Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 490
Filtrer
1.
Sci Rep ; 14(1): 21135, 2024 09 10.
Article de Anglais | MEDLINE | ID: mdl-39256510

RÉSUMÉ

Fatty acid-binding protein 4 (FABP4) plays an essential role in metabolism and inflammation. However, the role of FABP4 in alcoholic steatohepatitis (ASH) remains unclear. This study aimed to investigate the function and underlying mechanisms of FABP4 in the progression of ASH. We first obtained alcoholic hepatitis (AH) datasets from the National Center for Biotechnology Information-Gene Expression Omnibus database and conducted bioinformatics analysis to identify critical genes in the FABP family. We then established ASH models of the wild-type (WT) and Fabp4-deficient (Fabp4-/-) mice to investigate the role of FABP4 in ASH. Additionally, we performed transcriptional profiling of mouse liver tissue and analyzed the results using integrative bioinformatics. The FABP4-associated signaling pathway was further verified. FABP4 was upregulated in two AH datasets and was thus identified as a critical biomarker for AH. FABP4 expression was higher in the liver tissues of patients with alcoholic liver disease and ASH mice than in the corresponding control samples. Furthermore, the Fabp4-/- ASH mice showed reduced hepatic lipid deposition and inflammation compared with the WT ASH mice. Mechanistically, Fabp4 may be involved in regulating the p53 and sirtuin-1 signaling pathways, subsequently affecting lipid metabolism and macrophage polarization in the liver of ASH mice. Our results demonstrate that Fabp4 is involved in the progression of ASH and that Fabp4 deficiency may ameliorate ASH. Therefore, FABP4 may be a potential therapeutic target for ASH treatment.


Sujet(s)
Protéines de liaison aux acides gras , Stéatose hépatique alcoolique , Transduction du signal , Protéine p53 suppresseur de tumeur , Animaux , Protéines de liaison aux acides gras/génétique , Protéines de liaison aux acides gras/métabolisme , Souris , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique alcoolique/génétique , Stéatose hépatique alcoolique/anatomopathologie , Souris knockout , Humains , Mâle , Modèles animaux de maladie humaine , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée C57BL , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Métabolisme lipidique
2.
Int J Biol Macromol ; 279(Pt 4): 135565, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39270893

RÉSUMÉ

Alcoholic fatty liver disease (AFLD) is characterized by excessive lipid accumulation in the liver. This study aimed to investigate the protective effects and mechanisms of Polygala fallax Hemsl polysaccharides (PFPs) on AFLD. PFPs were purified and structurally characterized. An AFLD model was established in mice using alcohol and a high-fat diet. A significant reduction in hepatic steatosis was observed following PFPs treatment, evidenced by decreased fat deposition in liver tissues. Additionally, PFPs reduced various liver injury markers, increased levels of antioxidant enzymes, and improved significantly liver function. RNA sequencing revealed that PFPs improved lipid and CYP450 metabolic pathway abnormalities in AFLD mice. Furthermore, PFPs activated the AMPK pathway, reducing lipid accumulation and enhancing lipid metabolism. A HepG2 cell model treated with ethanol and oleic acid showed significant biochemical improvements with PFPs pretreatment, including reduced lipid accumulation and lower reactive oxygen species (ROS) levels. To further elucidate the AMPK and PFPs correlation in AFLD, an AMPK inhibitor (compound C) was used. In vitro and in vivo qRT-PCR and Western blot results confirmed that PFPs protected against AFLD by activating AMPK phosphorylation, regulating lipid synthesis, and inhibiting lipid accumulation. PFPs also modulated CYP2E1 and oxidative stress-related gene expression, affecting liver metabolism.


Sujet(s)
AMP-Activated Protein Kinases , Stéatose hépatique alcoolique , Métabolisme lipidique , Polygala , Polyosides , Animaux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Polyosides/pharmacologie , Polyosides/composition chimique , Souris , Humains , AMP-Activated Protein Kinases/métabolisme , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/métabolisme , Cellules HepG2 , Polygala/composition chimique , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Cytochrome P-450 CYP2E1/métabolisme , Cytochrome P-450 CYP2E1/génétique , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Souris de lignée C57BL
3.
J Sci Food Agric ; 104(13): 8230-8239, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38873964

RÉSUMÉ

BACKGROUND: Chronic excessive alcohol consumption can lead to alcoholic fatty liver, posing substantial health risks. l-Theanine (LTA) and epigallocatechin gallate (EGCG) in tea exert antioxidant and hepatoprotective effects. However, the combined effects of LTA and EGCG on rats with alcoholic fatty liver, and the underlying mechanisms of such effects, remain unclear. In this study, Sprague Dawley (SD) rats were fed with alcohol for 6 weeks to induce alcoholic fatty liver. Subsequently, for another 6 weeks, the rats were administered LTA (200 mg kg-1 day-1), EGCG (200 mg kg-1 day-1), or a combination of LTA with EGCG (40 mg kg-1 day-1 l-Thea +160 mg kg-1 day-1 EGCG), respectively. RESULTS: The combined use of LTA and EGCG for alcoholic fatty liver disease had more significant effects than their individual administration. This combination reduced the activity of serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) as well as the levels of hepatic triglyceride (TG), malondialdehyde (MDA), and reactive oxygen species (ROS) in the rats. The combined intervention also increased hepatic superoxide dismutase (SOD) and glutathione peroxidase activity. Reductions in hepatic fat accumulation and inflammatory responses were observed. The mechanism underlying these effects primarily involved the inhibition of fatty acid synthesis and the alleviation of lipid peroxidation through the downregulation of the mRNA and protein expression of TNF-α, SREBP1c, and CYP2E1 and the upregulation of the mRNA and protein expression of ADH1, ALDH2, Lipin-1, PPARαPPARα, AMPK, and PGC-1α, thereby promoting the oxidative decomposition of fatty acids and reducing the synthesis of cholesterol and glucose. CONCLUSION: l-Theanine and EGCG appear to be able to alleviate alcoholic fatty liver by modulating lipid metabolism and ameliorating oxidative stress, indicating their potential as natural active ingredients in anti-alcoholic fatty liver food products. © 2024 Society of Chemical Industry.


Sujet(s)
Alanine transaminase , Catéchine , Stéatose hépatique alcoolique , Glutamates , Foie , Malonaldéhyde , Rat Sprague-Dawley , Animaux , Catéchine/analogues et dérivés , Catéchine/administration et posologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Rats , Glutamates/administration et posologie , Mâle , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/métabolisme , Malonaldéhyde/métabolisme , Alanine transaminase/métabolisme , Alanine transaminase/sang , Agents protecteurs/administration et posologie , Agents protecteurs/pharmacologie , Aspartate aminotransferases/métabolisme , Aspartate aminotransferases/sang , Humains , Antioxydants , Triglycéride/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme
4.
Nutrients ; 16(8)2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38674865

RÉSUMÉ

Alcoholic Fatty Liver Disease (AFLD) is characterized by the accumulation of lipids in liver cells owing to the metabolism of ethanol. This process leads to a decrease in the NAD+/NADH ratio and the generation of reactive oxygen species. A systematic review and meta-analysis were conducted to investigate the role of oxidative stress in AFLD. A total of 201 eligible manuscripts were included, which revealed that animals with AFLD exhibited elevated expression of CYP2E1, decreased enzymatic activity of antioxidant enzymes, and reduced levels of the transcription factor Nrf2, which plays a pivotal role in the synthesis of antioxidant enzymes. Furthermore, animals with AFLD exhibited increased levels of lipid peroxidation markers and carbonylated proteins, collectively contributing to a weakened antioxidant defense and increased oxidative damage. The liver damage in AFLD was supported by significantly higher activity of alanine and aspartate aminotransferase enzymes. Moreover, animals with AFLD had increased levels of triacylglycerol in the serum and liver, likely due to reduced fatty acid metabolism caused by decreased PPAR-α expression, which is responsible for fatty acid oxidation, and increased expression of SREBP-1c, which is involved in fatty acid synthesis. With regard to inflammation, animals with AFLD exhibited elevated levels of pro-inflammatory cytokines, including TNF-a, IL-1ß, and IL-6. The heightened oxidative stress, along with inflammation, led to an upregulation of cell death markers, such as caspase-3, and an increased Bax/Bcl-2 ratio. Overall, the findings of the review and meta-analysis indicate that ethanol metabolism reduces important markers of antioxidant defense while increasing inflammatory and apoptotic markers, thereby contributing to the development of AFLD.


Sujet(s)
Stéatose hépatique alcoolique , Stress oxydatif , Animaux , Humains , Antioxydants/métabolisme , Cytochrome P-450 CYP2E1/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine , Stéatose hépatique alcoolique/métabolisme , Peroxydation lipidique , Foie/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Espèces réactives de l'oxygène/métabolisme
5.
Int J Mol Sci ; 25(6)2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38542468

RÉSUMÉ

This study was performed to investigate the protective effects of Allium ochotense on fatty liver and hepatitis in chronic alcohol-induced hepatotoxicity. The physiological compounds of a mixture of aqueous and 60% ethanol (2:8, w/w) extracts of A. ochotense (EA) were identified as kestose, raffinose, kaempferol and quercetin glucoside, and kaempferol di-glucoside by UPLC Q-TOF MSE. The EA regulated the levels of lipid metabolism-related biomarkers such as total cholesterol, triglyceride, low-density lipoprotein (LDL), and high-density lipoprotein (HDL)-cholesterol in serum. Also, EA ameliorated the levels of liver toxicity-related biomarkers such as glutamic oxaloacetic transaminase (GOT), glutamic pyruvic transaminase (GPT), and total bilirubin in serum. EA improved the antioxidant system by reducing malondialdehyde contents and increasing superoxide dismutase (SOD) levels and reduced glutathione content. EA improved the alcohol metabolizing enzymes such as alcohol dehydrogenase, acetaldehyde dehydrogenase, and cytochrome P450 2E1 (CYP2E1). Treatment with EA alleviated lipid accumulation-related protein expression by improving phosphorylation of AMP-activated protein kinase (p-AMPK) expression levels. Especially, EA reduced inflammatory response by regulating the toll-like receptor-4/nuclear factor kappa-light-chain-enhancer of activated B cells (TLR-4/NF-κB) signaling pathway. EA showed an anti-apoptotic effect by regulating the expression levels of B-cell lymphoma 2 (BCl-2), BCl-2-associated X protein (BAX), and caspase 3. Treatment with EA also ameliorated liver fibrosis via inhibition of transforming growth factor-beta 1/suppressor of mothers against decapentaplegic (TGF-ß1/Smad) pathway and alpha-smooth muscle actin (α-SMA). Therefore, these results suggest that EA might be a potential prophylactic agent for the treatment of alcoholic liver disease.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique , Souris , Animaux , Kaempférols/pharmacologie , Foie/métabolisme , Souris de lignée C57BL , Stéatose hépatique alcoolique/métabolisme , Éthanol/toxicité , Éthanol/métabolisme , Stéatose hépatique/métabolisme , Inflammation/métabolisme , Cholestérol/métabolisme , Glucosides/pharmacologie , Marqueurs biologiques/métabolisme , Stress oxydatif
6.
Cell Mol Biol Lett ; 29(1): 35, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38475733

RÉSUMÉ

BACKGROUND AND AIMS: Epidemiological evidence suggests that the phenotype of glutathione S-transferase mu 1 (GSTM1), a hepatic high-expressed phase II detoxification enzyme, is closely associated with the incidence of alcohol-related liver disease (ALD). However, whether and how hepatic GSTM1 determines the development of ALD is largely unclear. This study was designed to elucidate the role and potential mechanism(s) of hepatic GSTM1 in the pathological process of ALD. METHODS: GSTM1 was detected in the liver of various ALD mice models and cultured hepatocytes. Liver-specific GSTM1 or/and micro (miR)-743a-3p deficiency mice were generated by adenoassociated virus-8 delivered shRNA, respectively. The potential signal pathways involving in alcohol-regulated GSTM1 and GSTM1-associated ALD were explored via both genetic manipulation and pharmacological approaches. RESULTS: GSTM1 was significantly upregulated in both chronic alcohol-induced mice liver and ethanol-exposed murine primary hepatocytes. Alcohol-reduced miR-743a-3p directly contributed to the upregulation of GSTM1, since liver specific silencing miR-743a-3p enhanced GSTM1 and miR-743a-3p loss protected alcohol-induced liver dysfunctions, which was significantly blocked by GSTM1 knockdown. GSTM1 loss robustly aggravated alcohol-induced hepatic steatosis, oxidative stress, inflammation, and early fibrotic-like changes, which was associated with the activation of apoptosis signal-regulating kinase 1 (ASK1), c-Jun N-terminal kinase (JNK), and p38. GSTM1 antagonized ASK1 phosphorylation and its downstream JNK/p38 signaling pathway upon chronic alcohol consumption via binding with ASK1. ASK1 blockage significantly rescued hepatic GSTM1 loss-enhanced disorders in alcohol-fed mice liver. CONCLUSIONS: Chronic alcohol consumption-induced upregulation of GSTM1 in the liver provides a feedback protection against hepatic steatosis and liver injury by counteracting ASK1 activation. Down-regulation of miR-743a-3p improves alcohol intake-induced hepatic steatosis and liver injury via direct targeting on GSTM1. The miR-743a-3p-GSTM1 axis functions as an innate protective pathway to defend the early stage of ALD.


Sujet(s)
Stéatose hépatique alcoolique , Glutathione transferase , microARN , Animaux , Souris , Glutathione transferase/métabolisme , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Foie/anatomopathologie , microARN/métabolisme , Stéatose hépatique alcoolique/métabolisme
7.
J Tradit Chin Med ; 44(2): 277-288, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38504534

RÉSUMÉ

OBJECTIVE: To analyze the effect and molecular mechanism of Gehua Jiejiu Dizhi decoction (, GJDD) on alcoholic fatty live disease (AFLD) by using proteomic methods. METHODS: The male C57BL/6J mouse were randomly divided into four groups: control group, model group, GJDD group and resveratrol group. After the AFLD model was successfully prepared by intragastric administration of alcohol once on the basis of the Lieber-DeCarli classical method, the GJDD group and resveratrol group were intragastrically administered with GJDD (4900 mg/kg) and resveratrol (400 mg/kg) respectively, once a day for 9 d. The fat deposition of liver tissue was observed and evaluated by oil red O (ORO) staining. 4DLabel-free quantitative proteome method was used to determine and quantify the protein expression in liver tissue of each experimental group. The differentially expressed proteins were screened according to protein expression differential multiples, and then analyzed by Gene ontology classification and Kyoto Encyclopedia of Genes and Genomes pathway enrichment. Finally, expression validation of the differentially co-expressed proteins from control group, model group and GJDD group were verified by targeted proteomics quantification techniques. RESULTS: In semiquantitative analyses of ORO, all kinds of steatosis (ToS, MaS, and MiS) were evaluated higher in AFLD mice compared to those in GJDD or resveratrol-treated mice. 4DLabel-free proteomics analysis results showed that a total of 4513 proteins were identified, of which 3763 proteins were quantified and 946 differentially expressed proteins were screened. Compared with the control group, 145 proteins were up-regulated and 148 proteins were down-regulated in the liver tissue of model group. In addition, compared with the model group, 92 proteins were up-regulated and 135 proteins were down-regulated in the liver tissue of the GJDD group. 15 differentially co-expressed proteins were found between every two groups (model group vs control group, GJDD group vs model group and GJDD group vs control group), which were involved in many biological processes. Among them, 11 differentially co-expressed key proteins (Aox3, H1-5, Fabp5, Ces3a, Nudt7, Serpinb1a, Fkbp11, Rpl22l1, Keg1, Acss2 and Slco1a1) were further identified by targeted proteomic quantitative technology and their expression patterns were consistent with the results of 4D label-free proteomic analysis. CONCLUSIONS: Our study provided proteomics-based evidence that GJDD alleviated AFLD by modulating liver protein expression, likely through the modulation of lipid metabolism, bile acid metabolism and with exertion of antioxidant stress.


Sujet(s)
Stéatose hépatique alcoolique , Serpines , Souris , Mâle , Animaux , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/génétique , Stéatose hépatique alcoolique/métabolisme , Antioxydants/métabolisme , Protéomique/méthodes , Resvératrol/métabolisme , Effort physique , Souris de lignée C57BL , Foie/métabolisme , Métabolisme lipidique , Acides et sels biliaires/métabolisme , Lipides , Serpines/métabolisme , Aldehyde oxidoreductases/métabolisme
8.
Int Immunopharmacol ; 127: 111460, 2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38157696

RÉSUMÉ

BACKGROUND: Ligustilide (Lig) is the main active ingredient of Umbelliferae Angelicae Sinensis Radix (Chinese Angelica) and Chuanxiong Rhizoma (Sichuan lovase rhizome). Lig possesses various pharmacological properties and could treat obesity by regulating energy metabolism. However, the impact and regulatory mechanism of Lig on alcoholic hepatic steatosis remains unclear. PURPOSE: This study aimed to explore the therapeutic effect of Lig on alcoholic hepatic steatosis and its related pharmacological mechanism. RESULTS: With chronic and binge ethanol feeding, liver tissue damage and lipid accumulation in mice suffering alcoholic hepatic steatosis were significantly improved after Lig treatment. Lig effectively regulated the expression levels of lipid metabolism-related proteins in alcoholic hepatic steatosis. In addition, Lig reduced RXFP1 expression, inhibited the activation of NLRP3 inflammasome, and blocked NET formation. Lig reduced the infiltration of immune cells to the liver and the further prevented the occurrence of alcohol-stimulated inflammatory response in liver. Lig significantly regulated lipid accumulation in alcohol exposed AML12 cells via modulating PPARα and SREBP1. In MPMs, Lig decreased the expression of RXFP1, inhibited the activation of NLRP3 in macrophages stimulated by LPS/ATP, and slowed down the occurrence of inflammatory response. CONCLUSION: Lig sustained lipid metabolism homeostasis in alcoholic hepatic steatosis, through inhibiting the activation of NLRP3 inflammasomes and the formation of NETs, especially targeting RXFP1 in macrophages.


Sujet(s)
4-Butyrolactone/analogues et dérivés , Stéatose hépatique alcoolique , Protéine-3 de la famille des NLR contenant un domaine pyrine , Souris , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/métabolisme , Foie/métabolisme , Éthanol/usage thérapeutique , Inflammasomes , Lipides/usage thérapeutique , Souris de lignée C57BL
9.
Int J Biol Sci ; 19(16): 5074-5088, 2023.
Article de Anglais | MEDLINE | ID: mdl-37928262

RÉSUMÉ

Alcoholic liver disease (ALD) encompasses conditions ranging from simple steatosis to cirrhosis and even liver cancer. It has gained significant global attention in recent years. Despite this, effective pharmacological treatments for ALD remain elusive, and the core mechanisms underlying the disease are not yet fully comprehended. S100A16, a newly identified calcium-binding protein, is linked to lipid metabolism. Our research has discovered elevated levels of the S100A16 protein in both serum and liver tissue of ALD patients. A similar surge in hepatic S100A16 expression was noted in a Gao-binge alcohol feeding mouse model. S100a16 knockdown alleviated ethanol-induced liver injury, steatosis and inflammation. Conversely, S100a16 transgenic mice showed aggravating phenomenon. Mechanistically, we identify mesencephalic astrocyte-derived neurotrophic factor (MANF) as a regulated entity downstream of S100a16 deletion. MANF inhibited ER-stress signal transduction induced by alcohol stimulation. Meanwhile, MANF silencing suppressed the inhibition effect of S100a16 knockout on ethanol-induced lipid droplets accumulation in primary hepatocytes. Our data suggested that S100a16 deletion protects mice against alcoholic liver lipid accumulation and inflammation dependent on upregulating MANF and inhibiting ER stress. This offers a potential therapeutic avenue for ALD treatment.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique , Maladies alcooliques du foie , Humains , Souris , Animaux , Stéatose hépatique alcoolique/métabolisme , Maladies alcooliques du foie/métabolisme , Stéatose hépatique/métabolisme , Éthanol/toxicité , Inflammation/métabolisme , Facteurs de croissance nerveuse/génétique , Facteurs de croissance nerveuse/métabolisme
10.
Biosci Rep ; 43(11)2023 11 30.
Article de Anglais | MEDLINE | ID: mdl-37883219

RÉSUMÉ

OBJECTIVES: Alanine aminotransferase (ALT) expression is highly elevated in the serum of patients with hepatocellular carcinoma. However, the role of ALT isoenzymes in the total ALT activity remains unclear. In the present study, we systematically investigated the role of ALT isoenzymes in alcoholic and non-alcoholic fatty liver cancer. MATERIALS AND METHODS: The expression of ALT1 and ALT2 at the mRNA and protein levels in 25 paired primary liver cancer tissues was detected by reverse transcription quantitative PCR (RT-qPCR), Western blotting, and immunohistochemistry. Serum ALT activity was determined using an automated biochemical analyzer. RESULTS: The mRNA and protein expression levels of ALT1 and ALT2 were lower in the tissues of alcoholic and non-alcoholic fatty liver cancers than in the paracancerous tissues. Notably, ALT2 was highly expressed in non-alcoholic fatty liver cancer tissues compared with alcoholic fatty liver cancer tissues. Total serum ALT activity was mainly contributed by ALT1 in alcoholic fatty liver cancer, whereas ALT1 contributed only marginally more to the total ALT activity than ALT2 in non-alcoholic fatty liver cancer. ALT2/ALT1 ratio can well discriminate normal control group, alcoholic liver cancer and non-alcoholic liver cancer. CONCLUSION: ALT1 contributed more to the total ALT activity than ALT2 in both alcoholic and non-alcoholic fatty liver cancer. Serum ALT2 to ALT activity was higher in non-alcoholic fatty liver cancer than that in alcoholic fatty liver cancer. ALT2/ALT1 ratio has some diagnostic significance for alcoholic and non-alcoholic liver cancer.


Sujet(s)
Stéatose hépatique alcoolique , Tumeurs du foie , Stéatose hépatique non alcoolique , Humains , Alanine transaminase , Isoenzymes/métabolisme , Stéatose hépatique alcoolique/diagnostic , Stéatose hépatique alcoolique/génétique , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique non alcoolique/diagnostic , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/métabolisme , Tumeurs du foie/diagnostic , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , ARN messager/métabolisme , Foie/métabolisme
11.
Mol Med Rep ; 28(5)2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37772395

RÉSUMÉ

Alcoholic fatty liver disease (AFLD) is a disease with a high incidence rate among individuals who drink alcohol. Our previous study found that agarwood alcohol extracts (AAEs) have a protective effect against drug­induced liver damage via anti­inflammatory and antioxidant mechanisms. Therefore, we hypothesized that agarwood may have a protective effect against AFLD. The present study assessed the potential protective effects and the underlying mechanism of action of AAEs for the treatment of an AFL in vivo model. The AFLD mouse model was established by continuous high fat diet and alcohol gavage in C57 mice. After treatment with AAEs, blood was collected, liver and adipose tissues were removed and liver and adipose indexes were analyzed. The levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), triglyceride (TG) and cholesterol (CHO) in serum were detected. The liver tissue was assessed using pathological sections. Biochemical methods were used to detect the levels of oxidative stress in the supernatant of liver tissue homogenate. The levels of pro­inflammatory cytokines in the serum were detected by ELISA. The protein expression levels of nuclear erythroid 2­related factor 2 (Nrf2) and nuclear factor kappa­B (NF­κB) in liver tissues were detected using western blotting. AAE treatment decreased the liver and adipose indexes, reduced the levels of AST, ALT, TG and CHO, improved the liver pathological characteristics and enhanced antioxidant and anti­inflammatory activities. In addition, AAEs increased the protein expression level of Nrf2 and decreased the protein expression level of NF­κB compared with AFL mice. AAE­treated animals exhibited reduced metabolic enzyme and blood lipid levels, demonstrated improved liver function and relieved the pathological damage of AFLD induced by consuming a high fat and alcohol diet. AAEs have potential protective effects in AFLD via antioxidant and anti­inflammatory mechanisms.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique non alcoolique , Souris , Animaux , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/métabolisme , Antioxydants/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Foie/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Éthanol/pharmacologie , Cholestérol/métabolisme , Triglycéride/métabolisme , Obésité/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/métabolisme
12.
Cell Signal ; 112: 110905, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37743009

RÉSUMÉ

BACKGROUND: Alcoholic fatty liver disease (AFLD) is characterized by abnormal lipid droplet accumulation in liver. Epigenetic regulation plays an important role in the pathogenesis of AFLD. Comprehensive bioinformatics analysis revealed that an E3 ubiquitin ligase, F-box and leucine-rich repeats protein 5 (FBXL5), was significantly upregulated in AFLD mice. METHODS: The mouse model of AFLD was established by feeding Lieber-DeCarli liquid diet containing ethanol. An in vitro model of AFLD was established by treating HepG2 cells with ethanol (EtOH). The FBXL5 expression was assessed by quantitative real-time PCR (qRT-PCR) and western blotting assays. The levels of triglyceride (TG), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lipid accumulation were analyzed by enzyme-linked immunosorbent assay (ELISA) and Nile red staining. RESULTS: The FBXL5 expression was markedly up-regulated in in vivo and in vitro models of AFLD compared with controls. Functionally, FBXL5 knockdown alleviated lipid accumulation in EtOH-treated HepG2 cells. Mechanistically, FBXL5 directly interacted with transcription factor EB (TFEB) and accelerated its ubiquitination-mediated degradation. TFEB knockdown reversed the effect of FBXL5 inhibition on decreasing EtOH-induced lipid accumulation. CONCLUSION: Our data suggest that FBXL5 promotes lipid accumulation in AFLD by promoting the ubiquitination and degradation of TFEB.


Sujet(s)
Protéines F-box , Stéatose hépatique alcoolique , Animaux , Souris , Épigenèse génétique , Éthanol/toxicité , Éthanol/métabolisme , Protéines F-box/métabolisme , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique alcoolique/anatomopathologie , Lipides , Foie/métabolisme , Ubiquitination , Humains
13.
Biochem Biophys Res Commun ; 672: 36-44, 2023 09 10.
Article de Anglais | MEDLINE | ID: mdl-37336123

RÉSUMÉ

Lipid droplets are unique lipid storage organelles in hepatocytes. Lipophagy is a key mechanism of selective degradation of lipid droplets through lysosomes. It plays a crucial role in the prevention of metabolic liver disease, including nonalcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD), and is a potential therapeutic target for treating these dysfunctions. In this review, we highlighted recent research and discussed advances in key proteins and molecular mechanisms related to lipophagy in liver disease. Reactive oxygen species (ROS) is an inevitable product of metabolism in alcohol-treated or high-fat-treated cells. Under this light, the potential role of ROS in autophagy in lipid droplet removal was initially explored to provide insights into the link between oxidative stress and metabolic liver disease. Subsequently, the current measures and drugs that treat NAFLD and AFLD through lipophagy regulation were summarized. The complexity of molecular mechanisms underlying lipophagy in hepatocytes and the need for further studies for their elucidation, as well as the status and limitations of current therapeutic measures and drugs, were also discussed.


Sujet(s)
Stéatose hépatique alcoolique , Maladies métaboliques , Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique alcoolique/métabolisme , Espèces réactives de l'oxygène/métabolisme , Métabolisme lipidique/physiologie , Foie/métabolisme , Autophagie/physiologie , Maladies métaboliques/métabolisme , Gouttelettes lipidiques/métabolisme
14.
Biochem Pharmacol ; 213: 115613, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37209859

RÉSUMÉ

Fatty liver is the earliest response to excessive ethanol consumption, which increases the susceptibility of the liver to develop advanced stage of liver disease. Our previous studies have revealed that chronic alcohol administration alters metabolic hormone levels and their functions. Of current interest to our laboratory is glucagon-like peptide 1 (GLP-1), a widely studied hormone known to reduce insulin resistance and hepatic fat accumulation in patients with metabolic-associated fatty liver disease. In this study, we examined the beneficial effects of exendin-4 (a GLP-1 receptor agonist) in an experimental rat model of ALD. Male Wistar rats were pair-fed the Lieber-DeCarli control or ethanol diet. After 4 weeks of this feeding regimen, a subset of rats in each group were intraperitoneally injected every other day with either saline or exendin-4 at a dose of 3 nmol/kg/day (total 13 doses) while still being fed their respective diet. At the end of the treatment, rats were fasted for 6 h and glucose tolerance test was conducted. The following day, the rats were euthanized, and the blood and tissue samples collected for subsequent analysis. We found that exendin-4 treatment had no significant effect on body weight gain among the experimental groups. Exendin-4-treated ethanol rats exhibited improved alcohol-induced alterations in liver/body weight and adipose/body weight ratio, serum ALT, NEFA, insulin, adiponectin and hepatic triglyceride levels. Reduction in indices of hepatic steatosis in exendin-4 treated ethanol-fed rats was attributed to improved insulin signaling and fat metabolism. These results strongly suggest that exendin-4 mitigates alcohol-associated hepatic steatosis by regulating fat metabolism.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique non alcoolique , Rats , Mâle , Animaux , Exénatide/pharmacologie , Exénatide/usage thérapeutique , Rat Wistar , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/prévention et contrôle , Stéatose hépatique alcoolique/métabolisme , Insuline/métabolisme , Glucagon-like peptide 1/agonistes , Éthanol/toxicité , Obésité/métabolisme
15.
Transpl Immunol ; 78: 101837, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37031771

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) derived exosomes (MSCs/Exo) is considered a new strategy in cell free regenerative therapy. Curcumin preconditioning of MSCs reported to improve the anti- inflammatory and immunomodulatory properties of MSCs. We investigated the efficacy of exosome (Exo) obtained from curcumin-preconditioned MSCs (MSCs/Exo-Cur) vs. MSC/Exo without curcumin to ameliorate and prevent recurrence of non-alcoholic fatty liver (NASH) disease. METHODS AND RESULTS: In-vivo, methionine/choline-deficient diet (MCD) induced mice non-alcoholic fatty liver disease (NASH) were injected with MSCs/Exo without curcumin or MSCs/Exo-Cur with curcumin. We found that mice treated with MSCs/Exo-Cur had significantly ameliorated steatosis, inflammation, as evaluated by the reduced fibrosis in histopathological examination, decreased the serum level of liver enzymes (p < 0.001), liver triglycerides (TG) (p < 0.001) and cholesterol (Ch) (p < 0.001) and increased the lipid peroxidation (p < 0.001) compared to MSCs/Exo-treated mice. These effects remained for 3 months after treatment in MSCs/Exo-Cur-treated mice while features of NASH returned in MSCs/Exo-treated group. In vitro, HepG2 cells were cultured with palmitic acid (PA) and treated with MSCs/Exo or MSCs/Exo-Cur: the MSCs/Exo-Cur exposure reversed the lipotoxic effect from 4.5 to 1.7 fold vs 4.0 fold in MSCs/Exo and oxidative stress in PA-treated HepG2 cells (p < 0.001). We found that MSCs/Exo-Cur regulated the key markers of inflammatory and oxidative stress, genes responsible for fibrogenesis of the liver, key genes of lipid synthesis and transport. Interestingly, MSCs/Exo-Cur significantly down regulated the ASK-JNK-BAX genes involved in mitochondrial stress and apoptosis compared to MSCs/Exo (p < 0.001). CONCLUSION: Our study indicated that exosomes derived from curcumin preconditioned MSCs were able to ameliorate and protect against recurrence of NASH and regulated inflammatory, oxidative stress and mitochondrial-dependent apoptosis ASK-JNK-BAX genes.


Sujet(s)
Curcumine , Exosomes , Stéatose hépatique alcoolique , Cellules souches mésenchymateuses , Stéatose hépatique non alcoolique , Souris , Animaux , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/métabolisme , Curcumine/usage thérapeutique , Curcumine/métabolisme , Curcumine/pharmacologie , Exosomes/métabolisme , Stéatose hépatique alcoolique/métabolisme , Protéine Bax/métabolisme , Inflammation/thérapie , Inflammation/métabolisme , Stress oxydatif
16.
EMBO J ; 42(8): e112304, 2023 04 17.
Article de Anglais | MEDLINE | ID: mdl-36825429

RÉSUMÉ

The tumor suppressor p53 is critical for tumor suppression, but the regulatory role of p53 in alcohol-induced fatty liver remains unclear. Here, we show a role for p53 in regulating ethanol metabolism via acetaldehyde dehydrogenase 2 (ALDH2), a key enzyme responsible for the oxidization of alcohol. By repressing ethanol oxidization, p53 suppresses intracellular levels of acetyl-CoA and histone acetylation, leading to the inhibition of the stearoyl-CoA desaturase-1 (SCD1) gene expression. Mechanistically, p53 directly binds to ALDH2 and prevents the formation of its active tetramer and indirectly limits the production of pyruvate that promotes the activity of ALDH2. Notably, p53-deficient mice exhibit increased lipid accumulation, which can be reversed by ALDH2 depletion. Moreover, liver-specific knockdown of SCD1 alleviates ethanol-induced hepatic steatosis caused by p53 loss. By contrast, overexpression of SCD1 in liver promotes ethanol-induced fatty liver development in wild-type mice, while it has a mild effect on p53-/- or ALDH2-/- mice. Overall, our findings reveal a previously unrecognized function of p53 in alcohol-induced fatty liver and uncover pyruvate as a natural regulator of ALDH2.


Sujet(s)
Aldehyde dehydrogenase, mitochondrial , Stéatose hépatique alcoolique , Stéatose hépatique , Protéine p53 suppresseur de tumeur , Animaux , Souris , Aldehyde dehydrogenase, mitochondrial/génétique , Aldehyde dehydrogenase, mitochondrial/métabolisme , Éthanol/toxicité , Éthanol/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique alcoolique/génétique , Stéatose hépatique alcoolique/métabolisme , Foie/métabolisme , Pyruvates/métabolisme , Pyruvates/pharmacologie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
17.
J Ethnopharmacol ; 307: 116227, 2023 May 10.
Article de Anglais | MEDLINE | ID: mdl-36739928

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Verbenalin is a major compound in Verbena officinalis L. Verbena officinalis L was first recorded in the 'Supplementary Records of Famous Physicians.' Verbenalin (VE) is its active constituent and has been found to have many biological effects, including anti-obesity, anti-inflammatory, and antioxidant activities, removing jaundice, and treating malaria. It could treat lump accumulation, dysmenorrhea, throat obstruction, edema, jaundice, and malaria. Palmitic acid (PA), oleic acid (OA), ethanol, and acetaminophen liver injuries have been proven to benefit from verbenalin. AIM OF THE STUDY: To study the effects of verbenalin on the prevention of alcoholic steatohepatitis (ASH) through the regulation of oxidative stress and mitochondrial dysfunction by regulating MDMX (Murine double minute X)/PPARα (Peroxisome proliferator-activated receptor alpha)-mediated ferroptosis. MATERIAL AND METHODS: C57BL/6 mice treated with alcohol followed by the Gao-Binge protocol were administered verbenalin by gavage simultaneously. The mitochondrial mass and morphology were visualized using TEM. AML-12 cells were stimulated with ethanol to mimic ASH in vitro. Western blotting, co-immunoprecipitation, and kit determination were simultaneously performed. The target protein of verbenalin was identified by molecular docking, and cellular thermal shift assay (CETSA) further confirmed its interactions. RESULTS: Verbenalin alleviates oxidative stress and ferroptosis in alcohol-associated steatohepatitis. To elucidate the molecular mechanism by which verbenalin inhibits abnormal mitochondrial dysfunction, molecular docking was performed, and MDMX was identified as the target protein of verbenalin. CETSA assays revealed a specific interaction between MDMX and verbenalin. Co-immunoprecipitation demonstrated that PPARα played a critical role in promoting the ability of MDMX to affect ferroptosis. Verbenalin regulates MDMX/PPARα-mediated ferroptosis in AML-12 cells. CONCLUSION: Verbenalin regulates ferroptosis and highlights the therapeutic potential of verbenalin and ferroptosis inhibition in reducing alcoholic steatohepatitis.


Sujet(s)
Stéatose hépatique alcoolique , Ferroptose , Leucémie aigüe myéloïde , Stéatose hépatique non alcoolique , Animaux , Femelle , Souris , Éthanol/pharmacologie , Stéatose hépatique alcoolique/métabolisme , Leucémie aigüe myéloïde/métabolisme , Foie , Souris de lignée C57BL , Mitochondries/métabolisme , Simulation de docking moléculaire , Stéatose hépatique non alcoolique/métabolisme , Récepteur PPAR alpha/métabolisme , Protéines/métabolisme
18.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-36835145

RÉSUMÉ

Alcoholic liver disease (ALD) is a consequence of excessive alcohol use. According to many studies, alcohol represents a significant socioeconomic and health risk factor in today's population. According to data from the World Health Organization, there are about 75 million people who have alcohol disorders, and it is well known that its use leads to serious health problems. ALD is a multimodality spectrum that includes alcoholic fatty liver disease (AFL) and alcoholic steatohepatitis (ASH), consequently leading to liver fibrosis and cirrhosis. In addition, the rapid progression of alcoholic liver disease can lead to alcoholic hepatitis (AH). Alcohol metabolism produces toxic metabolites that lead to tissue and organ damage through an inflammatory cascade that includes numerous cytokines, chemokines, and reactive oxygen species (ROS). In the process of inflammation, mediators are cells of the immune system, but also resident cells of the liver, such as hepatocytes, hepatic stellate cells, and Kupffer cells. These cells are activated by exogenous and endogenous antigens, which are called pathogen and damage-associated molecular patterns (PAMPs, DAMPs). Both are recognized by Toll-like receptors (TLRs), which activation triggers the inflammatory pathways. It has been proven that intestinal dysbiosis and disturbed integrity of the intestinal barrier perform a role in the promotion of inflammatory liver damage. These phenomena are also found in chronic excessive use of alcohol. The intestinal microbiota has an important role in maintaining the homeostasis of the organism, and its role in the treatment of ALD has been widely investigated. Prebiotics, probiotics, postbiotics, and symbiotics represent therapeutic interventions that can have a significant effect on the prevention and treatment of ALD.


Sujet(s)
Stéatose hépatique alcoolique , Maladies alcooliques du foie , Microbiote , Humains , Maladies alcooliques du foie/métabolisme , Éthanol/métabolisme , Foie/métabolisme , Inflammation/métabolisme , Stéatose hépatique alcoolique/métabolisme
19.
Arch Toxicol ; 97(5): 1335-1353, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36826472

RÉSUMÉ

Despite the high prevalence of alcoholic liver disease, its identification and characterization remain poor, especially in early stages such as alcoholic fatty liver disease and alcoholic steatohepatitis. This latter implies diagnostic difficulties, few therapeutic options and unclear mechanisms of action. To elucidate the metabolic alterations and pinpoint affected biochemical pathways, alcoholic steatohepatitis was simulated in vitro by exposing HepaRG cells to ethanol (IC10, 368 mM) and tumor necrosis factor alpha (TNF-α, 50 ng/mL) for 24 h. This combined exposure was compared to solely ethanol-exposed as well as -nonexposed cells. Four different metabolomics platforms were used combining liquid chromatography, high-resolution mass spectrometry and drift tube ion mobility to elucidate both intracellular and extracellular metabolic alterations. Some of the key findings include the influence of TNF-α in the upregulation of hepatic triglycerides and the downregulation of hepatic phosphatidylethanolamines and phosphatidylcholines. S-Adenosylmethionine showed to play a central role in the progression of alcoholic steatohepatitis. In addition, fatty acyl esters of hydroxy fatty acid (FAHFA)-containing triglycerides were detected for the first time in human hepatocytes and their alterations showed a potentially important role during the progression of alcoholic steatohepatitis. Ethoxylated phosphorylcholine was identified as a potential new biomarker of ethanol exposure.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique non alcoolique , Humains , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique alcoolique/anatomopathologie , Éthanol/toxicité , Facteur de nécrose tumorale alpha/métabolisme , Chromatographie en phase liquide , Spectrométrie de masse en tandem , Foie/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Métabolomique , Triglycéride/métabolisme
20.
Toxicol Appl Pharmacol ; 461: 116406, 2023 02 15.
Article de Anglais | MEDLINE | ID: mdl-36708882

RÉSUMÉ

Steatosis is regarded as an early response of the liver to excessive alcohol consumption, which ultimately results in alcoholic liver disease (ALD). Hepatocytes are the primary drivers of the pathological process known as hepatic damage and steatosis, which is characterized by significant fat accumulation and an abundance of fat vacuoles. NLRs, a family member of pattern recognition receptors, have recently been found to be crucial in liver disorders. In this study, we examined the possible impact of NLRC5, the largest NLR family member, on alcohol-induced fatty liver development using a gene knock-out mouse model. The mouse liver was severely damaged and developed steatosis as a result of chronic and excessive ethanol use, and this damage was prevented by the lack of NLRC5. Additionally, NLRC5 deletion reversed ethanol's ability to increase the serum concentrations of TG, T-CHO, ALT, and AST. Absence of NLRC5 reduced ethanol-stimulated aberrant expression of the vital regulators of lipid synthesis and metabolism, SREBP-1c, FAS and PPAR-α. Furthermore, loss- and gain-of-function research indicated that NLRC5 might affect the autophagy pathway in alcohol-induced hepatic steatosis progression. The functional role of NLRC5 in ALD is obviously impacted by the autophagy inducer rapamycin as well as the autophagy inhibitor 3-MA. Our research showed that NLRC5 was involved in ethanol-induced injury and steatosis of the liver, and may be considered a suitable therapeutic target for treating ALD.


Sujet(s)
Stéatose hépatique alcoolique , Stéatose hépatique , Maladies alcooliques du foie , Souris , Animaux , Foie , Stéatose hépatique/traitement médicamenteux , Éthanol , Hépatocytes , Stéatose hépatique alcoolique/traitement médicamenteux , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique alcoolique/anatomopathologie , Maladies alcooliques du foie/métabolisme , Autophagie , Protéines et peptides de signalisation intracellulaire/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE