Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G66-G79, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29597356

RESUMEN

Complement plays a crucial role in microbial defense and clearance of apoptotic cells. Emerging evidence suggests complement is an important contributor to alcoholic liver disease. While complement component 1, Q subcomponent (C1q)-dependent complement activation contributes to ethanol-induced liver injury, the role of the alternative pathway in ethanol-induced injury is unknown. Activation of complement via the classical and alternative pathways was detected in alcoholic hepatitis patients. Female C57BL/6J [wild type (WT)], C1q-deficient ( C1qa-/-, lacking classical pathway activation), complement protein 4-deficient ( C4-/-, lacking classical and lectin pathway activation), complement factor D-deficient ( FD-/-, lacking alternative pathway activation), and C1qa/FD-/- (lacking classical and alternative pathway activation) mice were fed an ethanol-containing liquid diet or pair-fed control diet for 4 or 25 days. Following chronic ethanol exposure, liver injury, steatosis, and proinflammatory cytokine expression were increased in WT but not C1qa-/-, C4-/-, or C1qa/FD-/- mice. In contrast, liver injury, steatosis, and proinflammatory mediators were robustly increased in ethanol-fed FD-/- mice compared with WT mice. Complement activation, assessed by hepatic accumulation of C1q and complement protein 3 (C3) cleavage products (C3b/iC3b/C3c), was evident in livers of WT mice in response to both short-term and chronic ethanol. While C1q accumulated in ethanol-fed FD-/- mice (short term and chronic), C3 cleavage products were detected after short-term but not chronic ethanol. Consistent with impaired complement activation, chronic ethanol induced the accumulation of apoptotic cells and fibrogenic responses in the liver of FD-/- mice. These data highlight the protective role of complement factor D (FD) and suggest that FD-dependent amplification of complement is an adaptive response that promotes hepatic healing and recovery in response to chronic ethanol. NEW & NOTEWORTHY Complement, a component of the innate immune system, is an important pathophysiological contributor to ethanol-induced liver injury. We have identified a novel role for factor D, a component of the alternative pathway, in protecting the liver from ethanol-induced inflammation, accumulation of apoptotic hepatocytes, and profibrotic responses. These data indicate a dual role of complement with regard to inflammatory and protective responses and suggest that accumulation of apoptotic cells impairs hepatic healing/recovery during alcoholic liver disease.


Asunto(s)
Etanol , Inflamación , Hepatopatías Alcohólicas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Depresores del Sistema Nervioso Central/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Factor D del Complemento/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Vía Alternativa del Complemento/fisiología , Citocinas/inmunología , Etanol/metabolismo , Etanol/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/prevención & control , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/metabolismo
2.
Alcohol Clin Exp Res ; 36(7): 1139-47, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22273278

RESUMEN

BACKGROUND: Correlative evidence indicates that apoptosis is associated with the progression of alcoholic liver disease. If apoptosis contributes to ethanol (EtOH)-induced steatohepatitis and/or fibrosis, then mice deficient in Bid, a key pro-apoptotic Bcl-2 family member, or mice treated with a pan-caspase inhibitor (VX166) should be resistant to EtOH-induced liver injury. METHODS: This hypothesis was tested in mice using a model of chronic, heavy EtOH-induced liver injury, as well as in a model in which moderate EtOH feeding accelerated the appearance of early markers of hepatic fibrosis in response to acute carbon tetrachloride (CCl(4) ) exposure. RESULTS: Chronic EtOH feeding to mice increased TUNEL- and cytokeratin-18-positive cells in the liver, as well as the expression of receptor-interacting protein kinase 3 (RIP3), a marker of necroptosis. In this model, Bid-/- mice or wild-type mice treated with VX166 were protected from EtOH-induced apoptosis, but not EtOH-induced RIP3 expression. Bid deficiency or inhibition of caspase activity did not protect mice from EtOH-induced increases in plasma alanine and aspartate amino transferase activity, steatosis, or mRNA expression of some inflammatory cytokines. Moderate EtOH feeding to mice enhanced the response of mice to acute CCl(4) exposure, resulting in increased expression of α-smooth muscle actin and accumulation of extracellular matrix protein. VX166-treatment attenuated EtOH-mediated acceleration of these early indicators of CCl(4) -induced hepatic fibrosis, decreasing the expression of α-smooth muscle actin, and the accumulation of extracellular matrix protein. CONCLUSIONS: EtOH-induced apoptosis of hepatocytes was mediated by Bid. Apoptosis played a critical role in the accelerating the appearance of early markers of CCl(4) -induced fibrosis by moderate EtOH but did not contribute to EtOH-induced hepatocyte injury, steatosis, or expression of mRNA for some inflammatory cytokines.


Asunto(s)
Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/deficiencia , Tetracloruro de Carbono/toxicidad , Etanol/toxicidad , Hígado Graso Alcohólico/patología , Mediadores de Inflamación/toxicidad , Hepatopatías Alcohólicas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/prevención & control , Femenino , Hepatitis Alcohólica/metabolismo , Hepatitis Alcohólica/patología , Hepatitis Alcohólica/prevención & control , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática Alcohólica/metabolismo , Cirrosis Hepática Alcohólica/patología , Cirrosis Hepática Alcohólica/prevención & control , Hepatopatías Alcohólicas/patología , Hepatopatías Alcohólicas/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G697-702, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21350183

RESUMEN

Obesity is a global epidemic with more than 1 billion overweight adults and at least 300 million obese patients worldwide. Diabetes is characterized by a defect in insulin secretion or a decrease in sensitivity to insulin, which results in elevated fasting blood glucose. Both obesity and elevated fasting glucose are risk factors for nonalcoholic fatty liver disease, a disease spectrum that includes hepatic steatosis (nonalcoholic fatty liver), nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Increased adiposity and insulin resistance contribute to the progression from NASH to fibrosis through the development of a profibrotic mileau in the liver, including increased hepatocellular death, increased reactive oxygen species generation, and an altered adipokine/cytokine balance. This review will summarize recent advances in our understanding of the pathological interactions among excessive fat accumulation, insulin resistance, and hepatic fibrogenesis and discuss specific molecular pathways that may be of interest in the development of therapeutic interventions to prevent and/or reverse hepatic fibrosis.


Asunto(s)
Complicaciones de la Diabetes/patología , Diabetes Mellitus/patología , Cirrosis Hepática/complicaciones , Obesidad/complicaciones , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Animales , Matriz Extracelular/patología , Hígado Graso/complicaciones , Hígado Graso/patología , Humanos , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico , Obesidad/patología , Especies Reactivas de Oxígeno/metabolismo
4.
Mol Immunol ; 72: 9-18, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922040

RESUMEN

BACKGROUND: Ethanol feeding in mice activates complement via C1q binding to apoptotic cells in the liver; complement contributes to ethanol-induced inflammation and injury. Despite the critical role of C1q in ethanol-induced injury, the mechanism by which ethanol activates C1q remains poorly understood. Secretory IgM (sIgM), traditionally considered to act as an anti-microbial, also has critical housekeeping functions, facilitating clearance of apoptotic cells, at least in part through activation of C1q. Therefore, we hypothesized that (1) ethanol-induced apoptosis in the liver recruits sIgM, facilitating the activation of C1q and complement and (2) C1INH (C1 esterase inhibitor), which inhibits C1 functional activity, prevents complement activation and decreases ethanol-induced liver injury. METHODS: Female C57BL/6 wild-type, C1qa(-/-), BID(-/-) and sIgM(-/-) mice were fed ethanol containing liquid diets or pair-fed control diets. C1INH or vehicle was given via tail vein injection to ethanol- or pair-fed wild-type mice at 24 and 48h prior to euthanasia. RESULTS: Ethanol exposure increased apoptosis in the liver, as well as the accumulation of IgM in the liver. In the early stages of ethanol feeding, C1q co-localized with IgM in the peri-sinusoidal space of the liver and accumulation of IgM and C3b was dependent on ethanol-induced BID-dependent apoptosis. sIgM(-/-) mice were protected from both ethanol-induced activation of complement and early ethanol-induced liver injury when compared to wild-type mice. Treatment with C1INH also decreased hepatic C3b deposition and ethanol-induced injury. CONCLUSION: These data indicate that sIgM contributes to activation of complement and ethanol-induced increases in inflammatory cytokine expression and hepatocyte injury in the early stages of ethanol-induced liver injury.


Asunto(s)
Apoptosis , Activación de Complemento , Complemento C1q/inmunología , Inmunoglobulina M/inmunología , Hepatopatías Alcohólicas/inmunología , Animales , Proteínas Inactivadoras del Complemento 1/farmacología , Proteína Inhibidora del Complemento C1 , Complemento C1q/antagonistas & inhibidores , Citocinas/biosíntesis , Femenino , Hepatocitos/inmunología , Hepatopatías Alcohólicas/patología , Ratones , Ratones Endogámicos C57BL , Solubilidad
5.
Clin Liver Dis ; 18(1): 157-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24274871

RESUMEN

Alcoholic liver disease (ALD) remains a major cause of chronic liver diseases and liver failure. Population-based prospective studies and patient cohort studies have demonstrated that obesity and the metabolic syndrome exacerbate progression of ALD and increase hepatocellular carcinoma (HCC) incidence and mortality. Emerging evidence also suggests a synergism between alcohol and obesity in mortality and HCC incidence. Recognition of these increased risks and detection of early-stage liver disease may offer the opportunity to address these modifiable risk factors and prevent disease progression in these patients.


Asunto(s)
Hepatopatías Alcohólicas/complicaciones , Síndrome Metabólico/complicaciones , Obesidad/complicaciones , Carcinoma Hepatocelular/etiología , Complicaciones de la Diabetes/etiología , Progresión de la Enfermedad , Humanos , Hepatopatías Alcohólicas/mortalidad , Hepatopatías Alcohólicas/terapia , Neoplasias Hepáticas/etiología , Factores de Riesgo
6.
Pharmacol Res Perspect ; 2(5): e00061, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25089199

RESUMEN

The hypoxia-sensing transcriptional factor HIF1α is implicated in a variety of hepato-pathological conditions; however, the contribution of hepatocyte-derived HIF1α during progression of alcoholic liver injury is still controversial. HIF1α induces a variety of genes including those involved in apoptosis via p53 activation. Increased hepatocyte apoptosis is critical for progression of liver inflammation, stellate cell activation and fibrosis. Using hepatocyte-specific HIF1α-deficient mice (ΔHepHIF1α-/-), here we investigated the contribution of HIF1α to ethanol-induced hepatocyte apoptosis and its role in amplification of fibrosis after carbon tetrachloride (CCl4) exposure. Moderate ethanol feeding (11% of Kcal) induced accumulation of hypoxia-sensitive pimonidazole adducts and HIF1α expression in the liver within 4 days of ethanol feeding. Chronic CCl4 treatment increased M30-positive cells, a marker of hepatocyte apoptosis in pair-fed control mice. Concomitant ethanol feeding (11% of Kcal) amplified CCl4-induced hepatocyte apoptosis in livers of wild-type mice, associated with elevated p53K386acetylation, PUMA expression and Ly6c+ cell infiltration. Subsequent to increased apoptosis, ethanol enhanced induction of pro-fibrotic markers including stellate cell activation, collagen 1 expression and extracellular matrix deposition, following CCl4 exposure. Ethanol-induced exacerbation of hepatocyte apoptosis, p53K386 acetylation and PUMA expression following CCl4 exposure was attenuated in livers of ΔHepHIF1α-/- mice. This protection was also associated with a reduction in Ly6c+ cell infiltration and decreased fibrosis in livers of ΔHepHIF1α-/- mice. In summary, these results indicate that moderate ethanol exposure leads to hypoxia/HIF1α-mediated signaling in hepatocytes and induction of p53-dependent apoptosis of hepatocytes, resulting in increased hepatic fibrosis during chronic CCl4 exposure.

7.
PLoS One ; 8(7): e69114, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874883

RESUMEN

UNLABELLED: The effect of moderate alcohol consumption on liver fibrosis is not well understood, but evidence suggests that adenosine may play a role in mediating the effects of moderate ethanol on tissue injury. Ethanol increases the concentration of adenosine in the liver. Adenosine 2A receptor (A2AR) activation is known to enhance hepatic stellate cell (HSC) activation and A2AR deficient mice are protected from fibrosis in mice. Making use of a novel mouse model of moderate ethanol consumption in which female C57BL/6J mice were allowed continued access to 2% (vol/vol) ethanol (11% calories) or pair-fed control diets for 2 days, 2 weeks or 5 weeks and superimposed with exposure to CCl4, we tested the hypothesis that moderate ethanol consumption increases fibrosis in response to carbon tetrachloride (CCl4) and that treatment of mice with an A2AR antagonist prevents and/or reverses this ethanol-induced increase in liver fibrosis. Neither the expression or activity of CYP2E1, required for bio-activation of CCl4, nor AST and ALT activity in the plasma were affected by ethanol, indicating that moderate ethanol did not increase the direct hepatotoxicity of CCl4. However, ethanol feeding enhanced HSC activation and exacerbated liver fibrosis upon exposure to CCl4. This was associated with an increased sinusoidal angiogenic response in the liver. Treatment with A2AR antagonist both prevented and reversed the ability of ethanol to exacerbate liver fibrosis. CONCLUSION: Moderate ethanol consumption exacerbates hepatic fibrosis upon exposure to CCl4. A2AR antagonism may be a potential pharmaceutical intervention to decrease hepatic fibrosis in response to ethanol.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Bebidas Alcohólicas/efectos adversos , Tetracloruro de Carbono/toxicidad , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/prevención & control , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Femenino , Cirrosis Hepática/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Aceite de Oliva , Aceites de Plantas , Purinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA