Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Med (Berl) ; 101(1-2): 139-149, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36527474

RESUMO

This work aimed to investigate the role of nuclear factor peroxisome proliferator-activated receptor α (PPARα) in modification of circadian clock and their relevance to development of nonalcoholic fatty liver disease (NAFLD). Both male wild-type (WT) and Pparα-null (KO) mice treated with high-fat diet (HFD) were used to explore the effect of PPARα and lipid diet on the circadian rhythm. WT, KO, and PPARα-humanized (hPPARα) mice were treated with PPARα agonist fenofibrate to reveal the hPPARα dependence of circadian locomotor output cycles kaput (CLOCK) down-regulation. The mouse model and hepatocyte experiments were designed to verify the action of PPARα in down-regulating CLOCK and lipid accumulation in vivo and in vitro. Strongest NAFLD developed in mice fed 45%HFD, and it was inhibited in WT mice. The activity rhythm of WT mice was found to be different from that of the KO mice on normal diet and HFD. The core circadian factor CLOCK was down-regulated by HFD in both WT and KO mice in the liver, not in the hypothalamus. More interestingly, hepatic CLOCK was down-regulated by basal PPARα and activated PPARα in dose dependence of fenofibrate. Accordingly, CLOCK down-regulation dependent of PPARα activity was involved in inhibition of lipid metabolism in hepatocytes. Down-regulation of hepatic CLOCK by basal PPARα contributes to tolerance against development of NAFLD. Inhibition of CLOCK by activated PPARα is involved in inhibition of NAFLD by PPARα agonists. KEY MESSAGES: • PPARα inhibited NAFLD development induced by HFD. • PPARα mediated modifications of circadian rhythm and the hepatic circadian factor CLOCK in NAFLD models. • Down-regulation of hepatic CLOCK by basal PPARα contributed to tolerance against development of NAFLD. • Inhibition of CLOCK by activated PPARα was involved in therapeutic actions against fatty liver diseases by PPARα agonists.


Assuntos
Fenofibrato , Hepatopatia Gordurosa não Alcoólica , Masculino , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , PPAR alfa/metabolismo , Fenofibrato/metabolismo , Fenofibrato/farmacologia , Regulação para Baixo , Fígado/metabolismo , Metabolismo dos Lipídeos , Dieta Hiperlipídica , Lipídeos , Camundongos Endogâmicos C57BL
2.
Toxicol Lett ; 373: 184-193, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36460194

RESUMO

The development of cholestatic liver injury (CLI) involves inflammation, but the dominant pathway mediating the chemotaxis is not yet established. This work explored key signaling pathway mediating chemotaxis in CLI and the role of Kupffer cells in the inflammatory liver injury. Probe inhibitors T-5224 (100 mg/kg) for AP-1 and C188-9 (100 mg/kg) for STAT3 were used to validate key inflammatory pathways in alpha-naphthylisothiocyanate (ANIT, 100 mg/kg)-induced CLI. Two doses of GdCl3 (10 mg/kg and 40 mg/kg) were used to delete Kupffer cells and explore their role in CLI. Serum and liver samples were collected for biochemical and mechanism analysis. The liver injury in ANIT-treated mice were significantly increased supported by biochemical and histopathological changes, and neutrophils gathering around the necrotic loci. Inhibitor treatments down-regulated liver injury biomarkers except the level of total bile acid. The chemokine Ccl2 increased by 170-fold and to a less degree Cxcl2 by 45-fold after the ANIT treatment. p-c-Jun and p-STAT3 were activated in the group A but inhibited by the inhibitors in western blot analysis. The immunofluorescence results showed AP-1 not STAT3 responded to inhibitors in ANIT-induced CLI. With or without GdCl3, there was no significant difference in liver injury among the CLI groups. In necrotic loci in CLI, CXCL2 colocalized with hepatocyte biomarker Albumin, not with the F4/80 in Kupffer cells. Conclusively, AP-1 played a more critical role in the inflammation cascade than STAT3 in ANIT-induced CLI. Hepatocytes, not the Kupffer cells released chemotactic factors mediating the chemotaxis in CLI.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Quimiotaxia , Fator de Transcrição STAT3 , Fator de Transcrição AP-1 , Animais , Camundongos , 1-Naftilisotiocianato/toxicidade , Biomarcadores , Quimiotaxia/genética , Quimiotaxia/fisiologia , Colestase/metabolismo , Hepatócitos/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Necrose/patologia , Fator de Transcrição AP-1/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fator de Transcrição STAT3/metabolismo
3.
Pharmazie ; 76(2): 103-108, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33714287

RESUMO

Cholestatic liver fibrosis occurs in liver injuries accompanied by inflammation, which develops into cirrhosis if not effectively treated in early stage. The aim of the study is to explore the effect of fenofibrate on liver fibrosis in chronic cholestatic mice. In this study, wild-type (WT) and Pparα-null (KO) mice were dosed alpha-naphthylisothiocyanate (ANIT) diet to induce chronic cholestasis. Induced liver fibrosis was determined by pathological biomarkers. Then fenofibrate 25 mg/kg was orally administrated to mice twice/day for 14 days. Serum and liver samples were collected for analysis of biochemistry and fibrosis. In WT mice, cholestatic biomarkers were increased by 5-8-fold and the expression of tissue inhibitors of metalloproteinases 1 (TIMP-1), Monocyte chemoattractant protein 1 (MCP-1), Collagen protein I (Collagen I) was increased by more than 10-fold. Fenofibrate significantly downgraded the biochemical and fibrotic biomarkers. In Western blot analysis, levels of collagenI and alpha-smooth muscle actin (α-SMA) were strongly inhibited by fenofibrate. In KO mice, liver fibrosis was induced successfully, but no improvement after fenofibrate treatment was observed. These data showed low-dose fenofibrate reverses cholestatic liver fibrosis in WT mice but not in KO mice, suggesting the dependence of therapeutic action on peroxisome proliferator-activated receptor alpha (PPARα). The study offers an additional therapeutic strategy for cholestatic liver fibrosis in practice.


Assuntos
1-Naftilisotiocianato/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Colestase/metabolismo , Fenofibrato/farmacologia , Cirrose Hepática/tratamento farmacológico , 1-Naftilisotiocianato/efeitos adversos , Actinas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Colestase/induzido quimicamente , Colestase/patologia , Colágeno Tipo I/metabolismo , Inflamação/tratamento farmacológico , Fígado/efeitos dos fármacos , Cirrose Hepática/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , PPAR alfa/deficiência , Fragmentos de Peptídeos/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
4.
Int J Mol Med ; 46(6): 2271-2279, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33125089

RESUMO

SP600125 is a classic inhibitor of c­Jun­N­terminal kinase (JNK) that is widely used in numerous medicinal studies, but its administration regimen has yet to be optimized. In the present study, intraperitoneal (i.p.) and intragastric (i.g.) injections of 15 mg/kg SP600125 was performed in mice to compare the inhibitory effect against JNK signalling in cholestasis induced by α­naphthylisothiocyanate (ANIT). SP600125 at a dose of 15 mg/kg administered by i.p. substantially decreased ANIT­induced liver injury as observed by biochemical and histopathological examinations. The adaptation of bile acid synthesis was inhibited in the A­SP­i.p. group compared with that in the A­SP­i.g. group, as indicated by the expression analysis of CYP7A1 and CYP8B1. The transcription of the pro­inflammatory factors IL­6, IL­1ß, ICAM­1 and IL­10 supported the differential toxic responses. Western blot analysis revealed that JNK signalling activated by ANIT was inhibited more markedly in the A­SP­i.p. group than in the A­SP­i.g. group. The peak concentration and the AUC0­24 of SP600125 in the A­SP­i.p. group were 5­fold and 1.56­fold higher, respectively, compared with those in the A­SP­i.g. group. These data indicated that i.p. administration of SP600125 produced a high plasma exposure profile, which directly determined its efficacy of blocking the JNK signalling. This effect of SP600125 on the JNK pathway may provide an optimized design for future in vivo investigations.


Assuntos
Antracenos/farmacologia , Colestase/patologia , Inflamação/patologia , Fígado/lesões , 1-Naftilisotiocianato , Animais , Ácidos e Sais Biliares/metabolismo , Transporte Biológico/efeitos dos fármacos , Biomarcadores/metabolismo , Colestase/sangue , Colestase/complicações , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/sangue , Inflamação/complicações , Fígado/efeitos dos fármacos , Fígado/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
Theranostics ; 10(25): 11497-11506, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33052228

RESUMO

Rationale: Light pollution leads to high risk of obesity but the underlying mechanism is not known except for the influence of altered circadian rhythm. Peroxisome proliferator-activated receptor α (PPARα) regulates lipid metabolism, but its role in circadian-related obesity is not clear. Methods: Wild-type (WT) and Ppara-null (KO) mice on a high-fat diet (HFD) were treated with neon light at night for 6 weeks. Body weights were recorded and diet consumption measured. The hypothalamus, liver, adipose and serum were collected for mechanism experimentation. Results: WT mice on a HFD and exposed to night neon light gained about 19% body weight more than the WT control mice without light exposure and KO control mice on a HFD and exposed to night neon light. The increase in adipose tissue weight and adipocyte size led to the differences in body weights. Biochemical analysis suggested increased hepatic lipid accumulated and increased transport of lipid from the liver to peripheral tissues in the WT mice that gained weight under neon light exposure. Unlike KO mice, the expression of genes involved in lipid metabolism and the circadian factor circadian locomotor output cycles kaput (CLOCK) in both liver and adipose tissues were elevated in WT mice under neon light exposure. Conclusions: PPARα mediated weight gain of HFD-treated mice exposed to night neon light. More lipids were synthesized in the liver and transported to peripheral tissue leading to adaptive metabolism and lipid deposition in the adipose tissue. These data revealed an important mechanism of obesity induced by artificial light pollution where PPARα was implicated.


Assuntos
Ritmo Circadiano/efeitos da radiação , Iluminação/efeitos adversos , Obesidade/metabolismo , PPAR alfa/metabolismo , Aumento de Peso/efeitos da radiação , Adaptação Fisiológica , Adipócitos/metabolismo , Adipócitos/efeitos da radiação , Tecido Adiposo/metabolismo , Animais , Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Humanos , Iluminação/instrumentação , Metabolismo dos Lipídeos/fisiologia , Metabolismo dos Lipídeos/efeitos da radiação , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neônio/efeitos adversos , Obesidade/etiologia , PPAR alfa/genética , Fotoperíodo , Aumento de Peso/fisiologia
6.
Mol Med Rep ; 22(2): 1583-1593, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32626965

RESUMO

α­naphthylisothiocyanate (ANIT) is used to induce intrahepatic cholestasis and it is frequently used for investigations into the disease mechanism. The lithocholic acid (LCA) cholestatic model has also been extensively used in various studies; however, to the best of our knowledge, a comparative study determining the hepatotoxic mechanisms induced by these two models has not been previously conducted. In the present study, ICR mice were treated with ANIT or LCA to induce cholestatic liver injury. Biochemical analysis was used to determine the serum. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bile acid (TBA) levels, and histopathological assessment was used to examine the liver tissue. Metabolomic analysis was used for the serum biomarker identification. Reverse transcription­quantitative PCR analysis and western blotting were used to analyze the inflammation biomarkers. The serum metabolome of the ANIT group clustered away from of the LCA group, which was demonstrated by the different modifications of the BA components. ALP level was found to be preferentially increased in the ANIT group from 24 to 48 h. Total BA levels was only increased in the ANIT group at 24 h. In contrast, AST and ALT activity levels were preferentially increased in the LCA group. The bile ducts in the hepatic tissues of the ANIT group were observed to be severely dilated, whereas the presence of edematous hepatocytes around the necrotic lesions and neutrophil infiltration were identified in the LCA group. The expression levels of cholesterol 7α­hydroxylase and sterol 12α­hydroxylase genes were significantly downregulated in the ANIT group compared with the LCA group, where a stronger adaptation of BA metabolism was supported by major differences in the concentration of the BA components. Despite the aforementioned etiological differences in the cholestasis induced by each treatment, the activation of the JNK/STAT3 signaling pathway was similar between the two cholestatic models. In conclusion, these data suggested that the liver injury induced by ANIT may be cholestatic, while the liver injury caused in the LCA model may be hepatocellular. Moreover, the downstream cholestatic liver injury in both models was indicated to be mediated by the JNK/STAT3 signaling pathway.


Assuntos
1-Naftilisotiocianato/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Colestase Intra-Hepática , Ácido Litocólico/efeitos adversos , Metaboloma , Animais , Biomarcadores/análise , Colestase Intra-Hepática/induzido quimicamente , Colestase Intra-Hepática/metabolismo , Modelos Animais de Doenças , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos ICR
7.
Pharmacol Rep ; 72(4): 935-944, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32048256

RESUMO

BACKGROUND: Fenofibrate was reported to be beneficial for cholestasis in combination with ursodeoxycholic acid. However, its therapeutic action as single therapy for chronic cholestasis and the underlying mechanism are not known. METHODS: In the present study, wild-type (WT) mice were administered a 0.05% ANIT diet to mimic chronic cholestatic liver injury. Mice were dosed fenofibrate 25 mg/kg twice every day for 10 days to investigate the therapeutic action of fenofibrate on chronic cholestatic liver injury. Ppara-null (KO) mice were used to explore PPARα's role in the therapeutic outcome. RESULTS: Fenofibrate, administered at 25 mg/kg twice daily, substantially reversed ANIT-induced chronic cholestatic liver injury shown by biochemical and pathological end points. The modifications of bile acid metabolism were found to be adaptive responses. The JNK-AP1-CCL2/CXCL2 axis was activated in all the mice administered ANIT which developed chronic cholestatic liver injury. But it was substantially decreased by fenofibrate in WT mice rather than that in KO mice. CONCLUSIONS: Low-dose fenofibrate reversed chronic cholestatic liver injury in mice. The therapeutic action was dependent on PPARα activation and occurred by inhibiting chemotaxis via the JNK-AP1-CCL2/CXCL2 signaling. These data provided an exciting basis for optimization of therapeutic fenofibrate regimen in the clinic. Additionally, they suggested anti-chemotaxis of low-dose fenofibrate in single therapy to treat cholestatic liver diseases.


Assuntos
Quimiocina CCL2/antagonistas & inibidores , Quimiocina CXCL2/antagonistas & inibidores , Quimiotaxia/efeitos dos fármacos , Colestase/tratamento farmacológico , Doença Hepática Terminal/tratamento farmacológico , Fenofibrato/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , 1-Naftilisotiocianato/toxicidade , Animais , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/metabolismo , Quimiotaxia/fisiologia , Colestase/metabolismo , Doença Hepática Terminal/induzido quimicamente , Doença Hepática Terminal/metabolismo , Fenofibrato/farmacologia , Hipolipemiantes/farmacologia , Hipolipemiantes/uso terapêutico , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout
8.
Toxicol Lett ; 300: 31-39, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30352267

RESUMO

Cholestasis is one of the most challenging diseases to be treated in current hepatology. However little is known about the adaptation difference and the underlying mechanism between acute and chronic cholestasis. In this study, wild-type and Pparα-null mice were orally administered diet containing 0.05% ANIT to induce chronic cholestasis. Biochemistry, histopathology and serum metabolome analysis exhibited the similar toxic phenotype between wild-type and Pparα-null mice. Bile acid metabolism was strongly adapted in Pparα-null mice but not in wild-type mice. The Shp and Fxr mRNA was found to be doubled in cholestatic Pparα-null mice compared with the control group. Western blot confirmed the up-regulated expression of FXR in Pparα-null mice treated with ANIT. Inflammation was found to be stronger in Pparα-null mice than those in wild-type mice in chronic cholestasis. These data chain indicated that bile acid metabolism and inflammation signaling were different between wild-type and Pparα-null mice developing chronic cholestasis, although their toxic phenotypes could not be discriminated. So basal PPARα cross-talked with FXR and inhibited bile acid metabolism adaptation in chronic cholestasis.


Assuntos
Ácidos e Sais Biliares/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Colestase/induzido quimicamente , Colestase/fisiopatologia , Isocianatos/efeitos adversos , Fígado/metabolismo , Camundongos Knockout/genética , Naftalenos/efeitos adversos , Animais , Doença Crônica , Variação Genética , Masculino , Camundongos , Fenótipo
9.
J Pharm Pharmacol ; 70(12): 1630-1642, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30251457

RESUMO

OBJECTIVES: Metabolic syndrome (MS) is the concurrence of at least three of five medical conditions: obesity, high blood pressure, insulin resistance, high serum triglyceride (TG) and low serum high-density lipoprotein levels. While fibrates are used to treat disorders other than the lowering serum TG, the mechanism by which fibrates decrease MS has not been established. METHODS: In this study, wild-type and Ppara-null mice fed a medium-fat diet (MFD) were administered gemfibrozil and fenofibrate for 3 months respectively, to explore the effect and action mechanism. KEY FINDINGS: In Ppara-null mice, MFD treatment increased body weight, adipose tissue, serum TG and impaired glucose tolerance. These phenotypes were attenuated in two groups treated with gemfibrozil and fenofibrate. The STAT3 pathway was activated in adipose and hepatic tissues in positive control, and inhibited in groups treated with gemfibrozil and fenofibrate. The above phenotypes and inflammation were not observed in any wild-type group. In 3T3-L1 adipogenic stem cells treated with high glucose, STAT3 knockdown greatly decreased the number of lipid droplets. CONCLUSIONS: Low dose of clinical fibrates was effective against MS development independent of PPARα, and this action was mediated by STAT3 signalling inhibition in adipose tissue and, to a lesser extent, in hepatic tissues.


Assuntos
Ácidos Fíbricos/farmacologia , Hipolipemiantes/farmacologia , Síndrome Metabólica/fisiopatologia , PPAR alfa/farmacologia , Fator de Transcrição STAT3/efeitos dos fármacos , Células 3T3-L1 , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal , Fenofibrato/farmacologia , Genfibrozila/farmacologia , Intolerância à Glucose/fisiopatologia , Camundongos , Camundongos Knockout , Fator de Transcrição STAT3/metabolismo , Triglicerídeos/sangue
10.
J Proteome Res ; 17(4): 1500-1508, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29498526

RESUMO

α-Naphthylisothiocyanate (ANIT) is an experimental agent used to induce intrahepatic cholestasis. The Ppara-null mouse line is widely employed to explore the physiological and pathological roles of PPARα. However, little is known about how PPARα influences the hepatotoxicity of ANIT. In the present study, wild-type and Ppara-null mice were orally treated with ANIT to induce cholestasis. The serum metabolome of wild-type mice segregated from that of the Ppara-null mice, driven by changes of bile acid (BA) metabolites. Alkaline phosphatase and total BAs were elevated preferentially in Ppara-null mice, which correlated with changes in Cyp7a1, Cyp8b1, Mrp3, Cyp3a11, Cyp2b10, Ugt1a2, and Ugt1a5 genes and showed cross-talk between basal PPARα and potentially adaptive pathways. Il6, Tnfa, and target genes in the STAT3 pathway ( Socs3, Fga, Fgb, and Fgg) were up-regulated in Ppara-null mice but not in wild-type mice. The JNK pathway was activated in both mouse lines, while NF-κB and STAT3 were activated only in Ppara-null mice. These data suggest protection against cholestasis by basal PPARα involves regulation of BA metabolism and inhibition of NF-κB/STAT3 signaling. Considering studies on the protective effects of both basal and activated PPARα, caution should be exercised when one attempts to draw conclusions in which the PPARα is modified by genetic manipulation, fasting, or activation in pharmacological and toxicological studies.


Assuntos
Colestase/metabolismo , Metabolômica , PPAR alfa/fisiologia , 1-Naftilisotiocianato/farmacologia , Animais , Ácidos e Sais Biliares/antagonistas & inibidores , Ácidos e Sais Biliares/metabolismo , Colestase/induzido quimicamente , Camundongos Endogâmicos , Camundongos Knockout , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Substâncias Protetoras , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
11.
Biopharm Drug Dispos ; 38(9): 535-542, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28946176

RESUMO

Gemfibrozil is a fibrate drug used widely for dyslipidemia associated with atherosclerosis. Clinically, both gemfibrozil and its phase II metabolite gemfibrozil 1-O-ß-glucuronide (gem-glu) are involved in drug-drug interaction (DDI). But the DDI risk caused by gem-glu between human and mice has not been compared. In this study, six volunteers were recruited and took a therapeutic dose of gemfibrozil for 3 days for examination of the gemfibrozil and gem-glu level in human. Male mice were fed a gemfibrozil diet (0.75%) for 7 days, following which a cocktail-based inhibitory DDI experiment was performed. Plasma samples and liver tissues from mice were collected for determination of gemfibrozil, gem-glu concentration and cytochrome p450 enzyme (P450) induction analysis. In human, the molar ratio of gem-glu/gemfibrozil was 15% and 10% at the trough concentration and the concentration at 1.5 h after the 6th dose. In contrast, this molar ratio at steady state in mice was 91%, demonstrating a 6- to 9-fold difference compared with that in human. Interestingly, a net induction of P450 activity and in vivo inductive DDI potential in mice was revealed. The P450 activity was not inhibited although the gem-glu concentration was high. These data suggested species difference of relative gem-glu exposure between human and mice, as well as a net inductive DDI potential of gemfibrozil in mouse model.


Assuntos
Indutores das Enzimas do Citocromo P-450/farmacocinética , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Genfibrozila/análogos & derivados , Glucuronatos/farmacocinética , Hipolipemiantes/farmacocinética , Adulto , Animais , Indutores das Enzimas do Citocromo P-450/administração & dosagem , Indutores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Genfibrozila/farmacocinética , Genfibrozila/farmacologia , Glucuronatos/farmacologia , Humanos , Hipolipemiantes/administração & dosagem , Hipolipemiantes/farmacologia , Fígado/metabolismo , Masculino , Camundongos , Especificidade da Espécie , Fatores de Tempo , Adulto Jovem
12.
Br J Pharmacol ; 174(18): 3000-3017, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28646549

RESUMO

BACKGROUND AND PURPOSE: Fenofibrate, a PPARα agonist, is the most widely prescribed drug for treating hyperlipidaemia. Although fibrate drugs are reported to be beneficial for cholestasis, their underlying mechanism has not been determined. EXPERIMENTAL APPROACH: Wild-type mice and Pparα-null mice were pretreated orally with fenofibrate for 3 days, following which α-naphthylisothiocyanate (ANIT) was administered to induce cholestasis. The PPARα agonist WY14643 and JNK inhibitor SP600125 were used to determine the role of PPARα and the JNK pathway, respectively, in cholestatic liver injury. The same fenofibrate regimen was applied to investigate its beneficial effects on sclerosing cholangitis in a DDC-induced cholestatic model. KEY RESULTS: Fenofibrate, 25 mg·kg-1 twice a day, totally attenuated ANIT-induced cholestasis and liver injury as indicated by biochemical and histological analyses. This protection occurred in wild-type, but not in Pparα-null, mice. Alterations in bile acid synthesis and transport were found to be an adaptive response rather than a direct effect of fenofibrate. WY14643 attenuated ANIT-induced cholestasis and liver injury coincident with inhibition of JNK signalling. Although SP600125 did not affect cholestasis, it inhibited liver injury in the ANIT model when the dose of fenofibrate used was ineffective. Fenofibrate was also revealed to have a beneficial effect in the sclerosing cholangitis model. CONCLUSIONS AND IMPLICATIONS: These data suggest that the protective effects of fenofibrate against cholestasis-induced hepatic injury are dependent on PPARα and fenofibrate dose, and are mediated through inhibition of JNK signalling. This mechanism of fenofibrate protection against intrahepatic cholestasis may offer additional therapeutic opportunities for cholestatic liver diseases.


Assuntos
Antracenos/farmacologia , Colestase Intra-Hepática/tratamento farmacológico , Fenofibrato/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , PPAR alfa/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , 1-Naftilisotiocianato , Animais , Antracenos/química , Colestase Intra-Hepática/induzido quimicamente , Colestase Intra-Hepática/patologia , Relação Dose-Resposta a Droga , Fenofibrato/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Knockout , PPAR alfa/agonistas , Inibidores de Proteínas Quinases/química , Pirimidinas/química , Relação Estrutura-Atividade
13.
Basic Clin Pharmacol Toxicol ; 121(3): 169-174, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28374976

RESUMO

Gemfibrozil, a peroxisome proliferator-activated receptor α (PPARα) agonist, is widely used for hypertriglyceridaemia and mixed hyperlipidaemia. Drug-drug interaction of gemfibrozil and other PPARα agonists has been reported. However, the role of PPARα in cytochrome P450 (CYP) induction by fibrates is not well known. In this study, wild-type mice were first fed gemfibrozil-containing diets (0.375%, 0.75% and 1.5%) for 14 days to establish a dose-response relationship for CYP induction. Then, wild-type mice and Pparα-null mice were treated with a 0.75% gemfibrozil-containing diet for 7 days. CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. In Pparα-null mice, their mRNA level, protein level and activity were induced more than those in wild-type mice. So, gemfibrozil induced CYP, and this action was inhibited by activated PPARα. These data suggested that the induction potential of CYPs was suppressed by activated PPARα, showing a potential role of this receptor in drug-drug interactions and metabolic diseases treated with fibrates.


Assuntos
Inibidores do Citocromo P-450 CYP2C8/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Genfibrozila/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipolipemiantes/farmacologia , Fígado/efeitos dos fármacos , PPAR alfa/agonistas , Animais , Citocromo P-450 CYP2B1/química , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Inibidores do Citocromo P-450 CYP2C8/administração & dosagem , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/genética , Relação Dose-Resposta a Droga , Indução Enzimática/efeitos dos fármacos , Genfibrozila/administração & dosagem , Hipolipemiantes/administração & dosagem , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , RNA Mensageiro/metabolismo
14.
Arch Toxicol ; 91(2): 897-907, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27344344

RESUMO

Perfluorodecanoic acid (PFDA) is widely used in production of many daily necessities based on their surface properties and stability. It was assigned as a Persistent Organic Pollutant in 2009 and became a public concern partly because of its potential for activation of the peroxisome proliferator-activated receptor alpha (PPARα). In this study, wild-type and Ppara-null mice were administered PFDA (80 mg/kg). Blood and liver tissues were collected and subjected to systemic toxicological and mechanistic analysis. UPLC-ESI-QTOFMS-based metabolomics was used to explore the contributing components of the serum metabolome that led to variation between wild-type and Pparα-null mice. Bile acid homeostasis was disrupted, and slight hepatocyte injury in wild-type mice accompanied by adaptive regulation of bile acid synthesis and transport was observed. The serum metabolome in wild-type clustered differently from that in Pparα-null, featured by sharp increases in bile acid components. Differential toxicokinetic tendency was supported by regulation of UDP-glucuronosyltransferases dependent on PPARα, but it did not contribute to the hepatotoxic responses. Increase in Il-10 and activation of the JNK pathway indicated inflammation was induced by disruption of bile acid homeostasis in wild-type mice. Inhibition of p-p65 dependent on PPARα activation by PFDA stopped the inflammatory cascade, as indicated by negative response of Il-6, Tnf-α, and STAT3 signaling. These data suggest disruptive and protective role of PPARα in hepatic responses induced by PFDA.


Assuntos
Ácidos Decanoicos/toxicidade , Fluorocarbonos/toxicidade , Fígado/efeitos dos fármacos , PPAR alfa/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Homeostase/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Fígado/metabolismo , Fígado/patologia , Metaboloma/efeitos dos fármacos , Camundongos Mutantes , Camundongos Transgênicos , PPAR alfa/genética , Toxicocinética , Cisto do Úraco
15.
Environ Toxicol Pharmacol ; 49: 112-118, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27984778

RESUMO

TiO2 nano-particle (TiO2 NP) is widely used in industrial, household necessities, as well as medicinal products. However, the effect of TiO2 NP on liver metabolic function has not been reported. In this study, after mice were orally administered TiO2 NP (21nm) for 14days, the serum and liver tissues were assayed by biochemical analysis, real time quantitative polymerase chain reaction, western blot and transmission electron microscopy. The serum bilirubin was increased in a dose dependent manner. Deposition of TiO2 NP in hepatocytes and the abnormality of microstructures was observed. Expression of metabolic genes involved in the endogenous and exogenous metabolism was modified, supporting the toxic phenotype. Collectively, oral administration of TiO2 NP (21nm) led to deposition of particles in hepatocytes, mitochondrial edema, and the disturbance of liver metabolism function. These data suggested oral administration disrupts liver metabolic functions, which was more sensitive than regular approaches to detect material hepatotoxicity. This study provided useful information for risk analysis and regulation of TiO2 NPs by administration agencies.


Assuntos
Fígado/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Titânio/toxicidade , Administração Oral , Alanina Transaminase/sangue , Animais , Proteínas Reguladoras de Apoptose/genética , Aspartato Aminotransferases/sangue , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/genética , Fígado/metabolismo , Fígado/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , RNA Mensageiro/metabolismo
16.
J Pharm Pharmacol ; 68(9): 1203-13, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27367057

RESUMO

OBJECTIVES: Chlorogenic acid (CGA) is one of the most widely consumed polyphenols in diets and is recognized to be a natural hepatoprotective agent. Here, we evaluated the protective effect and the potential mechanism of CGA against ɑ-naphthylisothiocyanate (ANIT)-induced cholestasis and liver injury. METHODS: Twenty-five male 129/Sv mice were administered with CGA, and ANIT challenge was performed at 75 mg/kg on the 4th day. Blood was collected and subjected to biochemical analysis; the liver tissues were examined using histopathological analysis and signalling pathways. KEY FINDINGS: Chlorogenic acid almost totally attenuated the ANIT-induced liver damage and cholestasis, compared with the ANIT group. Dose of 50 mg/kg of CGA significantly prevented ANIT-induced changes in serum levels of alanine aminotransferase, alkaline phosphatases, total bile acid, direct bilirubin, indirect bilirubin (5.3-, 6.3-, 18.8-, 158-, 41.4-fold, P<0.001) and aspartate aminotransferase (4.6-fold, P<0.01). Expressions of the altered bile acid metabolism and transport-related genes were normalized by cotreatment with CGA. The expressions of interleukin 6, tumour necrosis factor-α and suppressor of cytokine signalling 3 were found to be significantly decreased (1.2-fold, ns; 11.0-fold, P<0.01; 4.4-fold, P<0.05) in the CGA/ANIT group. Western blot revealed that CGA inhibited the activation and expression of signal transducer and activator of transcription 3 and NFκB. CONCLUSIONS: These data suggest that CGA inhibits both ANIT-induced intrahepatic cholestasis and the liver injury. This protective effect involves down-regulation of STAT3 and NFκB signalling.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Ácido Clorogênico/farmacologia , Colestase/metabolismo , Fígado/efeitos dos fármacos , NF-kappa B/metabolismo , Fitoterapia , Fator de Transcrição STAT3/metabolismo , 1-Naftilisotiocianato , Animais , Ácidos e Sais Biliares/metabolismo , Bilirrubina/sangue , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Ácido Clorogênico/uso terapêutico , Colestase/prevenção & controle , Citocinas/sangue , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Polifenóis/farmacologia , Polifenóis/uso terapêutico , Transdução de Sinais
17.
Pharmazie ; 71(4): 205-12, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27209701

RESUMO

BACKGROUND: Concurrence of high glucose or diabetes in patients with dyslipidemia is presenting major challenges for clinicians. Although sporadically reported, a rational basis for the use of fibrates for the treatment of dyslipidemia with concurrent metabolic syndrome has not been established. METHODS: In this study, wild-type (WT) and Ppara-null (KO) mice were fed a serial gemfibrozil- and fenofibrate-containing diet under the same experimental conditions for 14 days. Glucose level in the blood, glycogen storage in the liver tissues, and the potential toxic responses were assayed. Genes involved in glucose metabolism were determined by quantitative polymerase chain reaction analysis. RESULTS: Both the blood glucose level and the glycogen content in the liver were down-regulated by gemfibrozil but not by fenofibrate in WT mice, in a dose-dependent manner. This decrement did not occur in KO mice for either fibrate agent. Secondary regulation on the transcription of pyruvate kinase, and gluconolactonase were observed following gemfibrozil treatment, which was differential between WT mice and KO mice. CONCLUSIONS: Gemfibrozil, not fenofibrate, down-regulates systemic glucose level and glycogen storage in the liver dependent on PPARα, suggesting its potential value for treatment of dyslipidemia with concurrent diabetes or high glucose levels.


Assuntos
Fenofibrato/farmacologia , Genfibrozila/farmacologia , Glucose/metabolismo , Hipolipemiantes/farmacologia , PPAR alfa/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Glicogênio/metabolismo , Hepatomegalia/genética , Hepatomegalia/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , PPAR alfa/genética , Piruvato Quinase/biossíntese
19.
Pharmacol Rep ; 67(2): 299-304, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25712654

RESUMO

BACKGROUND: The antiepileptic drug carbamazepine (CBZ) is a typical inducer of cytochrome P450 (CYP) 3A and 2C in the clinic. It is considered a strong constitutive androstane receptor activator, however both CBZ and its main metabolite CBZ 10, 11-epoxide have been reported to be pregnane X receptor (PXR) activators whose maximal efficacy and potency are comparable with the human PXR ligand rifampicin. It is unknown whether or not PXR plays a substantially important role in in vivo induction of CYP by CBZ administration. METHODS: In this study, wild type and Pxr-/- mice were administered with CBZ for 5 days. Serum and liver samples were collected and subjected to hepatotoxicity assessment and CYP induction analysis. RESULTS: CYP2b, 2c and 3a were induced similarly in terms of transcription level, enzyme activity and protein abundance in both wild type and Pxr-/- mice. Inductive profile of CYPs in mice by CBZ administration accorded with those reported in rats, but differed from clinically reported data. CONCLUSIONS: These data suggest that in vivo induction of CYP in mice by multiple administration of CBZ is independent of PXR. Knowledge of the featured CYP induction profile in mice helps us understand species related CYP induction profiles among rodents and humans resulting from administration of CBZ.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Carbamazepina/farmacologia , Citocromo P-450 CYP3A/biossíntese , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/efeitos dos fármacos , Proteínas de Membrana/biossíntese , Receptores de Esteroides/deficiência , Esteroide Hidroxilases/biossíntese , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Carbamazepina/sangue , Carbamazepina/farmacocinética , Família 2 do Citocromo P450 , Indução Enzimática/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Camundongos Knockout , Receptor de Pregnano X , Receptores de Esteroides/genética
20.
Chem Biol Interact ; 223: 80-6, 2014 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-25265579

RESUMO

Overdose of acetaminophen (APAP) can cause acute liver injury that is sometimes fatal, requiring efficient pharmacological intervention. The traditional Chinese herb Bupleurum falcatum has been widely used for the treatment of several liver diseases in eastern Asian countries, and saikosaponin d (SSd) is one of its major pharmacologically-active components. However, the efficacy of Bupleurum falcatum or SSd on APAP toxicity remains unclear. C57/BL6 mice were administered SSd intraperitoneally once daily for 5days, followed by APAP challenge. Biochemical and pathological analysis revealed that mice treated with SSd were protected against APAP-induced hepatotoxicity. SSd markedly suppressed phosphorylation of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) and reversed the APAP-induced increases in the target genes of NF-κB, such as pro-inflammatory cytokine Il6 and Ccl2, and those of STAT3, such as Socs3, Fga, Fgb and Fgg. SSd also enhanced the expression of the anti-inflammatory cytokine Il10 mRNA. Collectively, these results demonstrate that SSd protects mice from APAP-induced hepatotoxicity mainly through down-regulating NF-κB- and STAT3-mediated inflammatory signaling. This study unveils one of the possible mechanisms of hepatoprotection caused by Bupleurum falcatum and/or SSd.


Assuntos
Acetaminofen/antagonistas & inibidores , Acetaminofen/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Ácido Oleanólico/análogos & derivados , Saponinas/farmacologia , Analgésicos não Narcóticos/toxicidade , Animais , Antipiréticos/antagonistas & inibidores , Antipiréticos/toxicidade , Bupleurum , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/genética , Regulação para Baixo/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Inativação Metabólica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Ácido Oleanólico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/metabolismo , Plantas Medicinais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...