1
|
Li W, Wang F, Li Z, Feng W, Huang H, Kwan MP, Tse LA. Lipid profile and non-alcoholic fatty liver disease detected by ultrasonography: is systemic inflammation a necessary mediator? Ann Med 2025; 57:2480250. [PMID: 40098359 PMCID: PMC11921154 DOI: 10.1080/07853890.2025.2480250] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/11/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
AIMS To examine the relationship between lipid profile and non-alcoholic fatty liver (NAFL), compare the predictive strengths of different lipid indicators to NAFL, and explore the possible mechanisms. METHODS Male workers from a baseline survey of a cohort of workers in southern China were included. Basic information was collected through face-to-face interviews. Plasma concentrations of fasting plasma glucose (FPG), total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) were determined using a blood biochemical analyzer. Liver sonography was used to identify NAFL cases. Regression models were used to calculate ORs, and examine the association between C-reactive protein (CRP) levels and lipid profiles. Restricted cubic spline regression with four knots was used to examine the dose-response relationship, and mediation analysis was employed to examine the mediation effect. RESULTS h Among the 4016 male workers, 829 (20.64%) were diagnosed with NAFL. Compared with normal lipid profile, individuals with abnormal lipid profile had higher prevalence of NAFL (OR=2.27, 95%CI: 1.85-2.79 for TG; OR=1.45, 95%CI: 1.03-2.04 for TC; OR=1.56, 95%CI: 1.21-2.02 for HDL; OR=1.65, 95%CI: 1.25-2.18 for LDL; OR=2.28, 95%CI: 1.87-2.77 for dyslipidaemia) after adjusting for potential confounders. Dose-response relationships were observed among TG, HDL, and NAFL. In addition, no significant mediation effect of C-reactive protein (CRP) was found in the association between lipid profiles and NAFL. CONCLUSIONS Abnormal TG, TC, HDL, and LDL levels were all positively associated with NAFL, while CRP has no mediating effect, and TG tended to be a better predictor of NAFL.
Collapse
Affiliation(s)
- Wenzhen Li
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Centre for Public Health and Primary Care (Shenzhen), Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute of the Chinese University of Hong Kong, Shenzhen, China
| | - Feng Wang
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhimin Li
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Wenting Feng
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Hongying Huang
- Institute of Occupational Medicine, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, China
| | - Mei-Po Kwan
- Department of Geography and Resource Management, The Chinese University of Hong Kong, Hong Kong SAS, China
- Institute of Space and Earth Information Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lap Ah Tse
- Jockey Club School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Centre for Public Health and Primary Care (Shenzhen), Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute of the Chinese University of Hong Kong, Shenzhen, China
- Institute of Space and Earth Information Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Santos-Sánchez G, Cruz-Chamorro I. Plant-derived bioactive peptides and protein hydrolysates for managing MAFLD: A systematic review of in vivo effects. Food Chem 2025; 481:143956. [PMID: 40147387 DOI: 10.1016/j.foodchem.2025.143956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) represents a growing health concern worldwide. Among the pursuit of therapeutic interventions, interest in natural bioactive compounds has intensified because of their potential hepatoprotective effects. This systematic review aims to evaluate the impact of plant-derived hydrolysates and peptides on MAFLD through the current literatures, encompassing their mechanisms of action. Key outcomes evaluated included changes in liver enzymes, liver lipid content, inflammation markers, and histopathological improvements. Preliminary findings suggest a potential beneficial effect of plant-derived hydrolysates and peptides on the improvement of MAFLD-related parameters, with mechanisms implicating antioxidant, anti-inflammatory, and lipid-lowering properties. This review highlights emerging evidence supporting the potential therapeutic role of plant-derived hydrolysates and peptides in the management of MAFLD. However, more well-designed clinical trials with larger sample sizes and longer durations are warranted to elucidate their efficacy, optimal dose, and long-term safety.
Collapse
Affiliation(s)
- Guillermo Santos-Sánchez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), 28049 Madrid, Spain.
| | - Ivan Cruz-Chamorro
- Facultad de Enfermería, Universidad de Castilla-La Mancha, 02071 Albacete, Spain.
| |
Collapse
|
3
|
Kim YJ, Lee JR, Kim MR, Jeong JA, Kim JJ, Jeong KW. Protein kinase-mediated inhibition of autophagy by palmitic acid in hepatocytes. Eur J Pharmacol 2025; 998:177528. [PMID: 40113068 DOI: 10.1016/j.ejphar.2025.177528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
Steatosis is characterized by an increase in free fatty acids, such as palmitic acid (PA), in hepatocytes and the accumulation of triglycerides in the liver. However, the role of intracellular autophagy in PA accumulation-induced hepatotoxicity is not clearly understood. Therefore, in this study, we investigated the effects of PA on autophagy in hepatocytes and its underlying mechanism of action. Treatment of HepG2 cells with PA induced a significant increase in intracellular p62 and LC3-II levels, suggesting inhibition of autophagy. Furthermore, PA inhibited autophagic flux in HepG2 cells, as monitored using GFP-RFP-LC3. Mechanistically, PA increased the phosphorylation of the Ser12 and Thr29 residues of LC3, which are autophagy inhibition markers, through protein kinase A (PKA) and protein kinase C (PKC) signaling. Finally, PKA and PKC inhibitors restored PA-induced autophagic flux inhibition, reduced intracellular lipid accumulation, and rescued the altered expression of lipogenic genes, such as SREBP-1c, in HepG2 cells. Thus, our study demonstrates the mechanism of autophagy inhibition by PA in hepatocytes and provides a potential therapeutic approach for preventing and treating hepatic steatosis.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jae Rim Lee
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Myeong Ryeo Kim
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jin Ah Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jung Ju Kim
- Autophagy Sciences Inc., Seoul, 08376, Republic of Korea
| | - Kwang Won Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea.
| |
Collapse
|
4
|
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
|
5
|
Almpanidou S, Vachliotis ID, Goulas A, Polyzos SA. The potential role of adipokines and hepatokines in age-related ocular diseases. Metabol Open 2025; 26:100365. [PMID: 40330313 PMCID: PMC12053655 DOI: 10.1016/j.metop.2025.100365] [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: 12/30/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Age-related ocular diseases, including diabetic retinopathy (DR), age-related macular degeneration (AMD), cataract and glaucoma may lead to visual impairment and even to blindness. Metabolic diseases, such as obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) have emerged as potential risk factors of age-related ocular diseases, especially DR. Visceral adiposity has been associated with increased risk of DR and AMD in most clinical studies, although body mass index has to-date provided conflicting association with DR and AMD. In addition, obesity is recognized as a risk factor of cataract and glaucoma. Similarly to obesity, MASLD appears to be associated with DR in patients with type 1 diabetes mellitus, but probably not in those with type 2 diabetes mellitus. A potential positive association between MASLD and AMD, glaucoma and cataract is supported by limited evidence to-date, thus needing further investigation. Altered secretion patterns of adipokines (adiponectin, leptin, lipocalin-2, resistin) and hepatokines [adropin, fetuin-A, fibroblast growth factor (FGF)-21, retinol binding protein (RBP)-4] seem to disrupt ocular homeostasis and contribute to the development of age-related ocular diseases in the context of obesity and MASLD. In this regard, novel adipokine-based and hepatokine-based therapies may be added to the treatment options for ocular diseases in the future. This narrative review aimed to summarize evidence on the interconnection of obesity and MASLD with age-related ocular diseases, with a specific focus on the roles of adipokines and hepatokines as mediators of these potential associations.
Collapse
Affiliation(s)
- Stavroula Almpanidou
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ilias D. Vachliotis
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
6
|
Lee NK, Lee Y, Shin DS, Park E, Paik HD. Probiotic Lactiplantibacillus plantarum Lb41 alleviates high-fat diet-induced nonalcoholic fatty liver disease in mice. Nutrition 2025; 134:112735. [PMID: 40147059 DOI: 10.1016/j.nut.2025.112735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is mainly related to genetics, obesity, insulin resistance, and type 2 diabetes. Probiotic Lactiplantibacillus plantarum Lb41 (Lb41) has not been reported to have hepatoprotective effects. Therefore, the aim of this study is to investigate the preventive effects of Lb41 against NAFLD in high-fat diet (HFD)-fed mice for preventing NAFLD. METHOD To induce fatty liver, the mice were given HFD for 5 weeks, followed by silymarin (200 mg/kg) or Lb41 (108 or 109 colony forming units/day) with the HFD for 7 weeks. After 12 weeks, body weight, histological change, serum and hepatic lipid profiles, etc. was performed compared to control and silymarin. RESULTS Lb41 had significantly reduced body weight (4.87 g) and serum lipids (triglycerides (77.64%), total cholesterol (67.53%), and low-density lipoprotein (40.50%) compared with the HFD group (P < 0.05). Lb41 significantly relieved HFD-associated hepatic injury by reducing aspartate transaminase (0.49-0.57 fold), alanine transaminase (0.49-0.51 fold), and alkaline phosphatase (0.76-0.90 fold) (P < 0.05). Additionally, they had decreased expression levels of peroxisome proliferator-activated receptor (PPAR) γ and sterol regulatory element-binding protein 1c and increased the expression levels of acyl-CoA oxidase, PPARα, carnitine palmitoyltransferase 1, acetyl CoA carboxylase 1, and fatty acid synthase in liver cells. Insulin and leptin levels decreased in the Lb41 treatment group compared with those in the HFD group. Meanwhile, adiponectin levels increased, similar to those in the normal diet group. CONCLUSION Based on these findings, Lb41 probiotics have possible hepatoprotective effects and could be used as functional food materials.
Collapse
Affiliation(s)
- Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Korea
| | - Yunjung Lee
- Department of Food and Nutrition, Kyungnam University, Changwon, Korea
| | - Da-Soul Shin
- Department of Food and Nutrition, Kyungnam University, Changwon, Korea
| | - Eunju Park
- Department of Food and Nutrition, Kyungnam University, Changwon, Korea.
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Korea.
| |
Collapse
|
7
|
Frías M, Chicano-Gálvez E, Rivero-Juárez A, Gordon A, Corona-Mata D, Moyano JM, Peralbo-Molina Á, Camacho Á, Pérez-Valero I, Del Mar Malagón M, Rivero A. Afamin and Apolipoprotein F Associated With Liver Steatosis From People Living With HIV: A Discovery Study. Aliment Pharmacol Ther 2025; 61:1767-1774. [PMID: 40159812 DOI: 10.1111/apt.70119] [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: 12/19/2024] [Revised: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Liver steatosis (LS) is a condition that is characterised by hepatic fat accumulation unrelated to significant alcohol consumption. This study explored the serum proteomic profile associated with LS in people living with HIV (PLWH). METHODS The study cohort comprised 266 PLWH, 21.1% and 78.9% of whom had LS and no LS, respectively. Serum samples were analysed using liquid chromatography coupled with mass spectrometry (LC-MS). RESULTS Among the 220 proteins detected, afamin (AFM) and apolipoprotein F (APOF) were identified as proteins associated with LS. Differential expression of AFM and APOF was observed in under- and normoweight patients, emphasising their potential as biomarkers in patients without overweight or obesity. CONCLUSIONS These findings suggest that the identified proteins could serve as promising biomarkers of LS in PLWH, paving the way for further investigations into the roles of these proteins in LS development in this unique population.
Collapse
Affiliation(s)
- Mario Frías
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Chicano-Gálvez
- Mass Spectrometry and Molecular Imaging Unit, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Antonio Rivero-Juárez
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Gordon
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Diana Corona-Mata
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - José María Moyano
- Department of Computer Science and Artificial Intelligence, University of Sevilla, Sevilla, Spain
| | - Ángela Peralbo-Molina
- Mass Spectrometry and Molecular Imaging Unit, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Ángela Camacho
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Pérez-Valero
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - María Del Mar Malagón
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Rivero
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Clinical Virology and Zoonoses Research Group, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Panganiban J, Kehar M, Ibrahim SH, Hartmann P, Sood S, Hassan S, Ramirez CM, Kohli R, Censani M, Mauney E, Cuda S, Karjoo S. Metabolic dysfunction-associated steatotic liver disease (MASLD) in children with obesity: An Obesity Medicine Association (OMA) and expert joint perspective 2025. OBESITY PILLARS 2025; 14:100164. [PMID: 40230708 PMCID: PMC11995806 DOI: 10.1016/j.obpill.2025.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Introduction This Obesity Medicine Association (OMA) Expert Joint Perspective examines steatotic liver disease (SLD), which is composed of metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction-associated steatohepatitis (MASH) in children with obesity. The prevalence of obesity is increasing, rates have tripled since 1963 from 5 % to now 19 % of US children affected in 2018. MASLD, is the most common liver disease seen in children, can be a precursor to the development of Type 2 Diabetes (T2DM) and is the primary reason for liver transplant listing in young adults. We must be vigilant in prevention and treatment of MASLD in childhood to prevent further progression. Methods This joint clinical perspective is based upon scientific evidence, peer and clinical expertise. The medical literature was reviewed via PubMed search and appropriate articles were included in this review. This work was formulated from the collaboration of eight hepatologists/gastroenterologists with MASLD expertise and two physicians from the OMA. Results The authors who are experts in the field, determined sentinel questions often asked by clinicians regarding MASLD in children with obesity. They created a consensus and clinical guideline for clinicians on the screening, diagnosis, and treatment of MASLD associated with obesity in children. Conclusions Obesity and the comorbidity of MASLD is increasing in children, and this is a medical problem that needs to be addressed urgently. It is well known that children with metabolic associated chronic disease often continue to have these chronic diseases as adults, which leads to reduced life expectancy, quality of life, and increasing healthcare needs and financial burden. The authors of this paper recommend healthy weight reduction not only through lifestyle modification but through obesity pharmacotherapy and bariatric surgery. Therefore, this guidance reviews available therapies to achieve healthy weight reduction and reverse MASLD to prevent progressive liver fibrosis, and metabolic disease.
Collapse
Affiliation(s)
| | - Mohit Kehar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Samar H. Ibrahim
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Hepatology & Nutrition, Rady Children’s Hospital San Diego, San Diego, CA, USA
| | - Shilpa Sood
- Division of Pediatric Gastroenterology, Boston Children's Health Physicians, New York Medical College, Valhalla, NY, USA
| | - Sara Hassan
- University of Texas Southwestern, Dallas, TX, United States
| | | | - Rohit Kohli
- Children's Hospital Los Angeles, CA, United States
| | - Marisa Censani
- Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Erin Mauney
- Tufts Medical Center, Boston, MA, United States
| | - Suzanne Cuda
- Alamo City Healthy Kids and Families, San Antonio, TX, United States
| | - Sara Karjoo
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
- University of South Florida, Tampa, FL, United States
- Florida State University, Tallahassee, FL, United States
| |
Collapse
|
9
|
Si G, Lv X, Ge Y, Zhang R, Hao D, Chen X, Wang C, Li Y, Li X, Yuan X. Age at menarche and risk of non-alcoholic fatty liver disease: Insights from the NHANES 2017-2020 and Mendelian randomization analyses. Exp Gerontol 2025; 204:112748. [PMID: 40194671 DOI: 10.1016/j.exger.2025.112748] [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: 12/21/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND This study aimed to explore the causal relationship between age at menarche (AAM) and non-alcoholic fatty liver disease (NAFLD), leveraging data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES) alongside Mendelian randomization (MR) analyses. Notably, this research represents the first attempt to link AAM to NAFLD using genetic methodologies, thereby providing novel insights into the interplay between these two conditions. METHODS Cross-sectional data from 2730 participants were analyzed using logistic regression to evaluate the association between AAM and NAFLD risk. A two-sample MR study was performed to investigate causal relationships, utilizing genetic data from large-scale genome-wide association studies (GWASs). The inverse variance weighted (IVW) method served as the primary MR analysis approach. RESULTS A significant negative association between AAM and NAFLD was found in Model 3 (OR = 0.85, 95 % CI: 0.74-0.97). Participants in the highest AAM quintile exhibited a 68 % reduction of NAFLD prevalence compared to those in the lowest AAM quintile (OR = 0.32, 95 % CI: 0.11-0.97). MR analysis confirmed a potential negative causal association (discovery: OR = 0.81, 95 % CI: 0.73-0.90; validation: OR = 0.80, 95 % CI: 0.66-0.96). CONCLUSIONS Our findings indicate a potential causal association between AAM and NAFLD, suggesting that early AAM may serve as a potential risk marker for NAFLD. This highlights the importance of incorporating AAM into clinical risk assessment tools and developing targeted prevention strategies for at-risk populations. Further research is needed to clarify the underlying mechanisms and to explore the potential benefits of early intervention.
Collapse
Affiliation(s)
- Guifei Si
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xiaopan Lv
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Yuxin Ge
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Rui Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Dongxiao Hao
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xuemei Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Changchun Wang
- Department of Internal Medicine, Feicheng Street Health Centre, Linyi 276000, Shandong, China
| | - Yuquan Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xiuping Li
- Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xuemin Yuan
- Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China.
| |
Collapse
|
10
|
Sun LY, Lu TY, Jin YL, Zhang WS, Xu L. Association between lifestyle factors and liver function parameters in the middle-aged and older population. BMC Public Health 2025; 25:1947. [PMID: 40420081 PMCID: PMC12105280 DOI: 10.1186/s12889-025-22260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 03/10/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Increasing evidence supports a higher risk of abnormal liver function parameters due to unfavorable lifestyles. We therefore explored the synergistic effects of various lifestyle factors on liver function. METHODS 8710 participants from Guangzhou Biobank Cohort Study were included. Five lifestyle factors including non-smoking, non-alcohol use, physically active, non-central and non-general obesity were assessed and a Healthy Lifestyle Index (HLI) (0-5, a higher score indicates healthier lifestyle) was generated. Multivariable linear regression was used to examine the association of HLI with liver function parameters, yielding regression coefficients (βs) and 95% confidence intervals (CIs). RESULTS A total of 8710 participants with an average age of 64.67 years (standard deviation = 6.07) were included. Of them, 71.65% were women. After adjusting for sex, age, education, family income, and comorbidities, compared with those with HLI of zero, those with HLI scores of 1, 2, 3, 4, and 5 showed a lower ALT level by -5.85 IU/L (95% CI: -10.73, -0.97), -9.97 IU/L (95% CI: -14.53, -5.42), -11.34 IU/L(95% CI: -15.86, -6.82), -12.81 IU/L (95% CI: -17.33, -8.30), and - 14.15 IU/L (95% CI: -18.68, -9.62), respectively (P for trend < 0.001), and a lower AST level by 1.82 IU/L (95% CI: -4.85,1.21), -3.74 IU/L (95% CI: -6.57, -0.91), -4.47 IU/L (95% CI: -7.28, -1.66), -4.69 IU/L (95% CI: -7.49, -1.88), and - 4.75 IU/L (95% CI: -7.57, -1.94), respectively (P for trend < 0.001). Similar trends were observed for a higher ALB level with higher healthy lifestyle scores (P for trend = 0.003). CONCLUSIONS A healthy lifestyle was associated with optimal liver function parameters, highlighting the importance of advocating for health-conscious behaviors to potentially mitigate the incidence of liver dysfunction.
Collapse
Affiliation(s)
- Lin Ye Sun
- School of Public Health, Sun Yat-sen University, No.74, 2nd Zhongshan Road, Guangzhou, Guangdong, China
| | - Ting Yu Lu
- School of Public Health, Sun Yat-sen University, No.74, 2nd Zhongshan Road, Guangzhou, Guangdong, China
| | - Ya Li Jin
- Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Wei Sen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China.
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, No.74, 2nd Zhongshan Road, Guangzhou, Guangdong, China.
- School of Public Health, The University of Hong Kong, Hong Kong, China.
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK.
| |
Collapse
|
11
|
Shi X, Tian Z, Wang Y, Cheng X, Zhang Y, Guo X, Zhang Y, Hu B, Liang C, Wang J, Tao F, Yang L. Associations of non‑essential metals and their mixture with non-alcoholic fatty liver disease in Chinese older adults. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2025; 47:228. [PMID: 40413681 DOI: 10.1007/s10653-025-02539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Research investigating the impact of the non-essential metal (NEM) mixture on non-alcoholic fatty liver disease (NAFLD) among the elderly is presently insufficient. This study investigated the relationships between individual NEMs, their mixtures, and NAFLD in elderly individuals residing in Chinese communities. METHODS The analysis included 2741 participants drawn from the baseline survey of a longitudinal study. Urinary concentrations of aluminum (Al), gallium (Ga), arsenic (As), cesium (Cs), barium (Ba), thallium (Tl), uranium (U), and cadmium (Cd) were quantified using inductively coupled plasma mass spectrometry (ICP-MS). NAFLD diagnosis was determined using abdominal ultrasound imaging. Logistic regression and restricted cubic spline (RCS) models were utilized to evaluate the relationships between individual NEMs and NAFLD. Additionally, Bayesian kernel machine regression (BKMR) and quantile-based computation regression (QGC) models were employed to assess the impact of the NEM mixture on NAFLD. RESULTS After adjusting for covariates, Tl was significantly associated with an increased likelihood of NAFLD (OR 1.26, 95% CI 1.10-1.44). Both RCS and BKMR models confirmed a linear relationship between urine Tl and the risk of NAFLD. Additionally, both BKMR and QGC models highlighted a significant connection between the NEMs mixture and NAFLD, identifying Tl as the primary driver. Significant interactions were observed between Tl and Ba, as well as between Tl and hypertension (Pinteraction = 0.055) and Tl and central obesity (Pinteraction = 0.008), collectively demonstrating synergistic impacts on NAFLD risk. CONCLUSIONS The NEM mixture is associated with a higher risk of NAFLD in Chinese old adults, with Tl as the primary contributor. Additional investigation is required to validate these findings and shed light on underlying biological pathways through which co-exposure to NEMs contribute to NAFLD.
Collapse
Affiliation(s)
- Xue Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, 230032, Anhui, China
| | - Ziwei Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, 230032, Anhui, China
| | - Yuan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
- Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xuqiu Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
| | - Yuantao Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
| | - Xianwei Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
| | - Yan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China
| | - Bing Hu
- Fuyang Center for Diseases Prevention and Control, Fuyang, 236069, Anhui, China
| | - Changliu Liang
- Fuyang Center for Diseases Prevention and Control, Fuyang, 236069, Anhui, China
| | - Jun Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Fangbiao Tao
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, 230032, Anhui, China
| | - Linsheng Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Center for Big Data and Population Health of IHM, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, 230032, Anhui, China.
| |
Collapse
|
12
|
Yang Q, Zhang Y, Zhang H, Yang Z, Feng Y, Ye B, Gong P, Qian G, Li D. Advanced N-glycoproteomics and proteomics approach revealed sexually dimorphic molecular signatures in primary mouse hepatocyte. Anal Bioanal Chem 2025:10.1007/s00216-025-05912-1. [PMID: 40410350 DOI: 10.1007/s00216-025-05912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Sexual dimorphism plays a critical role in disease pathophysiology, but the subtlety and complexity of these differences, along with a lack of precise comparative methods, hinder the advancement of precision medicine and drug development. This limitation is particularly evident in metabolic dysfunction-associated steatotic liver disease (MASLD), where sex-specific molecular mechanisms remain insufficiently understood. To address this gap, we employed an advanced integrative N-glycoproteomics and proteomics approach to systematically analyze sex-biased molecular signatures in primary mouse hepatocytes (PMHs) under healthy and MASLD conditions. Our analysis identified 280 sex-biased proteins and 39 sex-biased N-glycosites, and KEGG enrichment revealed that female-biased molecules were primarily involved in lipid metabolism, while male-biased molecules were associated with inflammation and cytoskeletal remodeling. A combined dataset of 302 sex-biased molecules was further analyzed using protein-protein interaction (PPI) analysis and Rc value calculations, resulting in the identification of 21 hub proteins and 2 hub N-glycosites as MASLD-associated sex-biased signatures. Notably, MASLD amplified proteomic sex differences while attenuating them in N-glycosylation. Western blot validation of key signatures, including female-biased MVK and male-biased LGALS3, highlighted distinct molecular adaptations between the sexes in MASLD progression. Our study introduced an advanced analytical framework for high-resolution comparative molecular profiling by integrating N-glycoproteomics with proteomics, providing valuable insights into sex-biased molecular signatures, enhancing preclinical model development, and advancing sex-specific therapeutic strategies in MASLD research and broader biological systems.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yong Zhang
- Department of Nephrology, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanruyu Feng
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Ninth People's Hospital of Zhengzhou, Zhengzhou, 45000, China
| | - Bengui Ye
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Medical College of Tibet University, Lhasa, 850002, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, China
| | - Guangsheng Qian
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Dapeng Li
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Koliaki C, Dalamaga M, Kakounis K, Liatis S. Metabolically Healthy Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): Navigating the Controversies in Disease Development and Progression. Curr Obes Rep 2025; 14:46. [PMID: 40387999 PMCID: PMC12089219 DOI: 10.1007/s13679-025-00637-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
PURPOSE OF REVIEW The natural course of metabolic dysfunction-associated steatotic liver disease (MASLD) in the population with metabolically healthy obesity (MHO) has not been adequately explored. In the present narrative review, we summarize the evidence regarding the association between MHO and MASLD prevalence, incidence and progression. RECENT FINDINGS Cross-sectional, population-based, cohort studies have shown an increased prevalence of hepatic steatosis and fibrosis in subjects with MHO compared with metabolically healthy non-obese individuals (MHNO). In large-scale longitudinal cohort studies among metabolically healthy subjects, increasing body mass index (BMI) has been found to be independently associated with an increased incidence of MASLD and progressive hepatic fibrosis over a mean follow-up period of 2.2-7.7 years. With regard to advanced MASLD, the prevalence of steatohepatitis and clinically significant liver fibrosis is lower in MHO compared with subjects with metabolically unhealthy obesity (MUO). The presence of MASLD has been proposed as a strong risk factor for metabolic health deterioration in MHO. Furthermore, subjects with MHO and MASLD display an elevated 10-year cardiovascular risk and a three-fold increased risk of incident diabetes compared with MHO without MASLD. MASLD may also predict the failure to convert from MUO to MHO after a weight loss intervention.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Propaedeutic Department of Internal Medicine and Diabetes Center, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, Athens, 11527, Greece.
| | - Maria Dalamaga
- Department of Biologic Chemistry, Medical School, National Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Kakounis
- Department of Gastroenterology, Hippokration General Hospital of Athens, Athens, Greece
| | - Stavros Liatis
- First Propaedeutic Department of Internal Medicine and Diabetes Center, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, Athens, 11527, Greece
| |
Collapse
|
14
|
Wang L, Yang Y, Sun H, Fei M. Magnoflorine alleviates nonalcoholic fatty liver disease by modulating lipid metabolism, mitophagy and inflammation. Prostaglandins Other Lipid Mediat 2025; 178:106997. [PMID: 40378915 DOI: 10.1016/j.prostaglandins.2025.106997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent liver condition associated with metabolic syndrome, often aggravated by inflammation and mitochondrial dysfunction. This study aims to explore the therapeutic potential of magnoflorine, an alkaloid with known anti-inflammatory properties, in ameliorating NAFLD by modulating mitochondrial autophagy and inhibiting the NLRP3 inflammasome. METHODS Male C57BL/6 J mice were fed a high-fat diet (HFD) for 16 weeks to induce NAFLD. Magnoflorine (5 and 10 mg/kg) was administered by gavage daily for 16 weeks. Liver and serum samples were analyzed for lipid profiles, inflammation markers, and autophagy-related proteins, and liver histology was examined to assess changes. RESULTS Magnoflorine treatment improved dyslipidemia in NAFLD mice, shown by decreased serum triglycerides, total cholesterol, and LDL-C, and increased HDL-C. Histological analysis showed reduced hepatic steatosis and inflammation, with less lipid droplet accumulation and hepatocyte ballooning. Western blot results indicated upregulation of Parkin and PINK1, and downregulation of NLRP3, ASC, and caspase-1, with lower serum IL-1β levels, reflecting reduced inflammation. CONCLUSIONS Magnoflorine offers a promising approach for mitigating NAFLD progression through modulating mitochondrial autophagy and inhibiting inflammation.
Collapse
Affiliation(s)
- Liming Wang
- Department of Gastroenterology, the Second People's Hospital of Hefei, Guangde Road, Hefei, Anhui 230000, China.
| | - Yan Yang
- Department of Gastroenterology, the Second People's Hospital of Hefei, Guangde Road, Hefei, Anhui 230000, China
| | - Haibing Sun
- Department of Gastroenterology, the Second People's Hospital of Hefei, Guangde Road, Hefei, Anhui 230000, China
| | - Mengxue Fei
- Department of Gastroenterology, the Second People's Hospital of Hefei, Guangde Road, Hefei, Anhui 230000, China
| |
Collapse
|
15
|
Vrentzos E, Pavlidis G, Korakas E, Kountouri A, Pliouta L, Dimitriadis GD, Lambadiari V. Nutraceutical Strategies for Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Path to Liver Health. Nutrients 2025; 17:1657. [PMID: 40431398 PMCID: PMC12113997 DOI: 10.3390/nu17101657] [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: 04/08/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
MASLD (Metabolic Dysfunction-Associated Steatotic Liver Disease) is a growing global concern. Nutraceuticals offer an appealing approach by targeting key mechanisms, such as oxidative stress, inflammation, lipid metabolism, and insulin resistance. This narrative review examines the role of various nutraceuticals in MASLD treatment, including silymarin, vitamin E, omega-3, curcumin, berberine, and coenzyme Q10. Some of them show promising biochemical and metabolic changes, while others produce conflicting results due to relevant studies' design and endpoints. To bridge the gap between research and reality, we summarize the data, create an interpretation heatmap, and develop a practical supplement guide. Regardless of their potential, nutraceuticals should be viewed as add-ons to lifestyle interventions rather than standalone treatments. Future research should focus on well-designed, long-term studies to prove efficacy, dosing, and combination strategies for personalized MASLD management.
Collapse
Affiliation(s)
- Emmanouil Vrentzos
- 4th Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.V.); (G.P.)
| | - George Pavlidis
- 4th Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.V.); (G.P.)
| | - Emmanouil Korakas
- 2nd Department of Internal Medicine, Research Unit and Diabetes Center, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.K.); (A.K.); (L.P.); (G.D.D.)
| | - Aikaterini Kountouri
- 2nd Department of Internal Medicine, Research Unit and Diabetes Center, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.K.); (A.K.); (L.P.); (G.D.D.)
| | - Loukia Pliouta
- 2nd Department of Internal Medicine, Research Unit and Diabetes Center, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.K.); (A.K.); (L.P.); (G.D.D.)
| | - George D. Dimitriadis
- 2nd Department of Internal Medicine, Research Unit and Diabetes Center, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.K.); (A.K.); (L.P.); (G.D.D.)
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, Research Unit and Diabetes Center, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.K.); (A.K.); (L.P.); (G.D.D.)
| |
Collapse
|
16
|
Sun X. Dapagliflozin ameliorates metabolic and hepatic outcomes in a mouse model of metabolic dysfunction-associated steatotic liver disease and diabetes. Acta Diabetol 2025:10.1007/s00592-025-02488-1. [PMID: 40353918 DOI: 10.1007/s00592-025-02488-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/04/2025] [Indexed: 05/14/2025]
Abstract
AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty liver disease (NAFLD), has become a great public healthcare burden and is closely associated with type 2 diabetes (T2D) and insulin resistance. However, there is no specific treatment for MASLD. Recent clinical findings have indicated a possible beneficial effect of sodium-glucose cotransporter 2 (SGLT2) on MASLD. This study aimed to investigate the effects of dapagliflozin (Dapa) on MASLD in T2D mice. METHODS Four-week-old ob/ob mice were fed with a high-fat diet (HFD) for 8 weeks and then randomly divided into two groups supplemented with Dapa or vehicle for another 12 weeks. C57BL/6J mice fed with a standard chow diet (CD) were used as the control group. Metabolic outcomes, liver pathology, lipidomics and insulin signaling were assessed. RESULTS We showed that Dapa reduced body weight and ameliorated hyperglycemia and fatty liver in obese diabetic ob/ob mice. Compared with vehicle, dapa improved the NAFLD activity score mainly by attenuating fat deposition. Importantly, Dapa decreased the expression levels of mRNAs and proteins related to fatty acid synthesis and increased the expression levels of β-oxidation-related factors. We also found that Dapa treatment improved insulin signaling by increasing PI3K and Akt phosphorylation. CONCLUSIONS Dapa protects mice from diet-induced weight gain and improves hepatic lipotoxicity and insulin resistance in diabetic MASLD mice. Our results revealed that Dapa has a therapeutic effect on MASLD and could be a potential drug candidate for the treatment of MASLD.
Collapse
Affiliation(s)
- Xiaoya Sun
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
García-Ulloa AC, Pérez-Peralta L, Lugo-Bautista K, Martínez-Sánchez VA, Mehta R, Hernández-Jiménez S, On behalf of CAIPaDi study group. Metabolic Comorbidities Among Relatives of Type 2 Diabetes Patients Stratified by Weight: Implications for Prevention and Care. Diabetes Metab Syndr Obes 2025; 18:1539-1549. [PMID: 40365576 PMCID: PMC12071750 DOI: 10.2147/dmso.s483171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/21/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Diabetes, affecting 18.3% of young adults in Mexico (6), is influenced by both genetic factors and shared unhealthy habits within families. Objective To determine the metabolic abnormalities in relatives of people with T2D, stratified by body mass index. Materials and Methods This observational, descriptive study was conducted at the Center for Comprehensive Care for Patients with Diabetes (CAIPaDi). The study involved relatives of participants with type 2 diabetes mellitus (T2DM), recruited between June 2017 and December 2020. The relatives were people without diabetes, including spouses, siblings, offspring, or close family members aged 18 to 65 who spent over four days a week with the patient. Exclusion criteria included relatives diagnosed with diabetes, smokers, or any individual from a patient-relative pair that was excluded. All participants underwent laboratory tests and body measurements. Relatives were classified into three groups based on body weight: normal weight, overweight, and obesity. The relatives attended four monthly visits and then annual evaluations. Ethical approval was obtained. Results The study enrolled 220 relatives of people with T2DM, 69% women, median age 49±12 years; 19.5% with normal weight, 40.4% overweight, and 40% with obesity. Prediabetes (39.4%), dyslipidemia (67.2%), and abnormal liver function tests (32.2%) were prevalent. Higher levels of triglycerides and LDL cholesterol were associated with increased risk for comorbid conditions. Anxiety and depression showed no significant differences across weight categories. Conclusion These results highlight the importance of overweight and obesity as factors associated with the presence of comorbidities and the metabolic syndrome. It is essential to implement strategies to promote healthy habits among family members of people with diabetes, especially in those who are overweight or obese to reduce the risk of developing future metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Ana Cristina García-Ulloa
- Centro de Atención Integral del Paciente con Diabetes (CAIPaDi), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Liliana Pérez-Peralta
- Centro de Atención Integral del Paciente con Diabetes (CAIPaDi), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Karla Lugo-Bautista
- Endocrinology and Lipid Metabolism Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Víctor A Martínez-Sánchez
- Internal Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Roopa Mehta
- Endocrinology and Lipid Metabolism Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sergio Hernández-Jiménez
- Centro de Atención Integral del Paciente con Diabetes (CAIPaDi), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - On behalf of CAIPaDi study group
- Centro de Atención Integral del Paciente con Diabetes (CAIPaDi), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Endocrinology and Lipid Metabolism Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Internal Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
18
|
Jiang R, Dai L, Xu X, Zhang Z. Multiple machine learning algorithms identify 13 types of cell death-critical genes in large and multiple non-alcoholic steatohepatitis cohorts. Lipids Health Dis 2025; 24:169. [PMID: 40340817 PMCID: PMC12060327 DOI: 10.1186/s12944-025-02588-5] [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: 01/09/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Dysregulated programmed cell death pathways mechanistically contribute to hepatic inflammation and fibrogenesis in non-alcoholic steatohepatitis (NASH). Identification of cell death genes may offer insights into diagnostic and therapeutic strategies for NASH. METHODS Data from multiple NASH cohorts were integrated, and 12 machine learning algorithms were applied to identify key dysregulated cell death-related genes and develop a binary classification model for NASH. Spearman's rank correlation coefficients quantified associations between these genes and clinical markers, immune infiltration profiles, and signature genes encoding pro-inflammatory mediators, metabolic regulators, and fibrotic drivers. Gene set enrichment analysis (GSEA) was performed to delineate the mechanistic underpinnings of these key genes. Consensus clustering analysis was then used to stratify patients with NASH into distinct phenotypic subgroups based on expression levels of these genes. RESULTS A NASH prediction model, developed using the random forest (RF) algorithm, demonstrated high diagnostic accuracy across multiple cohorts. Four key genes, enriched in lipid metabolism and inflammation pathways, were identified. Their transcriptional levels were significantly correlated with the non-alcoholic fatty liver disease activity score (NAS), hepatic inflammatory infiltration, molecular signatures of metabolic dysregulation (lipid homeostasis regulators), and fibrosis progression. These genes also enabled accurate classification of patients with NASH into clusters reflecting varying disease severity. CONCLUSIONS A binary classification model, developed using the RF algorithm, accurately identified patients with NASH. The four cell death genes, identified through 12 machine learning algorithms, represent potential biomarkers and therapeutic targets for NASH. These genes contribute to inflammation-related immune cell activation, lipid metabolism dysregulation, and liver fibrosis, highlighting the complex interplay between cell death and NASH progression.
Collapse
Affiliation(s)
- Renao Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province, 230022, China
| | - Longfei Dai
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province, 230022, China
| | - Xinjian Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province, 230022, China
| | - Zhen Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
19
|
Yi Y, Qin S, Ding S, Fang J. Polysaccharides in the medicine and food homology to combat obesity via gut-liver axis: A review of possible mechanisms. Int J Biol Macromol 2025; 312:144044. [PMID: 40345304 DOI: 10.1016/j.ijbiomac.2025.144044] [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: 01/15/2025] [Revised: 04/13/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Polysaccharides, as macromolecular carbohydrates present in various medicine and food homology, have gained growing recognition for their potential in combating obesity through multiple mechanisms. Their natural origin and favorable safety profile have made polysaccharides from medicine and food homology (PMFH) an area of significant research interest, particularly in the context of developing effective, safe, and sustainable interventions for obesity management. This review summarized the classification and biological properties of PMFH and then elucidated the pathological characteristics of obesity. We primarily focused on the effects of PMFHs on obesity, with particular attention to the potential mechanisms mediated through the gut-liver axis. These mechanisms encompassed the improvement of fat metabolism imbalances, manager of appetite and energy balance, adjustment of intestinal microbial imbalances, and alleviation of oxidative stress and inflammation. The findings provided critical theoretical insights and data to support the development of anti-obesity dietary and pharmaceutical products. In brief, this review outlined future research directions regarding the potential mechanisms underlying the anti-obesity effects of PMFH, particularly those involving the gut-liver axis.
Collapse
Affiliation(s)
- Yuhang Yi
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China
| | - Si Qin
- Laboratory of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China.
| |
Collapse
|
20
|
Bhattacharjee P, Fadlaoui A, Ryan CE, Carlson CB, Zhang D, Sunny NE. Induction of Fructose Mediated De Novo Lipogenesis Coexists with the Upregulation of Mitochondrial Oxidative Function in Mice Livers. J Nutr 2025:S0022-3166(25)00276-7. [PMID: 40334788 DOI: 10.1016/j.tjnut.2025.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Dysfunctional mitochondrial metabolism and sustained de novo lipogenesis (DNL) are characteristics of metabolic dysfunction-associated steatotic liver disease (MASLD), a comorbidity of obesity and type 2 diabetes. Fructose, a common sweetener and a potent inducer of lipogenesis, contributes to the etiology of MASLD. OBJECTIVES Our goal was to determine whether higher rates of DNL, through its biochemical relationships with mitochondria, can contribute to dysfunctional induction of oxidative networks in the liver. METHODS Male C57BL/6JN mice were given a low-fat (10% fat kcal, 49.9% corn starch kcal), high-fat (HF; 60% fat kcal), or HF/high-fructose diet (HF/HFr; 25% fat kcal, 34.9% fructose kcal) for 24 wk. In a follow-up study, mice on normal feed pellets were provided either 30% fructose in drinking water (FW) to induce hepatic DNL or regular water (NW) for 14 d. Hepatic mitochondria and liver tissue were used to determine oxygen consumption, reactive oxygen species (ROS) generation, tricarboxylic acid (TCA) cycle activity, and gene/protein expression profiles. RESULTS Hepatic steatosis remained similar between HF and HF/HFr fed mice livers. However, lipogenic and lipid oxidation gene expression profiles and the induction of TCA cycle metabolism were all higher (P ≤ 0.05) in HF/HFr livers. Under fed conditions, the upregulation of DNL in FW livers occurred in concert with higher mitochondrial oxygen consumption (basal; 1.7 ± 0.21 compared with 3.3 ± 0.14 nmoles/min, P ≤ 0.05), higher ROS (0.87 ± 0.09 compared with 1.25 ± 0.12 μM, P ≤ 0.05) and higher flux through TCA cycle components P ≤0.05. Furthermore, TCA cycle activity and lipid oxidation remained higher during fasting in the FW livers P ≤ 0.05. CONCLUSIONS Our results show that fructose administration to mice led to the concurrent induction of mitochondrial oxidative networks and DNL in the liver. Sustained induction of both DNL and mitochondrial oxidative function could accelerate cellular stress and metabolic dysfunction during MASLD.
Collapse
Affiliation(s)
- Parama Bhattacharjee
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| | - Ayeesha Fadlaoui
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| | - Caitlin E Ryan
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| | - Courtney B Carlson
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| | - Daoning Zhang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, United States
| | - Nishanth E Sunny
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States.
| |
Collapse
|
21
|
Trapp HM, Machado-Júnior PAB, Pimentel SK. INCIDENTAL HEPATIC STEATOSIS IDENTIFIED ON ULTRASOUND IN PATIENTS UNDERGOING CHOLECYSTECTOMY: HIGH PREVALENCE AND INSUFFICIENT INVESTIGATIVE AND CLINICAL MANAGEMENT. ARQUIVOS DE GASTROENTEROLOGIA 2025; 62:e24118. [PMID: 40332311 PMCID: PMC12052268 DOI: 10.1590/s0004-2803.24612024-118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/07/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Steatotic liver disease (SLD) affects about 1 billion people globally, making its proper management essential to prevent progression to more severe stages. OBJECTIVE The aim of this study was to evaluate medical management concerning hepatic steatosis incidentally identified by ultrasound in patients undergoing elective cholecystectomy. METHODS This observational, cross-sectional, and retrospective study included patients aged 18 years or older who underwent elective cholecystectomy at Hospital do Trabalhador, in Curitiba/PR, between 2018 and 2022. Patients with external ultrasound reports or incomplete data in their medical records were excluded. Medical records, laboratory tests, and ultrasound reports were analyzed to evaluate the prevalence of steatosis in these patients. RESULTS The study sample consisted of 355 patients, and 103 (29.01%) of them presented steatosis on ultrasound. Older age (P=0.0022), male sex (P=0.03009), higher body mass index (P<0.001), obesity (P<0.001), hypertension (P<0.001), dyslipidemia (P=0.0072), and elevated levels of oxaloacetic and pyruvic aminotransferases (P=0.02112) were associated with the presence of this finding. No action was taken regarding the presence of steatosis in 60.19% of patients. Approximately 39.81% had the finding recorded in their medical records, 6.80% received lifestyle change counseling, and 4.85% were investigated for the stage of steatosis. CONCLUSION A significant prevalence of hepatic steatosis was incidentally identified in the ultrasound of patients undergoing cholecystectomy. However, the approach to this finding was insufficient, highlighting the need for substantial improvements on its management and investigation.
Collapse
Affiliation(s)
- Heloísa Mello Trapp
- Universidade Federal do Paraná, Faculdade de Medicina, Curitiba, PR, Brasil
- Hospital do Trabalhador, Serviço de Cirurgia Geral, Curitiba, PR, Brasil
| | | | - Silvania Klug Pimentel
- Universidade Federal do Paraná, Faculdade de Medicina, Curitiba, PR, Brasil
- Hospital do Trabalhador, Serviço de Cirurgia Geral, Curitiba, PR, Brasil
| |
Collapse
|
22
|
Ma L, Jiang H, Qu N. Mendelian randomization analysis of smoking, BMI, and nonalcoholic fatty liver disease in European descent populations. Medicine (Baltimore) 2025; 104:e42308. [PMID: 40324243 PMCID: PMC12055159 DOI: 10.1097/md.0000000000042308] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 12/27/2024] [Accepted: 04/14/2025] [Indexed: 05/07/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver condition with a steadily increasing prevalence. Evidence indicates that both smoking and obesity are significant risk factors for NAFLD, yet the extent to which smoking influences NAFLD through weight gain remains unclear. This study aimed to dissect the intricate relationship between smoking, body mass index (BMI), and NAFLD using Mendelian randomization (MR) analysis. We leveraged data from 30 genome-wide association studies involving over 1.2 million individuals, from which 123 single nucleotide polymorphisms were selected as instrumental variables for smoking. BMI data were sourced from the Genetic Investigation of Anthropometric Traits (GIANT) consortium, encompassing more than 700,000 individuals, with 521 single nucleotide polymorphisms serving as instrumental variables. NAFLD data were obtained from multiple databases, including the eMERGE Network, UK Biobank, Estonian Biobank, and FinnGen, comprising 8434 cases and 770,180 controls. All participants in this study were of European ancestry. We first applied univariate MR analysis to assess the causal relationship between smoking, NAFLD, and BMI. Subsequently, multivariate MR was used to assess the effect of smoking on NAFLD after adjusting for BMI. The coefficient product method was used to calculate the mediating effect of BMI. Results found that both smoking and high BMI were able to increase the risk of NAFLD, with odds ratios of 1.83 (95% confidence interval [CI]: 1.31-2.55) and 1.58 (95% CI: 1.42-1.77), respectively. BMI mediated 73.3% (95% CI: 62.3%-80.5%) of the effect of smoking on NAFLD. The findings support weight control and the encouragement of smoking cessation, especially in obese populations, as strategies to reduce the risk of NAFLD.
Collapse
Affiliation(s)
- Lei Ma
- The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
- The First Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, China
| | - Haixing Jiang
- The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Nanfang Qu
- The First Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
23
|
Sun M, Sun H. Recent prevalence and trends of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) among US adolescents: 1999 to 2020. Pediatr Obes 2025; 20:e70003. [PMID: 39967492 DOI: 10.1111/ijpo.70003] [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: 07/17/2024] [Revised: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is understudied among US adolescents despite rising obesity rates. METHODS This study analysed the prevalence and trends of obesity and MASLD among US adolescents aged 12-17 using data from the National Health and Nutrition Examination Survey (NHANES). We developed a new screening model utilizing FibroScan-measured controlled attenuation parameter (CAP) scores, body measurements and blood chemistry data from 2017 to 2020 to assist in analysing MASLD trends from 1999 to 2020. RESULTS Between 2017 and 2020, the prevalence of obesity and MASLD was approximately 20%, with about 70% of obese adolescents affected by MASLD. The condition was more common in boys, particularly among Mexican American adolescents. Additionally, 97.2% of those with NAFLD also had MASLD. Adolescents with MASLD had significantly higher body weight, waist circumference, triglyceride levels and alanine transaminase (ALT) levels, along with lower high-density lipoprotein (HDL) cholesterol and an increased risk of liver fibrosis. Insufficient physical activity and poor diet quality were key risk factors for developing MASLD. From 1999 to 2020, the prevalence of MASLD rose significantly, paralleling the increasing rates of obesity. CONCLUSIONS The study underscores the pressing need to screen at-risk adolescents for metabolic issues associated with steatotic liver diseases, given the rising obesity rates among adolescents. The high overlap between MASLD and NAFLD diagnoses indicates that the transition from NAFLD to MASLD can be effectively integrated into paediatric practice.
Collapse
Affiliation(s)
- Michael Sun
- Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Hongbing Sun
- Nutrition, Biostatistics and Health Study, Department of Earth and Chemical Sciences, Rider University, Lawrenceville, New Jersey, USA
| |
Collapse
|
24
|
Tuğal Aslan D, Göktaş Z. The Therapeutic Potential of Theobromine in Obesity: A Comprehensive Review. Nutr Rev 2025; 83:859-868. [PMID: 39271172 PMCID: PMC11986327 DOI: 10.1093/nutrit/nuae122] [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] [Indexed: 09/15/2024] Open
Abstract
Obesity, characterized by chronic low-grade inflammation, is a significant health concern. Phytochemicals found in plants are being explored for therapeutic use, particularly in combating obesity. Among these, theobromine, commonly found in cocoa and chocolate, shows promise. Although not as extensively studied as caffeine, theobromine exhibits positive effects on human health. It improves lipid profiles, aids in asthma treatment, lowers blood pressure, regulates gut microbiota, reduces tumor formation, moderates blood glucose levels, and acts as a neuroprotective agent. Studies demonstrate its anti-obesity effects through mechanisms such as browning of white adipose tissue, activation of brown adipose tissue, anti-inflammatory properties, and reduction of oxidative stress. This study aims to suggest theobromine as a potential therapeutic agent against obesity-related complications.
Collapse
Affiliation(s)
- Dilem Tuğal Aslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Altindag, Ankara, Turkiye
| | - Zeynep Göktaş
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Altindag, Ankara, Turkiye
| |
Collapse
|
25
|
Liu X, Ling J, Wu Y, Zhao H, Hu Y, Yan Z, Zhu W, Yu P, Wang J, Zhang Y, Bucci T, Lip GYH. Association between metabolically healthy obesity and atrial fibrillation: A systematic review and meta-analysis of longitudinal studies. Diabetes Metab Syndr 2025; 19:103228. [PMID: 40306065 DOI: 10.1016/j.dsx.2025.103228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/12/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Obesity is not a single diagnosis, and the association of 'metabolically unhealthy' obesity with cardiovascular disease is well-described. However, the relationship between metabolically healthy obesity (MHO) and atrial fibrillation (AF) is still debated. OBJECTIVE Our objective is to investigate the association between MHO and the risk of AF. METHODS A comprehensive search of databases, including PubMed, EMBASE, Web of Science, and the Cochrane Library regarding longitudinal studies of MHO and risk of AF was performed. Random effects were used to pool the effect estimates. RESULTS Nine cohort studies comprising 4,250,557 participants were included. The pooled results revealed that individuals with MHO were associated with a greater incidence of AF than those with a metabolically healthy normal weight (HR: 1.34, 95 % CI: 1.26 to 1.42) with moderate certainty according to the Grading of Recommendations Assessment, Development, and Evaluation assessment. Individuals with MHO were associated with a lower risk of AF compared with participants with metabolically unhealthy obesity (RR: 0.48, 95 % CI: 0.36 to 0.64). Individuals with MHO were not significantly associated with the risk of AF as compared to metabolically unhealthy normal weight (HR: 1.04, 95 % CI: 0.89 to 1.22). CONCLUSION MHO is associated with a greater incidence of AF, highlighting the importance of weight reduction in individuals without metabolic disorders in reducing the risk of AF. REGISTRATION PROSPERO - registration number CRD42023432195.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yifan Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huilei Zhao
- Anesthesiology Department, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuzhe Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhiwei Yan
- Department of Sports Rehabilitation, College of Human Kinesiology, Shenyang Sport University, Shenyang, China
| | - Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Jinfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Yuling Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Tommaso Bucci
- Liverpool Centre of Cardiovascular Science at University of Liverpool, Liverpool JohnMoores University and Liverpool Heart and Chest Hospital, Liverpool, UK; Department of General and Specialized Surgery, Sapienza University of Rome, Rome, Italy
| | - Gregory Y H Lip
- Liverpool Centre of Cardiovascular Science at University of Liverpool, Liverpool JohnMoores University and Liverpool Heart and Chest Hospital, Liverpool, UK; Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
26
|
Wang K, Dong L, Wang X, Wang Z, Qiu X, Xu H, Xu X. Outcomes and risk factors for liver transplantation using steatotic grafts for hepatocellular carcinoma: a multicenter study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110061. [PMID: 40288219 DOI: 10.1016/j.ejso.2025.110061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION A growing number of steatotic grafts have been used in liver transplantation (LT), including hepatocellular carcinoma (HCC) patients. However, the impact of steatotic grafts on the prognosis of HCC recipients remains unclear. This study aims to evaluate the impact of steatotic graft in long-term prognosis for HCC recipients and development an algorithm for minimizing the risk of these grafts. MATERIALS AND METHODS The clinicopathologic data of HCC patients undergoing LT from 2003 to 2022 in the United Network for Organ Sharing database was analyzed. The disease-free survival (DFS) and overall survival (OS) of recipients were compared between non-steatotic (macrosteatosis <30 %) and steatotic (macrosteatosis ≥30 %) graft groups after propensity score matching (PSM). Interaction analysis was conducted to identify factors that amplified the negative impact of steatotic grafts on DFS. RESULTS A total of 8345 eligible HCC patients were included. Three factors exhibited significant interaction effect with steatotic grafts: cold ischemia time ≥6h (HR = 1.447; P = 0.023), donor body mass index ≥40 (HR = 1.771; P = 0.018) and recipient with non-alcoholic fatty liver disease (HR = 1.632; P = 0.032). Hazard Associated with Macrosteatotic Liver (HAML) score was created based on these three factors. In HAML ≥1 cohort, the DFS and OS of steatotic graft group were significantly reduced compared to non-steatotic graft group. But in HAML = 0 cohort, no significant differences in DFS and OS were observed between the two groups. CONCLUSIONS The risk of steatotic grafts in LT for HCC could be minimized through evaluating HAML score.
Collapse
Affiliation(s)
- Kai Wang
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Libin Dong
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xiaobo Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Zhoucheng Wang
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xun Qiu
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Hanzhi Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
27
|
Arasteh B, Hamzehzadeh S, Toutounchi KS, Nikniaz Z, Amini L, Alizadeh L. Association of ultrasound signs of sarcopenia with serum ferritin levels and hepatic indices like NFS and FIB-4 in NAFLD patients. BMC Gastroenterol 2025; 25:261. [PMID: 40234765 PMCID: PMC12001710 DOI: 10.1186/s12876-025-03773-7] [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: 07/02/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease is one of the most common chronic diseases all around the world, which significantly correlates with metabolic disorders and inflammatory cycles. Sarcopenia is a decrease in the mass of skeletal muscles interacting with factors such as inflammatory processes and chronic diseases. It can also lead to the aggravation of chronic diseases. METHOD The study population was randomly selected and entered into the research based on exclusion and inclusion criteria. Non-alcoholic fatty liver disease was confirmed in all members of the study population by ultrasound. Patients' serum ferritin level was assessed, and their NFS and Fib 4 scores were calculated. Sarcopenia was diagnosed by measuring the thickness of the rectus femoris by ultrasonography. The correlation between these variables was evaluated and analyzed by statistical software. RESULTS According to statistical analysis, there is a significant association between the serum ferritin level and sarcopenia (P-value < 0.001). Besides, there is a significant association between NFS, Fib4, and sarcopenia (P-value = 0.024, 0.000). CONCLUSION This research's results reflect the correlation between serum ferritin and sarcopenia; however, it cannot conclude a cause-and-effect relationship between these variables.
Collapse
Affiliation(s)
- Bahar Arasteh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Hamzehzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Zeinab Nikniaz
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Amini
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Alizadeh
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Liu Y, Wang R. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio as a predictor of NAFLD prevalence and steatosis severity. Sci Rep 2025; 15:12990. [PMID: 40234442 PMCID: PMC12000293 DOI: 10.1038/s41598-024-82012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/02/2024] [Indexed: 04/17/2025] Open
Abstract
Altered lipid metabolism is a crucial jeopardy cause for developing non-alcoholic fatty liver disease (NAFLD). Among various lipid metrics, the ratio of non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol (NHHR) has recently emerged as a promising indicator showing significant potential in predicting the prevalence of NAFLD. This study aimed to provide novel insights for the prevention of NAFLD by examining the relationship between NHHR and the prevalence of NAFLD. To identify the connection between NHHR and NAFLD, this study utilized descriptive analysis, multivariate logistic regression, and restricted cubic spline regression to investigate data collected by the National Health and Nutrition Examination Survey performed from 2017 to 2020. Furthermore, the connection between NHHR and the controlled attenuation parameter (CAP) was assessed using multiple linear regression, smoothed curve fitting, and threshold effect analysis. The NAFLD group showed higher NHHR levels than the non-NAFLD group (2.990 vs 2.240, P < 0.001). Multiple logistic and linear regression analyses indicated significant positive associations between NHHR and its quartiles with both the prevalence of NAFLD and CAP levels. Additionally, NHHR was positively associated with the prevalence of NAFLD in a linear dose-response relationship. Furthermore, smoothed curve fitting demonstrated a positive relationship between NHHR and CAP, with a threshold effect at an inflection point of 3.398. Higher NHHR levels were significantly associated with the prevalence of NAFLD and steatosis, and maintaining NHHR in the appropriate range may reduce these conditions.
Collapse
Affiliation(s)
- Yajie Liu
- Department of Spleen, Stomach, Liver and Gallbladder Diseases, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450099, China
| | - Ruilin Wang
- Department of Traditional Chinese Medicine and Liver Diseases, Fifth Medical Center, PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
29
|
Huang H, Liu Z, Ruan J, Fang Z, Xu C. Laparoscopically confirmed endometriosis and the risk of incident NAFLD: a prospective cohort study. Reprod Biol Endocrinol 2025; 23:55. [PMID: 40205408 PMCID: PMC11983926 DOI: 10.1186/s12958-025-01391-2] [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: 01/07/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND To investigate whether endometriosis is associated with the risk of incident nonalcoholic fatty liver disease (NAFLD). METHODS Data were retrieved from Nurses' Health Study II with participants followed up from 1995 to 2017. A total of 61,649 participants were included in this prospective cohort study. The exposure of this study was laparoscopically confirmed endometriosis. We performed Cox proportional hazard regression analyses to estimate the hazard ratio (HR) and 95% confidence interval (95% CI) of the association between endometriosis and NAFLD. RESULTS A total of 4,774 incident NAFLD cases were recorded during a 1,313,067 person-years of follow-up. In the multivariable adjusted model, laparoscopically confirmed endometriosis was positively associated with the risk of NAFLD (HR: 1.17, 95% CI: 1.07 - 1.29). The results of the mediation analyses revealed that the association was partly attributable to hysterectomy/oophorectomy (31.6% mediated, 95% CI: 18.8-47.9%), hypercholesterolemia, hypertension and infertility. Further analysis revealed that the interaction effect of age was significant for the association between endometriosis and NAFLD (P = 0.01). CONCLUSIONS Laparoscopically confirmed endometriosis was positively associated with the risk of incident NAFLD. Awareness of the potential NAFLD risk should be raised for clinicians and patients during the regular follow-up of endometriosis.
Collapse
Affiliation(s)
- Hangkai Huang
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Centre for Digestive Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhening Liu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Centre for Digestive Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaqi Ruan
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Centre for Digestive Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zejun Fang
- Sanmenwan Branch, The First Affiliated Hospital, Zhejiang University School of Medicine, Taizhou, China
| | - Chengfu Xu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Centre for Digestive Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
30
|
Yu C, He S, Peng N, Kuang M, Huang X, Wang C, Sheng G, Wang W, Xie G, Zou Y. The fasting plasma glucose to high-density lipoprotein cholesterol ratio: a novel index for identifying NAFLD. BMC Gastroenterol 2025; 25:236. [PMID: 40205359 PMCID: PMC11984220 DOI: 10.1186/s12876-025-03831-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Previous research has indicated that the fasting plasma glucose to high-density lipoprotein cholesterol ratio (FHR) is a novel biomarker for assessing cardiovascular disease prognosis. The objective of this study is to investigate the potential relationship of FHR with non-alcoholic fatty liver disease (NAFLD). METHODS The present study employed a retrospective design and included 14,251 participants who underwent health examinations. Restricted cubic spline regression and multivariable logistic regression were used to assess the relationship of NAFLD with FHR. The performance of the FHR in identifying NAFLD was analyzed through receiver operating curve analysis. RESULTS After adjustment for confounders, a positive association of FHR with NAFLD was observed in the multivariable logistic regression model. Compared to the lowest FHR quartile, the odds ratios for the 2, 3, and 4 quartiles were 1.49 (1.14, 1.94), 2.03 (1.57, 2.63), and 2.34 (1.79, 3.06), respectively. Additionally, restricted cubic spline regression analysis revealed a nonlinear relationship of FHR with NAFLD prevalence with a potential threshold effect. The prevalence of NAFLD remained nearly unchanged when the FHR was less than 3, but increased progressively when the ratio exceeded 3. Further receiver operating curve analysis demonstrated that the FHR significantly outperformed fasting plasma glucose or high-density lipoprotein cholesterol alone in identifying NAFLD, with an optimal threshold identified at 3.89. CONCLUSION The FHR is a new index used to identify NAFLD, and the incorporation of the FHR into the risk stratification assessment system for NAFLD may be of significant help.
Collapse
Affiliation(s)
- Changhui Yu
- Department of Emergency, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Shiming He
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Nan Peng
- Department of Endocrinology, Shenshan Medical Center of Sun Yat-Sen Memorial Hospital, Shanwei, Guangzhou, China
| | - Maobin Kuang
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xin Huang
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Chao Wang
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Guotai Sheng
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Wei Wang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Guobo Xie
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| |
Collapse
|
31
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
32
|
Zhu X, Sun F, Gao X, Liu H, Luo Z, Sun Y, Fan L, Deng J. Predictive value of triglyceride glucose index in non-obese non-alcoholic fatty liver disease. BMJ Open 2025; 15:e083686. [PMID: 40204316 PMCID: PMC11979504 DOI: 10.1136/bmjopen-2023-083686] [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: 12/27/2023] [Accepted: 01/21/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVES A large number of patients with non-obese non-alcoholic fatty liver disease (NAFLD) in China remain undiagnosed and untreated due to insufficient awareness and ineffective pharmacotherapy. Therefore, a convenient, predictive marker and diagnostic tools are imperative. This study aimed to investigate the ability of the triglyceride glucose index (TyG) in predicting non-obese NAFLD. DESIGN An observational cross-sectional study. SETTING Department of Health Management, large urban academic medical centre and DRYAD database data. PARTICIPANTS This study included 456 patients with non-obese NAFLD and matched 456 non-fatty liver controls according to age, sex and body mass index (BMI). PRIMARY AND SECONDARY OUTCOME MEASURES The receiver operating characteristic (ROC) curve was used to evaluate the predictive role of the TyG index in non-obese NAFLD. Based on the TyG index, a clinical prediction model for non-obese NAFLD was constructed, then the prediction model was verified by the DRYAD database (n=11 562). RESULTS TyG in non-obese NAFLD was higher than that in controls (9.00 (8.66-9.40) vs 8.46 (8.10-8.83), p<0.001). Logistic regression analysis showed that TyG was an independent risk factor for non-obese NAFLD (OR=9.03, 95% CI: 5.46 to 14.94, p<0.001). ROC analysis showed that the area under the curve (AUC) was 0.78, the sensitivity was 82.5%, the specificity was 60.5%. Based on the TyG index, sex, age and BMI, the AUC of the predictive model for non-obese NAFLD was 0.78 (95% CI: 0.75 to 0.81, p<0.001). Using the DRYAD database to verify the prediction model, the AUC of the verification group was 0.85 (95% CI: 0.84 to 0.86, p<0.001). CONCLUSIONS The high level of the TyG may be an independent risk factor for non-obese NAFLD. The prediction model for non-obese NAFLD based on the TyG index has good clinical prediction value.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Fang Sun
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Xia Gao
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - He Liu
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - ZhongYan Luo
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Yijian Sun
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Liqi Fan
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Juan Deng
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, Chongqing, China
| |
Collapse
|
33
|
Ye C, Wang X, Lin J, Wu C, Gao Y, Guo C, Liao Y, Rao Z, Huang S, Chen W, Huang Y, Sun J, Zhao D, Jiang C. Systematical identification of regulatory GPCRs by single-cell trajectory inference reveals the role of ADGRD1 and GPR39 in adipogenesis. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1057-1072. [PMID: 39821834 DOI: 10.1007/s11427-024-2732-8] [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: 05/04/2024] [Accepted: 09/15/2024] [Indexed: 01/19/2025]
Abstract
Adipogenesis is the healthy expansion of white adipose tissue (WAT), serving as a compensatory response to maintain metabolic homeostasis in the presence of excess energy in the body. Therefore, the identification of novel regulatory molecules in adipogenesis, specifically membrane receptors such as G protein-coupled receptors (GPCRs), holds significant clinical promise. These receptors can serve as viable targets for pharmaceuticals, offering potential for restoring metabolic homeostasis in individuals with obesity. We utilized trajectory inference methods to analyze three distinct single-nucleus sequencing (sNuc-seq) datasets of adipose tissue and systematically identified GPCRs with the potential to regulate adipogenesis. Through verification in primary adipose progenitor cells (APCs) of mice, we discovered that ADGRD1 promoted the differentiation of APCs, while GPR39 inhibits this process. In the obese mouse model induced by a high-fat diet (HFD), both gain-of-function and loss-of-function studies validated that ADGRD1 promoted adipogenesis, thereby improving metabolic homeostasis, while GPR39 inhibited adipogenesis, leading to metabolic dysfunction. Additionally, through the analysis of 2,400 ChIP-seq data and 1,204 bulk RNA-seq data, we found that the transcription factors (TFs) MEF2D and TCF12 regulated the expression of ADGRD1 and GPR39, respectively. Our study revealed the regulatory role of GPCRs in adipogenesis, providing novel targets for clinical intervention of metabolic dysfunction in obese patients.
Collapse
Affiliation(s)
- Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jun Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Chenyang Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Yuhua Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Chenghao Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yunxi Liao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Ziyan Rao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Shaodong Huang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Weixuan Chen
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
| | - Ying Huang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, 100191, China
| | - Jinpeng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250100, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
34
|
Bertinat R, Holyoak T, Gatica R, Jara N, González-Chavarría I, Westermeier F. The neglected PCK1/glucagon (inter)action in nutrient homeostasis beyond gluconeogenesis: Disease pathogenesis and treatment. Mol Metab 2025; 94:102112. [PMID: 39954782 PMCID: PMC11909762 DOI: 10.1016/j.molmet.2025.102112] [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: 11/30/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Glucagon plays a central role in hepatic adaptation during fasting, with the upregulation of hepatic phosphoenolpyruvate carboxykinase 1 (PCK1) traditionally associated with increased gluconeogenesis. However, recent experimental models and clinical studies have challenged this view, suggesting a more complex interplay between PCK1 and glucagon, which extends beyond gluconeogenesis and has broader implications for metabolic regulation in health and disease. SCOPE OF REVIEW This review provides a comprehensive overview of the current evidence on the multifaceted roles of PCK1 in glucagon-dependent hepatic adaptation during fasting, which is crucial for maintaining systemic homeostasis not only of glucose, but also of lipids and amino acids. We explore the relationship between PCK1 deficiency and glucagon resistance in metabolic disorders, including inherited PCK1 deficiency and metabolic dysfunction-associated steatotic liver disease (MASLD), and compare findings from experimental animal models with whole-body or tissue-specific ablation of PCK1 or the glucagon receptor. We propose new research platforms to advance the therapeutic potential of targeting PCK1 in metabolic diseases. MAJOR CONCLUSIONS We propose that hepatic PCK1 deficiency might be an acquired metabolic disorder linking alterations in lipid metabolism with impaired glucagon signaling. Our findings highlight interesting links between glycerol, PCK1 deficiency, elevated plasma alanine levels and glucagon resistance. We conclude that the roles of PCK1 and glucagon in metabolic regulation are more complex than previously assumed. In this (un)expected scenario, hepatic PCK1 deficiency and glucagon resistance appear to exert limited control over glycemia, but have broader metabolic effects related to lipid and amino acid dysregulation. Given the shift in glucagon research from receptor inhibition to activation, we propose that a similar paradigm shift is needed in the study of hepatic PCK1. Understanding PCK1 expression and activity in the glucagon-dependent hepatic adaptation to fasting might provide new perspectives and therapeutic opportunities for metabolic diseases.
Collapse
Affiliation(s)
- Romina Bertinat
- Centro de Microscopía Avanzada, CMA-BIO BIO, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile; Laboratorio de Lipoproteínas y Cáncer, Departamento de Fisiopatología, Universidad de Concepción, Concepción, Chile.
| | - Todd Holyoak
- Department of Biology, Faculty of Science, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Rodrigo Gatica
- Escuela de Veterinaria, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Nery Jara
- Departamento de Farmacología, Universidad de Concepción, Concepción, Chile
| | - Iván González-Chavarría
- Laboratorio de Lipoproteínas y Cáncer, Departamento de Fisiopatología, Universidad de Concepción, Concepción, Chile
| | - Francisco Westermeier
- Institute of Biomedical Science, Department of Health Studies, FH JOANNEUM University of Applied Sciences, Graz, Austria; Centro de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile.
| |
Collapse
|
35
|
Wang H, Wang Z, Wang D, Nie K, Wu W, Gao Y, Chen S, Jiang X, Tang Y, Su H, Hu M, Fang K, Dong H. Berberine Attenuates Nonalcoholic Hepatic Steatosis by Regulating Lipid Droplet-Associated Proteins: In Vivo, In Vitro and Molecular Evidence. J Cell Mol Med 2025; 29:e70524. [PMID: 40194991 PMCID: PMC11975506 DOI: 10.1111/jcmm.70524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/27/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Hepatic lipid droplet (LD) accumulation is a hallmark of nonalcoholic fatty liver disease (NAFLD). Although the clinical efficacy of berberine (BBR) in treating NAFLD has been established, the mechanism remains uncertain. This study is to evaluate the effects of BBR on hepatic LDs and investigate the underlying mechanisms. Using high-fat diet-induced obese (DIO) mice as the model for NAFLD, BBR was administered daily by gavage for 4 weeks. Liver tissue was examined for changes in lipid deposition and histology. Transcriptomics was performed to screen differently expressed genes. The potential targets of BBR against NAFLD were then determined by Western Blot and immunostaining. In oleic acid (OA)-induced HepG2 cells, the link between BBR and potential targets was further elucidated through the activation or antagonism of PPARα. The binding of BBR to potential targets was predicted using molecular docking. BBR significantly reduced hepatic steatosis by decreasing LD size rather than number. Transcriptomics with validation demonstrated that BBR modulated the expression of LD-associated proteins CIDEA and PLIN4 in the liver. Further investigations revealed that BBR reversed the abnormal elevation of BSCL2 and PLIN2 in steatotic livers. Finally, we found that BBR reduced LD size in OA-induced HepG2 cells by regulating BSCL2 and PPARα-mediated CIDEA/PLIN4/PLIN2. Notably, BBR could bind well to PPARα and BSCL2. BBR can attenuate hepatic steatosis in DIO mice by reducing LD size through the regulation of LD-associated proteins.
Collapse
Affiliation(s)
- Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhi Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Dingkun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Wenbin Wu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Shen Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xinyue Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yueheng Tang
- Department of Rehabilitation Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Su
- Department of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Meilin Hu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
36
|
Li Q, Cheng J, Sun Y, He L, Li R. Protective Effects of Polygonatum sibiricum Polysaccharides Against Type 2 Diabetic Mice Induced by High-Fat Diet and Low-Dose Streptozotocin. TOXICS 2025; 13:255. [PMID: 40278571 PMCID: PMC12031623 DOI: 10.3390/toxics13040255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025]
Abstract
Polysaccharides possessing hypoglycemic effects have shown promising results in treating diabetes. Polygonatum sibiricum polysaccharide (PSP) is one of the most active ingredients in the Chinese medicine P. sibiricum Redoute with many biological activities. However, its efficacy in alleviating type 2 diabetes mellitus (T2DM) remains unexplored. Our aim is to evaluate the protective effect of PSP against T2DM by measuring body weight and serum biochemical indicators, examining the histopathological images of pancreatic and liver tissues, detecting fecal short-chain fatty acid (SCFA) content, and analyzing the intestinal flora diversity and the microbiota structure in T2DM mice. The findings indicated that PSP treatment in T2DM mice could obviously decrease the fasting blood glucose and fasting insulin levels, ameliorate glucose tolerance, insulin resistance, lipid, and inflammatory factor levels, attenuate pancreatic and liver damage, and increase the fecal SCFA content. In addition, PSP could modulate the composition of gut microbiota in T2DM mice, resulting in the relative abundance of Firmicutes decreasing and that of Bacteroidetes increasing, along with the abundance of beneficial flora significantly increasing, especially SCFA-producing bacteria. The findings indicate that PSP administration protected against diabetes by controlling disordered glucolipid metabolism and modulating the gut microbiota, which provides a valuable strategy for the utilization of PSP to treat T2DM.
Collapse
Affiliation(s)
- Qingxiangzi Li
- Laboratory Animal Center, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jufen Cheng
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | | | - Liang He
- Laboratory Animal Center, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Rui Li
- Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
37
|
Deng J, Ding K, Liu S, Chen F, Huang R, Xu B, Zhang X, Xie W. SOX9 Overexpression Ameliorates Metabolic Dysfunction-associated Steatohepatitis Through Activation of the AMPK Pathway. J Clin Transl Hepatol 2025; 13:189-199. [PMID: 40078197 PMCID: PMC11894392 DOI: 10.14218/jcth.2024.00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025] Open
Abstract
Background and Aims The transcription factor sex-determining region Y-related high-mobility group-box gene 9 (SOX9) plays a critical role in organ development. Although SOX9 has been implicated in regulating lipid metabolism in vitro, its specific role in metabolic dysfunction-associated steatohepatitis (MASH) remains poorly understood. This study aimed to investigate the role of SOX9 in MASH pathogenesis and explored the underlying mechanisms. Methods MASH models were established using mice fed either a methionine- and choline-deficient (MCD) diet or a high-fat, high-fructose diet. To evaluate the effects of SOX9, hepatocyte-specific SOX9 deletion or overexpression was performed. Lipidomic analyses were conducted to assess how SOX9 influences hepatic lipid metabolism. RNA sequencing was employed to identify pathways modulated by SOX9 during MASH progression. To elucidate the mechanism further, HepG2 cells were treated with an adenosine monophosphate-activated protein kinase (AMPK) inhibitor to test whether SOX9 acts via AMPK activation. Results SOX9 expression was significantly elevated in hepatocytes of MASH mice. Hepatocyte-specific SOX9 deletion exacerbated MCD-induced MASH, whereas overexpression of SOX9 mitigated high-fat, high-fructose-induced MASH. Lipidomic and RNA sequencing analyses revealed that SOX9 suppresses the expression of genes associated with lipid metabolism, inflammation, and fibrosis in MCD-fed mice. Furthermore, SOX9 deletion inhibited AMPK pathway activation, while SOX9 overexpression enhanced it. Notably, administration of an AMPK inhibitor negated the protective effects of SOX9 overexpression, leading to increased lipid accumulation in HepG2 cells. Conclusions Our findings demonstrate that SOX9 overexpression alleviates hepatic lipid accumulation in MASH by activating the AMPK pathway. These results highlight SOX9 as a promising therapeutic target for treating MASH.
Collapse
Affiliation(s)
- Juan Deng
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shuqing Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fei Chen
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ru Huang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bonan Xu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weifen Xie
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| |
Collapse
|
38
|
Zhang J, Wang Q, Zhou N, Liu J, Tao L, Peng Z, Hu G, Wang H, Fu L, Peng S. Fluorofenidone attenuates choline-deficient, l-amino acid-defined, high-fat diet-induced metabolic dysfunction-associated steatohepatitis in mice. Sci Rep 2025; 15:9863. [PMID: 40118958 PMCID: PMC11928590 DOI: 10.1038/s41598-025-94401-7] [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: 11/11/2024] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), a severe form of metabolic dysfunction-associated steatotic liver disease (MASLD), involves hepatic lipid accumulation, inflammation, and fibrosis. It can progress to cirrhosis or hepatocellular carcinoma without timely treatment. Current treatment options for MASH are limited. This study explores the therapeutic effects of fluorofenidone (AKF-PD), a novel small-molecule compound with antifibrotic and anti-inflammatory properties, on MASH in mouse model. Mice fed a choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) were treated with AKF-PD, resulting in reduced serum ALT, AST, hepatic lipid accumulation, liver inflammation, and fibrosis. Network pharmacology and RNA-sequencing analyses suggested that AKF-PD influenced multiple metabolic, inflammatory, and fibrosis-related pathways. Further experiments verified that AKF-PD activated hepatic AMPK signaling, leading to the inhibition of the downstream SREBF1/SCD1 pathway and the activation of autophagy. Additionally, AKF-PD suppressed the expression of various inflammatory factors, reduced macrophage infiltration, and inhibited NLRP3 inflammasome activation. Moreover, AKF-PD attenuated liver fibrosis by inhibiting TGFβ1/SMAD signaling. In conclusion, this study reveals that AKF-PD effectively decreases hepatic lipid accumulation, liver inflammation and fibrosis in a CDAHFD-induced MASH model, positioning AKF-PD as a promising candidate for the treatment of MASH.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qianbing Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Nianqi Zhou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinqing Liu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Gaoyun Hu
- Faculty of Pharmaceutical Sciences, Central South University, Changsha, 410008, Hunan, China
| | - Huiwen Wang
- Department of Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
39
|
Forouzesh P, Kheirouri S, Alizadeh M. Predicting hepatic steatosis degree in metabolic dysfunction-associated steatotic liver disease using obesity and lipid-related indices. Sci Rep 2025; 15:8612. [PMID: 40074727 PMCID: PMC11904216 DOI: 10.1038/s41598-024-73132-1] [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: 07/10/2024] [Accepted: 09/13/2024] [Indexed: 03/14/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as nonalcoholic fatty liver disease, represents a prevalent condition ranging from simple steatosis to advanced stages associated with liver cancer. Asymptomatic presentation in the majority of cases underscores the need for non-invasive, cost-effective methods to stratify degree of hepatic steatosis. This cross-sectional study aimed to assess the association between obesity and lipid-related indices with the degree of hepatic steatosis in MASLD patients. 150 individuals recently diagnosed with metabolic dysfunction-associated steatotic liver disease were recruited. Anthropometric measurements, including weight, height, and waist circumference (WC), were taken, alongside biochemical parameters such as alanine aminotransferase, aspartate aminotransferase, total cholesterol, triglycerides (TG), high-density lipoprotein, low-density lipoprotein, and fasting plasma glucose, following a 12-h fasting period. Various indicators of obesity and lipid metabolism, including body mass index, waist-to-height ratio (WHtR), a body shape index, lipid accumulation product (LAP), triglyceride-glucose index (TyG), visceral adiposity index, and hepatic steatosis index (HSI), were calculated. The diagnosis of MASLD and degree of hepatic steatosis were established through abdominal ultrasound examination. Data analysis was performed utilizing SPSS version 22. All the investigated indices displayed an area under the curve (AUC) surpassing 0.5, implying a correlation with the degree of hepatic steatosis. Notably, TyG-WC, TyG-WHtR, LAP, and a cardiometabolic obesity index showed the highest AUC values (> 0.7), indicating a relatively strong association with degree of hepatic steatosis. Specifically, in females, TyG-WC (AUC = 0.797, 95% CI 0.712-0.882, threshold = 865.991), while in males, LAP (AUC = 0.746, 95% CI 0.593-0.899, threshold = 74.290), demonstrated the highest AUC values. TyG-WHtR, TyG-WC, and LAP exhibited significant correlations with the degree of hepatic steatosis. Given their non-invasive nature and easy measurement, they hold promise for potential clinical utility, pending validation in additional studies.
Collapse
Affiliation(s)
- Paniz Forouzesh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran.
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Lian YE, Wang Y, Yang Y, Chen J. Weight-adjusted waist circumference index with hepatic steatosis and fibrosis in adult females: a cross-sectional, nationally representative study (NHANES 2017-2020). BMC Gastroenterol 2025; 25:137. [PMID: 40045243 PMCID: PMC11884151 DOI: 10.1186/s12876-025-03706-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Obesity is detrimental to liver health. Weight-adjusted waist circumference (WWI) is a new indicator of obesity that is superior to body mass index (BMI) and waist circumference (WC) in predicting obesity. There are limited studies on the relationship between Metabolic Associated Fatty Liver Disease (MASLD) and WWI. Therefore, this study aimed to investigate the association between WWI, Controlled Attenuation Parameters (CAP), and Liver Stiffness Measurement (LSM), with special attention to gender differences. METHODS This cross-sectional study included participants from the 2017 to 2020 National Health and Nutrition Examination Survey (NHANES). The study used multiple linear regression models, smoothed curves, and threshold effects analyses to describe the relationships between variables. Multiple regression analyses were used to examine the associations between the four obesity indicators and CAP and LSM. Subject work characteristics (ROC) curves were used to assess the predictive value of WWI and other traditional obesity indicators for hepatic steatosis and liver fibrosis, and predictive power was assessed by area under the curve (AUC). RESULTS The study involved 6713 participants, including 3072 men (46%) and 3641 women (54%). The results showed that among female participants, higher WWI was associated with hepatic steatosis (OR = 1.71, 95% CI: 1.43, 2.04; P < 0.0001) and hepatic fibrosis (OR = 2.11, 95% CI: 1.58, 2.84; P < 0.0001). Smoothed curve fitting of WWI versus CAP showed a statistically significant positive correlation between WWI in male and female participants There was a statistically significant positive correlation with CAP for both male and female participants. The same significant non-linear relationship was found between WWI and LSM, with no significant difference between males and females. WWI was also a good predictor of hepatic steatosis compared to other obesity indicators and was more pronounced in male participants (AUC = 0.8224). Whereas in the comparison of WWI with LSM, wBMI was a better predictor in female participants (AUC = 0.7751). CONCLUSIONS Based on this study, WWI was significantly associated with the risk of hepatic steatosis and hepatic fibrosis in women, suggesting the potential of WWI as a screening tool. Due to the cross-sectional design, causality cannot be inferred. Longitudinal studies are needed to validate our findings.
Collapse
Affiliation(s)
- Yu-E Lian
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China
| | - Yixuan Wang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China
| | - Yinyin Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, 730050, China
| | - Jiayu Chen
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China.
| |
Collapse
|
41
|
Do A, Zahrawi F, Mehal WZ. Therapeutic landscape of metabolic dysfunction-associated steatohepatitis (MASH). Nat Rev Drug Discov 2025; 24:171-189. [PMID: 39609545 DOI: 10.1038/s41573-024-01084-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/30/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its severe subgroup metabolic dysfunction-associated steatohepatitis (MASH) have become a global epidemic and are driven by chronic overnutrition and multiple genetic susceptibility factors. The physiological outcomes include hepatocyte death, liver inflammation and cirrhosis. The first therapeutic for MASLD and MASH, resmetirom, has recently been approved for clinical use and has energized this therapeutic space. However, there is still much to learn in clinical studies of MASH, such as the scale of placebo responses, optimal trial end points, the time required for fibrosis reversal and side effect profiles. This Review introduces aspects of disease pathogenesis related to drug development and discusses two main therapeutic approaches. Thyroid hormone receptor-β agonists, such as resmetirom, as well as fatty acid synthase inhibitors, target the liver and enable it to function within a toxic metabolic environment. In parallel, incretin analogues such as semaglutide improve metabolism, allowing the liver to self-regulate and reversing many aspects of MASH. We also discuss how combinations of therapeutics could potentially be used to treat patients.
Collapse
Affiliation(s)
- Albert Do
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Division of Gastroenterology, University of California, Davis, Davis, USA
| | - Frhaan Zahrawi
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- West Haven Veterans Hospital, West Haven, CT, USA.
| |
Collapse
|
42
|
Clark SL, Saade GA, Tolcher MC, Belfort MA, Rouse DJ, Silver RM, Kapadia A, Sundgren NC, Saridey SK, Sibai BM. An Organ-Specific Approach to the Management of Gestational Hypertension: Evidence versus Tradition. Am J Perinatol 2025; 42:546-554. [PMID: 39496325 DOI: 10.1055/a-2459-8748] [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] [Indexed: 11/06/2024]
Abstract
The management of hypertensive disease in pregnancy is currently guided by practice recommendations based largely on observational data from a half century ago and has changed only superficially since that time. These recommendations are both narrowly prescriptive (women without traditional features of severe disease should all be delivered at exactly 370/7 weeks) and at the same time frustratingly ambiguous (the presence of epigastric pain unresponsive to repeat analgesics precludes expectant management at any gestational age, regardless of laboratory studies). Guidelines that ignore recent data from the obstetric, pediatric, and internal medicine literature too often lead practitioners to be more aggressive than necessary in the delivery of very premature pregnancies, and, conversely, more complacent than patient safety would support in prolonging pregnancy with advanced fetal maturity. We present here an alternative, organ-specific-based approach to the management of gestational hypertension that allows and encourages practitioners to formulate a management plan based on a thoughtful and, when possible, evidence-based synthesis of the continuous variables of blood pressure, degree of organ dysfunction and response to treatment, gestational age, and patient balancing of maternal and fetal/neonatal risks. Such clinical care is more complex and nuanced than simply basing life-altering critical management decisions, including timing of delivery, on whether the patient does, or does not have any one of the conditions described by box 4 of the current American College of Obstetricians and Gynecologists practice guidelines. Nonetheless, we believe this approach will not only improve care but will also open the door to useful investigations into prevention and management of the various entities traditionally considered as the same disease process. KEY POINTS: · Traditional approaches to preeclampsia are not evidence based.. · The use of such approaches has resulted in stagnant maternal morbidity and mortality ratios.. · The consideration of disease severity as binary is particularly counterproductive.. · An organ-based approach will facilitate evidence-based individualization of care..
Collapse
Affiliation(s)
- Steven L Clark
- Department of Obstetrics and Gynecology, Nephrology, Neurology and Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - George A Saade
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Mary C Tolcher
- Department of Obstetrics and Gynecology, Nephrology, Neurology and Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Michael A Belfort
- Department of Obstetrics and Gynecology, Nephrology, Neurology and Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Dwight J Rouse
- Department of Obstetrics and Gynecology, Brown University, Providence, Rhode Island
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, Utah
| | | | | | | | - Baha M Sibai
- Department of Obstetrics and Gynecology, University of Texas, Houston, Texas
| |
Collapse
|
43
|
Farrera DO, Alaaldin MM, Lindberg P, Sample PA, Lenzen-Hammerel P, LaMadrid CS, Haymore R, Wright SH, Cherrington NJ. Alterations of valsartan pharmacokinetics in a rodent model of metabolic dysfunction-associated steatohepatitis. Drug Metab Dispos 2025; 53:100043. [PMID: 40054126 DOI: 10.1016/j.dmd.2025.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 03/30/2025] Open
Abstract
Valsartan (VAL) is commonly prescribed for patients with cardiovascular disease (CVD) to lower blood pressure, reduce heart failure risk, and prevent heart attacks or strokes by blocking the effects of angiotensin II. Many patients with CVD also suffer from metabolic dysfunction-associated steatohepatitis (MASH), which disrupts several xenobiotic transporters, affecting the pharmacokinetics of numerous drugs. Medications used in patients to treat comorbidities associated with MASH may be subject to this altered disposition and potential toxicity. This study aimed to assess how MASH alters the pharmacokinetics of VAL using a rodent model that mimics human MASH. MASH was induced in rats via a methionine- and choline-deficient (MCD) diet. Rats received VAL-a substrate of organic anion-transporting polypeptide (OATP) 1B1/1B3 and reported for multidrug resistance-associated protein-2-(2 mg/kg) through intravenous injection to isolate hepatic transport processes, and bile, serum, and liver concentrations measured using liquid chromatography-tandem mass spectrometry. Consistent with MASH progression, MCD rats presented with more gross pathology, including increased liver-to-body weight ratios, along with macrosteatosis, hepatocyte ballooning, and lobular inflammation. In MCD rats, the expression of Oatp1b2 was significantly reduced, and Mrp2 was internalized, resulting in higher systemic exposure to VAL compared with controls. Additionally, cumulative biliary excretion of VAL was lower in MCD rats. To further assess VAL disposition in MASH, transport kinetics were evaluated in human embryonic kidney 293 cells overexpressing OATP1B1 or OATP1B3, revealing similar affinity for VAL between both transporters. These findings suggest that changes in OATP function in MASH may alter VAL pharmacokinetics, which may have implications for personalized treatments. SIGNIFICANCE STATEMENT: Although expression of drug transporters in metabolic dysfunction-associated steatohepatitis (MASH) has been explored, the combined effect between MASH and genetic loss of transporters on the disposition of sartan drugs has not been determined. This study applied liquid chromatography-tandem mass spectrometry analyses and immunohistological staining to assess drug disposition and identify alterations to drug transporters in rodents on a methionine- and choline-deficient diet. The observations made in this study have significant implications regarding its disposition in the context of hepatic dysfunction associated with MASH.
Collapse
Affiliation(s)
- Dominique O Farrera
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Mina M Alaaldin
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Paige Lindberg
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Paxton A Sample
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Paige Lenzen-Hammerel
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Christopher S LaMadrid
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Ryan Haymore
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- College of Medicine, Department of Physiology, The University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology, The University of Arizona, Tucson, Arizona.
| |
Collapse
|
44
|
Wang K, Liu Z, Tang R, Sha Y, Wang Z, Chen Y, Chen G. Gallstones in the Era of Metabolic Syndrome: Pathophysiology, Risk Prediction, and Management. Cureus 2025; 17:e80541. [PMID: 40225487 PMCID: PMC11993725 DOI: 10.7759/cureus.80541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Gallstone disease (GSD) and metabolic syndrome (MetS) are increasingly prevalent conditions with significant global health implications. Recent evidence highlights a strong epidemiological association between these disorders, driven by shared pathophysiological mechanisms. This review provides a comprehensive analysis of the intricate relationship between MetS and GSD, focusing on the role of insulin resistance, dyslipidemia, obesity, and gut microbiota dysbiosis in gallstone formation. An integrated pathophysiological model is proposed, linking metabolic disturbances to bile cholesterol supersaturation, gallbladder dysmotility, and chronic inflammation. The review also explores clinical implications, including risk prediction models based on metabolic parameters, early detection biomarkers, and targeted interventions such as lifestyle modifications, pharmacological therapies, and microbiome modulation. By addressing the metabolic underpinnings of GSD, this synthesis offers a foundation for developing preventive and therapeutic strategies to mitigate the burden of these interconnected conditions. Future research directions are outlined to refine mechanistic insights and improve clinical outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Surgery, Wannan Medical College, Wuhu, CHN
| | - Zhigang Liu
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| | - Rongmei Tang
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| | | | | | - Yisheng Chen
- General Surgery, Wuhu Guangji Hospital, Wuhu, CHN
| | - Guangbin Chen
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| |
Collapse
|
45
|
Zhang X, Lau HCH, Yu J. Pharmacological treatment for metabolic dysfunction-associated steatotic liver disease and related disorders: Current and emerging therapeutic options. Pharmacol Rev 2025; 77:100018. [PMID: 40148030 DOI: 10.1016/j.pharmr.2024.100018] [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: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as nonalcoholic fatty liver disease) is a chronic liver disease affecting over a billion individuals worldwide. MASLD can gradually develop into more severe liver pathologies, including metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and liver malignancy. Notably, although being a global health problem, there are very limited therapeutic options against MASLD and its related diseases. While a thyroid hormone receptor agonist (resmetirom) is recently approved for MASH treatment, other efforts to control these diseases remain unsatisfactory. Given the projected rise in MASLD and MASH incidence, it is urgent to develop novel and effective therapeutic strategies against these prevalent liver diseases. In this article, the pathogenic mechanisms of MASLD and MASH, including insulin resistance, dysregulated nuclear receptor signaling, and genetic risk factors (eg, patatin-like phospholipase domain-containing 3 and hydroxysteroid 17-β dehydrogenase-13), are introduced. Various therapeutic interventions against MASH are then explored, including approved medication (resmetirom), drugs that are currently in clinical trials (eg, glucagon-like peptide 1 receptor agonist, fibroblast growth factor 21 analog, and PPAR agonist), and those failed in previous trials (eg, obeticholic acid and stearoyl-CoA desaturase 1 antagonist). Moreover, given that the role of gut microbes in MASLD is increasingly acknowledged, alterations in the gut microbiota and microbial mechanisms in MASLD development are elucidated. Therapeutic approaches that target the gut microbiota (eg, dietary intervention and probiotics) against MASLD and related diseases are further explored. With better understanding of the multifaceted pathogenic mechanisms, the development of innovative therapeutics that target the root causes of MASLD and MASH is greatly facilitated. The possibility of alleviating MASH and achieving better patient outcomes is within reach. SIGNIFICANCE STATEMENT: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, and it can progress to more severe pathologies, including steatohepatitis, cirrhosis, and liver cancer. Better understanding of the pathogenic mechanisms of these diseases has facilitated the development of innovative therapeutic strategies. Moreover, increasing evidence has illustrated the crucial role of gut microbiota in the pathogenesis of MASLD and related diseases. It may be clinically feasible to target gut microbes to alleviate MASLD in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
46
|
Lian R, Tang H, Chen Z, Chen X, Luo S, Jiang W, Jiang J, Yang M. Development and multi-center cross-setting validation of an explainable prediction model for sarcopenic obesity: a machine learning approach based on readily available clinical features. Aging Clin Exp Res 2025; 37:63. [PMID: 40021576 PMCID: PMC11870957 DOI: 10.1007/s40520-025-02975-z] [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: 11/28/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025]
Abstract
OBJECTIVES Sarcopenic obesity (SO), characterized by the coexistence of obesity and sarcopenia, is an increasingly prevalent condition in aging populations, associated with numerous adverse health outcomes. We aimed to identify and validate an explainable prediction model of SO using easily available clinical characteristics. SETTING AND PARTICIPANTS A preliminary cohort of 1,431 participants from three community regions in Ziyang city, China, was used for model development and internal validation. For external validation, we utilized data from 832 residents of multi-center nursing homes. MEASUREMENTS The diagnosis of SO was based on the European Society for Clinical Nutrition and Metabolism (ESPEN) and the European Association for the Study of Obesity (EASO) criteria. Five machine learning models (support vector machine, logistic regression, random forest, light gradient boosting machine, and extreme gradient boosting) were used to predict SO. The performance of these models was assessed by the area under the receiver operating characteristic curve (AUC). The SHapley Additive exPlanations (SHAP) approach was used for model interpretation. RESULTS After feature reduction, an 8-feature model demonstrated good predictive ability. Among the five models tested, the support vector machine (SVM) model performed best in SO prediction in both internal (AUC = 0.862) and external (AUC = 0.785) validation sets. The eight key predictors identified were BMI, gender, neck circumference, waist circumference, thigh circumference, time to full tandem standing, time to five-times sit-to-stand, and age. SHAP analysis revealed BMI and gender as the most influential predictors. To facilitate the utilization of the SVM model in clinical setting, we developed a web application ( https://svcpredictapp.streamlit.app/ ). CONCLUSIONS We developed an explainable machine learning model to predict SO in aging community and nursing populations. This model offers a novel, accessible, and interpretable approach to SO prediction with potential to enhance early detection and intervention strategies. Further studies are warranted to validate our model in diverse populations and evaluate its impact on patient outcomes when integrated into comprehensive geriatric assessments.
Collapse
Affiliation(s)
- Rongna Lian
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Huiyu Tang
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zecong Chen
- Department of Geriatric, Zigong Affiliated Hospital of Southwest Medical University, Zigong, China
| | - Xiaoyan Chen
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyue Luo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenhua Jiang
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaojiao Jiang
- Rehabilitation Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Ming Yang
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
47
|
Lan Y, Song R, Feng D, He J. Bioinformatic analysis of molecular expression patterns during the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Sci Rep 2025; 15:7294. [PMID: 40025132 PMCID: PMC11873118 DOI: 10.1038/s41598-025-90744-3] [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: 06/25/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
The global incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise, primarily driven by the escalating obesity epidemic worldwide. MASLD, a spectrum of liver disorders, can progress to more severe conditions, metabolic dysfunction-associated steatohepatitis (MASH), ultimately culminating in hepatocellular carcinoma (HCC). Given the complex nature of MASLD, there is an urgent need to develop robust risk prediction models and design specialized cancer screening initiatives tailored specifically for individuals with MASLD. This study aimed to identify genes exhibiting trending expression patterns that could serve as potential biomarkers or therapeutic targets. Our approach involved analyzing expression patterns across the five stages of MASLD development and progression. Notably, we introduced an innovative two-phase classification-MASLD occurrence and MASLD progression-instead of categorizing differentially expressed genes (DEGs) into multiple types. Leveraging LASSO regression models, we demonstrated their relatively strong capability to predict and distinguish both MASLD occurrence and progression. Furthermore, our analysis identified CYP7A1 and TNFRSF12A as significantly associated with the prognosis of MASLD progressing to HCC. These findings contribute to the understanding of gene expression dynamics in MASLD and may pave the way for the development of effective prognostic tools and targeted therapies in the realm of liver disease.
Collapse
Affiliation(s)
- Yuanfeng Lan
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
48
|
Somi M, Frounchi N, Zakavi SS, Ostadrahimi A, Gilani N, Faramarzi E, Sanaie S. The association between serum uric acid levels and the cardiometabolic phenotype among healthcare workers of Tabriz University of Medical Sciences. J Cardiovasc Thorac Res 2025; 17:40-48. [PMID: 40365515 PMCID: PMC12068801 DOI: 10.34172/jcvtr.32902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 03/09/2024] [Accepted: 01/01/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction It is unclear whether hyperuricemia can be considered as an independent risk factor or just as a marker to represent the correlation between uric acid levels and other risk factors of MetS. In this work, we intend to study the correlation between serum uric acid (SUA) and the cardiometabolic phenotype among Tabriz University of Medical Science healthcare workers. Methods In this cross-sectional study, anthropometric measurements, serum fasting blood sugar (FBS), triglyceride (TG), cholesterol, high-density lipoprotein (HDL), liver enzymes, blood urea nitrogen (BUN), SUA, creatinine (Cr), and blood pressures of 1,451 healthcare workers were evaluated. MetS was diagnosed based on ATP III. We classified the participants into four cardiometabolic phenotypes: metabolically-healthy lean (MHL), metabolically-unhealthy lean (MUHL), metabolically-healthy obese (MHO), and metabolically-unhealthy obese (MUHO). Results MHL (26.6%) and MHO (65.8 %) had the highest prevalence rates in the first and second SUA categories, respectively (P≤0.001). Compared to the lowest SUA category, the odds of MHO and MUHO increased by 3.13 (95% CI 2.21-4.44) and 5.50 (95%CI 3.53-8.57) in the highest category, respectively. This trend was not observed regarding the association between MUHL and the SUA classification. Conclusion We propose using the easily-measured SUA level as a marker for early diagnosis of at-risk MUHL and MHO individuals to administer proper interventions. Further prospective studies are needed to identify the effects of SUA on the progression of MetS in various body-size subgroups.
Collapse
Affiliation(s)
- Mohammadhossein Somi
- Liver and Gastrointestinal Diseases Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Frounchi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Sina Zakavi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Gilani
- Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Faramarzi
- Liver and Gastrointestinal Diseases Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Higa R, Pourteymour S, Kolan PS, Dankel SN, Fernø J, Mellgren G, Pan C, Seldin MM, Lusis AJ, Drevon CA, Dalen KT, Norheim FA. Hepatic lipid metabolism is altered in Ubiad1 +/- mice of both sexes. Sci Rep 2025; 15:7022. [PMID: 40016272 PMCID: PMC11868635 DOI: 10.1038/s41598-025-91283-7] [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: 05/13/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
UbiA prenyltransferase domain containing 1 (Ubiad1) has the potential to affect cholesterol and phospholipid levels in different cell types. We previously identified Ubiad1 as a candidate gene for regulating subcutaneous fat pad weight in a mouse genome-wide association study. Here we evaluated the relationship between Ubiad1 and obesity-related traits in cohorts of humans and mice, and in Ubiad1+/- mice fed a high-fat diet. In both humans and mice, adipose tissue Ubiad1 mRNA expression correlated negatively with adiposity and positively with mitochondria-related genes. To determine the role of Ubiad1 in high-fat diet-induced obesity, we disrupted the Ubiad1 gene in mice. Deletion of Ubiad1 was embryonically lethal in C57BL/6 N mice, preventing analysis of adult Ubiad1-/- mice. Thus, male and female Ubiad1+/+ and Ubiad1+/- mice were fed high-fat diet for 10 weeks, with no difference in weight gain and adipose tissue organ weights observed between the genotypes. Analysis of liver mRNA expression revealed that Ubiad1 heterozygosis (Ubiad1+/-) altered several pathways involved in lipid metabolism. Detailed lipid quantification with HPLC-qTOF/MS showed increased levels of hepatic ceramides in female Ubiad1+/- mice, whereas phosphatidylglycerols, phosohatidylinositol and lysophosphatidylethanolamines were reduced in male Ubiad1+/- mice. Our findings reveal sex-specific effects of Ubiad1 expression on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Ryoko Higa
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Shirin Pourteymour
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Pratibha S Kolan
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Simon N Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Vitas AS, Science Park, Gaustadalléen 21, 0349, Oslo, Norge
| | - Knut T Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Frode A Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway.
| |
Collapse
|
50
|
Zhu R, Xu C, Jiang S, Xia J, Wu B, Zhang S, Zhou J, Liu H, Li H, Lou J. Risk factor analysis and predictive model construction of lean MAFLD: a cross-sectional study of a health check-up population in China. Eur J Med Res 2025; 30:137. [PMID: 40001266 PMCID: PMC11863909 DOI: 10.1186/s40001-025-02373-1] [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: 09/05/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
AIM Cardiovascular disease morbidity and mortality rates are high in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). The objective of this study was to analyze the risk factors and differences between lean MAFLD and overweight MAFLD, and establish and validate a nomogram model for predicting lean MAFLD. METHODS This retrospective cross-sectional study included 4363 participants who underwent annual health checkup at Yuyao from 2019 to 2022. The study population was stratified into three groups: non-MAFLD, lean MAFLD (defined as the presence of fatty liver changes as determined by ultrasound in individuals with a BMI < 25 kg/m2), and overweight MAFLD (BMI ≥ 25.0 kg/m2). Subsequent modeling analysis was conducted in a population that included healthy subjects with < 25 kg/m2 (n = 2104) and subjects with lean MAFLD (n = 849). The study population was randomly split (7:3 ratio) to a training vs. a validation cohort. Risk factors for lean MAFLD was identify by multivariate regression of the training cohort, and used to construct a nomogram to estimate the probability of lean MAFLD. Model performance was examined using the receiver operating characteristic (ROC) curve analysis and k-fold cross-validation (k = 5). Decision curve analysis (DCA) was applied to evaluate the clinical usefulness of the prediction model. RESULTS The multivariate regression analysis indicated that the triglycerides and glucose index (TyG) was the most significant risk factor for lean MAFLD (OR: 4.03, 95% CI 2.806-5.786). The restricted cubic spline curves (RCS) regression model demonstrated that the relationships between systolic pressure (SBP), alanine aminotransferase (ALT), serum urate (UA), total cholesterol (TCHO), triglyceride (TG), triglyceride glucose (TyG) index, high density lipoprotein cholesterol (HDLC), and MAFLD were nonlinear and the cutoff values for lean MAFLD and overweight MAFLD were different. The nomogram was constructed based on seven predictors: glycosylated hemoglobin A1c (HbA1c), serum ferritin (SF), ALT, UA, BMI, TyG index, and age. In the validation cohort, the area under the ROC curve was 0.866 (95% CI 0.842-0.891), with 83.8% sensitivity and 76.6% specificity at the optimal cutoff. The PPV and NPV was 63.3% and 90.8%, respectively. Furthermore, we used fivefold cross-validation and the average area under the ROC curve was 0.866 (Figure S3). The calibration curves for the model's predictions and the actual outcomes were in good agreement. The DCA findings demonstrated that the nomogram model was clinically useful throughout a broad threshold probability range. CONCLUSIONS Lean and overweight MAFLD exhibit distinct metabolic profiles. The nomogram model developed in this study is designed to assist clinicians in the early identification of high-risk individuals with lean MAFLD, including those with a normal BMI but at metabolic risk, as well as those with abnormal blood lipid, glucose, uric acid or transaminase levels. In addition, this model enhances screening efforts in communities and medical screening centers, ultimately ensuring more timely and effective medical services for patients.
Collapse
Affiliation(s)
- Ruya Zhu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Caicai Xu
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China
| | - Suwen Jiang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jianping Xia
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Sijia Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| | - Jianjun Lou
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China.
| |
Collapse
|