Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
3.
J Biomed Sci ; 22: 63, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26231226

RESUMEN

BACKGROUND: Hepatic fibrosis is a dynamic process which ultimately leads to cirrhosis in almost patients with chronic hepatic injury. However, progressive fibrosis is a reversible scarring response. Activation of hepatic stellate cells (HSCs) is the prevailing process during hepatic fibrosis. Osthole is an active component majorly contained in the fruit of Cnidium monnieri (L.) Cusson. This present study investigated the therapeutic effects of osthole on rat liver fibrosis and HSC activation. RESULTS: We established the thioacetamide (TAA)-model of Sprague-Dawley (SD) rats to induce hepatic fibrosis. Rats were divided into three groups: control, TAA, and TAA + osthole (10 mg/kg). In vivo, osthole significantly reduced liver injury by diminishing levels of plasma AST and ALT, improving histological architecture, decreasing collagen and α-SMA accumulation, and improving hepatic fibrosis scores. Additionally, osthole reduced the expression of fibrosis-related genes significantly. Osthole also suppressed the production of fibrosis-related cytokines and chemokines. Moreover, nuclear translocation of p65 was significantly suppressed in osthole-treated liver. Osthole also ameliorated TAA-induced injury through reducing cellular oxidation. Osthole showed inhibitory effects in inflammation-related genes and chemokines production as well. In vitro, we assessed osthole effects in activated HSCs (HSC-T6 and LX-2). Osthole attenuated TGF-ß1-induced migration and invasion in HSCs. Furthermore, osthole decreased TNF-α-triggered NF-κB activities significantly. Besides, osthole alleviated TGF-ß1- or ET-1-induced HSCs contractility. CONCLUSIONS: Our study demonstrated that osthole improved TAA-caused liver injury, fibrogenesis and inflammation in rats. In addition, osthole suppressed HSCs activation in vitro significantly.


Asunto(s)
Cumarinas/farmacología , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Actinas/metabolismo , Animales , Citocinas/metabolismo , Células Estrelladas Hepáticas/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Oxidación-Reducción/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tioacetamida/toxicidad
4.
BMC Gastroenterol ; 15: 22, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25886887

RESUMEN

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is associated with hepatic fibrogenesis. Despite well-known cholesterol-lowering action of statins, their mechanisms against NASH-mediated fibrogenesis remain unclear. This study aimed at investigating the in vitro and in vivo anti-fibrotic properties of fluvastatin (Flu). METHODS: Palmitate (PA)-induced changes in intracellular hydrogen peroxide levels in primary rat hepatocytes (PRHs) and human hepatoma cell line (HepG2) were quantified by dichlorofluorescein diacetate (DCF-DA) dye assay, whereas changes in expressions of NADPH oxidase gp91 (phox) subunit, α-smooth muscle actin (α-SMA), and NFκB p65 nuclear translocation were quantified with Western blotting. Quantitative real-time polymerase chain reaction (q-PCR) was used to investigate mRNA expressions of pro-inflammatory genes (ICAM-1, IL-6, TNF-α). Conditioned medium (CM) from PA-treated PRHs was applied to cultured rat hepatic stellate cell line, HSC-T6, with or without Flu-pretreatment for 2 h. Pro-fibrogenic gene expressions (COL1, TIMP-1, TGF-ß1, α-SMA) and protein expression of α-SMA were analyzed. In vivo study using choline-deficient L-amino acid defined (CDAA) diet-induced rat NASH model was performed by randomly assigning Wistar rats (n = 28) to normal controls (n = 4), CDAA diet with vehicles, and CDAA diet with Flu (5 mg/kg or 10 mg/kg) (n = 8 each) through gavage for 4 or 8 weeks. Livers were harvested for histological, Western blot (α-SMA), and q-PCR analyses for expressions of pro-inflammatory (IL-6, iNOS, ICAM-1) and pro-fibrogenic (Col1, α-SMA, TIMP-1) genes. RESULTS: In vitro, Flu (1-20 µM) inhibited PA-induced free-radical production, gp91 (phox) expression, and NFκB p65 translocation in HepG2 and PRHs, while CM-induced α-SMA protein expression and pro-fibrogenic gene expressions in HSC-T6 were suppressed in Flu-pretreated cells compared to those without pretreatment. Moreover, α-SMA protein expression was significantly decreased in HSC-T6 cultured with CM from PA-Flu-treated PRHs compared to those cultured with CM from PA-treated PRHs. Flu also reduced steatosis and fibrosis scores, α-SMA protein expression, mRNA expression of pro-inflammatory and pro-fibrogenic genes in livers of CDAA rats. CONCLUSIONS: We demonstrated PA-induced HSC activation through paracrine effect of hepatocyte in vitro that was significantly suppressed by pre-treating HSC with Flu. In vivo, Flu alleviated steatosis-induced HSC activation and hepatic fibrogenesis through mitigating inflammation and oxidative stress, suggesting possible therapeutic role of Flu against NASH.


Asunto(s)
Ácidos Grasos Monoinsaturados/farmacología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Indoles/farmacología , Cirrosis Hepática/prevención & control , Comunicación Paracrina/efectos de los fármacos , Actinas/genética , Actinas/metabolismo , Animales , Colina/administración & dosificación , Colágeno Tipo I/genética , Medios de Cultivo Condicionados/farmacología , Dieta , Ácidos Grasos Monoinsaturados/uso terapéutico , Fluvastatina , Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/fisiología , Humanos , Peróxido de Hidrógeno/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Indoles/uso terapéutico , Molécula 1 de Adhesión Intercelular/genética , Interleucina-6/genética , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo/efectos de los fármacos , Ácido Palmítico/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Transcripción ReIA/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor de Necrosis Tumoral alfa/genética
5.
PLoS One ; 9(7): e103229, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25076488

RESUMEN

BACKGROUND: Anti-inflammation via inhibition of NF-κB pathways in hepatic stellate cells (HSCs) is one therapeutic approach to hepatic fibrosis. Tanshinone IIA (C19H18O3, Tan IIA) is a lipophilic diterpene isolated from Salvia miltiorrhiza Bunge, with reported anti-inflammatory activity. We tested whether Tan IIA could inhibit HSC activation. MATERIALS AND METHODS: The cell line of rat hepatic stellate cells (HSC-T6) was stimulated with lipopolysaccharide (LPS) (100 ng/ml). Cytotoxicity was assessed by MTT assay. HSC-T6 cells were pretreated with Tan IIA (1, 3 and 10 µM), then induced by LPS (100 ng/ml). NF-κB activity was evaluated by the luciferase reporter gene assay. Western blotting analysis was performed to measure NF-κB-p65, and phosphorylations of MAPKs (ERK, JNK, p38). Cell chemotaxis was assessed by both wound-healing assay and trans-well invasion assay. Quantitative real-time PCR was used to detect gene expression in HSC-T6 cells. RESULTS: All concentrations of drugs showed no cytotoxicity against HSC-T6 cells. LPS stimulated NF-κB luciferase activities, nuclear translocation of NF-κB-p65, and phosphorylations of ERK, JNK and p38, all of which were suppressed by Tan IIA. In addition, Tan IIA significantly inhibited LPS-induced HSCs chemotaxis, in both wound-healing and trans-well invasion assays. Moreover, Tan IIA attenuated LPS-induced mRNA expressions of CCL2, CCL3, CCL5, IL-1ß, TNF-α, IL-6, ICAM-1, iNOS, and α-SMA in HSC-T6 cells. CONCLUSION: Our results demonstrated that Tan IIA decreased LPS-induced HSC activation.


Asunto(s)
Abietanos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Actinas/metabolismo , Animales , Línea Celular Transformada , Quimiotaxis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Ratas
6.
Int J Med Sci ; 11(5): 528-37, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24693223

RESUMEN

This study investigated the antifatigue effects of rutin, a flavonoid extracted from the ethyl acetate extract of S. involucrata. Mice were subjected to a weight-loaded forced swim test (WFST) on alternate days for 3 wk. Rutin was administered orally to the mice for 7 days in dosages of 15, 30, and 60 mg/kg body weight, and several biomarkers of physical fatigue were evaluated: swimming time, change in body weight, lipid peroxidation, lactic acid (LA), glycogen, and the activities of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx). On Day 7, the rutin-treated mice had a 3-fold longer exhaustive swimming time than the control mice, as well as significantly reduced blood LA concentrations. The 15, 30, and 60 mg/kg body weight rutin-supplemented groups displayed 11.2%, 22.5%, and 37.7% reduced malondialdehyde (MDA) concentrations, respectively, in brain and muscle tissues compared with the control exercised group. Our results indicated that the administration of rutin protected the mice against the depletion of SOD and GPx activities significantly. Following 7 days of rutin treatment, we sacrificed the mice and analyzed their soleus muscle and brain for peroxisome proliferator-activated receptor-α coactivator (PGC-1α) and sirtuin 1 (SIRT1) mRNA expression. We observed that rutin treatment increased PGC-1α and SIRT1 mRNA and protein expression. The changes in these markers of mitochondrial biogenesis were associated with increased maximal endurance capacity. The application of 2D gel electrophoresis to analyze the rutin-responsive protein profiles in the WFST mouse brain further revealed the upregulation of the CB1 cannabinoid receptor-interacting protein 1, myelin basic protein, Rho GDP dissociation inhibitor (GDI) alpha, and TPI, indicating that rutin might inhibit anxiety through the upregulation of the expression of anxiety-associated proteins. Western blot analysis of MAPK expression further confirmed the antianxiety effects of rutin. Our study results thus indicate that rutin treatment ameliorates the various impairments associated with physical fatigue.


Asunto(s)
Antioxidantes/metabolismo , Fatiga/tratamiento farmacológico , Rutina/administración & dosificación , Saussurea/química , Animales , Peso Corporal/efectos de los fármacos , Fatiga/patología , Glutatión Peroxidasa/biosíntesis , Humanos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Condicionamiento Físico Animal , Rutina/química , Superóxido Dismutasa/biosíntesis , Natación
7.
J Gastroenterol Hepatol ; 29(5): 1073-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24325631

RESUMEN

BACKGROUND AND AIM: Vascular endothelial (VEGF) and fibroblast growth factor (FGF)-induced hepatic stellate (HSCs) and liver endothelial cells (LECs) activation accelerates hepatic fibrogenesis and angiogenesis, and hemodynamic dysarrangements in cirrhosis. VEGF targeting agents had been reported as potential drugs for cirrhosis. However, the evaluation of effects of dual VEGF/FGF targeting agent in cirrhosis is still limited. METHODS: Using hemodynamic parameters, blood chemistry, primary isolated HSCs and LECs, histology, and digital imaging, we assess the effects of 2-week brivanib alaninate, a dual VEGFR/FGFR inhibitor, treatment in the pathophysiology of bile duct-ligated-cirrhotic rats. RESULTS: Fibrogenic and angiogenic markers in the serum and liver of bile duct-ligated-cirrhotic rats, including hydroxyproline, transforming growth factor-ß1, angiopoietin-1, VEGF, FGF-2, endocan and phosphorylated-VEGFR2/VEGFR2, and phosphorylated-FGFR/FGFR together with hepatic CD31/angiopoietin-1 expressions (immunohistochemistry staining), angiogenesis (micro-computed tomography scan), microcirculatory dysfunction (in vivo miscroscopy and in situ liver perfusion study), portal hypertension, and hyperdynamic circulations (colored microsphere methods) were markedly suppressed and ameliorated by brivanib alaninate treatment. In in vitro study, acute brivanib alaninate incubation inhibited the transforming growth factor-ß1-induced HSCs contraction/migration and VEGF-induced LECs angiogenesis. Concomitantly, the overexpression of various fibrogenic and angiogenic markers in HSCs and LECs, and in their culture media, was increased in parallel and these changes were suppressed by acute brivanib alaninate incubation. CONCLUSIONS: This study demonstrated that brivanib alaninate targeting multiple mechanisms and working in the different pathogenic steps of the complications of cirrhotic rats with portal hypertension.


Asunto(s)
Alanina/análogos & derivados , Hipertensión Portal/tratamiento farmacológico , Hipertensión Portal/genética , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Triazinas/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/patología , Masculino , Terapia Molecular Dirigida , Ratas , Ratas Sprague-Dawley , Triazinas/farmacología
8.
Clin Sci (Lond) ; 126(2): 163-74, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23875703

RESUMEN

Besides cardioprotective effects, the AT1R (angiotensin-II type 1 receptor) antagonist losartan protects the liver from IRI [IR (ischaemia/reperfusion) injury], but the mechanism has not been fully determined. The HIF (hypoxia inducible factor)-1α and Wnt/ß-catenin signalling pathways have been reported to be involved in the mechanism of liver IRI. Therefore the aim of the present study was to determine whether the Wnt/HIF axis is part of the mechanism of the positive effect of AngII inhibition by losartan in liver IRI in rats. Various measurements were made in MCD/HF-NASH (methionine- and choline-deficient-diet/high-fat-diet-induced non-alcoholic steatohepatitis) rats with liver IRI. Acute losartan pre-administration markedly reversed the IR-suppressed levels of the hepatic-protective factors IL (interleukin)-6, IFN (interferon)-γ, Wnt3a, ß-catenin and HIF-1α, and decreased hepatic blood flow and IR-elevated serum ALT (alanine aminotransferase), hepatic TNF (tumour necrosis factor)-α, IL-1α, hepatic congestion, vacuolization and necrosis, hepatic Suzuki IRI scores, necrotic index and levels of TBARS (thiobarbituric acid-reacting substances) in MCD/HF-NASH rats. Furthermore, acute Wnt3a pre-treatment significantly inhibited IR-elevated serum ALT, hepatic Suzuki IRI scores and TBARS, and restored the IR-depleted ß-catenin/HIF-1α activity in MCD/HF-NASH rats. Simultaneous acute sFRP2 (secreted frizzled-related protein 2; a Wnt3a inhibitor) pre-treatment eliminated the losartan-related beneficial effects in MCD/HF-NASH rats with liver IRI, which was accompanied by a decrease in hepatic HIF-1α/ß-catenin activity. Losartan-induced up-regulation of HIF-1α and Wnt/ß-catenin signalling was associated with the recovery of IR-inhibited hepatic Bcl-2, Mn-SOD (manganese superoxide), Cu/Zn-SOD (copper/zinc superoxide) and GSH levels, and the suppression of IR-increased hepatic catalase and caspase 3/caspase 8 levels in MCD/HF-NASH rats. In conclusion, up-regulation of the HIF-1α and Wnt/ß-catenin signalling pathways are part of the mechanism of the positive effects of losartan-related AngII inhibition in MCD/HF-NASH rats with liver IRI. Our study highlights the potential of the dual-organ protective agent losartan in NASH patients with steatotic livers and cardiovascular risk.


Asunto(s)
Hígado Graso/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Losartán/farmacología , Daño por Reperfusión/prevención & control , Transducción de Señal/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología , Animales , Dieta Alta en Grasa , Glicoproteínas/farmacología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Enfermedad del Hígado Graso no Alcohólico , Ratas , Ratas Sprague-Dawley , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Regulación hacia Arriba , Proteína Wnt3A/farmacología
9.
Liver Int ; 34(4): 521-34, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23998651

RESUMEN

BACKGROUND & AIMS: Antivascular endothelial growth factor receptor (VEGFR) agents improve hepatic fibrosis and portal hypertension in cirrhosis. Detail interactions among recruited/activated leucocytes, hepatic angiogenesis and fibrogenesis, splanchnic blood pooling, decreased hepatic veins to fluctuated splanchnic blood volume (hepatic venous dysregulation), portal hypertensive syndrome and ascites have never explored in cirrhosis. Our study used two anti-VEGFR agents - brivanib and sorafenib - to elucidate the relationship between above abnormalities of nonalcoholic steatohepatitis (NASH)-cirrhotic rats. MATERIALS AND METHODS: NASH-cirrhotic rats received 2-week brivanib, sorafenib or vehicle (NASH-cirrhotic+briv, NASH-cirrhotic+soraf and NASH-cirrhotic rats) were included for various measurements. RESULTS: In comparison with NASH-cirrhotic rats, significant decreased plasma VEGF, fibroblast growth factor, platelet-derived growth factor, hepatic tumour necrosis factor (TNFα), IL-1ß, IL-6, IL-17 were accompanied by decreased leucocyte mass/activity ((99 m) Tc-phytate and (18) F-FDG SPECT/PET/CT scans), hepatic leucocytes recruitment/microvascular density (fluorescence-enhanced intravital microscopy) and hydroxyproline content, and increased hepatic blood flow in NASH-cirrhotic+briv and NASH-cirrhotic+soraf rats. In addition, increased hepatic microvasculatures compliance-related improved buffering effect of portal vein to acute mannitol infusion was associated with decreased circulating nitric oxide and aldosterone, plasma volume expansion (dye dilution method), splanchnic blood pooling ((99 m) Tc-RBC SPECT/PET/CT scans), peripheral hypotension, portal hypertension and ascites in brivanib and sorafenib-treated NASH-cirrhotic rats. CONCLUSION: Besides antifibrotic, antiangiogenic and portal hypertensive effects, chronic antagonism of anti-VEGFR with brivanib and sorafenib improves hepatic blood flow, hepatic venous dysregulation, inhibits leucocytes recruitment/activation, splanchnic blood pooling and ascites formation in NASH-cirrhotic rats. Thus, brivanib and sorafenib might be ideal therapeutic agents in cirrhotic patients suffering from severe haemodynamic disarrangement and ascites.


Asunto(s)
Alanina/análogos & derivados , Ascitis/tratamiento farmacológico , Hígado Graso/tratamiento farmacológico , Microcirculación/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Circulación Esplácnica/efectos de los fármacos , Triazinas/farmacología , Alanina/farmacología , Animales , Citocinas/metabolismo , Cartilla de ADN/genética , Hígado Graso/patología , Factores de Crecimiento de Fibroblastos/sangre , Microcirculación/fisiología , Niacinamida/farmacología , Enfermedad del Hígado Graso no Alcohólico , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Tomografía de Emisión de Positrones , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sorafenib , Factor A de Crecimiento Endotelial Vascular/sangre
10.
J Ethnopharmacol ; 150(3): 835-42, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24036165

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Liver fibrosis is the result of long-term liver damage and the wound-healing process, in which the hepatic stellate cell (HSC) plays a crucial role during fibrogenesis. The liver sinusoidal endothelial cell (LSEC) is a liver-resident scavenger, contributing to sinusoidal remodeling, HSC activation and liver fibrosis. Lipopolysaccharide (LPS) causes an inflammatory reaction associated with portal circulation and LSECs signaling. Scutellariae radix, the root of Scutellaria baicalensis Georgi, is a Chinese herb widely used for liver diseases. However, its effect on LSEC activation and HSC migration in liver fibrosis has not been investigated yet. AIM OF THIS STUDY: LPS-induced rat LSEC (rLSEC) activation was used as a model to screen and explore the active components of Scutellariae radix. The anti-fibrotic effect of Scutellariae radix on rLSEC activation and rHSC migration was further investigated. MATERIALS AND METHODS: LPS-induced rLSEC mRNA expression, including VEGF, VEGFR, MCP-1, and TGF-ß1, were examined by real-time PCR analyses. MCP-1 protein levels were measured by an ELISA kit. rLSEC conditioned medium on rHSC migration was measured by wound-healing assay and transwell chemoattraction assay. RESULTS: Results showed LPS-induced rLSEC activation with upregulated MCP-1 mRNA and protein expressions, and that rLSEC-condition medium enhanced rHSC migration. Both baicalein and wogonin from the active subfraction significantly reduced MCP-1 expression, but only baicalein markedly inhibited rHSC migration in rLSEC conditioned medium. CONCLUSION: This study demonstrated that Scutellariae radix attenuates LPS-induced rLSEC activation and HSC migration with downregulation of MCP-1 expression. The results provide supporting evidence that Scutellariae radix may be beneficial for the amelioration of liver fibrosis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Flavonoides/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Extractos Vegetales/farmacología , Scutellaria baicalensis , Animales , Movimiento Celular/fisiología , Quimiocina CCL2/genética , Células Endoteliales/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/fisiología , Lipopolisacáridos , Hígado/citología , Raíces de Plantas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Factor de Crecimiento Transformador beta1/genética , Factor A de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas
11.
J Cell Biochem ; 114(3): 541-50, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22961938

RESUMEN

Hepatitis C virus core protein (HCVcp), which is secreted by infected cells, is reported as an immunomodulator in immune cells. However, the effects of HCVcp on hepatic stellate cells (HSCs), the key cells in liver fibrosis, still remain unclear. In this study, we investigated the effects of HCVcp on obese receptor (ObR) related downstream signaling pathways and fibrogenic gene expression in HSCs. LX-2, a human HSC line, was incubated with HCVcp. Inhibitors and short interfering RNAs were used to interrogate the mechanisms of HCVcp action on HSCs. HCVcp (20-100 ng/ml) concentration-dependently stimulated α-smooth muscle actin (α-SMA) protein expression and mRNA expression of α-SMA, procollagen α2(I) and TGF-ß1 genes, with a plateau of 220% of controls at 100 ng/ml. HCVcp induced mRNA and protein expression of ObR. Blocking of Ob-Rb with a neutralizing antibody inhibited phosphorylation of signal transducer and activator of transcription 3 (STAT3) and AMPKα stimulated by HCVcp. Furthermore, knockdown of Ob-Rb down-regulated HCVcp-induced STAT3, AKT, and AMPKα phosphorylation, and reversed HCVcp-suppressed mRNA expression of matrix metalloproteinase (MMP)-1, peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element binding protein-1c (SREBP-1c) genes. AMPKα signaling blockade reversed HCVcp-suppressed SREBP-1c mRNA expression. HCVcp stimulated reactive oxygen species formation and gp91(phox) (a component of NADPH oxidase) protein expression, together with AKT phosphorylation, leading to suppression of PPARγ and SREBP-1c genes. Our results provide a new finding that HCVcp induced ObR-dependent Janus Kinase (JAK) 2-STAT3, AMPKα, and AKT signaling pathways and modulated downstream fibrogenetic gene expression in HSCs.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Antígenos de la Hepatitis C/metabolismo , Cirrosis Hepática , Receptores de Leptina/metabolismo , Proteínas del Núcleo Viral/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Actinas/biosíntesis , Actinas/genética , Anticuerpos Neutralizantes/inmunología , Línea Celular , Colágeno Tipo I/genética , Hepacivirus/genética , Hepacivirus/metabolismo , Células Estrelladas Hepáticas/virología , Antígenos de la Hepatitis C/genética , Humanos , Janus Quinasa 2/metabolismo , Metaloproteinasa 1 de la Matriz/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/inmunología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Factor de Crecimiento Transformador beta1/genética , Proteínas del Núcleo Viral/genética
12.
J Chin Med Assoc ; 75(10): 501-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23089401

RESUMEN

BACKGROUND: It is a well-accepted fact that angiotensin II (Ang II) contributes to increased vascular tone in cirrhotic livers. However, aliskiren attenuates the effect of Ang II through direct renin inhibition. Our study aimed to evaluate the effects of aliskiren on portal pressure and intrahepatic resistance in bile duct ligated (BDL) rats. METHODS: The effects of acute intravenous infusion (1 mg or 3 mg) or a course of 2-day oral administration of aliskiren (20 mg/kg/day) on blood pressure and portal pressure were evaluated in BDL and sham rats. Intrahepatic resistance was evaluated by a liver perfusion study isolated in situ. Ang II efflux was measured by Enzyme-linked immunosorbent assay (ELISA). The hepatic gene expression of angiotensinogen, renin, angiotensin-converting enzyme (ACE), Ang II type 1 receptor (AT(1)R) was analyzed with quantitative reverse transcription polymerase chain reaction. RESULTS: Aliskiren infusion intravenously reduced portal pressure with a minimal effect on blood pressure in BDL rats. Direct infusion of aliskiren in an isolated cirrhotic liver caused greater vasorelaxation and decreased hepatic production of Ang II. Two days of aliskiren treatment reduced portal pressure and hepatic ACE mRNA; in addition, it improved the vasodilator response to acetylcholine in the cirrhotic livers and decreased Ang II efflux. CONCLUSION: Aliskiren reduced portal pressure in cirrhotic rats. The portal hypotensive effect of aliskiren was related to the amelioration of the Ang II induced intrahepatic vasoconstriction.


Asunto(s)
Amidas/farmacología , Fumaratos/farmacología , Circulación Hepática/efectos de los fármacos , Presión Portal/efectos de los fármacos , Resistencia Vascular/efectos de los fármacos , Administración Oral , Amidas/administración & dosificación , Animales , Fumaratos/administración & dosificación , Infusiones Intravenosas , Cirrosis Hepática Experimental/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley
13.
Liver Transpl ; 18(11): 1371-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22847887

RESUMEN

During liver transplantation, nonalcoholic steatohepatitis (NASH) aggravates ischemia/reperfusion (IR) injury by activating various kinases and subsequently releasing cytokines and chemokines. Nonetheless, the effect of the multikinase inhibitor sorafenib on IR liver injury in rats with NASH has never been explored. Our study was designed to determine this effect and associated mechanisms in NASH rats. Sorafenib was acutely administered to NASH rats with IR liver injury that were or were not chronically pretreated with the Rho-kinase-specific inhibitor fasudil. Then, the following were evaluated: mean arterial pressure; hepatic blood flow and microcirculatory dysfunction; hepatic inflammation (serum alanine aminotransferase); necrosis; apoptosis; leukocyte infiltration; CD45 staining; caspase levels and DNA fragmentation; various serum and hepatic cytokines; and proteins and genes of the Raf/mitogen-activated protein-extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase, and apoptosis pathways. In NASH rats with IR liver injury, hepatic inflammation, necrosis, apoptosis, leukocyte infiltration, and microcirculatory dysfunction were significantly attenuated by the acute administration of sorafenib through the inhibition of the hepatic release of macrophage inflammatory protein 2, keratinocyte chemoattractant, granulocyte-monocyte colony-stimulating factor, and hepatic caspase-3 and caspase-9 as well as DNA fragmentation. Furthermore, there was decreased expression of p-Raf1 (where p indicates the phosphorylated form), p-MEK1/2, p-ERK1/2, p-Rho-kinase, B cell lymphoma 2-associated death promoter, and B cell lymphoma 2-associated X protein at the protein and messenger RNA levels. Notably, the aforementioned beneficial effects of sorafenib were significantly abolished by chronic pretreatment with the Rho-kinase-specific inhibitor fasudil. This study demonstrated that the multikinase inhibitor sorafenib protects NASH rats from IR injury by interfering with the inflammation, necrotic, and apoptotic responses causing leukocyte-dependent hepatic microcirculatory dysfunction. The hepatoprotective effects of sorafenib seem to work through the inhibition of the Rho-kinase-dependent Raf/MEK/ERK pathway, which is up-regulated during IR injury in the livers of NASH rats.


Asunto(s)
Hígado Graso/terapia , Hígado/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Daño por Reperfusión/tratamiento farmacológico , Quinasas Asociadas a rho/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Hemodinámica , Inflamación , Hígado/enzimología , Hígado/lesiones , Sistema de Señalización de MAP Quinasas , Masculino , Necrosis , Niacinamida/farmacología , Enfermedad del Hígado Graso no Alcohólico , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sorafenib , Trasplante Homólogo
14.
Clin Sci (Lond) ; 123(12): 669-80, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22734967

RESUMEN

Cirrhotic portal hypertension is characterized by increased hepatic oxidative stress, AA (arachidonic acid)-derived TXA(2) (thromboxane A(2)) release and exaggerated hepatic response to the α-adrenergic agonist MTX (methoxamine). Besides promoting hepatic fibrosis, the role of hyperleptinaemia in the modulation of vascular response in NASH (non-alcoholic steatohepatitis) rat livers remains unknown. The aim of the present study was to explore the possible links between hyperleptinaemia and the disarrangement in the hepatic microcirculation. NASH-cirrhosis with hyperleptinaemia was induced in lean rats by feeding with an HF/MCD (high-fat/methionine-choline-deficient) diet. Portal haemodynamics, various substances, protein and mRNA expression and PUFA (polyunsaturated fatty acid) composition were measured. Finally, the effects of leptin pre-infusion on TXA(2) release and concentration-PPP (portal perfusion pressure) curves in response to MTX were evaluated by simultaneously pre-treatment with the Kupffer cell inactivators GdCl(3) (gadolinium chloride) or EC (encapsulated clodronate), the TXS (TXA(2) synthase) inhibitor furegrelate, the TP receptor (TXA(2) receptor) antagonist SQ29548 and the dual TXS/TP receptor antagonist BM567. In HF/MCD+leptin-lean rats, cirrhosis-induced PPP and MTX hyper-responsiveness were associated with increased hepatic TXA(2) production, TBARS (thiobarbituric acid-reacting substances) levels and the AA (arachidonic acid)/n-3 PUFA ratio, and up-regulation of hepatic leptin, FAS (fatty acid synthase), NADPH oxidase subunits, TXS, TP receptor, TGFß(1) (transforming growth factor ß(1)) proteins and mRNAs. Pre-infusion of leptin significantly enhanced MTX-stimulated PPP elevation and TXA(2) release, which were attenuated by GdCl(3) and EC pre-treatment. Concomitantly pre-incubation with BM567, but not furegrelate or SQ29548, significantly abolished the leptin-enhanced MTX-stimulated increase in PPP in NASH-cirrhotic rats. Hyperleptinaemia plays an important role in hyper-responsiveness to MTX in NASH-cirrhotic rat livers with portal hypertension. The leptin-enhanced MTX-stimulated increase in PPP is mediated by increased oxidative stress and Kupffer-cell-activated AA-derived TXA(2) release in NASH-cirrhotic rats.


Asunto(s)
Hígado Graso/fisiopatología , Hipertensión Portal/fisiopatología , Macrófagos del Hígado/fisiología , Leptina/metabolismo , Metoxamina/metabolismo , Estrés Oxidativo/fisiología , Tromboxano A2/metabolismo , Análisis de Varianza , Animales , Ácido Araquidónico/metabolismo , Benzofuranos/farmacología , Colina/metabolismo , Ácido Clodrónico/farmacología , Cartilla de ADN/genética , Dieta Alta en Grasa , Ácidos Grasos Insaturados/metabolismo , Hígado Graso/complicaciones , Hígado Graso/metabolismo , Gadolinio/farmacología , Hemodinámica , Hipertensión Portal/etiología , Resistencia a la Insulina/fisiología , Macrófagos del Hígado/efectos de los fármacos , Leptina/sangre , Metionina/deficiencia , Microcirculación/fisiología , Enfermedad del Hígado Graso no Alcohólico , ARN Mensajero/metabolismo , Ratas , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Compuestos de Sulfonilurea/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
15.
Planta Med ; 78(4): 341-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22399273

RESUMEN

Hepatic stellate cells (HSCs) play a key role in the pathogenesis of liver fibrosis. In chronic liver injury, HSCs undergo transdifferentiation to an activated myofibroblastic phenotype and migrate to injured areas in response to chemotactic factors, producing extracellular matrix proteins such as collagen type I to repair the damage as well as overexpression of α-smooth muscle actin (α-SMA). Paeoniae Radix, the root of Paeonia lactiflora Pall, was investigated for PDGF-BB-induced HSC chemotaxis. Rat HSCs and LX-2, a human HSC cell line, were used for the in vitro experiments. Cell migration was analyzed by wound-healing and transwell assays. An ELISA and a Sircol collagen assay kit were used to detect the expressions of α-SMA and of collagen, respectively. Phosphorylations of mitogen-activated protein kinases, including ERK 1/2, p38, and JNK, were evaluated with immunoblotting. Results indicated that PDGF-BB increased migration as well as α-SMA and collagen expression in HSCs. Paeoniae Radix extracts and its active components, paeonol and 1,2,3,4,6-penta- O-galloyl- ß-D-glucose (PGG), inhibited PDGF-BB-induced HSC migration and α-SMA and collagen expressions in a concentration-dependent manner. The inhibitory effects were associated with downregulation of PDGF receptor- α, ERK, p38, and JNK activation. Both paeonol and PGG participate in HSC migration, but via differential mechanisms.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Paeonia/química , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-sis/antagonistas & inhibidores , Acetofenonas/farmacología , Actinas/antagonistas & inhibidores , Actinas/biosíntesis , Animales , Becaplermina , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Colágeno/antagonistas & inhibidores , Colágeno/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Humanos , Taninos Hidrolizables/farmacología , Masculino , Raíces de Plantas/química , Ratas , Ratas Sprague-Dawley
16.
J Ethnopharmacol ; 140(3): 568-86, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22373513

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: GP-TCM is the first EU-funded Coordination Action consortium dedicated to traditional Chinese medicine (TCM) research. One of the key deliverables of the Work Package 7 in GP-TCM was to investigate information of the existing requirements for registration of TCM products listed by global regulatory bodies. The paper aims to collate data and draw comparison of these regulations. Case studies are also presented to illustrate the problems involved in registering TCM products in different regions worldwide. MATERIALS AND METHODS: A collaborative network task force was established during the early stage of the GP-TCM project and operated through exchanges, teleconferences and focused discussions at annual meetings. The task force involved coordinators, academics who are actively involved with R&D of Chinese herbal medicines, experts on monographic standards of Chinese materia medica, representatives from regulatory agencies, experts from industries in marketing Chinese medicines/herbal medicines and natural products. The co-ordinators took turns to chair teleconferences, led discussions on specific issues at AGM discussion sessions, at joint workshops with other work-packages such as WP1 (quality issues), WP3 (toxicology issues) and WP6 (clinical trial issues). Collectively the authors were responsible for collating discussion outcomes and updating written information. RESULTS: A global overview of regulations on herbal registration has been compiled during the three years of the consortium. The regulatory requirements for registration of herbal products in the EU and China were compared, and this is extended to other regions/countries: Africa, Australia, Brazil, Canada, Japan, Russia, South Korea, Taiwan, and the United States. A wide variation of the regulations for the categories of herbal products exists: food (functional food, novel foods, dietary food for special medical purpose, foods for particular nutritional use, food supplement); cosmetic, traditional herbal medicine products; herbal medicines for human use and veterinary use. CONCLUSION: The regulatory issues for registration of herbal products are complicated among the countries and regions worldwide. The information summarised in the text is for reference only. Some regulations which are presented in this review are still in legislation process and may change in due course. Before taking any regulatory action, readers are advised to consult current official legislation and guidance and/or to seek appropriate professional advice. The lessons learnt from global regulation of TCM will provide valuable insights for regulation of other traditional medicine such as Ayurveda and Unani medicine, as well as other forms of indigenous medicine. The WHO is well placed to co-ordinate a consultation process with the aim of putting forward suggestions for harmonisation to key regulatory agencies.


Asunto(s)
Seguridad de Productos para el Consumidor/legislación & jurisprudencia , Medicamentos Herbarios Chinos , Regulación Gubernamental , Legislación de Medicamentos , Legislación Alimentaria , Medicina Tradicional China , Fitoterapia , África , Asia , Australia , Brasil , China , Unión Europea , Predicción , Humanos , Internacionalidad , Plantas Medicinales , Estados Unidos
17.
Phytother Res ; 26(3): 344-53, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21717514

RESUMEN

The aim of this study was to investigate if armepavine (Arm, C19H23O3N) could exert inhibitory effects against hepatic fibrosis in rats. A cell line of rat hepatic stellate cells (HSC-T6) was stimulated with tumour necrosis factor-α (TNF-α) to evaluate the inhibitory effects of Arm. Rats were injected with thioacetamide (TAA; 300 mg/kg, intraperitoneally) thrice a week for 4 weeks to induce hepatic fibrosis, with Arm (3 or 10 mg/kg) given by gavage twice a day. Liver sections were taken for western blotting, fibrosis scoring and immunofluorescence staining. Arm (1-10 µm) concentration-dependently attenuated TNF-α-stimulated: (i) protein expressions of α-smooth muscle actin (α-SMA), collagen type I and angiopoietin-1; (ii) H2O2 production; and (iii) NF-κB, JunD and C/EBPß (cytidine-cytidine-adenosine-adenosine-thymidine (CCAAT)/enhancer binding protein-ß (EBPß)) nuclear translocations in HSC-T6 cells. In vivo Arm treatment significantly reduced plasma aspartate transaminase and alanine transaminase levels, hepatic α-SMA expression and collagen contents, and fibrosis scores of TAA-injected rats. Moreover, Arm treatment decreased α-SMA- and NF-κB-positive cells in immunohistochemical staining, and mRNA expression levels of IL-6, TGF-ß1, TIMP-1, col1α2, iNOS and ICAM-1 genes, but up-regulated the metallothionein gene in the livers of TAA-injected rats. Our results indicated that Arm exerted both in vitro and in vivo antifibrotic effects in rats, with inhibition of NF-κB, JunD and C/EBPß pathways.


Asunto(s)
Bencilisoquinolinas/uso terapéutico , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Fitoterapia , Tioacetamida/efectos adversos , Actinas/genética , Actinas/metabolismo , Transporte Activo de Núcleo Celular , Alanina Transaminasa/sangre , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Aspartato Aminotransferasas/sangre , Bencilisoquinolinas/administración & dosificación , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Colágeno Tipo I/metabolismo , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Técnica del Anticuerpo Fluorescente , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Peróxido de Hidrógeno/metabolismo , Cirrosis Hepática/patología , Masculino , Metalotioneína/genética , Metalotioneína/metabolismo , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Tioacetamida/administración & dosificación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
18.
Liver Int ; 32(1): 48-57, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22098317

RESUMEN

BACKGROUND: Hepatic endothelial dysfunction (HED), which is caused by decreased hepatic nitric oxide (NO) bioavailability and increased lipid peroxidation, contributes to portal hypertension, which is a characteristic of cirrhosis. Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase (NOS), is involved in cirrhosis-related HED and portal hypertension. AIMS: We evaluated the effect of vitamin E treatment on the lipid peroxidation, HED and portal hypertension in cirrhotic rats. METHODS: The common bile duct ligation (BDL)-induced cirrhotic rats were treated orally either with vehicle or with vitamin E for 1 month immediately after BDL. Systemic and portal haemodynamics, the magnitude of the increase in portal pressure induced by volume expansion, HED, oxidative stress, levels of ADMA, various proteins and mRNAs were then measured. RESULTS: In the vitamin E-treated BDL rats, a decrease in portal pressure was associated with an attenuation of the increased portal pressure induced by volume expansion. In isolated and perfused BDL rat livers, the vitamin E treatment significantly inhibited the (paradoxical) vasoconstriction response to methoxamine and acetylcholine (HED), and this was abolished by the presence of NOS. Vitamin E decreased ADMA synthesizing enzyme PRMT1 expression and the level of thiobarbituric acid-reactive substances (TBARS) in the liver, while increasing the levels of hepatic ADMA metabolizing enzyme DDAH2, eNOS, phosphor-eNOS, ADMA level and superoxide dismutase activity. CONCLUSIONS: The administration of vitamin E suppressed hepatic ADMA and oxidative stress in the cirrhotic liver circulation, and therefore increases NO bioavailability, which improved HED and portal hypertension.


Asunto(s)
Antioxidantes/farmacología , Arginina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hipertensión Portal/tratamiento farmacológico , Cirrosis Hepática Experimental/tratamiento farmacológico , Vitamina E/farmacología , Animales , Arginina/farmacología , Sinergismo Farmacológico , Células Endoteliales/patología , Hemodinámica/efectos de los fármacos , Hipertensión Portal/metabolismo , Hipertensión Portal/patología , Técnicas In Vitro , Peroxidación de Lípido/efectos de los fármacos , Hígado/irrigación sanguínea , Hígado/efectos de los fármacos , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Masculino , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Perfusión , Ratas , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos
19.
Hepatology ; 55(5): 1540-50, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22183953

RESUMEN

UNLABELLED: Leptin, the ob gene product, is a protein released from adipocytes and has been detected in fibrotic and cirrhotic livers. Leptin in brain has an inhibitory effect on food intake. Nonalcoholic steatohepatitis (NASH) is characterized by hyperleptinemia. This study explores the possible mechanisms of hyperleptinemia in relation to increased intrahepatic resistance (IHR) and portal hypertension in NASH cirrhotic rats. NASH cirrhotic rats with hyperleptinemia were induced in Zucker (fa/fa) and lean rats by feeding the animals a high fat/methionine-choline-deficient (HF/MCD) diet with and without exogenous administration of recombinant leptin. Portal venous pressure (PVP), IHR, plasma and hepatic levels of various substances, histopathology of the liver, the hepatic hydroxyproline content, and the expression of various hepatic protein and messenger RNA (mRNA) were measured. Hepatic microcirculatory dysfunction and the vasoconstrictive response to endothelin-1 were also observed using a liver perfusion system and intravital microscopy. Finally, the effect of leptin on hepatic stellate cells (HSCs) was evaluated. Both in HF/MCD-Zucker and HF/MCD+leptin lean rats, significant hepatic fibrogenesis and cirrhosis, marked portal hypertension, microcirculatory dysfunction, an enhanced vasoconstrictive response to endothelin-1, and an increased IHR were found to be associated with higher levels of hepatic endothelin-1 and endocannabinoids, expression levels of the cannabinoid type 1 receptor, endothelin-1 type A receptor (ET(A) R), activator protein-1, transforming growth factor beta (TGF-ß)(1), osteopontin, tumor necrosis factor alpha (TNF-α), leptin, and the leptin receptor (OBRb). Interestingly, acute incubation of leptin directly increases the expression of ET(A) R, OBRb and activator protein-1 in HSCs. CONCLUSION: An HF/MCD diet and hyperleptinemia increase hepatic endocannabinoids production, promote hepatic fibrogenesis, enhance the hepatic vasoconstrictive response to endothelin-1, and aggravate hepatic microcirculatory dysfunction; these events subsequently increase IHR and portal hypertension in NASH cirrhotic rats.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Endotelina-1/metabolismo , Hígado Graso/fisiopatología , Hipertensión Portal/fisiopatología , Leptina/metabolismo , Hígado/metabolismo , Animales , Biopsia con Aguja , Peso Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/complicaciones , Células Estrelladas Hepáticas/metabolismo , Hipertensión Portal/complicaciones , Inmunohistoquímica , Resistencia a la Insulina/fisiología , Macrófagos del Hígado/metabolismo , Microcirculación , Enfermedad del Hígado Graso no Alcohólico , ARN Mensajero/análisis , Distribución Aleatoria , Ratas , Ratas Zucker , Estadísticas no Paramétricas
20.
Eur J Clin Invest ; 42(6): 607-16, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22103576

RESUMEN

BACKGROUND: We previously demonstrated that kaerophyllin, a lignan, isolated from a widely used traditional Chinese herb, Bupleurum scorzonerifolium, leading to the inhibition of hepatic stellate cells (HSCs) activation in vitro. This current study evaluated the in vivo role of kaerophyllin in protecting the liver against injury and fibrogenesis caused by thioacetamide (TAA) in rats and further explored the underlying mechanisms. MATERIALS AND METHODS: Liver fibrosis in Sprague-Dawley rats was induced by intraperitoneal injection of TAA (200 mg/kg) twice per week for 6 weeks. Animals were divided into five groups: vehicle control, TAA control, TAA + low dose kaerophyllin, TAA + high dose kaerophyllin and TAA + curcumin groups. Kaerophyllin (10 or 30 mg/kg) or curcumin (150 mg/mL) was given by gavage twice per day consecutively for 4 weeks starting 2 weeks after TAA injection. Rat HSCs were used to investigate the anti-inflammatory role of kaerophyllin against tumour necrosis factor α (TNF-α) in vitro. Peroxisome proliferator-activated receptor-γ (PPAR-γ) expression was knocked down in rat HSCs using PPAR-γ small interfering RNAs. RESULTS: Kaerophyllin significantly protected liver from injury by reducing serum aspartate transaminase and alanine transaminase levels and by improving the histological architecture and fibrosis score. In addition, kaerophyllin suppressed inflammation by reducing the mRNA of TNF-α, interleukin-1ß (IL-1ß) and monocyte chemoattractant protein-1 (MCP-1) genes. In HSCs, kaerophyllin elevated PPAR-γ activity and reduced TNF-α-stimulated mRNA levels of intracellular adhesion molecule-1 (ICAM-1), MCP-1 and IL-1ß genes, which were reversed by small interfering RNA knockdown of PPAR-γ gene. CONCLUSIONS: Our results demonstrated that kaerophyllin protected the rat liver from TAA-caused injury and fibrogenesis by suppressing hepatic inflammation and inhibiting HSC activation, possibly through upregulation of PPAR-γ expression.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Células Estrelladas Hepáticas/efectos de los fármacos , Lignanos/uso terapéutico , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Tioacetamida/efectos adversos , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Carcinógenos/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Masculino , Extractos Vegetales/metabolismo , Extractos Vegetales/uso terapéutico , Raíces de Plantas , Ratas , Ratas Sprague-Dawley , Tioacetamida/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...