1
|
Gbadamosi SO, Evans KA, Brady BL, Hoovler A. Noninvasive tests and diagnostic pathways to MASH diagnosis in the United States: a retrospective observational study. J Med Econ 2025; 28:314-322. [PMID: 39963742 DOI: 10.1080/13696998.2025.2468582] [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: 12/20/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/03/2025]
Abstract
AIM Although liver biopsy is considered the most reliable diagnostic tool for metabolic dysfunction-associated steatohepatitis (MASH), it is invasive and can be costly. Clinicians are increasingly relying on routine biomarkers and other noninvasive tests (NITs) for diagnosis. We examined real-world diagnostic pathways for patients newly diagnosed with MASH with a primary focus on NITs. MATERIALS AND METHODS This retrospective, observational study analyzed healthcare claims data (Merative MarketScan Commercial and Medicare Databases) from patients in the United States newly diagnosed with MASH from October 1, 2016, to March 31, 2023. Patients ≥18 years old with ≥12 months of continuous enrollment with medical and pharmacy benefits prior to diagnosis were included. Diagnostic pathways leading up to MASH diagnosis, including NITs (blood-based and imaging-based tests) and liver biopsies were assessed. Prevalence of comorbid conditions, MASH-associated medication use, and the diagnosing physician specialty were also examined. RESULTS A total of 18,396 patients were included in the analysis. Routine laboratory tests (alanine aminotransferase [ALT], albumin, aspartate aminotransferase [AST], cholesterol, complete blood count, and hemoglobin A1c) were performed among ≥70% of patients prior to MASH diagnosis, including 89% of patients with a liver enzyme test (ALT and/or AST). More than 75% of patients had necessary laboratory tests to calculate AST to platelet ratio index (APRI) and fibrosis-4 index (FIB-4) scores. The most common imaging performed was ultrasound (62%); liver biopsy was only performed in 10% of patients. There was a high prevalence of cardio metabolic risk factors such as hyperlipidemia (66%), hypertension (62%), obesity (58%), type 2 diabetes (40%), and cardiovascular disease (21%). Nearly half of the patients (49%) were diagnosed by a primary care physician. LIMITATIONS AND CONCLUSIONS This study highlights real-world diagnostic pathways among patients newly diagnosed with MASH, supporting previous findings that liver biopsies are infrequently used in favor of noninvasive methods.
Collapse
|
2
|
Polyzos SA, Mantzoros CS. Metabolic dysfunction-associated steatotic liver disease and malignancies: Unmasking a silent saboteur. Metabolism 2025; 168:156253. [PMID: 40164408 DOI: 10.1016/j.metabol.2025.156253] [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: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Not required for Editorials.
Collapse
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Christos S Mantzoros
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Internal Medicine, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Chaslin M, Maroun G, Durand E, Bonafos B, Assou S, Chaiyut J, Vaysse L, Ferrer V, Liengprayoon S, Brioche T, Pessemesse L, Macart M, Bertrand-Gaday C, Pers YM, Coudray C, Brondello JM, Casas F, Feillet-Coudray C. Furan fatty acids supplementation in obese mice reverses hepatic steatosis and protects against cartilage degradation. Biomed Pharmacother 2025; 187:118072. [PMID: 40253827 DOI: 10.1016/j.biopha.2025.118072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
Obesity is a major global health problem associated with numerous metabolic dysfunctions, an increased risk of developing Metabolic Associated Steatotic Liver Disease (MASLD) and osteoarthritis. Recently, we demonstrated that in Diet-induced-Obesity (DIO) mouse model, preventive furan fatty acids (FuFA-F2) supplementation, a natural compounds found in many foods, reduced the onset of metabolic disorders and increased muscle mass. Here, we aimed to determine whether a short FuFA-F2 supplementation is capable of providing beneficial health effects in obese mice, notably by reversing metabolic disorders and limiting cartilage degradation. 6-month-old obese C57Bl/6 J mice were fed for four additional weeks on a high-fat and high-sucrose (HFHS) diet, supplemented or not with FuFA-F2 (40 mg/day/kg of body weight). Liver triglyceride content and histologic analysis revealed that 4 weeks of FuFA-F2 supplementation fully reversed hepatic steatosis in obese mice. Liver RNA-sequencing analysis highlighted that FuFA-F2 partly reversed the gene expression signature induced by the HFHS diet and favorably changed the expression of many genes known to be involved in the development of hepatic steatosis such as Pcsk9, Stard4, Insig1 and Sulf2. We also found that FuFA-F2 supplementation increased skeletal muscle mass and protected against cartilage degradation and synovitis induced by obesity. Our findings demonstrated that FuFA-F2 supplementation for 4 weeks in obese mice was enough to reverse the development of MASLD, promote an increase in skeletal muscle mass and protect against cartilage degradation induced by the HFHS diet. This study highlights that nutritional supplementation with FuFA-F2 could be an effective approach to treat obesity-related disorders.
Collapse
Affiliation(s)
| | - Georges Maroun
- Institute of Regenerative Medicine and Biotherapies (IRMB), Univ Montpellier, INSERM, Montpellier, France
| | - Erwann Durand
- Qualisud, Univ Montpellier, CIRAD, Montpellier, France; Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | | | - Said Assou
- Institute of Regenerative Medicine and Biotherapies (IRMB), Univ Montpellier, INSERM, Montpellier, France
| | - Jatuporn Chaiyut
- Kasetsart agricultural and agro-industrial product improvement institute, Kasetsart University, Bangkok, Thailand
| | - Laurent Vaysse
- CIRAD, UPR BioWooEB, Montpellier, France; BioWooEB, Univ Montpellier, CIRAD, Montpellier, France
| | - Vincent Ferrer
- Kasetsart agricultural and agro-industrial product improvement institute, Kasetsart University, Bangkok, Thailand; CIRAD, UPR BioWooEB, Montpellier, France; BioWooEB, Univ Montpellier, CIRAD, Montpellier, France
| | - Siriluck Liengprayoon
- Kasetsart agricultural and agro-industrial product improvement institute, Kasetsart University, Bangkok, Thailand
| | | | | | | | | | - Yves-Marie Pers
- Institute of Regenerative Medicine and Biotherapies (IRMB), Univ Montpellier, INSERM, Montpellier, France; Montpellier University Hospital, Clinical immunology and osteoarticular diseases therapeutic Unit, Lapeyronie, Montpellier, France
| | | | - Jean-Marc Brondello
- Institute of Regenerative Medicine and Biotherapies (IRMB), Univ Montpellier, INSERM, Montpellier, France
| | | | | |
Collapse
|
4
|
Eid AH, Khachab M, Kobeissy F, Sahebkar A. Pharmacotherapeutic perspectives on nutraceuticals in the treatment of MASLD and MASH. Ther Adv Chronic Dis 2025; 16:20406223251339388. [PMID: 40415898 PMCID: PMC12103661 DOI: 10.1177/20406223251339388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Affiliation(s)
- Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Maha Khachab
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Zhang Y, Pan K, Xu A, Sun S, Huang Q, Wang Y, Wang H, Han Q, Li D, Ding Q, Li J. n-3 polyunsaturated fatty acids-enriched fish oil attenuates chronic alcohol-induced liver injury via a mechanism involving the upregulation of Retsat. J Nutr Biochem 2025:109971. [PMID: 40409513 DOI: 10.1016/j.jnutbio.2025.109971] [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: 11/19/2024] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
This study aimed to delineate the protective role of fish oil against alcoholic liver disease (ALD), identify the principal active component between eicosapentaenoic acid (EPA, C20:5 n-3) and docosahexaenoic acid (DHA, C22:6 n-3), and elucidate the molecular mechanisms. C57BL/6J mice were randomly assigned to receive either an alcohol-fed (AF) or pair-fed control (PF) diet, enriched with fish oil (FO) or corn oil (CO) for four weeks. Additionally, a series of in vitro experiments were performed using AML-12 cells to further investigate potential mechanisms. The results showed that plasma levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were significantly lower in the AF-FO group compared to the AF-CO group, indicating that fish oil alleviated alcohol-induced liver damage. Hepatic antioxidant markers, including glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT) were also higher in the AF-FO group than in the AF-CO group. Transcriptomic analysis revealed FO supplementation significantly affected genes involved in oxidoreductase activity and lipid metabolism pathways, with Retsat being the most up-regulated gene. The in vitro experiments indicated that DHA, but not EPA, markedly increased Retsat expression, cell viability, and the expression of genes related to oxidoreductase activity and lipid metabolism, compared to linoleic acid (LA, C18:2 n-6). Notably, knocking down Retsat abolished the protective effects of DHA. In conclusion, dietary fish oil mitigated chronic alcohol-induced liver injury primarily through DHA by upregulating Retsat and downstream genes associated with oxidoreductase function and lipid metabolism.
Collapse
Affiliation(s)
- Yuxuan Zhang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Kaixin Pan
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Angcheng Xu
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Shuzhen Sun
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Qingling Huang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Yicheng Wang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Hao Wang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Qiang Han
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Jiaomei Li
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China.
| |
Collapse
|
6
|
Direksunthorn T, Abdelgawwad El-Sehrawy AAM, Hjazi A, Obaidur Rab S, Suliman Maashi M. The association between lifelines diet score and metabolic associated fatty liver disease: a case-control study. Front Nutr 2025; 12:1569814. [PMID: 40444252 PMCID: PMC12119271 DOI: 10.3389/fnut.2025.1569814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Adherence to a healthy dietary pattern is a fundamental recommendation for the prevention of Metabolic Associated Fatty Liver Disease (MAFLD); however, conclusive evidence regarding the optimal dietary pattern remains elusive. Objectives The Lifelines Diet Score (LLDS) is a novel, evidence-based scoring system designed to evaluate diet quality. However, despite the extensive research on dietary patterns and liver health, the specific relationship between the LLDS and MAFLD remains underexplored. This study aims to investigate the association between LLDS and MAFLD, providing insights into how dietary adherence, as measured by LLDS, may influence the risk and prevalence of MAFLD. Methods This case-control study enrolled 215 individuals who had recently been diagnosed with MAFLD and 430 healthy controls at King Khalid University Hospital. All participants were aged between 20 and 60 years, with data collection occurring from February 2023 to January 2025. The dietary intake of the participants was assessed through the utilization of a validated semi-quantitative food frequency questionnaire, which comprised a total of 168 distinct food items. Logistic regression was used to estimate the association between LLDS and MAFLD. Results Out of 645 participants, 215 newly diagnosed MAFLD patients and 430 healthy controls were analyzed. After stratifying participants based on LLDS tertiles, those in the highest LLDS group had a 78% lower odds of MAFLD than those in the lowest tertile (odds ratio (OR): 0.22; 95% Confidence interval (CI): 0.12-0.36, p for trend <0.001). The association remained robust even after adjustment for major confounders. These findings highlight a novel and robust association between LLDS and MAFLD, providing evidence for dietary pattern assessment in liver health research. Conclusion Our study strengthens the evidence that adherence to a healthy dietary pattern (as measured by LLDS) is associated with a lower MAFLD risk, even after accounting for major confounders. However, further research integrating genetic and molecular data is needed to refine personalized dietary recommendations for MAFLD prevention.
Collapse
Affiliation(s)
- Thanyaporn Direksunthorn
- School of Medicine, Walailak University, Tha Sala District, Surat Skin Clinic, Surat Thani, Thailand
| | | | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Safia Obaidur Rab
- Central Labs, King Khalid University, AlQura’a, Abha, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Marwah Suliman Maashi
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit at King Fahd Medical Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Zhou R, Zhang X, Liu X, Huang R, Wang Y, Xia D, Li X, Wu Y, Shi Y. Association between dietary amino acid intake and the risk of metabolic dysfunction-associated steatotic liver disease. J Adv Res 2025:S2090-1232(25)00349-2. [PMID: 40381910 DOI: 10.1016/j.jare.2025.05.029] [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: 11/17/2024] [Revised: 04/26/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
INTRODUCTION Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is becoming the major chronic liver disease, and diet plays crucial role in MASLD prevention. OBJECTIVES The study aimed to explore the association between dietary amino acids with MASLD risk. METHODS Utilizing data from the U.S. NHANES (2017-2020) and UK Biobank (2006-2010), we investigated the association between dietary intake of combined or specific amino acids, and the risk of MASLD, severe MASLD and MASLD-related events. And we identified MASLD-risk amino acid intake patterns and their primary food sources. RESULTS The study included 5,568 participants from the U.S. NHANES and 48,261 from the UK Biobank, with MASLD prevalence of 34.3 % and 28.4 %, respectively. In QGC model, each quartile increase in combined amino acid intake was associated with a higher MASLD risk in the U.S. NHANES (aOR = 1.17, 95 % CI: 1.01-1.37, P = 0.035) and UK Biobank (aOR = 1.07, 95 % CI: 1.002-1.15, P = 0.042). Increased lysine intake was particularly linked to elevated MASLD risk in U.S. NHANES (aOR = 1.49, 95 % CI: 1.08-2.05, P = 0.023) and UK Biobank (aOR = 1.12, 95 % CI: 1.01-1.24, P = 0.032). Substituting lysine with other amino acids was associated with reduced MASLD risk. Lysine intake was also associated with a higher risk of severe MASLD (aHR = 1.13, 95 % CI: 1.04-1.23, P = 0.002) but not liver cirrhosis or HCC. The 'Lys-Met' pattern was identified as a MASLD-risk pattern, with red meats being a representative food source. CONCLUSION Lysine plays a major role in the association between amino acid intake and risk of MASLD. Dietary patterns rich in lysine, such as red meat, could be key targets for MASLD prevention strategies.
Collapse
Affiliation(s)
- Ruoqi Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinrong Zhang
- Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Xinxin Liu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Huang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Yuwei Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health and The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
8
|
Losasso MR, Parussolo MLC, Oliveira Silva A, Direito R, Quesada K, Penteado Detregiachi CR, Bechara MD, Méndez-Sánchez N, Abenavoli L, Araújo AC, de Alvares Goulart R, Guiger EL, Fornari Laurindo L, Maria Barbalho S. Unraveling the Metabolic Pathways Between Metabolic-Associated Fatty Liver Disease (MAFLD) and Sarcopenia. Int J Mol Sci 2025; 26:4673. [PMID: 40429815 PMCID: PMC12111209 DOI: 10.3390/ijms26104673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/10/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a public health concern that is constantly expanding, with a fast-growing prevalence, and it affects about a quarter of the world's population. This condition is a significant risk factor for cardiovascular, hepatic, and oncologic diseases, such as hypertension, hepatoma, and atherosclerosis. Sarcopenia was long considered to be an aging-related syndrome, but today, it is acknowledged to be secondarily related to chronic diseases such as metabolic syndrome, cardiovascular conditions, and liver diseases, among other comorbidities associated with insulin resistance and chronic inflammation, besides inactivity and poor nutrition. The physiopathology involving MAFLD and sarcopenia has still not been solved. Inflammation, oxidative stress, mitochondrial dysfunction, and insulin resistance seem to be some of the keys to this relationship since this hormone target is mainly the skeletal muscle. This review aimed to comprehensively discuss the main metabolic and physiological pathways involved in these conditions. MAFLD and sarcopenia are interconnected by a complex network of pathophysiological mechanisms, such as insulin resistance, skeletal muscle tissue production capacity, chronic inflammatory state, oxidative stress, and mitochondrial dysfunction, which are the main contributors to this relationship. In addition, in a clinical analysis, patients with sarcopenia and MAFLD manifest more severe hepatitis fibrosis when compared to patients with only MAFLD. These patients, with both disorders, also present clinical improvement in their MAFLD when treated for sarcopenia, reinforcing the association between them. Lifestyle changes accompanied by non-pharmacological interventions, such as dietary therapy and increased physical activity, undoubtedly improve this scenario.
Collapse
Affiliation(s)
- Marina Ribas Losasso
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Maria Luiza Cesto Parussolo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Antony Oliveira Silva
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Claudia Rucco Penteado Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Elen Landgraf Guiger
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, SP, Brazil
- Research Coordinator, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| |
Collapse
|
9
|
Zhang S, Zhao R, Wang R, Lu Y, Xu M, Lin X, Lan R, Zhang S, Tang H, Fan Q, Yang J, Liu L, Xu J. Weissella viridescens Attenuates Hepatic Injury, Oxidative Stress, and Inflammation in a Rat Model of High-Fat Diet-Induced MASLD. Nutrients 2025; 17:1585. [PMID: 40362894 PMCID: PMC12073722 DOI: 10.3390/nu17091585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disorder globally. Probiotic supplementation has shown promise in its prevention and treatment. Although Weissella viridescens, a lactic acid bacterium with immunomodulatory effects, has antibacterial and anti-inflammatory activities, there is a lack of direct evidence for its role in alleviating MASLD. This study aimed to investigate the protective effects of W. viridescens strain Wv2365, isolated from healthy human feces, in a high-fat diet (HFD)-induced rat model of MASLD. Methods: Rats were randomly assigned to a normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with W. viridescens Wv2365 (Wv2365) groups. All groups were fed their respective diets for 8 weeks. During this period, the NC and HFD groups received a daily oral gavage of PBS, while the Wv2365 group received a daily oral gavage of Wv2365. Results: Wv2365 supplementation significantly reduced HFD-induced body weight gain, improved NAFLD activity scores, alleviated hepatic injury, and restored lipid metabolism. A liver transcriptomic analysis revealed the downregulation of inflammation-related pathways, along with decreased serum levels of TNF-α, IL-1β, IL-6, MCP-1, and LPS. Wv2365 also activated the Nrf2/HO-1 antioxidant pathway, enhanced hepatic antioxidant enzyme activities and reduced malondialdehyde levels. A gut microbiota analysis showed the enrichment of beneficial genera, including Butyricicoccus, Akkermansia, and Blautia. Serum metabolomic profiling revealed increased levels of metabolites including indole-3-propionic acid, indoleacrylic acid, and glycolithocholic acid. Conclusions: Wv2365 attenuates hepatic injury, oxidative stress, and inflammation in a rat model of high-fat-diet-induced MASLD, supporting its potential as a probiotic candidate for the modulation of MASLD.
Collapse
Affiliation(s)
- Shuwei Zhang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiqing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruoshi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yao Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mingchao Xu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang 050010, China
| | - Xiaoying Lin
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Suping Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Huijing Tang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qianhua Fan
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Liyun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Jianguo Xu
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| |
Collapse
|
10
|
Mailloux RJ. Targeted Redox Regulation α-Ketoglutarate Dehydrogenase Complex for the Treatment of Human Diseases. Cells 2025; 14:653. [PMID: 40358176 PMCID: PMC12071522 DOI: 10.3390/cells14090653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
α-ketoglutarate dehydrogenase complex (KGDHc) is a crucial enzyme in the tricarboxylic acid (TCA) cycle that intersects monosaccharides, amino acids, and fatty acid catabolism with oxidative phosphorylation (OxPhos). A key feature of KGDHc is its ability to sense changes in the redox environment through the reversible oxidation of the vicinal lipoic acid thiols of its dihydrolipoamide succinyltransferase (DLST; E2) subunit, which controls its activity and, by extension, OxPhos. This characteristic inculcates KGDHc with redox regulatory properties for the modulation of metabolism and mediating of intra- and intercellular signals. The innate capacity of KGDHc to participate in the regulation of cell redox homeodynamics also occurs through the production of mitochondrial hydrogen peroxide (mtH2O2), which is generated by the dihydrolipoamide dehydrogenase (DLD; E3) downstream from the E2 subunit. Reversible covalent redox modification of the E2 subunit controls this mtH2O2 production by KGDHc, which not only protects from oxidative distress but also modulates oxidative eustress pathways. The importance of KGDHc in modulating redox homeodynamics is underscored by the pathogenesis of neurological and metabolic disorders that occur due to the hyper-generation of mtH2O2 by this enzyme complex. This also implies that the targeted redox modification of the E2 subunit could be a potential therapeutic strategy for limiting the oxidative distress triggered by KGDHc mtH2O2 hyper-generation. In this short article, I will discuss recent findings demonstrating KGDHc is a potent mtH2O2 source that can trigger the manifestation of several neurological and metabolic diseases, including non-alcoholic fatty liver disease (NAFLD), inflammation, and cancer, and the targeted redox modification of the E2 subunit could alleviate these syndromes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- School of Human Nutrition, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Quebec, QC H9X 3V9, Canada
| |
Collapse
|
11
|
Wu F, Zheng F, Li X, Wu D, Li H, Zeng Y, Tang Y, Liu S, Li A. Association between non-skimmed milk consumption and metabolic dysfunction-associated fatty liver disease in US adults: insights from NHANES data. BMC Gastroenterol 2025; 25:270. [PMID: 40251472 PMCID: PMC12007357 DOI: 10.1186/s12876-025-03834-x] [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: 01/30/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Previous studies on the association between non-skimmed milk consumption and non-alcoholic fatty liver disease (NAFLD) have reported inconsistent findings, with some suggesting an increased risk and others indicating a protective effect. Moreover, as the research focus has shifted globally from NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD), there remains limited evidence on the relationship between non-skimmed milk consumption and MAFLD. The objective of this cross-sectional study was to investigate this association using data from the National Health and Nutrition Examination Survey (NHANES). METHODS In this U.S. population-based study, adults with complete information on non-skimmed milk consumption and MAFLD diagnosis from the 2017-March 2020 Pre-Pandemic NHANES were included. MAFLD was defined using the controlled attenuation parameter (CAP). The association between non-skimmed milk consumption and MAFLD was assessed using weighted multivariable logistic regression, adjusting for potential confounders. Subgroup and sensitivity analyses were conducted to evaluate effect modifications and robustness. RESULTS The study involved 3,758 participants in total, 1,423 (37.87%) of whom had MAFLD according to the diagnosis. Frequent non-skimmed milk consumption was independently associated with higher MAFLD risk. Compared to the "Rarely" group (< 1 time/week), the "Sometimes" group (≥ 1 time/week but < 1 time/day) had an odds ratio (OR) of 1.67 (95% CI: 1.32-2.12, P = 0.004), and the "Often" group (≥ 1 time/day) had an OR of 1.36 (95% CI: 1.06-1.75, P = 0.046). Stratified analysis revealed that the association was significantly modified by education level (P for interaction = 0.010), with a stronger association observed among participants with higher education levels. Sensitivity analysis yielded consistent results, further supporting the robustness of the association. CONCLUSION Our findings suggest a significant association between frequent non-skimmed milk consumption and risk of MAFLD, particularly in highly educated individuals. These results highlight the importance of dietary modifications, specifically reducing non-skimmed milk intake, as a potential preventive strategy for MAFLD, especially in high-risk populations.
Collapse
Affiliation(s)
- Futao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuying Zheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danzhu Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Honghao Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yingyi Zeng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Gastroenterology, Zhuhai Peoples Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China.
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Wang Q, Chen R, Chen S, Wei B, Liu C, Jiang Z. Exploring the association between dietary indices and metabolic dysfunction-associated steatotic liver disease: Mediation analysis and evidence from NHANES. PLoS One 2025; 20:e0321251. [PMID: 40245006 PMCID: PMC12005519 DOI: 10.1371/journal.pone.0321251] [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: 01/15/2025] [Accepted: 02/27/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The association between dietary indices and metabolic dysfunction-associated steatotic liver disease (MASLD) has shown inconsistent results in previous studies. Additionally, the potential mediating variables linking dietary quality to MASLD have not been adequately explored. METHODS We analyzed data from 6,369 participants in the National Health and Nutrition Examination Survey (NHANES) 2007-2018. Three dietary indices-Healthy Eating Index (HEI), Energy-adjusted Dietary Inflammatory Index (EDII), and Composite Dietary Antioxidant Index (CDAI)-were evaluated for their associations with MASLD using logistic regression models adjusted for a comprehensive range of covariates. Mediation analysis was performed to evaluate the roles of potential mediators from four domains: insulin resistance (homeostatic model assessment of insulin resistance, HOMA-IR; metabolic score for insulin resistance, METS-IR), systemic inflammation (systemic inflammatory response index, SIRI; systemic immune-inflammation index, SII), obesity or visceral fat distribution (a body shape index, ABSI; body roundness index, BRI), and oxidative stress (Gamma-Glutamyltransferase, GGT; Bilirubin; Uric Acid). RESULTS After adjusting for all covariates, only HEI showed a consistent inverse association with MASLD, while EDII and CDAI showed no significant associations. Mediation analysis identified METS-IR, HOMA-IR, BRI, and ABSI as significant mediators in the relationship between HEI and MASLD, with mediation proportion accounting for 47.16%, 48.84%, 52.69%, and 13.84%, respectively. CONCLUSION Higher HEI is associated with a reduced risk of MASLD. The findings suggest that insulin resistance and visceral fat distribution partially mediate the relationship between HEI and MASLD, providing insights into potential mechanisms linking diet and liver health.
Collapse
Affiliation(s)
- Qiang Wang
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Rude Chen
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Shaohua Chen
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Bowen Wei
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Chunlan Liu
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Zongxing Jiang
- Xindu District People’s Hospital of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Polyzos SA, Targher G. Hepatic thyroid hormone receptor-β signalling: Mechanisms and recent advancements in the treatment of metabolic dysfunction-associated steatohepatitis. Diabetes Obes Metab 2025; 27:1635-1647. [PMID: 39658733 DOI: 10.1111/dom.16117] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/23/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024]
Abstract
The pharmacotherapy of metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, the metabolic dysfunction-associated steatohepatitis (MASH), remains a hot topic in research and a largely unmet need in clinical practice. As the first approval of a disease-specific drug, resmetirom, was regarded as a milestone for the management of this common liver disease, this comprehensive and updated review aimed to highlight the importance of the hepatic thyroid hormone (TH) receptor (THR)-β signalling for the treatment of MASH, with a special focus on resmetirom. First, the genomic and non-genomic actions of the liver-directed THR-β mediated mechanisms are summarized. THR-β has a key role in hepatic lipid and carbohydrate metabolism; disruption of THR-β signalling leads to dysmetabolism, thus promoting MASLD and possibly its progression to MASH and cirrhosis. In the clinical setting, this is translated into a significant association between primary hypothyroidism and MASLD, as confirmed by recent meta-analyses. An association between MASLD and subclinical intrahepatic hypothyroidism (i.e. a state of relatively low hepatic triiodothyronine concentrations, with circulating TH concentrations within the normal range) is also emerging and under investigation. In line with this, the favourable results of the phase 3 placebo-controlled MAESTRO trials led to the recent conditional approval of resmetirom by the US FDA for treating adults with MASH and moderate-to-advanced fibrosis. This conditional approval of resmetirom opened a new window to the management of this common and burdensome liver disease, thus bringing the global scientific community in front of new perspectives and challenges.
Collapse
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella (VR), Italy
| |
Collapse
|
14
|
Eslam M, Fan JG, Yu ML, Wong VWS, Cua IH, Liu CJ, Tanwandee T, Gani R, Seto WK, Alam S, Young DY, Hamid S, Zheng MH, Kawaguchi T, Chan WK, Payawal D, Tan SS, Goh GBB, Strasser SI, Viet HD, Kao JH, Kim W, Kim SU, Keating SE, Yilmaz Y, Kamani L, Wang CC, Fouad Y, Abbas Z, Treeprasertsuk S, Thanapirom K, Al Mahtab M, Lkhagvaa U, Baatarkhuu O, Choudhury AK, Stedman CAM, Chowdhury A, Dokmeci AK, Wang FS, Lin HC, Huang JF, Howell J, Jia J, Alboraie M, Roberts SK, Yoneda M, Ghazinian H, Mirijanyan A, Nan Y, Lesmana CRA, Adams LA, Shiha G, Kumar M, Örmeci N, Wei L, Lau G, Omata M, Sarin SK, George J. The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2025; 19:261-301. [PMID: 40016576 DOI: 10.1007/s12072-024-10774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/28/2024] [Indexed: 03/01/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) affects over one-fourth of the global adult population and is the leading cause of liver disease worldwide. To address this, the Asian Pacific Association for the Study of the Liver (APASL) has created clinical practice guidelines focused on MAFLD. The guidelines cover various aspects of the disease, such as its epidemiology, diagnosis, screening, assessment, and treatment. The guidelines aim to advance clinical practice, knowledge, and research on MAFLD, particularly in special groups. The guidelines are designed to advance clinical practice, to provide evidence-based recommendations to assist healthcare stakeholders in decision-making and to improve patient care and disease awareness. The guidelines take into account the burden of clinical management for the healthcare sector.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of MedicineSchool of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, Kaohsiung Medical University, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Global City, Philippines
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal MedicineHepatitis Research CenterGraduate Institute of Clinical Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tawesak Tanwandee
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rino Gani
- Department of Internal Medicine, Hepatobiliary Division, Dr. Cipto Mangunkusumo National General Hospital, Universitas Indonesia, Pangeran Diponegoro Road No. 71St, Central Jakarta, 10430, Indonesia
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Dan Yock Young
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Saeed Hamid
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Payawal
- Department of Medicine, Cardinal Santos Medical Center, Mandaluyong, Philippines
| | - Soek-Siam Tan
- Department of Hepatology, Selayang Hospital, Batu Caves, Malaysia
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Hang Dao Viet
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Jia-Horng Kao
- Graduate Institute of Clinical MedicineDepartment of Internal MedicineHepatitis Research CenterDepartment of Medical Research, National Taiwan University College of Medicine, National Taiwan University, National Taiwan University Hospital, 1 Chang-Te Street, 10002, Taipei, Taiwan
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Chia-Chi Wang
- Buddhist Tzu Chi Medical Foundation and School of Medicine, Taipei Tzu Chi Hospital, Tzu Chi University, Taipei, Taiwan
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Cairo, Egypt
| | - Zaigham Abbas
- Department of Hepatogastroenterology, Dr.Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | | | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Undram Lkhagvaa
- Department of Health Policy, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oidov Baatarkhuu
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ashok Kumar Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| | | | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - A Kadir Dokmeci
- Department of Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Institute of Clinical Medicine, School of Medicine, Taipei Veterans General Hospital, National Yang-Ming Chiao Tung University, No. 201, Section 2, Shipai RdNo. 155, Section 2, Linong St, Beitou District, Taipei City, 112, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jess Howell
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Clayton, VIC, 3008, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3165, Australia
| | - Jidong Jia
- Liver Research Center, Beijing Key Laboratory of Translational Medicine On Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, 11884, Egypt
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Central Clinical School, The Alfred, Monash University, Melbourne, Australia
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Aram Mirijanyan
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Leon A Adams
- Medical School, Faculty of Medicine and Health Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansoura, Egypt
| | - Manoj Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Necati Örmeci
- Department of Gastroenterohepatology, Istanbul Health and Technology University, Istanbul, Turkey
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - George Lau
- Humanity and Health Medical Group, Humanity and Health Clinical Trial Center, Hong Kong SAR, China
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
15
|
Khanmohammadi S, Fallahtafti P, Habibzadeh A, Ezzatollahi Tanha A, Alamdari AA, Fallahtafti P, Shafi Kuchay M. Effectiveness of body roundness index for the prediction of nonalcoholic fatty liver disease: a systematic review and meta-analysis. Lipids Health Dis 2025; 24:117. [PMID: 40148946 PMCID: PMC11948846 DOI: 10.1186/s12944-025-02544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Several anthropometric indices, such as body mass index and waist circumference, have been used as clinical screening tools for the prediction of nonalcoholic fatty liver disease (NAFLD). To further refine these clinical tools for NAFLD, the body roundness index (BRI) has recently been evaluated. In this systematic review and meta-analysis, the objective was to evaluate the relationship and predictive capability of the BRI in identifying NAFLD. METHODS A comprehensive search was conducted in PubMed, Embase, Web of Science, and Scopus up to December 31, 2024. Eligibility criteria included observational studies on adults (≥ 18 years old) with measured BRI and its association with NAFLD. The Joanna Briggs Institute tool was used for risk of bias assessment. Meta-analyses used random-effects models to pool data on mean difference, odds ratio, sensitivity, specificity, and the area under the curve (AUC), with heterogeneity and publication bias assessed. RESULTS Ten studies involving 59,466 participants were included. The pooled mean difference in BRI between the NAFLD and non-NAFLD groups was 1.73 (95% confidence interval [CI]: 1.31-2.15). The pooled sensitivity and specificity of BRI for diagnosing NAFLD were 0.806 and 0.692, respectively. The pooled AUC for BRI was 0.803 (95% CI: 0.775-0.830), indicating good diagnostic accuracy. Unlike subgroup analysis by country, subgroup analysis by sex showed no significant differences. Higher BRI values were associated with increased odds of NAFLD (pooled OR = 2.87, 95% CI: 1.39; 5.96). Studies provided mixed results on the predictive ability of BRI compared to other indices like body mass index, mostly favoring BRI over conventional indices. CONCLUSION BRI demonstrates a good diagnostic performance for NAFLD, suggesting it may be a valuable clinical tool for NAFLD assessment. Further research is necessary to validate these findings and strengthen the evidence base.
Collapse
Affiliation(s)
- Shaghayegh Khanmohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Parisa Fallahtafti
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Amir Ali Alamdari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parsa Fallahtafti
- School of Pharmacy and Pharmaceutical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Shafi Kuchay
- Divison of Endocrinology and Diabetes, Medanta the Medicity Hospital, Gurugram, Haryana, 122001, India
| |
Collapse
|
16
|
Lai S, Tang D, Feng J. Mitochondrial targeted therapies in MAFLD. Biochem Biophys Res Commun 2025; 753:151498. [PMID: 39986088 DOI: 10.1016/j.bbrc.2025.151498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a clinical-pathological syndrome primarily characterized by excessive accumulation of fat in hepatocytes, independent of alcohol consumption and other well-established hepatotoxic agents. Mitochondrial dysfunction is widely acknowledged as a pivotal factor in the pathogenesis of various diseases, including cardiovascular diseases, cancer, neurodegenerative disorders, and metabolic diseases such as obesity and obesity-associated MAFLD. Mitochondria are dynamic cellular organelles capable of modifying their functions and structures to accommodate the metabolic demands of cells. In the context of MAFLD, the excess production of reactive oxygen species induces oxidative stress, leading to mitochondrial dysfunction, which subsequently promotes metabolic disorders, fat accumulation, and the infiltration of inflammatory cells in liver and adipose tissue. This review aims to systematically analyze the role of mitochondria-targeted therapies in MAFLD, evaluate current therapeutic strategies, and explore future directions in this rapidly evolving field. We specifically focus on the molecular mechanisms underlying mitochondrial dysfunction, emerging therapeutic approaches, and their clinical implications. This is of significant importance for the development of new therapeutic approaches for these metabolic disorders.
Collapse
Affiliation(s)
- Sien Lai
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Dongsheng Tang
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Juan Feng
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| |
Collapse
|
17
|
Merheb C, Gerbal-Chaloin S, Casas F, Diab-Assaf M, Daujat-Chavanieu M, Feillet-Coudray C. Omega-3 Fatty Acids, Furan Fatty Acids, and Hydroxy Fatty Acid Esters: Dietary Bioactive Lipids with Potential Benefits for MAFLD and Liver Health. Nutrients 2025; 17:1031. [PMID: 40292496 PMCID: PMC11945187 DOI: 10.3390/nu17061031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common form of chronic liver disease, for which only resmetirom has recently received FDA approval. Prevention is crucial, as it can help manage and potentially reverse the progression of MAFLD to more severe stages. Omega-3 fatty acids, which are a type of polyunsaturated fatty acid (PUFA), have numerous beneficial effects in health and disease, including liver disease. Other bioactive lipids, such as furanic fatty acids (FuFA) and hydroxy fatty acid esters (FAHFA), have also demonstrated several benefits on relevant markers of liver dysfunction in animal and cell models. However, the effects of FAHFAs on hepatic steatosis are inconsistent, and studies on the impact of FuFAs in MAFLD are scarce. Further and more extensive research is required to better understand their role in liver health. The aim of this narrative review is to provide a brief overview of the potential effects of omega-3 fatty acids and other bioactive lipids, such as FuFAs and FAHFAs, on liver disease, with a focus on MAFLD.
Collapse
Affiliation(s)
- Camil Merheb
- Institute for Regenerative Medicine and Biotherapy (IRMB), University Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), F-34000 Montpellier, France; (C.M.); (S.G.-C.)
| | - Sabine Gerbal-Chaloin
- Institute for Regenerative Medicine and Biotherapy (IRMB), University Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), F-34000 Montpellier, France; (C.M.); (S.G.-C.)
| | - François Casas
- Dynamique du Muscle et Métabolisme (DMEM), University Montpellier, Institut National de Recherche pour L’agriculture, L’alimentation et L’environnement (INRAE), F-34295 Montpellier, France; (F.C.); (C.F.-C.)
| | - Mona Diab-Assaf
- Tumorigenesis Molecular and Anticancer Pharmacology, Faculty of Sciences-II, Lebanese University, Beyrouth 1500, Lebanon;
| | - Martine Daujat-Chavanieu
- Institute for Regenerative Medicine and Biotherapy (IRMB), University Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Montpellier, F-34000 Montpellier, France
| | - Christine Feillet-Coudray
- Dynamique du Muscle et Métabolisme (DMEM), University Montpellier, Institut National de Recherche pour L’agriculture, L’alimentation et L’environnement (INRAE), F-34295 Montpellier, France; (F.C.); (C.F.-C.)
| |
Collapse
|
18
|
Qiu X, Shen S, Jiang N, Lu D, Feng Y, Yang G, Xiang B. Adherence to the planetary health diet index and metabolic dysfunction-associated steatotic liver disease: a cross-sectional study. Front Nutr 2025; 12:1534604. [PMID: 40051965 PMCID: PMC11882404 DOI: 10.3389/fnut.2025.1534604] [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: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Backgrounds Adherence to the Planetary Health Diet Index (PHDI) has been shown to benefit both individual health and the planet. However, its impact on Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) remains unclear. This study aimed to investigate the relationship between PHDI adherence and the MASLD risk. Methods We analyzed a cohort of 15,865 adults (aged ≥18 years) using data from the National Health and Nutrition Examination Survey (NHANES, 2005-2018). The PHDI was derived from 24-h dietary assessments and comprised the scores of 15 food groups. Multivariate logistic regression was used to investigate the association between PHDI and MASLD, while restricted cubic spline (RCS) regression and threshold analysis were employed to explore potential non-linear relationship. Subgroup analyses were conducted to assess the influence of various demographic and clinical characteristics on the observed associations. Mediation analysis was performed to evaluate the indirect effect of PHDI on MASLD, and weighted quantile sum (WQS) regression was used to assess the influence of individual PHDI nutrients on MASLD. Results Among the cohort, 6,125 individuals were diagnosed with MASLD. Multivariate logistic regression revealed that a higher quintile of PHDI was significantly associated with reduced MASLD risk in the fully adjusted model (OR = 0.610, 95%CI 0.508-0.733, p < 0.001). Notably, nonlinear relationships between PHDI and MASLD risk were observed through RCS analysis (p = 0.002). Subgroup analyses indicated that PHDI was particularly effective in reducing MASLD risk among females, those with higher education attainment, and those living with a partner. WQS regression identified saturated fatty acids as the most significant factor contributing to MASLD risk (weight = 0.313). Additionally, BMI and waist circumference (81.47 and 87.66%, respectively) partially mediated the association between PHDI and MASLD risk, suggesting that the effect of PHDI on MASLD operates, in part, through its impact on BMI and waist circumference. The association between PHDI and MASLD remained robust across multiple sensitivity analyses. Conclusion Our findings indicate that adherence to PHDI is linked to a lower risk of MASLD, providing crucial insights for strategies aimed at mitigating the MASLD epidemic while simultaneously fostering environmental sustainability.
Collapse
Affiliation(s)
- Xin Qiu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shuang Shen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Nizhen Jiang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Donghong Lu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yifei Feng
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guodong Yang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| |
Collapse
|
19
|
Li J, Guo J, Yuen M, Yuen H, Peng Q. The comparative effects of ω-7 fatty acid-rich sea buckthorn oil and ω-3 fatty acid-rich DHA algal oil on improving high-fat diet-induced hyperlipidemia. Food Funct 2025; 16:1241-1253. [PMID: 39760431 DOI: 10.1039/d4fo04961f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
This study explores the therapeutic potential of ω-3 algal oil (rich in DHA) and ω-7 sea buckthorn oil (rich in palmitoleic acid) in addressing hyperlipidemia and associated metabolic disorders. These oils regulate lipid metabolism through the PPARγ-LXRα-ABCA1/ABCG1 signaling pathway, reducing cholesterol accumulation, oxidative stress, and inflammation. In high-fat diet-induced hyperlipidemic mice, supplementation with these oils significantly improved lipid profiles, alleviated hepatic steatosis, and promoted cardiovascular health. The combination of ω-3 and ω-7 fatty acids showed synergistic effects, offering greater efficacy compared to individual treatments. These findings suggest that algal and sea buckthorn oils could serve as dietary supplements or therapeutic interventions for managing hyperlipidemia, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases. This study highlights the potential of these oils as novel, natural solutions for metabolic health improvement.
Collapse
Affiliation(s)
- Jing Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Jiahan Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | | | | | - Qiang Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
20
|
Faienza MF, Baima J, Cecere V, Monteduro M, Farella I, Vitale R, Antoniotti V, Urbano F, Tini S, Lenzi FR, Prodam F. Fructose Intake and Unhealthy Eating Habits Are Associated with MASLD in Pediatric Obesity: A Cross-Sectional Pilot Study. Nutrients 2025; 17:631. [PMID: 40004960 PMCID: PMC11858415 DOI: 10.3390/nu17040631] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Fructose consumption in children is increasing, as is the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD). Despite evidence linking added sugars to metabolic syndrome, fructose's impact on liver disease in youth remains unclear, especially in pediatrics. Our study aimed to evaluate the role of fructose intake in metabolic and liver dysfunction in a cohort of pre-school children and adolescents with obesity. Methods: We recruited 41 children and adolescents with obesity (age range: 2.5-16 years, BMI SDS 2.6 ± 0.5 kg/m2). Clinical and biochemical parameters were assessed. Through ultrasound (US), MASLD, hepatorenal index (HRI), subcutaneous adipose tissue (scAT), and visceral adipose tissue (vAT) were assessed. Dietary intake was evaluated using the IDEFICS FFQ and a fructose-specific questionnaire. Results: Pubertal subjects had more scAT and vAT, higher insulin resistance, and higher liver fibrosis parameters than those prepubertal. MASLD was detected in 12 subjects, associated with higher scAT and vAT. Pubertal subjects had lower weekly fructose intake than prepubertal subjects (p < 0.02). However, they consumed less fructose from fruits (p < 0.04) and more from other sugars (p < 0.04) than younger children. Patients with MASLD reported higher fructose intake (p < 0.01), primarily from fruits (p < 0.003), likely due to misreporting, alongside higher consumption of unhealthy food, mainly rich in saturated fats. Conclusions: Fructose intake and unhealthy dietary habits were associated with MASLD in pre-school and adolescents with obesity. Advice to pay attention to fructose intake and foods rich in saturated fats is mandatory to decrease both obesity and MASLD. Further high-powered studies in any pediatric age and different geographical areas are needed to better evaluate the MASLD history.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, 70124 Bari, Italy;
| | - Jessica Baima
- Unit of Endocrinology, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (J.B.); (V.A.); (S.T.)
| | - Valentina Cecere
- Giovanni XXIII Pediatric Hospital, University of Bari “A. Moro”, 70124 Bari, Italy; (V.C.); (R.V.); (F.U.)
| | | | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
| | - Rossella Vitale
- Giovanni XXIII Pediatric Hospital, University of Bari “A. Moro”, 70124 Bari, Italy; (V.C.); (R.V.); (F.U.)
| | - Valentina Antoniotti
- Unit of Endocrinology, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (J.B.); (V.A.); (S.T.)
| | - Flavia Urbano
- Giovanni XXIII Pediatric Hospital, University of Bari “A. Moro”, 70124 Bari, Italy; (V.C.); (R.V.); (F.U.)
| | - Sabrina Tini
- Unit of Endocrinology, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (J.B.); (V.A.); (S.T.)
| | - Francesca Romana Lenzi
- Laboratory of Psychology and Social Processes in Sport, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy;
| | - Flavia Prodam
- Unit of Endocrinology, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (J.B.); (V.A.); (S.T.)
| |
Collapse
|
21
|
Hu L, Du H, Zhou Q, Liu C, Zhang T, Yuan M. Web of Science-Based Visualization of Metabolic Dysfunction-Associated Fatty Liver Disease in Pediatric and Adolescent Populations: A Bibliometric Study. Health Sci Rep 2025; 8:e70409. [PMID: 39897463 PMCID: PMC11779742 DOI: 10.1002/hsr2.70409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Background and Aims The prevalence of metabolism-associated fatty liver disease (MAFLD) in children is on the rise. This study employs visualization and analysis to evaluate the research implications, current advancements, and emerging trends in pediatric MAFLD, with the aim of elucidating its pathogenesis and informing the development of clinical treatment strategies. Methods Using visualization software, we conducted a visual analysis and mapping of the journal distribution, leading institutions, prominent authors, annual publication trends, and keyword frequencies among the 1179 scholarly articles retrieved from the Web of Science Core Collection for this study. Results The overall publication volume demonstrated an upward trend, with a total of 200 journals, contributions from 63 countries, 882 research institutions, and 5605 authors involved, including 84 who were identified as core authors. The main research team is led by Nobili, Valerio. The main research institutions are concentrated in Italy, the United States, and China. A total of 473 keywords were included, and the keywords with high frequency and medium centricity are insulin resistance, metabolic syndrome, children, steatohepatitis, adolescents, hepatic steatosis, nash, obesity, diagnosis, and fibrosis, which resulted in six keyword clusters. Conclusion MAFLD represents a significant public health concern. Research on children and adolescents with MAFLD continues to attract high interest. Noninvasive diagnostic methods, pathogenesis (intestinal microbiota research), disease prediction (gene research) are current research hotspots.
Collapse
Affiliation(s)
- Liangyu Hu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huarong Du
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - QianQian Zhou
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chunlei Liu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tiansong Zhang
- Department of Traditional Chinese Medicine, Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Min Yuan
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
22
|
Handu D, Stote K, Piemonte T. Evaluating Bioactive-Substance-Based Interventions for Adults with MASLD: Results from a Systematic Scoping Review. Nutrients 2025; 17:453. [PMID: 39940310 PMCID: PMC11820841 DOI: 10.3390/nu17030453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Objective: Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic condition affecting a broad population. This review aimed to identify and summarize the current evidence on bioactive-substance-based interventions for adults with MASLD, formerly known as nonalcoholic fatty liver disease (NAFLD), covering publications from 2000 to 2023. Methods: A search was conducted across six databases (MEDLINE, CINAHL, Cochrane CENTRAL, Cochrane Database of Systematic Reviews, Food Science Source, and SPORTDiscus) for randomized controlled trials and other study types (e.g., prospective cohort studies and systematic reviews), reflecting the scoping nature of this review. The search was limited to studies in adults (>18 years old), with an intervention of interest and at least one comparator group. Results: A total of 4572 articles were retrieved, with 201 full-text articles screened for eligibility. Of these, 131 primary studies and 49 systematic reviews were included in the scoping review. The most studied bioactive substances were Curcumin (Turmeric) (n = 25), Silymarin (Milk Thistle) (n = 17), Resveratrol (n = 10), Coffee (n = 7), Green Tea (n = 5), and Berberine (n = 5 each). Moreover, 46 studies reported on 36 other bioactive substances with 2 or fewer articles each. Among the included systematic reviews, 13 focused on Curcumin, 12 on Coffee or Tea, 10 on bioactive substance combinations, 6 on Resveratrol, and 2 each on Silymarin and Artichoke Leaf. The included studies showed substantial heterogeneity in reported outcomes, which primarily focused on hepatic health, body weight, adverse events, glycemic control, blood lipids, and body composition. Conclusions: This scoping review highlights a range of bioactive substances used in the treatment of MASLD. While evidence is abundant for bioactive substances like Curcumin and Silymarin, further research and synthesis of findings is necessary to establish the clinical efficacy of all bioactive substances.
Collapse
Affiliation(s)
- Deepa Handu
- Academy of Nutrition and Dietetics, Chicago, IL 60606, USA;
| | - Kim Stote
- Department of Allied Health Sciences, State University of New York, Empire State University, Saratoga Springs, NY 12866, USA;
| | - Tami Piemonte
- Academy of Nutrition and Dietetics, Chicago, IL 60606, USA;
| |
Collapse
|
23
|
Tonphu K, Mueangaun S, Lerkdumnernkit N, Sengking J, Tocharus J, Benjakul S, Mittal A, Tocharus C. Chitooligosaccharide-epigallocatechin gallate conjugate ameliorates lipid accumulation and promotes browning of white adipose tissue in high fat diet fed rats. Chem Biol Interact 2025; 406:111316. [PMID: 39577827 DOI: 10.1016/j.cbi.2024.111316] [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: 08/12/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
The prevalence of obesity has increased progressively worldwide. Obesity is characterized by excessive accumulation of fat in adipose tissues, leading to metabolic impairment. The anti-obese effects of chitooligosaccharide (COS) and epigallocatechin-3-gallate (EGCG) have been extensively clarified. This study aimed to investigate the effects and potential mechanisms of the COS-EGCG conjugate (CE) on anti-obesity, specifically by alleviating lipid accumulation and promoting the browning of white adipose tissue (WAT) in obese rats. Obesity as a consequence of a high-fat diet (HFD) was induced in male Wistar rats. The HFD was given for 16 weeks and the rats were then randomly subdivided into five groups namely: vehicle (control group), HFD plus CE at 150 mg/kg/day, HFD plus CE at 600 mg/kg/day, HFD plus COS at 600 mg/kg/day, and HFD plus atorvastatin at 10 mg/kg/day for 4 weeks. CE could reduce body weight, improve serum lipid profiles, and promote lipid metabolism via activation of AMP-activated protein kinase (AMPK) in WAT and enhance the processes of WAT browning by activating sirtuin 1 (Sirt 1), peroxisome proliferator-activated receptor-gamma coactivator (PGC1-α), and uncoupling the protein 1 (UCP1) signaling pathway. CE reduced obesity and promoted WAT browning in HFD-fed rats. Therefore, CE might be a new therapy for metabolic syndrome and obesity.
Collapse
Affiliation(s)
- Kanokrada Tonphu
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sirikul Mueangaun
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natcha Lerkdumnernkit
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jirakhamon Sengking
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, 90110, Thailand
| | - Ajay Mittal
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, 90110, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
24
|
Gillet R, Cerda-Drago TG, Brañes MC, Valenzuela R. Submicron Dispersions of Phytosterols Reverse Liver Steatosis with Higher Efficacy than Phytosterol Esters in a Diet Induced-Fatty Liver Murine Model. Int J Mol Sci 2025; 26:564. [PMID: 39859279 PMCID: PMC11766071 DOI: 10.3390/ijms26020564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Consumption of phytosterols is a nutritional strategy employed to reduce cholesterol absorption, but recent research shows that their biological activity might go beyond cholesterol reduction for the treatment of metabolic dysfunction-associated fatty liver disease (MAFLD), and novel phytosterol formulations, such as submicron dispersions, could improve these effects. We explored the therapeutic activity of phytosterols, either formulated as submicron dispersions of phytosterols (SDPs) or conventional phytosterol esters (PEs), in a mouse model of MAFLD. MAFLD was induced in mice by atherogenic diet (AD) feeding. The reversion of distorted serum and liver parameter values after a period of AD feeding was investigated after supplementation of the AD with SDPs, PEs, or a placebo (PT). Additionally, the metabolic parameters of fatty acid synthesis, fatty acid oxidation, and inflammation were studied to understand the mechanism of action of phytosterols. AD supplementation with SDPs was shown to reduce liver fat, along with showing a significant improvement in liver triglycerides (TGs), free fatty acids (FFAs), and liver cholesterol levels. These results were reinforced by the analyses of the liver steatosis scores, and liver histologies, where SDP intervention showed a consistent improvement. Treatment with PEs showed slighter effects in the same analyses, and no effects were observed with the PT treatment. Additionally, SDP intervention reversed, with a higher efficacy than PEs, the effect of AD on the serum levels of TGs, total- and LDL-cholesterol levels, and glucose levels. And, exceptionally, while SDP improved HDL-cholesterol serum levels, PEs did not show any effect on this parameter. We provide evidence for the therapeutical activity of phytosterols in MAFLD beyond the regulation of cholesterol levels, which is increased when the phytosterols are formulated as submicron dispersions compared to ester formulations.
Collapse
Affiliation(s)
- Raimundo Gillet
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Tomás G. Cerda-Drago
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - María C. Brañes
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| |
Collapse
|
25
|
Sorita GD, Favaro SP, Gambetta R, Ambrosi A, Di Luccio M. Macauba (Acrocomia ssp.) fruits: A comprehensive review of nutritional and phytochemical profiles, health benefits, and sustainable oil production. Compr Rev Food Sci Food Saf 2025; 24:e70097. [PMID: 39776255 DOI: 10.1111/1541-4337.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Macauba is an underexplored palm with significant potential for food-grade vegetable oil production. Its fruits yield two distinct sources of oil, the pulp and the kernel, each with its unique composition, emerging as a potential vegetable oil source with high competitiveness with well-established conventional oil sources. Besides the oil, macauba fruits are rich in essential nutrients, including proteins, minerals, vitamins, dietary fiber, and phytochemicals, with outstanding health benefits. Macauba processing generates valuable co-products, including the epicarp, pulp and kernel cakes, and endocarp, which have considerable potential for enhancing the macauba production chain. This review explores the nutritional and phytochemical profile of macauba, its health benefits, and the potential for exploiting its co-products. Innovative extraction methods and a comprehensive strategy for producing multiple products from macauba co-products are also highlighted as opportunities to achieve sustainable development goals and a circular economy in macauba fruit processing.
Collapse
Affiliation(s)
- Guilherme Dallarmi Sorita
- Laboratory of Membrane Processes (LABSEM), Department of Chemical and Food Engineering, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
- Brazilian Agricultural Research Corporation (EMBRAPA), Embrapa Agroenergia, Brasília, Federal District, Brazil
| | - Simone Palma Favaro
- Brazilian Agricultural Research Corporation (EMBRAPA), Embrapa Agroenergia, Brasília, Federal District, Brazil
| | - Rossano Gambetta
- Brazilian Agricultural Research Corporation (EMBRAPA), Embrapa Agroenergia, Brasília, Federal District, Brazil
| | - Alan Ambrosi
- Laboratory of Membrane Processes (LABSEM), Department of Chemical and Food Engineering, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Marco Di Luccio
- Laboratory of Membrane Processes (LABSEM), Department of Chemical and Food Engineering, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
26
|
He X, Zhang S, Bai Q, Pan M, Jiang Y, Liu W, Li W, Gong Y, Li X. Air pollution exposure and prevalence of non-alcoholic fatty liver disease and related cirrhosis: A systematic review and meta-analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117469. [PMID: 39657383 DOI: 10.1016/j.ecoenv.2024.117469] [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: 07/07/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND AND OBJECTIVE A systematic review and meta-analysis were used to investigate the relationship between air pollution exposure and the prevalence of non-alcoholic fatty liver disease (NAFLD) and its related cirrhosis. Through this study, we hope to clarify the potential public health risks of air pollution as an environmental exposure factor. METHODS Through a comprehensive and systematic search of the EMBASE, PubMed, Web of Science, and Cochrane library databases, studies published up to March 30, 2024, that met the eligibility criteria were identified. The meta-analysis aimed to determine the association between air pollution exposure and NAFLD risk. Subgroup analyses were conducted based on regional economic development after adjusting for confounding factors. The combined odds ratio (OR) was calculated, publication bias was assessed using funnel plots, and consideration was given to heterogeneity among study-specific relative risks. RESULTS This review included 14 observational studies (including 7 cohort studies and 7 cross-sectional studies) involving 43,475,41 participants. The pooled analysis showed that PM2.5, NOx, PM10, PM2.5-10, passive smoking, PM1, and air pollution from solid fuels were positively associated with the incidence and prevalence of NAFLD and its related cirrhosis. The risk ratios for PM2.5, NOx, PM10, PM2.5-10, passive smoking, and air pollution from solid fuels for NAFLD and its related cirrhosis were 1.33 (95 % CI: 1.25, 1.42), 1.19 (95 % CI: 1.14, 1.23), 1.27 (95 % CI: 1.05, 1.55), 1.05 (95 % CI: 1.00, 1.11), 1.53 (95 % CI: 1.12, 2.09), 1.50 (95 % CI: 0.86, 2.63), and 1.18 (95 % CI: 0.85, 1.63), respectively. In contrast, the risk ratio for O3 was 0.75 (95 % CI: 0.69, 0.83), suggesting that O3 may lower the incidence and prevalence of NAFLD and its related cirrhosis. We also conducted subgroup analyses based on the level of national development to examine the impact of PM2.5 on NAFLD and its related cirrhosis. The results showed that the risk of NAFLD and its related cirrhosis associated with PM2.5 in developing countries was 1.41 (95 % CI: 1.29, 1.53), which was higher than 1.20 (95 % CI: 1.12, 1.29) in developed countries. CONCLUSION The study findings show that PM2.5, NOx, PM10, PM2.5-10, passive smoking, PM1, and air pollution from solid fuels can increase an individual's risk of developing NAFLD and its related cirrhosis; while O3 can reduce the risk. In developing countries, the risk level of NAFLD and its related cirrhosis due to PM2.5 is higher than that in developed countries.
Collapse
Affiliation(s)
- Xingyi He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, PR China
| | - Shipeng Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, PR China
| | - Qinglin Bai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, PR China
| | - Moshen Pan
- School of Economics, Shanghai University of Finance and Economics, Shanghai 200433, PR China
| | - Yanjie Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No.157 Daming Road, Nanjing 210022, PR China
| | - Weiwei Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Wei Li
- Department of Intensive Care Medicine, Sichuan Hospital of Integrated Traditional Chinese and Western Medicine, Chengdu 610041, PR China
| | - Yuanyuan Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Xueping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
27
|
Karagiannakis DS, Stefanaki K, Paschou SA, Papatheodoridi M, Tsiodras S, Papanas N. Addressing the essentials of the recent guidelines for managing patients with metabolic dysfunction-associated steatotic liver disease. Hormones (Athens) 2024:10.1007/s42000-024-00625-z. [PMID: 39695010 DOI: 10.1007/s42000-024-00625-z] [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: 09/23/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of end-stage liver disease and liver transplantation in the Western world, with an approximate prevalence of 30% worldwide which is continuously rising. It is characterized by intrahepatic fat deposition along with at least one cardiometabolic risk factor, such as diabetes mellitus, obesity, hypertriglyceridemia, and hypertension. MASLD consists of a spectrum of liver diseases ranging from simple liver steatosis to steatohepatitis, liver fibrosis, and cirrhosis. Recently, the European Association for the Study of the Liver (EASL), the European Association for the Study of Diabetes (EASD), and the European Association for the Study of Obesity (EASO) released the latest guidelines regarding the management of patients with MASLD. This article highlights the critical points of these guidelines and emphasizes problematic issues that need further evaluation.
Collapse
Affiliation(s)
- Dimitrios S Karagiannakis
- Academic Department of Gastroenterology, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Katerina Stefanaki
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Margarita Papatheodoridi
- Academic Department of Gastroenterology, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Papanas
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, Thrace, Alexandroupolis, Greece
| |
Collapse
|
28
|
Li P, Wang T, Qiu H, Zhang R, Yu C, Wang J. 6-Gingerol Inhibits De Novo Lipogenesis by Targeting Stearoyl-CoA Desaturase to Alleviate Fructose-Induced Hepatic Steatosis. Int J Mol Sci 2024; 25:11289. [PMID: 39457074 PMCID: PMC11508832 DOI: 10.3390/ijms252011289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease (NAFLD), is a worldwide liver disease without definitive or widely used therapeutic drugs in clinical practice. In this study, we confirm that 6-gingerol (6-G), an active ingredient of ginger (Zingiber officinale Roscoe) in traditional Chinese medicine (TCM), can alleviate fructose-induced hepatic steatosis. It was found that 6-G significantly decreased hyperlipidemia caused by high-fructose diets (HFD) in rats, and reversed the increase in hepatic de novo lipogenesis (DNL) and triglyceride (TG) levels induced by HFD, both in vivo and in vitro. Mechanistically, chemical proteomics and cellular thermal shift assay (CETSA)-proteomics approaches revealed that stearoyl-CoA desaturase (SCD) is a direct binding target of 6-G, which was confirmed by further CETSA assay and molecular docking. Meanwhile, it was found that 6-G could not alter SCD expression (in either mRNA or protein levels), but inhibited SCD activity (decreasing the desaturation levels of fatty acids) in HFD-fed rats. Furthermore, SCD deficiency mimicked the ability of 6-G to reduce lipid accumulation in HF-induced HepG2 cells, and impaired the improvement in hepatic steatosis brought about by 6-G treatment in HFD supplemented with oleic acid diet-induced SCD1 knockout mice. Taken together, our present study demonstrated that 6-G inhibits DNL by targeting SCD to alleviate fructose diet-induced hepatic steatosis.
Collapse
Affiliation(s)
- Pan Li
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Tingting Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Hongmei Qiu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Ruoyu Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
| |
Collapse
|