mice after administration of the Lieber-De-Carli diet (4.5% volume/volume ethanol) or an isocaloric liquid control diet for 5 weeks. Alcohol feeding resulted in a significant increase in serum alanine aminotransferase levels, liver steatosis and triglyceride levels suggesting liver damage in WT, TLR2-KO, and MyD88-KO but not in TLR4-KO mice. Expression of inflammatory mediators (tumor necrosis factor-alpha and interleukin-6) and TLR4 coreceptors (CD14 and MD2) was significantly higher in livers of alcohol-fed WT, TLR2-KO, or MyD88-KO, but not in TLR4-KO mice, compared to controls. Reactive oxygen radicals produced by cytochrome P450 and the nicotinamide adenine dinucleotide phosphate complexes contribute to alcoholic liver damage. Alcohol feeding-induced expression and activation of cytochrome P450 and the nicotinamide adenine dinucleotide phosphate complex were prevented by TLR4-deficiency but not by MyD88-deficiency. Liver expression of interferon regulatory factor 3 (IRF3), a MyD88-independent signaling molecule, was not affected by chronic alcohol treatment in whole livers of WT mice or in any of the KO mice. However, the induction of IRF7, an IRF3-inducible gene, was found in Kupffer cells of alcohol-fed WT mice. Alcohol feeding also induced nuclear factor-kappaB activation in a TLR4-dependent MyD88-independent manner. CONCLUSION: While TLR4 deficiency was protective, MyD88 deficiency failed to prevent alcohol-induced liver damage and inflammation. These results suggest that the common TLR adapter, MyD88, is dispensable in TLR4-mediated liver injury in ALD.


Asunto(s)
Hepatopatías Alcohólicas/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Animales , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Etanol , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Hepatocitos/metabolismo , Hepatocitos/patología , Interleucina-6/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Hígado/metabolismo , Hígado/patología , Hepatopatías Alcohólicas/patología , Hepatopatías Alcohólicas/prevención & control , Antígeno 96 de los Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , NADP/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Crit Rev Immunol ; 28(1): 61-94, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18298384

RESUMEN

Viral infections represent a major source of acute and chronic human disease. The immune system plays a central role in the elimination of viruses through its ability to recognize pathogens and to induce virus-specific cellular activation, accompanied by a robust production of soluble molecules with antiviral effects. Interferons are among the most powerful natural soluble antiviral molecules. Upon viral infection, interferons are produced by a variety of cell types, with immune cells being the main contributors. The immune system works as a well-orchestrated team composed of multiple cell types. The mechanisms of intercellular cooperation that includes dendritic cells (DCs), their soluble factors, and different types of immune cells are yet to be fully understood. Further, the effects of viral infections on interimmune cooperation need to be investigated. In this review, we define the role of plasmacytoid dendritic cells (PDC) and PDC-derived interferon alpha (IFNalpha) during viral infections. Specifically, we address the mechanisms of IFNalpha induction and the cooperation between PDC, PDC-derived IFNalpha and T cells, B cells, NK, iNKT, and myeloid dendric cells in antiviral immune responses.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus ADN/inmunología , Células Dendríticas/inmunología , Interferón-alfa/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Virus ARN/inmunología , Subgrupos de Linfocitos T/inmunología , Receptores Toll-Like/metabolismo , Animales , Antivirales/inmunología , Antivirales/metabolismo , Linfocitos B/metabolismo , Infecciones por Virus ADN/virología , Células Dendríticas/metabolismo , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Interferón-alfa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Infecciones por Virus ARN/virología , Subgrupos de Linfocitos T/metabolismo , Receptores Toll-Like/inmunología , Virus/inmunología
18.
Alcohol Clin Exp Res ; 33(10): 1704-10, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19572984

RESUMEN

BACKGROUND: Alcoholic and nonalcoholic steatohepatitis are leading causes of liver diseases worldwide. While of different etiology, these share common pathophysiological mechanisms and feature abnormal fat metabolism, inflammation and fibrosis. MicroRNAs (miRNA) are highly conserved noncoding RNAs that control gene expression at the post-transcriptional level either via the degradation of target mRNAs or the inhibition of translation. Each miRNA controls the expression of multiple targets; miRNAs have been linked to regulation of lipid metabolism and inflammation. METHODS: We fed Lieber-DeCarli alcohol or methionine-choline-deficient (MCD) diets to C57Bl6 and analyzed livers for histopathology, cytokines by ELISA, alanine aminotransferase (ALT) by biochemical assay, and microRNA profile by microarray. RESULTS: Both Lieber-DeCarli and MCD diets lead to development of liver steatosis, liver injury, indicated by increased ALT, and elevated levels of serum TNFalpha, suggesting that animal models portray the pathophysiological features of alcoholic and nonalcoholic fatty liver, respectively. We identified that Lieber-deCarli diet up-regulated 1% and down-regulated 1% of known miRNA; MCD diet up-regulated 3% and down-regulated 1% of known miRNA, compared to controls. Of miRNAs that changed expression levels, 5 miRNAs were common in alcoholic and nonalcoholic fatty livers: the expression of both miR-705 and miR-1224 was increased after Lieber-DeCarli or MCD diet feeding. In contrast, miR-182, miR-183, and miR-199a-3p were down-regulated in Lieber-deCarli feeding, while MCD diet lead to their up-regulation, compared to corresponding controls. CONCLUSIONS: Our findings indicate etiology-specific changes in miRNA expression profile during steatohepatitis models, which opens new avenues for research in the pathophysiology of alcoholic and nonalcoholic fatty liver disease.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Deficiencia de Colina/genética , Deficiencia de Colina/patología , Etanol/toxicidad , Hígado Graso Alcohólico/genética , Hígado Graso/genética , Metionina/deficiencia , MicroARNs/biosíntesis , MicroARNs/genética , Animales , Citocinas/sangre , Dieta , Hígado Graso/patología , Hígado Graso Alcohólico/patología , Femenino , Inflamación/genética , Inflamación/patología , Metabolismo de los Lípidos/genética , Hígado/patología , Pruebas de Función Hepática , Ratones , Ratones Endogámicos C57BL
19.
J Leukoc Biol ; 84(3): 614-22, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18495782

RESUMEN

The mechanism behind the apparent lack of effective antiviral immune response in patients with chronic hepatitis C virus (HCV) infection is poorly understood. Although multiple levels of abnormalities have been identified in innate and adaptive immunity, it remains unclear if any of the subpopulations of T cells with regulatory capacity (Tregs) contribute to the induction and maintenance of HCV persistence. In this review, we summarize the current knowledge about Tregs as they relate to HCV infection.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/inmunología , Inmunidad Celular/inmunología , Linfocitos T Reguladores/inmunología , Animales , Hepacivirus/patogenicidad , Hepatitis C/virología , Humanos , Proteínas del Núcleo Viral/inmunología
20.
Gastroenterology ; 133(5): 1627-36, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17916356

RESUMEN

BACKGROUND & AIMS: Persistent inflammation contributes to progression of liver damage in chronic HCV (cHCV) infection. Repeated exposure to toll-like receptor (TLR) ligands results in tolerance, a protective mechanism aimed at limiting inflammation. METHODS: Monocytes/macrophages were repeatedly stimulated via proinflammatory cytokine-inducing TLRs and evaluated for activation markers. RESULTS: Unlike monocytes of controls or patients with nonalcoholic steatohepatitis, the monocytes of cHCV patients were hyperresponsive and failed to show homo- or heterotolerance to TLR ligands, manifested by elevated tumor necrosis factor (TNF)-alpha production. Serum levels of interferon (IFN)-gamma, endotoxin (TLR4 ligand), and HCV core protein (TLR2 ligand) were elevated in cHCV patients suggesting potential mechanisms for in vivo monocyte preactivation. Treatment of normal monocytes with IFN-gamma resulted in loss of tolerance to lipopolysaccharide (LPS) or HCV core protein. Furthermore, we found increased levels of MyD88-IRAK1 complexes and nuclear factor (NF)-kappaB activity both in monocytes of cHCV patients and in normal monocytes that lost TLR tolerance after IFN-gamma + LPS pretreatment. In vitro differentiation of TLR non-tolerant cHCV monocytes into macrophages restored their capacity to exhibit TLR tolerance to LPS and HCV core protein, and this could be reversed by administration of IFN-gamma. cHCV patients exhibited increased TNF-alpha in the circulation and in the liver. In cHCV livers, we found Kupffer cell/macrophage activation indicated by increased CD163 and CD33 expression. CONCLUSIONS: We identified that host-derived factors (IFN-gamma and endotoxin) and viral factors (HCV core protein) act in tandem to induce and maintain monocyte/macrophage activation, thus favoring persistent inflammation in patients with cHCV infection.


Asunto(s)
Endotoxinas/farmacología , Hepatitis C Crónica/inmunología , Interferón gamma/farmacología , Activación de Macrófagos/efectos de los fármacos , Receptores Toll-Like/inmunología , Proteínas del Núcleo Viral/farmacología , Adulto , Estudios de Casos y Controles , Endotoxinas/sangre , Femenino , Hepacivirus/metabolismo , Hepatitis C Crónica/sangre , Humanos , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/sangre , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Ligandos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , FN-kappa B/metabolismo , Receptor Toll-Like 2/sangre , Receptor Toll-Like 4/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas del Núcleo Viral/sangre
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda