Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791514

RESUMEN

Supplementation with fish oil rich in omega-3 polyunsaturated fatty acids (n-3 PUFAs) effectively reduces acute and chronic alcohol-induced hepatic steatosis. We aimed to find molecular mechanisms underlying the effects of n-3 PUFAs in alcohol-induced hepatic steatosis. Because free fatty acid receptor 4 (FFA4, also known as GPR120) has been found as a receptor for n-3 PUFAs in an ethanol-induced liver steatosis model, we investigated whether n-3 PUFAs protect against liver steatosis via FFA4 using AH7614, an FFA4 antagonist, and Ffa4 knockout (KO) mice. N-3 PUFAs and compound A (CpdA), a selective FFA4 agonist, reduced the ethanol-induced increase in lipid accumulation in hepatocytes, triglyceride content, and serum ALT levels, which were not observed in Ffa4 KO mice. N-3 PUFAs and CpdA also reduced the ethanol-induced increase in lipogenic sterol regulatory element-binding protein-1c expression in an FFA4-dependent manner. In Kupffer cells, treatment with n-3 PUFA and CpdA reversed the ethanol-induced increase in tumor necrosis factor-α, cyclooxygenase-2, and NLR family pyrin domain-containing 3 expression levels in an FFA4-dependent manner. In summary, n-3 PUFAs protect against ethanol-induced hepatic steatosis via the anti-inflammatory actions of FFA4 on Kupffer cells. Our findings suggest FFA4 as a therapeutic target for alcoholic hepatic steatosis.


Asunto(s)
Etanol , Ácidos Grasos Omega-3 , Hígado Graso Alcohólico , Macrófagos del Hígado , Receptores Acoplados a Proteínas G , Animales , Masculino , Ratones , Ácidos Grasos Omega-3/farmacología , Hígado Graso Alcohólico/tratamiento farmacológico , Hígado Graso Alcohólico/prevención & control , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Sustancias Protectoras/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Triglicéridos/metabolismo
2.
Hepatology ; 73(3): 952-967, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32418220

RESUMEN

BACKGROUND AND AIMS: Chronic alcohol consumption is accompanied by intestinal inflammation. However, little is known about how alterations to the intestinal immune system and sphingolipids contribute to the pathogenesis of alcohol-associated liver disease (ALD). APPROACH AND RESULTS: We used wild-type mice, retinoid-related orphan receptor gamma t (RORγt)-deficient mice, sphingosine kinase-deficient mice, and local gut anti-inflammatory, 5-aminosalicyclic acid-treated mice in a chronic-binge ethanol feeding model. Targeted lipidomics assessed the sphingolipids in gut and liver samples. Gut immune cell populations, the amounts of sphingolipids, and the level of liver injury were examined. Alcohol intake induces a pro-inflammatory shift in immune cell populations in the gut, including an increase in Th17 cells. Using RORγt-deficient mice, we found that Th17 cells are required for alcohol-associated gut inflammation and the development of ALD. Treatment with 5-aminosalicyclic acid decreases alcohol-induced liver injury and reverses gut inflammation by the suppression of CD4+ /RORγt+ /interleukin-17A+ cells. Increased Th17 cells were due to up-regulation of sphingosine kinase 1 activity and RORγt activation. We found that S1P/S1PR1 signaling is required for the development of Th17 cell-mediated ALD. Importantly, in vivo intervention blocking of S1P/S1PR1 signaling markedly attenuated alcohol-induced liver inflammation, steatosis, and damage. CONCLUSIONS: Gut inflammation is a functional alteration of immune cells in ALD. Reducing gut Th17 cells leads to reduced liver damage. S1P signaling was crucial in the pathogenesis of ALD in a Th17 cell-dependent manner. Furthermore, our findings suggest that compounds that reduce gut inflammation locally may represent a unique targeted approach in the treatment of ALD.


Asunto(s)
Etanol/efectos adversos , Hígado Graso Alcohólico/prevención & control , Lisofosfolípidos/farmacología , Esfingosina/análogos & derivados , Células Th17/fisiología , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Modelos Animales de Enfermedad , Hígado Graso Alcohólico/etiología , Femenino , Intestinos/citología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Masculino , Mesalamina/farmacología , Mesalamina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esfingosina/farmacología , Células Th17/efectos de los fármacos
3.
Molecules ; 26(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34834064

RESUMEN

Alcohol is metabolized in liver. Chronic alcohol abuse results in alcohol-induced fatty liver and liver injury. Red quinoa (Chenopodium formosanum) was a traditional staple food for Taiwanese aborigines. Red quinoa bran (RQB) included strong anti-oxidative and anti-inflammatory polyphenolic compounds, but it was usually regarded as the agricultural waste. Therefore, this study is to investigate the effect of water and ethanol extraction products of RQB on the prevention of liquid alcoholic diet-induced acute liver injury in mice. The mice were given whole grain powder of red quinoa (RQ-P), RQB ethanol extract (RQB-E), RQB water extract (RQB-W), and rutin orally for 6 weeks, respectively. The results indicated that RQB-E, RQB-W, and rutin decreased alcoholic diet-induced activities of aspartate aminotransferase and alanine aminotransferase, and the levels of serum triglyceride, total cholesterol, and hepatic triglyceride. Hematoxylin and eosin staining of liver tissues showed that RQB-E and RQB-W reduced lipid droplet accumulation and liver injury. However, ethanol extraction process can gain high rutin and antioxidative agents contents from red quinoa, that showed strong effects in preventing alcoholic fatty liver disease and liver injury via increasing superoxide dismutase/catalase antioxidative system and repressing the expressions of fatty acid synthesis enzyme acetyl-CoA carboxylase.


Asunto(s)
Antioxidantes/uso terapéutico , Chenopodium quinoa , Hígado Graso Alcohólico/prevención & control , Extractos Vegetales/uso terapéutico , Rutina/uso terapéutico , Animales , Antioxidantes/química , Chenopodium quinoa/química , Etanol/efectos adversos , Ácidos Grasos/metabolismo , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/metabolismo , Lipogénesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/química , Rutina/química
4.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G428-G438, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31928222

RESUMEN

Enhanced free fatty acid (FFA) flux from adipose tissue (AT) to liver plays an important role in the development of nonalcoholic steatohepatitis (NASH) and alcohol-associated liver disease (AALD). We determined the effectiveness of nanoformulated superoxide dismutase 1 (Nano) in attenuating liver injury in a mouse model exhibiting a combination of NASH and AALD. Male C57BL6/J mice were fed a chow diet (CD) or a high-fat diet (HF) for 10 wk followed by pair feeding of the Lieber-DeCarli control (control) or ethanol (ET) diet for 4 wk. Nano was administered once every other day for the last 2 wk of ET feeding. Mice were divided into 1) CD + control diet (CD + Cont), 2) high-fat diet (HF) + control diet (HF + Cont), 3) HF + Cont + Nano, 4) HF + ET diet (HF + ET), and 5) HF + ET + Nano. The total fat mass, visceral AT mass (VAT), and VAT perilipin 1 content were significantly lower only in HF + ET-fed mice but not in HF + ET + Nano-treated mice compared with controls. The HF + ET-fed mice showed an upregulation of VAT CYP2E1 protein, and Nano abrogated this effect. We noted a significant rise in plasma FFAs, ALT, and monocyte chemoattractant protein-1 in HF + ET-fed mice, which was blunted in HF + ET + Nano-treated mice. HF + ET-induced increases in hepatic steatosis and inflammatory markers were attenuated upon Nano treatment. Nano reduced hepatic CYP2E1 and enhanced catalase levels in HF + ET-fed mice with a concomitant increase in SOD1 protein and activity in liver. Nano was effective in attenuating AT and liver injury in mice exhibiting a combination of NASH and AALD, partly via reduced CYP2E1-mediated ET metabolism in these organs.NEW & NOTEWORTHY Increased free fatty acid flux from adipose tissue (AT) to liver accompanied by oxidative stress promotes nonalcoholic steatohepatitis (NASH) and alcohol-associated liver injury (AALD). Obesity increases the severity of AALD. Using a two-hit model involving a high-fat diet and chronic ethanol feeding to mice, and treating them with nanoformulated superoxide dismutase (nanoSOD), we have shown that nanoSOD improves AT lipid storage, reduces CYP2E1 in AT and liver, and attenuates the combined NASH/AALD in mice.


Asunto(s)
Citocromo P-450 CYP2E1/metabolismo , Hígado Graso Alcohólico/prevención & control , Grasa Intraabdominal/efectos de los fármacos , Hígado/efectos de los fármacos , Nanopartículas , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Superóxido Dismutasa-1/administración & dosificación , Adiposidad/efectos de los fármacos , Animales , Catalasa/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Composición de Medicamentos , Hígado Graso Alcohólico/enzimología , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Regulación de la Expresión Génica , Grasa Intraabdominal/enzimología , Grasa Intraabdominal/patología , Lipólisis/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Nanomedicina , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo/efectos de los fármacos , Perilipina-1/genética , Perilipina-1/metabolismo , Transducción de Señal , Superóxido Dismutasa-1/química
5.
Alcohol Clin Exp Res ; 44(5): 1046-1060, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32267550

RESUMEN

BACKGROUND: Excess alcohol (ethanol, EtOH) consumption is a significant cause of chronic liver disease, accounting for nearly half of the cirrhosis-associated deaths in the United States. EtOH-induced liver toxicity is linked to EtOH metabolism and its associated increase in proinflammatory cytokines, oxidative stress, and the subsequent activation of Kupffer cells. Dihydromyricetin (DHM), a bioflavonoid isolated from Hovenia dulcis, can reduce EtOH intoxication and potentially protect against chemical-induced liver injuries. But there remains a paucity of information regarding the effects of DHM on EtOH metabolism and liver protection. As such, the current study tests the hypothesis that DHM supplementation enhances EtOH metabolism and reduces EtOH-mediated lipid dysregulation, thus promoting hepatocellular health. METHODS: The hepatoprotective effect of DHM (5 and 10 mg/kg; intraperitoneal injection) was evaluated using male C57BL/6J mice and a forced drinking ad libitum EtOH feeding model and HepG2/VL-17A hepatoblastoma cell models. EtOH-mediated lipid accumulation and DHM effects against lipid deposits were determined via H&E stains, triglyceride measurements, and intracellular lipid dyes. Protein expression of phosphorylated/total proteins and serum and hepatic cytokines was determined via Western blot and protein array. Total NAD+ /NADH Assay of liver homogenates was used to detect NAD + levels. RESULTS: DHM reduced liver steatosis, liver triglycerides, and liver injury markers in mice chronically fed EtOH. DHM treatment resulted in increased activation of AMPK and downstream targets, carnitine palmitoyltransferase (CPT)-1a, and acetyl CoA carboxylase (ACC)-1. DHM induced expression of EtOH-metabolizing enzymes and reduced EtOH and acetaldehyde concentrations, effects that may be partly explained by changes in NAD+ . Furthermore, DHM reduced the expression of proinflammatory cytokines and chemokines in sera and cell models. CONCLUSION: In total, these findings support the utility of DHM as a dietary supplement to reduce EtOH-induced liver injury via changes in lipid metabolism, enhancement of EtOH metabolism, and suppressing inflammation responses to promote liver health.


Asunto(s)
Etanol/efectos adversos , Etanol/metabolismo , Flavonoles/administración & dosificación , Metabolismo de los Lípidos/efectos de los fármacos , Hepatopatías Alcohólicas/prevención & control , Hígado/metabolismo , Adenilato Quinasa/metabolismo , Animales , Suplementos Dietéticos , Activación Enzimática/efectos de los fármacos , Hígado Graso Alcohólico/prevención & control , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Triglicéridos/metabolismo
6.
J Proteome Res ; 18(11): 3821-3830, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31612718

RESUMEN

Alcoholic steatosis is one of the most prevalent forms of liver disease, and appropriate insight and application of anti-steatosis drugs must be considered. Geniposide, the major active constituent of the Gardenia jasminoides (Ellis) fruit, has been commonly used as a traditional herbal medicine for the treatment of liver diseases. However, its hepatoprotective effect on alcoholic steatosis has not been reported. Moreover, geniposide overdose-induced hepatotoxicity was demonstrated. Hence, its therapeutic effects and overdose-induced hepatotoxicity in rat models along with corresponding targets, especially the targets of transcription factors (TFs), were systematically investigated in this study by using a concatenated tandem array of consensus TF response elements. The results indicate that geniposide can attenuate alcoholic steatosis and liver injury by enhancing the transcriptional activities of peroxisome proliferator-activated receptor-α and hepatocyte nuclear factors 1α and 4α, while geniposide overdose perturbs other TFs. In addition, therapeutic doses and overdoses of geniposide have differentiated target TFs. This study is the first to provide a systematic insight into the difference of critical transcription factors between the actions of therapeutic doses and overdoses of geniposide, as well as much-needed attention to the important topic of alcoholic liver disease therapy.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado Graso Alcohólico/metabolismo , Iridoides/administración & dosificación , Proteómica/métodos , Factores de Transcripción/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Sobredosis de Droga/complicaciones , Hígado Graso Alcohólico/prevención & control , Frutas/química , Gardenia/química , Iridoides/efectos adversos , Masculino , PPAR alfa/metabolismo , Fitoterapia/efectos adversos , Fitoterapia/métodos , Proteoma/metabolismo , Ratas Sprague-Dawley
7.
IUBMB Life ; 71(11): 1740-1750, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31265202

RESUMEN

Alcoholic liver disease (ALD) is generated from excessive alcohol consumption, characterized by hepatic steatosis. Mechanistically, excessive hepatic lipid accumulation was attributed to the aberrant lipin-1 signaling during the development of alcoholic steatosis in rodent species and human. Dihydroartemisinin (DHA) has been recently identified to relieve hepatocytes necrosis and prevent from hepatic steatosis in alcohol-induced liver diseases; however, the role of DHA in ALD has not been elucidated completely. Therefore, this study was aimed to further identify the potential mechanisms of pharmacological effects of DHA on ALD. Results demonstrated that DHA regulated the expression and nucleocytoplasmic shuttling of lipin-1 in mice with chronic ethanol exposure. Results confirmed that the disruption of lipin-1 signaling abolished the suppression of DHA on alcohol-induced hepatic steatosis. Interestingly, DHA also significantly improved liver injury, and inflammation mediated by lipin-1 signaling in chronic alcohol-fed mice. in vivo experiments further consolidated the concept that DHA protected against hepatocyte lipoapoptosis dependent on the regulation of nucleocytoplasmic shuttling of lipin-1 signaling, resulting in attenuated ratio of Lpin1 ß/α. Obvious increases in cell apoptosis were observed in alcohol-treated lipin1ß-overexpressed mice. Although DHA attenuated cell apoptosis, overexpression of lipin-1ß neutralized DHA action. DHA ameliorated activation of endoplasmic reticulum stress through inhibiting activation of JNK and CHOP, which was abrogated by overexpression of lipin-1ß. In summary, DHA significantly improved liver injury, steatosis and hepatocyte lipoapoptosis in chronic alcohol-fed mice via regulation of lipin-1 signaling.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Hígado Graso Alcohólico/prevención & control , Regulación de la Expresión Génica/efectos de los fármacos , Fosfatidato Fosfatasa/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Fosfatidato Fosfatasa/genética
8.
Alcohol Clin Exp Res ; 43(7): 1376-1383, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30908637

RESUMEN

BACKGROUND: A mouse with hepatocyte-specific deiodinase type II inactivation (Alb-D2KO) is resistant to diet-induced obesity, hepatic steatosis, and hypertriglyceridemia due to perinatal epigenetic modifications in the liver. This phenotype is linked to low levels of Zfp125, a hepatic transcriptional repressor that promotes liver steatosis by inhibiting genes involved in packaging and secretion of very-low-density lipoprotein. METHODS: Here, we used chronic and binge ethanol (EtOH) in mice to cause liver steatosis. RESULTS: The EtOH treatment causes a 2.3-fold increase in hepatic triglyceride content; Zfp125 levels were approximately 50% higher in these animals. In contrast, Alb-D2KO mice did not develop EtOH-induced liver steatosis. They also failed to elevate Zfp125 to the same levels, despite being on the EtOH-containing diet for the same period of time. Their phenotype was associated with 1.3- to 2.9-fold up-regulation of hepatic genes involved in lipid transport and export that are normally repressed by Zfp125, that is, Mttp, Abca1, Ldlr, Apoc1, Apoc3, Apoe, Apoh, and Azgp1. Furthermore, genes involved in the EtOH metabolic pathway, that is, Aldh2 and Acss2, were also 1.6- to 3.1-fold up-regulated in Alb-D2KO EtOH mice compared with control animals kept on EtOH. CONCLUSIONS: EtOH consumption elevates expression of Zfp125. Alb-D2KO animals, which have lower levels of Zfp125, are much less susceptible to EtOH-induced liver steatosis.


Asunto(s)
Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/prevención & control , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Hígado/metabolismo , Alcoholismo/complicaciones , Alcoholismo/genética , Animales , Consumo Excesivo de Bebidas Alcohólicas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dieta , Etanol/metabolismo , Hígado Graso , Hígado Graso Alcohólico/metabolismo , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Redes y Vías Metabólicas/genética , Ratones , Ratones Noqueados , Triglicéridos/metabolismo , Yodotironina Deyodinasa Tipo II
9.
J Hepatol ; 66(3): 589-600, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27984176

RESUMEN

BACKGROUND & AIMS: Mitochondrial dysfunction, oxidative stress, inflammation, and metabolic reprograming are crucial contributors to hepatic injury and subsequent liver fibrosis. Poly(ADP-ribose) polymerases (PARP) and their interactions with sirtuins play an important role in regulating intermediary metabolism in this process. However, there is little research into whether PARP inhibition affects alcoholic and non-alcoholic steatohepatitis (ASH/NASH). METHODS: We investigated the effects of genetic deletion of PARP1 and pharmacological inhibition of PARP in models of early alcoholic steatohepatitis, as well as on Kupffer cell activation in vitro using biochemical assays, real-time PCR, and histological analyses. The effects of PARP inhibition were also evaluated in high fat or methionine and choline deficient diet-induced steatohepatitis models in mice. RESULTS: PARP activity was increased in livers due to excessive alcohol intake, which was associated with decreased NAD+ content and SIRT1 activity. Pharmacological inhibition of PARP restored the hepatic NAD+ content, attenuated the decrease in SIRT1 activation and beneficially affected the metabolic-, inflammatory-, and oxidative stress-related alterations due to alcohol feeding in the liver. PARP1-/- animals were protected against alcoholic steatohepatitis and pharmacological inhibition of PARP or genetic deletion of PARP1 also attenuated Kupffer cell activation in vitro. Furthermore, PARP inhibition decreased hepatic triglyceride accumulation, metabolic dysregulation, or inflammation and/or fibrosis in models of NASH. CONCLUSION: Our results suggests that PARP inhibition is a promising therapeutic strategy in steatohepatitis with high translational potential, considering the availability of PARP inhibitors for clinical treatment of cancer. LAY SUMMARY: Poly(ADP-ribose) polymerases (PARP) are the most abundant nuclear enzymes. The PARP inhibitor olaparib (Lynparza) is a recently FDA-approved therapy for cancer. This study shows that PARP is overactivated in livers of subjects with alcoholic liver disease and that pharmacological inhibition of this enzyme with 3 different PARP inhibitors, including olaparib, attenuates high fat or alcohol induced liver injury, abnormal metabolic alteration, fat accumulation, inflammation and/or fibrosis in preclinical models of liver disease. These results suggest that PARP inhibition is a promising therapeutic strategy in the treatment of alcoholic and non-alcoholic liver diseases.


Asunto(s)
Hígado Graso Alcohólico/prevención & control , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/metabolismo , Humanos , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NAD/metabolismo , Estrés Nitrosativo/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fenantrenos/farmacología , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/deficiencia , Poli(ADP-Ribosa) Polimerasa-1/genética , Quinolinas/farmacología , Sirtuina 1/metabolismo
10.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3190-3201, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847514

RESUMEN

Alcohol exposure induces adipose hyperlipolysis and causes excess fatty acid influx into the liver, leading to alcoholic steatosis. The impacts of omega-3 polyunsaturated fatty acids (n-3 PUFA) on ethanol-induced fatty liver are well documented. However, the role of n-3 PUFA in ethanol-induced adipose lipolysis has not been sufficiently addressed. In this study, the fat-1 transgenic mice that synthesizes endogenous n-3 from n-6 PUFA and their wild type littermates with an exogenous n-3 PUFA enriched diet were subjected to a chronic ethanol feeding plus a single binge as model to induce liver injury with adipose lipolysis. Additionally, the differentiated adipocytes from 3T3-L1 cells were treated with docosahexaenoic acid or eicosapentaenoic acid for mechanism studies. Our results demonstrated that endogenous and exogenous n-3 PUFA enrichment ameliorates ethanol-stimulated adipose lipolysis by increasing PDE3B activity and reducing cAMP accumulation in adipocyte, which was associated with activation of GPR120 and regulation of Ca2+/CaMKKß/AMPK signaling, resultantly blocking fatty acid trafficking from adipose tissue to the liver, which contributing to ameliorating ethanol-induced adipose dysfunction and liver injury. Our findings identify that endogenous and exogenous n-3 PUFA enrichment ameliorated alcoholic liver injury by activation of GPR120 to suppress ethanol-stimulated adipose lipolysis, which provides the new insight to the hepatoprotective effect of n-3 PUFA against alcoholic liver disease.


Asunto(s)
Adiposidad/efectos de los fármacos , Etanol/farmacología , Ácidos Grasos Omega-3/farmacología , Hepatopatías Alcohólicas/prevención & control , Sustancias Protectoras/farmacología , Células 3T3-L1 , Quinasas de la Proteína-Quinasa Activada por el AMP , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Hígado Graso Alcohólico/prevención & control , Femenino , Lipólisis/efectos de los fármacos , Hepatopatías Alcohólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
11.
IUBMB Life ; 69(7): 540-552, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28500689

RESUMEN

Alcoholic liver disease (ALD), characterized by excessive deposition of lipids in hepatocytes, causes heavy health burden personally and socially. Mechanistically, hedgehog signaling was activated during the development of ALD, and exerted compelling role in regulating lipometabolism. The current promising intervention strategy is inhibition of lipid accumulation and apoptosis in hepatocytes. Magnesium isoglycyrrhizinate (MgIG) has been widely used in various liver diseases for its good hepatoprotective activities. However, the role of MgIG in ALD has not been elucidated. Therefore, this study was aimed to explore the role of MgIG and further identify the potential mechanisms. We found for the first time that MgIG reduced lipid accumulation, including triglyceride, and total cholesterol, probably via inducing peroxisome proliferator-activated receptor-alpha and inhibiting sterol regulatory element-binding protein-1c. Further, MgIG alleviated ethanol-induced oxidative stress, evidenced by reduced abundance of reactive oxygen species and increased levels of glutathione, superoxide dismutase, and mitochondrial transmembrane potential. Besides, MgIG protected hepatocytes from ethanol-induced apoptosis. In addition, MgIG dose-dependently suppressed hedgehog signaling. Of note was that disruption of hedgehog signaling could mimic the effects of MgIG, whereas activation of hedgehog signaling abrogated the effects of MgIG. These findings suggested that MgIG prevented ethanol-induced hepatocyte steatosis and apoptosis via a hedgehog signaling inhibition-dependent mechanism. © 2017 IUBMB Life, 69(7):540-552, 2017.


Asunto(s)
Etanol/toxicidad , Hígado Graso Alcohólico/prevención & control , Proteínas Hedgehog/metabolismo , Hepatocitos/efectos de los fármacos , Saponinas/farmacología , Triterpenos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Hígado Graso Alcohólico/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína con Dedos de Zinc GLI1/metabolismo
12.
Toxicol Appl Pharmacol ; 315: 23-34, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27939985

RESUMEN

Alcoholic liver disease (ALD) is a common etiology of liver diseases, characterized by hepatic steatosis. We previously identified farnesoid X receptor (FXR) as a potential therapeutic target for ALD. Dihydroartemisinin (DHA) has been recently identified to possess potent pharmacological activities on liver diseases. This study was aimed to explore the impact of DHA on ALD and further elaborate the underlying mechanisms. Gain- or loss-of-function analyses of FXR were applied in both in vivo and in vitro studies. Results demonstrated that DHA rescued FXR expression and activity in alcoholic rat livers. DHA also reduced serodiagnostic markers of liver injury, including aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, and lactate dehydrogenase. DHA improved alcohol-induced liver histological lesions, expression of inflammation genes, and inflammatory cell infiltration. In addition, DHA not only attenuated hyperlipidemia but also reduced hepatic steatosis through regulating lipogenesis and lipolysis genes. In vitro experiments further consolidated the concept that DHA ameliorated ethanol-caused hepatocyte injury and steatosis. Noteworthily, DHA effects were reinforced by FXR agonist obeticholic acid or FXR expression plasmids but abrogated by FXR antagonist Z-guggulsterone or FXR siRNA. In summary, DHA significantly improved alcoholic liver injury by inhibiting hepatic steatosis, which was dependent on its activation of FXR in hepatocytes.


Asunto(s)
Artemisininas/farmacología , Hígado Graso Alcohólico/prevención & control , Hepatopatías Alcohólicas/prevención & control , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Masculino , Ratas , Ratas Sprague-Dawley
13.
Dig Dis Sci ; 62(8): 2021-2034, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28424943

RESUMEN

BACKGROUND AND AIMS: Bacterially derived factors from the gut play a major role in the activation of inflammatory pathways in the liver and in the pathogenesis of alcoholic liver disease. The intestinal brush-border enzyme intestinal alkaline phosphatase (IAP) detoxifies a variety of bacterial pro-inflammatory factors and also functions to preserve gut barrier function. The aim of this study was to investigate whether oral IAP supplementation could protect against alcohol-induced liver disease. METHODS: Mice underwent acute binge or chronic ethanol exposure to induce alcoholic liver injury and steatosis ± IAP supplementation. Liver tissue was assessed for biochemical, inflammatory, and histopathological changes. An ex vivo co-culture system was used to examine the effects of alcohol and IAP treatment in regard to the activation of hepatic stellate cells and their role in the development of alcoholic liver disease. RESULTS: Pretreatment with IAP resulted in significantly lower serum alanine aminotransferase compared to the ethanol alone group in the acute binge model. IAP treatment attenuated the development of alcohol-induced fatty liver, lowered hepatic pro-inflammatory cytokine and serum LPS levels, and prevented alcohol-induced gut barrier dysfunction. Finally, IAP ameliorated the activation of hepatic stellate cells and prevented their lipogenic effect on hepatocytes. CONCLUSIONS: IAP treatment protected mice from alcohol-induced hepatotoxicity and steatosis. Oral IAP supplementation could represent a novel therapy to prevent alcoholic-related liver disease in humans.


Asunto(s)
Fosfatasa Alcalina/administración & dosificación , Suplementos Dietéticos , Hígado Graso Alcohólico/prevención & control , Alanina Transaminasa/sangre , Animales , Técnicas de Cocultivo , Citocinas/análisis , Citocinas/sangre , Etanol , Hígado Graso Alcohólico/sangre , Hígado Graso Alcohólico/enzimología , Femenino , Células Estrelladas Hepáticas/enzimología , Hepatocitos/enzimología , Intestinos/enzimología , Lipogénesis , Lipopolisacáridos/sangre , Hígado/química , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Activador de Tejido Plasminógeno , Triglicéridos/análisis
14.
Gastroenterology ; 149(4): 1030-41.e6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26099526

RESUMEN

BACKGROUND & AIMS: Alcoholic steatohepatitis (ASH) is the progressive form of alcoholic liver disease and may lead to cirrhosis and hepatocellular carcinoma. We studied mouse models and human tissues to identify molecules associated with ASH progression and focused on the mouse fat-specific protein 27 (FSP-27)/human cell death-inducing DFF45-like effector C (CIDEC) protein, which is expressed in white adipose tissues and promotes formation of fat droplets. METHODS: C57BL/6N mice or mice with hepatocyte-specific disruption of Fsp27 (Fsp27(Hep-/-) mice) were fed the Lieber-Decarli ethanol liquid diet (5% ethanol) for 10 days to 12 weeks, followed by 1 or multiple binges of ethanol (5 or 6 g/kg) during the chronic feeding. Some mice were given an inhibitor (GW9662) of peroxisome proliferator-activated receptor γ (PPARG). Adenoviral vectors were used to express transgenes or small hairpin (sh) RNAs in cultured hepatocytes and in mice. Liver tissue samples were collected from ethanol-fed mice or from 31 patients with alcoholic hepatitis (AH) with biopsy-proved ASH and analyzed histologically and immunohistochemically and by transcriptome, immunoblotting, and real-time PCR analyses. RESULTS: Chronic-plus-binge ethanol feeding of mice, which mimics the drinking pattern of patients with AH, produced severe ASH and mild fibrosis. Microarray analyses revealed similar alterations in expression of many hepatic genes in ethanol-fed mice and humans with ASH, including up-regulation of mouse Fsp27 (also called Cidec) and human CIDEC. Fsp27(Hep-/-) mice and mice given injections of adenovirus-Fsp27shRNA had markedly reduced ASH following chronic-plus-binge ethanol feeding. Inhibition of PPARG and cyclic AMP-responsive element binding protein H (CREBH) prevented the increases in Fsp27α and FSP27ß mRNAs, respectively, and reduced liver injury in this chronic-plus-binge ethanol feeding model. Overexpression of FSP27 and ethanol exposure had synergistic effects in inducing production of mitochondrial reactive oxygen species and damage to hepatocytes in mice. Hepatic CIDEC mRNA expression was increased in patients with AH and correlated with the degree of hepatic steatosis and disease severity including mortality. CONCLUSIONS: In mice, chronic-plus-binge ethanol feeding induces ASH that mimics some histological and molecular features observed in patients with AH. Hepatic expression of FSP27/CIDEC is highly up-regulated in mice following chronic-plus-binge ethanol feeding and in patients with AH; this up-regulation contributes to alcohol-induced liver damage.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Proteínas/metabolismo , Adulto , Animales , Proteínas Reguladoras de la Apoptosis , Consumo Excesivo de Bebidas Alcohólicas , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Femenino , Perfilación de la Expresión Génica/métodos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mitocondrias Hepáticas/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Proteínas/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
15.
Alcohol Clin Exp Res ; 40(11): 2312-2319, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27581622

RESUMEN

BACKGROUND: Alcohol-induced reduction in the hepatocellular S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) ratio impairs the activities of many SAM-dependent methyltransferases. These impairments ultimately lead to the generation of several hallmark features of alcoholic liver injury including steatosis. Guanidinoacetate methyltransferase (GAMT) is an important enzyme that catalyzes the final reaction in the creatine biosynthetic process. The liver is a major site for creatine synthesis which places a substantial methylation burden on this organ as GAMT-mediated reactions consume as much as 40% of all the SAM-derived methyl groups. We hypothesized that dietary creatine supplementation could potentially spare SAM, preserve the hepatocellular SAM:SAH ratio, and thereby prevent the development of alcoholic steatosis and other consequences of impaired methylation reactions. METHODS: For these studies, male Wistar rats were pair-fed the Lieber-DeCarli control or ethanol (EtOH) diet with or without 1% creatine supplementation. At the end of 4 to 5 weeks of feeding, relevant biochemical and histological analyses were performed. RESULTS: We observed that creatine supplementation neither prevented alcoholic steatosis nor attenuated the alcohol-induced impairments in proteasome activity. The lower hepatocellular SAM:SAH ratio seen in the EtOH-fed rats was also not normalized or SAM levels spared when these rats were fed the creatine-supplemented EtOH diet. However, a >10-fold increased level of creatine was observed in the liver, serum, and hearts of rats fed the creatine-supplemented diets. CONCLUSIONS: Overall, dietary creatine supplementation did not prevent alcoholic liver injury despite its known efficacy in preventing high-fat-diet-induced steatosis. Betaine, a promethylating agent that maintains the hepatocellular SAM:SAH, still remains our best option for treating alcoholic steatosis.


Asunto(s)
Creatina/uso terapéutico , Hígado Graso Alcohólico/prevención & control , Amidinotransferasas/metabolismo , Animales , Suplementos Dietéticos , Guanidinoacetato N-Metiltransferasa/metabolismo , Riñón/enzimología , Hígado/enzimología , Masculino , Miocardio/metabolismo , Ratas Wistar , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo
16.
J Pathol ; 235(5): 710-20, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25557254

RESUMEN

Chronic ethanol consumption causes hepatic steatosis and inflammation, which are associated with liver hypoxia. Monocyte chemoattractant protein-1 (MCP-1) is a hypoxia response factor that determines recruitment and activation of monocytes to the site of tissue injury. The level of MCP-1 is elevated in the serum and liver of patients with alcoholic liver disease (ALD); however, the molecular details regarding the regulation of MCP-1 expression are not yet understood completely. Here, we show the role of liver X receptor α (LXRα) in the regulation of MCP-1 expression during the development of ethanol-induced fatty liver injury, using an antagonist, 22-S-hydroxycholesterol (22-S-HC). First, administration of 22-S-HC attenuated the signs of liver injury with decreased levels of MCP-1 and its receptor CCR2 in ethanol-fed mice. Second, hypoxic conditions or treatment with the LXRα agonist GW3965 significantly induced the expression of MCP-1, which was completely blocked by treatment with 22-S-HC or infection by shLXRα lentivirus in the primary hepatocytes. Third, over-expression of LXRα or GW3965 treatment increased MCP-1 promoter activity by increasing the binding of hypoxia-inducible factor-1α to the hypoxia response elements, together with LXRα. Finally, treatment with recombinant MCP-1 increased the level of expression of LXRα and LXRα-dependent lipid droplet accumulation in both hepatocytes and Kupffer cells. These data show that LXRα and its ligand-induced up-regulation of MCP-1 and MCP-1-induced LXRα-dependent lipogenesis play a key role in the autocrine and paracrine activation of MCP-1 in the pathogenesis of alcoholic fatty liver disease, and that this activation may provide a promising new target for ALD therapy.


Asunto(s)
Comunicación Autocrina/efectos de los fármacos , Quimiocina CCL2/metabolismo , Hígado Graso Alcohólico/prevención & control , Hidroxicolesteroles/farmacología , Hígado/efectos de los fármacos , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Comunicación Paracrina/efectos de los fármacos , Animales , Sitios de Unión , Hipoxia de la Célula , Células Cultivadas , Quimiocina CCL2/genética , Citoprotección , Modelos Animales de Enfermedad , Etanol , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Lipogénesis/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Receptores X del Hígado , Masculino , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal/efectos de los fármacos , Transfección , Regulación hacia Arriba
17.
BMC Complement Altern Med ; 16: 19, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26801973

RESUMEN

BACKGROUND: Our previous study suggested that licorice has anti-inflammatory activity in lipopolysaccharide-stimulated microglial cells and anti-oxidative activity in tert-butyl hydroperoxide-induced oxidative liver damage. In this study, we evaluated the effect of licorice on chronic alcohol-induced fatty liver injury mediated by inflammation and oxidative stress. METHODS: Raw licorice was extracted, and quantitative and qualitative analysis of its components was performed by using LC-MS/MS. Mice were fed a liquid alcohol diet with or without licorice for 4 weeks. RESULTS: We have standardized 70% fermented ethanol extracted licorice and confirmed by LC-MS/MS as glycyrrhizic acid (GA), 15.77 ± 0.34 µg/mg; liquiritin (LQ), 14.55 ± 0.42 µg/mg; and liquiritigenin (LG), 1.34 ± 0.02 µg/mg, respectively. Alcohol consumption increased serum alanine aminotransferase and aspartate aminotransferase activities and the levels of triglycerides and tumor necrosis factor (TNF)-α. Lipid accumulation in the liver was also markedly induced, whereas the glutathione level was reduced. All these alcohol-induced changes were effectively inhibited by licorice treatment. In particular, the hepatic glutathione level was restored and alcohol-induced TNF-α production was significantly inhibited by licorice. CONCLUSION: Taken together, our data suggests that protective effect of licorice against alcohol-induced liver injury may be attributed to its anti-inflammatory activity and enhancement of antioxidant defense.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Hígado Graso Alcohólico/prevención & control , Glycyrrhiza uralensis , Hígado/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Animales , Hígado Graso Alcohólico/sangre , Glycyrrhiza , Glycyrrhiza uralensis/química , Masculino , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/química , Raíces de Plantas/química , terc-Butilhidroperóxido
18.
J Nutr ; 145(12): 2690-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26468492

RESUMEN

BACKGROUND: Zinc deficiency has been well documented in alcoholic liver disease. OBJECTIVE: This study was undertaken to determine whether dietary zinc supplementation provides beneficial effects in treating alcohol-induced gut leakiness and endotoxemia. METHODS: Male Sprague Dawley rats were divided into 3 groups and pair-fed (PF) Lieber-DeCarli liquid diet for 8 wk: 1) control (PF); 2) alcohol-fed (AF; 5.00-5.42% wt:vol ethanol); and 3) AF with zinc supplementation (AF/Zn) at 220 ppm zinc sulfate heptahydrate. The PF and AF/Zn groups were pair-fed with the AF group. Hepatic inflammation and endotoxin signaling were determined by immunofluorescence and quantitative polymerase chain reaction (qPCR). Alterations in intestinal tight junctions and aldehyde dehydrogenases were assessed by qPCR and Western blot analysis. RESULTS: The AF rats had greater macrophage activation and cytokine production (P < 0.05) in the liver compared with the PF rats, whereas the AF/Zn rats showed no significant differences (P > 0.05). Plasma endotoxin concentrations of the AF rats were 136% greater than those of the PF rats, whereas the AF/Zn rats did not differ from the PF rats. Ileal permeability was 255% greater in the AF rats and 19% greater in the AF/Zn rats than in the PF rats. The AF group had reduced intestinal claudin-1, occludin, and zona occludens-1 (ZO-1) expression, and the AF/Zn group had upregulated claudin-1 and ZO-1 expression (P < 0.05) compared with the PF group. The intestinal epithelial expression and activity of aldehyde dehydrogenases were elevated (P < 0.05) in the AF/Zn rats compared with those of the AF rats. Furthermore, the ileal expression and function of hepatocyte nuclear factor 4α, which was impaired in the AF group, was significantly elevated in the AF/Zn group compared with the PF group. CONCLUSIONS: The results demonstrate that attenuating hepatic endotoxin signaling by preserving the intestinal barrier contributes to the protective effect of zinc on alcohol-induced steatohepatitis in rats.


Asunto(s)
Suplementos Dietéticos , Endotoxemia/prevención & control , Hígado Graso Alcohólico/prevención & control , Enfermedades Intestinales/prevención & control , Zinc/administración & dosificación , Aldehído Deshidrogenasa/metabolismo , Animales , Claudina-1/análisis , Citocinas/biosíntesis , Endotoxinas/análisis , Etanol/efectos adversos , Hígado Graso Alcohólico/fisiopatología , Enfermedades Intestinales/inducido químicamente , Mucosa Intestinal/metabolismo , Intestinos/química , Intestinos/enzimología , Hígado/patología , Hígado/fisiopatología , Activación de Macrófagos , Masculino , Ocludina/análisis , Permeabilidad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas de Uniones Estrechas/análisis , Zinc/deficiencia , Proteína de la Zonula Occludens-1/análisis
19.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G58-65, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24833709

RESUMEN

Sestrins (Sesns) are a family of stress-sensitive genes that have been suggested to regulate lipid metabolism. Chronic ethanol feeding is known to cause lipid accumulation in hepatocytes. This study was designed to investigate the role of Sesn3 in the pathogenesis of alcohol-induced hepatic steatosis. We demonstrated that ethanol inhibited the expression of Sesn3 in VL-17A cells. Overexpression of Sesn3 ameliorated triglyceride accumulation; downregulation using short hairpin RNA significantly deteriorated triglyceride accumulation in these cells. The expression of Sesn3 was also reduced in mice fed with ethanol for 4 wk. Overexpression of Sesn3 prevented hepatic steatosis, whereas knockdown of Sesn3 worsened hepatic steatosis in ethanol-fed mice. Overexpression of Sesn3 significantly reduced the expression of genes encoding for lipid synthesis through AMPK pathway. Overexpression of Sesn3 augmented the effect of ethanol on phospho-p70 S6 kinase. The levels of hepatic light chain 3, a marker for autophagy, expression were significantly decreased in ethanol-fed mice after Sesn3 gene was knocked down. Our findings suggest that inhibitory effect of ethanol on Sesn3 may play an important role in the development of ethanol-induced fatty liver.


Asunto(s)
Etanol , Hígado Graso Alcohólico/metabolismo , Proteínas de Choque Térmico/metabolismo , Hígado/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Proteínas de Choque Térmico/genética , Células Hep G2 , Humanos , Lipogénesis , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Interferencia de ARN , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factores de Tiempo , Transfección , Triglicéridos/metabolismo
20.
J Nutr ; 144(7): 1009-15, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24828027

RESUMEN

Ethanol consumption can lead to hepatic steatosis that contributes to late-stage liver diseases such as cirrhosis and hepatocellular carcinoma. In this study, we investigated the potential protective effect of a flavonoid, luteolin, on ethanol-induced fatty liver development and liver injury. Six-wk-old male C57BL/6 mice were divided into 3 groups: a control group; a group exposed to alcohol by using a chronic and binge ethanol feeding protocol (EtOH); and a group that was administered daily 50 mg/kg of luteolin in addition to ethanol exposure (EtOH + Lut). A chronic and binge ethanol feeding protocol was used, including chronic ethanol consumption (1%, 2%, and 4% for 3 d, and 5% for 9 d) and a binge (30% ethanol) on the last day. Compared with the control group, the EtOH group had a significant elevation in serum concentrations of alanine aminotransferase (ALT) (561%), triglyceride (TG) (47%), and LDL cholesterol (95%), together with lipid accumulation in the liver. Compared with the EtOH group, the EtOH + Lut group had significant reductions in serum concentrations of ALT (43%), TG (22%), LDL cholesterol (52%), and lipid accumulation in the liver. Ethanol elevated liver expression of lipogenic genes including sterol regulatory element-binding protein 1c (Srebp1c) (560%), fatty acid synthase (Fasn) (190%), acetyl-CoA carboxylase (Acc) (48%), and stearoyl-CoA desaturase 1 (Scd1) (286%). Luteolin reduced ethanol-induced expression of these genes in the liver: Srebp1c (79%), Fasn (80%), Acc (60%), and Scd1 (89%). In cultured hepatocytes, luteolin prevented alcohol-induced lipid accumulation and increase in the expression of lipogenic genes. The transcriptional activity of the master regulator of lipid synthesis, sterol regulatory element-binding protein (SREBP), was enhanced by ethanol treatment (160%) and reduced by luteolin administration (67%). In addition, ethanol-induced reduction of AMP-activated protein kinase and SREBP-1c phosphorylation was abrogated by luteolin. Collectively, our study indicates that luteolin is effective in ameliorating ethanol-induced hepatic steatosis and injury.


Asunto(s)
Alcoholismo/fisiopatología , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Hepatopatías Alcohólicas/prevención & control , Hígado/metabolismo , Luteolina/uso terapéutico , Animales , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Línea Celular , Etanol/antagonistas & inhibidores , Etanol/toxicidad , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Regulación de la Expresión Génica , Hepatitis Alcohólica/etiología , Hepatitis Alcohólica/metabolismo , Hepatitis Alcohólica/patología , Hepatitis Alcohólica/prevención & control , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Lipogénesis , Hígado/patología , Hígado/fisiopatología , Hepatopatías Alcohólicas/etiología , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Luteolina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda