1
|
Wen YQ, Zou ZY, Zhao GG, Zhang MJ, Zhang YX, Wang GH, Shi JJ, Wang YY, Song YY, Wang HX, Chen RY, Zheng DX, Duan XQ, Liu YM, Gonzalez FJ, Fan JG, Xie C. FXR activation remodels hepatic and intestinal transcriptional landscapes in metabolic dysfunction-associated steatohepatitis. Acta Pharmacol Sin 2024; 45:2313-2327. [PMID: 38992119 PMCID: PMC11489735 DOI: 10.1038/s41401-024-01329-1] [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: 01/15/2024] [Accepted: 05/28/2024] [Indexed: 07/13/2024] Open
Abstract
The escalating obesity epidemic and aging population have propelled metabolic dysfunction-associated steatohepatitis (MASH) to the forefront of public health concerns. The activation of FXR shows promise to combat MASH and its detrimental consequences. However, the specific alterations within the MASH-related transcriptional network remain elusive, hindering the development of more precise and effective therapeutic strategies. Through a comprehensive analysis of liver RNA-seq data from human and mouse MASH samples, we identified central perturbations within the MASH-associated transcriptional network, including disrupted cellular metabolism and mitochondrial function, decreased tissue repair capability, and increased inflammation and fibrosis. By employing integrated transcriptome profiling of diverse FXR agonists-treated mice, FXR liver-specific knockout mice, and open-source human datasets, we determined that hepatic FXR activation effectively ameliorated MASH by reversing the dysregulated metabolic and inflammatory networks implicated in MASH pathogenesis. This mitigation encompassed resolving fibrosis and reducing immune infiltration. By understanding the core regulatory network of FXR, which is directly correlated with disease severity and treatment response, we identified approximately one-third of the patients who could potentially benefit from FXR agonist therapy. A similar analysis involving intestinal RNA-seq data from FXR agonists-treated mice and FXR intestine-specific knockout mice revealed that intestinal FXR activation attenuates intestinal inflammation, and has promise in attenuating hepatic inflammation and fibrosis. Collectively, our study uncovers the intricate pathophysiological features of MASH at a transcriptional level and highlights the complex interplay between FXR activation and both MASH progression and regression. These findings contribute to precise drug development, utilization, and efficacy evaluation, ultimately aiming to improve patient outcomes.
Collapse
Affiliation(s)
- Ying-Quan Wen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zi-Yuan Zou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, 200092, China
| | - Guan-Guan Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Meng-Jiao Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yong-Xin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Gai-Hong Wang
- Cascade Pharmaceuticals, Inc, Shanghai, 201321, China
| | - Jing-Jing Shi
- Cascade Pharmaceuticals, Inc, Shanghai, 201321, China
| | - Yuan-Yang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Laboratory Medicine and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Ye-Yu Song
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, 200092, China
| | - Hui-Xia Wang
- Cascade Pharmaceuticals, Inc, Shanghai, 201321, China
| | - Ru-Ye Chen
- Cascade Pharmaceuticals, Inc, Shanghai, 201321, China
| | | | - Xiao-Qun Duan
- Industrial Technology Research Institute of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Ya-Meng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jian-Gao Fan
- Department of Gastroenterology, Center for Fatty Liver, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, 200092, China.
| | - Cen Xie
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Zhao B, Zhang J, Zhao K, Wang B, Liu J, Wang C, Zeng L, Zeng X, Luo Y. Effect of rapamycin on hepatic metabolomics of non-alcoholic fatty liver rats based on non-targeted platform. J Pharm Biomed Anal 2024; 253:116541. [PMID: 39489928 DOI: 10.1016/j.jpba.2024.116541] [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: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Rapamycin (Rapa) is an inhibitor of mTOR complex, and its therapeutic effect on liver function was examined in non-alcoholic fatty liver disease (NAFLD) rats here. And the possible mechanism of Rapa in NAFLD was preliminarily elucidated based on the non-targeted metabolomics analysis. Adult male SD rats were fed with a high-fat and high-cholesterol diet (HFD) to establish NAFLD model. For Rapa group, 0.8 mg/(kg.d) Rapa was given to the HFD rats. Ultra-performance liquid chromatography and Q-Tof-mass spectrometry (UPLC and Q-TOF/MS) analysis were applied for the identification of metabolites in the serum of rats, which were annotated using Kyoto Encyclopedia of Genes and Genomes (KEGG). NAFLD rats presented with disturbed liver function, lipid metabolism and oxidative stress, but Rapa exerted a mitigating influence on the disorders. The metabolite profile data identified 579 metabolites that varied remarkably between the Rapa and HFD groups, with the main classes of amino acids and peptides, benzene, lipids and fatty acids. The differential metabolites were mainly involved in biosynthesis of cofactors, bile secretion, and glycerophospholipid metabolism were mainly enriched. In conclusion, Rapa has a potential protective effect against HFD-induced NAFLD, its hepatoprotective effect may achieved through mediating bile secretion and glycerophospholipid metabolism.
Collapse
Affiliation(s)
- Baiyun Zhao
- Drug Clinical Trial Institution, Hangzhou Normal University Affiliated Hospital, Hangzhou, China; Department of Pharmacy, Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Kaiyue Zhao
- Medical Department, Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Bin Wang
- Department of Critical Care Medicine, Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Jing Liu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
| | - Chaoxuan Wang
- Drug Clinical Trial Institution, Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Ling Zeng
- Drug Clinical Trial Institution, Hangzhou Normal University Affiliated Hospital, Hangzhou, China
| | - Xin Zeng
- Traditional Chinese Pharmacy, China Pharmaceutical University School, Nanjing, China
| | - Yan Luo
- Department of Translational Medicine Center, Hangzhou Normal University Affiliated Hospital, Hangzhou, China.
| |
Collapse
|
3
|
Antelo-Cea DA, Martínez-Rojas L, Cabrerizo-Ibáñez I, Roudi Rashtabady A, Hernández-Alvarez MI. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:11237. [PMID: 39457018 PMCID: PMC11508381 DOI: 10.3390/ijms252011237] [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: 10/03/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity and type 2 diabetes (T2D) are widespread metabolic disorders that significantly impact global health today, affecting approximately 17% of adults worldwide with obesity and 9.3% with T2D. Both conditions are closely linked to disruptions in lipid metabolism, where peroxisomes play a pivotal role. Mitochondria and peroxisomes are vital organelles responsible for lipid and energy regulation, including the β-oxidation and oxidation of very long-chain fatty acids (VLCFAs), cholesterol biosynthesis, and bile acid metabolism. These processes are significantly influenced by estrogens, highlighting the interplay between these organelles' function and hormonal regulation in the development and progression of metabolic diseases, such as obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and T2D. Estrogens modulate lipid metabolism through interactions with nuclear receptors, like peroxisome proliferator-activated receptors (PPARs), which are crucial for maintaining metabolic balance. Estrogen deficiency, such as in postmenopausal women, impairs PPAR regulation, leading to lipid accumulation and increased risk of metabolic disorders. The disruption of peroxisomal-mitochondrial function and estrogen regulation exacerbates lipid imbalances, contributing to insulin resistance and ROS accumulation. This review emphasizes the critical role of these organelles and estrogens in lipid metabolism and their implications for metabolic health, suggesting that therapeutic strategies, including hormone replacement therapy, may offer potential benefits in treating and preventing metabolic diseases.
Collapse
Affiliation(s)
- Damián A. Antelo-Cea
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Laura Martínez-Rojas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Izan Cabrerizo-Ibáñez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Ayda Roudi Rashtabady
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Harris LD, Aponte RAL, Jiao W, Cameron SA, Weymouth-Wilson A, Furneaux RH, Compton BJ, Luxenburger A. An efficient regioconvergent synthesis of 3-aza-obeticholic acid. Steroids 2024; 212:109517. [PMID: 39322098 DOI: 10.1016/j.steroids.2024.109517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Bile acids (BAs) are steroidal molecules that play important roles in nutrient absorption, distribution, and excretion. They also act on specific receptors implicated in various metabolic and inflammatory diseases demonstrating their importance as potential drug candidates. Accordingly, there has been a concerted effort to develop new BA derivatives to probe structure-activity relationships with the goal of discovering BA analogues with enhanced pharmacological properties. Among the many steroidal derivatisations reported, the formation of endocyclic azasteroids appeals due to their potential to deliver altered biological responses with minimal change to the steroidal superstructure. Here, we report the synthesis of 3-aza-obeticholic acid (6) via a regioconvergent route. Ammoniolysis of lactones, formed from an m-CPBA-mediated Baeyer-Villiger reaction on a 3-keto-OCA derivative, furnished protected intermediate amido-alcohols which were separately elaborated to amino-alcohols via Hofmann degradation with BAIB. Upon individual N-Boc-protection, these underwent annulation to the 3-aza-A-ring when subjected to either mesylation or a Dess-Martin oxidation/hydrogenation sequence. Global deprotection of the 3-aza-intermediate delivered 3-aza-OCA in ten steps and an overall yield of up to 19%.
Collapse
Affiliation(s)
- Lawrence D Harris
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | - Roselis A Landaeta Aponte
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | - Wanting Jiao
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | - Scott A Cameron
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | | | - Richard H Furneaux
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand
| | - Andreas Luxenburger
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5040, New Zealand.
| |
Collapse
|
5
|
Yuan H, Jung ES, Chae SW, Jung SJ, Daily JW, Park S. Biomarkers for Health Functional Foods in Metabolic Dysfunction-Associated Steatotic Liver Disorder (MASLD) Prevention: An Integrative Analysis of Network Pharmacology, Gut Microbiota, and Multi-Omics. Nutrients 2024; 16:3061. [PMID: 39339660 PMCID: PMC11434757 DOI: 10.3390/nu16183061] [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: 08/07/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disorder (MASLD) is increasingly prevalent globally, highlighting the need for preventive strategies and early interventions. This comprehensive review explores the potential of health functional foods (HFFs) to maintain healthy liver function and prevent MASLD through an integrative analysis of network pharmacology, gut microbiota, and multi-omics approaches. We first examined the biomarkers associated with MASLD, emphasizing the complex interplay of genetic, environmental, and lifestyle factors. We then applied network pharmacology to identify food components with potential beneficial effects on liver health and metabolic function, elucidating their action mechanisms. This review identifies and evaluates strategies for halting or reversing the development of steatotic liver disease in the early stages, as well as biomarkers that can evaluate the success or failure of such strategies. The crucial role of the gut microbiota and its metabolites for MASLD prevention and metabolic homeostasis is discussed. We also cover state-of-the-art omics approaches, including transcriptomics, metabolomics, and integrated multi-omics analyses, in research on preventing MASLD. These advanced technologies provide deeper insights into physiological mechanisms and potential biomarkers for HFF development. The review concludes by proposing an integrated approach for developing HFFs targeting MASLD prevention, considering the Korean regulatory framework. We outline future research directions that bridge the gap between basic science and practical applications in health functional food development. This narrative review provides a foundation for researchers and food industry professionals interested in developing HFFs to support liver health. Emphasis is placed on maintaining metabolic balance and focusing on prevention and early-stage intervention strategies.
Collapse
Affiliation(s)
- Heng Yuan
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
| | - Eun-Soo Jung
- Clinical Trial Center for Functional Foods, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-W.C.); (S.-J.J.)
- Clinical Trial Center for K-FOOD Microbiome, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54907, Republic of Korea
| | - Soo-Wan Chae
- Clinical Trial Center for Functional Foods, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-W.C.); (S.-J.J.)
- Clinical Trial Center for K-FOOD Microbiome, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54907, Republic of Korea
| | - Su-Jin Jung
- Clinical Trial Center for Functional Foods, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-W.C.); (S.-J.J.)
- Clinical Trial Center for K-FOOD Microbiome, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54907, Republic of Korea
| | - James W. Daily
- Department of R&D, Daily Manufacturing Inc., Rockwell, NC 28138, USA;
| | - Sunmin Park
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 20 Hoseoro79bungil, Asan 31499, Republic of Korea
| |
Collapse
|
6
|
Wang L, Xu J, You N, Shao L, Zhuang Z, Zhuo L, Liu J, Shi J. Characteristics of intestinal flora in nonobese nonalcoholic fatty liver disease patients and the impact of ursodeoxycholic acid treatment on these features. Lipids 2024. [PMID: 39246185 DOI: 10.1002/lipd.12410] [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: 04/17/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024]
Abstract
The study aimed to investigate the alterations in gut microbiota among nonobese individuals with nonalcoholic fatty liver disease (NAFLD) and their response to treatment with ursodeoxycholic acid (UDCA). A total of 90 patients diagnosed with NAFLD and 36 healthy subjects were recruited to participate in this study. Among them, a subgroup of 14 nonobese nonalcoholic steatohepatitis (NASH) were treated with UDCA. Demographic and serologic data were collected for all participants, while stool samples were obtained for fecal microbiome analysis using 16S sequencing. In nonobese NAFLD patients, the alpha diversity of intestinal flora decreased (Shannon index, p < 0.05), and the composition of intestinal flora changed (beta diversity, p < 0.05). The abundance of 20 genera, including Fusobacterium, Lachnoclostridium, Klebsiella, etc., exhibited significant changes (p < 0.05). Among them, nine species including Fusobacterium, Lachnoclostridium, Klebsiella, etc. were found to be associated with abnormal liver enzymes and glucolipid metabolic disorders. Among the 14 NASH patients treated with UDCA, improvements were observed in terms of liver enzymes, CAP values, and E values (p < 0.05), however, no improve the glucolipid metabolism. While the alpha diversity of intestinal flora did not show significant changes after UDCA treatment, there was a notable alteration in the composition of intestinal flora (beta diversity, p < 0.05). Furthermore, UCDA treatment led to an improvement in the relative abundance of Alistipes, Holdemanella, Gilisia, etc. among nonobese NASH patients (p < 0.05). Nonobese NAFLD patients exhibit dysbiosis of the intestinal microbiota. UDCA can ameliorate hepatic enzyme abnormalities and reduce liver fat content in nonobese NASH patients, potentially through its ability to restore intestinal microbiota balance.
Collapse
Affiliation(s)
- Liyan Wang
- Department of Infectious diseases, The Second Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Jiali Xu
- Department of Endocrinology, The Second People's Hospital of Quzhou, Quzhou, Zhejiang, China
| | - Ningning You
- Department of Gastroenterology, Taizhou Enze Medical Center, Taizhou, Zhejiang, China
| | - Li Shao
- Institute of Translational Medicine, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Zhenjie Zhuang
- Institute of Translational Medicine, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Lili Zhuo
- Department of Endocrinology, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jing Liu
- Department of Hepatology, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Junping Shi
- Institute of Hepatology and Metabolic Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
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] [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
|
8
|
Léger T, Alilat S, Ferron PJ, Dec L, Bouceba T, Lanceleur R, Huet S, Devriendt-Renault Y, Parinet J, Clément B, Fessard V, Le Hégarat L. Chlordecone-induced hepatotoxicity and fibrosis are mediated by the proteasomal degradation of septins. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135177. [PMID: 39018595 DOI: 10.1016/j.jhazmat.2024.135177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Chlordecone (CLD) is a pesticide persisting in soils and contaminating food webs. CLD is sequestered in the liver and poorly metabolized into chlordecol (CLDOH). In vitro liver cell models were used to investigate the fate and mechanistic effects of CLD and CLDOH using multiomics. A 3D-cell model was used to investigate whether CLD and CLDOH can affect susceptibility to the metabolic dysfunction-associated steatotic liver disease (MASLD). Hepatocytes were more sensitive to CLD than CLDOH. CLDOH was intensively metabolized into a glucuronide conjugate, whereas CLD was sequestered. CLD but not CLDOH induced a depletion of Septin-2,- 7,- 9,- 10,- 11 due to proteasomal degradation. Septin binding with CLD and CLDOH was confirmed by surface plasmon resonance. CLD disrupted lipid droplet size and increased saturated long-chain dicarboxylic acid production by inhibiting stearoyl-CoA desaturase (SCD) abundance. Neither CLD nor CLDOH induced steatosis, but CLD induced fibrosis in the 3D model of MASLD. To conclude, CLD hepatoxicity is specifically driven by the degradation of septins. CLDOH, was too rapidly metabolized to induce septin degradation. We show that the conversion of CLD to CLDOH reduced hepatotoxicity and fibrosis in liver organoids. This suggests that protective strategies could be explored to reduce the hepatotoxicity of CLD.
Collapse
Affiliation(s)
- Thibaut Léger
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France.
| | - Sarah Alilat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Pierre-Jean Ferron
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Léonie Dec
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Tahar Bouceba
- Sorbonne University, CNRS, Institut de Biologie Paris-Seine (IBPS), Protein Engineering Platform, Molecular Interaction Service, Paris, France
| | - Rachelle Lanceleur
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Sylvie Huet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Yoann Devriendt-Renault
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Julien Parinet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Bruno Clément
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Valérie Fessard
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Ludovic Le Hégarat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| |
Collapse
|
9
|
Ziółkiewicz A, Niziński P, Soja J, Oniszczuk T, Combrzyński M, Kondracka A, Oniszczuk A. Potential of Chlorogenic Acid in the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): Animal Studies and Clinical Trials-A Narrative Review. Metabolites 2024; 14:346. [PMID: 38921480 PMCID: PMC11205996 DOI: 10.3390/metabo14060346] [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: 05/27/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Chlorogenic acid (CGA) is a natural polyphenol found in coffee, tea, vegetables, and fruits. It exhibits strong antioxidant activity and possesses several other biological properties, including anti-inflammatory effects, antimicrobial activity, and insulin-sensitizing properties. Moreover, it may improve lipid and glucose metabolism. This review summarizes the available information on the therapeutic effect of CGA in metabolic dysfunction-associated steatotic liver disease (MASLD). As the literature search engine, the browsers in the PubMed, Scopus, Web of Science databases, and ClinicalTrials.gov register were used. Animal trials and clinical studies suggest that CGA has promising therapeutic potential in treating MASLD and hepatic steatosis. Its mechanisms of action include antioxidant, anti-inflammatory, and anti-apoptotic effects via the activation of the Nrf2 signaling pathway and the inhibition of the TLR4/NF-κB signaling cascade. Furthermore, the alleviation of liver disease by CGA also involves other important molecules such as AMPK and important physiological processes such as the intestinal barrier and gut microbiota. Nevertheless, the specific target cell and key molecule to which CGA is directed remain unidentified and require further study.
Collapse
Affiliation(s)
- Agnieszka Ziółkiewicz
- Department of Inorganic Chemistry, Medical University of Lublin, Dr Witolda Chodźki 4a, 20-093 Lublin, Poland; (A.Z.); (A.O.)
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland
| | - Jakub Soja
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland; (J.S.); (T.O.); (M.C.)
| | - Tomasz Oniszczuk
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland; (J.S.); (T.O.); (M.C.)
| | - Maciej Combrzyński
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland; (J.S.); (T.O.); (M.C.)
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Dr Witolda Chodźki 4a, 20-093 Lublin, Poland; (A.Z.); (A.O.)
| |
Collapse
|
10
|
Hoang SH, Tveter KM, Mezhibovsky E, Roopchand DE. Proanthocyanidin B2 derived metabolites may be ligands for bile acid receptors S1PR2, PXR and CAR: an in silico approach. J Biomol Struct Dyn 2024; 42:4249-4262. [PMID: 37340688 PMCID: PMC10730774 DOI: 10.1080/07391102.2023.2224886] [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: 03/14/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Bile acids (BAs) act as signaling molecules via their interactions with various nuclear (FXR, VDR, PXR and CAR) and G-protein coupled (TGR5, M3R, S1PR2) BA receptors. Stimulation of these BA receptors influences several processes, including inflammatory responses and glucose and xenobiotic metabolism. BA profiles and BA receptor activity are deregulated in cardiometabolic diseases; however, dietary polyphenols were shown to alter BA profile and signaling in association with improved metabolic phenotypes. We previously reported that supplementing mice with a proanthocyanidin (PAC)-rich grape polyphenol (GP) extract attenuated symptoms of glucose intolerance in association with changes to BA profiles, BA receptor gene expression, and/or downstream markers of BA receptor activity. Exact mechanisms by which polyphenols modulate BA signaling are not known, but some hypotheses include modulation of the BA profile via changes to gut bacteria, or alteration of ligand-availability via BA sequestration. Herein, we used an in silico approach to investigate putative binding affinities of proanthocyanidin B2 (PACB2) and PACB2 metabolites to nuclear and G-protein coupled BA receptors. Molecular docking and dynamics simulations revealed that certain PACB2 metabolites had stable binding affinities to S1PR2, PXR and CAR, comparable to that of known natural and synthetic BA ligands. These findings suggest PACB2 metabolites may be novel ligands of S1PR2, CAR, and PXR receptors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Skyler H. Hoang
- Department of Food Science, New Jersey Institute for Food, Nutrition, and Health (Rutgers Center for Lipid Research and Center for Nutrition, Microbiome, and Health), Rutgers University, 61 Dudley Road, New Brunswick, New Jersey, 08901 USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Kevin M. Tveter
- Department of Food Science, New Jersey Institute for Food, Nutrition, and Health (Rutgers Center for Lipid Research and Center for Nutrition, Microbiome, and Health), Rutgers University, 61 Dudley Road, New Brunswick, New Jersey, 08901 USA
| | - Esther Mezhibovsky
- Department of Food Science, New Jersey Institute for Food, Nutrition, and Health (Rutgers Center for Lipid Research and Center for Nutrition, Microbiome, and Health), Rutgers University, 61 Dudley Road, New Brunswick, New Jersey, 08901 USA
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Diana E. Roopchand
- Department of Food Science, New Jersey Institute for Food, Nutrition, and Health (Rutgers Center for Lipid Research and Center for Nutrition, Microbiome, and Health), Rutgers University, 61 Dudley Road, New Brunswick, New Jersey, 08901 USA
| |
Collapse
|
11
|
Cheng Z, Chen Y, Schnabl B, Chu H, Yang L. Bile acid and nonalcoholic steatohepatitis: Molecular insights and therapeutic targets. J Adv Res 2024; 59:173-187. [PMID: 37356804 PMCID: PMC11081971 DOI: 10.1016/j.jare.2023.06.009] [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: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) has been the second most common cause of liver transplantation in the United States. To date, NASH pathogenesis has not been fully elucidated but is multifactorial, involving insulin resistance, obesity, metabolic disorders, diet, dysbiosis, and gene polymorphism. An effective and approved therapy for NASH has also not been established. Bile acid is long known to have physiological detergent function in emulsifying and absorbing lipids and lipid-soluble molecules within the intestinal lumen. With more and more in-depth understandings of bile acid, it has been deemed to be a pivotal signaling molecule, which is capable of regulating lipid and glucose metabolism, liver inflammation, and fibrosis. In recent years, a plethora of studies have delineated that disrupted bile acid homeostasis is intimately correlated with NASH disease severity. AIMS The review aims to clarify the role of bile acid in hepatic lipid and glucose metabolism, liver inflammation, as well as liver fibrosis, and discusses the safety and efficacy of some pharmacological agents targeting bile acid and its associated pathways for NASH. KEY SCIENTIFIC CONCEPTS OF REVIEW Bile acid has a salutary effect on hepatic metabolic disorders, which can ameliorate liver fat accumulation and insulin resistance mainly through activating Takeda G-protein coupled receptor 5 and farnesoid X receptor. Moreover, bile acid also exerts anti-inflammation and anti-fibrosis properties. Furthermore, bile acid has great potential in nonalcoholic liver disease stratification and treatment of NASH.
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
| | - Yixiong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Huikuan Chu
- 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.
| |
Collapse
|
12
|
Tang R, Liu R, Zha H, Cheng Y, Ling Z, Li L. Gut microbiota induced epigenetic modifications in the non-alcoholic fatty liver disease pathogenesis. Eng Life Sci 2024; 24:2300016. [PMID: 38708414 PMCID: PMC11065334 DOI: 10.1002/elsc.202300016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/29/2023] [Accepted: 05/22/2023] [Indexed: 05/07/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a growing global health concern that can lead to liver disease and cancer. It is characterized by an excessive accumulation of fat in the liver, unrelated to excessive alcohol consumption. Studies indicate that the gut microbiota-host crosstalk may play a causal role in NAFLD pathogenesis, with epigenetic modification serving as a key mechanism for regulating this interaction. In this review, we explore how the interplay between gut microbiota and the host epigenome impacts the development of NAFLD. Specifically, we discuss how gut microbiota-derived factors, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), can modulate the DNA methylation and histone acetylation of genes associated with NAFLD, subsequently affecting lipid metabolism and immune homeostasis. Although the current literature suggests a link between gut microbiota and NAFLD development, our understanding of the molecular mechanisms and signaling pathways underlying this crosstalk remains limited. Therefore, more comprehensive epigenomic and multi-omic studies, including broader clinical and animal experiments, are needed to further explore the mechanisms linking the gut microbiota to NAFLD-associated genes. These studies are anticipated to improve microbial markers based on epigenetic strategies and provide novel insights into the pathogenesis of NAFLD, ultimately addressing a significant unmet clinical need.
Collapse
Affiliation(s)
- Ruiqi Tang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Rongrong Liu
- Center of Pediatric Hematology‐oncologyPediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang ProvinceNational Clinical Research Center for Child HealthChildren's HospitalZhejiang University School of MedicineHangzhouChina
| | - Hua Zha
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yiwen Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Jinan Microecological Biomedicine Shandong LaboratoryJinanChina
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Jinan Microecological Biomedicine Shandong LaboratoryJinanChina
| |
Collapse
|
13
|
Adorini L, Rigbolt K, Feigh M, Roth J, Erickson M. Increased hepatoprotective effects of the novel farnesoid X receptor agonist INT-787 versus obeticholic acid in a mouse model of nonalcoholic steatohepatitis. PLoS One 2024; 19:e0300809. [PMID: 38662778 PMCID: PMC11045142 DOI: 10.1371/journal.pone.0300809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/05/2024] [Indexed: 04/28/2024] Open
Abstract
The nuclear farnesoid X receptor (FXR), a master regulator of bile acid and metabolic homeostasis, is a key target for treatment of nonalcoholic steatohepatitis (NASH). This study compared efficacy of FXR agonists obeticholic acid (OCA) and INT-787 by liver histopathology, plasma biomarkers of liver damage, and hepatic gene expression profiles in the Amylin liver NASH (AMLN) diet-induced and biopsy-confirmed Lepob/ob mouse model of NASH. Lepob/ob mice were fed the AMLN diet for 12 weeks before liver biopsy and subsequent treatment with vehicle, OCA, or INT-787 for 8 weeks. Hepatic steatosis, inflammation, and fibrosis (liver lipids, galectin-3, and collagen 1a1 [Col1a1], respectively), as well as plasma alanine transaminase (ALT) and aspartate transaminase (AST) levels, were assessed. Hepatic gene expression was assessed in Lepob/ob mice that were fed the AMLN diet for 14 weeks then treated with vehicle, OCA, or INT-787 for 2 weeks. INT-787, which is equipotent to OCA but more hydrophilic, significantly reduced liver lipids, galectin-3, and Col1a1 compared with vehicle, and to a greater extent than OCA. INT-787 significantly reduced plasma ALT and AST levels, whereas OCA did not. INT-787 modulated a substantially greater number of genes associated with FXR signaling, lipid metabolism, and stellate cell activation relative to OCA in hepatic tissue. These findings demonstrate greater efficacy of INT-787 treatment compared with OCA in improving liver histopathology, decreasing liver enzyme levels, and enhancing gene regulation, suggesting superior clinical potential of INT-787 for the treatment of NASH and other chronic liver diseases.
Collapse
Affiliation(s)
- Luciano Adorini
- Intercept Pharmaceuticals, Inc., San Diego, California, United States of America
| | | | | | - Jonathan Roth
- Intercept Pharmaceuticals, Inc., San Diego, California, United States of America
| | - Mary Erickson
- Intercept Pharmaceuticals, Inc., San Diego, California, United States of America
| |
Collapse
|
14
|
Zhu X, Zhou Z, Pan X. Research reviews and prospects of gut microbiota in liver cirrhosis: a bibliometric analysis (2001-2023). Front Microbiol 2024; 15:1342356. [PMID: 38550860 PMCID: PMC10972893 DOI: 10.3389/fmicb.2024.1342356] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/15/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION The gut-liver axis has emerged as a focal point in chronic liver disorders, prompting more research into the role of the gut microbiota in liver cirrhosis. In individuals with liver cirrhosis, changes in the structure and function of the gut microbiota are closely tied to clinical prognosis. However, there is a scarcity of bibliometric evaluations conducted in this particular field. METHODS This study is aiming to conduct a complete analysis of the knowledge structure and centers pertaining to gut microbiota in liver cirrhosis using bibliometric methods. Publications on gut microbiota and liver cirrhosis from 2001 to 2023 are sourced from the Web of Science Core Collection. For the bibliometric analysis, we employ VOSviewer, CiteSpace, and the R package "bibliometrix". RESULTS Our study encompasses a comprehensive collection of 3109 articles originating from 96 countries, with notable contributions from leading nations such as the United States and China. The quantity of publications concerning the gut microbiota of liver cirrhosis rises annually. The University of California San Diego, Virginia Commonwealth University, Zhejiang University are the primary research institutions. World Journal of Gastroenterology publishes the most papers in this field, while hepatology is the most frequently co-cited journal. These publications come from a total of 15,965 authors, and the most prolific authors are Bajaj Jasmohan S., Schnabl Bernd and Gillevet Patrick M., while the most co-cited authors are Bajaj Jasmohan S., Younossi Zobair M., and Reiner Wiest. In addition, "dysbiosis", "gut microbiota", "intestinal barrier", "fecal microbiota transplantation", and "complement-system" are the primary keywords of research trends in recent years. DISCUSSION This study offering a comprehensive insight into the research dynamics surrounding gut microbiota in patients with liver cirrhosis. It delineates the current research frontiers and hotspots, serving as a valuable guide for scholars.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Infectious Diseases, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Ziyuan Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaxia Pan
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
15
|
Murphy WA, Diehl AM, Loop MS, Fu D, Guy CD, Abdelmalek MF, Karachaliou GS, Sjöstedt N, Neuhoff S, Honkakoski P, Brouwer KLR. Alterations in zonal distribution and plasma membrane localization of hepatocyte bile acid transporters in patients with NAFLD. Hepatol Commun 2024; 8:e0377. [PMID: 38381537 PMCID: PMC10871794 DOI: 10.1097/hc9.0000000000000377] [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: 10/09/2023] [Accepted: 12/16/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND NAFLD is highly prevalent with limited treatment options. Bile acids (BAs) increase in the systemic circulation and liver during NAFLD progression. Changes in plasma membrane localization and zonal distribution of BA transporters can influence transport function and BA homeostasis. However, a thorough characterization of how NAFLD influences these factors is currently lacking. This study aimed to evaluate the impact of NAFLD and the accompanying histologic features on the functional capacity of key hepatocyte BA transporters across zonal regions in human liver biopsies. METHODS A novel machine learning image classification approach was used to quantify relative zonal abundance and plasma membrane localization of BA transporters (bile salt export pump [BSEP], sodium-taurocholate cotransporting polypeptide, organic anion transporting polypeptide [OATP] 1B1 and OATP1B3) in non-diseased (n = 10), NAFL (n = 9), and NASH (n = 11) liver biopsies. Based on these data, membrane-localized zonal abundance (MZA) measures were developed to estimate transporter functional capacity. RESULTS NAFLD diagnosis and histologic scoring were associated with changes in transporter membrane localization and zonation. Increased periportal BSEPMZA (mean proportional difference compared to non-diseased liver of 0.090) and decreased pericentral BSEPMZA (-0.065) were observed with NASH and also in biopsies with higher histologic scores. Compared to Non-diseased Liver, periportal OATP1B3MZA was increased in NAFL (0.041) and NASH (0.047). Grade 2 steatosis (mean proportional difference of 0.043 when compared to grade 0) and grade 1 lobular inflammation (0.043) were associated with increased periportal OATP1B3MZA. CONCLUSIONS These findings provide novel mechanistic insight into specific transporter alterations that impact BA homeostasis in NAFLD. Changes in BSEPMZA likely contribute to altered BA disposition and pericentral microcholestasis previously reported in some patients with NAFLD. BSEPMZA assessment could inform future development and optimization of NASH-related pharmacotherapies.
Collapse
Affiliation(s)
- William A. Murphy
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew Shane Loop
- Department of Health Outcomes Research and Policy, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Dong Fu
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cynthia D. Guy
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Manal F. Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Georgia Sofia Karachaliou
- Division of Gastroenterology and Hepatology, Duke University Medical Center, Durham, North Carolina, USA
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Paavo Honkakoski
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - 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
| |
Collapse
|
16
|
Xu H, Yuan M, Niu K, Yang W, Jiang M, Zhang L, Zhou J. Involvement of Bile Acid Metabolism and Gut Microbiota in the Amelioration of Experimental Metabolism-Associated Fatty Liver Disease by Nobiletin. Molecules 2024; 29:976. [PMID: 38474489 DOI: 10.3390/molecules29050976] [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: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolism-associated fatty liver disease (MAFLD), a growing health problem worldwide, is one of the major risks for the development of cirrhosis and liver cancer. Oral administration of nobiletin (NOB), a natural citrus flavonoid, modulates the gut microbes and their metabolites in mice. In the present study, we established a mouse model of MAFLD by subjecting mice to a high-fat diet (HFD) for 12 weeks. Throughout this timeframe, NOB was administered to investigate its potential benefits on gut microbial balance and bile acid (BA) metabolism using various techniques, including 16S rRNA sequencing, targeted metabolomics of BA, and biological assays. NOB effectively slowed the progression of MAFLD by reducing serum lipid levels, blood glucose levels, LPS levels, and hepatic IL-1β and TNF-α levels. Furthermore, NOB reinstated diversity within the gut microbial community, increasing the population of bacteria that produce bile salt hydrolase (BSH) to enhance BA excretion. By exploring further, we found NOB downregulated hepatic expression of the farnesoid X receptor (FXR) and its associated small heterodimer partner (SHP), and it increased the expression of downstream enzymes, including cholesterol 7α-hydroxylase (CYP7A1) and cytochrome P450 27A1 (CYP27A1). This acceleration in cholesterol conversion within the liver contributes to mitigating MAFLD. The present findings underscore the significant role of NOB in regulating gut microbial balance and BA metabolism, revealing that long-term intake of NOB plays beneficial roles in the prevention or intervention of MAFLD.
Collapse
Affiliation(s)
- Hongling Xu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mingming Yuan
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Kailin Niu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Yang
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Maoyuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Jing Zhou
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| |
Collapse
|
17
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [PMID: 37482263 DOI: 10.1016/j.jep.2023.116933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria-coptis herb couple (SC) is a classic herbal pair used in many Traditional Chinese Medicine (TCM) formulations in the treatment of endocrine and metabolic deseases. Diabetes mellitus and non-alcoholic steatohepatitis (NASH) are both endocrine and metabolic diseases. Previous studies have shown that SC has anti-diabetic effects. However, the effect and mechanism of SC against NASH remains unclear. AIM OF THE STUDY This study aimed to demonstrate the effect and mechanism of SC against NASH through the nuclear factor-erythroid 2-related factor 2 (Nrf2) and farnesoid X receptor (FXR) dual signaling pathways in vivo and in vitro. MATERIALS AND METHODS The high fat diet-fed rat model, and HepG2 and RAW264.7 cell models were used. Serum biochemical indexes and liver histopathological changes were examined. Metabolomics, transcriptomics, and flow cytometry were performed. RT-qPCR and western blot analysis were performed to provide expression of NRF2 and FXR pathway signal molecules during SC's anti-NASH treatment in vivo and in vitro. RESULTS SC had anti-NASH effects in vivo with significantly improvement of serum NASH biochemical index and hepatopathological structure; meanwhile, SC significantly elevated the expression levels of FXR protein in liver and intestinal tissues, and cholesterol 7a-hydroxylase (CYP7A1) protein in liver. The mRNA expression levels of Takeda G protein receptor 5 (TGR5), CYP7A1, fibroblast growth factor receptor-4 (FGFR4), FXR, small heterodimer partner (SHP), fibroblast growth factor 15/19 (FGF15/19) and glucagon-like peptide-1 (GLP-1) were significantly elevated by SC. SC reduced the levels of NorCA, isoLCA and α-MCA in the feces of NAFLD rats. In vitro, SC-containing serum (SC-CS) was found to significantly reduce intracellular lipid deposition, inhibit ROS production, reduce intracellular Malondialdehyde (MDA) and IL-1β levels, and enhance the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Six differential genes closely related to oxidative stress and Nrf2 were identified by transcriptomic analysis. SC-CS up-regulated the expression of NRF2, and reduced the expression of TXNIP and Caspase-1 genes in RAW264.7 cells. In addition, SC-CS reduced the expression of Keap1 and NF-κB, and up-regulated the expression of Nrf2, heme oxygenase-1 (HO-1), quinone oxidoreductase 1 (NQO1), and SOD; SC-CS elevated the protein level of NRF2, and reduced the protein level of TXNIP in HepG2 cells. CONCLUSIONS the mechanisms of SC action against NASH was closely related to the simultaneous activations of both NRF2 and FXR signaling pathways. These findings provide a new insight into the anti-NASH application of SC in clinical settings and demonstrate the potential of SC in the treatment of NASH.
Collapse
Affiliation(s)
- Yanan Xue
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Yue Wei
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Lan Cao
- Research Center of Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, PR China
| | - Min Shi
- College of Life Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Junqing Sheng
- College of Life Science, Nanchang University, Nanchang, 330031, PR China
| | - Qin Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ziwen Cheng
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Tao Luo
- First Affiliated Hospital of Nanchang University, 330006, PR China
| | - Quanhui Jiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ailan Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Lingyun Zhong
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Changhua Zhang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China; Nanchang Research Institute, Sun Yat-sen University, Jiangxi, 330096, PR China.
| |
Collapse
|
18
|
Jin S, Chen P, Yang J, Li D, Liu X, Zhang Y, Xia Q, Li Y, Chen G, Li Y, Tong Y, Yu W, Fan X, Lin H. Phocaeicola vulgatus alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA. Gut Microbes 2024; 16:2309683. [PMID: 38312099 PMCID: PMC10854360 DOI: 10.1080/19490976.2024.2309683] [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: 09/26/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify Phocaeicola vulgatus (P. vulgatus) as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of P. vulgatus mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by P. vulgatus, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of P. vulgatus in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.
Collapse
Affiliation(s)
- Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyin Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Xia
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiling Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoqiao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yixuan Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Tong
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weihua Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [DOI: https:/doi.org/10.1016/j.jep.2023.116933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2024]
|
20
|
Zhang M, Xiao B, Chen X, Ou B, Wang S. Physical exercise plays a role in rebalancing the bile acids of enterohepatic axis in non-alcoholic fatty liver disease. Acta Physiol (Oxf) 2024; 240:e14065. [PMID: 38037846 DOI: 10.1111/apha.14065] [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: 05/26/2023] [Revised: 10/09/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered as one of the most common diseases of lipid metabolism disorders, which is closely related to bile acids disorders and gut microbiota disorders. Bile acids are synthesized from cholesterol in the liver, and processed by gut microbiota in intestinal tract, and participate in metabolic regulation through the enterohepatic circulation. Bile acids not only promote the consumption and absorption of intestinal fat but also play an important role in biological metabolic signaling network, affecting fat metabolism and glucose metabolism. Studies have demonstrated that exercise plays an important role in regulating the composition and function of bile acid pool in enterohepatic axis, which maintains the homeostasis of the enterohepatic circulation and the health of the host gut microbiota. Exercise has been recommended by several health guidelines as the first-line intervention for patients with NAFLD. Can exercise alter bile acids through the microbiota in the enterohepatic axis? If so, regulating bile acids through exercise may be a promising treatment strategy for NAFLD. However, the specific mechanisms underlying this potential connection are largely unknown. Therefore, in this review, we tried to review the relationship among NAFLD, physical exercise, bile acids, and gut microbiota through the existing data and literature, highlighting the role of physical exercise in rebalancing bile acid and microbial dysbiosis.
Collapse
Affiliation(s)
- Minyu Zhang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Biyang Xiao
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Xiaoqi Chen
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Bingming Ou
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Songtao Wang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| |
Collapse
|
21
|
Pereira ENGDS, de Araujo BP, Rodrigues KL, Silvares RR, Guimarães FV, Martins CSM, Flores EEI, Silva PMRE, Daliry A. Cholesterol Exacerbates the Pathophysiology of Non-Alcoholic Steatohepatitis by Upregulating Hypoxia-Inducible Factor 1 and Modulating Microcirculatory Dysfunction. Nutrients 2023; 15:5034. [PMID: 38140293 PMCID: PMC10745917 DOI: 10.3390/nu15245034] [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: 11/04/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cholesterol is a pivotal lipotoxic molecule that contributes to the progression of Non-Alcoholic Steatohepatitis NASH). Additionally, microcirculatory changes are critical components of Non-Alcoholic Fatty Liver Disease (NAFLD) pathogenesis. This study aimed to investigate the role of cholesterol as an insult that modulates microcirculatory damage in NAFLD and the underlying mechanisms. The experimental model was established in male C57BL/6 mice fed a high-fat high-carbohydrate (HFHC) diet for 39 weeks. Between weeks 31-39, 2% cholesterol was added to the HFHC diet in a subgroup of mice. Leukocyte recruitment and hepatic stellate cells (HSC) activation in microcirculation were assessed using intravital microscopy. The hepatic microvascular blood flow (HMBF) was measured using laser speckle flowmetry. High cholesterol levels exacerbated hepatomegaly, hepatic steatosis, inflammation, fibrosis, and leukocyte recruitment compared to the HFHC group. In addition, cholesterol decreased the HMBF-cholesterol-induced activation of HSC and increased HIF1A expression in the liver. Furthermore, cholesterol promoted a pro-inflammatory cytokine profile with a Th1-type immune response (IFN-γ/IL-4). These findings suggest cholesterol exacerbates NAFLD progression through microcirculatory dysfunction and HIF1A upregulation through hypoxia and inflammation. This study highlights the importance of cholesterol-induced lipotoxicity, which causes microcirculatory dysfunction associated with NAFLD pathology, thus reinforcing the potential of lipotoxicity and microcirculation as therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Evelyn Nunes Goulart da Silva Pereira
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Beatriz Peres de Araujo
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Karine Lino Rodrigues
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Raquel Rangel Silvares
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Fernanda Verdini Guimarães
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Carolina Souza Machado Martins
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | - Edgar Eduardo Ilaquita Flores
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| | | | - Anissa Daliry
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (E.N.G.d.S.P.)
| |
Collapse
|
22
|
Georgieva M, Xenodochidis C, Krasteva N. Old age as a risk factor for liver diseases: Modern therapeutic approaches. Exp Gerontol 2023; 184:112334. [PMID: 37977514 DOI: 10.1016/j.exger.2023.112334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Recent scientific interest has been directed towards age-related diseases, driven by the significant increase in global life expectancy and the growing population of individuals aged 65 and above. The ageing process encompasses various biological, physiological, environmental, psychological, behavioural, and social changes, leading to an augmented susceptibility to chronic illnesses. Cardiovascular, neurological, musculoskeletal, liver and oncological diseases are prevalent in the elderly. Moreover, ageing individuals demonstrate reduced regenerative capacity and decreased tolerance towards therapeutic interventions, including organ transplantation. Liver diseases, such as non-alcoholic fatty liver disease, alcoholic liver disease, hepatitis, fibrosis, and cirrhosis, have emerged as significant public health concerns. Paradoxically, these conditions remain underestimated despite their substantial global impact. Age-related factors are closely associated with the severity and unfavorable prognosis of various liver diseases, warranting further investigation to enhance clinical management and develop novel therapeutic strategies. This comprehensive review focuses specifically on age-related liver diseases, their treatment strategies, and contemporary practices. It provides a detailed account of the global burden, types, molecular mechanisms, and epigenetic alterations underlying these liver pathologies.
Collapse
Affiliation(s)
- Milena Georgieva
- Institute of Molecular Biology "Acad. Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| | - Charilaos Xenodochidis
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| |
Collapse
|
23
|
Theys C, Ibrahim J, Mateiu L, Mposhi A, García-Pupo L, De Pooter T, De Rijk P, Strazisar M, İnce İA, Vintea I, Rots MG, Vanden Berghe W. Mitochondrial GpC and CpG DNA Hypermethylation Cause Metabolic Stress-Induced Mitophagy and Cholestophagy. Int J Mol Sci 2023; 24:16412. [PMID: 38003603 PMCID: PMC10671279 DOI: 10.3390/ijms242216412] [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: 09/14/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by a constant accumulation of lipids in the liver. This hepatic lipotoxicity is associated with a dysregulation of the first step in lipid catabolism, known as beta oxidation, which occurs in the mitochondrial matrix. Eventually, this dysregulation will lead to mitochondrial dysfunction. To evaluate the possible involvement of mitochondrial DNA methylation in this lipid metabolic dysfunction, we investigated the functional metabolic effects of mitochondrial overexpression of CpG (MSssI) and GpC (MCviPI) DNA methyltransferases in relation to gene expression and (mito)epigenetic signatures. Overall, the results show that mitochondrial GpC and, to a lesser extent, CpG methylation increase bile acid metabolic gene expression, inducing the onset of cholestasis through mito-nuclear epigenetic reprogramming. Moreover, both increase the expression of metabolic nuclear receptors and thereby induce basal overactivation of mitochondrial respiration. The latter promotes mitochondrial swelling, favoring lipid accumulation and metabolic-stress-induced mitophagy and autophagy stress responses. In conclusion, both mitochondrial GpC and CpG methylation create a metabolically challenging environment that induces mitochondrial dysfunction, which may contribute to the progression of MASLD.
Collapse
Affiliation(s)
- Claudia Theys
- Lab Protein Chemistry, Proteomics & Epigenetic Signaling (PPES), Department Biomedical Sciences, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium; (C.T.)
| | - Joe Ibrahim
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium
| | - Ligia Mateiu
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium
| | - Archibold Mposhi
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Laura García-Pupo
- Lab Protein Chemistry, Proteomics & Epigenetic Signaling (PPES), Department Biomedical Sciences, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium; (C.T.)
| | - Tim De Pooter
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Wilrijk, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Wirlijk, 2610 Antwerp, Belgium
| | - Peter De Rijk
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Wilrijk, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Wirlijk, 2610 Antwerp, Belgium
| | - Mojca Strazisar
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Wilrijk, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Wirlijk, 2610 Antwerp, Belgium
| | - İkbal Agah İnce
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Medical Microbiology, School of Medicine, Acıbadem Mehmet, Ali Aydınlar University, 34752 Ataşehir, İstanbul, Türkiye
| | - Iuliana Vintea
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium
| | - Marianne G. Rots
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Wim Vanden Berghe
- Lab Protein Chemistry, Proteomics & Epigenetic Signaling (PPES), Department Biomedical Sciences, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium; (C.T.)
| |
Collapse
|
24
|
Filipovic B, Marjanovic-Haljilji M, Mijac D, Lukic S, Kapor S, Kapor S, Starcevic A, Popovic D, Djokovic A. Molecular Aspects of MAFLD-New Insights on Pathogenesis and Treatment. Curr Issues Mol Biol 2023; 45:9132-9148. [PMID: 37998750 PMCID: PMC10669943 DOI: 10.3390/cimb45110573] [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: 10/05/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic-associated liver disease (MAFLD) affects up to 70% of overweight and more than 90% of morbidly obese people, and its pathogenesis is rather complex and multifactorial. The criteria for MAFLD include the presence of hepatic steatosis in addition to one of the following three criteria: overweight or obesity, presence of type 2 diabetes mellitus (T2DM), or evidence of metabolic dysregulation. If the specific criteria are present, the diagnosis of MAFLD can be made regardless of alcohol consumption and previous liver disease. The pathophysiological mechanisms of MAFLD, including inflammation, lipotoxicity, mitochondrial disfunction, and oxidative stress, as well as the impact of intestinal gut microbiota, are constantly being elucidated. Treatment strategies that are continually emerging are based on different key points in MAFLD pathogenesis. Yet, the ideal therapeutic option has still not been found and future research is of great importance, as MAFLD represents a multisystemic disease with numerous complications.
Collapse
Affiliation(s)
- Branka Filipovic
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
| | - Marija Marjanovic-Haljilji
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
| | - Dragana Mijac
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Clinic of Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia
| | - Snezana Lukic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Clinic of Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia;
| | - Slobodan Kapor
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Institute of Anatomy “Niko Miljanic”, Dr Subotica Starijeg 4/2, 11000 Belgrade, Serbia
| | - Ana Starcevic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Institute of Anatomy “Niko Miljanic”, Dr Subotica Starijeg 4/2, 11000 Belgrade, Serbia
| | - Dusan Popovic
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
| | - Aleksandra Djokovic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Department of Cardiology, Clinical and Hospital Center “Bezanijska Kosa”, Dr Zorza Matea s/n, 11080 Belgrade, Serbia
| |
Collapse
|
25
|
Arshad U, Husnain A, Poindexter MB, Zimpel R, Nelson CD, Santos JEP. Rumen-protected choline reduces hepatic lipidosis by increasing hepatic triacylglycerol-rich lipoprotein secretion in dairy cows. J Dairy Sci 2023; 106:7630-7650. [PMID: 37641262 DOI: 10.3168/jds.2022-23182] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/25/2023] [Indexed: 08/31/2023]
Abstract
Objectives were to determine the effects of supplementing rumen-protected choline (RPC) on hepatic composition and secretion of triacylglycerol-rich lipoprotein when cows were subjected to feed restriction to develop fatty liver. It was hypothesized that RPC reduces hepatic triacylglycerol by enhancing secretion of hepatic lipoprotein. Pregnant, nonlactating parous Holstein cows (n = 33) at mean (± standard deviation) 234 ± 2.2 d of gestation were blocked by body condition (3.79 ± 0.49) and assigned to receive 0 g/d (CON), 25.8 g/d choline ion from a RPC product containing 28.8% choline chloride (CC; treatment L25.8), or 25.8 g/d of choline ion from a RPC product containing 60.0% CC (H25.8). Cows were fed for ad libitum intake for the first 5 d and restricted to 41% of the net energy for lactation required for maintenance and pregnancy from d 6 to 13. Intake of metabolizable methionine was maintained at 18 g/d during feed restriction by supplying rumen-protected methionine. Hepatic tissue was sampled on d 6 and 13 and analyzed for triacylglycerol and glycogen, and mRNA expression of hepatic tissue was investigated. On d 14, cows were not fed and received a 10% solution of tyloxapol intravenously at 120 mg/kg of body weight to block hydrolysis of triacylglycerols in very low density lipoprotein (VLDL). Blood was sampled sequentially for 720 min and analyzed for concentration of triacylglycerol and total cholesterol. Lymph was sampled 6 h after tyloxapol infusion, and analyzed for concentrations of fatty acids, β-hydroxybutyrate, glucose, triacylglycerol, and total cholesterol. A sample of serum collected at 720 min after tyloxapol was assayed for the metabolome composition. The area under the curve (AUC) of serum triacylglycerol, VLDL cholesterol, and total cholesterol were calculated. Orthogonal contrasts evaluated the effect of supplementing RPC (CON vs. [1/2 L25.8 + 1/2 H25.8]) and source of RPC (L25.8 vs. H25.8). Least squares means and standard errors of the means are presented in sequence as CON, L25.8, H25.8. During feed restriction, supplementation of RPC reduced hepatic triacylglycerol (9.0 vs. 4.1 vs. 4.5 ± 0.6%) and increased glycogen contents (1.9 vs. 3.5 vs. 4.1 ± 0.2%). Similarly, supplementation of RPC increased the expression of transcripts involved in the synthesis and assembly of lipoproteins (MTTP), cellular autophagy (ATG3), and inflammation (TNFA), and reduced the expression of transcripts associated with mitochondrial oxidation of fatty acids (HADHA, MLYCD) and stabilization of lipid droplets (PLIN2). After infusion of tyloxapol, RPC increased the AUC for serum triacylglycerol (21,741 vs. 32,323 vs. 28,699 ± 3,706 mg/dL × min) and VLDL cholesterol (4,348 vs. 6,465 vs. 5,740 ± 741 mg/dL × min) but tended to reduce the concentrations of triacylglycerol in lymph (16.7 vs. 13.8 vs. 11.9 ± 1.9 mg/dL). Feeding RPC tended to increase the concentrations of 89 metabolites in serum, after adjusting for false discovery, including 3 acylcarnitines, 1 AA-related metabolite, 11 bile acids, 1 ceramide, 6 diacylglycerols, 2 dihydroceramides, 1 glycerophospholipid, and 64 triacylglycerols compared with CON. Feeding 25.8 g/d of choline ion as RPC mediated increased hepatic triacylglycerol secretion to promote lipotropic effects that reduced hepatic lipidosis in dairy cows.
Collapse
Affiliation(s)
- U Arshad
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611
| | - A Husnain
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611
| | - M B Poindexter
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611
| | - R Zimpel
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611
| | - C D Nelson
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611
| | - J E P Santos
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, FL 32611.
| |
Collapse
|
26
|
Pal SC, Castillo-Castañeda SM, Díaz-Orozco LE, Ramírez-Mejía MM, Dorantes-Heredia R, Alonso-Morales R, Eslam M, Lammert F, Méndez-Sánchez N. Molecular Mechanisms Involved in MAFLD in Cholecystectomized Patients: A Cohort Study. Genes (Basel) 2023; 14:1935. [PMID: 37895284 PMCID: PMC10606482 DOI: 10.3390/genes14101935] [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: 08/03/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Gallstone disease and metabolic dysfunction-associated fatty liver disease (MAFLD) share numerous common risk factors and progression determinants in that they both manifest as organ-specific consequences of metabolic dysfunction. Nevertheless, the precise molecular mechanisms underlying fibrosis development in cholecystectomized MAFLD patients remain inadequately defined. This study aimed to investigate the involvement of farnesoid X receptor 1 (FXR1) and fibroblast growth factor receptor 4 (FGFR4) in the progression of fibrosis in cholecystectomized MAFLD patients. A meticulously characterized cohort of 12 patients diagnosed with MAFLD, who had undergone liver biopsies during programmed cholecystectomies, participated in this study. All enrolled patients underwent a follow-up regimen at 1, 3, and 6 months post-cholecystectomy, during which metabolic biochemical markers were assessed, along with elastography, which served as indirect indicators of fibrosis. Additionally, the hepatic expression levels of FGFR4 and FXR1 were quantified using quantitative polymerase chain reaction (qPCR). Our findings revealed a robust correlation between hepatic FGFR4 expression and various histological features, including the steatosis degree (r = 0.779, p = 0.023), ballooning degeneration (r = 0.764, p = 0.027), interphase inflammation (r = 0.756, p = 0.030), and steatosis activity score (SAS) (r = 0.779, p = 0.023). Conversely, hepatic FXR1 expression did not exhibit any significant correlations with these histological features. In conclusion, our study highlights a substantial correlation between FGFR4 expression and histological liver damage, emphasizing its potential role in lipid and glucose metabolism. These findings suggest that FGFR4 may play a crucial role in the progression of fibrosis in cholecystectomized MAFLD patients. Further research is warranted to elucidate the exact mechanisms through which FGFR4 influences metabolic dysfunction and fibrosis in this patient population.
Collapse
Affiliation(s)
- Shreya C. Pal
- Faculty of Medicine, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico; (S.C.P.); (L.E.D.-O.)
| | - Stephany M. Castillo-Castañeda
- Medical, Dental and Health Sciences Master and Doctorate Program, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico;
- Liver Research Unit, Medica Sur Clinic & Foundation, Tlalpan, Mexico City 14050, Mexico; (M.M.R.-M.); (R.D.-H.)
| | - Luis E. Díaz-Orozco
- Faculty of Medicine, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico; (S.C.P.); (L.E.D.-O.)
| | - Mariana M. Ramírez-Mejía
- Liver Research Unit, Medica Sur Clinic & Foundation, Tlalpan, Mexico City 14050, Mexico; (M.M.R.-M.); (R.D.-H.)
- Plan of Combined Studies in Medicine, Faculty of Medicine, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico
| | - Rita Dorantes-Heredia
- Liver Research Unit, Medica Sur Clinic & Foundation, Tlalpan, Mexico City 14050, Mexico; (M.M.R.-M.); (R.D.-H.)
| | - Rogelio Alonso-Morales
- Genetic and Molecular Biology Laboratory, Faculty of Veterinary, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico;
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital, University of Sydney, Sydney, NSW 2145, Australia;
| | - Frank Lammert
- Health Sciences, Hannover Medical School (MHH), 30625 Hannover, Germany;
| | - Nahum Méndez-Sánchez
- Faculty of Medicine, National Autonomous University of Mexico, Tlalpan, Mexico City 04510, Mexico; (S.C.P.); (L.E.D.-O.)
- Liver Research Unit, Medica Sur Clinic & Foundation, Tlalpan, Mexico City 14050, Mexico; (M.M.R.-M.); (R.D.-H.)
| |
Collapse
|
27
|
Liu L, Deng L, Wei W, Li C, Lu Y, Bai J, Li L, Zhang H, Jin N, Li C, Zhao C. Lactiplantibacillus plantarum LPJZ-658 Improves Non-Alcoholic Steatohepatitis by Modulating Bile Acid Metabolism and Gut Microbiota in Mice. Int J Mol Sci 2023; 24:13997. [PMID: 37762300 PMCID: PMC10531215 DOI: 10.3390/ijms241813997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is one of the most prevalent diseases worldwide; it is characterized by hepatic lipid accumulation, inflammation, and progressive fibrosis. Here, a Western diet combined with low-dose weekly carbon tetrachloride was fed to C57BL/6J mice for 12 weeks to build a NASH model to investigate the attenuating effects and possible mechanisms of Lactiplantibacillus plantarum LPJZ-658. Hepatic pathology, lipid profiles, and gene expression were assessed. The metabolomic profiling of the serum was performed. The composition structure of gut microbiota was profiled using 16s rRNA sequencing. The results show that LPJZ-658 treatment significantly attenuated liver injury, steatosis, fibrosis, and inflammation in NASH mice. Metabolic pathway analysis revealed that several pathways, such as purine metabolism, glycerophospholipid metabolism, linoleic acid metabolism, and primary bile acid biosynthesis, were associated with NASH. Notably, we found that treatment with LPJZ-658 regulated the levels of bile acids (BAs) in the serum. Moreover, LPJZ-658 restored NASH-induced gut microbiota dysbiosis. The correlation analysis deduced obvious interactions between BAs and gut microbiota. The current study indicates that LPJZ-658 supplementation protects against NASH progression, which is accompanied by alternating BA metabolic and modulating gut microbiota.
Collapse
Affiliation(s)
- Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Liquan Deng
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Chunhua Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Yuting Lu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Jieying Bai
- College of Future Technology, Peking University, Beijing 100871, China;
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| |
Collapse
|
28
|
Zhong J, He X, Gao X, Liu Q, Zhao Y, Hong Y, Zhu W, Yan J, Li Y, Li Y, Zheng N, Bao Y, Wang H, Ma J, Huang W, Liu Z, Lyu Y, Ke X, Jia W, Xie C, Hu Y, Sheng L, Li H. Hyodeoxycholic acid ameliorates nonalcoholic fatty liver disease by inhibiting RAN-mediated PPARα nucleus-cytoplasm shuttling. Nat Commun 2023; 14:5451. [PMID: 37673856 PMCID: PMC10482907 DOI: 10.1038/s41467-023-41061-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is usually characterized with disrupted bile acid (BA) homeostasis. However, the exact role of certain BA in NAFLD is poorly understood. Here we show levels of serum hyodeoxycholic acid (HDCA) decrease in both NAFLD patients and mice, as well as in liver and intestinal contents of NAFLD mice compared to their healthy counterparts. Serum HDCA is also inversely correlated with NAFLD severity. Dietary HDCA supplementation ameliorates diet-induced NAFLD in male wild type mice by activating fatty acid oxidation in hepatic peroxisome proliferator-activated receptor α (PPARα)-dependent way because the anti-NAFLD effect of HDCA is abolished in hepatocyte-specific Pparα knockout mice. Mechanistically, HDCA facilitates nuclear localization of PPARα by directly interacting with RAN protein. This interaction disrupts the formation of RAN/CRM1/PPARα nucleus-cytoplasm shuttling heterotrimer. Our results demonstrate the therapeutic potential of HDCA for NAFLD and provide new insights of BAs on regulating fatty acid metabolism.
Collapse
Affiliation(s)
- Jing Zhong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, 313000, China
| | - Xiaofang He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinxin Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiaohong Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Hong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Juan Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yifan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Bao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hao Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Junli Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zekun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuanzhi Lyu
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Xisong Ke
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Jia
- Center for Translational Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, 999077, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
29
|
Lee SM, Jun DW, Yoon EL, Oh JH, Roh YJ, Lee EJ, Shin JH, Nam YD, Kim HS. Discovery biomarker to optimize obeticholic acid treatment for non-alcoholic fatty liver disease. Biol Direct 2023; 18:50. [PMID: 37626369 PMCID: PMC10463927 DOI: 10.1186/s13062-023-00407-4] [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: 01/28/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The response rate to obeticholic acid (OCA), a potential therapeutic agent for non-alcoholic fatty liver disease, is limited. This study demonstrated that upregulation of the alternative bile acid synthesis pathway increases the OCA treatment response rate. The hepatic transcriptome and bile acid metabolite profile analyses revealed that the alternative bile acid synthesis pathway (Cyp7b1 and muricholic acid) in the OCA-responder group were upregulated compared with those in the OCA-non-responder group. Intestinal microbiome analysis also revealed that the abundances of Bacteroidaceae, Parabacteroides, and Bacteroides, which were positively correlated with the alternative bile acid synthesis pathway, were higher in the OCA-responder group than in the non-responder group. Pre-study hepatic mRNA levels of Cyp8b1 (classic pathway) were downregulated in the OCA-responder group. The OCA response rate increased up to 80% in cases with a hepatic Cyp7b1/Cyp8b1 ratio ≥ 5.0. Therefore, the OCA therapeutic response can be evaluated based on the Cyp7b1/Cyp8b1 ratio or the alternative/classic bile acid synthesis pathway activity.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dae Won Jun
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Republic of Korea.
- Department of Internal Medicine, Hanyang University Hospital, Hanyang University College of Medicine, 17 Haengdang-dong, Sungdong-gu, Seoul, 133-792, Republic of Korea.
| | - Eileen Laurel Yoon
- Department of Internal Medicine, Hanyang University Hospital, Hanyang University College of Medicine, 17 Haengdang-dong, Sungdong-gu, Seoul, 133-792, Republic of Korea.
| | - Ju Hee Oh
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Severance Hospital, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Jin Roh
- Department of Dermatology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Eun Jeoung Lee
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Republic of Korea
| | - Ji-Hee Shin
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea
| | - Young-Do Nam
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea
| | - Hyun Sung Kim
- Pathology, Medical genetic, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
30
|
Wu R, Prachyathipsakul T, Zhuang J, Liu H, Han Y, Liu B, Gong S, Qiu J, Wong S, Ribbe A, Medeiros J, Bhagabati J, Gao J, Wu P, Dutta R, Herrera R, Faraci S, Xiao H, Thayumanavan S. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS NEXUS 2023; 2:pgad252. [PMID: 37649581 PMCID: PMC10465086 DOI: 10.1093/pnasnexus/pgad252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Optimization of metabolic regulation is a promising solution for many pathologies, including obesity, dyslipidemia, type 2 diabetes, and inflammatory liver disease. Synthetic thyroid hormone mimics-based regulation of metabolic balance in the liver showed promise but was hampered by the low biocompatibility and harmful effects on the extrahepatic axis. In this work, we show that specifically directing the thyromimetic to the liver utilizing a nanogel-based carrier substantially increased therapeutic efficacy in a diet-induced obesity mouse model, evidenced by the near-complete reversal of body weight gain, liver weight and inflammation, and cholesterol levels with no alteration in the thyroxine (T4) / thyroid stimulating hormone (TSH) axis. Mechanistically, the drug acts by binding to thyroid hormone receptor β (TRβ), a ligand-inducible transcription factor that interacts with thyroid hormone response elements and modulates target gene expression. The reverse cholesterol transport (RCT) pathway is specifically implicated in the observed therapeutic effect. Overall, the study demonstrates a unique approach to restoring metabolic regulation impacting obesity and related metabolic dysfunctions.
Collapse
Affiliation(s)
- Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Theeraphop Prachyathipsakul
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hongxu Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingyi Qiu
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siu Wong
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Alexander Ribbe
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jayashree Bhagabati
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Ranit Dutta
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | - Hang Xiao
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
31
|
Liu X, Wang J, Li M, Qiu J, Li X, Qi L, Liu J, Liu P, Xie G, Wang X. Farnesoid X receptor is an important target for the treatment of disorders of bile acid and fatty acid metabolism in mice with nonalcoholic fatty liver disease combined with cholestasis. J Gastroenterol Hepatol 2023; 38:1438-1446. [PMID: 37415275 DOI: 10.1111/jgh.16279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND AND AIM The prevalence of nonalcoholic fatty liver disease (NAFLD) has been rising globally. NAFLD patients combined with cholestasis have more obvious liver fibrosis, impaired bile acid (BA), and fatty acid (FA) metabolism and severer liver injury; however, its therapeutic options are limited, and the underlying metabolic mechanisms are understood. Here, we aimed to investigate the effects of farnesoid X receptor (FXR) on BA and FA metabolism in NAFLD combined with cholestasis and related signaling pathways. METHODS A mouse model of NAFLD combined with cholestasis was established by joint intervention with high-fat diet (HFD) and alpha-naphthylisothiocyanate. The effects of FXR on BA and FA metabolism were evaluated by serum biochemical analysis. Liver damage was identified by histopathology. The expression of nuclear hormone receptor, membrane receptor, FA transmembrane transporter, and BA transporter protein in mice were measured by western blot. RESULTS NAFLD mice combined with cholestasis developed more severe cholestasis and dysregulated BA and FA metabolism. Meanwhile, the expression of FXR protein was decreased in NAFLD mice combined with cholestasis compared to the controls. Fxr-/- mice showed liver injury. HFD aggravated the liver injury with decreased BSEP expression, increased expression of NTCP, LXRα, SREBP-1c, FAS, ACC1, and CD36, and significantly increased BA and FA accumulation. CONCLUSION All the results suggested that FXR plays a key role in both FA and BA metabolism in NAFLD combined with cholestasis and thus may be a potential target for the treatment of disorders of BA and FA metabolism in NAFLD combined with cholestasis.
Collapse
Affiliation(s)
- Xinzhu Liu
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiaxuan Wang
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Maogang Li
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China
| | - Jiannan Qiu
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xingying Li
- South China Normal University, Guangzhou, 510631, Guangdong, China
| | - Li Qi
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Liu
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping Liu
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guoxiang Xie
- Human Metabolomics Institute, Inc., Shenzhen, 518109, Guangdong, China
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiaoning Wang
- Basic Research Center of Traditional Chinese Medicine Prescription and Syndrome, Institute of Interdisciplinary Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
32
|
Kreimeyer H, Vogt K, Götze T, Best J, Götze O, Weigt J, Kahraman A, Özçürümez M, Kälsch J, Syn WK, Sydor S, Canbay A, Manka P. Influence of the Bile Acid Transporter Genes ABCB4, ABCB8, and ABCB11 and the Farnesoid X Receptor on the Response to Ursodeoxycholic Acid in Patients with Nonalcoholic Steatohepatitis. J Pers Med 2023; 13:1180. [PMID: 37511794 PMCID: PMC10381823 DOI: 10.3390/jpm13071180] [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: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The prevalence of NAFLD and NASH is increasing worldwide, and there is no approved medical treatment until now. Evidence has emerged that interfering with bile acid metabolism may lead to improvement in NASH. In this study, 28 patients with elevated cholestatic liver function tests (especially GGT) were screened for bile acid gene polymorphisms and treated with UDCA. All patients had a bile acid gene polymorphism in ABCB4 or ABCB11. Treatment with UDCA for 12 months significantly reduced GGT in all patients and ALT in homozygous patients. No difference in fibrosis was observed using FIb-4, NFS, and transient elastography (TE). PNPLA3 and TM6SF2 were the most common NASH-associated polymorphisms, and patients with TM6SF2 showed a significant reduction in GGT and ALT with the administration of UDCA. In conclusion, NASH patients with elevated GGT should be screened for bile acid gene polymorphisms, as UDCA therapy may improve liver function tests. However, no difference in clinical outcomes, such as progression to cirrhosis, has been observed using non-invasive tests (NITs).
Collapse
Affiliation(s)
- Henriette Kreimeyer
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Katharina Vogt
- Department of Gastroenterology and Hepatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Tobias Götze
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto-von-Guericke-University Hospital Magdeburg, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | - Jan Best
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Oliver Götze
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Jochen Weigt
- Department of Gastroenterology and Hepatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Alisan Kahraman
- Department of Gastroenterology and Hepatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Mustafa Özçürümez
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Julia Kälsch
- Department of Gastroenterology and Hepatology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Svenja Sydor
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Ali Canbay
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Paul Manka
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| |
Collapse
|
33
|
Lu H, Ma J, Li Y, Zhang J, An Y, Du W, Cai X. Bioinformatic and systems biology approach revealing the shared genes and molecular mechanisms between COVID-19 and non-alcoholic hepatitis. Front Mol Biosci 2023; 10:1164220. [PMID: 37405258 PMCID: PMC10315682 DOI: 10.3389/fmolb.2023.1164220] [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: 02/12/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction: Coronavirus disease 2019 (COVID-19) has become a global pandemic and poses a serious threat to human health. Many studies have shown that pre-existing nonalcoholic steatohepatitis (NASH) can worsen the clinical symptoms in patients suffering from COVID-19. However, the potential molecular mechanisms between NASH and COVID-19 remain unclear. To this end, key molecules and pathways between COVID-19 and NASH were herein explored by bioinformatic analysis. Methods: The common differentially expressed genes (DEGs) between NASH and COVID-19 were obtained by differential gene analysis. Enrichment analysis and protein-protein interaction (PPI) network analysis were carried out using the obtained common DEGs. The key modules and hub genes in PPI network were obtained by using the plug-in of Cytoscape software. Subsequently, the hub genes were verified using datasets of NASH (GSE180882) and COVID-19 (GSE150316), and further evaluated by principal component analysis (PCA) and receiver operating characteristic (ROC). Finally, the verified hub genes were analyzed by single-sample gene set enrichment analysis (ssGSEA) and NetworkAnalyst was used for the analysis of transcription factor (TF)-gene interactions, TF-microRNAs (miRNA) coregulatory network, and Protein-chemical Interactions. Results: A total of 120 DEGs between NASH and COVID-19 datasets were obtained, and the PPI network was constructed. Two key modules were obtained via the PPI network, and enrichment analysis of the key modules revealed the common association between NASH and COVID-19. In total, 16 hub genes were obtained by five algorithms, and six of them, namely, Kruppel-like factor 6 (KLF6), early growth response 1 (EGR1), growth arrest and DNA-damage-inducible 45 beta (GADD45B), JUNB, FOS, and FOS-like antigen 1 (FOSL1) were confirmed to be closely related to NASH and COVID-19. Finally, the relationship between hub genes and related pathways was analyzed, and the interaction network of six hub genes was constructed with TFs, miRNAs, and compounds. Conclusion: This study identified six hub genes related to COVID-19 and NASH, providing a new perspective for disease diagnosis and drug development.
Collapse
|
34
|
Henry Z, Meadows V, Guo GL. FXR and NASH: an avenue for tissue-specific regulation. Hepatol Commun 2023; 7:e0127. [PMID: 37058105 PMCID: PMC10109454 DOI: 10.1097/hc9.0000000000000127] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/23/2023] [Indexed: 04/15/2023] Open
Abstract
NASH is within the spectrum of NAFLD, a liver condition encompassing liver steatosis, inflammation, hepatocyte injury, and fibrosis. The prevalence of NASH-induced cirrhosis is rapidly rising and has become the leading indicator for liver transplantation in the US. There is no Food and Drug Administration (FDA)-approved pharmacological intervention for NASH. The farnesoid X receptor (FXR) is essential in regulating bile acid homeostasis, and dysregulation of bile acids has been implicated in the pathogenesis of NASH. As a result, modulators of FXR that show desirable effects in mitigating key characteristics of NASH have been developed as promising therapeutic approaches. However, global FXR activation causes adverse effects such as cholesterol homeostasis imbalance and pruritus. The development of targeted FXR modulation is necessary for ideal NASH therapeutics, but information regarding tissue-specific and cell-specific FXR functionality is limited. In this review, we highlight FXR activation in the regulation of bile acid homeostasis and NASH development, examine the current literature on tissue-specific regulation of nuclear receptors, and speculate on how FXR regulation will be beneficial in the treatment of NASH.
Collapse
Affiliation(s)
- Zakiyah Henry
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Vik Meadows
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, New Jersey, USA
- Department of Veterans Affairs New Jersey Health Care System, East Orange, New Jersey, USA
| |
Collapse
|
35
|
Buchynskyi M, Kamyshna I, Oksenych V, Zavidniuk N, Kamyshnyi A. The Intersection of COVID-19 and Metabolic-Associated Fatty Liver Disease: An Overview of the Current Evidence. Viruses 2023; 15:v15051072. [PMID: 37243158 DOI: 10.3390/v15051072] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The global population is currently experiencing the impact of the SARS-CoV-2 coronavirus, which has caused the Coronavirus Disease 2019 (COVID-19) pandemic. With our profound comprehension of COVID-19, encompassing the involvement sequence of the respiratory tract, gastrointestinal system, and cardiovascular apparatus, the multiorgan symptoms of this infectious disease have been discerned. Metabolic-associated fatty liver disease (MAFLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is a pervasive public health concern intricately linked with metabolic dysregulation and estimated to afflict one-fourth of the global adult population. The burgeoning focus on the association between COVID-19 and MAFLD is justified by the potential role of the latter as a risk factor for both SARS-CoV-2 infection and the subsequent emergence of severe COVID-19 symptoms. Investigations have suggested that changes in both innate and adaptive immune responses among MAFLD patients may play a role in determining the severity of COVID-19. The remarkable similarities observed in the cytokine pathways implicated in both diseases imply the existence of shared mechanisms governing the chronic inflammatory responses characterizing these conditions. The effect of MAFLD on the severity of COVID-19 illness remains uncertain, as indicated by conflicting results in cohort investigations.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Valentyn Oksenych
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7028 Trondheim, Norway
| | - Nataliia Zavidniuk
- Department of Infectious Diseases with Epidemiology, Dermatology and Venerology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| |
Collapse
|
36
|
Bertran L, Adalid L, Vilaró-Blay M, Barrientos-Riosalido A, Aguilar C, Martínez S, Sabench F, del Castillo D, Porras JA, Alibalic A, Richart C, Auguet T. Expression of STING in Women with Morbid Obesity and Nonalcoholic Fatty Liver Disease. Metabolites 2023; 13:metabo13040496. [PMID: 37110154 PMCID: PMC10146769 DOI: 10.3390/metabo13040496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic hepatic disease. Although mostly benign, this disease can evolve into nonalcoholic steatohepatitis (NASH). The stimulator of interferon genes (STING) plays an important role in the immune response against stressed cells, but this protein may also be involved in liver lipogenesis and microbiota composition. In this study, the role of STING in NAFLD was evaluated by RT–qPCR to analyze STING mRNA abundance and by immunohistochemical analysis to evaluate protein expression in liver biopsies from a cohort composed of 69 women with morbid obesity classified according to their liver involvement (normal liver, n = 27; simple steatosis (SS), n = 26; NASH, n = 16). The results showed that STING mRNA expression in the liver increases with the occurrence of NAFLD, specifically in the SS stage in which the degree of steatosis is mild or moderate. Protein analysis corroborated these results. Positive correlations were observed among hepatic STING mRNA abundance and gamma-glutamyl transferase and alkaline phosphatase levels, hepatic Toll-like receptor 9 expression and some circulating microbiota-derived bile acids. In conclusion, STING may be involved in the outcome and progression of NAFLD and may be related to hepatic lipid metabolism. However, further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Laia Bertran
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Laia Adalid
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona, Mallafré Guasch, 4, 43007 Tarragona, Spain
| | - Mercè Vilaró-Blay
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Andrea Barrientos-Riosalido
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Salomé Martínez
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona, Mallafré Guasch, 4, 43007 Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia i Anestèsia, Hospital Sant Joan de Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204 Reus, Spain
| | - Daniel del Castillo
- Servei de Cirurgia i Anestèsia, Hospital Sant Joan de Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204 Reus, Spain
| | - José Antonio Porras
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
| | - Ajla Alibalic
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
- Correspondence: ; Tel.: +34-977-29-58-33
| |
Collapse
|
37
|
Caballano-Infantes E, Ho-Plágaro A, López-Gómez C, Martín-Reyes F, Rodríguez-Pacheco F, Taminiau B, Daube G, Garrido-Sánchez L, Alcaín-Martínez G, Andrade RJ, García-Cortés M, Lucena MI, García-Fuentes E, Rodríguez-Díaz C. Membrane Vesicles of Toxigenic Clostridioides difficile Affect the Metabolism of Liver HepG2 Cells. Antioxidants (Basel) 2023; 12:antiox12040818. [PMID: 37107193 PMCID: PMC10135135 DOI: 10.3390/antiox12040818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Clostridioides difficile infection (CDI) appears to be associated with different liver diseases. C. difficile secretes membrane vesicles (MVs), which may be involved in the development of nonalcoholic fatty liver disease (NALFD) and drug-induced liver injury (DILI). In this study, we investigated the presence of C. difficile-derived MVs in patients with and without CDI, and analyzed their effects on pathways related to NAFLD and DILI in HepG2 cells. Fecal extracellular vesicles from CDI patients showed an increase of Clostridioides MVs. C. difficile-derived MVs that were internalized by HepG2 cells. Toxigenic C. difficile-derived MVs decreased mitochondrial membrane potential and increased intracellular ROS compared to non-toxigenic C. difficile-derived MVs. In addition, toxigenic C. difficile-derived MVs upregulated the expression of genes related to mitochondrial fission (FIS1 and DRP1), antioxidant status (GPX1), apoptosis (CASP3), glycolysis (HK2, PDK1, LDHA and PKM2) and β-oxidation (CPT1A), as well as anti- and pro-inflammatory genes (IL-6 and IL-10). However, non-toxigenic C. difficile-derived MVs did not produce changes in the expression of these genes, except for CPT1A, which was also increased. In conclusion, the metabolic and mitochondrial changes produced by MVs obtained from toxigenic C. difficile present in CDI feces are common pathophysiological features observed in the NAFLD spectrum and DILI.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Regeneration and Cell Therapy Andalusian, Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Junta de Andalucía, 41092 Seville, Spain
| | - Ailec Ho-Plágaro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Carlos López-Gómez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Flores Martín-Reyes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Francisca Rodríguez-Pacheco
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Bernard Taminiau
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Georges Daube
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Lourdes Garrido-Sánchez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Guillermo Alcaín-Martínez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Raúl J. Andrade
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miren García-Cortés
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - M. Isabel Lucena
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Departamento de Farmacología, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
- UICEC IBIMA, Plataforma SCReN (Spanish Clinical Research Network), Servicio de Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, 29010 Málaga, Spain
| | - Eduardo García-Fuentes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence:
| | - Cristina Rodríguez-Díaz
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, IBIMA Plataforma BIONAND, 29010 Málaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| |
Collapse
|
38
|
Ji J, Wu L, Wei J, Wu J, Guo C. The Gut Microbiome and Ferroptosis in MAFLD. J Clin Transl Hepatol 2023; 11:174-187. [PMID: 36406312 PMCID: PMC9647110 DOI: 10.14218/jcth.2022.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 12/04/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a new disease definition, and is proposed to replace the previous name, nonalcoholic fatty liver disease (NAFLD). Globally, MAFLD/NAFLD is the most common liver disease, with an incidence rate ranging from 6% to 35% in adult populations. The pathogenesis of MAFLD/NAFLD is closely related to insulin resistance (IR), and the genetic susceptibility to acquired metabolic stress-associated liver injury. Similarly, the gut microbiota in MAFLD/NAFLD is being revaluated by scientists, as the gut and liver influence each other via the gut-liver axis. Ferroptosis is a novel form of programmed cell death caused by iron-dependent lipid peroxidation. Emerging evidence suggests that ferroptosis has a key role in the pathological progression of MAFLD/NAFLD, and inhibition of ferroptosis may become a novel therapeutic strategy for the treatment of NAFLD. This review focuses on the main mechanisms behind the promotion of MAFLD/NAFLD occurrence and development by the intestinal microbiota and ferroptosis. It outlines new strategies to target the intestinal microbiota and ferroptosis to facilitate future MAFLD/NAFLD therapies.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liwei Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jue Wei
- Department of Gastroenterology Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University, Shanghai, China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence to: Chuanyong Guo, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, NO. 301, Middle Yanchang Road, Jing’an District, Shanghai 200072, China. ORCID: https://orcid.org/0000-0002-6527-4673. E-mail: ; Jianye Wu: Department of Gastroenterology, Putuo People’s Hospital, NO. 1291, Jiangning road, Putuo, Shanghai 200060, China. ORCID: https://orcid.org/0000-0003-2675-4241. E-mail:
| |
Collapse
|
39
|
Borel P, Dangles O, Kopec RE. Fat-soluble vitamin and phytochemical metabolites: Production, gastrointestinal absorption, and health effects. Prog Lipid Res 2023; 90:101220. [PMID: 36657621 DOI: 10.1016/j.plipres.2023.101220] [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: 06/07/2022] [Revised: 12/12/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Consumption of diets rich in fruits and vegetables, which provide some fat-soluble vitamins and many phytochemicals, is associated with a lower risk of developing certain degenerative diseases. It is well accepted that not only the parent compounds, but also their derivatives formed upon enzymatic or nonenzymatic transformations, can produce protective biological effects. These derivatives can be formed during food storage, processing, or cooking. They can also be formed in the lumen of the upper digestive tract during digestion, or via metabolism by microbiota in the colon. This review compiles the known metabolites of fat-soluble vitamins and fat-soluble phytochemicals (FSV and FSP) that have been identified in food and in the human digestive tract, or could potentially be present based on the known reactivity of the parent compounds in normal or pathological conditions, or following surgical interventions of the digestive tract or consumption of xenobiotics known to impair lipid absorption. It also covers the very limited data available on the bioavailability (absorption, intestinal mucosa metabolism) and summarizes their effects on health. Notably, despite great interest in identifying bioactive derivatives of FSV and FSP, studying their absorption, and probing their putative health effects, much research remains to be conducted to understand and capitalize on the potential of these molecules to preserve health.
Collapse
Affiliation(s)
- Patrick Borel
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France.
| | | | - Rachel E Kopec
- Human Nutrition Program, Department of Human Sciences, Foods for Health Discovery Theme, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
40
|
Paine A, Brookes PS, Bhattacharya S, Li D, De La Luz Garcia-Hernandez M, Tausk F, Ritchlin C. Dysregulation of Bile Acids, Lipids, and Nucleotides in Psoriatic Arthritis Revealed by Unbiased Profiling of Serum Metabolites. Arthritis Rheumatol 2023; 75:53-63. [PMID: 35818333 PMCID: PMC9797425 DOI: 10.1002/art.42288] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/29/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The transition from psoriasis to psoriatic arthritis (PsA) occurs in 20-30% of patients; however, the mechanisms underlying the emergence of musculoskeletal disease are not well understood. Metabolic disease is prevalent in psoriasis patients, but whether metabolic factors, other than obesity, increase arthritis risk in psoriasis patients is not known. This study was undertaken to investigate the link between metabolic changes and disease progression in psoriasis patients. METHODS To characterize the metabolic alterations during the progression of arthritis in psoriasis patients, we analyzed cross-sectional healthy controls and PsA samples and longitudinal psoriasis serum samples, before and after PsA onset. Nontargeted metabolomic profiling was performed using liquid chromatography mass spectrometry. RESULTS We identified several serum metabolites that differed between PsA patients, psoriasis patients, and healthy controls. Differentially abundant bile acids, purines, pyrimidines, glutathione, lipids, and amino acid metabolites were noted in these 3 groups. We also noted differences between psoriasis patients who progressed and those who did not progress to PsA. Bile acid and butyrate levels were depressed in those who progressed to PsA compared to those who did not, and the level of inflammatory lipid mediators increased following PsA diagnosis. In particular, the combination of leukotriene B4 and glycoursodeoxycholic acid sulfate were sensitive and specific predictors of PsA progression. CONCLUSION We observed notable differences in bile acid, purine, lipid, and amino acid-derived metabolites, among the healthy controls, psoriasis patients, and PsA patients and identified changes during the transition from psoriasis to PsA. The decreased bile acid and butyrate levels and elevated guanine levels in psoriasis patients at risk for PsA were particularly striking and may reflect gut microbial dysbiosis and dysregulated hepatic metabolism, leading to altered proliferation of immune cells and enhanced cytokine expression.
Collapse
Affiliation(s)
- Ananta Paine
- Division of Allergy, Immunology and Rheumatology, Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Paul S. Brookes
- Department of Anesthesiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Maria De La Luz Garcia-Hernandez
- Division of Allergy, Immunology and Rheumatology, Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Francisco Tausk
- Division of Allergy, Immunology and Rheumatology, Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Christopher Ritchlin
- Division of Allergy, Immunology and Rheumatology, Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| |
Collapse
|
41
|
Yang S, Yu D, Liu J, Qiao Y, Gu S, Yang R, Chai X, Wang W. Global publication trends and research hotspots of the gut-liver axis in NAFLD: A bibliometric analysis. Front Endocrinol (Lausanne) 2023; 14:1121540. [PMID: 36967792 PMCID: PMC10034112 DOI: 10.3389/fendo.2023.1121540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Nonalcoholic Fatty Liver Disease(NAFLD)refers to a spectrum of diseases ranging from simple liver steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. Bidirectional cross-talk between the gut-liver axis plays an important role in the pathogenesis of NAFLD. To learn more about the gut-liver axis in NAFLD, this study aims to provide a comprehensive analysis from a bibliometric perspective. METHOD Literature related to the gut-liver axis in NAFLD from 1989 to 2022 was extracted from the Web of Science Core Collection. Based on Microsoft Excel, CiteSpace and Vosviewer, we conducted to analyze the number of publications, countries/regions, institutions, authors, journals, references, and keywords. RESULTS A total of 1,891 literature since 2004 was included, with the rapid growth of the number of papers on the gut-liver axis in NAFLD annually. These publications were mainly from 66 countries and 442 institutions. Of the 638 authors analyzed, Bernd Schnabl was the one with the most publications, and Patrice D. Cani was the one with the most co-citations. International Journal of Molecular Sciences is the journal with the most articles published, and Hepatology is the journal with the most citations. The most common keywords are gut microbiota, inflammation, and insulin instance, which are current research hotspots. Short-chain fatty acid, in vitro, randomized controlled trial in clinical, and diabetes mellitus represent the research frontiers in this field and are in a stage of rapid development. CONCLUSION This is the first study to conduct a comprehensive bibliometric analysis of publications related to the gut-liver axis in NAFLD. This study reveals that gut microbiota, inflammation, insulin resistance, short-chain fatty acids, and randomized controlled trial will be the hotspots and new trends in the gut-liver axis in NAFLD research, which could provide researchers with key research information in this field and is helpful for further exploration of new research directions.
Collapse
Affiliation(s)
- Shuangjie Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Deshuai Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junjie Liu
- Department of Cardiology, Nanjing Pukou Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yanfang Qiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuxiao Gu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Ran Yang, ; Xinlou Chai, ; Wei Wang,
| | - Xinlou Chai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Ran Yang, ; Xinlou Chai, ; Wei Wang,
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Ran Yang, ; Xinlou Chai, ; Wei Wang,
| |
Collapse
|
42
|
Hassan HM, Onabote O, Isovic M, Passos DT, Dick FA, Torchia J. Regulation of Chromatin Accessibility by the Farnesoid X Receptor Is Essential for Circadian and Bile Acid Homeostasis In Vivo. Cancers (Basel) 2022; 14:cancers14246191. [PMID: 36551676 PMCID: PMC9777377 DOI: 10.3390/cancers14246191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The Farnesoid X Receptor (FXR) belongs to the nuclear receptor superfamily and is an essential bile acid (BA) receptor that regulates the expression of genes involved in the metabolism of BAs. FXR protects the liver from BA overload, which is a major etiology of hepatocellular carcinoma. Herein, we investigated the changes in gene expression and chromatin accessibility in hepatocytes by performing RNA-seq in combination with the Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) using a novel FXR knockout mouse model (Fxrex5Δ: Nr1h4ex5Δ/ex5Δ) generated through CRISPR/Cas9. Consistent with previous Fxr knockout models, we found that Fxrex5Δ mice develop late-onset HCC associated with increased serum and hepatic BAs. FXR deletion was associated with a dramatic loss of chromatin accessibility, primarily at promoter-associated transcription factor binding sites. Importantly, several genes involved in BA biosynthesis and circadian rhythm were downregulated following loss of FXR, also displayed reduced chromatin accessibility at their promoter regions. Altogether, these findings suggest that FXR helps to maintain a transcriptionally active state by regulating chromatin accessibility through its binding and recruitment of transcription factors and coactivators.
Collapse
Affiliation(s)
- Haider M. Hassan
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Oladapo Onabote
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Majdina Isovic
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Daniel T. Passos
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5C1, Canada
| | - Frederick A. Dick
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5C1, Canada
| | - Joseph Torchia
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Correspondence: ; Tel.: +519-685-8692
| |
Collapse
|
43
|
Ma S, Pang X, Tian S, Sun J, Hu Q, Li X, Lu Y. The protective effects of sulforaphane on high-fat diet-induced metabolic associated fatty liver disease in mice via mediating the FXR/LXRα pathway. Food Funct 2022; 13:12966-12982. [PMID: 36448414 DOI: 10.1039/d2fo02341e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is becoming the key factor in causing chronic liver disease all over the world. Sulforaphane (SFN) has been proven to be effective in alleviating many metabolic diseases, such as obesity and type 2 diabetes. In this study, C57BL/6 mice were fed a high-fat diet for 12 weeks to induce MAFLD and given SFN (10 mg per kg bw) daily. Our results showed that SFN not only improved the excessive accumulation of fat in the liver cells but also ameliorated liver and serum inflammatory and antioxidant levels. In addition, SFN can regulate bile-acid metabolism and fatty-acid synthesis by affecting their farnesoid X receptor (FXR)/liver X receptor alpha (LXRα) signaling pathway, ultimately alleviating MAFLD. Our study provides a theoretical basis for the mechanism by which SFN alleviates hepatic steatosis.
Collapse
Affiliation(s)
- Shaotong Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Qiaobin Hu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| |
Collapse
|
44
|
Keles U, Ow JR, Kuentzel KB, Zhao LN, Kaldis P. Liver-derived metabolites as signaling molecules in fatty liver disease. Cell Mol Life Sci 2022; 80:4. [PMID: 36477411 PMCID: PMC9729146 DOI: 10.1007/s00018-022-04658-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
Excessive fat accumulation in the liver has become a major health threat worldwide. Unresolved fat deposition in the liver can go undetected until it develops into fatty liver disease, followed by steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Lipid deposition in the liver is governed by complex communication, primarily between metabolic organs. This can be mediated by hormones, organokines, and also, as has been more recently discovered, metabolites. Although how metabolites from peripheral organs affect the liver is well documented, the effect of metabolic players released from the liver during the development of fatty liver disease or associated comorbidities needs further attention. Here we focus on interorgan crosstalk based on metabolites released from the liver and how these molecules act as signaling molecules in peripheral tissues. Due to the liver's specific role, we are covering lipid and bile mechanism-derived metabolites. We also discuss the high sucrose intake associated with uric acid release from the liver. Excessive fat deposition in the liver during fatty liver disease development reflects disrupted metabolic processes. As a response, the liver secretes a variety of signaling molecules as well as metabolites which act as a footprint of the metabolic disruption. In the coming years, the reciprocal exchange of metabolites between the liver and other metabolic organs will gain further importance and will help to better understand the development of fatty liver disease and associated diseases.
Collapse
Affiliation(s)
- Umur Keles
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Katharina Barbara Kuentzel
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Li Na Zhao
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Philipp Kaldis
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden. .,Lund University Diabetes Centre (LUDC), Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden.
| |
Collapse
|
45
|
Rivera-Andrade A, Álvarez CS. The importance of bile Acids in NAFLD: current evidence and future directions. Ann Hepatol 2022; 27:100773. [PMID: 36328687 DOI: 10.1016/j.aohep.2022.100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 02/04/2023]
Affiliation(s)
- Alvaro Rivera-Andrade
- Institute of Nutrition of Central America and Panama (INCAP) Research Center for the Prevention of Chronic Diseases, Institute of Nutrition of Central America and Panama, Guatemala City, Guatemala.
| | | |
Collapse
|
46
|
Şenol Y, Kaplan O, Varan C, Demirtürk N, Öncül S, Fidan BB, Ercan A, Bilensoy E, Çelebier M. Pharmacometabolomic assessment of vitamin E loaded human serum albumin nanoparticles on HepG2 cancer cell lines. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Research Progress on the Therapeutic Effect of Polysaccharides on Non-Alcoholic Fatty Liver Disease through the Regulation of the Gut–Liver Axis. Int J Mol Sci 2022; 23:ijms231911710. [PMID: 36233011 PMCID: PMC9570256 DOI: 10.3390/ijms231911710] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease affecting global public health at present, which can induce cirrhosis and liver cancer in serious cases. However, NAFLD is a multifactorial disease, and there is still a lack of research on its mechanism and therapeutic strategy. With the development of the gut–liver axis theory, the association between the gut–liver axis and the pathogenesis of NAFLD has been gradually disclosed. Polysaccharides, as a kind of natural product, have the advantages of low toxicity, multi-target and multi-pathway action. It has been reported that polysaccharides can affect the gut–liver axis at multiple interrelated levels, such as maintaining the ecological balance of gut microbiota (GM), regulating the metabolites of GM and improving the intestinal barrier function, which thereby plays a protective role in NAFLD. These studies have great scientific significance in understanding NAFLD based on the gut–liver axis and developing safe and effective medical treatments. Herein, we reviewed the recent progress of polysaccharides in improving nonalcoholic fatty liver disease (NAFLD) through the gut–liver axis.
Collapse
|
48
|
Takei H, Narushima S, Suzuki M, Kakiyama G, Sasaki T, Murai T, Yamashiro Y, Nittono H. Characterization of long-chain fatty acid-linked bile acids: a major conjugation form of 3β-hydroxy bile acids in feces. J Lipid Res 2022; 63:100275. [PMID: 36089004 PMCID: PMC9587409 DOI: 10.1016/j.jlr.2022.100275] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Although most bile acids (BAs) in feces are present in noncovalent forms that can be extracted with ethanol, non-negligible amounts of saponifiable BAs are also present. It is a major concern that such saponifiable BAs are routinely omitted from fecal BA measurements. We compared the BA profiles of healthy stools that were obtained with/without alkaline hydrolysis and found that as much as 29.7% (2.1–67.7%) of total BAs were saponifiable. Specifically, alkaline treatment led to significant elevations of isodeoxycholic acid (isoDCA) and isolithocholic acid (isoLCA) concentrations, suggesting that considerable proportions of isoDCA and isoLCA were esterified. Precursor ion scan data from LC/MS suggested the presence of long-chain FA-linked BAs. We chemically synthesized a series of fatty acid 3β-acyl conjugates of isoDCA and isoLCA as analytical standards and analyzed their fecal profiles from newborns to adults (n = 64) by LC/MS. FA-conjugated isobile acids (FA-isoBAs) were constantly present from 2 years of age to adulthood. C16- and C18-chain FA-isoBA esters were predominantly found regardless of age, but small amounts of acetic acid esters were also found. FA-isoBA concentrations were not correlated to fecal FA concentrations. Interestingly, there were some adults who did not have FA-isoBAs. Gut bacteria involved in the production of FA-isoBAs have not been identified yet. The present study provides insight into the establishment of early gut microbiota and the interactive development of esterified BAs.The contribution of FA-isoBAs to gut physiology and their role in pathophysiologic conditions such as inflammatory bowel disease are currently under investigation.
Collapse
Affiliation(s)
- Hajime Takei
- Junshin Clinic Bile Acid Institute, 2-1-24 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Seiko Narushima
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Genta Kakiyama
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, 1201 Broad Rock Blvd, Richmond, VA 23249, USA
| | - Takahiro Sasaki
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan
| | - Tsuyoshi Murai
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Juntendo University Graduate School of Medicine, 3rd floor, Hongo-Asakaze Bldg., 2-9-8 Hongo Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroshi Nittono
- Junshin Clinic Bile Acid Institute, 2-1-24 Haramachi, Meguro-ku, Tokyo 152-0011, Japan
| |
Collapse
|
49
|
Li X, Zhao W, Xiao M, Yu L, Chen Q, Hu X, Zhao Y, Xiong L, Chen X, Wang X, Ba Y, Guo Q, Wu X. Penthorum chinense Pursh. extract attenuates non-alcholic fatty liver disease by regulating gut microbiota and bile acid metabolism in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115333. [PMID: 35500802 DOI: 10.1016/j.jep.2022.115333] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Penthorum chinense Pursh. (PCP) is commonly used as a Miao ethnomedicine and health food for liver protection in China. Gansukeli (WS3-B-2526-97) is made from the extract of PCP (PCPE) for the treatment of viral hepatitis. In recent years, PCPE has been reported in the treatment of non-alcoholic fatty liver disease (NAFLD), however its potential mechanism is not fully elucidated. AIM OF THE STUDY To investigate the ameliorating effect of PCPE on high-fat diet (HFD)-induced NAFLD mice and demonstrate whether its protective effect is gut microbiota dependent and associated with bile acid (BA) metabolism. MATERIALS AND METHODS The alleviating effect of PCPE on NAFLD was conducted on male C57BL/6J mice fed an HFD for 16 weeks, and this effect associated with gut microbiota dependent was demonstrated by pseudo-germfree mice treated with antibiotics and fecal microbiota transplantation (FMT). The composition of the gut microbiota in the cecum contents was analyzed by 16S rRNA sequencing, and the levels of BAs in liver and fecal samples were determined by UPLC/MS-MS. RESULTS The results showed that administration of PCPE for 8 weeks could potently ameliorate HFD-induced NAFLD and alleviate dyslipidemia and insulin resistance. Moreover, PCPE treatment alleviated gut dysbiosis, especially reducing the relative abundance of bile salt hydrolase (BSH)-producing bacteria. Furthermore, PCPE significantly increased the levels of taurine-conjugated BAs in feces, such as tauro-β-muricholic acid (T-βMCA), tauroursodesoxycholic acid (TUDCA), and taurochenodeoxycholic acid (TCDCA), and increased hepatic chenodeoxycholic acid (CDCA). The protein and mRNA expression of farnesoid X receptor (FXR) and fibroblast growth factor 15 (FGF15) were decreased in intestine, increased taurine-conjugated BAs inhibited the intestinal signaling pathway, which was associated with increased genes expression of enzymes in the alternative BA synthesis pathway that reduced the levels of cholesterol. The increased CDCA produced via the alternative BA synthesis pathway promoted hepatic FXR activation and BA excretion. CONCLUSION Our study is the first time to demonstrate that PCPE could ameliorate NAFLD in HFD-induced mice by regulating the gut microbiota and BA metabolism, and from a novel perspective, to clarify the mechanism of PCPE in NAFLD.
Collapse
Affiliation(s)
- Xiaoxi Li
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Wenwen Zhao
- Department of Pharmacy, Beijing Children's hospital, Capital Medical University, National Center for Children Health, Beijing, 100045, China
| | - Meng Xiao
- National Institutes for Food and Drug Control, Beijing, 102629, China
| | - Lan Yu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Qijun Chen
- School of Pharmaceutical Sciences, Capital Medical University, 100069, Beijing, China
| | - Xiaolu Hu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Yimeng Zhao
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Lijuan Xiong
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Xiaoqing Chen
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Xing Wang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Yinying Ba
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Qiang Guo
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China
| | - Xia Wu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
50
|
Role of bile acids and their receptors in gastrointestinal and hepatic pathophysiology. Nat Rev Gastroenterol Hepatol 2022; 19:432-450. [PMID: 35165436 DOI: 10.1038/s41575-021-00566-7] [Citation(s) in RCA: 150] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2021] [Indexed: 02/06/2023]
Abstract
Bile acids (BAs) can regulate their own metabolism and transport as well as other key aspects of metabolic homeostasis via dedicated (nuclear and G protein-coupled) receptors. Disrupted BA transport and homeostasis results in the development of cholestatic disorders and contributes to a wide range of liver diseases, including nonalcoholic fatty liver disease and hepatocellular and cholangiocellular carcinoma. Furthermore, impaired BA homeostasis can also affect the intestine, contributing to the pathogenesis of irritable bowel syndrome, inflammatory bowel disease, and colorectal and oesophageal cancer. Here, we provide a summary of the role of BAs and their disrupted homeostasis in the development of gastrointestinal and hepatic disorders and present novel insights on how targeting BA pathways might contribute to novel treatment strategies for these disorders.
Collapse
|