Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.376
Filter
1.
Mol Nutr Food Res ; 68(12): e2300833, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38850176

ABSTRACT

SCOPE: Alcoholic liver disease (ALD) is a global public health concern. Nobiletin, a polymethoxyflavone abundant in citrus fruits, enhances circadian rhythms and ameliorates diet-induced hepatic steatosis, but its influences on ALD are unknown. This study investigates the role of brain and muscle Arnt-like protein-1 (Bmal1), a key regulator of the circadian clock, in nobiletin-alleviated ALD. METHODS AND RESULTS: This study uses chronic ethanol feeding plus an ethanol binge to establish ALD models in Bmal1flox/flox and Bmal1 liver-specific knockout (Bmal1LKO) mice. Nobiletin mitigates ethanol-induced liver injury (alanine aminotransferase [ALT]), glucose intolerance, hepatic apoptosis, and lipid deposition (triglyceride [TG], total cholesterol [TC]) in Bmal1flox/flox mice. Nobiletin fails to modulated liver injury (ALT, aspartate aminotransferase [AST]), apoptosis, and TG accumulation in Bmal1LKO mice. The expression of lipogenic genes (acetyl-CoA carboxylase alpha [Acaca], fatty acid synthase [Fasn]) and fatty acid oxidative genes (carnitine pamitoyltransferase [Cpt1a], cytochrome P450, family 4, subfamily a, polypeptide 10 [Cyp4a10], and cytochrome P450, family4, subfamily a, polypeptide 14 [Cyp4a14]) is inhibited, and the expression of proapoptotic genes (Bcl2 inteacting mediator of cell death [Bim]) is enhanced by ethanol in Bmal1flox/flox mice. Nobiletin antagonizes the expression of these genes in Bmal1flox/flox mice and not in Bmal1LKO mice. Nobiletin activates protein kinase B (PKB, also known as AKT) phosphorylation, increases the levels of the carbohydrate response element binding protein (ChREBP), ACC1, and FASN, and reduces the level of sterol-regulatory element binding protein 1 (SREBP1) and phosphorylation of ACC1 in a Bmal1-dependent manner. CONCLUSION: Nobiletin alleviates ALD by increasing the expression of genes involved in fatty acid oxidation by increasing AKT phosphorylation and lipogenesis in a Bmal1-dependent manner.


Subject(s)
ARNTL Transcription Factors , Flavones , Lipogenesis , Liver Diseases, Alcoholic , Mice, Knockout , Proto-Oncogene Proteins c-akt , Animals , Flavones/pharmacology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/drug therapy , Lipogenesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Male , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Mice , Protective Agents/pharmacology , Ethanol , Signal Transduction/drug effects , Apoptosis/drug effects
2.
Food Res Int ; 188: 114489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823872

ABSTRACT

Solanum nigrum L. (SN) berry is an edible berry containing abundant polyphenols and bioactive compounds, which possess antioxidant and antiinflammatory properties. However, the effects of SN on alcohol-induced biochemical changes in the enterohepatic axis remain unclear. In the current study, a chronic ethanol-fed mice ALD model was used to test the protective mechanisms of SN berries. Microbiota composition was determined via 16S rRNA sequencing, we found that SN berries extract (SNE) improved intestinal imbalance by reducing the Firmicutes to Bacteroides ratio, restoring the abundance of Akkermansia microbiota, and reducing the abundance of Allobaculum and Shigella. SNE restored the intestinal short-chain fatty acids content. In addition, liver transcriptome data analysis revealed that SNE primarily affected the genes involved in lipid metabolism and inflammatory responses. Furthermore, SNE ameliorated hepatic steatosis in alcohol-fed mice by activating AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), peroxisome proliferator-activated receptor α (PPAR-α). SNE reduced the expression of toll-like receptor 4 (TLR4), myeloid differentiation factor-88 (MyD88) nuclear factor kappa-B (NF-κB), which can indicate that SNE mainly adjusted LPS/TLR4/MyD88/NF-κB pathway to reduce liver inflammation. SNE enhanced hepatic antioxidant capacity by regulating NRF2-related protein expression. SNE alleviates alcoholic liver injury by regulating of gut microbiota, lipid metabolism, inflammation, and oxidative stress. This study may provide a reference for the development and utilization of SN resources.


Subject(s)
Fruit , Gastrointestinal Microbiome , Lipid Metabolism , Liver Diseases, Alcoholic , Oxidative Stress , Plant Extracts , Solanum nigrum , Animals , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Lipid Metabolism/drug effects , Plant Extracts/pharmacology , Mice , Fruit/chemistry , Solanum nigrum/chemistry , Male , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Mice, Inbred C57BL , Inflammation , Liver/drug effects , Liver/metabolism , Toll-Like Receptor 4/metabolism , Disease Models, Animal , PPAR alpha/metabolism , Antioxidants/pharmacology , Ethanol
3.
Medicine (Baltimore) ; 103(25): e38315, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905402

ABSTRACT

Gegensan (GGS) has been reported for the treatment of alcoholic liver disease (ALD), but its therapeutic mechanism is still unclear. This paper aims to determine the therapeutic mechanism and targets of action of GGS on alcoholic liver disease utilizing network pharmacology and bioinformatics. The active ingredients in GGS were screened in the literature and databases, and common targets of ALD were then obtained from public databases to construct the network diagram of traditional Chinese medicine-active ingredient targets. Based on the common targets, Gene Ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed to find target enrichment pathways, and the core targets were screened out by combining differential analysis and protein-protein interaction network analysis. Molecular docking was performed to verify the binding effect between the core targets and the corresponding active ingredients. ALD and GGS have 84 common targets, corresponding to 91 active ingredients. After subsequent differential analysis and protein-protein interaction network analysis, 10 core targets were identified. Gene Ontology and KEGG enrichment analyses showed that the main BPs corresponding to the common targets included the response to lipopolysaccharide, inflammatory response, etc. The KEGG pathways involved in the regulation of the common targets included the lipid-atherosclerosis pathway and the alcoholic liver disease pathway, etc. Further molecular docking showed that the core targets CYP1A1, CYP1A2, CXCL8, ADH1C, MMP1, SERPINE1, COL1A1, APOB, MMP1, and their corresponding 4 active ingredients, Naringenin, Kaempferol, Quercetin, and Stigmasterol, have a greater docking potential. The above results suggest that GGS can regulate lipid metabolism and inflammatory response in the ALD process, and alleviate the lipid accumulation and oxidative stress caused by ethanol. This study analyzed the core targets and mechanisms of action of GGS on ALD, which provides certain theoretical support for the further development of GGS in the treatment of ALD, and provides a reference for the subsequent research on the treatment of ALD.


Subject(s)
Computational Biology , Drugs, Chinese Herbal , Liver Diseases, Alcoholic , Molecular Docking Simulation , Network Pharmacology , Protein Interaction Maps , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Network Pharmacology/methods , Humans , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Computational Biology/methods , Medicine, Chinese Traditional/methods , Gene Ontology
4.
Commun Biol ; 7(1): 756, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907105

ABSTRACT

Tuberous sclerosis complex 2 (TSC2) crucially suppresses Rheb activity to prevent mTORC1 activation. However, mutations in TSC genes lead to mTORC1 overactivation, thereby causing various developmental disorders and cancer. Therefore, the discovery of novel Rheb inhibitors is vital to prevent mTOR overactivation. Here, we reveals that the anti-inflammatory cytokine IL-37d can bind to lysosomal Rheb and suppress its activity independent of TSC2, thereby preventing mTORC1 activation. The binding of IL-37d to Rheb switch-II subregion destabilizes the Rheb-mTOR and mTOR-S6K interactions, further halting mTORC1 signaling. Unlike TSC2, IL-37d is reduced under ethanol stimulation, which results in mitigating the suppression of lysosomal Rheb-mTORC1 activity. Consequently, the recombinant human IL-37d protein (rh-IL-37d) with a TAT peptide greatly improves alcohol-induced liver disorders by hindering Rheb-mTORC1 axis overactivation in a TSC2- independent manner. Together, IL-37d emerges as a novel Rheb suppressor independent of TSC2 to terminate mTORC1 activation and improve abnormal lipid metabolism in the liver.


Subject(s)
Liver Diseases, Alcoholic , Mechanistic Target of Rapamycin Complex 1 , Ras Homolog Enriched in Brain Protein , Signal Transduction , Tuberous Sclerosis Complex 2 Protein , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Ras Homolog Enriched in Brain Protein/genetics , Humans , Animals , Mice , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/genetics , Interleukin-1/metabolism , Interleukin-1/genetics , Mice, Inbred C57BL , Male , HEK293 Cells
5.
Gut Microbes ; 16(1): 2367342, 2024.
Article in English | MEDLINE | ID: mdl-38889450

ABSTRACT

Alcohol-related liver disease (ALD) is recognized as a global health crisis, contributing to approximately 20% of liver cancer-associated fatalities. Dysbiosis of the gut microbiome is associated with the development of ALD, with the gut microbial metabolite urolithin A (UA) exhibiting a potential for alleviating liver symptoms. However, the protective efficacy of UA against ALD and its underlying mechanism mediated by microbiota remain elusive. In this study, we provide evidence demonstrating that UA effectively ameliorates alcohol-induced metabolic disorders and hepatic endoplasmic reticulum (ER) stress through a specific gut-microbiota-liver axis mediated by major urinary protein 1 (MUP1). Moreover, UA exhibited the potential to restore alcohol-induced dysbiosis of the intestinal microbiota by enriching the abundance of Bacteroides sartorii (B. sartorii), Parabacteroides distasonis (P. distasonis), and Akkermansia muciniphila (A. muciniphila), along with their derived metabolite propionic acid. Partial attenuation of the hepatoprotective effects exerted by UA was observed upon depletion of gut microbiota using antibiotics. Subsequently, a fecal microbiota transplantation (FMT) experiment was conducted to evaluate the microbiota-dependent effects of UA in ALD. FMT derived from mice treated with UA exhibited comparable efficacy to direct UA treatment, as it effectively attenuated ER stress through modulation of MUP1. It was noteworthy that strong associations were observed among the hepatic MUP1, gut microbiome, and metabolome profiles affected by UA. Intriguingly, oral administration of UA-enriched B. sartorii, P. distasonis, and A. muciniphila can enhance propionic acid production to effectively suppress ER stress via MUP1, mimicking UA treatment. Collectively, these findings elucidate the causal mechanism that UA alleviated ALD through the gut-microbiota-liver axis. This unique mechanism sheds light on developing novel microbiome-targeted therapeutic strategies against ALD.


Subject(s)
Coumarins , Endoplasmic Reticulum Stress , Gastrointestinal Microbiome , Liver Diseases, Alcoholic , Liver , Mice, Inbred C57BL , Gastrointestinal Microbiome/drug effects , Animals , Mice , Liver/metabolism , Liver/drug effects , Liver Diseases, Alcoholic/microbiology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Male , Endoplasmic Reticulum Stress/drug effects , Coumarins/pharmacology , Coumarins/metabolism , Dysbiosis/microbiology , Humans , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
6.
Biochem Pharmacol ; 225: 116334, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38824967

ABSTRACT

Alcoholic liver injury (ALI) stands as a prevalent affliction within the spectrum of complex liver diseases. Prolonged and excessive alcohol consumption can pave the way for liver fibrosis, cirrhosis, and even hepatocellular carcinoma. Recent findings have unveiled the protective role of proline serine-threonine phosphatase interacting protein 2 (PSTPIP2) in combating liver ailments. However, the role of PSTPIP2 in ALI remains mostly unknown. This study aimed to determine the expression profile of PSTPIP2 in ALI and to uncover the mechanism through which PSTPIP2 affects the survival and apoptosis of hepatocytes in ALI, using both ethyl alcohol (EtOH)-fed mice and an EtOH-induced AML-12 cell model. We observed a consistent decrease in PSTPIP2 expression both in vivo and in vitro. Functionally, we assessed the impact of PSTPIP2 overexpression on ALI by administering adeno-associated virus 9 (AAV9)-PSTPIP2 into mice. The results demonstrated that augmenting PSTPIP2 expression significantly shielded against liver parenchymal distortion and curbed caspase-dependent hepatocyte apoptosis in EtOH-induced ALI mice. Furthermore, enforcing PSTPIP2 expression reduced hepatocyte apoptosis in a stable PSTPIP2-overexpressing AML-12 cell line established through lentivirus-PSTPIP2 transfection in vitro. Mechanistically, this study also identified signal transducer and activator of transcription 3 (STAT3) as a direct signaling pathway regulated by PSTPIP2 in ALI. In conclusion, our findings provide compelling evidence that PSTPIP2 has a regulatory role in hepatocyte apoptosis via the STAT3 pathway in ALI, suggesting PSTPIP2 as a promising therapeutic target for ALI.


Subject(s)
Apoptosis , Mice, Inbred C57BL , STAT3 Transcription Factor , Animals , Apoptosis/physiology , Apoptosis/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Mice , Male , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/prevention & control , Ethanol/toxicity , Ethanol/administration & dosage , Hepatocytes/metabolism , Hepatocytes/pathology , Cell Line , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
7.
Food Funct ; 15(13): 7124-7135, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881239

ABSTRACT

Alcoholic liver injury has become a leading threat to human health, with complicated pathogenesis and limited therapeutic options. Our previous study showed that Musculus senhousei peptides (MSPs) exhibit protective potential against early-stage alcoholic liver injury, although the underlying mechanism is not yet clear. In this study, histopathological analysis, mRNA abundance of injury-associated biomarkers, the gut microbiota, and faecal metabolome were evaluated using a mouse model subjected to acute alcohol exposure, aiming to identify the mechanism by which MSP can alleviate alcoholic hepatotoxicity. The results showed that MSP intervention significantly ameliorated symptoms of liver injury (suppressed serum ALT increment, hepatic lipid accumulation, and neutrophil infiltration in liver tissue), and reversed the abnormal mRNA abundance of biomarkers associated with oxidative stress (iNOS), inflammation (TNF-α, IL-1ß, MCP-1, TNF-R1, and TLR4), and apoptosis (Bax and Casp. 3) in the liver. Moreover, MSP improved intestinal barrier function by increasing the expression of tight junction proteins (Claudin-1 and Claudin-3). Further analysis of faecal microbiota and metabolome revealed that MSP promoted the growth of tryptophan-metabolizing bacteria (Clostridiales, Alistipes, and Odoribacter), leading to increased production of indole derivatives (indole-3-lactic acid and N-acetyltryptophan). These results suggested that MSPs may alleviate alcohol-induced liver injury targeting the gut-liver axis, and could be an effective option for the prevention of alcoholic liver injury.


Subject(s)
Gastrointestinal Microbiome , Liver Diseases, Alcoholic , Liver , Mice, Inbred C57BL , Animals , Mice , Gastrointestinal Microbiome/drug effects , Male , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Liver/metabolism , Liver/drug effects , Peptides/pharmacology , Oxidative Stress/drug effects , Disease Models, Animal
8.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167259, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38796918

ABSTRACT

BACKGROUND: Alcohol-associated liver disease (ALD) is a leading cause of liver disease-related deaths worldwide. Unfortunately, approved medications for the treatment of this condition are quite limited. One promising candidate is the anthocyanin, Cyanidin-3-O-glucoside (C3G), which has been reported to protect mice against hepatic lipid accumulation, as well as fibrosis in different animal models. However, the specific effects and mechanisms of C3G on ALD remain to be investigated. EXPERIMENTAL APPROACH: In this report, a Gao-binge mouse model of ALD was used to investigate the effects of C3G on ethanol-induced liver injury. The mechanisms of these C3G effects were assessed using AML12 hepatocytes. RESULTS: C3G administration ameliorated ethanol-induced liver injury by suppressing hepatic oxidative stress, as well as through reducing hepatic lipid accumulation and inflammation. Mechanistically, C3G activated the AMPK pathway and enhanced mitophagy to eliminate damaged mitochondria, thus reducing mitochondria-derived reactive oxidative species in ethanol-challenged hepatocytes. CONCLUSIONS: The results of this study indicate that mitophagy plays a potentially important role underlying the hepatoprotective action of C3G, as demonstrated in a Gao-binge mouse model of ALD. Accordingly, C3G may serve as a promising, new therapeutic drug candidate for use in ALD.


Subject(s)
Anthocyanins , Disease Models, Animal , Ethanol , Glucosides , Liver Diseases, Alcoholic , Mitophagy , Oxidative Stress , Animals , Anthocyanins/pharmacology , Mitophagy/drug effects , Mice , Glucosides/pharmacology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/prevention & control , Ethanol/toxicity , Ethanol/adverse effects , Oxidative Stress/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Mice, Inbred C57BL , Liver/metabolism , Liver/drug effects , Liver/pathology , Reactive Oxygen Species/metabolism , Lipid Metabolism/drug effects
9.
J Biomed Sci ; 31(1): 54, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38790021

ABSTRACT

BACKGROUND: Alcohol-related liver disease (ALD) is a major health concern worldwide, but effective therapeutics for ALD are still lacking. Tumor necrosis factor-inducible gene 6 protein (TSG-6), a cytokine released from mesenchymal stem cells, was shown to reduce liver fibrosis and promote successful liver repair in mice with chronically damaged livers. However, the effect of TSG-6 and the mechanism underlying its activity in ALD remain poorly understood. METHODS: To investigate its function in ALD mice with fibrosis, male mice chronically fed an ethanol (EtOH)-containing diet for 9 weeks were treated with TSG-6 (EtOH + TSG-6) or PBS (EtOH + Veh) for an additional 3 weeks. RESULTS: Severe hepatic injury in EtOH-treated mice was markedly decreased in TSG-6-treated mice fed EtOH. The EtOH + TSG-6 group had less fibrosis than the EtOH + Veh group. Activation of cluster of differentiation 44 (CD44) was reported to promote HSC activation. CD44 and nuclear CD44 intracellular domain (ICD), a CD44 activator which were upregulated in activated HSCs and ALD mice were significantly downregulated in TSG-6-exposed mice fed EtOH. TSG-6 interacted directly with the catalytic site of MMP14, a proteolytic enzyme that cleaves CD44, inhibited CD44 cleavage to CD44ICD, and reduced HSC activation and liver fibrosis in ALD mice. In addition, a novel peptide designed to include a region that binds to the catalytic site of MMP14 suppressed CD44 activation and attenuated alcohol-induced liver injury, including fibrosis, in mice. CONCLUSIONS: These results demonstrate that TSG-6 attenuates alcohol-induced liver damage and fibrosis by blocking CD44 cleavage to CD44ICD and suggest that TSG-6 and TSG-6-mimicking peptide could be used as therapeutics for ALD with fibrosis.


Subject(s)
Cell Adhesion Molecules , Hyaluronan Receptors , Liver Cirrhosis , Liver Diseases, Alcoholic , Animals , Male , Mice , Cell Adhesion Molecules/administration & dosage , Ethanol , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/chemically induced , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/drug therapy , Mice, Inbred C57BL , Peptides/pharmacology , Peptides/metabolism
10.
Int J Biol Macromol ; 270(Pt 1): 132093, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710247

ABSTRACT

Long-term and excessive alcohol consumption can lead to the development of alcoholic liver disease (ALD), characterized by oxidative damage, intestinal barrier injury, and disruption of intestinal microbiota. In this study, we extracted fucoidan (Aj-FUC) from Apostichopus japonicus using enzymatic methods and characterized its structure. The ALD model was established in male Balb/c mice using 56° Baijiu, with silymarin as a positive control. Mice were orally administered 100 mg/kg·bw and 300 mg/kg·bw of Aj-FUC for 28 days to evaluate its effects on liver injury in ALD mice and explore its potential role in modulating the gut-liver axis. The results showed significant improvements in histopathological changes and liver disease in the Aj-FUC group. Aj-FUC treatment significantly increased the levels of glutathione (GSH) and glutathione peroxidase (GSH-Px) while weakly reduced the elevation of malondialdehyde (MDA) induced by ALD. It also regulated the Nrf2/HO-1 signaling pathway, collectively alleviating hepatic oxidative stress. Aj-FUC intervention upregulated the expression of ZO-1 and Occludin, thus contributing to repair the intestinal barrier. Additionally, Aj-FUC increased the content of short-chain fatty acids (SCFAs) and regulated the imbalance in gut microbiota. These results suggested that Aj-FUC alleviates ALD by modulating the gut-liver axis homeostasis. It may prove to be a useful dietary supplement in the treatment of alcoholic liver damage.


Subject(s)
Homeostasis , Liver Diseases, Alcoholic , Liver , Oxidative Stress , Polysaccharides , Stichopus , Animals , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Polysaccharides/pharmacology , Polysaccharides/chemistry , Mice , Male , Liver/drug effects , Liver/metabolism , Liver/pathology , Homeostasis/drug effects , Oxidative Stress/drug effects , Stichopus/chemistry , Mice, Inbred BALB C , Malondialdehyde/metabolism , Gastrointestinal Microbiome/drug effects , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Disease Models, Animal , Glutathione Peroxidase/metabolism
11.
Cell Rep Methods ; 4(5): 100778, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38749443

ABSTRACT

Alcohol-associated liver disease (ALD) is a prevalent liver disease, yet research is hampered by the lack of suitable and reliable human ALD models. Herein, we generated human adipose stromal/stem cell (hASC)-derived hepatocellular organoids (hAHOs) and hASC-derived liver organoids (hALOs) in a three-dimensional system using hASC-derived hepatocyte-like cells and endodermal progenitor cells, respectively. The hAHOs were composed of major hepatocytes and cholangiocytes. The hALOs contained hepatocytes and nonparenchymal cells and possessed a more mature liver function than hAHOs. Upon ethanol treatment, both steatosis and inflammation were present in hAHOs and hALOs. The incubation of hALOs with ethanol resulted in increases in the levels of oxidative stress, the endoplasmic reticulum protein thioredoxin domain-containing protein 5 (TXNDC5), the alcohol-metabolizing enzymes ADH1B and ALDH1B1, and extracellular matrix accumulation, similar to those of liver tissues from patients with ALD. These results present a useful approach for understanding the pathogenesis of ALD in humans, thus facilitating the discovery of effective treatments.


Subject(s)
Adipose Tissue , Ethanol , Hepatocytes , Liver Diseases, Alcoholic , Organoids , Humans , Organoids/pathology , Organoids/drug effects , Ethanol/pharmacology , Ethanol/adverse effects , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/metabolism , Adipose Tissue/pathology , Adipose Tissue/cytology , Alcohol Dehydrogenase/metabolism , Oxidative Stress/drug effects , Liver/pathology , Liver/drug effects , Liver/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Models, Biological , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Stromal Cells/pathology , Stromal Cells/drug effects , Stromal Cells/metabolism , Thioredoxins/metabolism
12.
Free Radic Biol Med ; 220: 236-248, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38704052

ABSTRACT

Alcoholic liver disease (ALD) is a common chronic redox disease caused by increased alcohol consumption. Abstinence is a major challenge for people with alcohol dependence, and approved drugs have limited efficacy. Therefore, this study aimed to explore a new treatment strategy for ALD using ferroferric oxide endohedral fullerenol (Fe3O4@C60(OH)n) in combination with static magnetic and electric fields (sBE). The primary hepatocytes of 8-9-week-old female BALB/c mice were used to evaluate the efficacy of the proposed combination treatment. A mouse chronic binge ethanol feeding model was established to determine the alleviatory effect of Fe3O4@C60(OH)n on liver injury under sBE exposure. Furthermore, the ability of Fe3O4@C60(OH)n to eliminate •OH was evaluated. Alcohol-induced hepatocyte and mitochondrial damage were reversed in vitro. Additionally, the combination therapy reduced liver damage, alleviated oxidative stress by improving antioxidant levels, and effectively inhibited liver lipid accumulation in animal experiments. Here, we used a combination of magnetic derivatives of fullerenol and sBE to further improve the ROS clearance rate, thereby alleviating ALD. The developed combination treatment may effectively improve alcohol-induced liver damage and maintain redox balance without apparent toxicity, thereby enhancing therapy aimed at ALD and other redox diseases.


Subject(s)
Fullerenes , Hepatocytes , Liver Diseases, Alcoholic , Mice, Inbred BALB C , Oxidative Stress , Reactive Oxygen Species , Animals , Fullerenes/pharmacology , Fullerenes/chemistry , Fullerenes/therapeutic use , Mice , Reactive Oxygen Species/metabolism , Female , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Oxidative Stress/drug effects , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/drug therapy , Liver/metabolism , Liver/pathology , Liver/drug effects , Antioxidants/pharmacology , Disease Models, Animal , Humans , Oxidation-Reduction/drug effects , Ethanol/toxicity
13.
Clin Sci (Lond) ; 138(7): 435-487, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38571396

ABSTRACT

Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.


Subject(s)
Liver Diseases, Alcoholic , Non-alcoholic Fatty Liver Disease , Sepsis , Humans , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Liver Cirrhosis/metabolism , Liver Diseases, Alcoholic/metabolism , Sepsis/complications
14.
Biomed Pharmacother ; 174: 116595, 2024 May.
Article in English | MEDLINE | ID: mdl-38640709

ABSTRACT

Fatty liver is the earliest response of the liver to excessive alcohol consumption. Previously we identified that chronic alcohol administration increases levels of stomach-derived hormone, ghrelin, which by reducing circulating insulin levels, ultimately contributes to the development of alcohol-associated liver disease (ALD). In addition, ghrelin directly promotes fat accumulation in hepatocytes by enhancing de novo lipogenesis. Other than promoting ALD, ghrelin is known to increase alcohol craving and intake. In this study, we used a ghrelin receptor (GHSR) knockout (KO) rat model to characterize the specific contribution of ghrelin in the development of ALD with emphasis on energy homeostasis. Male Wistar wild type (WT) and GHSR-KO rats were pair-fed the Lieber-DeCarli control or ethanol diet for 6 weeks. At the end of the feeding period, glucose tolerance test was conducted, and tissue samples were collected. We observed reduced alcohol intake by GHSR-KOs compared to a previous study where WT rats were fed ethanol diet ad libitum. Further, when the WTs were pair-fed to GHSR-KOs, the KO rats exhibited resistance to develop ALD through improving insulin secretion/sensitivity to reduce adipose lipolysis and hepatic fatty acid uptake/synthesis and increase fatty acid oxidation. Furthermore, proteomic data revealed that ethanol-fed KO exhibit less alcohol-induced mitochondrial dysfunction and oxidative stress than WT rats. Proteomic data also confirmed that the ethanol-fed KOs are insulin sensitive and are resistant to hepatic steatosis development compared to WT rats. Together, these data confirm that inhibiting ghrelin action prevent alcohol-induced liver and adipose dysfunction independent of reducing alcohol intake.


Subject(s)
Ethanol , Ghrelin , Liver Diseases, Alcoholic , Liver , Rats, Wistar , Receptors, Ghrelin , Animals , Male , Rats , Alcohol Drinking , Fatty Acids/metabolism , Ghrelin/metabolism , Insulin/metabolism , Insulin/blood , Insulin Resistance , Liver/metabolism , Liver/drug effects , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Oxidative Stress/drug effects , Proteomics/methods , Receptors, Ghrelin/metabolism , Receptors, Ghrelin/genetics
15.
Biomolecules ; 14(4)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38672422

ABSTRACT

Alcohol-associated liver disease (ALD) is a substantial cause of morbidity and mortality worldwide and represents a spectrum of liver injury beginning with hepatic steatosis (fatty liver) progressing to inflammation and culminating in cirrhosis. Multiple factors contribute to ALD progression and disease severity. Here, we overview several crucial mechanisms related to ALD end-stage outcome development, such as epigenetic changes, cell death, hemolysis, hepatic stellate cells activation, and hepatic fatty acid binding protein 4. Additionally, in this review, we also present two clinically relevant models using human precision-cut liver slices and hepatic organoids to examine ALD pathogenesis and progression.


Subject(s)
Disease Progression , Liver Diseases, Alcoholic , Humans , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Animals , Liver/metabolism , Liver/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Epigenesis, Genetic
16.
Anal Biochem ; 691: 115534, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38621605

ABSTRACT

Xing 9 Ling tablet candy (X9LTC) effectively treats alcoholic liver disease (ALD), but its potential mechanism and molecular targets remain unstudied. We aimed to address this gap using network pharmacology. Furthermore, high-performance liquid chromatography (HPLC) and database analysis revealed a total of 35 active ingredients and 311 corresponding potential targets of X9LTC. Protein interaction analysis revealed PTGS2, JUN, and FOS as its core targets. Enrichment analysis indicated that chemical carcinogenesis-receptor activation, IL-17 and TNF signaling pathway were enriched by multiple core targets, which might be the main pathway of action. Further molecular docking validation showed that the core targets had good binding activities with the identified compounds. Animal experiments showed that X9LTC could reduce the high expression of ALT, AST and TG in the serum of ALD mice, alleviate the lesions in liver tissues, and reverse the high expression of PTGS2, JUN, and FOS proteins in the liver tissues. In this study, we established a method for the determination of X9LTC content for the first time, and predicted its active ingredient and mechanism of action in treating ALD, providing theoretical basis for further research.


Subject(s)
Drugs, Chinese Herbal , Liver Diseases, Alcoholic , Molecular Docking Simulation , Network Pharmacology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/drug therapy , Animals , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Male , Tablets , Cyclooxygenase 2/metabolism , Mice, Inbred C57BL , Chromatography, High Pressure Liquid , Liver/metabolism , Liver/drug effects
17.
Elife ; 122024 Apr 22.
Article in English | MEDLINE | ID: mdl-38648183

ABSTRACT

Recent genome-wide association studies (GWAS) have identified a link between single-nucleotide polymorphisms (SNPs) near the MBOAT7 gene and advanced liver diseases. Specifically, the common MBOAT7 variant (rs641738) associated with reduced MBOAT7 expression is implicated in non-alcoholic fatty liver disease (NAFLD), alcohol-associated liver disease (ALD), and liver fibrosis. However, the precise mechanism underlying MBOAT7-driven liver disease progression remains elusive. Previously, we identified MBOAT7-driven acylation of lysophosphatidylinositol lipids as key mechanism suppressing the progression of NAFLD (Gwag et al., 2019). Here, we show that MBOAT7 loss of function promotes ALD via reorganization of lysosomal lipid homeostasis. Circulating levels of MBOAT7 metabolic products are significantly reduced in heavy drinkers compared to healthy controls. Hepatocyte- (Mboat7-HSKO), but not myeloid-specific (Mboat7-MSKO), deletion of Mboat7 exacerbates ethanol-induced liver injury. Lipidomic profiling reveals a reorganization of the hepatic lipidome in Mboat7-HSKO mice, characterized by increased endosomal/lysosomal lipids. Ethanol-exposed Mboat7-HSKO mice exhibit dysregulated autophagic flux and lysosomal biogenesis, associated with impaired transcription factor EB-mediated lysosomal biogenesis and autophagosome accumulation. This study provides mechanistic insights into how MBOAT7 influences ALD progression through dysregulation of lysosomal biogenesis and autophagic flux, highlighting hepatocyte-specific MBOAT7 loss as a key driver of ethanol-induced liver injury.


Subject(s)
Acyltransferases , Homeostasis , Lipid Metabolism , Liver Diseases, Alcoholic , Lysosomes , Membrane Proteins , Animals , Humans , Male , Mice , Acyltransferases/genetics , Acyltransferases/metabolism , Hepatocytes/metabolism , Liver/metabolism , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/genetics , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Knockout
18.
J Pharm Biomed Anal ; 245: 116157, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38636192

ABSTRACT

Penthorum chinense Pursh (PCP), as a traditional medicine of Miao nationality in China, is often used for the treatment of various liver diseases. At present, information regarding the in vivo process of PCP is lacking. Herein, a sensitive and robust ultra-performance liquid chromatography tandem with mass spectrometry (UPLC-MS/MS) was developed and validated for the quantification of several components to study their pharmacokinetics, tissues distribution and excretion in normal and acute alcoholic liver injury (ALI) rats. Prepared samples were separated on a Thermo C18 column (4.6 mm × 50 mm, 2.4 µm) using water containing 0.1 % formic acid (A) and acetonitrile (B) as the mobile phase for gradient elution. Negative electrospray ionization was performed using multiple reaction monitoring (MRM) mode for each component. The validated UPLC-MS/MS assay gave good linearity, accuracy, precision, recovery rate, matrix effect and stability. This method was successfully applied to the pharmacokinetics, tissue distribution and excretion in normal and acute ALI rats. There were differences in pharmacokinetic process, tissue distribution and excretion characteristics, indicating that ALI had a significant influence on the in vivo process of PCP in rats. The research provided an experimental basis for the study of PCP quality control and further application in the clinic.


Subject(s)
Drugs, Chinese Herbal , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Animals , Tandem Mass Spectrometry/methods , Chromatography, High Pressure Liquid/methods , Rats , Male , Drugs, Chinese Herbal/pharmacokinetics , Tissue Distribution , Reproducibility of Results , Liver Diseases, Alcoholic/metabolism , Liquid Chromatography-Mass Spectrometry
19.
Food Chem ; 451: 139337, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38663243

ABSTRACT

Alcoholic liver disease (ALD) is a serious health threat. Soybean meal peptide (SMP) supplementation may protect against this damage; however, the potential mechanism underlying the specific sequence of SMPs is unclear. Protein-protein interaction and proteomic analyses are effective methods for studying functional ingredients in diseases. This study aimed to investigate the potential mechanism of action of the peptide Gly-Thr-Tyr-Trp (GTYW) on ALD using protein-protein interaction and proteomic analyses. These results demonstrate that GTYW influenced the targets of glutathione metabolism (glutathione-disulfide reductase, glutathione S-transferase pi 1, and glutathione S-transferase mu 2). It also regulated the expression of targets related to energy metabolism and amino acid conversion (trypsin-2, cysteine dioxygenase type-1, and F6SJM7). Amino acid and lipid metabolisms were identified based on Gene Ontology annotation. These results indicate that GTYW might affect alcohol-related liver disease signaling pathways. This study provides evidence of the protective and nutritional benefits of SMPs in ALD treatment.


Subject(s)
Glycine max , Liver Diseases, Alcoholic , Peptides , Proteomics , Animals , Mice , Glycine max/chemistry , Glycine max/metabolism , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/genetics , Male , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism , Peptides/administration & dosage , Humans , Mice, Inbred C57BL , Protective Agents/pharmacology , Protective Agents/administration & dosage , Protective Agents/chemistry , Liver/metabolism
20.
Environ Health Perspect ; 132(4): 47007, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38619879

ABSTRACT

BACKGROUND: Environmental pollutants, including polychlorinated biphenyls (PCBs) have been implicated in the pathogenesis of liver disease. Our group recently demonstrated that PCB126 promoted steatosis, hepatomegaly, and modulated intermediary metabolism in a rodent model of alcohol-associated liver disease (ALD). OBJECTIVE: To better understand how PCB126 promoted ALD in our previous model, the current study adopts multiple omics approaches to elucidate potential mechanistic hypotheses. METHODS: Briefly, male C57BL/6J mice were exposed to 0.2mg/kg polychlorinated biphenyl (PCB) 126 or corn oil vehicle prior to ethanol (EtOH) or control diet feeding in the chronic-binge alcohol feeding model. Liver tissues were collected and prepared for mRNA sequencing, phosphoproteomics, and inductively coupled plasma mass spectrometry for metals quantification. RESULTS: Principal component analysis showed that PCB126 uniquely modified the transcriptome in EtOH-fed mice. EtOH feeding alone resulted in >4,000 differentially expressed genes (DEGs), and PCB126 exposure resulted in more DEGs in the EtOH-fed group (907 DEGs) in comparison with the pair-fed group (503 DEGs). Top 20 significant gene ontology (GO) biological processes included "peptidyl tyrosine modifications," whereas top 25 significantly decreasing GO molecular functions included "metal/ion/zinc binding." Quantitative, label-free phosphoproteomics and western blot analysis revealed no major significant PCB126 effects on total phosphorylated tyrosine residues in EtOH-fed mice. Quantified hepatic essential metal levels were primarily significantly lower in EtOH-fed mice. PCB126-exposed mice had significantly lower magnesium, cobalt, and zinc levels in EtOH-fed mice. DISCUSSION: Previous work has demonstrated that PCB126 is a modifying factor in metabolic dysfunction-associated steatotic liver disease (MASLD), and our current work suggests that pollutants also modify ALD. PCB126 may, in part, be contributing to the malnutrition aspect of ALD, where metal deficiency is known to contribute and worsen prognosis. https://doi.org/10.1289/EHP14132.


Subject(s)
Environmental Pollutants , Fatty Liver , Liver Diseases, Alcoholic , Polychlorinated Biphenyls , Male , Mice , Animals , Multiomics , Mice, Inbred C57BL , Ethanol/toxicity , Ethanol/metabolism , Liver/metabolism , Polychlorinated Biphenyls/toxicity , Polychlorinated Biphenyls/metabolism , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Environmental Pollutants/toxicity , Environmental Pollutants/metabolism , Zinc/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...