51
|
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: 0] [Impact Index Per Article: 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
|
52
|
Wang Y, Xu H, Zhou X, Chen W, Zhou H. Dysregulated bile acid homeostasis: unveiling its role in metabolic diseases. MEDICAL REVIEW (2021) 2024; 4:262-283. [PMID: 39135605 PMCID: PMC11317083 DOI: 10.1515/mr-2024-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/13/2024] [Indexed: 08/15/2024]
Abstract
Maintaining bile acid homeostasis is essential for metabolic health. Bile acid homeostasis encompasses a complex interplay between biosynthesis, conjugation, secretion, and reabsorption. Beyond their vital role in digestion and absorption of lipid-soluble nutrients, bile acids are pivotal in systemic metabolic regulation. Recent studies have linked bile acid dysregulation to the pathogenesis of metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), and metabolic dysfunction-associated steatotic liver disease (MASLD). Bile acids are essential signaling molecules that regulate many critical biological processes, including lipid metabolism, energy expenditure, insulin sensitivity, and glucose metabolism. Disruption in bile acid homeostasis contributes to metabolic disease via altered bile acid feedback mechanisms, hormonal dysregulation, interactions with the gut microbiota, and changes in the expression and function of bile acid transporters and receptors. This review summarized the essential molecular pathways and regulatory mechanisms through which bile acid dysregulation contributes to the pathogenesis and progression of obesity, T2DM, and MASLD. We aim to underscore the significance of bile acids as potential diagnostic markers and therapeutic agents in the context of metabolic diseases, providing insights into their application in translational medicine.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, VA, USA
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Huangru Xu
- School of Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weidong Chen
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
53
|
Vujic E, Ferguson SS, Brouwer KLR. Effects of PFAS on human liver transporters: implications for health outcomes. Toxicol Sci 2024; 200:213-227. [PMID: 38724241 DOI: 10.1093/toxsci/kfae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) have become internationally recognized over the past three decades as persistent organic pollutants used in the production of various consumer and industrial goods. Research efforts continue to gauge the risk that historically used, and newly produced, PFAS may cause to human health. Numerous studies report toxic effects of PFAS on the human liver as well as increased serum cholesterol levels in adults. A major concern with PFAS, also dubbed "forever chemicals," is that they accumulate in the liver and kidney and persist in serum. The mechanisms responsible for their disposition and excretion in humans are poorly understood. A better understanding of the interaction of PFAS with liver transporters, as it pertains to the disposition of PFAS and other xenobiotics, could provide mechanistic insight into human health effects and guide efforts toward risk assessment of compounds in development. This review summarizes the current state of the literature on the emerging relationships (eg, substrates, inhibitors, modulators of gene expression) between PFAS and specific hepatic transporters. The adaptive and toxicological responses of hepatocytes to PFAS that reveal linkages to pathologies and epidemiological findings are highlighted. The evidence suggests that our understanding of the molecular landscape of PFAS must improve to determine their impact on the expression and function of hepatocyte transporters that play a key role in PFAS or other xenobiotic disposition. From here, we can assess what role these changes may have in documented human health outcomes.
Collapse
Affiliation(s)
- Ena Vujic
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
54
|
Lin YN, Hsu JR, Wang CL, Huang YC, Wang JY, Wu CY, Wu LL. Nuclear factor interleukin 3 and metabolic dysfunction-associated fatty liver disease development. Commun Biol 2024; 7:897. [PMID: 39048678 PMCID: PMC11269659 DOI: 10.1038/s42003-024-06565-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
This study investigates sex-specific effects in a gain-of-function model to evaluate Nfil3 function in relation to high-fat diet (HFD)-induced metabolic dysfunction-associated steatotic liver disease (MASLD) and gut microbiota (GM)-induced alterations in the bile acid (BA) profile. MASLD is induced in both wild type and Nfil3-deficient (NKO) C57BL/6 J mice through an HFD. The hepatic immune response is evaluated using flow cytometry, revealing that NKO mice exhibit lower body weight, serum triglyceride (TG) levels, tissue injury, inflammation, and fat accumulation. The Nfil3 deletion reduces macrophage counts in fibrotic liver tissues, decreases proinflammatory gene and protein expression, and diminishes gut barrier function. Alpha and beta diversity analysis reveal increased GM alpha diversity across different sexes. The Nfil3 gene deletion modifies the BA profile, suggesting that negative feedback through the Nfil3-FXR-FGF15 axis facilitates BA recycling from the liver via enterohepatic circulation. Therefore, inhibiting Nfil3 in the liver offers a viable treatment approach for MASLD.
Collapse
Grants
- CI-110-22 Yen Tjing Ling Medical Foundation
- 11210 Ministry of Health and Welfare (Ministry of Health and Welfare, Taiwan)
- National Science and Technology Council (NSTC), Taiwan (nos. 108-2320-B-010-045-MY3, 110-2320-B-002-080-MY3, MOST 111-2314-B-A49-072, and NSTC 112-2314-B-A49-028-MY3 to L.L.W and NSTC 112-2740-B-A49-002, NSTC 112-2327-B-A49-005–, NSTC 112-2321-B-A49-005–, MOHW112-TDU-B-221-124007, and MOHW113-TDU-B-221-13400 to C.Y. Wu), Yen Tjing Ling Medical Foundation (nos.CI-110-22 and CI-111-24 to L.L.W), and the TYGH-NYCU Joint Research Program (no. PTH110001) and Ministry of Health and Welfare (No. 11210).
Collapse
Affiliation(s)
- Yung-Ni Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jia-Rou Hsu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Lin Wang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chen Huang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jzy-Yu Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chun-Ying Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Health Innovation Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Microbiota Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Translational Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Public Health, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Li-Ling Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Health Innovation Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Microbiota Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
55
|
Newman NK, Monnier PM, Rodrigues RR, Gurung M, Vasquez-Perez S, Hioki KA, Greer RL, Brown K, Morgun A, Shulzhenko N. Host response to cholestyramine can be mediated by the gut microbiota. MICROBIOME RESEARCH REPORTS 2024; 3:40. [PMID: 39741955 PMCID: PMC11684918 DOI: 10.20517/mrr.2023.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/09/2024] [Accepted: 06/24/2024] [Indexed: 01/03/2025]
Abstract
Background: The gut microbiota has been implicated as a major factor contributing to metabolic diseases and the response to drugs used for the treatment of such diseases. In this study, we tested the effect of cholestyramine, a bile acid sequestrant that reduces blood cholesterol, on the murine gut microbiota and metabolism. We also explored the hypothesis that some effects of this drug on systemic metabolism can be attributed to alterations in the gut microbiota. Methods: We used a Western diet (WD) for 8 weeks to induce metabolic disease in mice, then treated some mice with cholestyramine added to WD. Metabolic phenotyping, gene expression in liver and ileum, and microbiota 16S rRNA genes were analyzed. Then, transkingdom network analysis was used to find candidate microbes for the cholestyramine effect. Results: We observed that cholestyramine decreased glucose and epididymal fat levels and detected dysregulation of genes known to be regulated by cholestyramine in the liver and ileum. Analysis of gut microbiota showed increased alpha diversity in cholestyramine-treated mice, with fourteen taxa showing restoration of relative abundance to levels resembling those in mice fed a control diet. Using transkingdom network analysis, we inferred two amplicon sequence variants (ASVs), one from the Lachnospiraceae family (ASV49) and the other from the Muribaculaceae family (ASV1), as potential regulators of cholestyramine effects. ASV49 was also negatively linked with glucose levels, further indicating its beneficial role. Conclusion: Our results indicate that the gut microbiota has a role in the beneficial effects of cholestyramine and suggest specific microbes as targets of future investigations.
Collapse
Affiliation(s)
- Nolan K. Newman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Philip M. Monnier
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Richard R. Rodrigues
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Manoj Gurung
- Department of Biomedical Sciences, Carson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Stephany Vasquez-Perez
- Department of Biomedical Sciences, Carson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Kaito A. Hioki
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Renee L. Greer
- Department of Biomedical Sciences, Carson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Kevin Brown
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Andrey Morgun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Natalia Shulzhenko
- Department of Biomedical Sciences, Carson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
56
|
Zhou Y, Bai F, Xiao R, Chen M, Sun Y, Ye J. Proteomics and Its Combined Analysis with Transcriptomics: Liver Fat-Lowering Effect of Taurine in High-Fat Fed Grouper ( Epinephelus coioides). Animals (Basel) 2024; 14:2039. [PMID: 39061501 PMCID: PMC11274106 DOI: 10.3390/ani14142039] [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: 06/17/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
In order to understand the intervention effect of taurine on liver fat deposition induced by high fat intake in the orange-spotted grouper (Epinephelus coioides), we performed proteomic analysis and association analysis with previously obtained transcriptomic data. Three isoproteic (47% crude protein) diets were designed to contain two levels of fat and were named as the 10% fat diet (10F), 15% fat diet (15F), and 15% fat with 1% taurine (15FT). The 10F diet was used as the control diet. After 8 weeks of feeding, the 15F diet exhibited comparable weight gain, feed conversion ratio, and hepatosomatic index as the 10F diet, but the former increased liver fat content vs. the latter. Feeding with the 15FT diet resulted in an improvement in weight gain and a reduction in feed conversion ratio, hepatosomatic index, and liver fat content compared with feeding the 15F diet. When comparing liver proteomic data between the 15F and 15FT groups, a total of 133 differentially expressed proteins (DEPs) were identified, of which 51 were upregulated DEPs and 82 were downregulated DEPs. Among these DEPs, cholesterol 27-hydroxylase, phosphatidate phosphatase LPIN, phosphatidylinositol phospholipase C, and 6-phosphofructo-2-kinase were further screened out and were involved in primary bile acid biosynthesis, glycerolipid metabolism, the phosphatidylinositol signaling system, and the AMPK signaling pathway as key DEPs in terms of alleviating liver fat deposition of taurine in high-fat fed fish. With the association analysis of transcriptomic and proteomic data through KEGG, three differentially expressed genes (atp1a, arf1_2, and plcd) and four DEPs (CYP27α1, LPIN, PLCD, and PTK2B) were co-enriched into five pathways related to fat metabolism including primary bile acid synthesis, bile secretion, glycerolipid metabolism, phospholipid D signaling, or/and phosphatidylinositol signaling. The results showed that dietary taurine intervention could trigger activation of bile acid biosynthesis and inhibition of triglyceride biosynthesis, thereby mediating the liver fat-lowering effects in high-fat fed orange-spotted grouper. The present study contributes some novel insight into the liver fat-lowering effects of dietary taurine in high-fat fed groupers.
Collapse
Affiliation(s)
| | | | | | | | | | - Jidan Ye
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen 361021, China; (Y.Z.); (F.B.); (R.X.); (M.C.); (Y.S.)
| |
Collapse
|
57
|
Yang X, Qiu K, Jiang Y, Huang Y, Zhang Y, Liao Y. Metabolic Crosstalk between Liver and Brain: From Diseases to Mechanisms. Int J Mol Sci 2024; 25:7621. [PMID: 39062868 PMCID: PMC11277155 DOI: 10.3390/ijms25147621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver and brain is implicated in numerous metabolic and neurodegenerative disorders. Recent insights have positioned the liver not only as a central metabolic hub but also as an endocrine organ, capable of secreting hepatokines that transmit metabolic signals throughout the body via the bloodstream. Metabolites from the liver or gut microbiota also facilitate a complex dialogue between liver and brain. In parallel to humoral factors, the neural pathways, particularly the hypothalamic nuclei and autonomic nervous system, are pivotal in modulating the bilateral metabolic interplay between the cerebral and hepatic compartments. The term "liver-brain axis" vividly portrays this interaction. At the end of this review, we summarize cutting-edge technical advancements that have enabled the observation and manipulation of these signals, including genetic engineering, molecular tracing, and delivery technologies. These innovations are paving the way for a deeper understanding of the liver-brain axis and its role in metabolic homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
58
|
Cheng Y, Wang S, Zhu W, Xu Z, Xiao L, Wu J, Meng Y, Zhang J, Cheng C. Deoxycholic acid inducing chronic atrophic gastritis with colonic mucosal lesion correlated to mucosal immune dysfunction in rats. Sci Rep 2024; 14:15798. [PMID: 38982226 PMCID: PMC11233621 DOI: 10.1038/s41598-024-66660-3] [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: 11/27/2023] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
The present study aimed to explore the underlying mechanism of bile reflux-inducing chronic atrophic gastritis (CAG) with colonic mucosal lesion. The rat model of CAG with colonic mucosal lesion was induced by free-drinking 20 mmol/L sodium deoxycholate to simulate bile reflux and 2% cold sodium salicylate for 12 weeks. In comparison to the control group, the model rats had increased abundances of Bacteroidetes and Firmicutes but had decreased abundances of Proteobacteria and Fusobacterium. Several gut bacteria with bile acids transformation ability were enriched in the model group, such as Blautia, Phascolarctobacter, and Enterococcus. The cytotoxic deoxycholic acid and lithocholic acid were significantly increased in the model group. Transcriptome analysis of colonic tissues presented that the down-regulated genes enriched in T cell receptor signaling pathway, antigen processing and presentation, Th17 cell differentiation, Th1 and Th2 cell differentiation, and intestinal immune network for IgA production in the model group. These results suggest that bile reflux-inducing CAG with colonic mucosal lesion accompanied by gut dysbacteriosis, mucosal immunocompromise, and increased gene expressions related to repair of intestinal mucosal injury.
Collapse
Affiliation(s)
- Yuqin Cheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Shuaishuai Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wenfei Zhu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zijing Xu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Ling Xiao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jianping Wu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Yufen Meng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Junfeng Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| | - Chun Cheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
59
|
Cheng Z, Yang L, Chu H. The role of gut microbiota, exosomes, and their interaction in the pathogenesis of ALD. J Adv Res 2024:S2090-1232(24)00268-6. [PMID: 38969094 DOI: 10.1016/j.jare.2024.07.002] [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: 05/06/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND The liver disorders caused by alcohol abuse are termed alcoholic-related liver disease (ALD), including alcoholic steatosis, alcoholic steatohepatitis, alcoholic hepatitis, and alcoholic cirrhosis, posing a significant threat to human health. Currently, ALD pathogenesis has not been completely clarified, which is likely to be related to the direct damage caused by alcohol and its metabolic products, oxidative stress, gut dysbiosis, and exosomes. AIMS The existing studies suggest that both the gut microbiota and exosomes contribute to the development of ALD. Moreover, there exists an interaction between the gut microbiota and exosomes. We discuss whether this interaction plays a role in the pathogenesis of ALD and whether it can be a potential therapeutic target for ALD treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW Chronic alcohol intake alters the diversity and composition of gut microbiota, which greatly contributes to ALD's progression. Some approaches targeting the gut microbiota, including probiotics, fecal microbiota transplantation, and phage therapy, have been confirmed to effectively ameliorate ALD in many animal experiments and/or several clinical trials. In ALD, the levels of exosomes and the expression profile of microRNA have also changed, which affects the pathogenesis of ALD. Moreover, there is an interplay between exosomes and the gut microbiota, which also putatively acts as a pathogenic factor of ALD.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
60
|
Vue Z, Murphy A, Le H, Neikirk K, Garza-Lopez E, Marshall AG, Mungai M, Jenkins B, Vang L, Beasley HK, Ezedimma M, Manus S, Whiteside A, Forni MF, Harris C, Crabtree A, Albritton CF, Jamison S, Demirci M, Prasad P, Oliver A, Actkins KV, Shao J, Zaganjor E, Scudese E, Rodriguez B, Koh A, Rabago I, Moore JE, Nguyen D, Aftab M, Kirk B, Li Y, Wandira N, Ahmad T, Saleem M, Kadam A, Katti P, Koh HJ, Evans C, Koo YD, Wang E, Smith Q, Tomar D, Williams CR, Sweetwyne MT, Quintana AM, Phillips MA, Hubert D, Kirabo A, Dash C, Jadiya P, Kinder A, Ajijola OA, Miller-Fleming TW, McReynolds MR, Hinton A. MICOS Complex Loss Governs Age-Associated Murine Mitochondrial Architecture and Metabolism in the Liver, While Sam50 Dictates Diet Changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599846. [PMID: 38979162 PMCID: PMC11230271 DOI: 10.1101/2024.06.20.599846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The liver, the largest internal organ and a metabolic hub, undergoes significant declines due to aging, affecting mitochondrial function and increasing the risk of systemic liver diseases. How the mitochondrial three-dimensional (3D) structure changes in the liver across aging, and the biological mechanisms regulating such changes confers remain unclear. In this study, we employed Serial Block Face-Scanning Electron Microscopy (SBF-SEM) to achieve high-resolution 3D reconstructions of murine liver mitochondria to observe diverse phenotypes and structural alterations that occur with age, marked by a reduction in size and complexity. We also show concomitant metabolomic and lipidomic changes in aged samples. Aged human samples reflected altered disease risk. To find potential regulators of this change, we examined the Mitochondrial Contact Site and Cristae Organizing System (MICOS) complex, which plays a crucial role in maintaining mitochondrial architecture. We observe that the MICOS complex is lost during aging, but not Sam50. Sam50 is a component of the sorting and assembly machinery (SAM) complex that acts in tandem with the MICOS complex to modulate cristae morphology. In murine models subjected to a high-fat diet, there is a marked depletion of the mitochondrial protein SAM50. This reduction in Sam50 expression may heighten the susceptibility to liver disease, as our human biobank studies corroborate that Sam50 plays a genetically regulated role in the predisposition to multiple liver diseases. We further show that changes in mitochondrial calcium dysregulation and oxidative stress accompany the disruption of the MICOS complex. Together, we establish that a decrease in mitochondrial complexity and dysregulated metabolism occur with murine liver aging. While these changes are partially be regulated by age-related loss of the MICOS complex, the confluence of a murine high-fat diet can also cause loss of Sam50, which contributes to liver diseases. In summary, our study reveals potential regulators that affect age-related changes in mitochondrial structure and metabolism, which can be targeted in future therapeutic techniques.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mariaassumpta Ezedimma
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Maria Fernanda Forni
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Claude F. Albritton
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ky’Era V. Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Izabella Rabago
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Johnathan E. Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Desiree Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Muhammad Aftab
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Yahang Li
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Nelson Wandira
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Taseer Ahmad
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Chantell Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, Iowa, USA1
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Dhanendra Tomar
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, 37232, USA
- Vanderbilt Institute for Global Health, Nashville, TN, 37232, USA
| | - Chandravanu Dash
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, NC
| | - André Kinder
- Artur Sá Earp Neto University Center – UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center, University of California, Los Angeles, CA, USA
| | - Tyne W. Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
61
|
Rodriguez Rodriguez ER, Nordvang RT, Petersson M, Rendsvig JKH, Arendrup EW, Fernández Quintero ML, Jenkins TP, Laustsen AH, Thrane SW. Fit-for-purpose heterodivalent single-domain antibody for gastrointestinal targeting of toxin B from Clostridium difficile. Protein Sci 2024; 33:e5035. [PMID: 38923049 PMCID: PMC11201815 DOI: 10.1002/pro.5035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024]
Abstract
Single-domain antibodies (sdAbs), such as VHHs, are increasingly being developed for gastrointestinal (GI) applications against pathogens to strengthen gut health. However, what constitutes a suitable developability profile for applying these proteins in a gastrointestinal setting remains poorly explored. Here, we describe an in vitro methodology for the identification of sdAb derivatives, more specifically divalent VHH constructs, that display extraordinary developability properties for oral delivery and functionality in the GI environment. We showcase this by developing a heterodivalent VHH construct that cross-inhibits the toxic activity of the glycosyltransferase domains (GTDs) from three different toxinotypes of cytotoxin B (TcdB) from lineages of Clostridium difficile. We show that the VHH construct possesses high stability and binding activity under gastric conditions, in the presence of bile salts, and at high temperatures. We suggest that the incorporation of early developability assessment could significantly aid in the efficient discovery of VHHs and related constructs fit for oral delivery and GI applications.
Collapse
Affiliation(s)
| | | | - Marcus Petersson
- Bactolife A/SCopenhagen EastDenmark
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | | | | | | | - Timothy P. Jenkins
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | - Andreas H. Laustsen
- Bactolife A/SCopenhagen EastDenmark
- Department of Biotechnology and BiomedicineTechnical University of DenmarkLyngbyDenmark
| | | |
Collapse
|
62
|
Mohanty I, Allaband C, Mannochio-Russo H, El Abiead Y, Hagey LR, Knight R, Dorrestein PC. The changing metabolic landscape of bile acids - keys to metabolism and immune regulation. Nat Rev Gastroenterol Hepatol 2024; 21:493-516. [PMID: 38575682 DOI: 10.1038/s41575-024-00914-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 04/06/2024]
Abstract
Bile acids regulate nutrient absorption and mitochondrial function, they establish and maintain gut microbial community composition and mediate inflammation, and they serve as signalling molecules that regulate appetite and energy homeostasis. The observation that there are hundreds of bile acids, especially many amidated bile acids, necessitates a revision of many of the classical descriptions of bile acids and bile acid enzyme functions. For example, bile salt hydrolases also have transferase activity. There are now hundreds of known modifications to bile acids and thousands of bile acid-associated genes, especially when including the microbiome, distributed throughout the human body (for example, there are >2,400 bile salt hydrolases alone). The fact that so much of our genetic and small-molecule repertoire, in both amount and diversity, is dedicated to bile acid function highlights the centrality of bile acids as key regulators of metabolism and immune homeostasis, which is, in large part, communicated via the gut microbiome.
Collapse
Affiliation(s)
- Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Celeste Allaband
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Helena Mannochio-Russo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lee R Hagey
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
63
|
Du K, Wang L, Jun JH, Dutta RK, Maeso-Díaz R, Oh SH, Ko DC, Diehl AM. Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress. NATURE AGING 2024; 4:949-968. [PMID: 38918603 DOI: 10.1038/s43587-024-00652-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/17/2024] [Indexed: 06/27/2024]
Abstract
Susceptibility to the biological consequences of aging varies among organs and individuals. We analyzed hepatocyte transcriptomes of healthy young and aged male mice to generate an aging hepatocyte gene signature, used it to deconvolute transcriptomic data from humans and mice with metabolic dysfunction-associated liver disease, validated findings with functional studies in mice and applied the signature to transcriptomic data from other organs to determine whether aging-sensitive degenerative mechanisms are conserved. We discovered that the signature enriches in diseased livers in parallel with degeneration. It is also enriched in failing human hearts, diseased kidneys and pancreatic islets from individuals with diabetes. The signature includes genes that control ferroptosis. Aged mice develop more hepatocyte ferroptosis and liver degeneration than young mice when fed diets that induce metabolic stress. Inhibiting ferroptosis shifts the liver transcriptome of old mice toward that of young mice and reverses aging-exacerbated liver damage, identifying ferroptosis as a tractable, conserved mechanism for aging-related tissue degeneration.
Collapse
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ji Hye Jun
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rajesh K Dutta
- Department of Medicine, Duke University, Durham, NC, USA
| | | | - Seh Hoon Oh
- Department of Medicine, Duke University, Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
64
|
Francini E, Orlandoni P, Sparvoli D, Jukic Peladic N, Cardelli M, Recchioni R, Silvi S, Stocchi V, Donati Zeppa S, Procopio AD, Capalbo M, Lattanzio F, Olivieri F, Marchegiani F. Possible Role of Tauroursodeoxycholic Acid (TUDCA) and Antibiotic Administration in Modulating Human Gut Microbiota in Home Enteral Nutrition Therapy for the Elderly: A Case Report. Int J Mol Sci 2024; 25:7115. [PMID: 39000220 PMCID: PMC11240908 DOI: 10.3390/ijms25137115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Tauroursodeoxycholic acid (TUDCA) increases the influx of primary bile acids into the gut. Results obtained on animal models suggested that Firmicutes and Proteobacteria phyla are more resistant to bile acids in rats. As part of a pilot study investigating the role of probiotics supplementation in elderly people with home enteral nutrition (HEN), a case of a 92-year-old woman with HEN is reported in the present study. She lives in a nursing home and suffers from Alzheimer's disease (AD); the patient had been prescribed TUDCA for lithiasis cholangitis. The aim of this case report is therefore to investigate whether long-term TUDCA administration may play a role in altering the patient's gut microbiota (GM) and the impact of an antibiotic therapy on the diversity of microbial species. Using next generation sequencing (NGS) analysis of the bacterial 16S ribosomal RNA (rRNA) gene a dominant shift toward Firmicutes and a remodeling in Proteobacteria abundance was observed in the woman's gut microbiota. Considering the patient's age, health status and type of diet, we would have expected to find a GM with a prevalence of Bacteroidetes phylum. This represents the first study investigating the possible TUDCA's effect on human GM.
Collapse
Affiliation(s)
- Emanuele Francini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (R.R.); (A.D.P.); (F.M.)
| | - Paolo Orlandoni
- Clinical Nutrition, IRCCS INRCA, 60127 Ancona, Italy; (P.O.); (D.S.); (N.J.P.)
| | - Debora Sparvoli
- Clinical Nutrition, IRCCS INRCA, 60127 Ancona, Italy; (P.O.); (D.S.); (N.J.P.)
| | | | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (M.C.); (F.O.)
| | - Rina Recchioni
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (R.R.); (A.D.P.); (F.M.)
| | - Stefania Silvi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Vilberto Stocchi
- Department of Human Science and Promotion of Quality of Life, San Raffaele Rome Telematic University, 00166 Rome, Italy;
| | - Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | - Antonio Domenico Procopio
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (R.R.); (A.D.P.); (F.M.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Maria Capalbo
- General Direction, IRCCS INRCA, 60124 Ancona, Italy;
| | | | - Fabiola Olivieri
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (M.C.); (F.O.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Francesca Marchegiani
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (R.R.); (A.D.P.); (F.M.)
| |
Collapse
|
65
|
Wu HT, Tsai CS, Chao TH, Ou HY, Tsai LM. A Novel Antioxidant, Hydrogen-Rich Coral Calcium Alters Gut Microbiome and Bile Acid Synthesis to Improve Methionine-and-Choline-Deficient Diet-Induced Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2024; 13:746. [PMID: 38929185 PMCID: PMC11201271 DOI: 10.3390/antiox13060746] [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: 04/11/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) has dramatically increased in recent years, and it is highly associated with metabolic diseases, as well as the development of hepatocellular carcinoma. However, effective therapeutic strategies for the treatment of NAFLD are still scarce. Although hydrogen-rich water shows beneficial effects for hepatic steatosis, the inconvenience limits the application of this antioxidant. In light of this, hydrogen-rich coral calcium (HRCC) was developed due to its convenience and quantifiable characteristics. However, the effects of HRCC on NAFLD are still unknown. In the present study, we found that HRCC treatment improved methionine-and-choline-deficient diet (MCD)-induced hepatic steatosis, increased aspartate aminotransferase and alanine aminotransferase levels, and elevated hepatic inflammatory factor expressions in mice. In addition to the increased expressions of antioxidative enzymes, we found that HRCC increased the expressions of bile acid biosynthesis-related genes, including Cyp8b1 and Cyp27a1. Increased hepatic bile acid contents, such as muricholic acids, 23 nor-deoxycholic acid, glycoursodeoxycholic acid, and cholic acids, were also confirmed in MCD mice treated with HRCC. Since the biogenesis of bile acids is associated with the constitution of gut microbiome, the alterations in gut microbiome by HRCC were evaluated. We found that HRCC significantly changed the constitution of gut microbiome in MCD mice and increased the contents of Anaerobacterium, Acutalibacter, Anaerosacchariphilus, and Corynebacterium. Taken together, HRCC improved MCD-induced NAFLD through anti-inflammatory mechanisms and by increasing antioxidative activities. Additionally, HRCC might alter gut microbiome to change hepatic bile acid contents, exerting beneficial effects for the treatment of NAFLD.
Collapse
Affiliation(s)
- Hung-Tsung Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (H.-T.W.); (T.-H.C.); (H.-Y.O.)
- Tong-Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chin-Shiang Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ting-Hsing Chao
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (H.-T.W.); (T.-H.C.); (H.-Y.O.)
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Horng-Yih Ou
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (H.-T.W.); (T.-H.C.); (H.-Y.O.)
- Tong-Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Liang-Miin Tsai
- Department of Internal Medicine, Tainan Municipal Hospital (Managed by Show-Chwan Medical Care Corporation), Tainan 701, Taiwan
| |
Collapse
|
66
|
Zhang L, Lu L, Jiang S, Yin Z, Tan G, Ning F, Qin Z, Huang J, Huang M, Jin J. Salvianolic acid extract prevents Tripterygium wilfordii polyglycosides-induced acute liver injury by modulating bile acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117939. [PMID: 38382651 DOI: 10.1016/j.jep.2024.117939] [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: 12/17/2023] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii polyglycosides (TWP) tablet is the most widely used traditional Chinese medicine preparation for the treatment of rheumatoid arthritis (RA), but the hepatotoxicity often limits its widespread application. In traditional use, Salvia miltiorrhiza has cardioprotective and hepatoprotective effects. Salvianolic acid extract (SA) is a hydrophilic component of Salvia miltiorrhiza and has significant antioxidant and hepatoprotective effects. AIM OF THE STUDY To investigate the protective effects of SA on the TWP-induced acute liver injury in rats and to explore the related mechanisms by integration of metabolomics and transcriptomics. MATERIALS AND METHODS SA and TWP extracts were identified by UPLC-Q/TOF-MS. SA (200 mg/kg) was administered for consecutive 7 days. On day 7, TWP (360 mg/kg) was administered by gavage to induce the acute liver injury in rats. Serum biochemical assay and H&E staining were used to evaluate liver damage. Liver metabolomics and transcriptomics were used to explore the potential mechanisms, and further molecular biological experiments such as qPCR and IHC were utilized to validate the relevant signaling pathways. RESULTS SA can prevent liver injury symptoms caused by TWP, such as elevated liver index, elevated ALT and AST, and pathological changes in liver tissue. Liver metabolomics studies showed that TWP can significantly alter the content of individual bile acid in the liver and SA had the most significant impact on the biosynthetic pathway of bile acids. The transcriptomics results of the liver indicated that the genes changed in the SA + TWP group were mainly involved in sterol metabolism, lipid regulation and bile acid homeostasis pathways. The gene expression of Nr1h4, which encodes farnesoid X receptor (FXR), an important regulator of bile acid homeostasis, was significantly changed. Further studies confirmed that SA can prevent the downregulation of FXR and its downstream signaling induced by TWP, thereby regulating bile acid metabolism, ultimately preventing acute liver injury caused by TWP. CONCLUSION Our results demonstrated that SA could protect the liver from TWP-induced hepatic injury by modulation of the bile acid metabolic pathway. SA may provide a new strategy for the protection against TWP-induced acute liver injury.
Collapse
Affiliation(s)
- Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Langqing Lu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhaokun Yin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guoyao Tan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Fangqing Ning
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhiyan Qin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Junyuan Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
67
|
Baumeister T, Proaño-Vasco A, Metwaly A, Kleigrewe K, Kuznetsov A, Schömig L, Borgmann M, Khiat M, Anand A, Böttcher K, Haller D, Dunkel A, Somoza V, Reiter S, Meng C, Thimme R, Schmid RM, Patil DT, Burgermeister E, Huang Y, Sun Y, Wang HH, Wang TC, Abrams JA, Quante M. Microbiota metabolized Bile Acids accelerate Gastroesophageal Adenocarcinoma via FXR inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598405. [PMID: 38915718 PMCID: PMC11195123 DOI: 10.1101/2024.06.11.598405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Background The incidence of Barrett esophagus (BE) and Gastroesophageal Adenocarcinoma (GEAC) correlates with obesity and a diet rich in fat. Bile acids (BA) support fat digestion and undergo microbial metabolization in the gut. The farnesoid X receptor (FXR) is an important modulator of the BA homeostasis. The capacity of inhibiting cancer-related processes when activated, make FXR an appealing therapeutic target. In this work, we assess the role of diet on the microbiota-BA axis and evaluate the role of FXR in disease progression. Results Here we show that high fat diet (HFD) accelerated tumorigenesis in L2-IL1B mice (BE- and GEAC- mouse model) while increasing BA levels and enriching gut microbiota that convert primary to secondary BA. While upregulated in BE, expression of FXR was downregulated in GEAC in mice and humans. In L2-IL1B mice, FXR knockout enhanced the dysplastic phenotype and increased Lgr5 progenitor cell numbers. Treatment of murine organoids and L2-IL1B mice with the FXR agonist obeticholic acid (OCA) deacelerated GEAC progression. Conclusion We provide a novel concept of GEAC carcinogenesis being accelerated via the diet-microbiome-metabolome axis and FXR inhibition on progenitor cells. Further, FXR activation protected with OCA ameliorated the phenotype in vitro and in vivo, suggesting that FXR agonists have potential as differentiation therapy in GEAC prevention.
Collapse
Affiliation(s)
- Theresa Baumeister
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich; Germany
| | - Andrea Proaño-Vasco
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; Germany
- Faculty of Biology, University of Freiburg; Germany
| | - Amira Metwaly
- Chair of Nutrition and Immunology; Technical University of Munich; Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich; Germany
| | - Alexander Kuznetsov
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; Germany
- Faculty of Biology, University of Freiburg; Germany
| | - Linus Schömig
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; Germany
- Faculty of Biology, University of Freiburg; Germany
| | - Martin Borgmann
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; Germany
- Faculty of Biology, University of Freiburg; Germany
| | - Mohammed Khiat
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
| | - Akanksha Anand
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich; Germany
| | - Katrin Böttcher
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich; Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology; Technical University of Munich; Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich; Germany
- Chair of Food Chemistry and Molecular Sensory Science, TUM School of Life Sciences, Technical University of Munich; Germany
| | - Veronika Somoza
- Leibniz-Institute for Food Systems Biology, Technical University of Munich; Germany
| | - Sinah Reiter
- Chair of Food Chemistry and Molecular Sensory Science, TUM School of Life Sciences, Technical University of Munich; Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich; Germany
| | - Robert Thimme
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
| | - Roland M. Schmid
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich; Germany
| | - Deepa T. Patil
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, USA
| | - Elke Burgermeister
- Dept. of Internal Medicine II, Medical Faculty Mannheim, Heidelberg University; Germany
| | - Yiming Huang
- Systems & Synthetic Biology, Columbia University Medical Center; New York, NY, USA
| | - Yiwei Sun
- Systems & Synthetic Biology, Columbia University Medical Center; New York, NY, USA
| | - Harris H. Wang
- Systems & Synthetic Biology, Columbia University Medical Center; New York, NY, USA
| | - Timothy C. Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Julian A. Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Michael Quante
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg; Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; Germany
| |
Collapse
|
68
|
Aliu A, Bosch DHCA, Keszthelyi D, Rezazadeh Ardabili A, Colombel JF, Sawyer R, Törnblom H, Hart A, Jonkers DMAE, Pierik MJ, Mujagic Z. Review article: A practical approach to persistent gastrointestinal symptoms in inflammatory bowel disease in remission. Aliment Pharmacol Ther 2024; 59:1470-1488. [PMID: 38590140 DOI: 10.1111/apt.17988] [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: 10/16/2023] [Revised: 11/29/2023] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Persistent gastrointestinal symptoms are prevalent in adult patients with inflammatory bowel disease (IBD), even when endoscopic remission is reached. These symptoms can have profound negative effects on the quality of life of affected patients and can be difficult to treat. They may be caused by IBD-related complications or comorbid disorders, but they can also be explained by irritable bowel syndrome (IBS)-like symptoms. AIMS To provide a practical step-by-step guide to diagnose and treat persistent gastrointestinal symptoms in patients with IBD in remission via a personalised approach. METHODS We scrutinised relevant literature on causes, diagnostics and treatment of persistent gastrointestinal symptoms (abdominal pain or discomfort, bloating, abdominal distension, diarrhoea, constipation and faecal incontinence) in patients with IBD in remission. RESULTS A graphical practical guide for several steps in diagnosing, identifying potential triggers and adequate treatment of persistent gastrointestinal symptoms in IBD in remission is provided based on supporting literature. The first part of this review focuses on the diagnostic and treatment approaches for potential IBD-related complications and comorbidities. The second part describes the approach to IBS-like symptoms in IBD in remission. CONCLUSIONS Persistent gastrointestinal symptoms in IBD in remission can be traced back to potential pathophysiological mechanisms in individual patients and can be treated adequately. For both IBD-related complications and comorbidities and IBS-like symptoms in IBD in remission, pharmacological, dietary, lifestyle or psychological treatments can be effective. A systematic and personalised approach is required to reduce the burden for patients, healthcare systems, and society.
Collapse
Affiliation(s)
- Arta Aliu
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Daan H C A Bosch
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Daniel Keszthelyi
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Ashkan Rezazadeh Ardabili
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Jean-Frederic Colombel
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rachel Sawyer
- IBD Patient Advocacy, Founder of the Bottom Line IBD and IBD Women, UK
| | - Hans Törnblom
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ailsa Hart
- IBD Unit, St Mark's Hospital & Imperial College, London, UK
| | - Daisy M A E Jonkers
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Marieke J Pierik
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Zlatan Mujagic
- Department Gastroenterology and Hepatology, Maastricht University Medical Center+, Maastricht, the Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
69
|
Lee NY, Choi MG, Lee EJ, Koo JH. Interplay between YAP/TAZ and metabolic dysfunction-associated steatotic liver disease progression. Arch Pharm Res 2024; 47:558-570. [PMID: 38874747 PMCID: PMC11217110 DOI: 10.1007/s12272-024-01501-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming an increasingly pressing global health challenge, with increasing mortality rates showing an upward trend. Two million deaths occur annually from cirrhosis and liver cancer together each year. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key effectors of the Hippo signaling pathway, critically regulate tissue homeostasis and disease progression in the liver. While initial studies have shown that YAP expression is normally restricted to cholangiocytes in healthy livers, the activation of YAP/TAZ is observed in other hepatic cells during chronic liver disease. The disease-driven dysregulation of YAP/TAZ appears to be a critical element in the MASLD progression, contributing to hepatocyte dysfunction, inflammation, and fibrosis. In this study, we focused on the complex roles of YAP/TAZ in MASLD and explored how the YAP/TAZ dysregulation of YAP/TAZ drives steatosis, inflammation, fibrosis, and cirrhosis. Finally, the cell-type-specific functions of YAP/TAZ in different types of hepatic cells, such as hepatocytes, hepatic stellate cells, hepatic macrophages, and biliary epithelial cells are discussed, highlighting the multifaceted impact of YAP/TAZ on liver physiology and pathology.
Collapse
Affiliation(s)
- Na Young Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Myeung Gi Choi
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Eui Jin Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Ja Hyun Koo
- Research Institute of Pharmaceutical Sciences and Natural Products Research Institute, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
70
|
Tang Y, Fan Y, Wang Y, Wang D, Huang Q, Chen T, Cao X, Wen C, Shen X, Li J, You Y. A Current Understanding of FXR in NAFLD: The multifaceted regulatory role of FXR and novel lead discovery for drug development. Biomed Pharmacother 2024; 175:116658. [PMID: 38701562 DOI: 10.1016/j.biopha.2024.116658] [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: 02/13/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
The global prevalence of nonalcoholic fatty liver disease (NAFLD) has reached 30 %, with an annual increase. The incidence of NAFLD-induced cirrhosis is rapidly rising and has become the leading indicator for liver transplantation in the US. However, there are currently no US Food and Drug Administration-approved drugs for NAFLD. Increasing evidence underscores the close association between NAFLD and bile acid metabolism disorder, highlighting the feasibility of targeting the bile acid signaling pathway for NAFLD treatment. The farnesoid X receptor (FXR) is an endogenous receptor for bile acids that exhibits favorable effects in ameliorating the metabolic imbalance of bile acids, lipid disorders, and disruption of intestinal homeostasis, all of which are key characteristics of NAFLD, making FXR a promising therapeutic target for NAFLD. The present review provides a comprehensive overview of the diverse mechanisms through which FXR improves NAFLD, with particular emphasis on its involvement in regulating bile acid homeostasis and the recent advancements in drug development targeting FXR for NAFLD treatment.
Collapse
Affiliation(s)
- Yuhong Tang
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Yujuan Fan
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Yiming Wang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dong Wang
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Qingyu Huang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongqing Chen
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Xinyue Cao
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Cailing Wen
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Shen
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China.
| | - Jian Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Yan You
- School of Pharmacy & Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
71
|
Malin SK, Syeda UA. Exercise Training Independent of Intensity Lowers Plasma Bile Acids in Prediabetes. Med Sci Sports Exerc 2024; 56:1009-1017. [PMID: 38190376 PMCID: PMC11096085 DOI: 10.1249/mss.0000000000003384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
INTRODUCTION People with obesity have high circulating bile acids (BA). Although aerobic fitness favors low circulating BA, the effect of training intensity before clinically meaningful weight loss on BA is unclear. This study aimed to test the hypothesis that 2 wk of interval (INT) versus continuous (CONT) exercise would lower plasma BA in relation to insulin sensitivity. METHODS Twenty-three older adults with prediabetes (ADA criteria) were randomized to 12 work-matched bouts of INT ( n = 11, 60.3 ± 2.4 yr, 32.1 ± 1.2 kg·m -2 ) at 3 min at 50% HR peak and 3 min at 90% HR peak or CONT ( n = 12, 60.8 ± 2.4 yr, 34.0 ± 1.7 kg·m -2 ) at 70% HR peak cycling training for 60 min·d -1 over 2 wk. A 180-min 75-g oral glucose tolerance test (OGTT) was performed to assess glucose tolerance (tAUC), insulin sensitivity (Siis), and metabolic flexibility (RER postprandial -RER fast ; indirect calorimetry). BA ( n = 8 conjugated and 7 unconjugated) were analyzed at 0, 30, and 60 min of the OGTT. Anthropometrics and fitness (V̇O 2peak ) were also assessed. RESULTS INT and CONT comparably reduced body mass index (BMI; P < 0.001) and fasting RER ( P < 0.001) but raised insulin sensitivity ( P = 0.03). INT increased V̇O 2peak as compared with CONT ( P = 0.01). Exercise decreased the unconjugated BA chenodeoxycholic acid iAUC 60min ( P < 0.001), deoxycholic acid iAUC 60min ( P < 0.001), lithocholic acid iAUC 60min ( P < 0.001), and glycodeoxycholic acid (GCDCA) iAUC 60min ( P < 0.001). Comparable reductions were also seen in the conjugated BA hyodeoxycholic acid iAUC 60min ( P = 0.01) and taurolithocholic acid iAUC 60min ( P = 0.007). Increased V̇O 2peak was associated with lowered UDCA 0min ( r = -0.56, P = 0.02) and cholic acid iAUC 60min ( r = -0.60, P = 0.005), whereas reduced BMI was related to higher GDCA 0min ( r = 0.60, P = 0.005) and GCDCA 0min ( r = 0.53, P = 0.01). Improved insulin sensitivity correlated with lower GCDCA iAUC 60min ( r = -0.45, P = 0.03) and GDCA iAUC 60min ( r = -0.48, P = 0.02), whereas increased metabolic flexibility was related to deoxycholic acid iAUC 60min ( r = 0.64, P = 0.004) and GCDCA iAUC 60min ( r = 0.43, P = 0.05). CONCLUSIONS Short-term training lowers some BA in relation to insulin sensitivity independent of intensity.
Collapse
Affiliation(s)
- Steven K. Malin
- Department of Kinesiology & Health, New Brunswick, NJ
- Department of Kinesiology, University of Virginia, Charlottesville, VA
- Division of Endocrinology, Metabolism & Nutrition; Department of Medicine, Rutgers University, New Brunswick, NJ
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, NJ
| | | |
Collapse
|
72
|
Shi J, Zhu L, Tang BY, Yang WQ, Xi SY, Zhang CL, Li PF, Wang YJ, Guo KH, Huang JR, Huang CR, Yu ZX, Yu BK, Zhang CF, Zhang YM. Regulatory effect of Yinchenhao decoction on bile acid metabolism to improve the inflammatory microenvironment of hepatocellular carcinoma in mice. J Nat Med 2024; 78:633-643. [PMID: 38704807 DOI: 10.1007/s11418-024-01812-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with extremely high mortality. The tumor microenvironment is the "soil" of its occurrence and development, and the inflammatory microenvironment is an important part of the "soil". Bile acid is closely related to the occurrence of HCC. Bile acid metabolism disorder is not only directly involved in the occurrence and development of HCC but also affects the inflammatory microenvironment of HCC. Yinchenhao decoction, a traditional Chinese medicine formula, can regulate bile acid metabolism and may affect the inflammatory microenvironment of HCC. To determine the effect of Yinchenhao decoction on bile acid metabolism in mice with HCC and to explore the possible mechanism by which Yinchenhao decoction improves the inflammatory microenvironment of HCC by regulating bile acid metabolism, we established mice model of orthotopic transplantation of hepatocellular carcinoma. These mice were treated with three doses of Yinchenhao decoction, then liver samples were collected and tested. Yinchenhao decoction can regulate the disorder of bile acid metabolism in liver cancer mice. Besides, it can improve inflammatory reactions, reduce hepatocyte degeneration and necrosis, and even reduce liver weight and the liver index. Taurochenodeoxycholic acid, hyodeoxycholic acid, and taurohyodeoxycholic acid are important molecules in the regulation of the liver inflammatory microenvironment, laying a foundation for the regulation of the liver tumor inflammatory microenvironment based on bile acids. Yinchenhao decoction may improve the inflammatory microenvironment of mice with HCC by ameliorating hepatic bile acid metabolism.
Collapse
Affiliation(s)
- Jian Shi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Zhu
- Department of Traditional Chinese Medicine, Binhai County People's Hospital, Yancheng, China
| | - Bang-Yi Tang
- Department of Science and Education, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, China
| | - Wan-Qing Yang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Sheng-Yan Xi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chen-Long Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Peng-Fei Li
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yu-Jie Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Kai-Hang Guo
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Jing-Ru Huang
- Central Laboratory, School of Medicine, Xiamen University, Xiamen, China
| | - Chen-Rui Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou-Xin Yu
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Bao-Kang Yu
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chun-Fang Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China.
| | - Yu-Mei Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
73
|
Liu G, Yi Y, Wang Y, Feng Y, Lin M, Yan X, Wang J, Ning X, Ma N. Assessment of the Risk of Malnutrition or Frailty Among Patients Undergoing Liver Transplantation: A Hospital-Based Prospective Study. Int J Gen Med 2024; 17:2347-2354. [PMID: 38799201 PMCID: PMC11128220 DOI: 10.2147/ijgm.s448154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Objective We aimed to explore the status of nutritional and frailty in patients undergoing liver transplantation and the associated influencing factors. Methods We conducted a follow-up analysis of 44 patients who underwent liver transplantation between 2021 and 2022. We followed up and recorded the nutritional status and risk of weakness at different time-points (days 1, 2, 3, 6, 9, and 12) postoperatively. Patient information regarding demographics, physical examination, medical history, and perioperative blood tests were collected. Binary logistic regression was applied to identify risk factors for weakness after liver transplantation. Results The cohort comprised 44 liver transplant recipients, with a mean age of 47.66 years (standard deviation=9.49 years). Initial analysis revealed that, compared to the group without nutritional risks, the group with nutritional risks displayed elevated age and preoperative blood ammonia levels one week post-surgery. Moreover, this group had reduced levels of albumin and total bile acid preoperatively. Patients with preoperative nutritional risks were also prone to similar risks 2 weeks postoperatively. Further, a correlation was observed between preoperative pulmonary infections and increased frailty risk 6 days postoperatively. At both 9 and 12 days postoperatively, patients with frailty risk exhibited higher preoperative white blood cell counts and ammonia levels than those without. Multivariable analysis, controlling for confounding factors, indicated a significant association between preoperative nutritional status and nutritional risk 2 weeks postoperatively, as well as a link between preoperative white blood cell count and frailty risk at 12 days postoperatively. Conclusion There was a significant correlation between preoperative nutritional status and nutritional risk 2 weeks after liver transplantation, and preoperative white blood cell count was an independent risk factor for weakness 12 days postoperatively. Preoperative nutritional management for patients could potentially mitigate the likelihood of adverse clinical outcomes.
Collapse
Affiliation(s)
- Guiqing Liu
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Yuanyuan Yi
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Yanni Wang
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Yuru Feng
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Minyi Lin
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xu Yan
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Jinghua Wang
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xianjia Ning
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Nan Ma
- Department of Liver Surgery (Liver Transplantation), Shenzhen Third People’s Hospital and the Second Hospital Affiliated with the Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
74
|
Chen LM, Qian ST, Li ZQ, He MF, Li HJ. Psoralen and Isopsoralen, Two Estrogen -Like Natural Products from Psoraleae Fructus, Induced Cholestasis via Activation of ERK1/2. Chem Res Toxicol 2024; 37:804-813. [PMID: 38646980 DOI: 10.1021/acs.chemrestox.4c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
With the increasing use of oral contraceptives and estrogen replacement therapy, the incidence of estrogen-induced cholestasis (EC) has tended to rise. Psoralen (P) and isopsoralen (IP) are the major bioactive components in Psoraleae Fructus, and their estrogen-like activities have already been recognized. Recent studies have also reported that ERK1/2 plays a critical role in EC in mice. This study aimed to investigate whether P and IP induce EC and reveal specific mechanisms. It was found that P and IP increased the expression of esr1, cyp19a1b and the levels of E2 and VTG at 80 μM in zebrafish larvae. Exemestane (Exe), an aromatase antagonist, blocked estrogen-like activities of P and IP. At the same time, P and IP induced cholestatic hepatotoxicity in zebrafish larvae with increasing liver fluorescence areas and bile flow inhibition rates. Further mechanistic analysis revealed that P and IP significantly decreased the expression of bile acids (BAs) synthesis genes cyp7a1 and cyp8b1, BAs transport genes abcb11b and slc10a1, and BAs receptor genes nr1h4 and nr0b2a. In addition, P and IP caused EC by increasing the level of phosphorylation of ERK1/2. The ERK1/2 antagonists GDC0994 and Exe both showed significant rescue effects in terms of cholestatic liver injury. In conclusion, we comprehensively studied the specific mechanisms of P- and IP-induced EC and speculated that ERK1/2 may represent an important therapeutic target for EC induced by phytoestrogens.
Collapse
Affiliation(s)
- Liang-Min Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Si-Tong Qian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Zhuo-Qing Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ming-Fang He
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
75
|
Xue C, Jia H, Cao R, Cai W, Hong W, Tu J, Wang S, Jiang Q, Bi C, Shan A, Dong N. Oleanolic acid improved intestinal immune function by activating and potentiating bile acids receptor signaling in E. coli-challenged piglets. J Anim Sci Biotechnol 2024; 15:79. [PMID: 38760843 PMCID: PMC11102245 DOI: 10.1186/s40104-024-01037-0] [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: 12/30/2023] [Accepted: 04/18/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Infection with pathogenic bacteria during nonantibiotic breeding is one of the main causes of animal intestinal diseases. Oleanolic acid (OA) is a pentacyclic triterpene that is ubiquitous in plants. Our previous work demonstrated the protective effect of OA on intestinal health, but the underlying molecular mechanisms remain unclear. This study investigated whether dietary supplementation with OA can prevent diarrhea and intestinal immune dysregulation caused by enterotoxigenic Escherichia coli (ETEC) in piglets. The key molecular role of bile acid receptor signaling in this process has also been explored. RESULTS Our results demonstrated that OA supplementation alleviated the disturbance of bile acid metabolism in ETEC-infected piglets (P < 0.05). OA supplementation stabilized the composition of the bile acid pool in piglets by regulating the enterohepatic circulation of bile acids and significantly increased the contents of UDCA and CDCA in the ileum and cecum (P < 0.05). This may also explain why OA can maintain the stability of the intestinal microbiota structure in ETEC-challenged piglets. In addition, as a natural ligand of bile acid receptors, OA can reduce the severity of intestinal inflammation and enhance the strength of intestinal epithelial cell antimicrobial programs through the bile acid receptors TGR5 and FXR (P < 0.05). Specifically, OA inhibited NF-κB-mediated intestinal inflammation by directly activating TGR5 and its downstream cAMP-PKA-CREB signaling pathway (P < 0.05). Furthermore, OA enhanced CDCA-mediated MEK-ERK signaling in intestinal epithelial cells by upregulating the expression of FXR (P < 0.05), thereby upregulating the expression of endogenous defense molecules in intestinal epithelial cells. CONCLUSIONS In conclusion, our findings suggest that OA-mediated regulation of bile acid metabolism plays an important role in the innate immune response, which provides a new diet-based intervention for intestinal diseases caused by pathogenic bacterial infections in piglets.
Collapse
Affiliation(s)
- Chenyu Xue
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Hongpeng Jia
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Rujing Cao
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Wenjie Cai
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Weichen Hong
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Jianing Tu
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Songtao Wang
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Qianzhi Jiang
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Chongpeng Bi
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Anshan Shan
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Na Dong
- The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China.
| |
Collapse
|
76
|
Zhang W, Jiang Y, Ni Z, Zhou M, Liu L, Li X, Su S, Wang C. Identification of Copy Number Variations and Selection Signatures in Wannan Spotted Pigs by Whole Genome Sequencing Data: A Preliminary Study. Animals (Basel) 2024; 14:1419. [PMID: 38791637 PMCID: PMC11117326 DOI: 10.3390/ani14101419] [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: 04/07/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Copy number variation (CNV) is an important structural variation used to elucidate complex economic traits. In this study, we sequenced 25 Wannan spotted pigs (WSPs) to detect their CNVs and identify their selection signatures compared with those of 10 Asian wild boars. A total of 14,161 CNVs were detected in the WSPs, accounting for 0.72% of the porcine genome. The fixation index (Fst) was used to identify the selection signatures, and 195 CNVs with the top 1% of the Fst value were selected. Eighty genes were identified in the selected CNV regions. Functional GO and KEGG analyses revealed that the genes within these selected CNVs are associated with key traits such as reproduction (GAL3ST1 and SETD2), fatty acid composition (PRKG1, ACACA, ACSL3, UGT8), immune system (LYZ), ear size (WIF1), and feed efficiency (VIPR2). The findings of this study contribute novel insights into the genetic CNVs underlying WSP characteristics and provide essential information for the protection and utilization of WSP populations.
Collapse
Affiliation(s)
- Wei Zhang
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Yao Jiang
- National Animal Husbandry Service, Beijing 100125, China;
| | - Zelan Ni
- Anhui Provincial Livestock and Poultry Genetic Resources Conservation Center, Hefei 231283, China;
| | - Mei Zhou
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Linqing Liu
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Xiaoyu Li
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Shiguang Su
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Chonglong Wang
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| |
Collapse
|
77
|
Okada J, Landgraf A, Xiaoli AM, Liu L, Horton M, Schuster VL, Yang F, Sidoli S, Qiu Y, Kurland IJ, Eliscovich C, Shinoda K, Pessin JE. Spatial hepatocyte plasticity of gluconeogenesis during the metabolic transitions between fed, fasted and starvation states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591168. [PMID: 38746329 PMCID: PMC11092462 DOI: 10.1101/2024.04.29.591168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The liver acts as a master regulator of metabolic homeostasis in part by performing gluconeogenesis. This process is dysregulated in type 2 diabetes, leading to elevated hepatic glucose output. The parenchymal cells of the liver (hepatocytes) are heterogeneous, existing on an axis between the portal triad and the central vein, and perform distinct functions depending on location in the lobule. Here, using single cell analysis of hepatocytes across the liver lobule, we demonstrate that gluconeogenic gene expression ( Pck1 and G6pc ) is relatively low in the fed state and gradually increases first in the periportal hepatocytes during the initial fasting period. As the time of fasting progresses, pericentral hepatocyte gluconeogenic gene expression increases, and following entry into the starvation state, the pericentral hepatocytes show similar gluconeogenic gene expression to the periportal hepatocytes. Similarly, pyruvate-dependent gluconeogenic activity is approximately 10-fold higher in the periportal hepatocytes during the initial fasting state but only 1.5-fold higher in the starvation state. In parallel, starvation suppresses canonical beta-catenin signaling and modulates expression of pericentral and periportal glutamine synthetase and glutaminase, resulting in an enhanced pericentral glutamine-dependent gluconeogenesis. These findings demonstrate that hepatocyte gluconeogenic gene expression and gluconeogenic activity are highly spatially and temporally plastic across the liver lobule, underscoring the critical importance of using well-defined feeding and fasting conditions to define the basis of hepatic insulin resistance and glucose production.
Collapse
|
78
|
Fleishman JS, Kumar S. Bile acid metabolism and signaling in health and disease: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:97. [PMID: 38664391 PMCID: PMC11045871 DOI: 10.1038/s41392-024-01811-6] [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: 11/28/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.
Collapse
Affiliation(s)
- Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
79
|
Zhang F, Lo EKK, Chen J, Wang K, Felicianna, Ismaiah MJ, Leung HKM, Zhao D, Lee JCY, El-Nezami H. Probiotic Mixture Ameliorates a Diet-Induced MASLD/MASH Murine Model through the Regulation of Hepatic Lipid Metabolism and the Gut Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8536-8549. [PMID: 38575146 PMCID: PMC11037262 DOI: 10.1021/acs.jafc.3c08910] [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: 11/28/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disease that has no effective treatment. Our proprietary probiotic mixture, Prohep, has been proven in a previous study to be helpful in reducing hepatocellular carcinoma (HCC) in vivo. However, its prospective benefits on the treatment of other liver diseases such as MASLD, which is one of the major risk factors in the development of HCC, are unclear. To investigate the potential of Prohep in modulating the development and progression of MASLD, we first explored the effect of Prohep supplementation via voluntary intake in a high-fat diet (HFD)-induced MASLD/metabolic dysfunction-associated steatohepatitis (MASH) murine model. Our results indicated that Prohep alleviated HFD-induced liver steatosis and reduced excessive hepatic lipid accumulation and improved the plasma lipid profile when compared with HFD-fed control mice through suppressing hepatic de novo lipogenesis and cholesterol biosynthesis gene expressions. In addition, Prohep was able to modulate the gut microbiome, modify the bile acid (BA) profile, and elevate fecal short-chain fatty acid (SCFA) levels. Next, in a prolonged HFD-feeding MASLD/MASH model, we observed the effectiveness of Prohep in preventing the transition from MASLD to MASH via amelioration in hepatic steatosis, inflammation, and fibrosis. Taken together, Prohep could ameliorate HFD-induced MASLD and control the MASLD-to-MASH progression in mice. Our findings provide distinctive insights into the development of novel microbial therapy for the management of MASLD and MASH.
Collapse
Affiliation(s)
- Fangfei Zhang
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Emily Kwun Kwan Lo
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Jiarui Chen
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong
Kong 000, S.A.R., China
- Department
of Medicine, The University of Hong Kong, Hong Kong 000, S.A.R., China
- Leibniz
Institute for Natural Product Research and Infection Biology, Hans
Knöll Institute-Microbiome Dynamics, Jena D-07745, Germany
| | - Ke Wang
- Department
of Food Science and Nutrition, The Hong
Kong Polytechnic University, Hong
Kong 000, S.A.R., China
- Research
Institute for Future Food, The Hong Kong
Polytechnic University, Hong Kong 000, S.A.R., China
| | - Felicianna
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Marsena Jasiel Ismaiah
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Hoi Kit Matthew Leung
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Danyue Zhao
- Department
of Food Science and Nutrition, The Hong
Kong Polytechnic University, Hong
Kong 000, S.A.R., China
- Research
Institute for Future Food, The Hong Kong
Polytechnic University, Hong Kong 000, S.A.R., China
| | - Jetty Chung-Yung Lee
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Hani El-Nezami
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
- Institute
of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio FI-70211, Finland
| |
Collapse
|
80
|
Abdul Waheed MI, Jaiswal A, Yelne S, Nandanwar V. Navigating Perinatal Challenges: A Comprehensive Review of Cholestasis of Pregnancy and Its Impact on Maternal and Fetal Health. Cureus 2024; 16:e58699. [PMID: 38779244 PMCID: PMC11109475 DOI: 10.7759/cureus.58699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Cholestasis of pregnancy (CP), or intrahepatic CP (ICP), represents a condition peculiar to pregnancy, marked by impaired bile acid flow and consequent accumulation in the maternal bloodstream. Primarily emerging in the third trimester, CP is linked with considerable risks to both the mother and fetus, including heightened incidences of preterm birth, fetal distress, and stillbirth, alongside maternal complications such as intense pruritus and liver dysfunction. Despite its clinical significance, the etiology of CP, which involves genetic, hormonal, and environmental factors, remains partially understood. This comprehensive review delves into the physiology and pathophysiology of CP, outlines its clinical manifestations and diagnostic criteria, and discusses the associated maternal and fetal complications. Furthermore, it evaluates current management strategies, prognostic implications, and potential long-term effects on maternal and child health. It also explores future research directions, emphasizing the need for advancements in understanding the pathophysiology of CP, developing novel therapeutic interventions, and improving risk stratification models. By offering a thorough overview of CP, this review aims to enhance clinical awareness, guide management practices, and identify areas requiring further investigation, ultimately contributing to better health outcomes for affected women and their babies.
Collapse
Affiliation(s)
- Mohammed Irfan Abdul Waheed
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arpita Jaiswal
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Seema Yelne
- Nursing, Shalinitai Meghe College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Varsha Nandanwar
- Nursing, Shalinitai Meghe College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
81
|
HAN M, YI X, YOU S, WU X, WANG S, HE D. Gehua Jiejiu Dizhi decoction ameliorates alcoholic fatty liver in mice by regulating lipid and bile acid metabolism and with exertion of antioxidant stress based on 4DLabel-free quantitative proteomic study. J TRADIT CHIN MED 2024; 44:277-288. [PMID: 38504534 PMCID: PMC10927405 DOI: 10.19852/j.cnki.jtcm.20231018.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/27/2023] [Indexed: 03/21/2024]
Abstract
OBJECTIVE To analyze the effect and molecular mechanism of Gehua Jiejiu Dizhi decoction (, GJDD) on alcoholic fatty live disease (AFLD) by using proteomic methods. METHODS The male C57BL/6J mouse were randomly divided into four groups: control group, model group, GJDD group and resveratrol group. After the AFLD model was successfully prepared by intragastric administration of alcohol once on the basis of the Lieber-DeCarli classical method, the GJDD group and resveratrol group were intragastrically administered with GJDD (4900 mg/kg) and resveratrol (400 mg/kg) respectively, once a day for 9 d. The fat deposition of liver tissue was observed and evaluated by oil red O (ORO) staining. 4DLabel-free quantitative proteome method was used to determine and quantify the protein expression in liver tissue of each experimental group. The differentially expressed proteins were screened according to protein expression differential multiples, and then analyzed by Gene ontology classification and Kyoto Encyclopedia of Genes and Genomes pathway enrichment. Finally, expression validation of the differentially co-expressed proteins from control group, model group and GJDD group were verified by targeted proteomics quantification techniques. RESULTS In semiquantitative analyses of ORO, all kinds of steatosis (ToS, MaS, and MiS) were evaluated higher in AFLD mice compared to those in GJDD or resveratrol-treated mice. 4DLabel-free proteomics analysis results showed that a total of 4513 proteins were identified, of which 3763 proteins were quantified and 946 differentially expressed proteins were screened. Compared with the control group, 145 proteins were up-regulated and 148 proteins were down-regulated in the liver tissue of model group. In addition, compared with the model group, 92 proteins were up-regulated and 135 proteins were down-regulated in the liver tissue of the GJDD group. 15 differentially co-expressed proteins were found between every two groups (model group vs control group, GJDD group vs model group and GJDD group vs control group), which were involved in many biological processes. Among them, 11 differentially co-expressed key proteins (Aox3, H1-5, Fabp5, Ces3a, Nudt7, Serpinb1a, Fkbp11, Rpl22l1, Keg1, Acss2 and Slco1a1) were further identified by targeted proteomic quantitative technology and their expression patterns were consistent with the results of 4D label-free proteomic analysis. CONCLUSIONS Our study provided proteomics-based evidence that GJDD alleviated AFLD by modulating liver protein expression, likely through the modulation of lipid metabolism, bile acid metabolism and with exertion of antioxidant stress.
Collapse
Affiliation(s)
- Min HAN
- 1 Guizhou University of Traditional Chinese Medicine, Graduate School, Guiyang 550025, China
| | - Xu YI
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Shaowei YOU
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Xueli WU
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Shuoshi WANG
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Diancheng HE
- 2 Department of Clinical medical laboratory, Department of Gastroenterology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| |
Collapse
|
82
|
Chen J, Qin Y, Li Z, Shan A, Ma Q. Aromatic Amino Acids Promote Lipid Metabolism Disorders by Increasing Hepatic Bile Acid Synthesis. J Nutr 2024; 154:1321-1332. [PMID: 38582699 DOI: 10.1016/j.tjnut.2023.12.047] [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: 10/19/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Obesity is a progressive metabolic disease that begins with lipid metabolism disorders. Aromatic amino acids (AAAs), including tryptophan, phenylalanine, and tyrosine, have diverse biological activities as nutrients. However, the underlying mechanisms by which AAAs affect lipid metabolism are unclear. OBJECTIVES This study was designed to investigate the possible roles and underlying molecular mechanisms of AAA in the pathogenesis of lipid metabolism disorders. METHODS We added an AAA mixture to the high-fat diet (HFD) of mice. Glucose tolerance test was recorded. Protein expression of hepatic bile acid (BA) synthase and mRNA expression of BA metabolism-related genes were determined. Hepatic BA profiles and gut microbial were also determined in mice. RESULTS The results showed that AAA significantly increased body weight and white adipose tissue, aggravated liver injury, impaired glucose tolerance and intestinal integrity, and significantly increased hepatic BA synthesis by inhibiting intestinal farnesoid X receptor (FXR). Moreover, AAA increased the content of total BA in the liver and altered the hepatic BA profile, with elevated levels of lithocholic acid, glycochenodeoxycholic acid, and glycoursodeoxycholic acid. AAA markedly increased the levels of proteins involved in BA synthesis (cholesterol 7α-hydroxylase and oxysterol 7α-hydroxylase) and inhibited the intestinal FXR. Gut microbial composition also changed, reducing the abundance of some beneficial bacteria, such as Parvibacter and Lactobacillus. CONCLUSIONS Under HFD conditions, AAAs stimulate BA synthesis in both the classical and alternative pathways, leading to aggravation of liver injury and fat deposition. Excessive intake of AAA disrupts BA metabolism and contributes to the development of lipid metabolism disorders, suggesting that AAA may be a causative agent of lipid metabolism disorders.
Collapse
Affiliation(s)
- Jiayi Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yingjie Qin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
83
|
Mohanty I, Mannochio-Russo H, Schweer JV, El Abiead Y, Bittremieux W, Xing S, Schmid R, Zuffa S, Vasquez F, Muti VB, Zemlin J, Tovar-Herrera OE, Moraïs S, Desai D, Amin S, Koo I, Turck CW, Mizrahi I, Kris-Etherton PM, Petersen KS, Fleming JA, Huan T, Patterson AD, Siegel D, Hagey LR, Wang M, Aron AT, Dorrestein PC. The underappreciated diversity of bile acid modifications. Cell 2024; 187:1801-1818.e20. [PMID: 38471500 DOI: 10.1016/j.cell.2024.02.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/30/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
The repertoire of modifications to bile acids and related steroidal lipids by host and microbial metabolism remains incompletely characterized. To address this knowledge gap, we created a reusable resource of tandem mass spectrometry (MS/MS) spectra by filtering 1.2 billion publicly available MS/MS spectra for bile-acid-selective ion patterns. Thousands of modifications are distributed throughout animal and human bodies as well as microbial cultures. We employed this MS/MS library to identify polyamine bile amidates, prevalent in carnivores. They are present in humans, and their levels alter with a diet change from a Mediterranean to a typical American diet. This work highlights the existence of many more bile acid modifications than previously recognized and the value of leveraging public large-scale untargeted metabolomics data to discover metabolites. The availability of a modification-centric bile acid MS/MS library will inform future studies investigating bile acid roles in health and disease.
Collapse
Affiliation(s)
- Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Helena Mannochio-Russo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Joshua V Schweer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Department of Chemistry and Biochemistry, University of California, San Diego, San Diego, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Wout Bittremieux
- Department of Computer Science, University of Antwerp, 2020 Antwerpen, Belgium
| | - Shipei Xing
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Department of Chemistry, Faculty of Science, University of British Columbia, Vancouver Campus, Vancouver, BC, Canada
| | - Robin Schmid
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Felipe Vasquez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Valentina B Muti
- Department of Computer Science and Engineering, University of California, Riverside, Riverside, CA, USA; Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80210, USA
| | - Jasmine Zemlin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - Omar E Tovar-Herrera
- Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel; Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Sarah Moraïs
- Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel; Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Dhimant Desai
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, USA
| | - Shantu Amin
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, USA
| | - Imhoi Koo
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Christoph W Turck
- Max Planck Institute of Psychiatry, Proteomics and Biomarkers, Kraepelinstrasse 2-10, Munich 80804, Germany; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Itzhak Mizrahi
- Department of Life Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel; Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Kristina S Petersen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Jennifer A Fleming
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Tao Huan
- Department of Chemistry, Faculty of Science, University of British Columbia, Vancouver Campus, Vancouver, BC, Canada
| | - Andrew D Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Lee R Hagey
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Mingxun Wang
- Department of Computer Science and Engineering, University of California, Riverside, Riverside, CA, USA
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80210, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
84
|
Wang J, Yang N, Xu Y. Natural Products in the Modulation of Farnesoid X Receptor Against Nonalcoholic Fatty Liver Disease. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:291-314. [PMID: 38480498 DOI: 10.1142/s0192415x24500137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health concern with a high prevalence and increasing economic burden, but official medicine remains unavailable. Farnesoid X receptor (FXR), a nuclear receptor member, is one of the most promising drug targets for NAFLD therapy that plays a crucial role in modulating bile acid, glucose, and lipid homeostasis, as well as inhibits hepatic inflammation and fibrosis. However, the rejection of the FXR agonist, obecholic acid, by the Food and Drug Administration for treating hepatic fibrosis raises a question about the functions of FXR in NAFLD progression and the therapeutic strategy to be used. Natural products, such as FXR modulators, have become the focus of attention for NAFLD therapy with fewer adverse reactions. The anti-NAFLD mechanisms seem to act as FXR agonists and antagonists or are involved in the FXR signaling pathway activation, indicating a promising target of FXR therapeutic prospects using natural products. This review discusses the effective mechanisms of FXR in NAFLD alleviation, and summarizes currently available natural products such as silymarin, glycyrrhizin, cycloastragenol, berberine, and gypenosides, for targeting FXR, which can facilitate development of naturally targeted drug by medicinal specialists for effective treatment of NAFLD.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, P. R. China
| | - Na Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, P. R. China
| | - Yu Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Cailun Road 1200, Shanghai 201203, P. R. China
| |
Collapse
|
85
|
Li Y, Zhu C, Yao J, Zhu C, Li Z, Liu HY, Zhu M, Li K, Ahmed AA, Li S, Hu P, Cai D. Lithocholic Acid Alleviates Deoxynivalenol-Induced Inflammation and Oxidative Stress via PPARγ-Mediated Epigenetically Transcriptional Reprogramming in Porcine Intestinal Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5452-5462. [PMID: 38428036 DOI: 10.1021/acs.jafc.3c08044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Deoxynivalenol (DON) is a common mycotoxin that induces intestinal inflammation and oxidative damage in humans and animals. Given that lithocholic acid (LCA) has been suggested to inhibit intestinal inflammation, we aimed to investigate the protective effects of LCA on DON-exposed porcine intestinal epithelial IPI-2I cells and the underlying mechanisms. Indeed, LCA rescued DON-induced cell death in IPI-2I cells and reduced DON-stimulated inflammatory cytokine levels and oxidative stress. Importantly, the nuclear receptor PPARγ was identified as a key transcriptional factor involved in the DON-induced inflammation and oxidative stress processes in IPI-2I cells. The PPARγ function was found compromised, likely due to the hyperphosphorylation of the p38 and ERK signaling pathways. In contrast, the DON-induced inflammatory responses and oxidative stress were restrained by LCA via PPARγ-mediated reprogramming of the core inflammatory and antioxidant genes. Notably, the PPARγ-modulated transcriptional regulations could be attributed to the altered recruitments of coactivator SRC-1/3 and corepressor NCOR1/2, along with the modified histone marks H3K27ac and H3K18la. This study emphasizes the protective actions of LCA on DON-induced inflammatory damage and oxidative stress in intestinal epithelial cells via PPARγ-mediated epigenetically transcriptional reprogramming, including histone acetylation and lactylation.
Collapse
Affiliation(s)
- Yanwei Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Chuyang Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Jiacheng Yao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Cuipeng Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Zhaojian Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Miaonan Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Kaiqi Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
| | - Abdelkareem A Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Natural Resources, Gaborone 0027, Botswana
| | - Shicheng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, P. R. China
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, P. R. China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, P. R. China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, P. R. China
| |
Collapse
|
86
|
Nabizadeh F, Valizadeh P, Fallahi MS. Bile acid profile associated with CSF and PET biomarkers in Alzheimer's disease. Aging Clin Exp Res 2024; 36:62. [PMID: 38451317 PMCID: PMC10920417 DOI: 10.1007/s40520-024-02729-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Recent studies have shown that gut microbiota can affect the development of Alzheimer's disease (AD) through various mechanisms. Bile acids (BAs), which are the final byproducts of cholesterol metabolism created through both the human body and gut microbiome, appear to be influenced by gut microbiota and may impact AD pathological characteristics such as the accumulation of tau and amyloid-β. We aimed to investigate the associations between various serum BAs and CSF biomarkers (including Aβ, total tau, and p-tau). Additionally, we sought to examine the longitudinal changes in brain Aβ and tau through PET imaging in relation to BAs profile. METHODS The data of 828 subjects including 491 diagnosed with mild cognitive impairment (MCI), 119 patients diagnosed with AD, and 267 cognitively normal (CN) participants were obtained from ADNI. The baseline and longitudinal [18F] florbetapir and [18F] flortaucipir PET standard uptake value ratios (SUVR) measures were obtained to assess the accumulation of tau and Aβ. Moreover, baseline levels of serum BAs and CSF Aβ1-42, tau, and p-tau were used. RESULTS After FDR correction we observed that five BAs level and relevant calculated ratios were associated with CSF p-tau and tau, three with CSF Aβ1-42. Furthermore, three BAs level and relevant calculated ratios were associated with the tau-PET rate of change, and two with the Aβ rate of change. CONCLUSION The findings from our study suggest a correlation between altered profiles of BAs and CSF and imaging biomarkers associated with AD. These results provide supporting evidence for the link between the gut microbiome and the pathological features of AD.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Parya Valizadeh
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
87
|
Talavera Andújar B, Mary A, Venegas C, Cheng T, Zaslavsky L, Bolton EE, Heneka MT, Schymanski EL. Can Small Molecules Provide Clues on Disease Progression in Cerebrospinal Fluid from Mild Cognitive Impairment and Alzheimer's Disease Patients? ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:4181-4192. [PMID: 38373301 PMCID: PMC10919072 DOI: 10.1021/acs.est.3c10490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/21/2024]
Abstract
Alzheimer's disease (AD) is a complex and multifactorial neurodegenerative disease, which is currently diagnosed via clinical symptoms and nonspecific biomarkers (such as Aβ1-42, t-Tau, and p-Tau) measured in cerebrospinal fluid (CSF), which alone do not provide sufficient insights into disease progression. In this pilot study, these biomarkers were complemented with small-molecule analysis using non-target high-resolution mass spectrometry coupled with liquid chromatography (LC) on the CSF of three groups: AD, mild cognitive impairment (MCI) due to AD, and a non-demented (ND) control group. An open-source cheminformatics pipeline based on MS-DIAL and patRoon was enhanced using CSF- and AD-specific suspect lists to assist in data interpretation. Chemical Similarity Enrichment Analysis revealed a significant increase of hydroxybutyrates in AD, including 3-hydroxybutanoic acid, which was found at higher levels in AD compared to MCI and ND. Furthermore, a highly sensitive target LC-MS method was used to quantify 35 bile acids (BAs) in the CSF, revealing several statistically significant differences including higher dehydrolithocholic acid levels and decreased conjugated BA levels in AD. This work provides several promising small-molecule hypotheses that could be used to help track the progression of AD in CSF samples.
Collapse
Affiliation(s)
- Begoña Talavera Andújar
- Luxembourg
Centre for Systems Biomedicine (LCSB), University
of Luxembourg, Avenue du Swing 6, L-4367 Belvaux, Luxembourg
| | - Arnaud Mary
- Luxembourg
Centre for Systems Biomedicine (LCSB), University
of Luxembourg, Avenue du Swing 6, L-4367 Belvaux, Luxembourg
| | - Carmen Venegas
- Luxembourg
Centre for Systems Biomedicine (LCSB), University
of Luxembourg, Avenue du Swing 6, L-4367 Belvaux, Luxembourg
| | - Tiejun Cheng
- National
Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, United States
| | - Leonid Zaslavsky
- National
Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, United States
| | - Evan E. Bolton
- National
Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, United States
| | - Michael T. Heneka
- Luxembourg
Centre for Systems Biomedicine (LCSB), University
of Luxembourg, Avenue du Swing 6, L-4367 Belvaux, Luxembourg
| | - Emma L. Schymanski
- Luxembourg
Centre for Systems Biomedicine (LCSB), University
of Luxembourg, Avenue du Swing 6, L-4367 Belvaux, Luxembourg
| |
Collapse
|
88
|
Morley-Fletcher S, Gaetano A, Gao V, Gatta E, Van Camp G, Bouwalerh H, Thomas P, Nicoletti F, Maccari S. Postpartum Oxytocin Treatment via the Mother Reprograms Long-Term Behavioral Disorders Induced by Early Life Stress on the Plasma and Brain Metabolome in the Rat. Int J Mol Sci 2024; 25:3014. [PMID: 38474260 DOI: 10.3390/ijms25053014] [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: 01/31/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The rat model of perinatal stress (PRS), in which exposure of pregnant dams to restraint stress reduces maternal behavior, is characterized by a metabolic profile that is reminiscent of the "metabolic syndrome". We aimed to identify plasma metabolomic signatures linked to long-term programming induced by PRS in aged male rats. This study was conducted in the plasma and frontal cortex. We also investigated the reversal effect of postpartum carbetocin (Cbt) on these signatures, along with its impact on deficits in cognitive, social, and exploratory behavior. We found that PRS induced long-lasting changes in biomarkers of secondary bile acid metabolism in the plasma and glutathione metabolism in the frontal cortex. Cbt treatment demonstrated disease-dependent effects by reversing the metabolite alterations. The metabolomic signatures of PRS were associated with long-term cognitive and emotional alterations alongside endocrinological disturbances. Our findings represent the first evidence of how early life stress may alter the metabolomic profile in aged individuals, thereby increasing vulnerability to CNS disorders. This raises the intriguing prospect that the pharmacological activation of oxytocin receptors soon after delivery through the mother may rectify these alterations.
Collapse
Affiliation(s)
- Sara Morley-Fletcher
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Alessandra Gaetano
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Vance Gao
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Eleonora Gatta
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Gilles Van Camp
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Hammou Bouwalerh
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
| | - Pierre Thomas
- INSERM (U-1172) Laboratoire Lille Neuroscience & Cognition, équipe Plasticity & Subjectivity, Plateforme CURE, Hôpital Fontan, CHU de Lille, Psychiatry Department, Université de Lille, F-59000 Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology "V. Erspamer", University Sapienza of Rome, 00185 Roma, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Stefania Maccari
- Unité de Glycobiologie Structurale et Fonctionnelle, GlycoStress Team, CNRS, UMR 8576, UGSF, Université de Lille, F-59000 Lille, France
- Department of Science and Medical-Surgical Biotechnology, University Sapienza of Rome, 00185 Roma, Italy
| |
Collapse
|
89
|
Zhang KX, Zhu Y, Song SX, Bu QY, You XY, Zou H, Zhao GP. Ginsenoside Rb1, Compound K and 20(S)-Protopanaxadiol Attenuate High-Fat Diet-Induced Hyperlipidemia in Rats via Modulation of Gut Microbiota and Bile Acid Metabolism. Molecules 2024; 29:1108. [PMID: 38474620 DOI: 10.3390/molecules29051108] [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: 02/03/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Hyperlipidemia, characterized by elevated serum lipid concentrations resulting from lipid metabolism dysfunction, represents a prevalent global health concern. Ginsenoside Rb1, compound K (CK), and 20(S)-protopanaxadiol (PPD), bioactive constituents derived from Panax ginseng, have shown promise in mitigating lipid metabolism disorders. However, the comparative efficacy and underlying mechanisms of these compounds in hyperlipidemia prevention remain inadequately explored. This study investigates the impact of ginsenoside Rb1, CK, and PPD supplementation on hyperlipidemia in rats induced by a high-fat diet. Our findings demonstrate that ginsenoside Rb1 significantly decreased body weight and body weight gain, ameliorated hepatic steatosis, and improved dyslipidemia in HFD-fed rats, outperforming CK and PPD. Moreover, ginsenoside Rb1, CK, and PPD distinctly modified gut microbiota composition and function. Ginsenoside Rb1 increased the relative abundance of Blautia and Eubacterium, while PPD elevated Akkermansia levels. Both CK and PPD increased Prevotella and Bacteroides, whereas Clostridium-sensu-stricto and Lactobacillus were reduced following treatment with all three compounds. Notably, only ginsenoside Rb1 enhanced lipid metabolism by modulating the PPARγ/ACC/FAS signaling pathway and promoting fatty acid β-oxidation. Additionally, all three ginsenosides markedly improved bile acid enterohepatic circulation via the FXR/CYP7A1 pathway, reducing hepatic and serum total bile acids and modulating bile acid pool composition by decreasing primary/unconjugated bile acids (CA, CDCA, and β-MCA) and increasing conjugated bile acids (TCDCA, GCDCA, GDCA, and TUDCA), correlated with gut microbiota changes. In conclusion, our results suggest that ginsenoside Rb1, CK, and PPD supplementation offer promising prebiotic interventions for managing HFD-induced hyperlipidemia in rats, with ginsenoside Rb1 demonstrating superior efficacy.
Collapse
Affiliation(s)
- Kang-Xi Zhang
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
| | - Yue Zhu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Shu-Xia Song
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Qing-Yun Bu
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300308, China
| | - Xiao-Yan You
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Hong Zou
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guo-Ping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
90
|
Li X, Lu W, Kharitonenkov A, Luo Y. Targeting the FGF19-FGFR4 pathway for cholestatic, metabolic, and cancerous diseases. J Intern Med 2024; 295:292-312. [PMID: 38212977 DOI: 10.1111/joim.13767] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Human fibroblast growth factor 19 (FGF19, or FGF15 in rodents) plays a central role in controlling bile acid (BA) synthesis through a negative feedback mechanism. This process involves a postprandial crosstalk between the BA-activated ileal farnesoid X receptor and the hepatic Klotho beta (KLB) coreceptor complexed with fibrobalst growth factor receptor 4 (FGFR4) kinase. Additionally, FGF19 regulates glucose, lipid, and energy metabolism by coordinating responses from functional KLB and FGFR1-3 receptor complexes on the periphery. Pharmacologically, native FGF19 or its analogs decrease elevated BA levels, fat content, and collateral tissue damage. This makes them effective in treating both cholestatic diseases such as primary biliary or sclerosing cholangitis (PBC or PSC) and metabolic abnormalities such as nonalcoholic steatohepatitis (NASH). However, chronic administration of FGF19 drives oncogenesis in mice by activating the FGFR4-dependent mitogenic or hepatic regenerative pathway, which could be a concern in humans. Agents that block FGF19 or FGFR4 signaling have shown great potency in preventing FGF19-responsive hepatocellular carcinoma (HCC) development in animal models. Recent phase 1/2 clinical trials have demonstrated promising results for several FGF19-based agents in selectively treating patients with PBC, PSC, NASH, or HCC. This review aims to provide an update on the clinical development of both analogs and antagonists targeting the FGF19-FGFR4 signaling pathway for patients with cholestatic, metabolic, and cancer diseases. We will also analyze potential safety and mechanistic concerns that should guide future research and advanced trials.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Pharmacological Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas, USA
| | | | - Yongde Luo
- School of Pharmacological Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
91
|
Gao SY, Zhao JC, Xia Q, Sun C, Aili M, Talifu A, Huo SX, Zhang Y, Li ZJ. Evaluation of the hepatotoxicity of Psoralea corylifolia L. based on a zebrafish model. Front Pharmacol 2024; 15:1308655. [PMID: 38449808 PMCID: PMC10914953 DOI: 10.3389/fphar.2024.1308655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Objective: Psoralea corylifolia L. (FP) has received increasing attention due to its potential hepatotoxicity. Methods: In this study, zebrafish were treated with different concentrations of an aqueous extract of FP (AEFP; 40, 50, or 60 μg/mL), and the hepatotoxic effects of tonicity were determined by the mortality rate, liver morphology, fluorescence area and intensity of the liver, biochemical indices, and pathological tissue staining. The mRNA expression of target genes in the bile acid metabolic signaling pathway and lipid metabolic pathway was detected by qPCR, and the mechanism of toxicity was initially investigated. AEFP (50 μg/mL) was administered in combination with FXR or a peroxisome proliferator-activated receptor α (PPARα) agonist/inhibitor to further define the target of toxicity. Results: Experiments on toxic effects showed that, compared with no treatment, AEFP administration resulted in liver atrophy, a smaller fluorescence area in the liver, and a lower fluorescence intensity (p < 0.05); alanine transaminase (ALT), aspartate transaminase (AST), and γ-GT levels were significantly elevated in zebrafish (p < 0.01), and TBA, TBIL, total cholesterol (TC), TG, low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) levels were elevated to different degrees (p < 0.05); and increased lipid droplets in the liver appeared as fatty deposits. Molecular biological validation revealed that AEFP inhibited the expression of the FXR gene, causing an increase in the expression of the downstream genes SHP, CYP7A1, CYP8B1, BSEP, MRP2, NTCP, peroxisome proliferator-activated receptor γ (PPARγ), ME-1, SCD-1, lipoprotein lipase (LPL), CPT-1, and CPT-2 and a decrease in the expression of PPARα (p < 0.05). Conclusion: This study demonstrated that tonic acid extracts are hepatotoxic to zebrafish through the inhibition of FXR and PPARα expression, thereby causing bile acid and lipid metabolism disorders.
Collapse
Affiliation(s)
- Shu-Yan Gao
- Uyghur Medical Hospital of Xinjiang Uyghur Autonomous Region, Ürümqi, China
- Xinjiang Key Laboratory of Evidence-Based and Translation, Hospital Preparation of Traditional Chinese Medicine, Ürümqi, China
| | - Jing-Cheng Zhao
- College of Pharmacy, Xinjiang Medical University, Ürümqi, China
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Chen Sun
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Maimaiti Aili
- Uyghur Medical Hospital of Xinjiang Uyghur Autonomous Region, Ürümqi, China
- Xinjiang Key Laboratory of Evidence-Based and Translation, Hospital Preparation of Traditional Chinese Medicine, Ürümqi, China
| | - Ainiwaer Talifu
- Uyghur Medical Hospital of Xinjiang Uyghur Autonomous Region, Ürümqi, China
- Xinjiang Key Laboratory of Evidence-Based and Translation, Hospital Preparation of Traditional Chinese Medicine, Ürümqi, China
| | - Shi-Xia Huo
- Uyghur Medical Hospital of Xinjiang Uyghur Autonomous Region, Ürümqi, China
- Xinjiang Key Laboratory of Evidence-Based and Translation, Hospital Preparation of Traditional Chinese Medicine, Ürümqi, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Zhi-Jian Li
- Uyghur Medical Hospital of Xinjiang Uyghur Autonomous Region, Ürümqi, China
- Xinjiang Key Laboratory of Evidence-Based and Translation, Hospital Preparation of Traditional Chinese Medicine, Ürümqi, China
| |
Collapse
|
92
|
Zhao M, Kuang W, Yang J, Liu Y, Yang M, Chen Y, Zhu H, Yang Y. Cholesterol lowering in diet-induced hypercholesterolemic mice using Lactobacillus bile salt hydrolases with different substrate specificities. Food Funct 2024; 15:1340-1354. [PMID: 38205623 DOI: 10.1039/d3fo04871c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
The cholesterol-lowering effect of lactic acid bacteria with high activity of bile salt hydrolase (BSH) is unclear. We believe that distinguishing BSH substrate specificity is necessary to study the effect of various BSH enzymes. We engineered a BSH mutant enzyme recombinant strain named F67A, which exclusively hydrolyzes taurocholic acid (TCA) using site-directed mutagenesis, and a previously lab-constructed BSH recombinant strain, YB81 that exclusively hydrolyzes glycocholic acid (GCA). We also constructed the recombinant strain named NB5462, which carries the empty pSIP411 plasmid and was used as a blank control strain. The intestinal flora in pseudo-germ-free (PGF) mice in which intestinal flora were eliminated via antibiotics, and F67A successfully reduced serum cholesterol levels in high-cholesterol diet-fed mice, whereas YB81 did not yield the same results. However, YB81 regained its cholesterol-lowering capacity in specific pathogen-free (SPF) mice with intact intestinal flora. The cholesterol-lowering mechanism of F67A involved modifying the bile acid pool through BSH enzyme activity. This adjustment regulated the expression of intestinal farnesoid X receptor and subsequently elevated hepatic cholesterol 7α-hydroxylase (CYP7A1), effectively reducing cholesterol levels. Conversely, GCA, the substrate of YB81, was found in minimal quantities in mice, preventing it from inducing changes in bile acid pools. In the presence of intestinal flora, the YB81 BSH enzyme induced notable alterations in bile acids by regulating changes in the intestinal flora and BSH within the flora, ultimately resulting in cholesterol reduction. This is the first study investigating the substrate specificity of BSH, demonstrating that different substrate-specific BSH enzymes exhibit cholesterol-lowering properties. Additionally, we elaborate on the mechanism of BSH-mediated enterohepatic axis regulation.
Collapse
Affiliation(s)
- Menghuan Zhao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 2 Xuelin Road, Nanjing 210046, China.
| | - Weijia Kuang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 2 Xuelin Road, Nanjing 210046, China.
| | - Jiaxin Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 2 Xuelin Road, Nanjing 210046, China.
- Nanjing Institute of Product Quality Inspection, Nanjing 210019, China
| | - Yanrong Liu
- Nanjing Institute of Product Quality Inspection, Nanjing 210019, China
| | - Miao Yang
- Nanjing Institute of Product Quality Inspection, Nanjing 210019, China
| | - Ying Chen
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Huanjing Zhu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 2 Xuelin Road, Nanjing 210046, China.
| | - Yao Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 2 Xuelin Road, Nanjing 210046, China.
| |
Collapse
|
93
|
Antony F, Brough Z, Zhao Z, Duong van Hoa F. Capture of the Mouse Organ Membrane Proteome Specificity in Peptidisc Libraries. J Proteome Res 2024; 23:857-867. [PMID: 38232390 DOI: 10.1021/acs.jproteome.3c00825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Membrane proteins, particularly those on the cell surface, play pivotal roles in diverse physiological processes, and their dysfunction is linked to a broad spectrum of diseases. Despite being crucial biomarkers and therapeutic drug targets, their low abundance and hydrophobic nature pose challenges in isolation and quantification, especially when extracted from tissues and organs. To overcome these hurdles, we developed the membrane-mimicking peptidisc, enabling the isolation of the membrane proteome in a water-soluble library conducive to swift identification through liquid chromatography with tandem mass spectrometry. This study applies the method across five mice organs, capturing between 200 and 450 plasma membrane proteins in each case. More than just membrane protein identification, the peptidisc is used to estimate the relative abundance across organs, linking cell-surface protein molecular functions to organ biological roles, thereby contributing to the ongoing discourse on organ specificity. This contribution holds substantial potential for unveiling new avenues in the exploration of biomarkers and downstream applications involving knowledge of the organ cell-surface proteome.
Collapse
Affiliation(s)
- Frank Antony
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Zora Brough
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Zhiyu Zhao
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Franck Duong van Hoa
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
94
|
Li ZY, Shen QM, Wang J, Tuo JY, Tan YT, Li HL, Xiang YB. Prediagnostic plasma metabolite concentrations and liver cancer risk: a population-based study of Chinese men. EBioMedicine 2024; 100:104990. [PMID: 38306896 PMCID: PMC10847612 DOI: 10.1016/j.ebiom.2024.104990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Previous metabolic profiling of liver cancer has mostly used untargeted metabolomic approaches and was unable to quantitate the absolute concentrations of metabolites. In this study, we examined the association between the concentrations of 186 targeted metabolites and liver cancer risk using prediagnostic plasma samples collected up to 14 years prior to the clinical diagnosis of liver cancer. METHODS We conducted a nested case-control study (n = 322 liver cancer cases, n = 322 matched controls) within the Shanghai Men's Health Study. Conditional logistic regression models adjusted for demographics, lifestyle factors, dietary habits, and related medical histories were used to estimate the odds ratios. Restricted cubic spline functions were used to characterise the dose-response relationships between metabolite concentrations and liver cancer risk. FINDINGS After adjusting for potential confounders and correcting for multiple testing, 28 metabolites were associated with liver cancer risk. Significant non-linear relationships were observed for 22 metabolites. The primary bile acid biosynthesis and phenylalanine, tyrosine and tryptophan biosynthesis were found to be important pathways involved in the aetiology of liver cancer. A metabolic score consisting of 10 metabolites significantly improved the predictive ability of traditional epidemiological risk factors for liver cancer, with an optimism-corrected AUC increased from 0.84 (95% CI: 0.81-0.87) to 0.89 (95% CI: 0.86-0.91). INTERPRETATION This study characterised the dose-response relationships between metabolites and liver cancer risk, providing insights into the complex metabolic perturbations prior to the clinical diagnosis of liver cancer. The metabolic score may serve as a candidate risk predictor for liver cancer. FUNDING National Key Project of Research and Development Program of China [2021YFC2500404, 2021YFC2500405]; US National Institutes of Health [subcontract of UM1 CA173640].
Collapse
Affiliation(s)
- Zhuo-Ying Li
- School of Public Health, Fudan University, Shanghai, 200032, China; State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Qiu-Ming Shen
- State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Jing Wang
- State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Jia-Yi Tuo
- State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China; School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Yu-Ting Tan
- State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Hong-Lan Li
- State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Yong-Bing Xiang
- School of Public Health, Fudan University, Shanghai, 200032, China; State Key Laboratory of System Medicine for Cancer & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China; School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
95
|
Lun W, Yan Q, Guo X, Zhou M, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. Mechanism of action of the bile acid receptor TGR5 in obesity. Acta Pharm Sin B 2024; 14:468-491. [PMID: 38322325 PMCID: PMC10840437 DOI: 10.1016/j.apsb.2023.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/17/2023] [Accepted: 10/24/2023] [Indexed: 02/08/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of membrane protein receptors, and Takeda G protein-coupled receptor 5 (TGR5) is a member of this family. As a membrane receptor, TGR5 is widely distributed in different parts of the human body and plays a vital role in regulating metabolism, including the processes of energy consumption, weight loss and blood glucose homeostasis. Recent studies have shown that TGR5 plays an important role in glucose and lipid metabolism disorders such as fatty liver, obesity and diabetes. With the global obesity situation becoming more and more serious, a comprehensive explanation of the mechanism of TGR5 and filling the gaps in knowledge concerning clinical ligand drugs are urgently needed. In this review, we mainly explain the anti-obesity mechanism of TGR5 to promote the further study of this target, and show the electron microscope structure of TGR5 and review recent studies on TGR5 ligands to illustrate the specific binding between TGR5 receptor binding sites and ligands, which can effectively provide new ideas for ligand research and promote drug research.
Collapse
Affiliation(s)
- Weijun Lun
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qihao Yan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinghua Guo
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Minchuan Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Science City, Guangzhou 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
96
|
Qin S, Tian J, Zhao Y, Wang L, Wang J, Liu S, Meng J, Wang F, Liu C, Han J, Pan C, Zhang Y, Yi Y, Li C, Liu M, Liang A. Gardenia extract protects against intrahepatic cholestasis by regulating bile acid enterohepatic circulation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117083. [PMID: 37634748 DOI: 10.1016/j.jep.2023.117083] [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: 03/29/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cholestasis is the main manifestation of cholestatic liver disease, which has a risk of progression to end-stage liver disease. Gardeniae Fructus is the dried fruit of Gardeniae jasminoides Ellis, a plant of the Rubiaceae family. Gardeniae Fructus has shown therapeutic potential in cholestasis-related liver diseases and it is generally believed that Gardeniae Fructus ameliorates cholestasis, which could be related to its influence on bile acids (BAs) metabolism. However, the specific targets of Gardeniae Fructus and its impact on enterohepatic circulation of BAs have not yet been fully elucidated. AIM OF THE STUDY To systematically elucidate the mechanism by which Gardenia extract (GE, total iridoids in Gardeniae Fructus, which contains the predominant and characteristic phytoconstituents of Gardeniae Fructus) ameliorates alpha-naphthylisothiocyanate (ANIT)-induced cholestatic liver injury. MATERIALS AND METHODS Sprague-Dawley rats were orally administered water, obeticholic acid (OCA, 2 mg/kg), or GE (21 and 42 mg/kg) once daily for five days. On the third day, the model was established by administration of a single dose of ANIT (40 mg/kg) by oral gavage. Biochemical and pathological analyses, BA metabolomics, transcriptomics, and qRT-PCR were performed. RESULTS The profile of BAs in serum and liver confirmed that GE attenuated ANIT-induced acute cholestasis by affecting BA metabolism in a dose-dependent manner. Liver transcriptomic analysis indicated that GE mainly influenced the primary bile acid (PBA) biosynthesis and bile secretion pathways. GE mainly affected PBA biosynthesis in liver by upregulating Cyp8b1 gene expression, thereby significantly reducing the level of total bile acids (TBA). GE mainly promoted PBA excretion from liver into duodenum by upregulating Fxr and Oatp1 gene expression, thereby increasing the excretion of PBA in feces, and inhibiting PBA in liver entering the blood by alternative routes to reduce TBA levels in serum and urine and improve the enterohepatic circulation of BAs. CONCLUSION GE attenuated ANIT-induced hepatotoxicity and cholestasis in rats by upregulating Cyp8b1 expression to inhibit BA synthesis in the liver, while also promoting BA excretion via the intestinal-fecal route, and improving enterohepatic circulation of BAs.
Collapse
Affiliation(s)
- Shasha Qin
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Jingzhuo Tian
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Yong Zhao
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Lianmei Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Jinyu Wang
- Research Center for Traditional Chinese Medicine Preparations, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Suyan Liu
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Jing Meng
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Fang Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Chenyue Liu
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Jiayin Han
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Chen Pan
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Yushi Zhang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Yan Yi
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Chunying Li
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Meiting Liu
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| | - Aihua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| |
Collapse
|
97
|
Li Y, Peng X, Wang G, Zan B, Wang Y, Zou J, Tian T, Meng Q, Shi R, Wang T, Wu J, Ma Y. Identifying hepatoprotective mechanism and effective components of Yinchenzhufu decoction in chronic cholestatic liver injury using a comprehensive strategy based on metabolomics, molecular biology, pharmacokinetics, and cytology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117060. [PMID: 37598769 DOI: 10.1016/j.jep.2023.117060] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Traditional Chinese Medicine (TCM), cholestasis liver disease belongs to jaundice. Yinchenzhufu decoction (YCZFD) is a classic formula used for treating jaundice. AIM OF THE STUDY This study was aimed to investigate the potential mechanism and effective components of YCZFD in chronic cholestatic liver injury (CCLI). MATERIALS AND METHODS A chronic cholestatic mouse model induced by 3, 5-diethoxycarbonyl-1, 4-dihydroxychollidine was used to investigate the effect of YCZFD. Then, metabolomics was used to investigate the metabolites influenced by YCZFD. Serum and liver bile acid (BA) levels were measured using liquid chromatography coupled with triple quadruple mass spectrometry (LC-MS/MS), and the gene and protein expressions of BA transporters and metabolic enzymes were detected. Additionally, the pharmacokinetics of multiple components of YCZFD was explored to clarify the potential effective components. The effects of absorbed components of YCZFD on BA metabolism and transporter function, inflammation, and farnesoid X receptor (FXR) and pregnane X receptor (PXR) activation were analyzed using sandwich cultured rat hepatocytes, AML12 cells, and dual-luciferase receptor systems, respectively. RESULTS YCZFD decreased the liver damage in chronic cholestatic mice. Serum metabolomics results indicated that the main pathways influenced by YCZFD involved primary BA biosynthesis and arachidonic acid metabolism. YCZFD upregulated the expression of FXR, PXR, and BA efflux transporters and the metabolic enzymes of liver tissues, promoting BA excretion and metabolism in cholestatic mice. Additionally, YCZFD downregulated the expression of genes and proteins of the toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway and decreased liver inflammation. The pharmacokinetic study indicated that multiple components showed different pharmacokinetic properties. Among the absorbed components of YCZFD, multiple components activated the transcription of FXR and PXR, regulated BA transporters and metabolic enzyme function, and reduced the gene expression of TLR4 and NF-κB1. CONCLUSION YCZFD can ameliorate CCLI by promoting the excretion and metabolism of BAs and inhibiting inflammation via the TLR4/NF-κB signaling pathway. The multiple components of YCZFD could act on BA homeostasis regulation and anti-inflammation, exhibiting a combined effect against CCLI.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Xiaotian Peng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Guofeng Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Bin Zan
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Yahang Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Juan Zou
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Tian Tian
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Qian Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China; Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
98
|
Manudhane AP, Leupold MD, Shah HW, Shah R, Han SY, Lee PJ, Burlen JJ, Papachristou GI, Krishna SG. A Review on Endoscopic Management of Acute Cholecystitis: Endoscopic Ultrasound-Guided Gallbladder Drainage and Endoscopic Transpapillary Gallbladder Drainage. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:212. [PMID: 38399500 PMCID: PMC10890498 DOI: 10.3390/medicina60020212] [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: 01/04/2024] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024]
Abstract
A percutaneous cholecystostomy tube (PCT) is the conventionally favored nonoperative intervention for treating acute cholecystitis. However, PCT is beset by high adverse event rates, need for scheduled reintervention, and inadvertent dislodgement, as well as patient dissatisfaction with a percutaneous drain. Recent advances in endoscopic therapy involve the implementation of endoscopic transpapillary drainage (ETP-GBD) and endoscopic ultrasound-guided gallbladder drainage (EUS-GBD), which are increasingly preferred over PCT due to their favorable technical and clinical success combined with lower complication rates. In this article, we provide a comprehensive review of the literature on EUS-GBD and ETP-GBD, delineating instances when clinicians should opt for endoscopic management and highlighting potential risks associated with each approach.
Collapse
Affiliation(s)
- Albert P. Manudhane
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Matthew D. Leupold
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hamza W. Shah
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Raj Shah
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Samuel Y. Han
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Peter J. Lee
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Jordan J. Burlen
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Georgios I. Papachristou
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, 395 W. 12th Avenue, Suite 262, Columbus, OH 43210, USA; (A.P.M.)
| |
Collapse
|
99
|
Qu Y, Gong X, Zhao Z, Zhang Z, Zhang Q, Huang Y, Xie Q, Liu Y, Wei J, Du H. Establishment and Validation of Novel Prognostic Subtypes in Hepatocellular Carcinoma Based on Bile Acid Metabolism Gene Signatures Using Bulk and Single-Cell RNA-Seq Data. Int J Mol Sci 2024; 25:919. [PMID: 38255993 PMCID: PMC10815120 DOI: 10.3390/ijms25020919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly detrimental cancer type and has limited therapeutic options, posing significant threats to human health. The development of HCC has been associated with a disorder in bile acid (BA) metabolism. In this study, we employed an integrative approach, combining various datasets and omics analyses, to comprehensively characterize the tumor microenvironment in HCC based on genes related to BA metabolism. Our analysis resulted in the classification of HCC samples into four subtypes (C1, C2a, C2b, and C3). Notably, subtype C2a, characterized by the highest bile acid metabolism score (BAMS), exhibited the highest survival probability. This subtype also demonstrated increased immune cell infiltration, lower cell cycle scores, reduced AFP levels, and a lower risk of metastasis compared to subtypes C1 and C3. Subtype C1 displayed poorer survival probability and elevated cell cycle scores. Importantly, the identified subtypes based on BAMS showed potential relevance to the gene expression of drug targets in currently approved drugs and those under clinical research. Genes encoding VEGFR (FLT4 and KDR) and MET were elevated in C2, while genes such as TGFBR1, TGFB1, ADORA3, SRC, BRAF, RET, FLT3, KIT, PDGFRA, and PDGFRB were elevated in C1. Additionally, FGFR2 and FGFR3, along with immune target genes including PDCD1 and CTLA4, were higher in C3. This suggests that subtypes C1, C2, and C3 might represent distinct potential candidates for TGFB1 inhibitors, VEGFR inhibitors, and immune checkpoint blockade treatments, respectively. Significantly, both bulk and single-cell transcriptome analyses unveiled a negative correlation between BA metabolism and cell cycle-related pathways. In vitro experiments further confirmed that the treatment of HCC cell lines with BA receptor agonist ursodeoxycholic acid led to the downregulation of the expression of cell cycle-related genes. Our findings suggest a plausible involvement of BA metabolism in liver carcinogenesis, potentially mediated through the regulation of tumor cell cycles and the immune microenvironment. This preliminary understanding lays the groundwork for future investigations to validate and elucidate the specific mechanisms underlying this potential association. Furthermore, this study provides a novel foundation for future precise molecular typing and the design of systemic clinical trials for HCC therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (Y.Q.); (X.G.); (Z.Z.); (Z.Z.); (Q.Z.); (Y.H.); (Q.X.); (Y.L.)
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (Y.Q.); (X.G.); (Z.Z.); (Z.Z.); (Q.Z.); (Y.H.); (Q.X.); (Y.L.)
| |
Collapse
|
100
|
Jin C, Zhou T, Duan Z, Deng Y, Zhang X, Xiao C, He J, He G, Zhou Y, Li S. Effect of chin brick tea [Camellia sinensis (L.) Kuntze] on lipid metabolism and inflammation by modulating intestinal flora and bile acids in mice with non-alcoholic fatty liver disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116950. [PMID: 37506781 DOI: 10.1016/j.jep.2023.116950] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tea (Camellia sinensis) has been consumed for centuries as a traditional remedy for various metabolic diseases. The pharmacological mechanisms of many conventional medicines, including tea, often need to be clarified. Chin brick tea is a unique Chinese black tea grown in Hubei, China, rich in tea elements such as tea polyphenols and tea polysaccharides. AIM OF THE STUDY We focus on the effects of commercial chin brick tea on non-alcoholic fatty liver disease by altering intestinal flora and its metabolite, bile acids. MATERIALS AND METHODS Targeted UPLC-MS/MS was employed to quantify the tea elements in commercial chin brick tea. In this study, we performed an integrated approach of animal experiments, 16 S rDNA, and ultra-performance liquid chromatography-tandem mass spectrometry to explore the potential mechanism of action of chin brick tea in preventing non-alcoholic fatty liver disease (NAFLD). RESULTS After 14 weeks of administration, CBT extract could signiffcantly decrease the levels of body weight, liver weight, LDL-C, TC, ALT, IL-1β and IL-18, and slight increase HDL-C levels in NAFLD mice. The results indicated that the interventional impact of CBT with high-fat diet-induced NAFLD might depend on intestinal flora and its metabolites bile acids. Moreover, sequencing of 16 S rRNA genes demonstrated that CBT could signiffcantly improve the intestinal flora disorder of NAFLD mice. Speciffcally, CBT increased the levels of Lactobacillus, Alloprevotella, and Ruminococcaceae, while reducing the levels of Bacteroides in NAFLD mice. Then, a total of 23 bile acids were identified, 17 differential bile acids were obtained by screening, and CBT increase the primary bile acids/secondary bile acids ratio in NAFLD mice. Additionally, correlation analysis revealed that Bacteroides was negatively correlated with DCA and ωMCA, Lactobacillus was positively correlated with DCA and ωMCA, Bacteroides was positively correlated with NAFLD, Lactobacillus was negatively associated with NAFLD, and DCA and ωMCA were negatively correlated with NAFLD. CONCLUSION CBT extract has a good interventional impact on NAFLD mice. The mechanism by which this extract exerts its action is, at least partly, related to its regulation of intestinal flora and its metabolites bile acids.
Collapse
Affiliation(s)
- Can Jin
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Tingting Zhou
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Zhihao Duan
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China; Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Ying Deng
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Xiaoli Zhang
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Changyi Xiao
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China; Xinding Biological Technology Co., Ltd, Yichang, 443000, China; Hubei Province Changshengchuan chin brick tea Research Institute, Yichang, 443000, China
| | - Jiangang He
- Xinding Biological Technology Co., Ltd, Yichang, 443000, China; Hubei Province Changshengchuan chin brick tea Research Institute, Yichang, 443000, China
| | - Gongwei He
- Xinding Biological Technology Co., Ltd, Yichang, 443000, China; Hubei Province Changshengchuan chin brick tea Research Institute, Yichang, 443000, China
| | - You Zhou
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China.
| | - Shigang Li
- China Three Gorges University, Griffith Health, Yichang, 443000, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China; Xinding Biological Technology Co., Ltd, Yichang, 443000, China; Hubei Province Changshengchuan chin brick tea Research Institute, Yichang, 443000, China.
| |
Collapse
|