1
|
Dong L, Zhang H, Kang Y, Wang F, Bai T, Yang Y. NLRP3 and Gut-Liver Axis: New Possibility for the Treatment of Alcohol-Associated Liver Disease. J Gastroenterol Hepatol 2025; 40:1070-1078. [PMID: 40091479 DOI: 10.1111/jgh.16935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/04/2025] [Accepted: 03/01/2025] [Indexed: 03/19/2025]
Abstract
Alcohol-associated liver disease (ALD) is one of the most prevalent chronic diseases worldwide, with persistently high morbidity and mortality rates. Previous studies have identified NLRP3 inflammasome as a class of receptors of intracellular intrinsic immunity. These receptors can be activated by both intrinsic and extracellular danger signals, leading to the release of downstream pro-inflammatory factors, including interleukin IL-1β and IL-18. These vesicles are critical for maintaining host defense. Concurrently, researchers have identified a close relationship between the microbiome, gut-liver axis, and NLRP3 inflammasome with ALD. Consequently, the present study focus on the structure and activation of the NLRP3 inflammasome, the gut-liver axis, and intestinal microecological regulation, as well as the relationship between bile acid metabolism and the gut-liver axis. The objective of this study is to provide a foundation of knowledge and references for the development of targeted therapeutic interventions of ALD that are informed by the dynamic interplay between the NLRP3 inflammasome and the gut-liver axis.
Collapse
Affiliation(s)
- Lu Dong
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| | - Haotian Zhang
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| | - Yanyu Kang
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| | - Fei Wang
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| | - Ting Bai
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| | - Yong Yang
- Dalian key Laboratory of Chronic Disease Research Center, Dalian University, Dalian, Liaoning Province, China
| |
Collapse
|
2
|
Wolf PG, Welsh C, Binion B, Dai H, Oliveira ML, Hamm A, Goldberg S, Buobu PS, Schering T, Vergis S, Kessee N, Gomez SL, Yazici C, Maienschein-Cline M, Byrd DA, Gaskins HR, Ridlon JM, Mutlu E, Greening C, Tussing-Humphreys L. Secondary Bile Acid Derivatives Are Contributors to the Fecal Bile Acid Pool and Associated With Bile Acid-Modulating Nutrients. J Nutr 2025; 155:826-838. [PMID: 39805403 PMCID: PMC11934243 DOI: 10.1016/j.tjnut.2024.12.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Accumulation of hydrophobic bile acids (BAs) is linked with cancer development. However, derivatives of deoxycholic acid (DCA) and lithocholic acid (LCA) produced via bacterial metabolism may mitigate the proinflammatory and cytotoxic effects of hydrophobic BAs. The impact of diet on secondary BA derivative production has not been determined. OBJECTIVES This study aimed to study the associations between BA-modulating nutrients and the composition of secondary BAs and their derivatives. METHODS Stool and blood were collected from 138 participants aged 45-75 y that self-identified as Black or non-Hispanic White. BAs were extracted from stool and serum and quantified using LC/ESI-MS/MS. Energy, macronutrients, micronutrients, and specific dietary nutrients were estimated from two 24-h diet recalls. The abundance of genes for microbial BA metabolism was assessed from stool metagenomes. Kendall τ correlation and regression-based modeling were performed to determine associations between BA categories, microbial genes, and select energy-adjusted dietary variables (alcohol, calcium, coffee, fiber, fat, and protein). RESULTS Participants had a mean age of 60 y and a mean BMI of 31 kg/m2. BA derivatives were present in all participant stools, with lagodeoxycholic acid being the most abundant derivative quantified. Analysis of stool microbial metagenomes revealed the presence of genes for secondary BA derivative production in all participants. Protein is positively associated with the accumulation of secondary BAs. monounsaturated fatty acids (MUFA)s were negatively associated with high abundant derivatives of DCA in regression models. Total fiber and coffee intake were positively correlated with increased conversion of BAs to derivatives. Race and smoking status were significant predictors of associations between dietary variables and BA derivatives. CONCLUSION Protein, MUFAs, total fiber and coffee are significantly associated with concentrations of secondary BAs and their derivatives. Future work should account for social and structural influences on dietary intake and its relationship with BA-elicited cancer risk.
Collapse
Affiliation(s)
- Patricia G Wolf
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States; Institute for Cancer Research, Purdue University, West Lafayette, IN, United States.
| | - Caitlin Welsh
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Briawna Binion
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Hanchu Dai
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
| | - Manoela Lima Oliveira
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Alyshia Hamm
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Sarah Goldberg
- Department of Clinical Nutrition, Rush University, Chicago, IL, United States
| | - Pius Sarfo Buobu
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States; Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Teresa Schering
- University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Sevasti Vergis
- University of Illinois Cancer Center, Chicago, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Nicollette Kessee
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States
| | - Sandra L Gomez
- Department of Clinical Nutrition, Rush University, Chicago, IL, United States
| | - Cemal Yazici
- Department of Medicine, University of Illinois Chicago, Chicago, IL
| | | | - Doratha A Byrd
- Department of Cancer Epidemiology, Division of Population Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Ece Mutlu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Chris Greening
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States.
| |
Collapse
|
3
|
Shukla S, Hsu CL. Alcohol Use Disorder and the Gut-Brain Axis: A Narrative Review of the Role of Gut Microbiota and Implications for Treatment. Microorganisms 2025; 13:67. [PMID: 39858835 PMCID: PMC11767426 DOI: 10.3390/microorganisms13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/21/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Alcohol use disorder (AUD) affects millions of people worldwide and can lead to deleterious physical and social consequences. Recent research has highlighted not only the effect of alcohol on the gut microbiome, but also the role of the gut microbiome and the gut-brain axis in the development and maintenance of alcohol use disorder. This review provides an overview of the reciprocal relationship between alcohol consumption and the gut microbiome, including the effects of alcohol on gut microbial composition, changes in gut microbial metabolites in response to alcohol consumption, and how gut microbial metabolites may modulate alcohol use behavior. We also discuss the gut-mediated mechanisms of neuroinflammation that contribute to and result from AUD, including disruption of the intestinal barrier, toll-like receptor signaling, and the activation of glial cells and immune cells. Finally, we review the current evidence on gut microbial-directed therapies for AUD and discuss the implications of this research for our understanding of the pathophysiology of AUD and future research directions.
Collapse
Affiliation(s)
- Shikha Shukla
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Cynthia L. Hsu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
4
|
Wang L, Zheng W, Sun Y, Ren X, Yan C, Song S, Ai C. Fucoidan ameliorates alcohol-induced liver injury in mice through Parabacteroides distasonis-mediated regulation of the gut-liver axis. Int J Biol Macromol 2024; 279:135309. [PMID: 39236962 DOI: 10.1016/j.ijbiomac.2024.135309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Polysaccharides can benefit the liver via modulation of the gut microbiota, but the exact mechanism is still unclear. This study demonstrated that the effect of Scytosiphon lomentaria fucoidan (SLF) on alcohol-induced liver injury can be closely related to the level of Parabacteroides distasonis (Pd) via in vivo and in vitro models. Further mice experiment showed that Pd alleviated liver injury and inflammation by suppressing the NF-κB/MAPK pathways and activating Nrf2 pathway. The underlying mechanism can be closely associated with modulation of the gut microbiota, particularly an increase in microbiota diversity and beneficial bacteria and a reduction in Proteobacteria. Targeted metabolomics indicated that Pd ameliorated alcohol-induced dysbiosis of microbiota metabolites profile, primarily affecting amino acid metabolism. Moreover, Pd reduced the level of total bile acids (BAs) and improved BAs profile, affecting the expression levels of BA-associated genes in the liver and ileum involved in BA synthesis, transport, and reabsorption. This study suggests that SLF can benefit alcohol-induced liver injury via P. distasonis-mediated regulation of the gut-liver axis.
Collapse
Affiliation(s)
- Lu Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong 037009, PR China
| | - Yiyun Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunhong Yan
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
5
|
Sun Y, Men Q, Ren X, Yan C, Song S, Ai C. Low molecular fucoidan alleviated alcohol-induced liver injury in BALB/c mice by regulating the gut microbiota-bile acid-liver axis. Int J Biol Macromol 2024; 282:136930. [PMID: 39490864 DOI: 10.1016/j.ijbiomac.2024.136930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Fucoidan has attracted significant attention owing to its remarkable bioactivities, but the effect of molecular weight (Mw) on its activities in the context of alcoholic liver diseases (ALD) is poorly understood. In this study, low Mw fucoidan (OSLF) was prepared, and its protective effect against alcohol-induced liver injury was assessed in a mouse model. OSLF increased weight gain and colon length, improved lipid disorders, and reduced oxidative stress in mice exposed to alcohol, alleviating liver injury. OSLF alleviated inflammation in the liver by inhibiting alcohol-activated NF-κB and MAPK pathways. The underlying mechanism can be attributed to the improvement of alcohol-induced dysbiosis of the gut microbiota, including a decrease in Proteobacteria and Bacteroidetes and an increase in microbiota diversity, as well as the abundances of Parabacteroides, Bacteroides, and Faecalibaculum. Metabolomics results showed that OSLF improved alcohol-induced abnormalities of microbiota metabolites, primarily involving amino acid metabolism and short chain fatty acids production. In addition, OSLF ameliorated bile acid metabolism in the gut and regulated the expression of bile acid-associated genes in the liver, affecting bile acid synthesis, regulation, and transport. It suggested that OSLF had the potential for the management of ALD by regulating the gut microbiota-bile acid-liver axis.
Collapse
Affiliation(s)
- Yiyun Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunhong Yan
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
6
|
Zhou H, Wang X, She Z, Huang L, Wei H, Yang S, Wei Z, Chen H, Yang B, Hu Z, Feng X, Zhu P, Li Z, Shen J, Liu H, Dong H, Chen G, Zhang Q. Combining bioinformatics and multiomics strategies to investigate the key microbiota and active components of Liupao tea ameliorating hyperlipidemia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118438. [PMID: 38848972 DOI: 10.1016/j.jep.2024.118438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hyperlipidemia as a major health issue has attracted much public attention. As a geographical indication product of China, Liupao tea (LPT) is a typical representative of traditional Chinese dark tea that has shown good potential in regulating glucose and lipid metabolism. LPT has important medicinal value in hyperlipidemia prevention. However, the active ingredients and metabolic mechanisms by which LPT alleviates hyperlipidemia remain unclear. AIM OF THE STUDY This study aimed to systematically investigate the metabolic mechanisms and active ingredients of LPT extract in alleviating hyperlipidemia. MATERIALS AND METHODS Firstly, we developed a mouse model of hyperlipidemia to study the pharmacodynamics of LPT. Subsequently, network pharmacology and molecular docking were performed to predict the potential key active ingredients and core targets of LPT against hyperlipidemia. LC-MS/MS was used to validate the identity of key active ingredients in LPT with chemical standards. Finally, the effect and metabolic mechanisms of LPT extract in alleviating hyperlipidemia were investigated by integrating metabolomic, lipidomic, and gut microbiome analyses. RESULTS Results showed that LPT extract effectively improved hyperlipidemia by suppressing weight gain, remedying dysregulation of glucose and lipid metabolism, and reducing hepatic damage. Network pharmacology analysis and molecular docking suggested that four potential active ingredients and seven potential core targets were closely associated with roles for hyperlipidemia treatment. Ellagic acid, catechin, and naringenin were considered to be the key active ingredients of LPT alleviating hyperlipidemia. Additionally, LPT extract modulated the mRNA expression levels of Fxr, Cyp7a1, Cyp8b1, and Cyp27a1 associated with bile acid (BA) metabolism, mitigated the disturbances of BA and glycerophospholipid (GP) metabolism in hyperlipidemia mice. Combining fecal microbiota transplantation and correlation analysis, LPT extract effectively improved species diversity and abundance of gut microbiota, particularly the BA and GP metabolism-related gut microbiota, in the hyperlipidemia mice. CONCLUSIONS LPT extract ameliorated hyperlipidemia by modulating GP and BA metabolism by regulating Lactobacillus and Dubosiella, thereby alleviating hyperlipidemia. Three active ingredients of LPT served as the key factors in exerting an improvement on hyperlipidemia. These findings provide new insights into the active ingredients and metabolic mechanisms of LPT in improving hyperlipidemia, suggesting that LPT can be used to prevent and therapeutic hyperlipidemia.
Collapse
Affiliation(s)
- Hailin Zhou
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Xuancheng Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Zhiyong She
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Li Huang
- College of Light Industry and Food Engineering, Guangxi University, Guangxi, China.
| | - Huijie Wei
- College of Light Industry and Food Engineering, Guangxi University, Guangxi, China.
| | - Shanyi Yang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Zhijuan Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Hongwei Chen
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Bao Yang
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China.
| | - Zehua Hu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China.
| | - Xue Feng
- Center for Instrumental Analysis, Guangxi University, Guangxi, China.
| | - Pingchuan Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Guangxi, China.
| | - Zijian Li
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Jiahui Shen
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Huan Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Huanxiao Dong
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Guanghui Chen
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Guangxi, China.
| | - Qisong Zhang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China; Center for Instrumental Analysis, Guangxi University, Guangxi, China.
| |
Collapse
|
7
|
Guo W, Zhong W, He L, Wei X, Hao L, Dong H, Yue R, Sun X, Yin X, Zhao J, Zhang X, Zhou Z. Reversal of hepatic accumulation of nordeoxycholic acid underlines the beneficial effects of cholestyramine on alcohol-associated liver disease in mice. Hepatol Commun 2024; 8:e0507. [PMID: 39082957 DOI: 10.1097/hc9.0000000000000507] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/31/2024] [Indexed: 08/30/2024] Open
Abstract
BACKGROUND Dysregulation of bile acids (BAs) has been reported in alcohol-associated liver disease. However, the causal relationship between BA dyshomeostasis and alcohol-associated liver disease remains unclear. The study aimed to determine whether correcting BA perturbation protects against alcohol-associated liver disease and elucidate the underlying mechanism. METHODS BA sequestrant cholestyramine (CTM) was administered to C57BL/6J mice fed alcohol for 8 weeks to assess its protective effect and explore potential BA targets. The causal relationship between identified BA metabolite and cellular damage was examined in hepatocytes, with further manipulation of the detoxifying enzyme cytochrome p450 3A11. The toxicity of the BA metabolite was further validated in mice in an acute study. RESULTS We found that CTM effectively reversed hepatic BA accumulation, leading to a reversal of alcohol-induced hepatic inflammation, cell death, endoplasmic reticulum stress, and autophagy dysfunction. Specifically, nordeoxycholic acid (NorDCA), a hydrophobic BA metabolite, was identified as predominantly upregulated by alcohol and reduced by CTM. Hepatic cytochrome p450 3A11 expression was in parallel with NorDCA levels, being upregulated by alcohol and reduced by CTM. Moreover, CTM reversed alcohol-induced gut barrier disruption and endotoxin translocation. Mechanistically, NorDCA was implicated in causing endoplasmic reticulum stress, suppressing autophagy flux, and inducing cell injury, and such deleterious effects could be mitigated by cytochrome p450 3A11 overexpression. Acute NorDCA administration in mice significantly induced hepatic inflammation and injury along with disrupting gut barrier integrity, leading to subsequent endotoxemia. CONCLUSIONS Our study demonstrated that CTM treatment effectively reversed alcohol-induced liver injury in mice. The beneficial effects of BA sequestrant involve lowering toxic NorDCA levels. NorDCA not only worsens hepatic endoplasmic reticulum stress and inhibits autophagy but also mediates gut barrier disruption and systemic translocation of pathogen-associated molecular patterns in mice.
Collapse
Affiliation(s)
- Wei Guo
- Center for Translational Biomedical Research
| | - Wei Zhong
- Center for Translational Biomedical Research
- Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, USA
| | - Liqing He
- Department of Chemistry, University of Louisville, Louisville, Kentucky, USA
| | - Xiaoyuan Wei
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Liuyi Hao
- Center for Translational Biomedical Research
| | - Haibo Dong
- Center for Translational Biomedical Research
| | - Ruichao Yue
- Center for Translational Biomedical Research
| | - Xinguo Sun
- Center for Translational Biomedical Research
| | - Xinmin Yin
- Department of Chemistry, University of Louisville, Louisville, Kentucky, USA
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, Kentucky, USA
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research
- Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, USA
| |
Collapse
|
8
|
Wang Y, Zou Z, Wang S, Ren A, Ding Z, Li Y, Wang Y, Qian Z, Bian B, Huang B, Xu G, Cui G. Golden bile powder prevents drunkenness and alcohol-induced liver injury in mice via the gut microbiota and metabolic modulation. Chin Med 2024; 19:39. [PMID: 38431607 PMCID: PMC10908100 DOI: 10.1186/s13020-024-00912-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Drunkenness and alcoholic liver disease (ALD) are critical public health issues associated with significant morbidity and mortality due to chronic overconsumption of alcohol. Traditional remedies, such as bear bile powder, have been historically acclaimed for their hepatoprotective properties. This study assessed the efficacy of a biotransformed bear bile powder known as golden bile powder (GBP) in alleviating alcohol-induced drunkenness and ALD. METHODS A murine model was engineered to simulate alcohol drunkenness and acute hepatic injury through the administration of a 50% ethanol solution. Intervention with GBP and its effects on alcohol-related symptoms were scrutinized, by employing an integrative approach that encompasses serum metabolomics, network medicine, and gut microbiota profiling to elucidate the protective mechanisms of GBP. RESULTS GBP administration significantly delayed the onset of drunkenness and decreased the duration of ethanol-induced inebriation in mice. Enhanced liver cell recovery was indicated by increased hepatic aldehyde dehydrogenase levels and superoxide dismutase activity, along with significant decreases in the serum alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, triglyceride, and total cholesterol levels (P < 0.05). These biochemical alterations suggest diminished hepatic damage and enhanced lipid homeostasis. Microbiota analysis via 16S rDNA sequencing revealed significant changes in gut microbial diversity and composition following alcohol exposure, and these changes were effectively reversed by GBP treatment. Metabolomic analyses demonstrated that GBP normalized the alcohol-induced perturbations in phospholipids, fatty acids, and bile acids. Correlation assessments linked distinct microbial genera to serum bile acid profiles, indicating that the protective efficacy of GBP may be attributable to modulatory effects on metabolism and the gut microbiota composition. Network medicine insights suggest the prominence of two active agents in GBP as critical for addressing drunkenness and ALD. CONCLUSION GBP is a potent intervention for alcohol-induced pathology and offers hepatoprotective benefits, at least in part, through the modulation of the gut microbiota and related metabolic cascades.
Collapse
Affiliation(s)
- Yarong Wang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Zhenzhuang Zou
- Department of Pediatrics, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Sihua Wang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Airong Ren
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Zhaolin Ding
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Yingying Li
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Yifang Wang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Zhengming Qian
- College of Medical Imaging Laboratory and Rehabilitation, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bo Huang
- Department of Pediatrics, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Guiwei Xu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Guozhen Cui
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
9
|
Sosnowski K, Przybyłkowski A. Ethanol-induced changes to the gut microbiome compromise the intestinal homeostasis: a review. Gut Microbes 2024; 16:2393272. [PMID: 39224006 PMCID: PMC11376419 DOI: 10.1080/19490976.2024.2393272] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The intestine is the largest organ in terms of surface area in the human body. It is responsible not only for absorbing nutrients but also for protection against the external world. The gut microbiota is essential in maintaining a properly functioning intestinal barrier, primarily through producing its metabolites: short-chain fatty acids, bile acids, and tryptophan derivatives. Ethanol overconsumption poses a significant threat to intestinal health. Not only does it damage the intestinal epithelium, but, maybe foremostly, it changes the gut microbiome. Those ethanol-driven changes shift its metabolome, depriving the host of the protective effect the physiological gut microbiota has. This literature review discusses the impact of ethanol consumption on the gut, the gut microbiota, and its metabolome, providing a comprehensive overview of the mechanisms through which ethanol disrupts intestinal homeostasis and discussing potential avenues for new therapeutic intervention.
Collapse
Affiliation(s)
- Konrad Sosnowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Xiao L, Xu G, Chen S, He Y, Peng F, Yuan C. Kaempferol ameliorated alcoholic liver disease through inhibiting hepatic bile acid synthesis by targeting intestinal FXR-FGF15 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155055. [PMID: 37678053 DOI: 10.1016/j.phymed.2023.155055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/16/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is characterized by the disturbance of bile acids homeostasis, which further deteriorates ALD. Bile acid metabolism and its related signal molecules have become new therapeutic targets for alcoholic liver disease. This study aimed to investigate the impact of kaempferol (KAE) on ALD and elucidate its underlying mechanisms. METHODS C57BL/6 N mice were utilized to establish Binge-on-Chronic alcohol exposure mice model. KAE was administered as an interventional drug to chronic alcohol-fed mice for four weeks to assess its effects on liver damage and bile acid metabolism. And Z-Guggulsterone (Z-Gu), a global FXR inhibitor, was used to investigate the impact of intestinal FXR-FGF15 signal in ALD mice. Additionally, intestinal epithelial cells were exposed to alcohol or specific bile acid to induce the damage of FXR activity in vitro. The dual luciferase activity assay was employed to ascertain the interplay between KAE and FXR activity. RESULTS The results indicated that KAE treatment exhibited a significant hepatoprotective effect against chronic alcohol-fed mice. Accompanied by the intestinal FXR activation, the administration of KAE suppressed hepatic bile acid synthesis and promoted intestinal bile acid excretion in chronic ALD mice. And the notable alterations in total bile acid levels and composition were observed in mice after chronic alcohol feeding, which were reversed by KAE supplementation. And more, the protective effects of KAE on ALD mice were deprived by the inhibition of intestinal FXR activation. In vitro experiments demonstrated that KAE effectively activated FXR-FGF15 signaling, mitigated the damage to FXR activity in intestinal epithelial cells caused by alcohol or specific bile acids. Additionally, luciferase activity assays revealed that KAE directly promoted FXR expression, thereby enhancing FXR activity. CONCLUSION KAE treatment inhibited hepatic bile acids synthesis, maintained bile acids homeostasis in ALD mice by directly activating intestinal FXR-FGF15 signaling, which effectively alleviated liver injury induced by chronic alcohol consumption.
Collapse
Affiliation(s)
- Li Xiao
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
| | - Guangfu Xu
- College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Silong Chen
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
| | - Yumin He
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
| | - Fan Peng
- College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Chengfu Yuan
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, China.
| |
Collapse
|
11
|
Ceci L, Han Y, Krutsinger K, Baiocchi L, Wu N, Kundu D, Kyritsi K, Zhou T, Gaudio E, Francis H, Alpini G, Kennedy L. Gallstone and Gallbladder Disease: Biliary Tract and Cholangiopathies. Compr Physiol 2023; 13:4909-4943. [PMID: 37358507 DOI: 10.1002/cphy.c220028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Cholestatic liver diseases are named primarily due to the blockage of bile flow and buildup of bile acids in the liver. Cholestasis can occur in cholangiopathies, fatty liver diseases, and during COVID-19 infection. Most literature evaluates damage occurring to the intrahepatic biliary tree during cholestasis; however, there may be associations between liver damage and gallbladder damage. Gallbladder damage can manifest as acute or chronic inflammation, perforation, polyps, cancer, and most commonly gallstones. Considering the gallbladder is an extension of the intrahepatic biliary network, and both tissues are lined by biliary epithelial cells that share common mechanisms and properties, it is worth further evaluation to understand the association between bile duct and gallbladder damage. In this comprehensive article, we discuss background information of the biliary tree and gallbladder, from function, damage, and therapeutic approaches. We then discuss published findings that identify gallbladder disorders in various liver diseases. Lastly, we provide the clinical aspect of gallbladder disorders in liver diseases and ways to enhance diagnostic and therapeutic approaches for congruent diagnosis. © 2023 American Physiological Society. Compr Physiol 13:4909-4943, 2023.
Collapse
Affiliation(s)
- Ludovica Ceci
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Rome, Italy
| | - Yuyan Han
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, USA
| | - Kelsey Krutsinger
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, USA
| | | | - Nan Wu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Debjyoti Kundu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tianhao Zhou
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Rome, Italy
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
12
|
Liu F, Nong X, Qu W, Li X. Pharmacokinetics and tissue distribution of 12 major active components in normal and chronic gastritis rats after oral administration of Weikangling capsules. JOURNAL OF ETHNOPHARMACOLOGY 2023:116722. [PMID: 37271330 DOI: 10.1016/j.jep.2023.116722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Weikangling Capsules (WKLCs) have been used in the clinic for the treatment of gastrointestinal disorders for more than 30 years. However, the pharmacokinetic characteristics and tissue distribution of its major bioactive components in rats under different physiological and pathological conditions are unclear. AIM OF THE STUDY In this study, we aimed to clarify the differences in pharmacokinetic parameters and tissue distribution of the major active components in WKLCs under physiological and pathological states. MATERIALS AND METHOD Normal and ethanol-induced chronic gastritis rats received 2.16 g/kg WKLCs by gavage, and urine, feces, plasma, and tissue (heart, liver, spleen, lung, kidney, stomach, and small intestine) samples were obtained. The active components in urine, feces and plasma were detected by ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOF-MS/MS). A rapid and sensitive analytical method, ultra-high-performance liquid chromatography coupled with triple-quadrupole linear ion-trap tandem mass spectrometry (UHPLC-QTRAP-MS/MS), was established and validated to clarify and compare the pharmacokinetics and tissue distribution of the major active components in normal and chronic gastritis rats. RESULTS A total of 36 chemical components in the feces, urine, and plasma of chronic gastritis rats were identified by UHPLC-Q-TOF-MS/MS. Among them, 20 were the prototype components of WKLCs, and 16 were metabolites. The pharmacokinetic characteristics and tissue distribution of 12 prototype components were successfully analyzed by UHPLC-QTRAP-MS/MS. The pharmacokinetic results showed that the Cmax, AUC0-t, and AUC0-∞ of paeoniflorin, glycyrrhizic acid, and glycyrrhetinic acid were distinctly higher than those of the other components in normal and chronic gastritis rats. Compared to normal rats, the Cmax, AUC0-t, and AUC0-∞ of albiflorin, liquiritin apioside, liquiritin, isoliquiritin, ononin, isoliquiritigenin, dactylorhin A, and glycyrrhizic acid were significantly increased in chronic gastritis rats (P < 0.05), while the Cmax, AUC0-t and AUC0-∞ of militarine and liquiritigenin had significantly lower decreases in chronic gastritis rats (P < 0.05). The results of the tissue distribution showed that the 12 components were widely distributed in the heart, liver, spleen, lung, kidney, stomach, and small intestine of rats, of which the liver, kidney, stomach, and small intestine were the main accumulative organs. Compared with normal rats, the concentrations of 12 components in the liver, kidney, stomach, and small intestine of chronic gastritis rats were widely higher than those of normal rats at the same time points. CONCLUSION The pharmacokinetic characteristics and tissue distribution of 12 active components of WKLCs were comprehensively characterized and elucidated in normal and chronic gastritis rats. These findings laid a solid foundation for revealing the pharmacodynamic material basis of WKLCs in treating gastrointestinal disorders.
Collapse
Affiliation(s)
- Feng Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaojing Nong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenhua Qu
- Heilongjiang Sunflower Pharmaceutical Co. Ltd., Heilongjiang, 150070, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
13
|
Zhang G, Chen H, Ren W, Huang J. Efficacy of bile acid profiles in diagnosing and staging of alcoholic liver disease. Scand J Clin Lab Invest 2023; 83:8-17. [PMID: 36484775 DOI: 10.1080/00365513.2022.2151508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM The diagnosis of alcoholic liver disease (ALD) is still a great challenge. Therefore, the purpose of this study is to identify and characterize new metabolomic biomarkers for the diagnosis and staging of ALD. METHODS A total of 127 patients with early liver injury, 40 patients with alcoholic cirrhosis (ALC) and 40 healthy controls were included in this study. Patients with early liver injury included 45 patients with alcoholic liver disease (ALD), 40 patients with non-alcoholic fatty liver disease (NAFLD) and 40 patients with viral liver disease (VLD). The differential metabolites in serum samples were analyzed using ultra-high-performance liquid chromatography-quadrupole/time-of-flight mass spectrometry, and partial metabolites in the differential metabolic pathway were identified by liquid chromatography- tandem mass spectrometry. RESULTS A total of 40 differential metabolites and five differential metabolic pathways in the four groups of patients with early liver disease and healthy controls were found, and the metabolic pathway of primary bile acid (BA) biosynthesis was the pathway that included the most differential metabolites. Therefore, 22 BA profiles were detected. The results revealed that the changes of BA profiles were most pronounced in patients with ALD compared with patients with NAFLD and VLD, in whom 12 differential BAs were diagnostic markers of ALD (AUC = 0.883). The 19 differential BAs in ALC and ALD were diagnostic markers of the stage of alcoholic hepatic fibrosis (AUC = 0.868). CONCLUSION BA profiles are potential indicators in the diagnosis of ALD and evaluation of different stages.
Collapse
Affiliation(s)
- Gaixia Zhang
- Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Haizhen Chen
- Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Wenbo Ren
- Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Jing Huang
- Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Qi L, Chen Y. Circulating Bile Acids as Biomarkers for Disease Diagnosis and Prevention. J Clin Endocrinol Metab 2023; 108:251-270. [PMID: 36374935 DOI: 10.1210/clinem/dgac659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022]
Abstract
CONTEXT Bile acids (BAs) are pivotal signaling molecules that regulate energy metabolism and inflammation. Recent epidemiological studies have reported specific alterations in circulating BA profiles in certain disease states, including obesity, type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD), and Alzheimer disease (AD). In the past decade, breakthroughs have been made regarding the translation of BA profiling into clinical use for disease prediction. In this review, we summarize and synthesize recent data on variation in circulating BA profiles in patients with various diseases to evaluate the value of these biomarkers in human plasma for early diagnosis. EVIDENCE ACQUISITION This review is based on a collection of primary and review literature gathered from a PubMed search for BAs, obesity, T2DM, insulin resistance (IR), NAFLD, hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), colon cancer, and AD, among other keywords. EVIDENCE SYNTHESIS Individuals with obesity, T2DM, HCC, CCA, or AD showed specific alterations in circulating BA profiles. These alterations may have existed long before the initial diagnosis of these diseases. The intricate relationship between obesity, IR, and NAFLD complicates the establishment of clear and independent associations between BA profiles and nonalcoholic steatohepatitis. Alterations in the levels of total BAs and several BA species were seen across the entire spectrum of NAFLD, demonstrating significant increases with the worsening of histological features. CONCLUSIONS Aberrant circulating BA profiles are an early event in the onset and progression of obesity, T2DM, HCC, and AD. The pleiotropic effects of BAs explain these broad connections. Circulating BA profiles could provide a basis for the development of biomarkers for the diagnosis and prevention of a wide range of diseases.
Collapse
Affiliation(s)
- Li Qi
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
| | - Yongsheng Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
15
|
Liu Y, Liu T, Zhao X, Gao Y. New insights into the bile acid-based regulatory mechanisms and therapeutic perspectives in alcohol-related liver disease. Cell Mol Life Sci 2022; 79:486. [PMID: 35978227 PMCID: PMC11073206 DOI: 10.1007/s00018-022-04509-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/27/2022]
Abstract
Cholestasis is a key causative factor in alcohol-related liver disease (ALD) and variable degrees of cholestasis occur in all stages of ALD. However, the pathogenetic mechanisms and biomarkers associated with cholestasis are not well characterized. Cholestatic disease is marked by the disruption of bile acids (BA) transport and homeostasis. Consequently, in both human and experimental ALD, the disease shows a direct correlation with an imbalance in BA equilibrium, which in turn may also affect the severity of the disease. Modulation of BA metabolism or signaling pathways is increasingly considered as a potential therapeutic strategy for ALD in humans. In this paper, we highlight the key advances made in the past two decades in characterizing the molecular regulatory mechanisms of BA synthesis, enterohepatic circulation, and BA homeostasis. We summarize recent insights into the nature of the linkage between BA dysregulation and ALD, including the abnormal expression of genes involved in BA metabolism, abnormal changes in receptors that regulate BA metabolism, and disturbance in the gut flora engaged in BA metabolism caused by alcohol consumption. Additionally, we provide novel perspectives on the changes in BAs in various stages of ALD. Finally, we propose potential pharmacological therapies for ALD targeting BA metabolism and signaling.
Collapse
Affiliation(s)
- Yali Liu
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Tao Liu
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Xu Zhao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Yanhang Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China.
- Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
16
|
Way GW, Jackson KG, Muscu SR, Zhou H. Key Signaling in Alcohol-Associated Liver Disease: The Role of Bile Acids. Cells 2022; 11:1374. [PMID: 35456053 PMCID: PMC9031669 DOI: 10.3390/cells11081374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023] Open
Abstract
Alcohol-associated liver disease (ALD) is a spectrum of diseases, the onset and progression of which are due to chronic alcohol use. ALD ranges, by increasing severity, from hepatic steatosis to alcoholic hepatitis (AH) and alcohol-associated cirrhosis (AC), and in some cases, can lead to the development of hepatocellular carcinoma (HCC). ALD continues to be a significant health burden and is now the main cause of liver transplantations in the United States. ALD leads to biological, microbial, physical, metabolic, and inflammatory changes in patients that vary depending on disease severity. ALD deaths have been increasing in recent years and are projected to continue to increase. Current treatment centers focus on abstinence and symptom management, with little in the way of resolving disease progression. Due to the metabolic disruption and gut dysbiosis in ALD, bile acid (BA) signaling and metabolism are also notably affected and play a prominent role in disease progression in ALD, as well as other liver disease states, such as non-alcoholic fatty liver disease (NAFLD). In this review, we summarize the recent advances in the understanding of the mechanisms by which alcohol consumption induces hepatic injury and the role of BA-mediated signaling in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Grayson W. Way
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Kaitlyn G. Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
| | - Shreya R. Muscu
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA; (K.G.J.); (S.R.M.)
- Central Virginia Veterans Healthcare System, Richmond, VA 23249, USA
| |
Collapse
|
17
|
Ruan Y, Liu R, Gong L. Investigation of dysregulated lipid metabolism in diabetic mice via targeted metabolomics of bile acids in enterohepatic circulation. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9236. [PMID: 34897861 DOI: 10.1002/rcm.9236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
RATIONALE The mechanism of lipid metabolism disorder in type 2 diabetes (T2DM) remains unclear. This study aimed to reveal the mechanism underlying dysregulated lipid metabolism in T2DM through bile acid metabolism. METHODS A db/db mouse model was employed to investigate the alteration of bile acid profiles in T2DM. Ultrahigh-performance liquid chromatography with tandem mass spectrometry was used to quantify the detailed bile acid levels in each compartment of enterohepatic circulation. The pathological change of mouse liver was assessed by liver histology and serum biochemical assays. The expression level of bile acid-related transporters and synthases was measured with Western blot analysis. RESULTS The results showed that T2DM can result in severe liver fat accumulation and liver damage. In addition, compared to the control group, in T2DM mice, bile acid synthesis is reduced, while the level of bile acids is increased at the storage sites and the reabsorption sites, but there are subtle gender differences. Further, the ratio of conjugated bile acids in total bile acid in the liver of T2DM mice increased significantly relative to the control group for both female and male mice. CONCLUSIONS In T2DM, bile acid metabolism is disordered in both male and female mice, which could be the underlying mechanism of dysregulated lipid metabolism in T2DM.
Collapse
Affiliation(s)
- Yanjiao Ruan
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingzhi Gong
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Modulation of the Bile Acid Enterohepatic Cycle by Intestinal Microbiota Alleviates Alcohol Liver Disease. Cells 2022; 11:cells11060968. [PMID: 35326419 PMCID: PMC8946080 DOI: 10.3390/cells11060968] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Reshaping the intestinal microbiota by the ingestion of fiber, such as pectin, improves alcohol-induced liver lesions in mice by modulating bacterial metabolites, including indoles, as well as bile acids (BAs). In this context, we aimed to elucidate how oral supplementation of pectin affects BA metabolism in alcohol-challenged mice receiving feces from patients with alcoholic hepatitis. Pectin reduced alcohol liver disease. This beneficial effect correlated with lower BA levels in the plasma and liver but higher levels in the caecum, suggesting that pectin stimulated BA excretion. Pectin modified the overall BA composition, favoring an augmentation in the proportion of hydrophilic forms in the liver, plasma, and gut. This effect was linked to an imbalance between hydrophobic and hydrophilic (less toxic) BAs in the gut. Pectin induced the enrichment of intestinal bacteria harboring genes that encode BA-metabolizing enzymes. The modulation of BA content by pectin inhibited farnesoid X receptor signaling in the ileum and the subsequent upregulation of Cyp7a1 in the liver. Despite an increase in BA synthesis, pectin reduced BA serum levels by promoting their intestinal excretion. In conclusion, pectin alleviates alcohol liver disease by modifying the BA cycle through effects on the intestinal microbiota and enhanced BA excretion.
Collapse
|
19
|
Chen W, Zhang Q, Ding M, Yao J, Guo Y, Yan W, Yu S, Shen Q, Huang M, Zheng Y, Lin Y, Wang Y, Liu Z, Lu L. Alcohol triggered bile acid disequilibrium by suppressing BSEP to sustain hepatocellular carcinoma progression. Chem Biol Interact 2022; 356:109847. [PMID: 35149083 DOI: 10.1016/j.cbi.2022.109847] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022]
Abstract
Bile acids (BAs), the most important components of bile, attribute predominately to maintain metabolic homeostasis. In hepatocellular carcinoma (HCC) patients, the BAs homeostasis was seriously disturbed, especially in those patients with alcohol-intake history. However, whether alcohol consumption could promote HCC progression via influencing BAs homeostasis and the precise mechanism underlying are still unclear. In our study, by collecting HCC specimens from both alcohol-drinkers (n = 15) and non-alcohol drinkers (n = 22), we found that compared to non-alcohol intake HCC patients, BAs homeostasis was disturbed in HCC patients who drank alcohol. Furthermore, ethanol treatment was also found to promote HCC progression by markedly activating oncogenes (RAS, MYC, MET, and HER2), while remarkably suppressing tumor suppressor genes (BRCA2 and APC). We evaluated 14 key functional genes that maintain the homeostasis of BAs and found that either in alcohol-intake HCC patients (n = 15), or in ethanol-treated mice, BSEP, rate-limiting transporter governing excreting BAs from liver into bile duct, was remarkably decreased when exposed to alcohol. Moreover, by screening for changes in the epigenetic landscape of liver cancer cells exposed to alcohol, we strikingly found that histone methyltransferases (RBBP-5, Suv39h1, ASH2L, and SET7/9) were increased, and KMT3B, KMT4, and KMT7 gene expression was also elevated, while histone demethyltransferases (JARID1a, JARID1b, JARID1c) were decreased. In summary, we found that alcohol could trigger BAs disequilibrium to initiate and promote HCC progression. Our study provided a novel and supplementary mechanism to determine the important role of alcohol-intake in HCC development regarding from the perspective of BAs homeostasis.
Collapse
Affiliation(s)
- Wenbo Chen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qisong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Ming Ding
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jingjing Yao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yajuan Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Wenxin Yan
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Shaofang Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qinghong Shen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Min Huang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yaqiu Zheng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuefang Lin
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, SAR, China.
| | - Linlin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, SAR, China.
| |
Collapse
|
20
|
Bile Acids, Gut Microbes, and the Neighborhood Food Environment-a Potential Driver of Colorectal Cancer Health Disparities. mSystems 2022; 7:e0117421. [PMID: 35103491 PMCID: PMC8805634 DOI: 10.1128/msystems.01174-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bile acids (BAs) facilitate nutrient digestion and absorption and act as signaling molecules in a number of metabolic and inflammatory pathways. Expansion of the BA pool and increased exposure to microbial BA metabolites has been associated with increased colorectal cancer (CRC) risk. It is well established that diet influences systemic BA concentrations and microbial BA metabolism. Therefore, consumption of nutrients that reduce colonic exposure to BAs and microbial BA metabolites may be an effective method for reducing CRC risk, particularly in populations disproportionately burdened by CRC. Individuals who identify as Black/African American (AA/B) have the highest CRC incidence and death in the United States and are more likely to live in a food environment with an inequitable access to BA mitigating nutrients. Thus, this review discusses the current evidence supporting diet as a contributor to CRC disparities through BA-mediated mechanisms and relationships between these mechanisms and barriers to maintaining a low-risk diet.
Collapse
|
21
|
Frazier R, Cai X, Lee J, Bundy JD, Jovanovich A, Chen J, Deo R, Lash JP, Anderson AH, Go AS, Feldman HI, Shafi T, Rhee EP, Miyazaki M, Chonchol M, Isakova T. Deoxycholic Acid and Risks of Cardiovascular Events, ESKD, and Mortality in CKD: The CRIC Study. Kidney Med 2022; 4:100387. [PMID: 35072049 PMCID: PMC8767130 DOI: 10.1016/j.xkme.2021.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rationale & Objective Elevated levels of deoxycholic acid (DCA) are associated with adverse outcomes and may contribute to vascular calcification in patients with chronic kidney disease (CKD). We tested the hypothesis that elevated levels of DCA were associated with increased risks of cardiovascular disease, CKD progression, and death in patients with CKD. Study Design Prospective observational cohort study. Setting & Participants We included 3,147 Chronic Renal Insufficiency Cohort study participants who had fasting DCA levels. The average age was 59 ± 11 years, 45.3% were women, 40.6% were African American, and the mean estimated glomerular filtration rate was 42.5 ± 16.0 mL/min/1.73 m2. Predictor Fasting DCA levels in Chronic Renal Insufficiency Cohort study participants. Outcomes Risks of atherosclerotic and heart failure events, end-stage kidney disease (ESKD), and all-cause mortality. Analytical Approach We used Tobit regression to identify predictors of DCA levels. We used Cox regression to examine the association between fasting DCA levels and clinical outcomes. Results The strongest predictors of elevated DCA levels in adjusted models were increased age and nonuse of statins. The associations between log-transformed DCA levels and clinical outcomes were nonlinear. After adjustment, DCA levels above the median were independently associated with higher risks of ESKD (HR, 2.67; 95% CI, 1.51-4.74) and all-cause mortality (HR, 2.13; 95% CI, 1.25-3.64). DCA levels above the median were not associated with atherosclerotic and heart failure events, and DCA levels below the median were not associated with clinical outcomes. Limitations We were unable to measure DCA longitudinally or in urinary or fecal samples, and we were unable to measure other bile acids. We also could not measure many factors that affect DCA levels. Conclusions In 3,147 participants with CKD stages 2-4, DCA levels above the median were independently associated with ESKD and all-cause mortality.
Collapse
|
22
|
Li X, Khan I, Huang G, Lu Y, Wang L, Liu Y, Lu L, Hsiao WW, Liu Z. Kaempferol acts on bile acid signaling and gut microbiota to attenuate the tumor burden in ApcMin/+ mice. Eur J Pharmacol 2022; 918:174773. [DOI: 10.1016/j.ejphar.2022.174773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/16/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022]
|
23
|
Fan M, Wang Y, Jin L, Fang Z, Peng J, Tu J, Liu Y, Zhang E, Xu S, Liu X, Huo Y, Sun Z, Chao X, Ding WX, Yan Q, Huang W. Bile Acid-Mediated Activation of Brown Fat Protects From Alcohol-Induced Steatosis and Liver Injury in Mice. Cell Mol Gastroenterol Hepatol 2021; 13:809-826. [PMID: 34896286 PMCID: PMC8802063 DOI: 10.1016/j.jcmgh.2021.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Alcohol-associated liver disease (AALD) is one of the most common causes of liver injury and failure. Limited knowledge of the mechanisms underlying AALD impedes the development of efficacious therapies. Bile acid (BA) signaling was shown to participate in the progression of AALD. However, the mechanisms remain poorly understood. METHODS C57BL/6J wild-type (WT), Takeda G-protein-coupled bile acid receptor 5 (TGR5) knockout (KO) and brown adipose tissue (BAT)-specific TGR5 knockdown mice were subjected to ethanol feeding-induced AALD. Liver samples from alcoholic hepatitis patients were used to examine the BA circulation signaling. Human Embryonic Kidney Cells 293 were used for the TGR5 reporter assay. 23(S)-methyl-lithocholic acid was used as a molecular tool to confirm the regulatory functions of BAT in the AALD mouse model. RESULTS Ethanol feeding increased the expression of the thermogenesis genes downstream of TGR5 in BAT of WT, but not TGR5 KO, mice. TGR5 deficiency significantly blocked BAT activity and energy expenditure in mice after ethanol feeding. Alcohol increased serum BA levels in mice and human beings through altering BA transportation, and the altered BAs activated TGR5 signaling to regulate metabolism. Compared with ethanol-fed WT mice, ethanol-fed TGR5 KO mice showed less free fatty acid (FFA) β-oxidation in BAT, leading to higher levels of FFA in the circulation, increased liver uptake of FFAs, and exacerbated AALD. BAT-specific TGR5 knockdown mice showed similar results with TGR5 KO mice in AALD. Agonist treatment significantly activated TGR5 signaling in BAT, increased thermogenesis, reduced serum FFA level, and ameliorated hepatic steatosis and injury in AALD mice, while these effects were lost in TGR5 KO mice. CONCLUSIONS BA signaling plays a protective role in AALD by enhancing BAT thermogenesis. Targeting TGR5 in BAT may be a promising approach for the treatment of AALD.
Collapse
Affiliation(s)
- Mingjie Fan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang, China,Department of Diabetes Complications and Metabolism, Duarte, California
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Lihua Jin
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Zhipeng Fang
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Jiangling Peng
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Jui Tu
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Yanjun Liu
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Eryun Zhang
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Senlin Xu
- Department of Diabetes Complications and Metabolism, Duarte, California,Graduate School of Biological Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Xiaoqian Liu
- Department of Diabetes Complications and Metabolism, Duarte, California
| | - Yuqing Huo
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Qingfeng Yan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang, China,Qingfeng Yan, PhD, College of Life Science, Zhejiang University, Hangzhou, 310058 Zhejiang, China. fax: 01186-571-88206646.
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Duarte, California,Graduate School of Biological Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, California,Correspondence Address correspondence to: Wendong Huang, PhD, Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, California 91010. fax: (626) 256-8704.
| |
Collapse
|
24
|
Yue H, Cai W, Li Y, Feng X, Dong P, Xue C, Wang J. A Novel Sialoglycopeptide from Gadus morhua Eggs Prevents Liver Fibrosis Induced by CCl 4 via Downregulating FXR/FGF15 and TLR4/TGF-β/Smad Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13093-13101. [PMID: 34714650 DOI: 10.1021/acs.jafc.1c05411] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Liver fibrosis plays a critical role in liver disease progression. A sialoglycopeptide from the Gadus morhua eggs (Gm-SGPP) was identified having a 7000 Da molecular weight with a core pentasaccharide structure and osteogenesis activity. However, whether Gm-SGPP is beneficial to liver fibrosis remains unknown. In this study, mice with liver fibrosis were intraperitoneally injected with 2.5% CCl4 (10 mL/kg) and orally administered with Gm-SGPP (500 mg/kg) for 30 days. Results showed that Gm-SGPP alleviated oxidative liver damage and lipid metabolism disorder and reduced hepatocyte necrosis and lipid droplet accumulation. Notably, we found that Gm-SGPP increased the number and changed the composition of bile acids via increasing cholesterol 7a-hydroxylase (CYP7A1) and sterol 27-hydroxylase (CYP27A1) expression, which caused inhibition of ileum farnesoid X receptor (FXR) expression and accelerated the cholesterol conversion. Cholesterol accumulation is a risk factor for liver fibrosis. Masson staining showed that Gm-SGPP significantly reduced the degree of collagen deposition. Western blotting further suggested that Gm-SGPP downregulated the key gene of the toll-like receptor 4 (TLR4)-mediated transforming growth factor-β (TGF-β)/Smad pathway. To our best knowledge, this is the first report that Gm-SGPP prevented liver fibrosis via attenuating cholesterol accumulation. Our present results provide new ideas for the Gadus morhua egg's high-value utilization.
Collapse
Affiliation(s)
- Hao Yue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| | - Weizhen Cai
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| | - Yanqi Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| | - Xiaoxuan Feng
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| | - Ping Dong
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province 266237, P.R. China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shangdong 266003, China
| |
Collapse
|
25
|
Simbrunner B, Trauner M, Reiberger T. Review article: therapeutic aspects of bile acid signalling in the gut-liver axis. Aliment Pharmacol Ther 2021; 54:1243-1262. [PMID: 34555862 PMCID: PMC9290708 DOI: 10.1111/apt.16602] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Bile acids are important endocrine modulators of intestinal and hepatic signalling cascades orchestrating critical pathophysiological processes in various liver diseases. Increasing knowledge on bile acid signalling has stimulated the development of synthetic ligands of nuclear bile acid receptors and other bile acid analogues. AIM This review summarises important aspects of bile acid-mediated crosstalk between the gut and the liver ("gut-liver axis") as well as recent findings from experimental and clinical studies. METHODS We performed a literature review on bile acid signalling, and therapeutic applications in chronic liver disease. RESULTS Intestinal and hepatic bile acid signalling pathways maintain bile acid homeostasis. Perturbations of bile acid-mediated gut-liver crosstalk dysregulate transcriptional networks involved in inflammation, fibrosis and endothelial dysfunction. Bile acids induce enterohepatic feedback signalling by the release of intestinal hormones, and regulate enterohepatic circulation. Importantly, bile acid signalling plays a central role in maintaining intestinal barrier integrity and antibacterial defense, which is particularly relevant in cirrhosis, where bacterial translocation has a profound impact on disease progression. The nuclear bile acid farnesoid X receptor (FXR) is a central intersection in bile acid signalling and has emerged as a relevant therapeutic target. CONCLUSIONS Experimental evidence suggests that bile acid signalling improves the intestinal barrier and protects against bacterial translocation in cirrhosis. FXR agonists have displayed efficacy for the treatment of cholestatic and metabolic liver disease in randomised controlled clinical trials. However, similar effects remain to be shown in advanced liver disease, particularly in patients with decompensated cirrhosis.
Collapse
Affiliation(s)
- Benedikt Simbrunner
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria,Vienna Hepatic Hemodynamic LabMedical University of ViennaViennaAustria,Christian‐Doppler Laboratory for Portal Hypertension and Liver FibrosisMedical University of ViennaViennaAustria
| | - Michael Trauner
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Thomas Reiberger
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria,Vienna Hepatic Hemodynamic LabMedical University of ViennaViennaAustria,Christian‐Doppler Laboratory for Portal Hypertension and Liver FibrosisMedical University of ViennaViennaAustria
| |
Collapse
|