1
|
Wang D, Miao J, Zhang L, Zhang L. Research advances in the diagnosis and treatment of MASLD/MASH. Ann Med 2025; 57. [DOI: 10.1080/07853890.2024.2445780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Dekai Wang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinxian Miao
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihua Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
He Y, Ye M, Xia Y, Zhong Z, Wang W, Li Q. The role of cytokines as predictors for NAFLD-related diseases: A bidirectional Mendelian randomization study. Clin Res Hepatol Gastroenterol 2025; 49:102545. [PMID: 39900199 DOI: 10.1016/j.clinre.2025.102545] [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: 11/28/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Prior research has highlighted associations between inflammatory cytokines and non-alcoholic fatty liver disease (NAFLD), but causal relationships remain unclear. Employing the Mendelian randomization (MR) approach, this investigation aims to explore the connection between 41 inflammatory cytokines and NAFLD-related diseases. METHODS Our research implemented bidirectional study focusing on 41 cytokines in 8,293 Finns, predicting genetic associations with NAFLD, nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. We primarily utilized the inverse variance weighted (IVW) method to evaluate the bidirectional relationships. Additionally, a sensitivity analysis was carried out to ensure the reliability of our findings. RESULTS An elevated risk for NAFLD was correlated with both IL-2 (OR = 1.226, 95 % CI = 1.018-1.477, p = 0.031) and TNF-β (OR = 1.151, 95 % CI = 1.011-1.310, p = 0.033). IL-16 is associated with decreased NAFLD risk (OR = 0.820, 95 % CI = 0.719-0.934, p = 0.033). β-NGF (OR = 2.495, 95 % CI = 1.019-6.108, p = 0.045) and SCGFβ (OR = 1.541, 95 % CI = 1.052-2.256, p = 0.026) are linked to higher NASH risk. No significant associations were found for fibrosis and cirrhosis. Furthermore, the causal relationship between genetic predisposition to NAFLD-related diseases and various inflammatory cytokines was established. CONCLUSIONS Our MR analysis identifies specific cytokines as genetic predictors for NAFLD and NASH. IL-2 and TNF-β increase NAFLD risk, IL-16 appears protective, and β-NGF and SCGFβ are associated with greater NASH risk. These insights are crucial for understanding the etiology and treatment of NAFLD-related diseases.
Collapse
Affiliation(s)
- Yijia He
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Miaomin Ye
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yin Xia
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyi Zhong
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weiping Wang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Coyne ES, Nie Y, Lee D, Pandovski S, Yang T, Zhou H, Rosahl TW, Carballo-Jane E, Abdurrachim D, Zhou Y, Hendra C, Ali AAB, Meyers S, Blumenschein W, Gongol B, Liu Y, Zhou Y, Talukdar S. Loss of mitochondrial amidoxime-reducing component 1 (mARC1) prevents disease progression by reducing fibrosis in multiple mouse models of chronic liver disease. Hepatol Commun 2025; 9:e0637. [PMID: 39927988 PMCID: PMC11809980 DOI: 10.1097/hc9.0000000000000637] [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: 09/05/2024] [Accepted: 12/02/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease is a prevalent disease that affects nearly one-third of the global population. Recent genome-wide association studies revealed that a common missense variant in the gene encoding mitochondrial amidoxime reducing component 1 (mARC1) is associated with protection from metabolic dysfunction-associated steatotic liver disease, all-cause cirrhosis, and liver-related mortality suggesting a role for mARC1 in liver pathophysiology; however, little is known about its function in the liver. In this study, we aimed to evaluate the impact of mARC1 hepatoprotective variants on protein function, the effect of loss of mARC1 on cellular lipotoxic stress response, and the effect of global or hepatocyte-specific loss of mARC1 in various mouse models of metabolic dysfunction-associated steatohepatitis and liver fibrosis. METHODS AND RESULTS Expression and characterization of mARC1 hepatoprotective variants in cells and mouse liver revealed that the mARC1 p.A165T exhibited lower protein levels but maintained its mitochondrial localization. In cells, the knockdown of mARC1 improved cellular bioenergetics and decreased mitochondrial superoxide production in response to lipotoxic stress. Global genetic deletion and hepatocyte-specific knockdown of mARC1 in mice significantly reduced liver steatosis and fibrosis in multiple mouse models of metabolic dysfunction-associated steatohepatitis and liver fibrosis. Furthermore, RNA-seq analysis revealed that the pathways involved in extracellular matrix remodeling and collagen formation were downregulated in the liver, and the plasma lipidome was significantly altered in response to the loss of mARC1 in mice. CONCLUSIONS Overall, we have demonstrated that loss of mARC1 alters hepatocyte response to lipotoxic stress and protects mice from diet-induced MASH and liver fibrosis consistent with findings from human genetics.
Collapse
Affiliation(s)
- Erin S. Coyne
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Yilin Nie
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Darwin Lee
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Sentibel Pandovski
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Tiffany Yang
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Heather Zhou
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Thomas W. Rosahl
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Ester Carballo-Jane
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Desiree Abdurrachim
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Yongqi Zhou
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Christopher Hendra
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Asad Abu Bakar Ali
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Stacey Meyers
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Wendy Blumenschein
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Brendan Gongol
- Department of Data, AI & Genome Sciences, MSD, Singapore
| | - Yang Liu
- Department of Data, AI & Genome Sciences, MSD, Singapore
| | - Yingjiang Zhou
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Saswata Talukdar
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| |
Collapse
|
4
|
Pirola CJ, Fernández Gianotti T, Sookoian S. The Proteomics of MASLD Progression: Insights From Functional Analysis to Drive the Development of New Therapeutic Solutions. Aliment Pharmacol Ther 2025; 61:614-627. [PMID: 39744897 DOI: 10.1111/apt.18468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/22/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading chronic liver disease worldwide, with alarming prevalence reaching epidemic proportions. AIMS AND METHODS The objective of this study is to provide a comprehensive review of the latest blood proteomics studies on MASLD and metabolic dysfunction-associated steatohepatitis (MASH), with emphasis on fibrosis. Furthermore, our objective is to conduct an analysis of protein pathways and interactions by integrating proteomics data using functional enrichment analysis of the deregulated proteins. RESULTS Notwithstanding the considerable discrepancies in the methodology and the number of proteins examined in the circulation, the analysis reveals a consistent pattern among the list of proteins that are decreased or increased in the blood of the affected patients. The relevant biological processes (BP) associated with down- and upregulated proteins are high-density lipoprotein remodelling and complement activation, respectively. The protein families identified include not only those expected to be involved in the immune system and cell adhesion and migration but also ligands of glycoproteins expressed in cells that have been subjected to stress and proteins containing the Sushi domain. CONCLUSIONS The application of cutting-edge methodologies to investigate the blood proteome in MASH is yielding insights that facilitate the elucidation of disease mechanisms and the identification of optimal noninvasive biomarkers. However, several challenges remain to be addressed in future research, including the generalisation of results on a global scale, the optimisation of analytical technologies and the implementation of large longitudinal studies to gain insights into the molecular mechanisms that underpin the development of advanced disease.
Collapse
Affiliation(s)
- Carlos José Pirola
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| | - Tomas Fernández Gianotti
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| | - Silvia Sookoian
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Research in Health Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
5
|
Wong SW, Yang YY, Chen H, Xie L, Shen XZ, Zhang NP, Wu J. New advances in novel pharmacotherapeutic candidates for the treatment of metabolic dysfunction-associated steatohepatitis (MASH) between 2022 and 2024. Acta Pharmacol Sin 2025:10.1038/s41401-024-01466-7. [PMID: 39870846 DOI: 10.1038/s41401-024-01466-7] [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: 09/14/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) covers a broad spectrum of profile from simple fatty liver, evolving to metabolic dysfunction-associated steatohepatitis (MASH), to hepatic fibrosis, further progressing to cirrhosis and hepatocellular carcinoma (HCC). MASLD has become a prevalent disease with 25% in average over the world. MASH is an active stage, and requires pharmacological intervention when there is necroptotic damage with fibrotic progression. Although there is an increased understanding of MASH pathogenesis and newly approved resmetirom, given its complexity and heterogeneous pathophysiology, there is a strong necessity to develop more drug candidates with better therapeutic efficacy and well-tolerated safety profile. With an increased list of pharmaceutical candidates in the pipeline, it is anticipated to witness successful approval of more potential candidates in this fast-evolving field, thereby offering different categories of medications for selective patient populations. In this review, we update the advances in MASH pharmacotherapeutics that have completed phase II or III clinical trials with potential application in clinical practice during the latest 2 years, focusing on effectiveness and safety issues. The overview of fast-evolving status of pharmacotherapeutic candidates for MASH treatment confers deep insights into the key issues, such as molecular targets, endpoint selection and validation, clinical trial design and execution, interaction with drug administration authority, real-world data feedback and further adjustment in clinical application.
Collapse
Affiliation(s)
- Shu Wei Wong
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Yong-Yu Yang
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Hui Chen
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Li Xie
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Xi-Zhong Shen
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Ning-Ping Zhang
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| | - Jian Wu
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, 200032, China.
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| |
Collapse
|
6
|
Nuri A, Jeilu S, Teklu Y, Abdu K, Muhye A, Getachew MS, Dagne I, Yigezu M, Adugna SA, Nambiar VS, Oumer A. Magnitude, patterns, and factors associated with liver disease among clinically suspected clients in Eastern Ethiopia: hidden public health tragedy. BMC Gastroenterol 2025; 25:36. [PMID: 39856553 PMCID: PMC11760694 DOI: 10.1186/s12876-025-03628-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Liver diseases are a public health problem in developing regions, and the majority of them are asymptomatic. Evidence on the current burden of liver disease and associated context-specific risk factors is scarce in the study area, where various risk factors for liver disease, including khat chewing, smoking, and aflatoxin exposure, are common. This study was to assess the magnitude, patterns, and factors associated with liver disease among patients visiting governmental hospitals in eastern Ethiopia. METHODS A facility-based cross-sectional study was conducted among randomly selected 388 clinically suspected adults for liver disease visiting a public hospital in Dire Dawa, Ethiopia. Data was collected using a combination of structured questionnaires, physical examinations, and investigations through interviews, medical chart reviews, liver enzymes, and the results of ultrasound examinations. Bivariable and multivariable logistic regression analyses were employed to determine the factors associated with liver disease. Candidate variables in bivariable analysis were selected based on p-values < 0.25, biological plausibility, and evidence of associations from previous studies. The level of significance was considered at a P-value < 0.05, and the adjusted odds ratio with a 95% confidence interval was reported. RESULTS A total of 388 (92%) adults were included and liver disease was diagnosed among 295 (76%; 71.5-80.2) of the study participants, where chronic liver disease due to non-viral (65.1%) and viral causes (34.9%) were prevalent. The risk of liver disease was higher among females (AOR = 3.43; 1.80-6.55) and under grade twelve (AOR = 3.90; 1.95-7.82) compared to counterparts. Furthermore, alcohol consumption (AOR = 3.14; 1.33-7.32), khat chewing (AOR = 1.69; 0.91-3.15), a history of hospitalization (AOR = 4.20; 2.29-7.11), and a history of intestinal parasite infection (AOR = 1.17; 0.55-2.49) were found to be positively associated with an increased risk of liver disease among adults. CONCLUSIONS Liver disease detection rate was found to be high and could be associated with substance use (alcohol and khat), history of hospitalization, and worm infection, which can be incorporated into the prevention strategies of liver diseases.
Collapse
Affiliation(s)
- Aliya Nuri
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Sufian Jeilu
- Department of Internal Medicine, Sabian General Hospital, Dire Dawa, Ethiopia
| | - Yared Teklu
- School of Medicine, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Kadir Abdu
- Department of Nursing, College of Health Sciences, Oda Bultum University, Chiro, Ethiopia
| | - Ahmed Muhye
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Milkiyas Solomon Getachew
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Imam Dagne
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Muluken Yigezu
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Sewmehon Amsalu Adugna
- Department of Midwifery, College of Health Sciences, Oda Bultum University, Chiro, Ethiopia
| | - Vanisha S Nambiar
- Department of Foods and Nutrition, Faculty of Family and Community Sciences, the Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Abdu Oumer
- Department of Public Health, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia.
| |
Collapse
|
7
|
Shi L, Li Z, Ma X, Wang J, Wu Y, Zhu Y, Wang Y, Yang Y, Luo M, Li J, Sun X, He S. Effects of ultra-processed foods on the liver: insights from gut microbiome and metabolomics studies in rats. Front Nutr 2025; 11:1503879. [PMID: 39912061 PMCID: PMC11794082 DOI: 10.3389/fnut.2024.1503879] [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: 09/29/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose High consumption of Ultra-processed foods (UPF) have been identified as a potential risk factor for Non-alcoholic fatty liver disease (NAFLD). Nevertheless, there is limited empirical evidence regarding the impact of UPF, which are typical combination of processed foods, on liver health through alterations in gut microbiota and metabolic processes. We aim to examine the potential impact of UPF on liver health and to explore the role of gut microbiota and metabolites. Methods This study used Sprague-Dawley rats to mimic modern UPF diets for 90 days. Some serum biochemical indices, inflammatory factors, oxidative stress markers, hematoxylin-eosin (HE) staining of the liver, 16S ribosomal RNA (rRNA) and Liquid chromatography-mass spectrometry (LC-MS) of rat feces were detected. Results The UPF diet-induced simple steatosis of the liver in rats without affecting the levels of IL-6, GSH, MDA, and SOD. Additionally, it modified the gut microbiota, increasing potentially harmful bacteria, such as norank_f__Desulfovibrionaceae and Staphylococcus, while also elevating the relative abundance of potentially beneficial bacteria, including Dubosiella and Allobaculum. Furthermore, the consumption of UPF led to a metabolomic disorder characterized by disruptions in the sphingolipid signaling pathway, sulfur relay system, and arachidonic acid metabolism. Conclusion In conclusion, the findings of this study indicate that the consumption of UPF influences the development of simple hepatic steatosis, potentially through alterations in gut microbiota and metabolomics.
Collapse
Affiliation(s)
- Liping Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Zhuoyuan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Xiaojun Ma
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Junru Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yueping Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yongbin Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yanrong Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yue Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Minxiu Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Xian Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, China
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
8
|
Liu Y, Zhou F, Zhao H, Song J, Song M, Zhu J, Wang Y, Man Hoi MP, Lin L, Zhang Q. Dimeric guaianolide sesquiterpenoids from the flowers of Chrysanthemum indicum ameliorate hepatic steatosis through mitigating SIRT1-mediated lipid accumulation and ferroptosis. J Adv Res 2025:S2090-1232(24)00625-8. [PMID: 39788286 DOI: 10.1016/j.jare.2024.12.047] [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: 08/09/2024] [Revised: 10/04/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) acts as the primary contributor to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and potentially hepatocellular carcinoma. The flowers of Chrysanthemum indicum, a traditional edible medicinal herb, have been widely used in China for more than 2000 years. However, the function of C. indicum in managing NAFLD has seldom been investigated. OBJECTIVES To reveal the novel active components and underlying mechanisms of C. indicum in treating NAFLD. METHODS An MS/MS-based molecular networking-guided strategy was used for the chemical investigation. The structure identification of the new compounds involved high resolution electrospray ionization mass spectrometry (HRESIMS), 1D and 2D nuclear magnetic resonance (NMR) spectra, electronic circular dichroism (ECD), and X-ray crystallographic analysis. The biological evaluation was performed using Nile Red staining, flow cytometry, commercial kits, western blotting, co-immunoprecipitation, isothermal titration calorimetry, cellular thermal shift assay, drug affinity responsive target stability assay, molecular docking, and confocal immunofluorescence. RESULTS A total of 27 new dimeric sesquiterpenoids, chryindicolides A-Z (1-26) and chrysanthemolide C (27), together with seven known compounds, were isolated from the flowers of C. indicum under the guide of MS/MS-based molecular networking. Among them, compounds 1-7 were rare chlorine-containing guaianolide dimers. Chryindicolide O (15) directly bound and activated the deacetylase Sirtuin 1 (SIRT1) to reduce de novo lipogenesis, enhance fatty acid β-oxidation, and inhibit ferroptosis in palmitic acid and oleic acid (P/O)-induced AML12 hepatocytes. In addition, chryindicolide O significantly ameliorated liver steatosis in high-fat diet-fed zebrafish. CONCLUSION Novel guaianolide dimers from C. indicum alleviated hepatic steatosis through mitigating SIRT1-mediated lipid accumulation and ferroptosis, suggesting that they could be further developed as candidates against NAFLD.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Fei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Haoyu Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Jianguo Song
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Min Song
- Guangdong Provincial Engineering Research Center for Modernization of TCM, Jinan University, Guangzhou 510632, China
| | - Jianzhong Zhu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Ying Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China.
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China.
| |
Collapse
|
9
|
Ramasamy J. Diagnostic Utility of Serum Ferritin in Identifying Liver Fibrosis in Patients with Non-Alcoholic Fatty Liver Disease (NAFLD): A Cross-Sectional Study Using National Health and Nutrition Examination Survey (NHANES-2017-2020) Data. Indian J Clin Biochem 2025; 40:80-88. [PMID: 39835224 PMCID: PMC11741978 DOI: 10.1007/s12291-023-01159-8] [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: 09/21/2023] [Accepted: 10/01/2023] [Indexed: 01/22/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the major causes of chronic liver disease worldwide. There are conflicting reports on the association of serum ferritin levels and its utility in discriminating various stages of liver fibrosis in patients with NAFLD. This study is done to address the conflicts by analysing the National Health and Nutritional Examination Survey 2017-2020 (NHANES 2017-2020) data. The survey data was screened, and the eligible participants (n = 5901) were categorized based on the controlled attenuation parameter (CAP) obtained from Vibration Controlled and Transient Elastography (VCTE) scan into 2 groups, NAFLD (CAP ≥ 274 dB/m, n = 2528) and non-NAFLD (CAP < 274 dB/m, n = 3373). The NAFLD group had a significantly higher proportion of obese, diabetic, and metabolic syndrome participants. Serum ferritin levels were significantly higher in the NAFLD group, and the levels showed an upward trend from simple steatosis to significant fibrosis. Multivariate regression analysis showed that ferritin has a significant positive association with the median liver stiffness parameter (LSM), which is the index of liver fibrosis. Receiver operator characteristic curve (ROC) analysis showed that serum ferritin levels are a poor predictor of fibrosis (LSM > 8 kPA) in those with NAFLD (AUC = 0.59, Sensitivity = 53.5%, Specificity = 60.3%). To conclude, serum ferritin levels were positively associated and it trended upward as the disease progressed from simple steatosis to fibrosis in NAFLD. However, its diagnostic utility in discriminating liver fibrosis in patients with NAFLD is limited. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-023-01159-8.
Collapse
Affiliation(s)
- Jagadish Ramasamy
- Department of Biochemistry, Velammal Medical College Hospital and Research Institute, Madurai, Tamil Nadu 632002 India
| |
Collapse
|
10
|
Cui X, Li H, Li L, Xie C, Gao J, Chen Y, Zhang H, Hao W, Fu J, Guo H. Rodent model of metabolic dysfunction-associated fatty liver disease: a systematic review. J Gastroenterol Hepatol 2025; 40:48-66. [PMID: 39322221 PMCID: PMC11771679 DOI: 10.1111/jgh.16749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Although significant progress has been made in developing preclinical models for metabolic dysfunction-associated steatotic liver disease (MASLD), few have encapsulated the essential biological and clinical outcome elements reflective of the human condition. We conducted a comprehensive literature review of English-language original research articles published from 1990 to 2023, sourced from PubMed, Embase, and Web of Science, aiming to collate studies that provided a comparative analysis of physiological, metabolic, and hepatic histological characteristics between MASLD models and control groups. The establishment of a robust metabolic dysfunction-associated steatotic liver rodent model hinges on various factors, including animal species and strains, sex, induction agents and methodologies, and the duration of induction. Through this review, we aim to guide researchers in selecting suitable induction methods and animal species for constructing preclinical models aligned with their specific research objectives and laboratory conditions. Future studies should strive to develop simple, reliable, and reproducible models, considering the model's sensitivity to factors such as light-dark cycles, housing conditions, and environmental temperature. Additionally, the potential of diverse in vitro models, including 3D models and liver organ technology, warrants further exploration as valuable tools for unraveling the cellular mechanisms underlying fatty liver disease.
Collapse
Affiliation(s)
- Xiao‐Shan Cui
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hong‐Zheng Li
- Guang'an men HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Liang Li
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Cheng‐Zhi Xie
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Jia‐Ming Gao
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuan‐Yuan Chen
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hui‐Yu Zhang
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Wei Hao
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Jian‐Hua Fu
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hao Guo
- Safety Laboratory, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
11
|
Pirola CJ, Landa MS, Schuman M, García SI, Salatino A, Sookoian S. Metabolic dysfunction-associated steatotic liver disease exhibits sex-specific microbial heterogeneity within intestinal compartments. Clin Mol Hepatol 2025; 31:179-195. [PMID: 39391907 PMCID: PMC11791572 DOI: 10.3350/cmh.2024.0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/28/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND/AIMS Evidence suggests that the gastrointestinal microbiome plays a significant role in the biology of metabolic dysfunction-associated steatotic liver disease (MASLD). However, it remains unclear whether disparities in the gut microbiome across intestinal tissular compartments between the sexes lead to MASLD pathogenesis. METHODS Sex-specific analyses of microbiome composition in two anatomically distinct regions of the gut, the small intestine and colon, were performed using an experimental model of MASLD. The study involved male and female spontaneously hypertensive rats and the Wistar-Kyoto control rat strain, which were fed either a standard chow diet or a high-fat diet for 12 weeks to induce MASLD (12 rats per group). High-throughput 16S sequencing was used for microbiome analysis. RESULTS There were significant differences in the overall microbiome composition of male and female rats with MASLD, including variations in topographical gut regions. The beta diversity of the jejunal and colon microbiomes was higher in female rats than in male rats (PERMANOVA p-value=0.001). Sex-specific analysis and discriminant features using LEfSe showed considerable variation in bacterial abundance, along with distinct functional properties, in the jejunum and colon of animals with MASLD. Significantly elevated levels of lipopolysaccharide and protein expression of Toll-like receptor 4 were observed in the livers of male rats with MASLD compared with their female counterparts. CONCLUSION This study uncovered sexual dimorphism in the gut microbiome of MASLD and identified microbial heterogeneity within intestinal compartments. Insights into sex-specific variations in gut microbiome composition could facilitate customised treatment strategies.
Collapse
Affiliation(s)
- Carlos Jose Pirola
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES). Maimónides University, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Faculty of Health Science, Maimónides University, Buenos Aires, Argentina
| | - Maria Silvina Landa
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
| | - Mariano Schuman
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
| | - Silvia Inés García
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | - Adrian Salatino
- Max Planck Institute for Immunobiology and Epigenetics, Bioinformatics Facility, Frieburg, Germany
| | - Silvia Sookoian
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Faculty of Health Science, Maimónides University, Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Research in Health Center (CENITRES). Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
12
|
Eskes ECB, van Dussen L, Brands MMMG, Vaz FM, Aerts JMFG, van Kuilenburg ABP, Sjouke B, Hollak CEM. Natural disease course of chronic visceral acid sphingomyelinase deficiency in adults: A first step toward treatment criteria. J Inherit Metab Dis 2025; 48:e12789. [PMID: 39177062 PMCID: PMC11670446 DOI: 10.1002/jimd.12789] [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: 05/10/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Acid sphingomyelinase deficiency (ASMD) is an ultra-rare lysosomal storage disease with a broad spectrum of manifestations ranging from severe neuropathic forms to attenuated, chronic visceral forms. Manifestations of the chronic visceral subtype are variable and encompass different degrees of hepatosplenomegaly, pulmonary disease and dyslipidemia. The aim of this study was to provide insights into the natural course of adult patients with the chronic visceral subtype. Based on these insights, we proposed tentative criteria for initiation and follow-up of enzyme replacement therapy (ERT). The data of 23 adult patients were collected in a prospective study. Clinical, genetic and demographic data, plasma measurements, abdominal imaging, pulmonary imaging, pulmonary function tests and quality of life questionnaires were collected. Stability of disease based on several clinical, biochemical and radiological markers (i.e., spleen volume, platelet levels, liver volume, alanine aminotransferase [ALT] levels, diffusion capacity of the lungs for carbon monoxide [DLCO] chitotriosidase activity and lysosphingomyelin [LSM]) was assessed. Cardiovascular risk was estimated based on sex, age, smoking, systolic blood pressure and lipid profile. Quality of life was evaluated with the 36-Item Short Form Health Survey and the Health Assessment Questionnaire. Median follow-up was 6.1 years (range 1.3-19.5 years). The most common manifestations were splenomegaly (100%), decreased high-density lipoprotein cholesterol (HDL-C) plasma levels (83%), (signs of) steatosis measured with transient elastography (82%), thrombocytopenia (64%), hepatomegaly (52%) and decreased diffusion capacity (45%). The majority of markers remained stable during follow-up. Twelve patients showed progression of disease: four for spleen volume, two for liver volume, three for DLCO, seven for chitotriosidase activity and three for LSM. One patient showed progression of disease based on four markers, although this patient did not report any problems at the last visit. Cardiovascular risk was estimated and was increased in half of the patients older than 40 years. Patient-reported quality of life did not differ from the general population, but differences in median 36-Item Short Form Health Survey (SF-36) scores of patients with severe pulmonary involvement and those of patients without pulmonary involvement were observed. Tentative criteria for initiation and effect of therapy were proposed. In conclusion, the chronic visceral subtype of ASMD showed a predominantly stable disease course in this cohort. We propose that ERT should be initiated on an individual basis and only in case of progression or symptomatic disease. Collection and analysis of real world data are necessary to refine start, stop and follow-up criteria in the future.
Collapse
Affiliation(s)
- Eline C. B. Eskes
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of MetabolismAmsterdamThe Netherlands
| | - Laura van Dussen
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of MetabolismAmsterdamThe Netherlands
| | - Marion M. M. G. Brands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of MetabolismAmsterdamThe Netherlands
- Department of Pediatric Metabolic DiseasesAmsterdam UMC, University of Amsterdam, Emma Children's HospitalAmsterdamThe Netherlands
| | - Frédéric M. Vaz
- Laboratory Genetic Metabolic DiseasesAmsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Johannes M. F. G. Aerts
- Department of Medical BiochemistryLeiden Institute of Chemistry, University of LeidenLeidenThe Netherlands
| | - André B. P. van Kuilenburg
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of MetabolismAmsterdamThe Netherlands
- Laboratory Genetic Metabolic DiseasesAmsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Barbara Sjouke
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Department of Internal MedicineRadboud UMCNijmegenNetherlands
| | - Carla E. M. Hollak
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of MetabolismAmsterdamThe Netherlands
| |
Collapse
|
13
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
14
|
Tak J, Kim YS, Kim SG. Roles of X-box binding protein 1 in liver pathogenesis. Clin Mol Hepatol 2025; 31:1-31. [PMID: 39355873 PMCID: PMC11791611 DOI: 10.3350/cmh.2024.0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
The prevalence of drug-induced liver injury (DILI) and viral liver infections presents significant challenges in modern healthcare and contributes to considerable morbidity and mortality worldwide. Concurrently, metabolic dysfunctionassociated steatotic liver disease (MASLD) has emerged as a major public health concern, reflecting the increasing rates of obesity and leading to more severe complications such as fibrosis and hepatocellular carcinoma. X-box binding protein 1 (XBP1) is a distinct transcription factor with a basic-region leucine zipper structure, whose activity is regulated by alternative splicing in response to disruptions in endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR) activation. XBP1 interacts with a key signaling component of the highly conserved UPR and is critical in determining cell fate when responding to ER stress in liver diseases. This review aims to elucidate the emerging roles and molecular mechanisms of XBP1 in liver pathogenesis, focusing on its involvement in DILI, viral liver infections, MASLD, fibrosis/cirrhosis, and liver cancer. Understanding the multifaceted functions of XBP1 in these liver diseases offers insights into potential therapeutic strategies to restore ER homeostasis and mitigate liver damage.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
15
|
Lam R, Jain D, Deng Y, Acharya E, Lim JK. Advanced Liver Fibrosis Predicts Liver Outcomes in Biopsy-proven Metabolic Dysfunction-associated Steatotic Liver Disease: A U.S.-based Single-center Retrospective Cohort Study. J Clin Transl Hepatol 2024; 12:988-996. [PMID: 39649030 PMCID: PMC11622200 DOI: 10.14218/jcth.2024.00189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 12/10/2024] Open
Abstract
Background and Aims Data regarding risk factors and long-term outcomes of U.S. patients with biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD) are limited. This study aimed to investigate the role of clinical and histologic risk factors on long-term outcomes in patients with MASLD. Methods A retrospective cohort study of 451 adults with biopsy-proven MASLD was conducted at a U.S. academic hospital from 2012 to 2020. An experienced pathologist evaluated the index liver biopsy. Patients with a prior liver transplant or alternative etiologies of chronic liver disease were excluded. The duration of the risk exposure was determined from the date of the index liver biopsy to an outcome event or the last follow-up examination. Outcome events of interest included incident liver-related events, liver decompensation, and all-cause mortality. Results In the final cohort of 406 patients followed for a median of 3.7 years (interquartile range: 4.8 years), 35 patients died, 41 developed hepatic decompensation, and 70 experienced a liver-related event. Among histologic risk factors, stage 3 (adjusted Hazard ratio (aHR) 2.68, 95% confidence interval (CI) 1.18-6.11) and stage 4 (aHR 6.96, 95% CI 3.55-13.64) fibrosis were associated with incident liver-related events compared to stage 0-1 fibrosis. Stage 4 (aHR 8.46, 95% CI 3.26-21.99) fibrosis alone was associated with incident liver decompensation events compared to stage 0-1 fibrosis. Among clinical risk factors, hypertension (aHR 2.58, 95% CI 1.05-6.34) was associated with incident liver decompensation. Conclusions In a U.S. single-center cohort of patients with biopsy-proven MASLD, advanced fibrosis was the primary risk factor for incident liver decompensation and liver-related events.
Collapse
Affiliation(s)
- Robert Lam
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Dhanpat Jain
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yanhong Deng
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | | | - Joseph K. Lim
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Jiang H, Wang H, Guo Y, Zhu Y, Dai H, Liang C, Gao J. Characterization of the hepatic flora and metabolome in nonalcoholic fatty liver disease. Front Microbiol 2024; 15:1528258. [PMID: 39760085 PMCID: PMC11697427 DOI: 10.3389/fmicb.2024.1528258] [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: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background/aim The purpose of this study was to examine the hepatic bacterial composition and metabolome characteristics of patients with NAFLD using 16S rDNA sequencing and metabolomics. The results of the study revealed substantial differences in hepatic bacterial composition and metabolites between the NAFLD group and the control group. These differences were used to identify potential biomarkers that could be employed to diagnose NAFLD. Subjects/methods Liver tissues from 13 patients in the NAFLD group and 12 patients in the control group were collected for microbiota examination. Results The bacterial DNA profiles of the liver were significantly different between NAFLD patients and controls. NAFLD patients exhibited an enrichment of Enterobacterales, Mycobacteriales, Pseudomonadales, Flavobacteriales and Xanthomonadales, Sphingomonadales, Lysobact, which was characterised by a lack of erales. At the genus level, the abundance values of Escherichia-Shigella, Rhodococcus, and Chryseobacterium in the NAFLD group were significantly elevated, while the abundance values of Stenotrophomonas, Lawsonella and Sphingobium were significantly reduced. A total of 402 distinct metabolites were identified between the two groups, with 78 metabolites that were up-regulated and 14 metabolites that were down-regulated. The enrichment of metabolic pathways indicated that linoleic acid metabolism was the most significant contributor to the metabolic differences, and lipid metabolism was substantially differentiated. The hepatic metabolite levels were substantially correlated with the changes in hepatic microflora, as demonstrated by the correlation analysis. Conclusion Differences in pathogenesis and host physiological function of NAFLD may be attributed to the hepatic flora and metabolomic characteristics. In the future, this presents new opportunities for the investigation of prospective diagnostic and therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Hui Wang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yangfan Guo
- Central Laboratory of Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yankun Zhu
- Department of Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Hui Dai
- Department of Oncology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Chenchen Liang
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Jianpeng Gao
- Department of Gastroenterology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| |
Collapse
|
17
|
Bucci T, Nabrdalik K, Baratta F, Pastori D, Pignatelli P, Hydes T, Alam U, Violi F, Lip GYH. Risk of Adverse Events in Anticoagulated Patients With Atrial Fibrillation and Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2024; 110:208-217. [PMID: 38864452 PMCID: PMC11651694 DOI: 10.1210/clinem/dgae394] [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: 03/12/2024] [Revised: 05/15/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND The clinical impact of nonalcoholic fatty liver disease (NAFLD) in patients with atrial fibrillation (AF) is still controversial. AIM To evaluate the 1-year risk of all-cause death, thromboembolic events, and bleeding in patients with AF-NAFLD. METHODS Retrospective study with a health research network (TriNetX). Patients with AF on oral anticoagulation (OAC) were categorized according to the presence of NAFLD into 2 groups. The primary outcomes were the 1-year risks of (1) a composite cardiovascular outcome (all-cause death, myocardial infarction, stroke, cardiac arrest, and pulmonary embolism) and (2) a composite hemorrhagic outcome (intracranial hemorrhage and gastrointestinal bleeding). Cox regression analysis before and after propensity score matching was used to estimate hazard ratio (HR) and 95% 95% CI,. Sensitivity analyses investigated the risk associated with cirrhosis, thrombocytopenia, and type of OAC (warfarin vs non-vitamin K antagonist oral anticoagulants (NOACs). RESULTS We identified 22 636 patients with AF-NAFLD (69 ± 12 years, 46.7% females) and 391 014 patients with AF and without liver disease (72 ± 12 years, 42.7% females). NAFLD was associated with a higher risk of composite cardiovascular (HR, 1.54; 95% CI, 1.47-1.61) and hemorrhagic (HR, 1.56; 95% CI, 1.42-1.72) outcomes. This was consistent also for all the single outcomes. Cirrhotic and thrombocytopenic patients with AF-NAFLD showed the highest risks. Compared to patients with AF-NAFLD on NOACs, those on warfarin were associated with a higher risk of cardiovascular and hemorrhagic outcomes. CONCLUSION In patients with AF, NAFLD is associated with a higher 1-year risk of adverse events, with the risk of adverse events progressively increasing from noncirrhotic to cirrhotic and from nonthrombocytopenic to thrombocytopenic patients. NOACs were associated with a better effectiveness and safety profile compared to warfarin.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Katarzyna Nabrdalik
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice 40-055, Poland
| | - Francesco Baratta
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Theresa Hydes
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Uazman Alam
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg 9000, Denmark
| |
Collapse
|
18
|
Gunawan S, Soetikno V, Purwaningsih EH, Ferdinal F, Wuyung PE, Ramadhani D. 6-Gingerol, a Bioactive Compound of Zingiber officinale, Ameliorates High-Fat High-Fructose Diet-Induced Non-Alcoholic Related Fatty Liver Disease in Rats. J Exp Pharmacol 2024; 16:455-466. [PMID: 39712345 PMCID: PMC11662909 DOI: 10.2147/jep.s492971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024] Open
Abstract
Purpose Endoplasmic reticulum (ER) stress has a prominent role in the pathogenesis of high-fat diet-induced non-alcohol related fatty liver disease (NAFLD). The aim of this study is to investigate the effects of 6-G on the reduction of ER stress-induced NAFLD in metabolic syndrome (MetS) rats. Methods Twenty-five male Sprague-Dawley rats were fed with a high-fat high-fructose (HFHF) diet for 16 weeks. The rats were treated orally with 6-G (50,100, and 200 mg/kgBW) once daily for eight weeks. At Week 16, all animals were sacrificed, and serum and liver tissue were harvested for biochemical and structural analysis. Results NAFLD liver rats were shown to have elevated protein expression of GRP78, and ER-associated apoptotic protein, such as IRE1, TRAF2, p-JNK, and p-NF-κB, which were considerably reduced by the 6-G at three doses treatment. Furthermore, a significant increase in liver apoptosis and non-alcoholic steatohepatitis (NAS) score were observed in the NAFLD rat liver and which were also attenuated by the 6-G treatment at three doses. 6-G treatment also reduced ALT, AST, and ALP serum levels. Conclusion Considering all the findings, it is suggested that the 6-G treatment could be a potential candidate therapy in treating ER stress-induced NAFLD in rats.
Collapse
Affiliation(s)
- Shirly Gunawan
- Department of Pharmacology, Faculty of Medicine, Universitas Tarumanagara, Jakarta, Indonesia
| | - Vivian Soetikno
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Frans Ferdinal
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Tarumanagara, Jakarta, Indonesia
| | - Puspita Eka Wuyung
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Animal Research Facility, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Dwi Ramadhani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Banten, Indonesia
| |
Collapse
|
19
|
Al Maawali A, Al Salmi I, Al Saadi T, Al Balushi H. The Hyperdense Right Hemidiaphragm Sign: A Novel Radiological Indicator of Diffuse Fatty Liver Infiltration on Non-Enhanced CT. Cureus 2024; 16:e75340. [PMID: 39781126 PMCID: PMC11710877 DOI: 10.7759/cureus.75340] [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] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
Objectives The primary objective of this study is to describe and evaluate the diagnostic performance of the hyperdense right hemidiaphragm sign (HRHS) as a novel radiological indicator for diffuse fatty infiltration of the liver on non-enhanced CT (NECT) scans. This includes assessing its sensitivity, specificity, positive predictive value, and negative predictive value, and comparing these metrics with other established NECT signs. Methods This cross-sectional multicenter retrospective study included all patients over 12 years of age who underwent both abdominal MRI and NECT scans of the abdomen within a period not exceeding six months at two tertiary hospitals (The Royal Hospital and Armed Forces Hospital, Muscat, Sultanate of Oman) between January 2010 and December 2022. Two readers reviewed the NECT scan images for the following signs: HRHS, liver density of less than 10 HU compared to the spleen, absolute liver density of <40 HU, and hyperattenuated intrahepatic vessels. The drop in signal on the out-phase sequence compared to the in-phase sequence on MRI was used as the gold standard reference for diagnosing diffuse fatty infiltration of the liver. Data were analyzed using IBM SPSS Statistics for Windows, Version 26.0 (Released 2019; IBM Corp., Armonk, NY, USA) and MedCalc software (MedCalc Software Ltd., Oostende, Belgium), with sensitivity, specificity, and positive and negative predictive values calculated for each NECT sign. Results A total of 340 patients were included in this study, with a mean age of 54 years. Among these patients, 45 were diagnosed with diffuse fatty infiltration of the liver by MRI. Thirty-six patients were identified as positive for fatty liver infiltration on unenhanced CT based on the criterion of the HRHS, with a sensitivity of 91%, specificity of 99%, positive predictive value of 94%, and negative predictive value of 99%. In contrast, 45 patients were positive using the criterion of liver density less than 10 HU compared to the spleen, yielding a sensitivity of 83%, a specificity of 98%, a positive predictive value of 88%, and a negative predictive value of 97%. Using the criterion of absolute liver density <40 HU, 30 patients were positive, with a sensitivity of 72%, specificity of 98%, positive predictive value of 90%, and negative predictive value of 96%. Lastly, 20 patients were positive using the criterion of hyperattenuated intrahepatic vessels, with a sensitivity of 51%, specificity of 100%, positive predictive value of 100%, and negative predictive value of 93%. Conclusions The proposed qualitative evaluation using the HRHS for diffuse fatty infiltration of the liver on NECT scans demonstrates the highest sensitivity compared to other previously described NECT signs.
Collapse
|
20
|
Ziqubu K, Dludla PV, Mthembu SX, Nkambule B, Mazibuko-Mbeje SE. Low circulating levels of neuregulin 4 as a potential biomarker associated with the severity and prognosis of obesity-related metabolic diseases: a systematic review. Adipocyte 2024; 13:2390833. [PMID: 39162358 PMCID: PMC11340757 DOI: 10.1080/21623945.2024.2390833] [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: 04/30/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Neuregulin 4 (Nrg4) is a brown adipose tissue-derived adipokine that greatly affects systemic metabolism and improves metabolic derangements. Although abnormal circulating levels of Nrg4 are common in obesity, it remains elusive whether low or elevated levels of this batokine are associated with the onset of metabolic diseases. AIM To assess Nrg4 levels and its role as a feasible biomarker to predict the severity of obesity, gestational diabetes mellitus (GDM), type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVD). METHODS A search for relevant studies was performed systematically using prominent search engines, including PubMed, Google Scholar, and Embase, by following PRISMA guidelines. RESULTS Ample clinical evidence reported low serum/plasma levels of Nrg4 in obesity and these were inversely proportional to the indices of metabolic syndrome, including body mass index, waist circumference, triglycerides, fasting plasma glucose, and homoeostatic model assessment for insulin resistance as well as high-sensitivity C-reactive protein. Low circulating Nrg4 levels may aid in the prediction of morbid obesity, and subsequent GDM, T2DM, NAFLD, and CVD. CONCLUSION Current clinical evidence emphasizes that the circulating levels of Nrg4 are decreased in morbid obesity, and it also highlights that Nrg4 May serve as a potential prognostic biomarker for obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho, South Africa
| | - Phiwayinkosi V. Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | | | - Bongani Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | |
Collapse
|
21
|
Heyens L, Kenjic H, Dagnelie P, Schalkwijk C, Stehouwer C, Meex S, Kooman J, Bekers O, van Greevenbroek M, Savelberg H, Robaeys G, de Galan B, Koster A, van Dongen M, Eussen S, Koek G. Forns index and fatty liver index, but not FIB-4, are associated with indices of glycaemia, pre-diabetes and type 2 diabetes: analysis of The Maastricht Study. BMJ Open Gastroenterol 2024; 11:e001466. [PMID: 39615896 PMCID: PMC11624825 DOI: 10.1136/bmjgast-2024-001466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/07/2024] [Indexed: 12/09/2024] Open
Abstract
OBJECTIVE Glucose metabolism status (GMS) is linked to non-alcoholic fatty liver disease (NAFLD). Higher levels of advanced glycation end products (AGEs) are observed in people with type 2 diabetes mellitus (T2DM) and NAFLD. We examined the association between GMS, non-invasive tests and AGEs, with liver steatosis and fibrosis. METHODS Data from The Maastricht Study, a population-based cohort, were analysed. Participants with alcohol overconsumption or missing data were excluded. GMS was determined via an oral glucose tolerance test. AGEs, measured by skin autofluorescence (SAF), were assessed using an AGE Reader. Associations of GMS and SAF with the fibrosis-4 score (FIB-4), Forns index (FI) and fatty liver index (FLI) were investigated using multivariable linear regression, adjusted for sociodemographic, lifestyle and clinical variables. RESULTS 1955 participants (56.6%) were analysed: 598 (30.6%) had T2DM, 264 (13.5%) had pre-diabetes and 1069 (54.7%) had normal glucose metabolism. Pre-diabetes was significantly associated with FLI (standardised regression coefficient (Stβ) 0.396, 95% CI 0.323 to 0.471) and FI (Stβ 0.145, 95% CI 0.059 to 0.232) but not FIB-4. T2DM was significantly associated with FLI (Stβ 0.623, 95% CI 0.552 to 0.694) and FI (Stβ 0.307, 95% CI 0.226 to 0.388) but not FIB-4. SAF was significantly associated with FLI (Stβ 0.083, 95% CI 0.036 to 0.129), FI (Stβ 0.106, 95% CI 0.069 to 0.143) and FIB-4 (Stβ 0.087, 95% CI 0.037 to 0.137). CONCLUSION The study showed that adverse GMS and higher glycaemia are positively associated with steatosis. FI, but not FIB-4, was related to adverse GMS concerning fibrosis. This study is the first to demonstrate that SAF is positively associated with steatosis and fibrosis.
Collapse
Affiliation(s)
- Leen Heyens
- Hasselt University Faculty of Medicine and Life Sciences, Diepenbeek, Belgium
- Maastricht University Faculty of Health Medicine and Life Sciences, Maastricht, The Netherlands
| | - Hanna Kenjic
- School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Pieter Dagnelie
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Maastricht University Medical Centre+ Internal Medicine, Maastricht, The Netherlands
| | - Casper Schalkwijk
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Maastricht University Medical Centre+ Internal Medicine, Maastricht, The Netherlands
| | - Coen Stehouwer
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Steven Meex
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Department Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jeroen Kooman
- School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
- Maastricht University Medical Centre+ Internal Medicine, Maastricht, The Netherlands
| | - Otto Bekers
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Department Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marleen van Greevenbroek
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Maastricht University Medical Centre+ Internal Medicine, Maastricht, The Netherlands
| | - Hans Savelberg
- School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Geert Robaeys
- Hasselt University Faculty of Medicine and Life Sciences, Diepenbeek, Belgium
| | - Bastiaan de Galan
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
| | - Annemarie Koster
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Martien van Dongen
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Simone Eussen
- CARIM Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
| | - Ger Koek
- School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
- Maastricht University Medical Centre+ Internal Medicine, Maastricht, The Netherlands
| |
Collapse
|
22
|
Wang J, Li Q, Huo Y, Liu X, Shi Y, Xie B. ASPP2 deficiency promotes the progression of metabolic dysfunction-associated steatohepatitis via ACSL4 upregulation. Sci Rep 2024; 14:29177. [PMID: 39587161 PMCID: PMC11589572 DOI: 10.1038/s41598-024-80415-0] [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: 07/24/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
As a member of the p53-binding protein family, apoptosis-stimulating protein p53 2 (ASPP2) is closely related to autophagy and apoptosis. However, the mechanistic role of ASPP2 in the development of metabolic dysfunction-associated steatohepatitis (MASH) remains elusive. Therefore, we investigated the role and underlying mechanisms of ASPP2 in MASH progression in a mouse model of MASH and a cellular model of metabolic dysfunction-associated fatty liver disease. ASPP2 deficiency significantly promoted the inflammatory response, steatosis, and MASH progression in mice. Through transcriptomic analysis, increased ACSL4 expression was identified as a potential key factor. Further elucidation of the underlying mechanisms demonstrated that ASPP2 deficiency increased lipid accumulation and inhibited mitochondrial respiration capacity in HepG2 cells induced by oleic acid. However, silencing of ACSL4 reversed these effects. Thus, our study indicates that ASPP2 is an important regulator of MASH progression through ACSL4 upregulation, highlighting its potential as an alternative approach to MASH treatment.
Collapse
Affiliation(s)
- Jinming Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Quanwei Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yunfei Huo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiaoni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Ying Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
23
|
Toma T, Miyakawa N, Arakaki Y, Watanabe T, Nakahara R, Ali TFS, Biswas T, Todaka M, Kondo T, Fujita M, Otsuka M, Araki E, Tateishi H. An antifibrotic compound that ameliorates hyperglycaemia and fat accumulation in cell and HFD mouse models. Diabetologia 2024; 67:2568-2584. [PMID: 39251430 DOI: 10.1007/s00125-024-06260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/03/2024] [Indexed: 09/11/2024]
Abstract
AIMS/HYPOTHESIS Appropriate management of blood glucose levels and the prevention of complications are important in the treatment of diabetes. We have previously reported on a compound named HPH-15 that is not only antifibrotic but also AMP-activated protein kinase (AMPK)-activating. In this study, we evaluated whether HPH-15 is useful as a therapeutic medication for diabetes. METHODS We examined the effects of HPH-15 on AMPK activation, glucose uptake, fat accumulation and lactic acid production in L6-GLUT4, HepG2 and 3T3-L1 cells, as a model of muscle, liver and fat tissue, respectively. Additionally, we investigated the glucose-lowering, fat-accumulation-suppressing, antifibrotic and AMPK-activating effect of HPH-15 in mice fed a high-fat diet (HFD). RESULTS HPH-15 at a concentration of 10 µmol/l increased AMPK activation, glucose uptake and membrane translocation of GLUT4 in each cell model to the same extent as metformin at 2 mmol/l. The production of lactic acid (which causes lactic acidosis) in HPH-15-treated cells was equal to or less than that observed in metformin-treated cells. In HFD-fed mice, HPH-15 lowered blood glucose from 11.1±0.3 mmol/l to 8.2±0.4 mmol/l (10 mg/kg) and 7.9±0.4 mmol/l (100 mg/kg) and improved insulin resistance. The HPH-15 (10 mg/kg) group showed the same level of AMPK activation as the metformin (300 mg/kg) group in all organs. The HPH-15-treated HFD-fed mice also showed suppression of fat accumulation and fibrosis in the liver and fat tissue; these effects were more significant than those obtained with metformin. Mice treated with high doses of HPH-15 also exhibited a 44% reduction in subcutaneous fat. CONCLUSIONS/INTERPRETATION HPH-15 activated AMPK at lower concentrations than metformin in vitro and in vivo and improved blood glucose levels and insulin resistance in vivo. In addition, HPH-15 was more effective than metformin at ameliorating fatty liver and adipocyte hypertrophy in HFD-fed mice. HPH-15 could be effective in preventing fatty liver, a common complication in diabetic individuals. Additionally, in contrast to metformin, high doses of HPH-15 reduced subcutaneous fat in HFD-fed mice. Presumably, HPH-15 has a stronger inhibitory effect on fat accumulation and fibrosis than metformin, accounting for the reduction of subcutaneous fat. Therefore, HPH-15 is potentially a glucose-lowering medication that can lower blood glucose, inhibit fat accumulation and ameliorate liver fibrosis.
Collapse
Affiliation(s)
- Tsugumasa Toma
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobukazu Miyakawa
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuiichi Arakaki
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takuro Watanabe
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ryosei Nakahara
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Taha F S Ali
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Tanima Biswas
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Tatsuya Kondo
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Research and Development, Science Farm Ltd, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
- Kikuchi Medical Association Hospital, Kumamoto, Japan.
- Research Center for Health and Sport Sciences, Kumamoto Health Science University, Kumamoto, Japan.
| | - Hiroshi Tateishi
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
- Research and Development Department, Research and Development Headquarters, Hirata Corporation, Kumamoto, Japan.
| |
Collapse
|
24
|
Taranto D, Kloosterman DJ, Akkari L. Macrophages and T cells in metabolic disorder-associated cancers. Nat Rev Cancer 2024; 24:744-767. [PMID: 39354070 DOI: 10.1038/s41568-024-00743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
Cancer and metabolic disorders have emerged as major global health challenges, reaching epidemic levels in recent decades. Often viewed as separate issues, metabolic disorders are shown by mounting evidence to heighten cancer risk and incidence. The intricacies underlying this connection are still being unraveled and encompass a complex interplay between metabolites, cancer cells and immune cells within the tumour microenvironment (TME). Here, we outline the interplay between metabolic and immune cell dysfunction in the context of three highly prevalent metabolic disorders, namely obesity; two associated liver diseases, metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH); and type 2 diabetes. We focus primarily on macrophages and T cells, the critical roles of which in dictating inflammatory response and immune surveillance in metabolic disorder-associated cancers are widely reported. Moreover, considering the ever-increasing number of patients prescribed with metabolism disorder-altering drugs and diets in recent years, we discuss how these therapies modulate systemic and local immune phenotypes, consequently impacting cancer malignancy. Collectively, unraveling the determinants of metabolic disorder-associated immune landscape and their role in fuelling cancer malignancy will provide a framework essential to therapeutically address these highly prevalent diseases.
Collapse
Affiliation(s)
- Daniel Taranto
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Fang B, Luo J, Cui Z, Liu R, Wang P, Zhang J. Pea Albumin Alleviates Oleic Acid-Induced Lipid Accumulation in LO2 Cells Through Modulating Lipid Metabolism and Fatty Acid Oxidation Pathways. Foods 2024; 13:3482. [PMID: 39517266 PMCID: PMC11545291 DOI: 10.3390/foods13213482] [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: 09/14/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Excessive lipid accumulation in the liver can cause NAFLD, leading to chronic liver injury. To relieve liver lipid accumulation by dietary proteins, this study used oleic acid (OA) induction to establish a stable in vitro LO2 cell lipid accumulation model. This model was used to explore the mechanism by which pea albumin (PA) regulates lipid levels in LO2 cells. PA has been shown to ameliorate OA-induced lipid accumulation in LO2 cells by reducing the aggregation of intracellular lipid droplets and lowering cell TG and TC levels. In addition, it can alleviate OA-induced LO2 cell damage and oxidative stress, reduce cellular ALT and AST secretion, lower cellular MDA levels, and increase GSH-Px viability. Regulation of lipid metabolism in LO2 cells involves inhibiting the cellular lipid synthesis pathway and activating the expression of proteins related to the triglyceride catabolic and fatty acid oxidation pathways. PA contributes to regulating lipid accumulation in LO2 cells. This study provides new insights into alleviating liver fat accumulation and a theoretical basis for exploring the mechanism of protein regulation of liver cell lipid metabolism.
Collapse
Affiliation(s)
- Bing Fang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (B.F.); (Z.C.); (R.L.); (P.W.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Jie Luo
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410114, China;
| | - Zhengwu Cui
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (B.F.); (Z.C.); (R.L.); (P.W.)
| | - Rong Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (B.F.); (Z.C.); (R.L.); (P.W.)
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (B.F.); (Z.C.); (R.L.); (P.W.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Jian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (B.F.); (Z.C.); (R.L.); (P.W.)
| |
Collapse
|
26
|
Li H, Ye J, Dong Y, Kong W, Qian G, Xie Y. U-shaped association of serum vitamin A concentrations with all-cause mortality in patients with NAFLD: results from the NHANES database prospective cohort study. Front Nutr 2024; 11:1467659. [PMID: 39539372 PMCID: PMC11558463 DOI: 10.3389/fnut.2024.1467659] [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: 07/20/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Background Previous studies have demonstrated a significant association between serum vitamin A concentration and non-alcoholic fatty liver disease (NAFLD) development. However, the long-term prognostic implications of serum vitamin A in patients with NAFLD remain underexplored. This study aims to investigate whether there exists a correlation between serum vitamin A concentrations and overall mortality among subjects diagnosed with NAFLD. Methods To investigate the association between serum vitamin A concentrations and NAFLD outcomes, we conducted prospective cohort studies using data from the 1999-2006 and 2017-2018 National Health and Nutrition Examination Survey (NHANES). We utilized a multivariate Cox regression model to explore the relationship between serum vitamin A levels and all-cause mortality. Survival curves related to serum vitamin A were constructed using the Kaplan-Meier method. Additionally, the restricted cubic splines (RCS) method was applied to examine potential nonlinear relationships between serum vitamin A concentrations and all-cause mortality of NAFLD. Results Over a median follow-up period of 10.3 years, a total of 1,399 all-cause deaths were recorded. The weighted average concentration of serum vitamin A was 61.48 ± 0.37 μg/dL. After adjusting for potential confounders, a significant U-shaped relationship was identified between serum vitamin A concentrations and the risk of all-cause mortality in NAFLD patients. This relationship was particularly pronounced in men and elderly individuals aged 60 to 85. Conclusion Our study reveals a significant non-linear relationship between serum vitamin A concentrations and the risk of all-cause mortality in patients with NAFLD. These findings underscore the importance of monitoring and maintaining optimal serum vitamin A levels to potentially improve survival outcomes in NAFLD patients.
Collapse
Affiliation(s)
- Hui Li
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Jiayuan Ye
- Department of Infectious Diseases, Shangyu People's Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Yitian Dong
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Weiliang Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guoqing Qian
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yilian Xie
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
27
|
Weiss U, Mungo E, Haß M, Benning D, Gurke R, Hahnefeld L, Dorochow E, Schlaudraff J, Schmid T, Kuntschar S, Meyer S, Medert R, Freichel M, Geisslinger G, Niederberger E. Knock-Out of IKKepsilon Ameliorates Atherosclerosis and Fatty Liver Disease by Alterations of Lipid Metabolism in the PCSK9 Model in Mice. Int J Mol Sci 2024; 25:10721. [PMID: 39409049 PMCID: PMC11476531 DOI: 10.3390/ijms251910721] [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] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
The inhibitor-kappaB kinase epsilon (IKKε) represents a non-canonical IκB kinase that modulates NF-κB activity and interferon I responses. Inhibition of this pathway has been linked with atherosclerosis and metabolic dysfunction-associated steatotic liver disease (MASLD), yet the results are contradictory. In this study, we employed a combined model of hepatic PCSK9D377Y overexpression and a high-fat diet for 16 weeks to induce atherosclerosis and liver steatosis. The development of atherosclerotic plaques, serum lipid concentrations, and lipid metabolism in the liver and adipose tissue were compared between wild-type and IKKε knock-out mice. The formation and progression of plaques were markedly reduced in IKKε knockout mice, accompanied by reduced serum cholesterol levels, fat deposition, and macrophage infiltration within the plaque. Additionally, the development of a fatty liver was diminished in these mice, which may be attributed to decreased levels of multiple lipid species, particularly monounsaturated fatty acids, triglycerides, and ceramides in the serum. The modulation of several proteins within the liver and adipose tissue suggests that de novo lipogenesis and the inflammatory response are suppressed as a consequence of IKKε inhibition. In conclusion, our data suggest that the knockout of IKKε is involved in mechanisms of both atherosclerosis and MASLD. Inhibition of this pathway may therefore represent a novel approach to the treatment of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ulrike Weiss
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Eleonora Mungo
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Michelle Haß
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Denis Benning
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Robert Gurke
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Erika Dorochow
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Jessica Schlaudraff
- Goethe University Frankfurt, Faculty of Medicine, Institute of Neuroanatomy, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany;
| | - Tobias Schmid
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Silvia Kuntschar
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Sofie Meyer
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Rebekka Medert
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Marc Freichel
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Gerd Geisslinger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Ellen Niederberger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
28
|
Yang B, Tang G, Wang M, Ni Y, Tong J, Hu C, Zhou M, Jiao K, Li Z. Trimethylamine N-oxide induces non-alcoholic fatty liver disease by activating the PERK. Toxicol Lett 2024; 400:93-103. [PMID: 39153559 DOI: 10.1016/j.toxlet.2024.08.009] [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: 09/26/2023] [Revised: 06/17/2024] [Accepted: 08/15/2024] [Indexed: 08/19/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a liver disease causing different progressive pathological changes. Trimethylamine N-oxide (TMAO), a product of gut microbiota metabolism, is a specific agonist of the protein kinase R-like endoplasmic reticulum kinase (PERK) pathway, one of the endoplasmic reticulum stress (ERS) pathways. TMAO has been associated with the occurrence and development of NAFLD based on the results of previous studies, but whether the simple consumption of TMAO can directly induce NAFLD and its underlying mechanism remain unclear. To investigate this question, we constructed an animal model in which adult male zebrafish were fed a controlled diet containing 1 % or 3 % TMAO for 20 weeks. Eventually, we observed that TMAO caused lipid accumulation, inflammatory infiltration, liver injury and liver fibrosis in zebrafish livers; meanwhile, the PERK signaling pathway was activated in the zebrafish livers. This finding was further confirmed in HepG2 cells and hepatic stellate cells models. In conclusion, this study found that TMAO directly induced different pathological states of NAFLD in zebrafish liver, and the activation of PERK pathway is an important mechanism, which may provide crucial strategies for the diagnosis and treatment of NAFLD.
Collapse
Affiliation(s)
- Bingmo Yang
- Suqian Center for Disease Control and Prevention, Suqian, Jiangsu 223800, China
| | - Guomin Tang
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mengting Wang
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yifan Ni
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jiali Tong
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chunyan Hu
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ming Zhou
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Kailin Jiao
- Department of Nutrition, The Second Affiliated Hospital, Air Force Medical University, Xi'an 710038, China.
| | - Zhong Li
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
29
|
Hwang J, Okada J, Liu L, Pessin JE, Schwartz GJ, Jo YH. The development of hepatic steatosis depends on the presence of liver-innervating parasympathetic cholinergic neurons in mice fed a high-fat diet. PLoS Biol 2024; 22:e3002865. [PMID: 39436946 PMCID: PMC11530026 DOI: 10.1371/journal.pbio.3002865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/01/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Hepatic lipid metabolism is regulated by the autonomic nervous system of the liver, with the sympathetic innervation being extensively studied, while the parasympathetic efferent innervation is less understood despite its potential importance. In this study, we investigate the consequences of disrupted brain-liver communication on hepatic lipid metabolism in mice exposed to obesogenic conditions. We found that a subset of hepatocytes and cholangiocytes are innervated by parasympathetic nerve terminals originating from the dorsal motor nucleus of the vagus. The elimination of the brain-liver axis by deleting parasympathetic cholinergic neurons innervating the liver prevents hepatic steatosis and promotes browning of inguinal white adipose tissue (ingWAT). The loss of liver-innervating cholinergic neurons increases hepatic Cyp7b1 expression and fasting serum bile acid levels. Furthermore, knockdown of the G protein-coupled bile acid receptor 1 gene in ingWAT reverses the beneficial effects of the loss of liver-innervating cholinergic neurons, leading to the reappearance of hepatic steatosis. Deleting liver-innervating cholinergic neurons has a small but significant effect on body weight, which is accompanied by an increase in energy expenditure. Taken together, these data suggest that targeting the parasympathetic cholinergic innervation of the liver is a potential therapeutic approach for enhancing hepatic lipid metabolism in obesity and diabetes.
Collapse
Affiliation(s)
- Jiyeon Hwang
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Junichi Okada
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Li Liu
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jeffrey E. Pessin
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Gary J. Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscince, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
30
|
Zhu J, Zhang M, Yue Y, Zhu J, Li D, Sun G, Chen X, Zhang H. Toxic Beauty: Parabens and benzophenone-type UV Filters linked to increased non-alcoholic fatty liver disease risk. CHEMOSPHERE 2024; 366:143555. [PMID: 39424158 DOI: 10.1016/j.chemosphere.2024.143555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) has increased concomitantly with heightened exposure to environmental chemicals, such as benzophenone-type ultraviolet (BP-type UV) filters and parabens, which are prevalent in personal care products. This study aimed to investigate the potential link between the exposure to these chemicals and the risk of developing NAFLD. We conducted a case-control study involving 228 participants from South China, encompassing individuals diagnosed with NAFLD and healthy controls. Blood samples were collected and analyzed for the presence of 11 parabens and 8 BP-type UV filters. The findings revealed significantly elevated concentrations of several parabens and BP-type UV filters in the blood of patients with NAFLD compared with the healthy cohort. Notably, methylparaben (MeP), ethylparaben (EtP), isopropylparaben (iPrP), butylparaben (BuP), isobutylparaben (iBuP), 3,4-dihydroxybenzoic acid (3,4-DHB), total parabens (Σparabens), BP1, BP3, BP4, and 4-hydroxybenzophenone (4-OH-BP) were identified as significant predictors of NAFLD prevalence. Through multiple regression analyses, the blood levels of iBuP, Σparabens, and BP4 were found to be significantly associated with elevated triglycerides (TG) (β = 0.59 mmol/L, 95% CI = 0.11-1.59), total bilirubin (TBIL) (β = 2.81 μmol/L, 95% CI = 0.46-15.6) or direct bilirubin (DBIL) (β = 1.89 μmol/L, 95% CI = 0.47-10.2), and reduced globulins (GLB) (β = -0.29 g/L, 95% CI = -0.07 to -5.45), respectively, which are indicators of liver damage. Moreover, TBIL and DBIL were found to mediate 26.7% and 24.6% of the increase in NAFLD prevalence associated with Σparabens, respectively. In conclusion, this study offers pioneering insights into human exposure to parabens and BP-type UV filters as well as their hepatotoxic potential.
Collapse
Affiliation(s)
- Jing Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
| | - Mingyue Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China
| | - Yuhan Yue
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Jinsen Zhu
- Department of Internal Medicine, Licheng Street Community Health Servic Center (Licheng hospital), Guangzhou, 511399, China
| | - Dehai Li
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Guodong Sun
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China; Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Xiaomei Chen
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Hua Zhang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
31
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
32
|
Hwang J, Okada J, Liu L, Pessin JE, Schwartz GJ, Jo YH. The development of hepatic steatosis depends on the presence of liver-innervating parasympathetic cholinergic neurons in mice fed a high-fat diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565494. [PMID: 38260695 PMCID: PMC10802435 DOI: 10.1101/2023.11.03.565494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Hepatic lipid metabolism is regulated by the autonomic nervous system of the liver, with the sympathetic innervation being extensively studied, while the parasympathetic efferent innervation is less understood despite its potential importance. In this study, we investigate the consequences of disrupted brain-liver communication on hepatic lipid metabolism in mice exposed to obesogenic conditions. We found that a subset of hepatocytes and cholangiocytes are innervated by parasympathetic nerve terminals originating from the dorsal motor nucleus of the vagus. The elimination of the brain-liver axis by deleting parasympathetic cholinergic neurons innervating the liver prevents hepatic steatosis and promotes browning of inguinal white adipose tissue (ingWAT). The loss of liver-innervating cholinergic neurons increases hepatic Cyp7b1 expression and fasting serum bile acid levels. Furthermore, knockdown of the G protein-coupled bile acid receptor 1 gene in ingWAT reverses the beneficial effects of the loss of liver-innervating cholinergic neurons, leading to the reappearance of hepatic steatosis. Deleting liver-innervating cholinergic neurons has a small but significant effect on body weight, which is accompanied by an increase in energy expenditure. Taken together, these data suggest that targeting the parasympathetic cholinergic innervation of the liver is a potential therapeutic approach for enhancing hepatic lipid metabolism in obesity and diabetes.
Collapse
|
33
|
Bott S, Lallement J, Marino A, Daskalopoulos EP, Beauloye C, Esfahani H, Dessy C, Leclercq IA. When the liver is in poor condition, so is the heart - cardiac remodelling in MASH mouse models. Clin Sci (Lond) 2024; 138:1151-1171. [PMID: 39206703 PMCID: PMC11405860 DOI: 10.1042/cs20240833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/15/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) confers a risk for cardiovascular diseases in patients. Animal models may help exploring the mechanisms linking liver and heart diseases. Hence, we explored the cardiac phenotype in two MASH mouse models: foz/foz mice fed a high-fat diet (HFD) for 24 or 60 weeks and C57BL/6J mice fed a high-fat-, high-cholesterol-, and high-fructose diet for 60 weeks. Angiotensin II (AngII) was used as an additional cardiovascular stressor for 4 weeks in 10 weeks HFD-fed foz/foz mice. Foz/foz mice with fibrosing MASH developed cardiac hypertrophy with adverse cardiac remodelling not seen in WT similarly fed the HFD. AngII caused hypertension and up-regulated the expression of genes contributing to pathological cardiac hypertrophy (Nppa, Myh7) more severely so in foz/foz mice than in controls. After 60 weeks of HFD, while liver disease had progressed to burn-out non steatotic MASH with hepatocellular carcinoma in 50% of the animals, the cardiomyopathy did not. In an independent model (C57BL/6J mice fed a fat-, cholesterol- and fructose-rich diet), moderate fibrosing MASH is associated with cardiac fibrosis and dysregulation of genes involved in pathological remodelling (Col1a1, Col3a1, Vim, Myh6, Slc2a1). Thus, animals with MASH present consistent adverse structural changes in the heart with no patent alteration of cardiac function even when stressed with exogenous AngII. Liver disease, and likely not overfeeding or aging alone, is associated with this cardiac phenotype. Our findings support foz/foz mice as suitable for studying links between MASH and heart structural changes ahead of heart failure.
Collapse
Affiliation(s)
- Sebastian Bott
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Justine Lallement
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Alice Marino
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Christophe Beauloye
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Hrag Esfahani
- Platform of Integrated Physiology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Anne Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
| |
Collapse
|
34
|
Liu W, Shi L, Yuan M, Zhang Y, Li Y, Cheng C, Liu J, Yue H, An L. Strong associations between fasting lipids and glucose concentrations and ALT levels strengthened with increasing ALT quantiles. Lipids Health Dis 2024; 23:295. [PMID: 39267040 PMCID: PMC11391808 DOI: 10.1186/s12944-024-02281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND A persistent redox state and excessive reactive species involved in carbohydrate and lipid metabolism lead to oxidative damage in the liver, however, how fasting plasma concentrations of lipids and glucose are associated with fasting blood levels of alanine transaminase (ALT) and aspartate transaminase (AST) remains to be evaluated in large-scale population. METHODS A cross-sectional study with 182,971 residents aged 18 to 92 years; multidimensional stratified analyses including quantile linear regression analysis and sex stratification were adopted to improve the quality of the evidence. RESULTS The associations between the concentrations of non-HDL-C and triglyceride and ALT levels were positive, stronger in males in each quantile of ALT levels and the coefficients expanded with increasing ALT levels at slopes of 3.610 and 5.678 in males and 2.977 and 5.165 in females, respectively. The associations between the HDL-C concentrations and ALT levels were negative, also stronger in males in each quantile and the coefficients expanded with increasing ALT levels at slopes of -7.839 in females and - 5.797 in males. The associations between glucose concentrations and ALT levels were positive, but stronger in females in each quantile and the coefficients expanded with increasing ALT levels at slopes of 1.736 in males and 2.177 in females, respectively. Similar pattern consist of relatively weaker coefficients and slops were observed between concentrations of non-HDL-C, triglyceride and glucose and AST levels. The associations between albumin concentration and concentrations of blood lipids and glucose were relatively steady across all quantiles. CONCLUSIONS The dose dependent effect between blood concentrations of lipids and glucose and liver function changes suggests that excessive carbohydrate and lipid metabolism may cause subclinical liver damage. Long term sustained primary and secondary inflammatory factors produced in the liver might be transmitted to adjacent organs, such as the heart, kidneys, and lungs, to cause and/or exacerbate pathological changes in these visceral organs.
Collapse
Affiliation(s)
- Wei Liu
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Lipu Shi
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Mengmeng Yuan
- Department of Gynecology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Yonghui Zhang
- Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Yalong Li
- Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Chaofei Cheng
- Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Junping Liu
- Department of Infectious Diseases, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Han Yue
- Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.
| | - Lemei An
- Department of Rheumatology and Clinical Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
35
|
Denova-Gutiérrez E, Rivera-Paredez B, Quezada-Sánchez AD, Armenta-Guirado BI, Muñoz-Aguirre P, Flores YN, Velázquez-Cruz R, Salmerón J. Soft drink consumption and increased risk of nonalcoholic fatty liver disease: Results from the health workers cohort study. Ann Hepatol 2024; 30:101566. [PMID: 39276986 DOI: 10.1016/j.aohep.2024.101566] [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: 01/18/2024] [Revised: 05/27/2024] [Accepted: 07/17/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION AND OBJECTIVES Nonalcoholic fatty liver disease (NAFLD) is a common clinical condition and an important public health problem. Some epidemiological studies have suggested that soft drinks (SD) intake is associated with NAFLD. However, the evidence is inconsistent. Our objective was to assess the association between SD consumption and the risk of NAFLD in a Mexican adult population. MATERIALS AND METHODS A total of 1,759 participants from the Health Workers Cohort Study (HWCS) were included in the analyses. SD intake was measured using a validated food frequency questionnaire. We classified SD consumption as follows: a) less than 1 serving per week, b) 1 to less than 3.5 servings per week, and c) 3.5 or more servings per week. Hepatic steatosis index (HSI) was calculated based on sex, BMI, and blood transaminase levels, and was categorized as NAFLD ≥ 36. To assess the relation between SD and NAFLD, we followed two approaches: fixed effects logistic regression and generalized estimating equations. RESULTS After adjusting for demographic characteristics, lifestyle factors, and dietary intake, the odds ratio (OR) and 95 % confidence interval (95 % CI) for NAFLD were 1.26 (95 % CI: 1.08, 1.48) for 1 to less than 3.5 servings per week and 1.42 (95 % CI: 1.19, 1.69) for ≥3.5 servings/week category in both sexes. When stratifying the analysis by sex, we observed that the association tended to be greater in men than in women. CONCLUSIONS The results from our prospective study indicate that SD consumption is associated with an increased risk of NAFLD.
Collapse
Affiliation(s)
- Edgar Denova-Gutiérrez
- Center for Research in Nutrition and Health, Mexican National Institute of Public Health, Cuernavaca, Mexico.
| | - Berenice Rivera-Paredez
- Center for Research in Policies, Population and Health, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| | - Amado D Quezada-Sánchez
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Mexico
| | | | - Paloma Muñoz-Aguirre
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Center for Population Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Yvonne N Flores
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Instituto Mexicano del Seguro Social, Cuernavaca, Mexico; UCLA Department of Health Policy and Management, Center for Cancer Prevention and Control Research, and UCLA Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, USA
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Jorge Salmerón
- Center for Research in Policies, Population and Health, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico; Unidad de Investigación Epidemiológica y en Servicios de Salud, Instituto Mexicano del Seguro Social, Cuernavaca, Mexico
| |
Collapse
|
36
|
Fu K, Dai S, Ma C, Zhang Y, Zhang S, Wang C, Gong L, Zhou H, Li Y. Lignans are the main active components of
Schisandrae Chinensis Fructus for liver disease treatment: a review. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:2425-2444. [DOI: 10.26599/fshw.2022.9250200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Wang Y, Li X, Gao Y, Zhang X, Liu Y, Wu Q. Risk Factors for Non-Alcoholic Fatty Liver Disease in Patients with Bipolar Disorder: A Cross-Sectional Retrospective Study. Diabetes Metab Syndr Obes 2024; 17:3053-3061. [PMID: 39170901 PMCID: PMC11338168 DOI: 10.2147/dmso.s463335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Purpose The co-morbidity of non-alcoholic fatty liver disease (NAFLD) in patients with bipolar disorder (BD) has a negative impact on patient treatment and prognosis. This study aimed to identify the prevalence of NAFLD in patients with BD and investigate the risk factors of NAFLD. Patients and Methods A total of 678 patients with BD were included in the study. Clinical data were obtained from the hospital's electronic health record system. Data included fasting blood glucose, alanine aminotransferase, triglycerides, aspartate aminotransferase, high-density lipoprotein cholesterol (HDL), alkaline phosphatase, total cholesterol, glutamine transpeptidase, uric acid, apolipoprotein A1, apolipoprotein B, and liver ultrasound findings. Results The prevalence of NAFLD was 43.66% in patients with BD. Significant differences in body mass index (BMI), mean age, diabetes prevalence, course of BD, fasting blood glucose, alanine aminotransferase, HDL, alkaline phosphatase, triglycerides, aspartate aminotransferase, uric acid, glutamine transpeptidase, apolipoprotein B, total cholesterol, and apolipoprotein A1 were seen between the groups (all P<0.01). Male sex, age, BMI, course of BD, alanine aminotransferase, fasting blood glucose, aspartate aminotransferase, diabetes, glutamine transpeptidase, total cholesterol, alkaline phosphatase, triglycerides, uric acid, apolipoprotein B, HDL, and apolipoprotein A1 levels were correlated with NAFLD (all P<0.05). In patients with BD, diabetes (OR=6.412, 95% CI=1.049-39.21), BMI (OR=1.398, 95% CI=1.306-1.497), triglycerides (OR=1.456, 95% CI=1.036-2.045), and apolipoprotein A1 (OR=0.272, 95% CI=0.110-0.672) were risk factors for NAFLD (all P<0.05). Conclusion Risk factors for NAFLD in patients with BD include diabetes, BMI, course of BD, and a low level of apolipoprotein A1. A proactive approach to disease management, such as appropriate physical activity and adoption of a healthy diet, and regular monitoring of changes in patient markers should be adopted to reduce the prevalence of NAFLD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Psychiatry, Affiliated Psychological Hospital of Anhui Medical University, Anhui Mental Health Center, Hefei Fourth People’s Hospital, Hefei, Anhui, People’s Republic of China
| | - Xuelong Li
- Qingdao Mental Health Center, Qingdao, Shandong, People’s Republic of China
| | - Yakun Gao
- Qingdao Mental Health Center, Qingdao, Shandong, People’s Republic of China
| | - Xun Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Yiyi Liu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Qing Wu
- Department of Psychiatry, Affiliated Psychological Hospital of Anhui Medical University, Anhui Mental Health Center, Hefei Fourth People’s Hospital, Hefei, Anhui, People’s Republic of China
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, People’s Republic of China
| |
Collapse
|
38
|
Panyod S, Wu WK, Hsieh YC, Tseng YJ, Peng SY, Chen RA, Huang HS, Chen YH, Shen TCD, Ho CT, Liu CJ, Chuang HL, Huang CC, Wu MS, Sheen LY. Ginger essential oil prevents NASH progression by blocking the NLRP3 inflammasome and remodeling the gut microbiota-LPS-TLR4 pathway in mice. Nutr Diabetes 2024; 14:65. [PMID: 39152116 PMCID: PMC11329514 DOI: 10.1038/s41387-024-00306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Diet and gut microbiota contribute to non-alcoholic steatohepatitis (NASH) progression. High-fat diets (HFDs) change gut microbiota compositions, induce gut dysbiosis, and intestinal barrier leakage, which facilitates portal influx of pathogen-associated molecular patterns including lipopolysaccharides (LPS) to the liver and triggers inflammation in NASH. Current therapeutic drugs for NASH have adverse side effects; however, several foods and herbs that exhibit hepatoprotection could be an alternative method to prevent NASH. METHODS We investigated ginger essential oil (GEO) against palm oil-containing HFDs in LPS-injected murine NASH model. RESULTS GEO reduced plasma alanine aminotransferase levels and hepatic pro-inflammatory cytokine levels; and increased antioxidant catalase, glutathione reductase, and glutathione levels to prevent NASH. GEO alleviated hepatic inflammation through mediated NLR family pyrin domain-containing 3 (NLRP3) inflammasome and LPS/Toll-like receptor four (TLR4) signaling pathways. GEO further increased beneficial bacterial abundance and reduced NASH-associated bacterial abundance. CONCLUSION This study demonstrated that GEO prevents NASH progression which is probably associated with the alterations of gut microbiota and inhibition of the LPS/TLR4/NF-κB pathway. Hence, GEO may offer a promising application as a dietary supplement for the prevention of NASH.
Collapse
Affiliation(s)
- Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei, Taiwan, ROC
| | - Ya-Chi Hsieh
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Yea-Jing Tseng
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Sin-Yi Peng
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Yi-Hsun Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Ting-Chin David Shen
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Chun-Jen Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan City, Taiwan, ROC
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC.
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
39
|
Yang H, Ou F, Chang Q, Jiang J, Liu Y, Ji C, Chen L, Xia Y, Zhao Y. Physical frailty, genetic predisposition, and the risks of severe non-alcoholic fatty liver disease and cirrhosis: a cohort study. J Cachexia Sarcopenia Muscle 2024; 15:1491-1500. [PMID: 38887910 PMCID: PMC11294048 DOI: 10.1002/jcsm.13506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Frailty, defined as a phenotype of decreased physiological reserves and diminished ability to respond to stressors, has been linked to the development of chronic diseases. Epidemiological evidence connecting frailty to non-alcoholic fatty liver disease (NAFLD) and cirrhosis risks remain sparse. We aimed to assess the longitudinal associations of frailty with the risks of severe NAFLD and cirrhosis in middle-aged to older adults and further explore the modification role of genetic risk on these associations. METHODS This study included a total of 398 386 participants from the UK Biobank. Incident cases of severe NAFLD and cirrhosis were ascertained through linked hospital records and death registries. Frailty status was assessed by a modified version of the frailty phenotype, encompassing five key components: weight loss, tiredness, physical activity, gait speed, and grip strength. Participants were classified as pre-frailty if they met one or two of these criteria, and as frailty if they met three or more. Genetic predisposition to NAFLD and cirrhosis was estimated by genetic risk score (GRS) and further categorized into high, intermediate, and low genetic risk levels according to tertiles of GRSs. Cox proportional hazards regression model was employed to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for their associations. RESULTS The mean (standard deviation) age of the study population was 56.6 (8.03) years. 214 408 (53.8%) of the participants was female; 14 924 (3.75%) of participants met the criteria for frailty, 170 498 (42.8%) for pre-frailty, and 212 964 (53.5%) for non-frailty. Over a median follow-up of 12.0 years, we documented 4439 incident severe NAFLD and 3323 incident cirrhosis cases, respectively. Compared with non-frailty, both pre-frailty (HR: 1.50; 95% CI: 1.40-1.60) and frailty (HR: 1.98; 95% CI: 1.77-2.21) were associated with increased risk of NAFLD. Similar associations were observed for cirrhosis, the corresponding HRs (95% CIs) for non-frailty, pre-frailty, and frailty were 1.00 (reference), 1.29 (1.20, 1.38), and 1.90 (1.66, 2.18). Such associations were consistent across all genetic risk levels, with no observed interactions between frailty and GRSs (all P for interactions ≥0.10). Compared with participants with frailty and a low level of genetic risk, the greatest risk increasement in developing severe NAFLD (HR: 3.36; 95% CI: 2.83-3.99) and cirrhosis (HR: 2.81; 95% CI: 2.29-3.44) was both observed in those with frailty and a high level of genetic risk. CONCLUSIONS Our findings indicate that frailty is a significant predictor of severe NAFLD and cirrhosis, irrespective of genetic predisposition.
Collapse
Affiliation(s)
- Honghao Yang
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Fengrong Ou
- School of Public HealthChina Medical UniversityShenyangChina
| | - Qing Chang
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Jinguo Jiang
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Yashu Liu
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Chao Ji
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yang Xia
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| | - Yuhong Zhao
- Department of Clinical EpidemiologyShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic DiseaseShenyangChina
| |
Collapse
|
40
|
Rieman-Klingler MC, Jung J, Tesfai K, Loomba R, Non AL. Integrating genetic and socioeconomic data to predict the progression of nonalcoholic fatty liver disease. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 184:e24979. [PMID: 38778456 DOI: 10.1002/ajpa.24979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVES Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease globally, with an estimated prevalence exceeding 25%. Variants in the PNPLA3 and HSD17B13 genes have been a focus of investigations surrounding the etiology and progression of NAFLD and are believed to contribute to a greater burden of disease experienced by Hispanic Americans. However, little is known about socioeconomic factors influencing NAFLD progression or its increased prevalence among Hispanics. MATERIALS AND METHODS We cross-sectionally analyzed 264 patients to assess the role of genetic and socioeconomic variables in the development of advanced liver fibrosis in individuals at risk for NAFLD. RESULTS Adjusting for age, sex, body mass index, and PNPLA3 genotype, lacking a college degree was associated with 3.3 times higher odds of advanced fibrosis (95% confidence interval [CI]: 1.21-8.76, p = 0.019), an effect comparable to that of possessing the major PNPLA3 risk variant. Notably, the effect of PNPLA3 genotype on advanced fibrosis was attenuated to nonsignificance following adjustment for education and other socioeconomic markers. The effect of the protective HSD17B13 variant, moreover, diminished after adjustment for education (odds ratio [OR]: 0.39 [95% CI: 0.13-1.16, p = 0.092]), while lower education continued to predict advanced fibrosis following multivariable adjustment with an OR of 8.0 (95% CI: 1.91-33.86, p = 0.005). DISCUSSION Adjusting for education attenuated the effects of genotype and Hispanic ethnicity on liver fibrosis, suggesting that social factors-rather than genes or ethnicity-may be driving disease severity within some populations. Findings reveal the importance of including socioenvironmental controls when considering the role of genetics or ethnicity in complex disease.
Collapse
Affiliation(s)
- Maria C Rieman-Klingler
- Department of Anthropology, University of California, San Diego, La Jolla, California, USA
- School of Medicine, University of California, San Diego, La Jolla, California, USA
- Medical Scientist Training (MD/PhD) Program, University of California, San Diego, La Jolla, California, USA
| | - Jinho Jung
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kaleb Tesfai
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, California, USA
| | - Amy L Non
- Department of Anthropology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
41
|
Pompili S, Cappariello A, Vetuschi A, Sferra R. G-Protein-Coupled Receptor 120 Agonist Mitigates Steatotic and Fibrotic Features Triggered in Obese Mice by the Administration of a High-Fat and High-Carbohydrate Diet. ACS OMEGA 2024; 9:31899-31909. [PMID: 39072106 PMCID: PMC11270546 DOI: 10.1021/acsomega.4c03507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024]
Abstract
Nonalcoholic fatty liver disease (NALFD) represents a complex condition ranging from simple steatosis (nonalcoholic fatty liver, NAFL) to inflammation, and fibrosis is one of the main features of nonalcoholic steatohepatitis (NASH). The pathogenesis of NAFLD is not well established but involves several factors (i.e., predisposition of genetic variants, obesity, and unhealthy lifestyle as unbalanced diets) that lead to an alteration of lipid homeostasis and consequently to an abnormal accumulation of triglycerides and other lipids in the liver parenchyma. Currently, no resolutive pharmacological treatment for NAFLD is available, and the only therapeutic approach is a healthy diet and physical exercise. In this study, we investigated the potential beneficial effect of GprA, a new synthetic agonist of G-protein-coupled receptor 120/free fatty acid receptor 4 (GPR120/FFAR4), in the progression of NAFL/NASH in mice fed for different periods (26 weeks and 30 weeks), with a high-fat (40% kcal) and high-carbohydrate diet, also called a Western-style diet (WSD). In our experimental model, the histological, protein, and transcriptomic analyses highlighted that the GprA can reduce signs of steatosis in WSD-fed mice. Furthermore, in 30 week-treated mice, GprA is also effective in the reduction of collagen deposition and fibrosis development. Altogether, our data validate the central role of FFAR4 in the context of NAFL/NASH onset and progression and reveal that GprA could represent an interesting candidate for the development of a new therapeutic approach in NAFLD treatment.
Collapse
Affiliation(s)
- Simona Pompili
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Alfredo Cappariello
- Department
of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Antonella Vetuschi
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Roberta Sferra
- Department
of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
42
|
Botacin EC, Duarte SMB, Stefano JT, Barbosa MED, Pessoa MG, Oliveira CP. ASSOCIATION BETWEEN ANXIETY AND DEPRESSION IN METABOLIC DYSFUNCTION-ASSOCIATED STEATOTIC LIVER DISEASE (MASLD). ARQUIVOS DE GASTROENTEROLOGIA 2024; 61:e23128. [PMID: 39045999 DOI: 10.1590/s0004-2803.24612023-128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/09/2023] [Indexed: 07/25/2024]
Abstract
BACKGROUND This study aimed to assess the frequency and intensity of anxious and depressive symptoms in patients diagnosed with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS This is a descriptive and cross-sectional study, resulting from 106 patients from the Hepatology outpatient clinic at the Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo, Brazil without a history of alcohol abuse, verified by the alcohol use disorders identification test (AUDIT). These were assessed using the sociodemographic data sheet, Hospital Anxiety and Depression Scale (HADS), Hamilton Anxiety Rating Scale (HAM-A), and Hamilton Depression Scale (HAM-D). RESULTS A total of 69.8% were women and 30.2% were men, with a mean age of 61 years. The majority (71.7%) discovered MASLD through routine exams, presenting as comorbidities: Type 2 diabetes mellitus (59.4%), Dyslipidemia (49.1%), Arterial hypertension (68.9%), Obesity (61.3%) and Metabolic syndrome [MetS (63.2%)]. The HADS scale indicates 34% probability of anxiety and 33% depressive symptoms. The Hamilton's scales of intensity indicates 63.9% severe anxiety and 54.3% severe depression. There is also a relationship between anxiety, depression and the female gender, as well as between depression and MetS. CONCLUSION The findings point to the presence of anxiety and depression in more than one third of MASLD patients, most with severe symptoms. The group is concentrated in the elderly, with many comorbidities, including MetS. There was a positive correlation between anxiety, depression and being female; also, being significant between MetS and depression.
Collapse
Affiliation(s)
- Eloyse Cristina Botacin
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| | - Sebastião Mauro Bezerra Duarte
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| | - José Tadeu Stefano
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| | - Mary Ellen Dias Barbosa
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| | - Mario Guimarães Pessoa
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| | - Cláudia P Oliveira
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Laboratório de Gastroenterologia Clínica e Experimental (LIM-07), Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Departamento de Gastroenterologia e Hepatologia, São Paulo, SP, Brasil
| |
Collapse
|
43
|
Zhang S, Cui Z, Zhang H, Wang P, Wang F, Zhang J. Pea Albumin Extracted from Pea ( Pisum sativum L.) Seeds Ameliorates High-Fat-Diet-Induced Non-Alcoholic Fatty Liver Disease by Regulating Lipogenesis and Lipolysis Pathways. Nutrients 2024; 16:2232. [PMID: 39064674 PMCID: PMC11280122 DOI: 10.3390/nu16142232] [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: 06/21/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is now recognized as the most prevalent liver disease globally. Pea albumin (PA) has demonstrated positive impacts on reducing obesity and improving glucose metabolism. In this research, a mouse model of NAFLD induced by a high-fat diet (HFD) was employed to examine the impact of PA on NAFLD and explore its potential mechanisms. The findings revealed that mice subjected to a HFD developed pronounced fatty liver alterations. The intervention with PA significantly lowered serum TC by 26.81%, TG by 43.55%, and LDL-C by 57.79%. It also elevated HDL-C levels by 1.2 fold and reduced serum ALT by 37.94% and AST by 31.21% in mice fed a HFD. These changes contributed to the reduction in hepatic steatosis and lipid accumulation. Additionally, PA improved insulin resistance and inhibited hepatic oxidative stress and inflammatory responses. Mechanistic studies revealed that PA alleviated lipid accumulation in HFD-induced NAFLD by activating the phosphorylation of AMPKα and ACC, inhibiting the expression of SREBF1 and FASN to reduce hepatic lipogenesis, and increasing the expression of ATGL, PPARα, and PPARγ to promote lipolysis and fatty acid oxidation. These results indicate that PA could serve as a dietary supplement for alleviating NAFLD, offering a theoretical foundation for the rational intake of PA in NAFLD intervention.
Collapse
Affiliation(s)
- Shucheng Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (S.Z.); (H.Z.)
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Zhengwu Cui
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Hao Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (S.Z.); (H.Z.)
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| | - Pengjie Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (S.Z.); (H.Z.)
| | - Fuqing Wang
- Tibet Tianhong Science and Technology Co., Ltd., Lhasa 850000, China;
| | - Jian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
44
|
Liang Y, Luo S, Bell S, Mo JMY, He B, Zhou Y, Bai X, Au Yeung SL. Do iron homeostasis biomarkers mediate the associations of liability to type 2 diabetes and glycemic traits in liver steatosis and cirrhosis: a two-step Mendelian randomization study. BMC Med 2024; 22:270. [PMID: 38926684 PMCID: PMC11210020 DOI: 10.1186/s12916-024-03486-w] [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: 12/02/2023] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Previous studies, including Mendelian randomization (MR), have demonstrated type 2 diabetes (T2D) and glycemic traits are associated with increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD). However, few studies have explored the underlying pathway, such as the role of iron homeostasis. METHODS We used a two-step MR approach to investigate the associations of genetic liability to T2D, glycemic traits, iron biomarkers, and liver diseases. We analyzed summary statistics from various genome-wide association studies of T2D (n = 933,970), glycemic traits (n ≤ 209,605), iron biomarkers (n ≤ 246,139), MASLD (n ≤ 972,707), and related biomarkers (alanine aminotransferase (ALT) and proton density fat fraction (PDFF)). Our primary analysis was based on inverse-variance weighting, followed by several sensitivity analyses. We also conducted mediation analyses and explored the role of liver iron in post hoc analysis. RESULTS Genetic liability to T2D and elevated fasting insulin (FI) likely increased risk of liver steatosis (ORliability to T2D: 1.14 per doubling in the prevalence, 95% CI: 1.10, 1.19; ORFI: 3.31 per log pmol/l, 95% CI: 1.92, 5.72) and related biomarkers. Liability to T2D also likely increased the risk of developing liver cirrhosis. Genetically elevated ferritin, serum iron, and liver iron were associated with higher risk of liver steatosis (ORferritin: 1.25 per SD, 95% CI 1.07, 1.46; ORliver iron: 1.15 per SD, 95% CI: 1.05, 1.26) and liver cirrhosis (ORserum iron: 1.31, 95% CI: 1.06, 1.63; ORliver iron: 1.34, 95% CI: 1.07, 1.68). Ferritin partially mediated the association between FI and liver steatosis (proportion mediated: 7%, 95% CI: 2-12%). CONCLUSIONS Our study provides credible evidence on the causal role of T2D and elevated insulin in liver steatosis and cirrhosis risk and indicates ferritin may play a mediating role in this association.
Collapse
Affiliation(s)
- Ying Liang
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Shan Luo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Steven Bell
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Jacky Man Yuen Mo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Baoting He
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yangzhong Zhou
- Department of Rheumatology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
| | - Xiaoyin Bai
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shiu Lun Au Yeung
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
45
|
Jiang FY, Yue SR, Tan YY, Tang N, Xu YS, Zhang BJ, Mao YJ, Xue ZS, Lu AP, Liu BC, Wang RR. Gynostemma pentaphyllum Extract Alleviates NASH in Mice: Exploration of Inflammation and Gut Microbiota. Nutrients 2024; 16:1782. [PMID: 38892715 PMCID: PMC11174846 DOI: 10.3390/nu16111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
NASH (non-alcoholic steatohepatitis) is a severe liver disease characterized by hepatic chronic inflammation that can be associated with the gut microbiota. In this study, we explored the therapeutic effect of Gynostemma pentaphyllum extract (GPE), a Chinese herbal extract, on methionine- and choline-deficient (MCD) diet-induced NASH mice. Based on the peak area, the top ten compounds in GPE were hydroxylinolenic acid, rutin, hydroxylinoleic acid, vanillic acid, methyl vanillate, quercetin, pheophorbide A, protocatechuic acid, aurantiamide acetate, and iso-rhamnetin. We found that four weeks of GPE treatment alleviated hepatic confluent zone inflammation, hepatocyte lipid accumulation, and lipid peroxidation in the mouse model. According to the 16S rRNA gene V3-V4 region sequencing of the colonic contents, the gut microbiota structure of the mice was significantly changed after GPE supplementation. Especially, GPE enriched the abundance of potentially beneficial bacteria such as Akkerrmansia and decreased the abundance of opportunistic pathogens such as Klebsiella. Moreover, RNA sequencing revealed that the GPE group showed an anti-inflammatory liver characterized by the repression of the NF-kappa B signaling pathway compared with the MCD group. Ingenuity Pathway Analysis (IPA) also showed that GPE downregulated the pathogen-induced cytokine storm pathway, which was associated with inflammation. A high dose of GPE (HGPE) significantly downregulated the expression levels of the tumor necrosis factor-α (TNF-α), myeloid differentiation factor 88 (Myd88), cluster of differentiation 14 (CD14), and Toll-like receptor 4 (TLR4) genes, as verified by real-time quantitative PCR (RT-qPCR). Our results suggested that the therapeutic potential of GPE for NASH mice may be related to improvements in the intestinal microenvironment and a reduction in liver inflammation.
Collapse
Affiliation(s)
- Feng-Yan Jiang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Si-Ran Yue
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Yi-Yun Tan
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Nan Tang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Yue-Song Xu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Bao-Jun Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Yue-Jian Mao
- China Mengniu Dairy Company Limited, Hohhot 010000, China; (Y.-J.M.); (Z.-S.X.)
| | - Zheng-Sheng Xue
- China Mengniu Dairy Company Limited, Hohhot 010000, China; (Y.-J.M.); (Z.-S.X.)
| | - Ai-Ping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China;
| | - Bao-Cheng Liu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| | - Rui-Rui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (F.-Y.J.); (S.-R.Y.); (Y.-Y.T.); (N.T.); (Y.-S.X.); (B.-J.Z.)
| |
Collapse
|
46
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
47
|
Zhang X, Zhou C, Hu J, Hu J, Ding Y, Chen S, Wang X, Xu L, Gou Z, Zhang S, Shi W. Six-gene prognostic signature for non-alcoholic fatty liver disease susceptibility using machine learning. Medicine (Baltimore) 2024; 103:e38076. [PMID: 38728481 PMCID: PMC11081587 DOI: 10.1097/md.0000000000038076] [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: 12/28/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND nonalcoholic fatty liver disease (NAFLD) is a common liver disease affecting the global population and its impact on human health will continue to increase. Genetic susceptibility is an important factor influencing its onset and progression, and there is a lack of reliable methods to predict the susceptibility of normal populations to NAFLD using appropriate genes. METHODS RNA sequencing data relating to nonalcoholic fatty liver disease was analyzed using the "limma" package within the R software. Differentially expressed genes were obtained through preliminary intersection screening. Core genes were analyzed and obtained by establishing and comparing 4 machine learning models, then a prediction model for NAFLD was constructed. The effectiveness of the model was then evaluated, and its applicability and reliability verified. Finally, we conducted further gene correlation analysis, analysis of biological function and analysis of immune infiltration. RESULTS By comparing 4 machine learning algorithms, we identified SVM as the optimal model, with the first 6 genes (CD247, S100A9, CSF3R, DIP2C, OXCT 2 and PRAMEF16) as predictive genes. The nomogram was found to have good reliability and effectiveness. Six genes' receiver operating characteristic curves (ROC) suggest an essential role in NAFLD pathogenesis, and they exhibit a high predictive value. Further analysis of immunology demonstrated that these 6 genes were closely connected to various immune cells and pathways. CONCLUSION This study has successfully constructed an advanced and reliable prediction model based on 6 diagnostic gene markers to predict the susceptibility of normal populations to NAFLD, while also providing insights for potential targeted therapies.
Collapse
Affiliation(s)
- Xiang Zhang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunzi Zhou
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingwen Hu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingwen Hu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueping Ding
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shiqi Chen
- Lishui Hospital of Traditional Chinese Medicine, Lishui, China
| | - Xu Wang
- Shanghai Jinshan TCM-Integrated Hospital, Shanghai, China
| | - Lei Xu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Gou
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqun Shi
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
48
|
Yang S, Li J, Yan L, Wu Y, Zhang L, Li B, Tong H, Lin X. Molecular Mechanisms of Fucoxanthin in Alleviating Lipid Deposition in Metabolic Associated Fatty Liver Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10391-10405. [PMID: 38669300 DOI: 10.1021/acs.jafc.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is witnessing a global surge; however, it still lacks effective pharmacological interventions. Fucoxanthin, a natural bioactive metabolite derived from marine brown algae, exhibits promising pharmacological functions, particularly in ameliorating metabolic disorders. However, the mechanisms underlying its therapeutic efficacy in addressing MAFLD remain elusive. Our present findings indicated that fucoxanthin significantly alleviated palmitic acid (PA)-induced hepatic lipid deposition in vitro and obesity-induced hepatic steatosis in ob/ob mice. Moreover, at both the protein and transcriptional levels, fucoxanthin effectively increased the expression of PPARα and CPT1 (involved in fatty acid oxidation) and suppressed FASN and SREBP1c (associated with lipogenesis) in both PA-induced HepG2 cells and hepatic tissues in ob/ob mice. This modulation was accompanied by the activation of AMPK. The capacity of fucoxanthin to improve hepatic lipid deposition was significantly attenuated when utilizing the AMPK inhibitor or siRNA-mediated AMPK silencing. Mechanistically, fucoxanthin activates AMPK, subsequently regulating the KEAP1/Nrf2/ARE signaling pathway to exert antioxidative effects and stimulating the PGC1α/NRF1 axis to enhance mitochondrial biogenesis. These collective actions contribute to fucoxanthin's amelioration of hepatic steatosis induced by metabolic perturbations. These findings offer valuable insights into the prospective utilization of fucoxanthin as a therapeutic strategy for managing MAFLD.
Collapse
Affiliation(s)
- Shouxing Yang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Jinhai Li
- Department of Liver and Gall Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, P.R. China
| | - Liping Yan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Yu Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Lin Zhang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Boyang Li
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, P.R. China
| | - Xiaochun Lin
- Department of Pediatrics Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China
| |
Collapse
|
49
|
Tong L, Chen Z, Li Y, Wang X, Yang C, Li Y, Zhu Y, Lu Y, Liu Q, Xu N, Shao S, Wu L, Zhang P, Wu G, Wu X, Chen X, Fang J, Jia R, Xu T, Li B, Zheng L, Liu J, Tong X. Transketolase promotes MAFLD by limiting inosine-induced mitochondrial activity. Cell Metab 2024; 36:1013-1029.e5. [PMID: 38547864 DOI: 10.1016/j.cmet.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/10/2024] [Accepted: 03/06/2024] [Indexed: 05/12/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has a global prevalence of about 25% and no approved therapy. Using metabolomic and proteomic analyses, we identified high expression of hepatic transketolase (TKT), a metabolic enzyme of the pentose phosphate pathway, in human and mouse MAFLD. Hyperinsulinemia promoted TKT expression through the insulin receptor-CCAAT/enhancer-binding protein alpha axis. Utilizing liver-specific TKT overexpression and knockout mouse models, we demonstrated that TKT was sufficient and required for MAFLD progression. Further metabolic flux analysis revealed that Tkt deletion increased hepatic inosine levels to activate the protein kinase A-cAMP response element binding protein cascade, promote phosphatidylcholine synthesis, and improve mitochondrial function. Moreover, insulin induced hepatic TKT to limit inosine-dependent mitochondrial activity. Importantly, N-acetylgalactosamine (GalNAc)-siRNA conjugates targeting hepatic TKT showed promising therapeutic effects on mouse MAFLD. Our study uncovers how hyperinsulinemia regulates TKT-orchestrated inosine metabolism and mitochondrial function and provides a novel therapeutic strategy for MAFLD prevention and treatment.
Collapse
Affiliation(s)
- Lingfeng Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhangbing Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yangyang Li
- Unit of Immune and Metabolic Regulation, School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Xinxia Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Changjie Yang
- Department of Liver Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yakui Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Qi Liu
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Nannan Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sijia Shao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guangyu Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoyu Wu
- Key Laboratory of Pediatric Hematology and Oncology, Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiaosong Chen
- Department of Liver Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Junwei Fang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200032, China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tianle Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liang Zheng
- Key Laboratory of Pediatric Hematology and Oncology, Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Synvida Biotechnology Co., Ltd, Shanghai, China.
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
50
|
Alvarado-Ojeda ZA, Trejo-Moreno C, Ferat-Osorio E, Méndez-Martínez M, Fragoso G, Rosas-Salgado G. Role of Angiotensin II in Non-Alcoholic Steatosis Development. Arch Med Res 2024; 55:102986. [PMID: 38492325 DOI: 10.1016/j.arcmed.2024.102986] [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: 08/30/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Fatty liver is a multifactorial disease characterized by excessive accumulation of lipids in hepatocytes (steatosis), insulin resistance, oxidative stress, and inflammation. This disease has a major public health impact because it is the first stage of a chronic and degenerative process in the liver that can lead to steatohepatitis, cirrhosis, and liver cancer. Although this disease is mainly diagnosed in patients with obesity, type 2 diabetes mellitus, and dyslipidemia, recent evidence indicates that vasoactive hormones such as angiotensin II (ANGII) not only promote endothelial dysfunction (ED) and hypertension, but also cause fatty liver, increase adipose tissue, and develop a pro-steatotic environment characterized by a low-grade systemic pro-inflammatory and pro-oxidant state, with elevated blood lipid levels. The role of ANGII in lipid accumulation has been little studied, so this review aims to summarize existing reports on the possible mechanism of action of ANGII in inducing lipid accumulation in hepatocytes.
Collapse
Affiliation(s)
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico
| | - Eduardo Ferat-Osorio
- División de Investigación en Salud, Unidad de Investigación en Epidemiología Clínica, Hospital de Especialidades, Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico.
| |
Collapse
|