Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Nutrients ; 13(5)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064981

RESUMEN

Alcoholic liver disease (ALD) is one type of liver disease, causing a global healthcare problem and mortality. The liver undergoes tissue damage by chronic alcohol consumption because it is the main site for metabolism of ethanol. Chronic alcohol exposure progresses from alcoholic fatty liver (AFL) to alcoholic steatohepatitis (ASH), which further lead to fibrosis, cirrhosis, and even hepatocellular cancer. Therapeutic interventions to combat ALD are very limited such as use of corticosteroids. However, these therapeutic drugs are not effective for long-term usage. Therefore, additional effective and safe therapies to cope with ALD are urgently needed. Previous studies confirmed that edible food plants and their bioactive compounds exert a protective effect against ALD. In this review article, we summarized the hepatoprotective potential of edible food plants and their bioactive compounds. The underlying mechanism for the prevention of ALD by edible food plants was as follows: anti-oxidation, anti-inflammation, lipid regulation, inhibition of apoptosis, gut microbiota composition modulation, and anti-fibrosis.


Asunto(s)
Hepatopatías Alcohólicas/terapia , Plantas Comestibles/química , Polifenoles/uso terapéutico , Sustancias Protectoras/uso terapéutico , Consumo de Bebidas Alcohólicas , Animales , Etanol/efectos adversos , Etanol/metabolismo , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/terapia , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hepatopatías Alcohólicas/metabolismo , Neoplasias Hepáticas , Fitoquímicos/química , Fitoquímicos/uso terapéutico , Extractos Vegetales/uso terapéutico , Sustancias Protectoras/química
2.
Cell Host Microbe ; 28(2): 233-244, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32791115

RESUMEN

The liver communicates with the intestine via the portal vein, biliary system, and mediators in the circulation. Microbes in the intestine maintain liver homeostasis but can also serve as a source of pathogens and molecules that contribute to fatty liver diseases. We review changes in the gut microbiota that can promote development or progression of alcohol-associated and non-alcoholic fatty liver disease-the most common chronic liver diseases in Western countries. We discuss how microbes and their products contribute to liver disease pathogenesis, putative microbial biomarkers of disease, and potential treatment approaches based on manipulation of the gut microbiota. Increasing our understanding of interactions between the intestinal microbiome and liver might help us identify patients with specific disease subtypes and select specific microbiota-based therapies.


Asunto(s)
Hígado Graso Alcohólico/microbiología , Hígado Graso Alcohólico/patología , Microbioma Gastrointestinal/fisiología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Disbiosis/microbiología , Hígado Graso Alcohólico/terapia , Interacciones Microbiota-Huesped/fisiología , Humanos , Intestinos/microbiología , Intestinos/patología , Hígado/patología , Ratones , Enfermedad del Hígado Graso no Alcohólico/terapia , Probióticos/uso terapéutico
4.
Chin J Nat Med ; 18(3): 169-177, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32245586

RESUMEN

The objective of this study was to verify the protective effect of Bifidobacterium longum (BL) and the synergistical effect of Selenium and BL on alcohol plus high fat diet (HFD) induced hepatic injury in mice. We also want to explore the mechanism of Selenium-enriched Bifidobacterium longum (SeBL). C57BL/6 mice were treated with alcohol plus HFD with or without different dosage of BL or SeBL for 4 weeks. Serum levels of ALT, AST, TC, TG, LDL-C, HDL-C, FFAs, TNF-α, IL-6 and IL-1ß, hepatic MDA level, SOD activity, the mRNA levels of AMPK, PPAR-α and SREBP1 were invested. SeBL inhibited lipid accumulation in hepatocytes; reduced serum AST and ALT levels; improved dyslipidemia; decreased serum FFAs, TC, TG and LDL-C levels. SeBL also inhibited alcohol plus HFD-induced hepatocyte oxidative stress through decrease in hepatic MDA levels and increase in SOD activity. SeBL also regulated lipid metabolism related genes such as AMPK, PPAR-α and SREBP1. Although BL had similar effect as SeBL, SeBL is more effective than BL. SeBL protected mice from alcohol plus HFD-induced hepatic injury in mice because of its inhibitory effect on hepatocellular oxidative stress, lipogenesis and inflammation. Selenium enhanced the protective effect of BL.


Asunto(s)
Bifidobacterium longum , Hígado Graso Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/terapia , Estrés Oxidativo , Probióticos/uso terapéutico , Selenio/química , Animales , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Etanol/efectos adversos , Metabolismo de los Lípidos , Hígado/efectos de los fármacos , Hígado/fisiopatología , Pruebas de Función Hepática , Masculino , Ratones Endogámicos C57BL
6.
Cell Host Microbe ; 27(1): 25-40.e6, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31866426

RESUMEN

Although a link between the gut microbiota and alcohol-related liver diseases (ALDs) has previously been suggested, the causative effects of specific taxa and their functions have not been fully investigated to date. Here, we analyze the gut microbiota of 410 fecal samples from 212 Korean twins by using the Alcohol Use Disorders Identification Test (AUDIT) scales to adjust for host genetics. This analysis revealed a strong association between low AUDIT scores and the abundance of the butyrate-producing genus Roseburia. When Roseburia spp. are administered to ALD murine models, both hepatic steatosis and inflammation significantly improve regardless of bacterial viability. Specifically, the flagellin of R. intestinalis, possibly through Toll-like receptor 5 (TLR5) recognition, recovers gut barrier integrity through upregulation of the tight junction protein Occludin and helps to restore the gut microbiota through elevated expression of IL-22 and REG3γ. Our study demonstrates that Roseburia spp. improve the gut ecosystem and prevent leaky gut, leading to ameliorated ALDs.


Asunto(s)
Clostridiales/metabolismo , Hígado Graso Alcohólico/terapia , Microbioma Gastrointestinal , Adulto , Consumo de Bebidas Alcohólicas/efectos adversos , Trastornos Relacionados con Alcohol/patología , Animales , Clostridiales/aislamiento & purificación , Disbiosis/microbiología , Hígado Graso Alcohólico/metabolismo , Heces/microbiología , Femenino , Flagelina/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ocludina/metabolismo
7.
Biomaterials ; 227: 119570, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31670032

RESUMEN

Excessive release of interleukin-1ß (IL-1ß) is well-known to provoke cascades of inflammatory responses thus contributing to the pathogenesis of alcohol-induced steatohepatitis (ASH), but the cellular mechanism that regulates IL-1ß release during ASH remains unclear. Herein, we identified that gasdermin D (GSDMD) membrane pore is critical in mediating IL-1ß hypersecretion from chronic ethanol or acetaldehyde-stimulated macrophages. Deletion of GSDMD reduced IL-1ß release and ameliorated alcoholic steatohepatitis in vivo. These findings uncovered a novel mechanism regarding the IL-1ß release in ASH, and also indicated the therapeutic potential of IL-1ß blockade. Interleukin-1 receptor antagonist (IL-1Ra) is protective to ASH by blocking IL-1ß, but it has a short biological half-life (4-6 h) and lower liver concentrations. Thus, we constructed a therapeutic plasmid pVAX1-IL-1Ra-ApoAI (pVAX1-IA) encoding IL-1Ra anchored to the liver-targeting protein apolipoprotein A-I (ApoAI), and developed hepatocyte-specific nanobiologics (Glipo-pVAX1-IA) by galactose functionalization for local and prolonged expression of IL-1Ra in liver. Data presented here showed that Glipo-pVAX1-IA facilitated efficient uptake of gene cargos by hepatocytes. The biodistribution studies confirmed a predominant hepatocytes internalization, but a minimal kupffer cells uptake of Glipo-pVAX1-IA following intravenous injection. The locally secreted IL-1Ra attenuated alcohol-induced steatohepatisis and infiltration of inflammatory cells. Together, our results unraveled the critical role of GSDMD membrane pore in IL-1ß hypersecretion and highlighted the hepatocyte-specific Glipo-pVAX1-IA nanobiologics as a promising therapeutic strategy for ASH.


Asunto(s)
Hígado Graso Alcohólico , Interleucina-1beta , Animales , Hígado Graso Alcohólico/terapia , Hepatocitos/metabolismo , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Macrófagos del Hígado/metabolismo , Ratones , Proteínas de Unión a Fosfato/metabolismo , Distribución Tisular
10.
PLoS One ; 14(2): e0212523, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30794635

RESUMEN

Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in mammalian nicotinamide adenine dinucleotide (NAD)+ biosynthesis. Through its NAD+-biosynthetic activity, NAMPT influences the activity of NAD+-dependent enzymes, such as sirtuins. NAMPT is able to modulate processes involved in the pathogenesis of non-alcohol induced fatty liver disease (NAFLD), but the roles NAMPT plays in development of alcoholic liver disease (ALD) still remain unknown. Here, we show that ethanol treatment suppresses the expression of Nampt in hepatocytes. Consistently, chronic ethanol administration also reduces Nampt expression in the mouse liver. We next demonstrate that hepatocytes infected with Ad-NAMPT adenovirus exhibit significantly elevated intracellular NAD+ levels and decreased ethanol-induced triglyceride (TG) accumulation. Similarly, adenovirus-mediated overexpression of NAMPT in mice ameliorates ethanol induced hepatic steatosis. Moreover, we demonstrate that SIRT1 is required to mediate the effects of NAMPT on reduction of hepatic TG accumulation and serum ALT, AST levels in ethanol-fed mice. Our results provide important insights in targeting NAMPT for treating alcoholic fatty liver disease.


Asunto(s)
Citocinas/metabolismo , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/terapia , Nicotinamida Fosforribosiltransferasa/metabolismo , Adenoviridae/genética , Animales , Células Cultivadas , Citocinas/genética , Citocinas/uso terapéutico , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Etanol/toxicidad , Hígado Graso Alcohólico/genética , Técnicas de Silenciamiento del Gen , Terapia Genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/uso terapéutico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuinas/deficiencia , Sirtuinas/genética , Sirtuinas/metabolismo , Triglicéridos/metabolismo , Regulación hacia Arriba
12.
J Hepatol ; 68(5): 1025-1032, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29343427

RESUMEN

BACKGROUND & AIMS: Controlled attenuation parameter (CAP) is a novel non-invasive measure of hepatic steatosis, but it has not been evaluated in alcoholic liver disease. Therefore, we aimed to validate CAP for the assessment of biopsy-verified alcoholic steatosis and to study the effect of alcohol detoxification on CAP. METHODS: This was a cross-sectional biopsy-controlled diagnostic study in four European liver centres. Consecutive alcohol-overusing patients underwent concomitant CAP, regular ultrasound, and liver biopsy. In addition, we measured CAP before and after admission for detoxification in a separate single-centre cohort. RESULTS: A total of 562 patients were included in the study: 269 patients in the diagnostic cohort with steatosis scores S0, S1, S2, and S3 = 77 (28%), 94 (35%), 64 (24%), and 34 (13%), respectively. CAP diagnosed any steatosis and moderate steatosis with fair accuracy (area under the receiver operating characteristic curve [AUC] ≥S1 = 0.77; 0.71-0.83 and AUC ≥S2 = 0.78; 0.72-0.83), and severe steatosis with good accuracy (AUC S3 = 0.82; 0.75-0.88). CAP was superior to bright liver echo pattern by regular ultrasound. CAP above 290 dB/m ruled in any steatosis with 88% specificity and 92% positive predictive value, while CAP below 220 dB/m ruled out steatosis with 90% sensitivity, but 62% negative predictive value. In the 293 patients who were admitted 6.3 days (interquartile range 4-6) for detoxification, CAP decreased by 32 ±â€¯47 dB/m (p <0.001). Body mass index predicted higher CAP in both cohorts, irrespective of drinking pattern. Obese patients with body mass index ≥30 kg/m2 had a significantly higher CAP, which did not decrease significantly during detoxification. CONCLUSIONS: CAP has a good diagnostic accuracy for diagnosing severe alcoholic liver steatosis and can be used to rule in any steatosis. In non-obese but not in obese, patients, CAP rapidly declines after alcohol withdrawal. LAY SUMMARY: CAP is a new ultrasound-based technique for measuring fat content in the liver, but has never been tested for fatty liver caused by alcohol. Herein, we examined 562 patients in a multicentre setting. We show that CAP highly correlates with liver fat, and patients with a CAP value above 290 dB/m were highly likely to have more than 5% fat in their livers, determined by liver biopsy. CAP was also better than regular ultrasound for determining the severity of alcoholic fatty-liver disease. Finally, we show that three in four (non-obese) patients rapidly decrease in CAP after short-term alcohol withdrawal. In contrast, obese alcohol-overusing patients were more likely to have higher CAP values than lean patients, irrespective of drinking.


Asunto(s)
Abstinencia de Alcohol , Hígado Graso Alcohólico/diagnóstico por imagen , Hígado Graso Alcohólico/terapia , Ultrasonografía/métodos , Adulto , Alcoholismo/diagnóstico por imagen , Biopsia , Estudios de Cohortes , Estudios Transversales , Diagnóstico por Imagen de Elasticidad , Femenino , Humanos , Hígado/diagnóstico por imagen , Masculino , Síndrome Metabólico/diagnóstico por imagen , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Factores de Riesgo
13.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878132

RESUMEN

Alcoholic steatohepatitis (ASH) and nonalcoholic steatohepatitis (NASH) are among the most frequent causes of chronic liver disease in the United States. Although the two entities are triggered by different etiologies - chronic alcohol consumption (ASH) and obesity-associated lipotoxicity (NASH) - they share overlapping histological and clinical features owing to common pathogenic mechanisms. These pathogenic processes include altered hepatocyte lipid metabolism, organelle dysfunction (i.e., ER stress), hepatocyte apoptosis, innate immune system activation, and hepatic stellate cell activation. Nonetheless, there are several disease-specific molecular signaling pathways, such as differential pathway activation downstream of TLR4 (MyD88-dependence in NASH versus MyD88-independence in ASH), inflammasome activation and IL-1ß signaling in ASH, insulin resistance and lipotoxicity in NASH, and dysregulation of different microRNAs, which clearly highlight that ASH and NASH are two distinct biological entities. Both pathogenic similarities and differences have therapeutic implications. In this Review, we discuss these pathogenic mechanisms and their therapeutic implications for each disease, focusing on both shared and distinct targets.


Asunto(s)
Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/terapia , Hígado Graso Alcohólico/metabolismo , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo
15.
Am J Physiol Gastrointest Liver Physiol ; 311(6): G1018-G1036, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27686615

RESUMEN

Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) represent a major health burden in industrialized countries. Although alcohol abuse and nutrition play a central role in disease pathogenesis, preclinical models support a contribution of the gut microbiota to ALD and NAFLD. This review describes changes in the intestinal microbiota compositions related to ALD and NAFLD. Findings from in vitro, animal, and human studies are used to explain how intestinal pathology contributes to disease progression. This review summarizes the effects of untargeted microbiome modifications using antibiotics and probiotics on liver disease in animals and humans. While both affect humoral inflammation, regression of advanced liver disease or mortality has not been demonstrated. This review further describes products secreted by Lactobacillus- and microbiota-derived metabolites, such as fatty acids and antioxidants, that could be used for precision medicine in the treatment of liver disease. A better understanding of host-microbial interactions is allowing discovery of novel therapeutic targets in the gut microbiota, enabling new treatment options that restore the intestinal ecosystem precisely and influence liver disease. The modulation options of the gut microbiota and precision medicine employing the gut microbiota presented in this review have excellent prospects to improve treatment of liver disease.


Asunto(s)
Hígado Graso Alcohólico/terapia , Microbioma Gastrointestinal , Enfermedad del Hígado Graso no Alcohólico/terapia , Medicina de Precisión/métodos , Animales , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/microbiología , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología , Probióticos/uso terapéutico
16.
Sci Rep ; 6: 29352, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27404390

RESUMEN

Alcohol consumption is one of the major causes of hepatic steatosis, fibrosis, cirrhosis, and superimposed hepatocellular carcinoma. Ethanol metabolism alters the NAD(+)/NADH ratio, thereby suppressing the activity of sirtuin family proteins, which may affect lipid metabolism in liver cells. However, it is not clear how long-term ingestion of ethanol eventually causes lipid accumulation in liver. Here, we demonstrate that chronic ethanol ingestion activates peroxisome proliferator-activated receptor γ (PPARγ) and its target gene, monoacylglycerol O-acyltransferase 1 (MGAT1). During ethanol metabolism, a low NAD(+)/NADH ratio repressed NAD-dependent deacetylase sirtuin 1 (SIRT1) activity, concomitantly resulting in increased acetylated PPARγ with high transcriptional activity. Accordingly, SIRT1 transgenic mice exhibited a low level of acetylated PPARγ and were protected from hepatic steatosis driven by alcohol or PPARγ2 overexpression, suggesting that ethanol metabolism causes lipid accumulation through activation of PPARγ through acetylation. Among the genes induced by PPARγ upon alcohol consumption, MGAT1 has been shown to be involved in triglyceride synthesis. Thus, we tested the effect of MGAT1 knockdown in mice following ethanol consumption, and found a significant reduction in alcohol-induced hepatic lipid accumulation. These results suggest that MGAT1 may afford a promising approach to the treatment of fatty liver disease.


Asunto(s)
Aciltransferasas/metabolismo , Hígado Graso Alcohólico/terapia , PPAR gamma/metabolismo , Aciltransferasas/genética , Animales , Etanol/metabolismo , Hígado Graso Alcohólico/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NAD/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sirtuina 1/genética , Sirtuina 1/metabolismo
18.
Ann Hepatol ; 15(4): 463-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27236145

RESUMEN

 The burden of alcoholic liver disease continues to be a major public health problem worldwide. The spectrum of disease ranges from fatty liver to cirrhosis and hepatocellular carcinoma. Alcoholic hepatitis (AH) is a type of acute-on-chronic liver failure and the most severe form of alcoholic liver disease. Severe AH carries a poor short-term prognosis and its management is still challenging, with scarce advances in the last decades. Corticosteroids are still the first line of therapy in severe cases. Unfortunately, many patients do not respond and novel targeted therapies are urgently needed. Liver transplantation has shown extraordinary results in non-responders to corticosteroids however; its applicability is very low. This review summarizes the epidemiology, natural history, risk factors and pathogenesis of alcoholic liver disease with special focus on the latest advances in prognostic stratification and therapy of patients with alcoholic hepatitis.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada/fisiopatología , Hígado Graso Alcohólico/fisiopatología , Hepatitis Alcohólica/fisiopatología , Insuficiencia Hepática Crónica Agudizada/epidemiología , Insuficiencia Hepática Crónica Agudizada/terapia , Corticoesteroides/uso terapéutico , Hígado Graso Alcohólico/epidemiología , Hígado Graso Alcohólico/terapia , Hepatitis Alcohólica/epidemiología , Hepatitis Alcohólica/terapia , Humanos , Cirrosis Hepática Alcohólica/epidemiología , Cirrosis Hepática Alcohólica/fisiopatología , Cirrosis Hepática Alcohólica/terapia , Trasplante de Hígado , Pronóstico , Factores de Riesgo
19.
J Microbiol ; 53(12): 856-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26626356

RESUMEN

Lactobacillus rhamnosus CCFM1107 was screened for high antioxidative activity from 55 lactobacilli. The present study attempted to explore the protective properties of L. rhamnosus CCFM1107 in alcoholic liver injury. A mouse model was induced by orally feeding alcohol when simultaneously treated with L. rhamnosus CCFM1107, the drug Hu-Gan- Pian (HGP), L. rhamnosus GG (LGG), and L. plantarum CCFM1112 for 3 months. Biochemical analysis was performed for both serum and liver homogenate. Detailed intestinal flora and histological analyses were also carried out. Our results indicated that the administration of L. rhamnosus CCFM1107 significantly inhibited the increase in the levels of serum aminotransferase and endotoxin, as well as the levels of triglyceride (TG) and cholesterol (CHO) in the serum and in the liver. Glutathione (GSH), glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) were elevated while the levels of malondialdehyde (MDA) were decreased. The enteric dysbiosis caused by alcohol was restored by increasing the numbers of both lactobacilli and bifidobacteria and decreasing the numbers of both enterococci and enterobacter. Histological analysis confirmed the protective effect of L. rhamnosus CCFM1107. Compared with the other lactobacilli and to the drug Hu-Gan-Pian, there is a high chance that L. rhamnosus CCFM1107 provides protective effects on alcoholic liver injury by reducing oxidative stress and restoring the intestinal flora.


Asunto(s)
Terapia Biológica/métodos , Etanol/efectos adversos , Hígado Graso Alcohólico/terapia , Lacticaseibacillus rhamnosus , Hígado/efectos de los fármacos , Probióticos/uso terapéutico , Alanina Transaminasa/sangre , Animales , Colesterol/sangre , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Disbiosis/inducido químicamente , Endotoxinas/sangre , Hígado Graso Alcohólico/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Glutatión/sangre , Glutatión Peroxidasa/sangre , Hígado/patología , Masculino , Malondialdehído/sangre , Ratones , Estrés Oxidativo/efectos de los fármacos , Superóxido Dismutasa/sangre , Triglicéridos/sangre
20.
J Nutr Biochem ; 26(4): 337-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25622859

RESUMEN

BACKGROUND: We have previously demonstrated that Lactobacillus rhamnosus GG culture supernatant (LGGs) prevents acute-alcohol-exposure-induced hepatic steatosis and injury. The protective effects of LGGs were attributed to the improved intestinal barrier function leading to decreased endotoxemia. The purpose of this study was to determine whether LGGs was effective in protecting against chronic-alcohol-induced hepatic steatosis and injury and to evaluate the underlying mechanisms of LGGs on hepatic lipid metabolism. METHODS: C57BL/6N mice were fed liquid diet containing 5% alcohol or pair-fed isocaloric maltose dextrin for 4 weeks. LGGs at a dose equivalent to 10(9) CFU/day/mouse was given in the liquid diet. Hepatic steatosis, liver enzymes and hepatic apoptosis were analyzed. RESULTS: LGGs prevented alcohol-mediated increase in hepatic expression of lipogenic genes, sterol regulatory element binding protein-1 and stearoyl-CoA desaturase-1 and increased the expression of peroxisome proliferator activated receptor-α, peroxisome proliferator-activated receptor gamma coactivator protein-1α and carnitine palmitoyltransferase-1, leading to increased fatty acid ß-oxidation. Importantly, chronic alcohol exposure decreased adenosine-monophosphate-activated protein kinase (AMPK) phosphorylation and increased acetyl-CoA carboxylase activity, which were attenuated by LGGs administration. LGGs also decreased Bax expression and increased Bcl-2 expression, which attenuated alcohol-induced hepatic apoptosis. These LGGs-regulated molecular changes resulted in the attenuation of chronic-alcohol-exposure-mediated increase in hepatic fat accumulation and liver injury. CONCLUSIONS: Probiotic LGG culture supernatant is effective in the prevention of chronic-alcohol-exposure-induced hepatic steatosis and injury. LGGs likely exerts its beneficial effects, at least in part, through modulation of hepatic AMPK activation and Bax/Bcl-2-mediated apoptosis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Consumo de Bebidas Alcohólicas/efectos adversos , Medios de Cultivo Condicionados/farmacología , Hígado Graso Alcohólico/microbiología , Hígado Graso Alcohólico/terapia , Lacticaseibacillus rhamnosus/química , Proteínas Quinasas Activadas por AMP/genética , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Fosforilación , Probióticos/administración & dosificación , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Regulación hacia Arriba , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...