Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Drug Metab Dispos ; 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38050055

RESUMEN

The role of inflammatory cells and other components of the immune system in acetaminophen (APAP)-induced liver injury and repair has been extensively investigated. Although this has resulted in a wealth of information regarding the function and regulation of immune cells in the liver after injury, apparent contradictions have fueled controversy around the central question of whether the immune system is beneficial or detrimental after APAP overdose. Ultimately, this may not be a simple assignment of "good" or "bad." Clinical studies have clearly demonstrated an association between immune dysregulation and a poor outcome in patients with severe liver damage/liver failure induced by APAP overdose. To date, studies in mice have not uniformly replicated this connection. The apparent disconnect between clinical and experimental studies has perhaps stymied progress and further complicated investigation of the immune system in APAP-induced liver injury. Mouse models are often dismissed as not recapitulating the clinical scenario. Moreover, clinical investigation is most often focused on the most severe APAP overdose patients, those with liver failure. Notably, recent studies have made it apparent that the functional role of the immune system in the pathogenesis of APAP-induced liver injury is highly context dependent and greatly influenced by the experimental conditions. In this review, we highlight some of these recent findings, and suggest strategies seeking to resolve and build on existing disconnects in the literature. Significance Statement Acetaminophen overdose is the most frequent cause of acute liver failure in the United States. Studies indicate that dysregulated innate immunity contributes to the transition from acute liver injury to acute liver failure. In this review, we discuss the evidence for this and the potential underlying causes.

2.
Am J Pathol ; 189(10): 1986-2001, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31381887

RESUMEN

Kupffer cells and monocyte-derived macrophages are critical for liver repair after acetaminophen (APAP) overdose. These cells produce promitogenic cytokines and growth factors, and they phagocytose dead cell debris, a process that is critical for resolution of inflammation. The factors that regulate these dynamic functions of macrophages after APAP overdose, however, are not fully understood. We tested the hypothesis that the fibrinolytic enzyme, plasmin, is a key regulator of macrophage function after APAP-induced liver injury. In these studies, inhibition of plasmin in mice with tranexamic acid delayed up-regulation of proinflammatory cytokines after APAP overdose. In culture, plasmin directly, and in synergy with high-mobility group B1, stimulated Kupffer cells and bone marrow-derived macrophages to produce cytokines by a mechanism that required NF-κB. Inhibition of plasmin in vivo also prevented trafficking of monocyte-derived macrophages into necrotic lesions after APAP overdose. This prevented phagocytic removal of dead cells, prevented maturation of monocyte-derived macrophages into F4/80-expressing macrophages, and prevented termination of proinflammatory cytokine production. Our studies reveal further that phagocytosis is an important stimulus for cessation of proinflammatory cytokine production as treatment of proinflammatory, monocyte-derived macrophages, isolated from APAP-treated mice, with necrotic hepatocytes decreased expression of proinflammatory cytokines. Collectively, these studies demonstrate that plasmin is an important regulator of macrophage function after APAP overdose.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Fibrinolisina/metabolismo , Macrófagos del Hígado/patología , Macrófagos/patología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Sobredosis de Droga , Mediadores de Inflamación/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis
3.
Cytokine ; 127: 154986, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31951966

RESUMEN

INTRODUCTION: Cxcl12, or stromal-derived factor-1, is a chemokine produced by several hepatic cell types, including hepatocytes, after liver injury and surgical resection. Studies have revealed that Cxcl12 is important for regeneration of the liver after surgical resection and for development of liver fibrosis during chronic liver injury. While the function of Cxcl12 in the liver is well established, the mechanism by which Cxcl12 is upregulated is not fully understood. Because regions of hypoxia develop in the liver following injury, we tested the hypothesis that hypoxia upregulates Cxcl12 in hepatocytes by a hypoxia-inducible factor (HIF)-dependent mechanism. METHODS: To test this hypothesis, primary mouse hepatocytes were isolated from the livers of HIF-1α-deficient mice or HIF-1ß-deficient mice and exposed to 1% oxygen. Cxcl12 expression was increased following exposure of primary mouse hepatocytes to 1% oxygen. Previously we have shown, that in addition to HIFs, transforming growth factor-ß is required for upregulation of a subset of genes in hypoxic hepatocytes. To examine the role of TGF-ß in regulation of Cxcl12 during hypoxia, hepatocytes were pretreated with the TGF-ß receptor I inhibitor, SB431542. RESULTS: Upregulation of Cxcl12 by hypoxia was partially prevented in hepatocytes from HIF-1α-deficient mice and completely prevented in hepatocytes from HIF-1ß-deficient hepatocytes. This suggests that under hypoxic conditions, both HIF-1α and HIF-2α regulate Cxcl12 in hepatocytes. Pretreatment of hepatocytes with SB431542 completely prevented upregulation Cxcl12 by hypoxia. Further, treatment of hepatocytes with recombinant TGF-ß1 upregulated Cxcl12 in hepatocytes cultured in room air. CONCLUSION: Collectively, these studies demonstrate that hypoxia upregulates Cxcl12 in primary mouse hepatocytes by a mechanism that involves HIFs and TGF-ß.


Asunto(s)
Quimiocina CXCL12/metabolismo , Hepatocitos/metabolismo , Hipoxia/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/fisiología , Animales , Células Cultivadas , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Activación Transcripcional/fisiología
4.
Pharmacol Res ; 104: 9-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26706784

RESUMEN

For many years, bile acids were thought to only function as detergents which solubilize fats and facilitate the uptake of fat-soluble vitamins in the intestine. Many early observations; however, demonstrated that bile acids regulate more complex processes, such as bile acids synthesis and immune cell function through activation of signal transduction pathways. These studies were the first to suggest that receptors may exist for bile acids. Ultimately, seminal studies by many investigators led to the discovery of several bile acid-activated receptors including the farnesoid X receptor, the vitamin D receptor, the pregnane X receptor, TGR5, α5 ß1 integrin, and sphingosine-1-phosphate receptor 2. Several of these receptors are expressed outside of the gastrointestinal system, indicating that bile acids may have diverse functions throughout the body. Characterization of the functions of these receptors over the last two decades has identified many important roles for these receptors in regulation of bile acid synthesis, transport, and detoxification; regulation of glucose utilization; regulation of fatty acid synthesis and oxidation; regulation of immune cell function; regulation of energy expenditure; and regulation of neural processes such as gastric motility. Through these many functions, bile acids regulate many aspects of digestion ranging from uptake of essential vitamins to proper utilization of nutrients. Accordingly, within a short time period, bile acids moved beyond simple detergents and into the realm of complex signaling molecules. Because of the important processes that bile acids regulate through activation of receptors, drugs that target these receptors are under development for the treatment of several diseases, including cholestatic liver disease and metabolic syndrome. In this review, we will describe the various bile acid receptors, the signal transduction pathways activated by these receptors, and briefly discuss the physiological processes that these receptors regulate.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Humanos , Transducción de Señal
5.
J Immunol ; 192(8): 3847-3857, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24639359

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is activated in hepatic stellate cells (HSCs) by hypoxia and regulates genes important for tissue repair. Whether HIF-1α is activated in HSCs after acute injury and contributes to liver regeneration, however, is not known. To investigate this, mice were generated with reduced levels of HIF-1α in HSCs by crossing HIF-1α floxed mice with mice that express Cre recombinase under control of the glial fibrillary acidic protein (GFAP) promoter (i.e., HIF-1α-GFAP Cre+ mice). These mice and control mice (i.e., HIF-1α-GFAP Cre- mice) were treated with a single dose of carbon tetrachloride, and liver injury and repair were assessed. After carbon tetrachloride, HIF-1α was activated in HSCs. Although liver injury was not different between the two strains of mice, during resolution of injury, clearance of necrotic cells was decreased in HIF-1α-GFAP Cre+ mice. In these mice, the persistence of necrotic cells stimulated a fibrotic response characterized by extensive collagen deposition. Hepatic accumulation of macrophages, which clear necrotic cells from the liver after carbon tetrachloride, was not affected by HIF-1α deletion in HSCs. Conversion of macrophages to M1-like, proinflammatory macrophages, which have increased phagocytic activity, however, was reduced in HIF-1α-GFAP Cre+ mice as indicated by a decrease in proinflammatory cytokines and a decrease in the percentage of Gr1(hi) macrophages. Collectively, these studies have identified a novel function for HSCs and HIF-1α in orchestrating the clearance of necrotic cells from the liver and demonstrated a key role for HSCs in modulating macrophage phenotype during acute liver injury.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Fenotipo , Animales , Tetracloruro de Carbono/farmacología , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Eliminación de Gen , Células Estrelladas Hepáticas/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Macrófagos/patología , Ratones , Ratones Transgénicos , Necrosis , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
6.
Am J Pathol ; 183(5): 1498-1507, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24012680

RESUMEN

During obstructive cholestasis, increased concentrations of bile acids activate ERK1/2 in hepatocytes, which up-regulates early growth response factor 1, a key regulator of proinflammatory cytokines, such as macrophage inflammatory protein 2 (MIP-2), which, in turn, exacerbates cholestatic liver injury. Recent studies have indicated that IL-17A contributes to hepatic inflammation during obstructive cholestasis, suggesting that bile acids and IL-17A may interact to regulate hepatic inflammatory responses. We treated mice with an IL-17A neutralizing antibody or control IgG and subjected them to bile duct ligation. Neutralization of IL-17A prevented up-regulation of proinflammatory cytokines, hepatic neutrophil accumulation, and liver injury, indicating an important role for IL-17A in neutrophilic inflammation during cholestasis. Treatment of primary mouse hepatocytes with taurocholic acid (TCA) increased the expression of MIP-2. Co-treatment with IL-17A synergistically enhanced up-regulation of MIP-2 by TCA. In contrast to MIP-2, IL-17A did not affect up-regulation of Egr-1 by TCA, indicating that IL-17A does not affect bile acid-induced activation of signaling pathways upstream of early growth response factor 1. In addition, bile acids increased expression of IL-23, a key regulator of IL-17A production in hepatocytes in vitro and in vivo. Collectively, these data identify bile acids as novel triggers of the IL-23/IL-17A axis and suggest that IL-17A promotes hepatic inflammation during cholestasis by synergistically enhancing bile acid-induced production of proinflammatory cytokines by hepatocytes.


Asunto(s)
Colestasis/metabolismo , Colestasis/patología , Inflamación/metabolismo , Inflamación/patología , Interleucina-17/metabolismo , Actinas/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Ácidos y Sales Biliares/administración & dosificación , Conductos Biliares/efectos de los fármacos , Conductos Biliares/patología , Biomarcadores/metabolismo , Recuento de Células , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Colestasis/complicaciones , Colágeno Tipo I/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Inflamación/complicaciones , Interleucina-23/genética , Interleucina-23/metabolismo , Ligadura , Hígado/efectos de los fármacos , Hígado/lesiones , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Neutralización , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Ácido Taurocólico/farmacología , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Pathol ; 181(4): 1287-95, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22841818

RESUMEN

Thrombin generation is increased in patients with nonalcoholic fatty liver disease (NAFLD) and in mouse models of diet-induced obesity. Deficiency in the thrombin receptor protease activated receptor-1 reduces hepatic inflammation and steatosis in mice fed a Western diet. However, it is currently unclear whether thrombin inhibitors can modify the pathogenesis of established NAFLD. We tested the hypothesis that thrombin inhibition could reverse hepatic steatosis and inflammation in mice with established diet-induced NAFLD. Low-density lipoprotein receptor-deficient LDLr(-/-) mice were fed a control diet or a Western diet for 19 weeks. Mice were given the direct thrombin inhibitor argatroban ∼15 mg/kg/day or its vehicle via a miniosmotic pump for the final 4 weeks of the study. Argatroban administration significantly reduced hepatic proinflammatory cytokine expression and reduced macrophage and neutrophil accumulation in livers of mice fed a Western diet. Argatroban did not significantly impact hepatic steatosis, as indicated by histopathology, Oil Red O staining, and hepatic triglyceride levels. Argatroban reduced serum triglyceride and cholesterol levels in mice fed a Western diet. Argatroban reduced both α-smooth muscle actin expression and Type 1 collagen mRNA levels in livers of mice fed a Western diet, indicating reduced activation of hepatic stellate cells. This study indicates that therapeutic intervention with a thrombin inhibitor attenuates hepatic inflammation and several profibrogenic changes in mice fed a Western diet.


Asunto(s)
Hígado Graso/complicaciones , Hígado Graso/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Ácidos Pipecólicos/farmacología , Ácidos Pipecólicos/uso terapéutico , Trombina/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Colágeno/metabolismo , Dieta , Hígado Graso/sangre , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/sangre , Cirrosis Hepática/complicaciones , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Ácidos Pipecólicos/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo , Sulfonamidas , Trombina/metabolismo , Aumento de Peso/efectos de los fármacos
8.
Front Immunol ; 14: 1303921, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38094302

RESUMEN

Introduction: Systemic levels of the anti-inflammatory cytokine interleukin 10 (IL-10) are highest in acetaminophen (APAP)-induced acute liver failure (ALF) patients with the poorest prognosis. The mechanistic basis for this counterintuitive finding is not known, as induction of IL-10 is hypothesized to temper the pathological effects of immune cell activation. Aberrant production of IL-10 after severe liver injury could conceivably interfere with the beneficial, pro-reparative actions of immune cells, such as monocytes. Methods: To test this possibility, we determined whether IL-10 levels are dysregulated in mice with APAP-induced ALF and further evaluated whether aberrant production of IL-10 prevents monocyte recruitment and/or the resolution of necrotic lesions by these cells. Results: Our studies demonstrate that in mice challenged with 300 mg/kg acetaminophen (APAP), a hepatotoxic dose of APAP that fails to produce ALF (i.e., APAP-induced acute liver injury; AALI), Ly6Chi monocytes were recruited to the liver and infiltrated the necrotic lesions by 48 hours coincident with the clearance of dead cell debris. At 72 hours, IL-10 was upregulated, culminating in the resolution of hepatic inflammation. By contrast, in mice treated with 600 mg/kg APAP, a dose that produces clinical features of ALF (i.e., APAP-induced ALF; AALF), IL-10 levels were markedly elevated by 24 hours. Early induction of IL-10 was associated with a reduction in the hepatic numbers of Ly6Chi monocytes resulting in the persistence of dead cell debris. Inhibition of IL-10 in AALF mice, beginning at 24 hours after APAP treatment, increased the hepatic numbers of monocytes which coincided with a reduction in the necrotic area. Moreover, pharmacologic elevation of systemic IL-10 levels in AALI mice reduced hepatic myeloid cell numbers and increased the area of necrosis. Discussion: Collectively, these results indicate that during ALF, aberrant production of IL-10 disrupts the hepatic recruitment of monocytes, which prevents the clearance of dead cell debris. These are the first studies to document a mechanistic basis for the link between high IL-10 levels and poor outcome in patients with ALF.


Asunto(s)
Acetaminofén , Fallo Hepático Agudo , Humanos , Animales , Ratones , Acetaminofén/efectos adversos , Interleucina-10 , Monocitos , Necrosis/inducido químicamente
9.
J Pharmacol Exp Ther ; 341(2): 307-16, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22271822

RESUMEN

Macrophages play an integral role in the development of liver fibrosis by releasing mediators, such as platelet-derived growth factor-B (PDGF-B) and transforming growth factor-ß1, which stimulate hepatic stellate cell proliferation, chemotaxis, and collagen production. However, the mechanism by which chronic liver injury stimulates macrophages to release these mediators is not completely understood. We tested the hypothesis that chronic liver injury activates hypoxia-inducible factor (HIF) transcription factors in macrophages that regulate the production of mediators that promote fibrosis. To test this hypothesis, Cre/lox technology was used to generate myeloid cell-specific HIF-1α or HIF-1ß knockout mice. When these mice were subjected to bile duct ligation (BDL), levels of α-smooth muscle actin and type I collagen in the liver were reduced compared with those of mice with normal levels of HIFs. The deficiency of HIFs in macrophages did not affect liver injury or inflammation after BDL but reduced PDGF-B mRNA and protein, suggesting that HIF activation in macrophages may promote fibrosis by regulating the production of PDGF-B. Consistent with a role for HIFs in liver fibrosis in cholestatic liver disease, nuclear HIF-1α protein was present in macrophages, hepatocytes, and fibroblasts in the livers from patients with primary biliary cirrhosis and primary sclerosing cholangitis. These studies demonstrate that HIFs are important regulators of profibrotic mediator production by macrophages during the development of liver fibrosis and suggest that HIFs may be a novel therapeutic target for the treatment of chronic liver disease in patients.


Asunto(s)
Colestasis/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Cirrosis Hepática/patología , Células Mieloides/metabolismo , Actinas/genética , Actinas/metabolismo , Adulto , Anciano , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Conductos Biliares/metabolismo , Hipoxia de la Célula/fisiología , Colestasis/genética , Colestasis/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Femenino , Fibroblastos/metabolismo , Hepatocitos/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación/genética , Inflamación/metabolismo , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Células Mieloides/patología , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/genética
10.
Am J Pathol ; 178(1): 175-86, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224055

RESUMEN

Inflammation contributes to liver injury during cholestasis. The mechanism by which cholestasis initiates an inflammatory response in the liver, however, is not known. Two hypotheses were investigated in the present studies. First, activation of Toll-like receptor 4 (TLR4), either by bacterial lipopolysaccharide or by damage-associated molecular pattern molecules released from dead hepatocytes, triggers an inflammatory response. Second, bile acids act as inflammagens, and directly activate signaling pathways in hepatocytes that stimulate production of proinflammatory mediators. Liver inflammation was not affected in lipopolysaccharide-resistant C3H/HeJ mice after bile duct ligation, indicating that Toll-like receptor 4 is not required for initiation of inflammation. Treatment of hepatocytes with bile acids did not directly cause cell toxicity but increased the expression of numerous proinflammatory mediators, including cytokines, chemokines, adhesion molecules, and other proteins that influence immune cell levels and function. Up-regulation of several of these genes in hepatocytes and in the liver after bile duct ligation required early growth response factor-1, but not farnesoid X receptor. In addition, early growth response factor-1 was up-regulated in the livers of patients with cholestasis and correlated with levels of inflammatory mediators. These data demonstrate that Toll-like receptor 4 is not required for the initiation of acute inflammation during cholestasis. In contrast, bile acids directly activate a signaling network in hepatocytes that promotes hepatic inflammation during cholestasis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis/complicaciones , Colestasis/patología , Regulación de la Expresión Génica , Hepatitis/genética , Hepatocitos/metabolismo , Animales , Ácidos y Sales Biliares/farmacología , Supervivencia Celular , Quimiocina CCL7/genética , Quimiocina CXCL2/genética , Colestasis/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Hepatitis/etiología , Hepatitis/inmunología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Molécula 1 de Adhesión Intercelular/genética , Lipopolisacáridos/inmunología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Serpina E2/genética , Receptor Toll-Like 4/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , ras-GRF1/genética
11.
Am J Pathol ; 178(3): 1117-25, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356363

RESUMEN

Chronic cholestatic liver injury induced by cholestasis in rodents is associated with hepatic fibrin deposition, and we found evidence of fibrin deposition in livers of patients with cholestasis. Key components of the fibrinolytic pathway modulate cholestatic liver injury by regulating activation of hepatocyte growth factor. However, the exact role of hepatic fibrin deposition in chronic cholestasis is not known. We tested the hypothesis that fibrinogen (Fbg) deficiency worsens liver injury induced by cholestasis. Fbg-deficient mice (Fbgα(-/-) mice) and heterozygous control mice (Fbgα(+/-) mice) were fed either the control diet or a diet containing 0.025% α-naphthylisothiocyanate (ANIT), which selectively injures bile duct epithelial cells in the liver, for 2 weeks. Hepatic fibrin and collagen deposits were evident in livers of heterozygous control mice fed the ANIT diet. Complete Fbg deficiency was associated with elevated serum bile acids, periportal necrosis, and increased serum alanine aminotransferase activity in mice fed the ANIT diet. Fbg deficiency was associated with enhanced hepatic expression of the transcription factor early growth response-1 (Egr-1) and enhanced induction of genes encoding the Egr-1-regulated proinflammatory chemokines monocyte chemotactic protein-1, KC growth-regulated protein, and macrophage inflammatory protein-2. Interestingly, peribiliary collagen deposition was not evident near necrotic areas in Fbg-deficient mice. The results suggest that in this model of chronic cholestasis, fibrin constrains the release of bile constituents from injured intrahepatic bile ducts, thereby limiting the progression of hepatic inflammation and hepatocellular injury.


Asunto(s)
Afibrinogenemia/complicaciones , Afibrinogenemia/metabolismo , Colestasis/complicaciones , Colestasis/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Fibrinógeno/metabolismo , Hígado/patología , 1-Naftilisotiocianato/administración & dosificación , Afibrinogenemia/patología , Anciano , Animales , Conductos Biliares/patología , Colestasis/inducido químicamente , Colestasis/patología , Enfermedad Crónica , Colágeno/metabolismo , Dieta , Modelos Animales de Enfermedad , Conducta Alimentaria , Femenino , Fibrina/deficiencia , Fibrina/metabolismo , Regulación de la Expresión Génica , Humanos , Hiperplasia , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Hígado/metabolismo , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , Persona de Mediana Edad , Neutrófilos/metabolismo , Xenobióticos
12.
Liver Int ; 32(1): 58-69, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22098667

RESUMEN

BACKGROUND: Cholestatic liver diseases can be caused by genetic defects, drug toxicities, hepatobiliary malignancies or obstruction of the biliary tract. Cholestasis leads to accumulation of bile acids (BAs) in hepatocytes. Direct toxicity of BAs is currently the most accepted hypothesis for cholestatic liver injury. However, information on which bile acids are actually accumulating during cholestasis is limited. AIM: To assess the BA composition in liver and serum after bile duct ligation (BDL) in male C57Bl/6 mice between 6 h and 14 days and evaluate toxicity of the most abundant BAs. RESULTS: Bile acid concentrations increased in liver (27-fold) and serum (1400-fold) within 6 h after surgery and remained elevated up to 14 days. BAs in livers of BDL mice became more hydrophilic than sham controls, mainly because of increased 6ß-hydroxylation and taurine conjugation. Among the eight unconjugated and 16 conjugated BAs identified in serum and liver, only taurocholic acid (TCA), ß-muricholic acid (ßMCA) and TßMCA were substantially elevated representing >95% of these BAs over the entire time course. Although glycochenodeoxycholic acid and other conjugated BAs increased in BDL animals, the changes were several orders of magnitude lower compared with TCA, ßMCA and TßMCA. A mixture of these BAs did not cause apoptosis or necrosis, but induced inflammatory gene expression in cultured murine hepatocytes. CONCLUSION: The concentrations of cytotoxic BAs are insufficient to cause hepatocellular injury. In contrast, TCA, ßMCA and TßMCA are able to induce pro-inflammatory mediators in hepatocytes. Thus, BAs act as inflammagens and not as cytotoxic mediators after BDL in mice.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Extrahepática/sangre , Hígado/metabolismo , Animales , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/farmacología , Conductos Biliares Extrahepáticos/cirugía , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Colestasis Extrahepática/etiología , Colestasis Extrahepática/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Mediadores de Inflamación/metabolismo , Ligadura , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
13.
J Pharmacol Exp Ther ; 339(2): 487-98, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21856859

RESUMEN

Fatty acid-induced lipotoxicity plays a critical role in the pathogenesis of nonalcoholic liver disease. Saturated fatty acids and unsaturated fatty acids have differential effects on cell death and steatosis, but the mechanisms responsible for these differences are not known. Using cultured HepG2 cells and primary mouse hepatocytes, we found that unsaturated and saturated fatty acids differentially regulate autophagy and apoptosis. The unsaturated fatty acid, oleic acid, promoted the formation of triglyceride-enriched lipid droplets and induced autophagy but had a minimal effect on apoptosis. In contrast, the saturated fatty acid, palmitic acid, was poorly converted into triglyceride-enriched lipid droplets, suppressed autophagy, and significantly induced apoptosis. Subsequent studies revealed that palmitic acid-induced apoptosis suppressed autophagy by inducing caspase-dependent Beclin 1 cleavage, indicating cross-talk between apoptosis and autophagy. Moreover, our data suggest that the formation of triglyceride-enriched lipid droplets and induction of autophagy are protective mechanisms against fatty acid-induced lipotoxicity. In line with our in vitro findings, we found that high-fat diet-induced hepatic steatosis was associated with autophagy in the mouse liver. Potential modulation of autophagy may be a novel approach that has therapeutic benefits for obesity-induced steatosis and liver injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Ácidos Grasos Insaturados/farmacología , Hígado Graso/fisiopatología , Hepatocitos/fisiología , Ácido Oléico/farmacología , Ácido Palmítico/farmacología , Adenoviridae/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Caspasa 3/metabolismo , Dieta Alta en Grasa , Ácidos Grasos Insaturados/fisiología , Hígado Graso/patología , Células Hep G2 , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Triglicéridos/análisis
14.
J Pharmacol Exp Ther ; 338(2): 492-502, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21576378

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is a critical transcription factor that controls oxygen homeostasis in response to hypoxia, inflammation, and oxidative stress. HIF has been implicated in the pathogenesis of liver injury in which these events play a role, including acetaminophen (APAP) overdose, which is the leading cause of acute liver failure in the United States. APAP overdose has been reported to activate HIF-1α in mouse livers and isolated hepatocytes downstream of oxidative stress. HIF-1α signaling controls many factors that contribute to APAP hepatotoxicity, including mitochondrial cell death, inflammation, and hemostasis. Therefore, we tested the hypothesis that HIF-1α contributes to APAP hepatotoxicity. Conditional HIF-1α deletion was generated in mice using an inducible Cre-lox system. Control (HIF-1α-sufficient) mice developed severe liver injury 6 and 24 h after APAP overdose (400 mg/kg). HIF-1α-deficient mice were protected from APAP hepatotoxicity at 6 h, but developed severe liver injury by 24 h, suggesting that HIF-1α is involved in the early stage of APAP toxicity. In further studies, HIF-1α-deficient mice had attenuated thrombin generation and reduced plasminogen activator inhibitor-1 production compared with control mice, indicating that HIF-1α signaling contributes to hemostasis in APAP hepatotoxicity. Finally, HIF-1α-deficient animals had decreased hepatic neutrophil accumulation and plasma concentrations of interleukin-6, keratinocyte chemoattractant, and regulated upon activation normal T cell expressed and secreted compared with control mice, suggesting an altered inflammatory response. HIF-1α contributes to hemostasis, sterile inflammation, and early hepatocellular necrosis during the pathogenesis of APAP toxicity.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Hepatocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
15.
Liver Int ; 31(2): 230-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20880076

RESUMEN

BACKGROUND/AIMS: Several studies have shown that regions of hypoxia develop in the liver during chronic injury. Furthermore, it has been demonstrated that hypoxia stimulates the release of mediators from hepatic stellate cells (HSCs) that may affect the progression of fibrosis. The mechanism by which hypoxia modulates gene expression in HSCs is not known. Recent studies demonstrated that the hypoxia-activated transcription factor, hypoxia-inducible factor (HIF)-1α, is critical for the development of fibrosis. Accordingly, the hypothesis was tested that HIF-1α is activated in HSCs and regulates the expression of genes important for HSC activation and liver fibrosis. METHODS: Hepatic stellate cells were isolated from mice and exposed to hypoxia. HIF-1α and HIF-2α activation were measured, and gene expression was analysed by gene array analysis. To identify the genes regulated by HIF-1α, HSCs were isolated from control and HIF-1α-deficient mice. RESULTS: Exposure of primary mouse HSCs to 0.5% oxygen activated HIF-1α and HIF-2α. mRNA levels of numerous genes were increased in HSCs exposed to 0.5% oxygen, many of which are important for HSC function, angiogenesis and collagen synthesis. Of the mRNAs increased, chemokine receptor (Ccr) 1, Ccr5, macrophage migration inhibitory factor, interleukin-13 receptor α1 and prolyl-4-hydroxylase α2 (P4h α2) were completely HIF-1α dependent. Upregulation of the vascular endothelial growth factor and the placental growth factor was partially HIF-1α dependent and upregulation of angiopoietin-like 4 and P4h α1 was HIF-1α independent. CONCLUSIONS: Results from these studies demonstrate that hypoxia, through activation of HIF-1α, regulates the expression of genes that may alter the sensitivity of HSCs to certain activators and chemotaxins, and regulates the expression of genes important for angiogenesis and collagen synthesis.


Asunto(s)
Hipoxia de la Célula/fisiología , Regulación de la Expresión Génica/fisiología , Células Estrelladas Hepáticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Cirrosis Hepática/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Cartilla de ADN/genética , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Cirrosis Hepática/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Receptores CCR1/metabolismo , Receptores CCR5/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Adv Pharmacol ; 91: 213-228, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34099109

RESUMEN

Liver fibrosis is a leading cause of death worldwide, accounting for approximately 2 million deaths annually. Despite its wide prevalence, there are currently no pharmacological therapies that directly reverse the fibrotic process in patients. Studies over the last decade have revealed that liver fibrosis is reversible in patients and in animal models. Further, studies aimed at elucidating the mechanism of fibrosis reversal have revealed that macrophages are central to this process. During resolution of fibrosis, proinflammatory macrophages shift phenotype to pro-resolution macrophages which produce matrix degrading enzymes and mediators that inactivate hepatic stellate cells, the cell type principally involved in matrix production during fibrosis development. Since fibrosis reversal begins when disease-causing macrophages transition to disease-reversing macrophages, studies have focused on identifying pharmacological agents that stimulate this process to occur. If successful, these "drugs" would constitute a first-in-class, macrophage-targeted therapeutic approach to reverse liver fibrosis. In the following review, we summarize the current approaches under investigation to modify macrophage phenotype for liver disease treatment. Further we discuss the potential of other approaches to identify novel macrophage-targeted drugs that modify the phenotype of these cells.


Asunto(s)
Cirrosis Hepática , Macrófagos , Animales , Fibrosis , Células Estrelladas Hepáticas/patología , Humanos , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Macrófagos/patología , Fenotipo
17.
Semin Liver Dis ; 30(2): 195-204, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20422501

RESUMEN

Cholestasis is a reduction in bile flow that occurs from a variety of causes in humans. This produces hepatocellular injury and fibrosis. Considering that there are limited therapies for this disease, there has been interest in understanding the mechanism by which cholestasis produces injury. Studies have demonstrated that oxidative stress occurs in livers of humans with cholestasis. In vitro studies have demonstrated that bile acids kill hepatocytes by a mechanism that depends upon reactive oxygen species (ROS). Further studies, however, have demonstrated that this mechanism is of limited importance in vivo. Cholestasis also initiates an inflammatory response resulting in accumulation of neutrophils in the liver. Inhibition of neutrophil function reduces oxidative stress and liver injury suggesting that neutrophils are an important source of damaging ROS in vivo. Furthermore, inhibition of ROS during cholestasis reduces fibrosis. Collectively, these studies suggest that ROS are important for pathologic changes that occur during cholestasis.


Asunto(s)
Colestasis/fisiopatología , Hígado/fisiopatología , Estrés Oxidativo/fisiología , Animales , Colestasis/complicaciones , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/fisiología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Hígado/metabolismo , Cirrosis Hepática/etiología , Cirrosis Hepática/fisiopatología , Mitocondrias Hepáticas/fisiología , Neutrófilos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , S-Adenosilmetionina/análogos & derivados , S-Adenosilmetionina/fisiología
18.
Toxicol Appl Pharmacol ; 243(1): 63-7, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19931294

RESUMEN

Cholestasis results when excretion of bile acids from the liver is interrupted. Liver injury occurs during cholestasis, and recent studies showed that inflammation is required for injury. Our previous studies demonstrated that early growth response factor-1 (Egr-1) is required for development of inflammation in liver during cholestasis, and that bile acids upregulate Egr-1 in hepatocytes. What remains unclear is the mechanism by which bile acids upregulate Egr-1. Bile acids modulate gene expression in hepatocytes by activating the farnesoid X receptor (FXR) and through activation of mitogen-activated protein kinase (MAPK) signaling. Accordingly, the hypothesis was tested that bile acids upregulate Egr-1 in hepatocytes by FXR and/or MAPK-dependent mechanisms. Deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA) stimulated upregulation of Egr-1 to the same extent in hepatocytes isolated from wild-type mice and FXR knockout mice. Similarly, upregulation of Egr-1 in the livers of bile duct-ligated (BDL) wild-type and FXR knockout mice was not different. Upregulation of Egr-1 in hepatocytes by DCA and CDCA was prevented by the MEK inhibitors U0126 and SL-327. Furthermore, pretreatment of mice with U0126 prevented upregulation of Egr-1 in the liver after BDL. Results from these studies demonstrate that activation of MAPK signaling is required for upregulation of Egr-1 by bile acids in hepatocytes and for upregulation of Egr-1 in the liver during cholestasis. These studies suggest that inhibition of MAPK signaling may be a novel therapy to prevent upregulation of Egr-1 in liver during cholestasis.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Regulación hacia Arriba/efectos de los fármacos , Animales , Ácido Quenodesoxicólico/farmacología , Ácido Desoxicólico/farmacología , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/metabolismo
19.
Liver Int ; 30(5): 669-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20158611

RESUMEN

BACKGROUND/AIMS: During development of liver fibrosis, an important source of myofibroblasts is hepatocytes, which differentiate into myofibroblasts by epithelial to mesenchymal transition (EMT). In epithelial tumours and kidney fibrosis, hypoxia, through activation of hypoxia-inducible factors (HIFs), is an important stimulus of EMT. Our recent studies demonstrated that HIF-1alpha is important for the development of liver fibrosis. Accordingly, the hypothesis was tested that hypoxia stimulates hepatocyte EMT by a HIF-dependent mechanism. METHODS: Primary mouse hepatocytes were exposed to room air or 1% oxygen and EMT evaluated. In addition, bile duct ligations (BDLs) were performed in control and HIF-1alpha-deficient mice and EMT quantified. RESULTS: Exposure of hepatocytes to 1% oxygen increased expression of alpha-smooth muscle actin, vimentin, Snail and fibroblast-specific protein-1 (FSP-1). Levels of E-cadherin and zona occludens-1 were decreased. Upregulation of FSP-1 and Snail by hypoxia was completely prevented in HIF-1beta-deficient hepatocytes and by pretreatment with SB431542, a transforming growth factor-beta (TGF-beta) receptor inhibitor. HIFs promoted TGF-beta-dependent EMT by stimulating activation of latent TGF-beta1. To determine whether HIF-1alpha contributes to EMT in the liver during the development of fibrosis, control and HIF-1alpha-deficient mice were subjected to BDL. FSP-1 was increased to a greater extent in the livers of control mice when compared with HIF-1alpha-deficient mice. CONCLUSIONS: Results from these studies demonstrate that hypoxia stimulates hepatocyte EMT by a HIF and TGF-beta-dependent mechanism. Furthermore, these studies suggest that HIF-1alpha is important for EMT in the liver during the development of fibrosis.


Asunto(s)
Hipoxia de la Célula , Células Epiteliales/patología , Hepatocitos/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Mesodermo/patología , Factor de Crecimiento Transformador beta1/fisiología , Actinas/química , Animales , Cadherinas/análisis , Proteínas de Unión al Calcio/análisis , Queratina-19/análisis , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/análisis , Proteína de Unión al Calcio S100A4 , Proteínas S100 , Proteína de la Zonula Occludens-1
20.
Liver Int ; 30(3): 406-15, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20040050

RESUMEN

BACKGROUND: Maternal metabolic demands change dramatically during the course of gestation and must be co-ordinated with the needs of the developing placenta and fetus. The liver is critically involved in metabolism and other important functions. However, maternal hepatic adjustments to pregnancy are poorly understood. AIM: The aim of the study was to evaluate the influences of pregnancy on the maternal liver growth and gene expression profile. METHODS: Holtzman Sprague-Dawley rats were mated and sacrificed at various stages of gestation and post-partum. The maternal livers were analysed in gravimetric response, DNA content by PicoGreen dsDNA quantitation reagent, hepatocyte ploidy by flow cytometry and hepatocyte proliferation by ki-67 immunostaining. Gene expression profiling of non-pregnant and gestation d18.5 maternal hepatic tissue was analysed using a DNA microarray approach and partially verified by northern blot or quantitative real-time PCR analysis. RESULTS: During pregnancy, the liver exhibited approximately an 80% increase in size, proportional to the increase in body weight of the pregnant animals. The pregnancy-induced hepatomegaly was a physiological event of liver growth manifested by increases in maternal hepatic DNA content and hepatocyte proliferation. Pregnancy did not affect hepatocyte polyploidization. Pregnancy-dependent changes in hepatic expression were noted for a number of genes, including those associated with cell proliferation, cytokine signalling, liver regeneration and metabolism. CONCLUSIONS: The metabolic demands of pregnancy cause marked adjustments in maternal liver physiology. Central to these adjustments are an expansion in hepatic capacity and changes in hepatic gene expression. Our findings provide insights into pregnancy-dependent hepatic adaptations.


Asunto(s)
Adaptación Fisiológica , Perfilación de la Expresión Génica , Hígado/fisiología , Embarazo/genética , Animales , Northern Blotting , Proliferación Celular , ADN/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Antígeno Ki-67/metabolismo , Hígado/anatomía & histología , Hígado/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo/fisiología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA