1
|
Wang D, Miao J, Zhang L, Zhang L. Research advances in the diagnosis and treatment of MASLD/MASH. Ann Med 2025; 57. [DOI: 10.1080/07853890.2024.2445780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Dekai Wang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinxian Miao
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihua Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Jiang L, Yi R, Chen H, Wu S. Quercetin alleviates metabolic-associated fatty liver disease by tuning hepatic lipid metabolism, oxidative stress and inflammation. Anim Biotechnol 2025; 36:2442351. [PMID: 39718035 DOI: 10.1080/10495398.2024.2442351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
The natural flavonoid quercetin, which exhibits a range of biological activities, has been implicated in liver disease resistance in recent research. In vivo study attesting to quercetin's protective effect against metabolic-associated fatty liver disease (MAFLD) is inadequate, however. Here, our investigation explored the potential benefits of quercetin in preventing MAFLD in C57BL/6 mice fed a high-fat diet (HFD). The results revealed that quercetin ameliorated the aberrant enhancement of body and liver weight. The hepatic histological anomalie induced by MAFLD were also mitigated by quercetin. HFD-induced imbalance in serum LDL, HDL, AST, ALT, TG, and LDH was mitigated by quercetin. Mechanically, we found that quercetin improved lipid metabolism by reducing lipogenesis proteins including ACC, FASN, and SREBP-1c and enhancing β-oxidation proteins including PPARα and CPT1A. In vitro study demonstrated that quercetin regulated hepatic lipid metabolism by targeting SREBP-1c and PPARα. Additionally, quercetin enhanced the antioxidant capacity in HFD-treated mice by downregulating Nrf2 and HO-1 expressions and upregulating SOD and GPX1 expressions. The hyper-activation of inflammation was also restored by quercetin via eliminating the phosphorylation of IκBα and NF-κB p65. Collectively, our observations highlight that quercetin exerts hepatoprotective properties in MAFLD mice by regulating hepatic lipid metabolism, oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Ling Jiang
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Rong Yi
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Huan Chen
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Shuwu Wu
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| |
Collapse
|
3
|
Tanaka M, Akiyama Y, Mori K, Hosaka I, Endo K, Ogawa T, Sato T, Suzuki T, Yano T, Ohnishi H, Hanawa N, Furuhashi M. Machine learning-based analyses of contributing factors for the development of hypertension: a comparative study. Clin Exp Hypertens 2025; 47:2449613. [PMID: 39773295 DOI: 10.1080/10641963.2025.2449613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVES Sufficient attention has not been given to machine learning (ML) models using longitudinal data for investigating important predictors of new onset of hypertension. We investigated the predictive ability of several ML models for the development of hypertension. METHODS A total of 15 965 Japanese participants (men/women: 9,466/6,499, mean age: 45 years) who received annual health examinations were randomly divided into a training group (70%, n = 11,175) and a test group (30%, n = 4,790). The predictive abilities of 58 candidates including fatty liver index (FLI), which is calculated by using body mass index, waist circumference and levels of γ-glutamyl transferase and triglycerides, were investigated by statistics analogous to the area under the curve (AUC) in receiver operating characteristic curve analyses using ML models including logistic regression, random forest, naïve Bayes, extreme gradient boosting and artificial neural network. RESULTS During a 10-year period (mean period: 6.1 years), 2,132 subjects (19.1%) in the training group and 917 subjects (19.1%) in the test group had new onset of hypertension. Among the 58 parameters, systolic blood pressure, age and FLI were identified as important candidates by random forest feature selection with 10-fold cross-validation. The AUCs of ML models were 0.765-0.825, and discriminatory capacity was significantly improved in the artificial neural network model compared to that in the logistic regression model. CONCLUSIONS The development of hypertension can be simply and accurately predicted by each ML model using systolic blood pressure, age and FLI as selected features. By building multiple ML models, more practical prediction might be possible.
Collapse
Affiliation(s)
- Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Tanaka Medical Clinic, Yoichi, Japan
| | - Yukinori Akiyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Japan
| | - Kazuma Mori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Itaru Hosaka
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Endo
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toru Suzuki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Natori Toru Internal Medicine and Diabetes Clinic, Natori, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Nagisa Hanawa
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
4
|
Mekontso JG, Nnang JY, Tembi TB, Kortim AB, Nguefang GL, Wagner J, Bernstein M. Efficacy, Safety, and Tolerability of Farnesoid X Receptor Agonists in the Treatment of Metabolic Dysfunction-associated Steatotic Liver Disease: A Systematic Review and Meta-analysis. J Clin Exp Hepatol 2025; 15:102563. [PMID: 40337255 PMCID: PMC12053700 DOI: 10.1016/j.jceh.2025.102563] [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: 02/17/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
Background/Aims Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of chronic liver disease. Farnesoid X receptor (FXR) agonists are emerging as promising therapies for fibrosis, steatosis, and metabolic dysfunctions. However, its efficacy and safety remain unclear. Methods A systematic search of PubMed, Embase, and Cochrane databases identified randomized controlled trials (RCTs) comparing FXR agonists with placebo in patients with MASLD. The main outcomes included improvement in fibrosis without worsening steatohepatitis, changes in liver chemistry and lipid profiles, and liver fat content (LFC). The safety outcomes assessed included side effects and treatment discontinuation rates. Heterogeneity was evaluated using I² statistics, with a random-effects model applied to the pooled analyses. Results Ten RCTs involving 3,779 patients were included, of which 2,527 (67%) were randomized to receive FXR agonists. FXR agonists significantly improved fibrosis by ≥ 1 stage (RR, 1.52; 95% CI: [1.23, 1.88]; P < 0.0001) and reduced LFC (mean difference: -4.9%; 95% CI: [-8.26, -1.55]; P < 0.001). A higher proportion of patients achieved a ≥30% reduction in LFC (42.8% vs. 18.4%; RR, 2.42; 95% CI: [1.69, 3.46]; P < 0.00001). Significant reductions in alanine aminotransferase and gamma glutamyltransferase levels were observed, whereas alkaline phosphatase levels were increased. FXR agonists were associated with a slight reduction in High-Density Lipoprotein (HDL) cholesterol levels and a higher incidence of pruritus (37.8% vs. 18.7%; RR, 2.67; 95% CI: [1.63, 4.38]; P < 0.00001), leading to higher treatment discontinuation rates. Conclusion FXR agonists have the potential to improve fibrosis and steatosis in MASLD patients. However, safety concerns still remain. Further research is required to determine the long-term efficacy and tolerability of these drugs.
Collapse
Affiliation(s)
- Joel G.K. Mekontso
- New York City Health and Hospitals, South Brooklyn Health, Brooklyn, NY, USA
| | - Joseph Y.B. Nnang
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Ticha B.T. Tembi
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | - Guy L. Nguefang
- Texas Tech University Health Sciences Center, Odessa, TX, USA
| | - Justin Wagner
- New York City Health and Hospitals, South Brooklyn Health, Brooklyn, NY, USA
| | - Michael Bernstein
- New York City Health and Hospitals, South Brooklyn Health, Brooklyn, NY, USA
| |
Collapse
|
5
|
Chen Y, Liao Z, Mao J, Wang W, Liu Y, Dai W, Wen Z, Liu S, Chen Y, Ma Y, Wang X, Li Z. Discovery of the first-in-class FABP/PPAR multiple modulator for the treatment of metabolic dysfunction-associated steatohepatitis. Eur J Med Chem 2025; 291:117635. [PMID: 40279770 DOI: 10.1016/j.ejmech.2025.117635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a complex metabolic syndrome, and the development of new drugs is urgently needed. Fatty acid binding proteins (FABPs) and peroxisome proliferator-activated receptors (PPARs) play an important role in the regulation of lipid absorption, metabolism and inflammation. Considering the synergistic effect of FABP and PPAR in the regulation of MASH pathophysiology, the development of FABP/PPAR multiple modulators might be a promising anti-MASH strategy. Herein, the first-in-class FABP/PPAR multiple modulators were designed by hybrid resveratrol and PPARs agonist Elafibranor. Among them, the compound 27 was identified as the optimal FABP/PPAR multiple modulator (FABP1 IC50 = 0.65 μM, FABP4 IC50 = 1.08 μM, PPARα EC50 = 9.19 μM, PPARγ EC50 = 2.20 μM, PPARδ EC50 = 1.58 μM). Further MST assay confirmed the direct interaction of compound 27 and FABP1, providing a robust validation of its target specificity. In MASH mice, compound 27 exhibited a better therapeutic effect than clinical candidate obeticholic acid in ameliorating multiple pathological features of MASH. This study reported the successful discovery of the first-in-class FABP/PPAR multiple modulators, which provided preliminary evidence that such multi-target agents have broad medical prospects.
Collapse
Affiliation(s)
- Ya Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zibin Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jianming Mao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wenxin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yuxia Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wei Dai
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
| | - Zheng Wen
- Department of Emergency, Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, PR China
| | - Sishi Liu
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Yayi Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yiming Ma
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoying Wang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
6
|
Zhou H, Chen H, Wu D, Lu H, Wu B, Dong Z, Yang J. Exercise self-efficacy in older adults with metabolic-associated fatty liver disease: A latent profile analysis. SPORTS MEDICINE AND HEALTH SCIENCE 2025; 7:285-291. [PMID: 40264834 PMCID: PMC12010361 DOI: 10.1016/j.smhs.2024.05.001] [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: 11/30/2023] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 04/24/2025] Open
Abstract
China has a high prevalence rate of Metabolic-associated fatty liver disease (MAFLD), and there is currently limited understanding of the levels of exercise self-efficacy (ESE) among individuals with MAFLD. The objective was to explore the potential ESE patterns in older adults with MAFLD. A cross-sectional study was conducted on 800 older adults with fatty liver disease from five communities from April 20, 2023 to August 15, 2023. Latent profile analysis (LPA) and k-means clustering were used to determine the optimal number of ESE groups. Using univariate analysis and multivariate logistic regression to investigate the factors influencing profiles of ESE. A sample of 775 subjects met the diagnostic criteria for MAFLD. LPA yielded three profiles: the low-ESE, mild-ESE, and high-ESE groups, which comprised 25%, 28%, and 47% of the sample, respectively. K-means clustering further supported the categorization of ESE into three distinct classes. The multivariate logistic regression analysis revealed that diabetes, arthritis and/or arthrosis, as well as companionship during PA were significant influencing factors for the different profiles (p < 0.05). Our findings suggest that the ESE of older patients with MAFLD is primarily at a moderate level or above. There was population heterogeneity in ESE among older patients with MAFLD. Diabetes mellitus, arthritis, and/or arthrosis, as well as companionship during PA were significant factors in influencing the likelihood of having high ESE.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
- Department of Medicine, Jiangnan University, Jiangsu, China
| | - Haiyan Chen
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
- Department of Medicine, Jiangnan University, Jiangsu, China
| | - Di Wu
- Xingcheng Special Care Rehabilitation Center, Liaoning, China
| | - Hanxiao Lu
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
- Department of Medicine, Jiangnan University, Jiangsu, China
| | - Bo Wu
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
- Department of Medicine, Jiangnan University, Jiangsu, China
| | - Zhixia Dong
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Jun Yang
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Jiangsu, China
| |
Collapse
|
7
|
Askeland-Gjerde DE, Westlye LT, Andersson P, Korbmacher M, de Lange AM, van der Meer D, Smeland OB, Halvorsen S, Andreassen OA, Gurholt TP. Mediation Analyses Link Cardiometabolic Factors and Liver Fat With White Matter Hyperintensities and Cognitive Performance: A UK Biobank Study. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100488. [PMID: 40330223 PMCID: PMC12052680 DOI: 10.1016/j.bpsgos.2025.100488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 05/08/2025] Open
Abstract
Background Liver fat is associated with cardiometabolic disease, cerebrovascular disease, and dementia. Cerebrovascular disease, most often cerebral small vessel disease, identified by magnetic resonance imaging as white matter hyperintensities (WMHs) often contributes to dementia. However, liver fat's role in the relationship between cardiometabolic risk, WMHs, and cognitive performance is unclear. Methods In the UK Biobank cohort (N = 32,461, 52.6% female; mean age 64.2 ± 7.7 years; n = 23,354 in the cognitive performance subsample), we used linear regression to investigate associations between cardiometabolic factors measured at baseline and liver fat, WMHs, and cognitive performance measured at follow-up, which was 9.3 ± 2.0 years later on average. We used structural equation modeling to investigate whether liver fat mediated associations between cardiometabolic factors and WMHs and whether WMHs mediated associations between liver fat and cognitive performance. Results Nearly all cardiometabolic factors were significantly associated with liver fat (|r| range = 0.03-0.41, p = 3.4 × 10-8 to 0) and WMHs (|r| = 0.04-0.15, p = 5.8 × 10-13 to 7.0 × 10-159) in regression models. Liver fat was associated with WMHs (r = 0.11, p = 4.3 × 10-82) and cognitive performance (r = -0.03, p = 1.6 × 10-7). Liver fat mediated the associations between cardiometabolic factors and WMHs (|βmediation| = 0.003-0.027, p mediation = 1.9 × 10-8 to 0), and WMHs mediated the associations between liver fat and cognitive performance (βmediation = -0.01, p mediation = 0). Conclusions Our findings indicate that liver fat mediates associations between cardiometabolic factors and WMHs and that WMHs mediate the association between liver fat and cognitive performance. This suggests that liver fat may be important for understanding the effects of cardiometabolic factors on cerebrovascular disease and cognitive function. Experimental studies are warranted to determine relevant targets for preventing vascular-driven cognitive impairment.
Collapse
Affiliation(s)
- Daniel E. Askeland-Gjerde
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | | | - Max Korbmacher
- Neuro-SysMed Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Ann-Marie de Lange
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Dennis van der Meer
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Olav B. Smeland
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Halvorsen
- Department of Cardiology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Tiril P. Gurholt
- Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
8
|
Sui X, Zhao J, Yang Y, Yang Y, Li K, Wang Z, Liu Z, Lu R, Zhang G. Epidemiological Dynamics of Burden and Health Inequalities in Metabolic Dysfunction-associated Steatotic Liver Disease in Adolescents at Global, Regional, and National Levels, 1990-2021. J Clin Exp Hepatol 2025; 15:102537. [PMID: 40226388 PMCID: PMC11987614 DOI: 10.1016/j.jceh.2025.102537] [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: 12/11/2024] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD) has become one of the major causes of chronic liver disease among adolescents. However, epidemiological studies on MASLD in adolescents are still insufficient. In this study, we aim to investigate the global burden and the trend of MASLD in adolescents from 1990 to 2021. Methods The age-standardized incidence, prevalence, mortality, and disability-adjusted life years (DALYs) of MASLD were calculated based on the Global Burden of Disease (GBD) 2021 study and stratified by sex, socio-demographic index (SDI), GBD regions, and countries. The temporal trends were examined using the average annual percentage change (AAPC) and joinpoint regression. Results From 1990 to 2021, the global trends of age-standardized incidence rate (ASIR) and age-standardized prevalence rate (ASPR) of MASLD show notable increase, and the male is significantly higher than the female in adolescents. According to the incidence and prevalence, nations with low SDI confront a higher burden of MASLD. Besides, the inequality of incidence and prevalence between different SDI regions have shrunk in 2021, but the inequality of DALYs and mortality are still exacerbated. Decomposition analysis revealed that population growth and epidemiological changes were the main reasons for the increase in the incidence of MASLD. Conclusion From 1990 to 2021, there is a significant upward trend in the incidence of MASLD among adolescents worldwide. Of particular note are male adolescents, East Asian regions, and groups living in high SDI countries.
Collapse
Affiliation(s)
- Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Junde Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Yuxin Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Yikun Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Kaifeng Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Zuocheng Wang
- Australian National University Research School of Biology, Canberra, 2601, Australia
| | - Ziqi Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Ruining Lu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| | - Guiju Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250011, China
| |
Collapse
|
9
|
Liu Z, Hou J, Tian M, Zhang Y, Huang D, Zhao N, Ma Y, Cui S. Hypoxia ameliorates high-fat-diet-induced hepatic lipid accumulation by modulating the HIF2α/PP4C signaling. Cell Signal 2025; 131:111751. [PMID: 40112904 DOI: 10.1016/j.cellsig.2025.111751] [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: 12/07/2024] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Hepatic lipid accumulation is a hallmark of metabolically associated fatty liver disease (MAFLD), which contributes to the progression of cirrhosis and even hepatoma. However, the underlying mechanisms remain poorly understood. Protein phosphatase 4C (PP4C) is an important enzyme that exists widely in the body and participates in cell metabolism. Hypoxia can affect the development of metabolic diseases. In this study, we investigated the role of PP4C in hepatic lipid metabolism under hypoxia in vivo and in vitro. Hypoxia-inducible factor 2α (HIF2α), PP4C, phosphorylated AU-rich element RNA-binding factor 1(pAUF1), acetyl-CoA carboxylase 1 (ACC1), and carnitine palmitoyl transferase-1 (CPT1) were analyzed via western blotting and immunofluorescence. The mechanism by which PP4C affects hepatic lipid accumulation under hypoxia was evaluated in stable transfected cell lines. Compared with those in the 2200 m HFD group, body weight, triglyceride (TG), total cholesterol (TC), amino alanine transferase (ALT), aspartate transaminase (AST), and lipid accumulation were lower in the 4500 m HFD group (P < 0.05). Compared with those in the 4500 m ND group, ACC1 and PP4C levels were lower than in the 4500 m HFD group, but HIF2α, pAUF1, and CPT1 levels were greater (P < 0.05). Knockdown of HIF2α prevented the hypoxia-induced reduction of PP4C, confirming the regulatory role of the HIF2α-PP4C axis in hepatic lipid metabolism. PP4C could affect the phosphorylation and expression localization of AU-rich element RNA-binding factor 1 (AUF1). PP4C enhanced lipid accumulation by reducing pAUF1, while the knockdown of PP4C had the opposite effect; pAUF1 had no change. Compared with those in the control group, ACC1 levels were decreased and CPT1 levels were increased in the AUF1 overexpression group, whereas ACC1 and CPT1 levels were not altered in the AUF1 knockdown group (P < 0.05). In conclusion, hypoxia might improve lipid accumulation by downregulating PP4C via HIF2a. PP4C is involved in hepatic lipid metabolism by regulating AUF1 phosphorylation under different oxygen concentrations. PP4C might be a promising target for treating hepatic lipid accumulation.
Collapse
Affiliation(s)
- Zhe Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining 810000, China; Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Xining 810000, China; Department of Gynecology, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Jing Hou
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - MeiYuan Tian
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - YaoGang Zhang
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - DengLiang Huang
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Na Zhao
- Graduate School of Qinghai University, Qinghai University, Xining 810000, China
| | - Yanyan Ma
- Central Laboratory/Research Key Laboratory for Echinococcosis, Affiliated Hospital of Qinghai University, Xining 810000, China; Department of Scientific Research Office, Affiliated Hospital of Qinghai University, Xining 810000, China.
| | - Sen Cui
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining 810000, China.
| |
Collapse
|
10
|
Li L, Gao W, Yao F, Li J, Sang W, Zhang R. Innovative nanomedicine approaches for the management of nonalcoholic fatty liver disease. J Control Release 2025; 382:113680. [PMID: 40180250 DOI: 10.1016/j.jconrel.2025.113680] [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: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disorder globally. The prevalence of NAFLD in the general population is estimated to be 25-30 %, making it the most common chronic liver condition in China as well as worldwide. Given the escalating disease burden and the scarcity of effective therapeutic interventions, there is a pressing unmet clinical need. Consequently, the development of novel pharmaceuticals has emerged as a pivotal research focus in recent years. Moreover, the advent of nano-delivery technology offers innovative solutions for NAFLD drug therapy. This paper presents a comprehensive examination of the pathogenesis and therapeutic targets of NAFLD. It critically reviews the latest advancements in nanomedicine research pertinent to NAFLD treatment. The review synthesizes a broad range of research findings to bridge the gap between current knowledge and emerging therapeutic strategies, and aims to inform and guide future research directions in NAFLD management.
Collapse
Affiliation(s)
- Limeng Li
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Weiqi Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China; Shanxi Academy of Advanced Research and Innovation (SAARl), Taiyuan, 030032, China
| | - Fengyang Yao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Jiayi Li
- School of Forensic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Wei Sang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Institute of Medical Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
11
|
Jin H, Liang Z, Hu X, Li X, Liu Z, Qiao Y, Cheng Y, Yao H, Liu Y. Comparative association of MAFLD/MASLD and Subtypes with Cardiovascular Diseases Outcomes. Nutr Metab Cardiovasc Dis 2025; 35:104024. [PMID: 40189471 DOI: 10.1016/j.numecd.2025.104024] [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: 11/20/2024] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) acts as an alternative for demarcating metabolic dysfunction-associated fatty liver disease (MAFLD). This study aimed to investigate the factors that significantly influence the relationship between MAFLD and MASLD in relation to the incidence of major cardiovascular outcomes. METHODS AND RESULTS A total of 340,998 participants in the UK Biobank study were included. Multivariable Cox proportional hazards models were used to estimate the effect of MAFLD and MASLD on the outcomes of cardiovascular diseases (CVDs) (coronary artery disease, stroke, heart failure, and CVD-related death) with hazard ratios (HRs) and 95 % confidence intervals (CIs). A total of 126,077 (36.97 %) participants had MAFLD and 97,418 (28.57 %) had MASLD. Over a median follow-up of 13.5 years (interquartile range 12.6-14.2), there were 41,548 new events of CVDs recorded. MAFLD (HR = 1.52; 95 % CI: 1.49-1.55) and MASLD (HR = 1.42; 95 % CI: 1.39-1.45) were associated with high risks of CVDs. Among the subtypes of MAFLD and steatotic liver disease (SLD), MAFLD diabetes subtype (HR = 2.26; 95 % CI: 2.17-2.35) and alcohol-associated liver disease (ALD) (HR = 1.65; 95 % CI: 1.55-1.76) exhibited the highest risk of CVDs. MAFLD overweight without MD subtype were not associated with CVDs. The effect of MAFLD on the CVD outcomes was consistent regardless of the presence of MASLD. CONCLUSION The metabolic health status and alcohol consumption function as more critical factors than obesity in assessing CVD outcomes in participants with MAFLD or MASLD.
Collapse
Affiliation(s)
- Huizhen Jin
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Zhuoshuai Liang
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Xinmeng Hu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Xiaoyang Li
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Zhantong Liu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Yichun Qiao
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China
| | - Yi Cheng
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hanxin Yao
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yawen Liu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun, 130021, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, School of Public Health, Jilin University, Changchun, 130062, China.
| |
Collapse
|
12
|
Kim JH, Lee Y, Nam CM, Kwon YJ, Lee JW. Impact of cardiometabolic risk factors for metabolic dysfunction-associated steatotic liver disease on mortality. Nutr Metab Cardiovasc Dis 2025; 35:103965. [PMID: 40187915 DOI: 10.1016/j.numecd.2025.103965] [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: 11/16/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a potential independent risk factor for cardiovascular disease (CVD)-associated and all-cause mortalities as they share common risk factors. We investigated the association between cardiometabolic risk factors for MASLD and CVD-associated and all-cause mortality risks in middle-aged and older Korean adults. METHODS AND RESULTS We used data from the Korean Genome and Epidemiology Study, a population-based prospective cohort study. Five cardiometabolic risk factors were assessed. MASLD was defined as liver steatosis with a fatty liver index (FLI) ≥60 and at least one cardiometabolic risk factor. The non-MASLD group included individuals with a FLI <60 or FLI ≥60 without cardiometabolic risk factors. The primary outcomes were CVD-associated and all-cause mortalities. Cox proportional hazard models were used to evaluate the association between cardiometabolic risk factors for MASLD and mortalities, adjusting for covariates. Multivariable Cox regression analysis revealed that the MASLD group had increased CVD-associated and all-cause mortality risks compared to the non-MASLD group. The presence of three or more and one or more cardiometabolic risk factors significantly increased the CVD-associated and all-cause mortality rate, respectively. The combination of hypertriglyceridemia, low high-density lipoprotein cholesterol (HDL-C), and high glucose concentrations significantly increased both CVD-associated (hazard ratio [HR] 3.64; 95 % confidence interval [CI] 1.44-9.22; p = 0.006) and all-cause (HR 4.57; 95 % CI: 1.74-12.05; p = 0.002) mortality risks. CONCLUSION Cardiometabolic risk factors for MASLD are strongly associated with higher CVD-associated and all-cause mortality risks, highlighting the need to manage hypertriglyceridemia, low HDL-C, and high glucose concentrations.
Collapse
Affiliation(s)
- Jung-Hwan Kim
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yaeji Lee
- Department of Biostatistics and Computing, Yonsei University, Seoul, 03722, Republic of Korea
| | - Chung-Mo Nam
- Department of Health Informatics and Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, 16995, Republic of Korea.
| | - Ji-Won Lee
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
13
|
Ni D, Qi Z, Ma S, Wang Y, Liang D, Zhang X, Man Y, Chen J, Dou K, Li G. Membrane-associated ring-CH-type finger 2 protects against metabolic dysfunction-associated fatty liver disease by targeting fatty acid synthase. Mol Metab 2025; 96:102137. [PMID: 40189099 DOI: 10.1016/j.molmet.2025.102137] [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: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as an important public health concern that poses a significant threat to human health and imposes a substantial economic burden. Research has demonstrated that ubiquitin ligase-mediated substrate protein ubiquitination is a pivotal factor influencing liver lipid homeostasis and metabolic abnormalities in MAFLD. Nevertheless, the specific enzyme molecules implicated in this regulatory process remain to be elucidated. We have published a transcriptome-overexpressing ubiquitin ligase, membrane-associated ring-CH-type finger 2 (MARCH2), in HepG2 cells, and subsequent reanalysis of these transcriptome data revealed a close association between MARCH2 and lipid metabolism. METHODS By employing a range of methodologies, including recombinant adeno-associated virus (rAAV) transduction, lentiviral transduction, immunoblotting, quantitative PCR, tissue section staining, ubiquitination assays, serum biochemical analysis, immunoprecipitation, and mass spectrometry, this study investigated the functions and mechanisms of MARCH2 in the progression of MAFLD at the molecular, cellular, and organismal levels. RESULTS Overexpression of MARCH2, but not its catalytically inactive ligase variant, inhibited lipid accumulation in HepG2 cells. Additionally, MARCH2 undergoes K48-linked self-polyubiquitination and subsequent proteasomal degradation in response to oleic acid/palmitic acid stimulation. Furthermore, knockout of MARCH2 exacerbates the progression of MAFLD-related phenotypes, including increased body weight, impaired glucose tolerance, reduced insulin sensitivity, hypercholesterolemia, hepatic lipid accumulation, and steatosis, in high-fat diet-fed mice, irrespective of sex. Mechanistically, MARCH2 facilitates the polyubiquitination and degradation of fatty acid synthase (FASN) in the de novo lipogenesis pathway. And liver-specific overexpression of MARCH2 by rAAV effectively reduces FASN levels and further ameliorates MAFLD in ob/ob mice. CONCLUSIONS MARCH2 undergoes self-ubiquitination and plays an important role in maintaining the liver lipid homeostasis of MAFLD, and drug intervention in the MARCH2-FASN axis is a promising approach for treating systemic metabolic abnormalities in MAFLD.
Collapse
Affiliation(s)
- Dongsheng Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Zhaolai Qi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Shuang Ma
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Yuefeng Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Dehuan Liang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Fifth School of Clinical Medicine (Beijing Hospital), Peking University, Beijing, 100730, PR China
| | - Xiyue Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Yong Man
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, PR China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, PR China.
| | - Kefei Dou
- Cardiometabolic Medicine Center, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, PR China.
| | - Guoping Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
14
|
Liu X, Yu H, Hu T, He Y, Li Y, Yuan Q, Dong M, Liu D, Xu Y, Mao L. G3BP1, a stress granule core protein, ameliorates metabolic dysfunction-associated fatty liver disease by attenuating hepatocyte lipid deposition. Diabetes Obes Metab 2025; 27:2985-2995. [PMID: 40230220 DOI: 10.1111/dom.16302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 04/16/2025]
Abstract
AIM Abnormal lipid accumulation is an important cause of metabolic dysfunction-associated fatty liver disease (MAFLD) progression and can induce several stress responses within cells. This study is the first to explore the role and molecular mechanism of stress granules (SGs) in MAFLD. METHODS A gene knock-down model of G3BP1, a core SG molecule in mice and HepG2 cells, was constructed to explore the role of SGs in MAFLD induced in vivo by a high-fat diet or in vitro by palmitic acid (PA). Methods included metabolic phenotyping; western blotting; qPCR; and immunofluorescence, haematoxylin/eosin and masson staining. The downstream molecules of G3BP1 and its specific molecular mechanism were screened using RNA sequencing (RNA-seq). RESULTS G3BP1 and TIA1 expression were upregulated in high-fat diet-fed mouse liver tissues and PA-induced HepG2 cells, and the two molecules showed significantly increased colocalisation. G3BP1 knock-down slightly increased TIA1 expression in the livers of obese mice but not in lean mice. G3BP1 deficiency aggravated liver lipid deposition and insulin resistance in obese mice, and this phenotype was confirmed in vitro in PA-induced hepatocytes. RNA-seq demonstrated that G3BP1 slowed down MAFLD progression by inhibiting APOC3, possibly through a mechanistic suppression of APOC3 entry into the nucleus. CONCLUSION This study reveals for the first time a protective role for SGs in MAFLD. Specifically, knocking down the core G3BP1 molecule in SGs aggravated the progression of fatty acid-induced MAFLD through a mechanism that may involve the nuclear entry of APOC3. These findings provide a new therapeutic direction for MAFLD.
Collapse
Affiliation(s)
- Xingjing Liu
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Huimei Yu
- Department of Endocrinology, Huai'an Hospital Affiliated to Yangzhou University, China
| | - Tongtong Hu
- Department of Cardiovascular Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Yu He
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Yiming Li
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Qi Yuan
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Meijuan Dong
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Dezhen Liu
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| | - Yue Xu
- Department of Endocrinology, Huai'an Hospital Affiliated to Yangzhou University, China
| | - Li Mao
- Department of Endocrinology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, China
| |
Collapse
|
15
|
Li R, Wang T, Luo H, Fan Y, Guan Y, Tian Y. Causal effects and mediating pathways of metabolic dysfunction-associated fatty liver disease on novel subtypes of adult-onset diabetes: A two-step Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103976. [PMID: 40180825 DOI: 10.1016/j.numecd.2025.103976] [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] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND AND AIM Based on an in-depth understanding of diabetes heterogeneity, five novel subtypes of adult-onset diabetes have been identified. This study investigates the differential impact of metabolic dysfunction-associated fatty liver disease (MAFLD) on these subtypes using a Mendelian randomization (MR) approach, while also exploring modifiable mediating factors. METHODS AND RESULTS Genetic variants associated with MAFLD were selected at the genome-wide significance threshold (P < 5 × 10-8), with 16 variants used to assess causal associations with the risk of severe autoimmune diabetes (SAID), mild obesity-related diabetes (MOD), severe insulin-resistant diabetes (SIRD), severe insulin-deficient diabetes (SIDD), and mild age-related diabetes (MARD). Two-step MR was used to estimate total, direct, and mediated effects, analyzing 55 potential mediators across five domains. The primary method used was inverse variance weighting (IVW) along with a series of sensitivity analyses to ensure robustness of the results. Genetically predicted MAFLD was significantly associated with increased risk of MARD (OR = 1.171, 95 % CI 1.091-1.256), SIDD (OR = 1.158, 95 % CI 1.056-1.270), and SIRD (OR = 1.267, 95 % CI 1.119-1.434), with suggestive evidence for SAID (OR = 1.161, 95 % CI 1.016-1.327), but no association with MOD. Among all subtypes, waist-to-hip ratio adjusted for BMI (WHRadjBMI) was a common mediator, with liver fat, alanine aminotransferase (ALT), gamma-glutamyl transferase, fasting insulin (FI), and BMI mediating at least three subtypes. Liver fat accounted for the largest proportion of mediation (43.63 %-73.09 %), followed by ALT (8.99 %-17.93 %) and FI (6.81 %-13.04 %). CONCLUSION This study underscores the causal relationship between MAFLD and specific diabetes subtypes, highlighting the importance of integrated management of liver lipid metabolism, abdominal obesity, and blood glucose regulation. These findings support personalized intervention strategies.
Collapse
Affiliation(s)
- Rongrong Li
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Ting Wang
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Hongping Luo
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yawei Fan
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yan Guan
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Ye Tian
- Nursing Department, Tongii Hospital, Tongii Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
16
|
Shora HA, El-Deen IM, El-Lithy NM, Abo-Elmatty DM, Khirallah SM. Growth differentiation factor-15: A marker for diabetic kidney disease in patients with metabolic-associated fatty liver disease. J Diabetes Complications 2025; 39:109037. [PMID: 40233467 DOI: 10.1016/j.jdiacomp.2025.109037] [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: 01/30/2025] [Revised: 03/23/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Growth/differentiation factor-15 (GDF-15) plays a crucial role in modulating inflammation and fibrosis and is emerging as a potential biomarker in metabolic diseases. This study investigated the association between circulating GDF-15 levels and the development of diabetic kidney disease (DKD) in patients with type 2 diabetes mellitus (T2DM), with or without metabolic dysfunction-associated fatty liver disease (MAFLD). METHODS In this prospective observational study, participants were stratified into three groups: those with T2DM and MAFLD (Group A) and those with T2DM and no MAFLD (Group B), alongside a healthy control group (Group C). Comprehensive clinical evaluations and laboratory assessments, including measurements of GDF-15, metabolic profiles, liver enzymes, and renal function, were performed. RESULTS GDF-15 levels demonstrated a stepwise elevation from controls to T2DM patients, with the highest levels observed in patients with MAFLD (1929 ± 412 pg/mL in Group A, 1655 ± 368 pg/mL in Group B, and 1246 ± 245 pg/mL in Group C; p < 0.001). DKD was diagnosed in 51.4 % of patients in Group A compared to 37.1 % in Group B (p < 0.001). Multivariate analysis identified GDF-15 as the sole independent predictor of DKD (p = 0.01). ROC curve analysis revealed that a GDF-15 cutoff value of ≥1890.51 pg/mL provided an AUC of 0.951, with a sensitivity of 94.2 % and specificity of 87.1 %. CONCLUSION Elevated GDF-15 levels are independently associated with an increased risk of DKD in T2DM patients, particularly in those with MAFLD. These findings highlight the potential of GDF-15 as an early biomarker for DKD.
Collapse
Affiliation(s)
| | - Ibrahim M El-Deen
- Chemistry Department, Faculty of Science, Port Said University, Port Said 42526, Egypt
| | - Naglaa M El-Lithy
- Chemistry Department (Biochemistry Division), Faculty of Science, Port Said University, Port Said 42526, Egypt
| | - Dina M Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Salma M Khirallah
- Chemistry Department (Biochemistry Division), Faculty of Science, Port Said University, Port Said 42526, Egypt.
| |
Collapse
|
17
|
Gogola T, Pitkänen S, Huovinen M, Laitinen H, Küblbeck J. Association between phthalate exposure and metabolic dysfunction-associated steatotic liver disease (MASLD) - Systematic literature review. ENVIRONMENTAL RESEARCH 2025; 273:121186. [PMID: 39986424 DOI: 10.1016/j.envres.2025.121186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rising globally. Recent studies have suggested connections between exposure to endocrine disrupting chemicals (EDCs) and the development of MASLD. Phthalates, which are commonly utilized as plasticizers, in building materials and consumer items, exhibit endocrine disrupting effects and have been shown to interfere with lipid metabolism in mechanistic studies. The objective of this systematic review was to examine the association between MASLD and exposure to phthalates in the adult human populations. We searched PubMed, Scopus and Web of Science for studies published from the inception of each database until December 12, 2024. The literature search yielded 10 cross-sectional studies, which were analyzed in detail. The key findings of this study indicate a potential correlation between the prevalence of MASLD and exposure to certain phthalates. Among the phthalates examined, the metabolites of bis(2-ethylhexyl) phthalate (DEHP) - namely MECPP, MEHHP, and MEOHP, demonstrated the strongest and most frequent associations with MASLD. All the current studies followed cross-sectional study designs, which limits the possibility to establish a causal relationship between MASLD and phthalate exposure. Therefore, longitudinal studies are needed to corroborate these findings and shed light on the involvement of phthalate exposure in MASLD.
Collapse
Affiliation(s)
- Tomasz Gogola
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Sini Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland.
| | - Marjo Huovinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | | | - Jenni Küblbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| |
Collapse
|
18
|
Bhattacharya I, Maity DK, Kumar A, Sarkar S, Bhattacharya T, Sahu A, Sreedhar R, Arumugam S. Beyond obesity: lean metabolic dysfunction-associated steatohepatitis from unveiling molecular pathogenesis to therapeutic advancement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04257-x. [PMID: 40366398 DOI: 10.1007/s00210-025-04257-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD), now known by the name of metabolic dysfunction-associated fatty liver disease (MAFLD), with increased global incidence, has been recognized as a significant metabolic disorder. NAFLD includes a spectrum liver disease from hepatocellular fat accumulation (isolated steatosis) to an advanced form of liver injury known as nonalcoholic steatohepatitis (NASH), which refers to distinct histologic features, including hepatocellular steatosis and injury, necroinflammation, and eventually fibrosis. Nonobese or lean individuals associated with metabolic dysregulation usually demonstrated diverse risk factors compared to obese MAFLD. The presence of normal range body mass index (BMI) and excess visceral adiposity with increased cardiometabolic and renal comorbidities, along with sarcopenia, has been evidenced to be associated with lean MASH. Genetic predispositions accompanying lifestyle and environmental factors contribute to disease initiation and progression. The genetic influence in pathophysiology indicated the significant contributions of the following genes: PNPLA3, TM6SF2, APOB, LIPA, MBOAT7, and HSD17B13, and the impact of their disease-specific variants in the development of obesity-independent MASH. The epigenetic modifications exhibited differential DNA methylation patterns in the genes involved in lipid metabolism, particularly hypomethylation of PEMT. Diet-induced and genetic animal models of lean MASH, including Slc: Wistar/ST rats, PPAR-α, PTEN, and MAT1A knockout mice models, are indicated to be pivotal in the exploration of disease progression and observing the effect of therapeutic interventions. This comprehensive review comprises the molecular and genetic pathophysiology, molecular diagnostics, and therapeutic aspects of lean MASH to enunciate a diagnostic approach that combines detailed clinical phenotyping regarding genomic analysis.
Collapse
Affiliation(s)
- Indrajit Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Deep Kumar Maity
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amit Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Sampriti Sarkar
- School of Biosciences & Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - Teeshyo Bhattacharya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Amrita Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Remya Sreedhar
- School of Pharmacy, Sister Nivedita University, DG Block, Action Area I, 1/2, Newtown, Kolkata, 700156, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
19
|
Borghese MM, Feng J, Liang CL, Kienapple N, Manz KE, Fisher M, Arbuckle TE, Atlas E, Braun JM, Bouchard MF, Foster W, Ashley-Martin J. Legacy, alternative, and precursor PFAS and associations with lipids and liver function biomarkers: results from a cross-sectional analysis of adult females in the MIREC-ENDO study. Int J Hyg Environ Health 2025; 267:114592. [PMID: 40359778 DOI: 10.1016/j.ijheh.2025.114592] [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: 12/16/2024] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Legacy per- and polyfluoroalkyl substances (PFAS) can promote dyslipidemia; however, evidence is lacking for alternative and precursor PFAS. We investigated associations between serum concentrations of 31 PFAS and concurrently measured lipids and liver function biomarkers. METHODS PFAS, lipids, and liver function biomarkers were analyzed in serum samples provided by 282 adult females participating in a 2018-2021 follow-up study of a Canadian pregnancy cohort. We examined percent differences in outcomes continuously for 17 PFAS with >50% detection and as detected vs. not detected for 14 PFAS with 10-50% detection. We also examined associations with the sum of 7 PFAS recommended by the National Academies of Sciences, Engineering, and Medicine guidance report on PFAS testing and 17 PFAS. We used weighted quantile sum (WQS) and quantile g-computation models to estimate joint associations. RESULTS Each two-fold increase in concentrations of PFHxS, PFOS, PFNA, PFDA, PFHpS, and Σ7PFAS were associated with up to 7% higher total and LDL cholesterol and the TC:HDL ratio. Individuals with detectable concentrations of N-EtFOSA, N-MeFOSA, PFBS, and 9Cl-PF3ONS had up to 17% higher total and LDL cholesterol and TC:HDL. Each one-quartile increase in the mixture of 7 PFAS was associated with up to 10% higher total and LDL cholesterol. Adding additional PFAS to the mixture (17 PFAS) made estimates less precise in WQS models and attenuated associations to the null in quantile g-computation models. CONCLUSION Alternative and precursor PFAS, including replacements for legacy PFAS, are associated with higher cholesterol levels; prospective studies are required to confirm these findings.
Collapse
Affiliation(s)
- Michael M Borghese
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Jingxue Feng
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Chun Lei Liang
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Natasha Kienapple
- Health Products and Food Branch, Health Canada, 251 Sir Frederick Banting Driveway, Ottawa, ON, Canada.
| | - Katherine E Manz
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, USA.
| | - Mandy Fisher
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Tye E Arbuckle
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| | - Joseph M Braun
- Department of Epidemiology, Brown University, 121 S Main St, Providence, RI, USA.
| | - Maryse F Bouchard
- Institut National de la Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, Québec, Canada; CHU Sainte-Justine, 3175, Chem. de la Côte-Sainte-Catherine, Montréal, Québec, Canada.
| | - Warren Foster
- Department of Obstetrics and Gynecology, McMaster University, 1280 Main Street West, Hamilton, ON, Canada.
| | - Jillian Ashley-Martin
- Environmental Health Science and Research Bureau, Health Canada, 269 Laurier Ave W, Ottawa, ON, Canada.
| |
Collapse
|
20
|
Nikparast A, Sohouli MH, Forouzan K, Farani MA, Dehghan P, Rohani P, Asghari G. The association between total, animal, and plant protein intake and metabolic dysfunction-associated fatty liver disease in overweight and obese children and adolescents. Nutr J 2025; 24:75. [PMID: 40349058 PMCID: PMC12066057 DOI: 10.1186/s12937-025-01142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Dietary protein plays a crucial role in the growth and development of children and adolescents. However, recent evidence has shown inconsistent findings regarding the impact of dietary protein sources on health outcomes. This study aimed to investigate the association between total, animal, and plant protein intake and the odds of metabolic dysfunction-associated fatty liver disease (MAFLD) in overweight and obese children and adolescents. METHODS This cross-sectional study included 505 participants (52.9% males) aged 6-18 years, with a body mass index (BMI)-for-age z-score ≥ 1 based on WHO standards. MAFLD diagnosis followed established consensus definitions. Dietary intake of total, animal, and plant protein was assessed using a validated 147-item food frequency questionnaire. Adjusted logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for MAFLD across protein intake and subtype quartiles. RESULTS The participants had a mean age of 10.0 ± 2.3 years and a mean BMI-for-age z-score of 2.70 ± 1.01. Higher animal protein intake was significantly associated with an increased likelihood of MAFLD (highest vs. lowest quartile OR: 2.31; 95% CI: 1.01-5.30). Conversely, higher plant protein intake was significantly associated with reduced odds of MAFLD (highest vs. lowest quartile OR:0.48;95% CI:0.23-0.96). No significant relationship was found between total protein intake and MAFLD odds. CONCLUSIONS Our findings highlight the significance of dietary protein source in the odds of MAFLD among overweight and obese children and adolescents. Further studies are warranted to confirm these findings and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Ali Nikparast
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Sohouli
- Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Centre of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kimia Forouzan
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Amani Farani
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pooneh Dehghan
- Department of Imaging, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pejman Rohani
- Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Centre of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Golaleh Asghari
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Kido H, Mizukoshi E, Yanagi M, Shihui L, Seike T, Nakagawa H, Yamashima T, Shiraishi Y, Ozaki N, Harada K, Okada H, Goto H, Kimura K, Yamamoto Y, Yamashita T. Abnormalities of intracellular organelles in metabolic dysfunction-associated steatotic disease. J Gastroenterol 2025:10.1007/s00535-025-02257-5. [PMID: 40343540 DOI: 10.1007/s00535-025-02257-5] [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: 09/13/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The concept of metabolic dysfunction-associated steatotic disease (MASLD) is increasingly being recognized. The mechanisms contributing to hepatocellular injury include oxidative stress owing to mitochondrial dysfunction, endoplasmic reticulum (ER) stress owing to abnormal protein accumulation in the rough ER, and disruption of cellular homeostasis and metabolic regulation to autophagic dysfunction. However, the morphological abnormalities of these intracellular organelles remain unclear. METHODS Liver tissues from model mice of MASLD, patients with MASLD, and respective controls were subjected to histopathological examination using light microscopy, and intracellular organelles were analyzed via transmission electron microscopy (TEM). RESULTS In model mice of MASLD, the progression of MASLD pathology was associated with abnormalities in mitochondria, glycogen granules, and rough ER. Based on these findings, the electron microscopic observations of these intracellular organelles were classified, weighted, and evaluated in liver tissues of patients with MASLD. The electron microscopic findings were significantly relatively frequent in patients with MASLD and correlated with existing histopathological scoring. CONCLUSIONS Using TEM, we identified characteristic abnormalities in intracellular organelles specific to MASLD. These findings contribute to the understanding of the mechanisms underlying hepatocellular injury in MASLD.
Collapse
Affiliation(s)
- Hidenori Kido
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan.
| | - Masahiro Yanagi
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Li Shihui
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takuya Seike
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hidetoshi Nakagawa
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Tetsumori Yamashima
- Department of Psychiatry and Behavioral Science, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yoshitake Shiraishi
- Department of Functional Anatomy, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kenichi Harada
- Department of Human Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hikari Okada
- Information-Based Medicine Development, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hisanori Goto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kumi Kimura
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
22
|
Pérez-Díaz AJ, Ros-Madrid I, Martínez-Sánchez MA, Rico-Chazarra S, Oliva-Bolarín A, Balaguer-Román A, Fernández-Ruiz VE, Martínez CM, Yuste JE, Ferrer-Gómez M, Llamoza-Torres CJ, Frutos MD, Núñez-Sánchez MÁ, Ramos-Molina B. Alterations in hepatic amino acid metabolism related to MASLD in individuals with obesity. J Physiol Biochem 2025:10.1007/s13105-025-01086-7. [PMID: 40335876 DOI: 10.1007/s13105-025-01086-7] [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: 02/04/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Deregulation of amino acid (AA) metabolism has been reported in several pathological conditions, including metabolic diseases (e.g., obesity and diabetes), cardiovascular diseases, and cancer. However, the role of alterations in AA levels in chronic liver disorders such as metabolic dysfunction-associated steatotic liver disease (MASLD) remains largely unexplored. In this study we aimed to evaluate the hepatic AA composition in patients with different stages of MASLD, and their relationship with MASLD-related risk factors. A case-control study was conducted in 40 patients with obesity undergoing bariatric surgery at Virgen de la Arrixaca University Hospital (Murcia, Spain), where MASLD diagnosis was confirmed by histological analysis of liver biopsies, and hepatic AA levels were measured using ultra-performance liquid chromatography high-resolution time-of-flight mass spectrometry. Our results revealed that the hepatic AA profile was significantly altered in patients with MASLD. More specifically, comparison between MASLD patients revealed a significant increase in hepatic levels of arginine, glycine and cystine in MASH samples compared to steatotic livers. In addition, hepatic concentrations of arginine, lysine and cystine positively correlated with histopathological diagnosis and other MASLD-related parameters, including transaminases and CK-18 levels. These findings suggest that alterations in certain hepatic AA levels such as arginine, lysine, glycine and cystine in MASLD patients could have translational relevance in understanding the onset of this disease.
Collapse
Affiliation(s)
| | - Inmaculada Ros-Madrid
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
- Department of Endocrinology and Nutrition, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - María A Martínez-Sánchez
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Sara Rico-Chazarra
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alba Oliva-Bolarín
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Andrés Balaguer-Román
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Virginia E Fernández-Ruiz
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
- Department of Endocrinology and Nutrition, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Carlos M Martínez
- Experimental Pathology Platform, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - José E Yuste
- Metabolomics Platform of CEBAS-CSIC, Campus Universitario de Espinardo, Murcia, Spain
| | - Mercedes Ferrer-Gómez
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
- Department of Endocrinology and Nutrition, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Camilo J Llamoza-Torres
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
- Division of Liver Diseases, Department of Gastroenterology and Hepatology, University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - María D Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - María Á Núñez-Sánchez
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain.
- Biomedical Research Institute of Murcia (IMIB) Edificio LAIB, Carretera Buenavista s/n, Murcia, Spain.
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain.
- Biomedical Research Institute of Murcia (IMIB) Edificio LAIB, Carretera Buenavista s/n, Murcia, Spain.
| |
Collapse
|
23
|
Zhu X, Yin H, Han J, Zhang X, Han Q, Sun W, Liu Y, Tao W, Liu X, Wang G, Li L. Association Between Uric Acid to HDL-C Ratio and Metabolic Dysfunction-Associated Steatotic Liver Disease in Type 2 Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2025; 18:1459-1466. [PMID: 40356713 PMCID: PMC12067459 DOI: 10.2147/dmso.s520688] [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: 02/04/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Background Patients with type 2 diabetes mellitus (T2DM) exhibit an elevated risk of developing metabolic dysfunction-associated steatotic liver disease (MASLD). The uric acid to high-density lipoprotein cholesterol ratio (UHR) has emerged as a novel metabolic biomarker implicated in MASLD pathogenesis. This study aimed to evaluate the association between UHR and MASLD in a T2DM population. Methods In this cross-sectional study, we analyzed clinical data from 1081 T2DM patients (464 without MASLD, 617 with MASLD). Physiological and biochemical parameters were collected and analyzed. UHR was calculated as [uric acid (mg/dL)/HDL-C (mg/dL)] × 100%. Univariate and multivariate logistic regression analyses were performed to examine the association between UHR and MASLD. Results T2DM patients with MASLD had significantly higher UHR levels than those without MASLD (12.12[9.06-16.83] vs 10.36[7.65-14.08], p<0.001). UHR showed a strong positive correlation with TG/HDL (r =0.673, p < 0.001), moderate correlations with TG (r = 0.516, p < 0.001) and TC/HDL (r =0.548, p < 0.001), weak but significant associations with BMI (r = 0.330), WHR (r = 0.289), HOMA-IR (r = 0.121), ALT (r = 0.123), and GGT (r = 0.267) (all p < 0.05). Multivariate logistic regression showed that elevated UHR levels were significantly associated with increased MASLD risk (adjusted OR = 1.057, 95% CI: 1.016-1.100, p = 0.006), after adjusting for age, diabetes duration, BMI, blood pressure, and biochemical confounders. Conclusion Elevated UHR is independently associated with MASLD in T2DM patients, suggesting its clinical relevance in MASLD screening among this high-risk population.
Collapse
Affiliation(s)
- Xiangyun Zhu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Han Yin
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Jing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xiaoyan Zhang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Qing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Yijun Liu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Wenxuan Tao
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xinliang Liu
- Department of Endocrinology, Lianyungang Affiliated Hospital of Nanjing Medical University, Lianyungang, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Xuyi People’s Hospital of Clinical College of Yangzhou University, Huai’an, Jiangsu, 211700, People’s Republic of China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| |
Collapse
|
24
|
Duan S, Chen M, Chen J, Shao Y, Jin X, Wang C, Feng P, Teng X, Yu Z. Application of insulin resistance score in type 2 diabetes mellitus complicated with fatty liver and liver fibrosis. Eur J Gastroenterol Hepatol 2025:00042737-990000000-00531. [PMID: 40359287 DOI: 10.1097/meg.0000000000002998] [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] [Indexed: 05/15/2025]
Abstract
BACKGROUND Insulin resistance plays a pivotal role in the progression of type 2 diabetes mellitus (T2DM). An in-depth investigation into the role of insulin resistance scores in evaluating T2DM combined with metabolic-associated fatty liver disease (MAFLD) and liver fibrosis holds significant importance for clinical decisions and personalized treatment. METHODS The study screened patients with diabetes from Taizhou Central Hospital from June 2020 to May 2024. In conjunction with the National Health and Nutrition Examination Survey (NHANES) database, various statistical methods such as logistic regression analysis, restricted cubic spline, and receiver operating characteristic curves were employed to complete data analysis. RESULTS This study encompassed 3776 patients with T2DM, including 1074 diagnosed with MAFLD. Insulin resistance scores in the MAFLD group were significantly elevated. Compared with nonfibrotic patients, those with T2DM and liver fibrosis exhibited notably higher Chinese visceral adiposity index (CVAI) scores and notably lower triglyceride-glucose index and visceral adiposity index scores; the incidence of hypertension, coronary heart disease, stroke, and peripheral arterial disease were significantly elevated. Among other insulin resistance scores, the CVAI score demonstrated the highest value for correlating with the MAFLD and liver fibrosis in patients with T2DM. The NHANES database, encompassing data from 6763 individuals, validated the aforementioned findings, further affirming that the CVAI score exhibited optimal consistency with the risk of T2DM with MAFLD and liver fibrosis. CONCLUSION The insulin resistance scores were significantly elevated in T2DM combined with MAFLD. The CVAI score demonstrated the best predictive effect on MAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Shaojie Duan
- Department of Gastroenterology
- Department of Geriatrics
| | | | - Jie Chen
- Endoscopy Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | | | | | | | | | | | - Zhenjun Yu
- Department of Gastroenterology
- Department of Geriatrics
| |
Collapse
|
25
|
Ren Z, Fan H, Xue Y, Yang X, Liu X, Luo J, Zhao J, Wang L, Zhang Y, Liang B. Mediational role of metabolic syndrome between physical activity, sedentary behavior and non-alcoholic fatty liver disease: a cross-sectional study. BMC Public Health 2025; 25:1661. [PMID: 40329313 PMCID: PMC12054284 DOI: 10.1186/s12889-025-22925-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Physical activity (PA), sedentary behavior (SB), metabolic syndrome (MetS), and non-alcoholic fatty liver disease (NAFLD) have been linked in previous studies. Nevertheless, it is unclear whether MetS has a mediating influence on the relationships among physical activity, sedentary behavior, and non-alcoholic fatty liver disease. This study aims to assess the connections between physical activity, sedentary behavior, and non-alcoholic fatty liver disease and to explore the extent to which metabolic syndrome acts as a mediator in this association. METHODS A total of 3351 adults from the National Health and Nutrition Examination Survey (NHANES) from 2017 to 2018 were included in our study. Physical activity and sedentary behavior were categorized as work activity (WA), recreational activity (RA), walking/bicycling (for commuting) and sedentary behavior to investigate the association with metabolic syndrome and non-alcoholic fatty liver disease. Besides, mediation analysis was utilized to determine the extent to which metabolic syndrome mediates the relationships among inadequate physical activity, prolonged sedentary behavior, and non-alcoholic fatty liver disease. RESULTS Regression analysis revealed that a reduced risk of developing NAFLD was associated with sufficient recreational activity (OR = 0.61, 95% CI: 0.44-0.83, P = 0.004), while an increased risk of MetS was observed in sedentary behavior group (OR = 1.28, 95% CI: 1.00-1.64, P < 0.05). In addition, strong associations were detected between MetS and NAFLD. Mediation analysis indicated that metabolic syndrome accounts for 17.9% of the influence that recreational activity has on the risk of NAFLD. Subgroup analysis indicated sex differences in these associations. Specifically, recreational activity may not significantly influence the risk of developing NAFLD in females, and the mediating role of MetS was no longer significant in both sex-specific subgroups. CONCLUSION In the general adult population, metabolic syndrome may account for nearly 18% of the association between insufficient recreational activity and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Zhaoyu Ren
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Hongxuan Fan
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yaya Xue
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Xinyu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Xuchang Liu
- The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Luo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Jianqi Zhao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Leigang Wang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Yao Zhang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Wuyi Road, Taiyuan, Shanxi, 030000, China.
| |
Collapse
|
26
|
Yi H, Zhang Y, Zhou Z, Sun W, Wang Y, Tao W, Yu H, Yao L, Li J, Li L. Diagnostic performance of noninvasive tests for identifying advanced fibrosis in metabolic dysfunction-associated fatty liver disease with mixed etiologies. Endocr Pract 2025:S1530-891X(25)00140-5. [PMID: 40334939 DOI: 10.1016/j.eprac.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/10/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVES To assess the performance of fibrosis-4 index (FIB-4), non-alcoholic fatty liver disease fibrosis score (NFS) and aspartate aminotransferase to platelet ratio index (APRI) for advanced fibrosis in metabolic dysfunction-associated fatty liver disease (MAFLD) subgroups categorized by concomitant liver conditions. METHODS We conducted a multicentered study comprising inpatients with type 2 diabetes mellitus (T2DM) and MAFLD. Participants were categorized into two groups: MAFLD with pure metabolic etiologies (MAFLD-P) and MAFLD with mixed etiologies (MAFLD-M). Diagnostic performance of FIB-4, NFS and APRI was assessed by area under receiver operating characteristic curve (AUC), sensitivity and specificity. RESULTS This study comprised a total of 1475 participants, with a mean (SD) age of 58.4 (13) years and 835 (56.6%) males. FIB-4 and APRI had higher AUCs for advanced fibrosis in the MAFLD-M group than in the MAFLD-P group (MAFLD-M vs MAFLD-P: FIB-4 0.680 vs 0.591, p = 0.0442; APRI 0.723 vs 0.631, p = 0.0363). No significant difference was observed in the AUC of NFS between the two subgroups (MAFLD-M 0.572 vs MAFLD-P 0.617; p = 0.3237). Besides, the sensitivity of FIB-4 (69.6% vs 54.0%; p = 0.019) and APRI (43.5% vs 26.1%; p = 0.005) was higher in the MAFLD-M group. However, no significant difference in sensitivity of NFS and specificity of FIB-4, NFS and APRI was observed between subgroups. CONCLUSIONS In this diagnostic study of the T2DM population, FIB-4 and APRI showed better performance for identifying advanced fibrosis in MAFLD with mixed etiologies.
Collapse
Affiliation(s)
- He Yi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Yan Zhang
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Ziwei Zhou
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Yifan Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Wenxuan Tao
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Hekai Yu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China
| | - Liqin Yao
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, 214200, China.
| | - Jia Li
- Department of Ultrasound, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
27
|
Huang C, Gao Z, Huang Z, Xu J. Nonlinear association between body roundness index and metabolic dysfunction associated steatotic liver disease in nondiabetic Japanese adults. Sci Rep 2025; 15:15442. [PMID: 40316694 PMCID: PMC12048529 DOI: 10.1038/s41598-025-99540-5] [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: 12/14/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
The global rise in obesity and diabetes has been paralleled by a rising incidence of metabolic dysfunction-associated steatotic liver disease (MASLD). Although previous studies have explored the association between body roundness index (BRI) and MASLD, the specific relationship in non-diabetic Japanese adults requires further investigation. This study analyzed data from 15,299 participants enrolled in the NAGALA cohort (2004-2015) to explore the association between BRI and MASLD through multivariable logistic regression, stratified analysis, and restricted cubic spline modeling. The prevalence of MASLD was 14.46%, with 13.73% occurring in non-obese individuals (BMI < 30). After adjusting for all confounding factors, BRI demonstrated a significant association with MASLD, yielding an adjusted odds ratio of 1.72 (95% CI 1.48-1.99). The restricted cubic spline model revealed a nonlinear relationship, with an inflection point at 3.06. Stratified analyses revealed stronger associations in individuals with lower BMI (≤ 24 kg/m2).
Collapse
Affiliation(s)
- Cheng Huang
- Department of Colorectal Surgery, First People's Hospital of Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Zhichao Gao
- Department of Neurosurgery, First People's Hospital of Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Zhenxia Huang
- Department of Infectious Diseases, First People's Hospital of Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Junfeng Xu
- Department of Colorectal Surgery, First People's Hospital of Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| |
Collapse
|
28
|
Ye D, Wang J, Shi J, Ma Y, Li Y, Li Q, Hu X, Chen J, Bao Z. Prevalence of MAFLD in the U.S. based on NHANES 2009-2018: differences in demographic characteristics, physical indices and lifestyle conditions. BMC Gastroenterol 2025; 25:329. [PMID: 40316899 PMCID: PMC12046859 DOI: 10.1186/s12876-025-03956-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND The incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) is high among U.S. adults, but studies on its occurrence in different ethnic and age groups are limited. The aim of the present study was to assess MAFLD occurrence among the U.S. adults by considering demographic characteristics, physical indices, and lifestyle conditions. METHODS This study utilized the National Health and Nutrition Examination Survey (NHANES) data 2009-2018 from 23,546 participants aged ≥ 20 years. Variables such as age, sex, race, body mass index (BMI), waist circumference (WC), blood pressure, sedentary behavior, sleep, and depression were analyzed. RESULTS Among 9933 participants, 3562 had MAFLD (34.1%), with notably higher percentages of Mexican-Americans (54.1%) and lower percentages of blacks (20.5%). The incidence of MAFLD was significantly greater (P < 0.001) in males (39%) than in females (29.2%), which was particularly evident within the 36-40 years age group. The MAFLD incidence exhibited an age-dependent pattern, initially increasing and subsequently declining (except for whites). Compared to white MAFLD patients, black MAFLD patients exhibited greater BMI, WC, systolic blood pressure (SBP), and diastolic blood pressure (DBP) values, whereas values for these measures were lower among Mexican-American patients. Logistic regression analysis adjusting for age and sex revealed that depression was more common among MAFLD patients (P < 0.001), except for severe depression (P > 0.05). Notably, the MAFLD incidence was not significantly associated with sedentary behavior or sleep duration. CONCLUSIONS The MAFLD incidence varies across different racial, age, and sex groups, and targeted interventions are essential for reducing the burden of MAFLD. However, further research is necessary to explore the correlations among MAFLD incidence, sleep patterns, and an inactive lifestyle.
Collapse
Affiliation(s)
- Dan Ye
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jiaofeng Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Shanghai Institute of Geriatrics and Gerontology, Shanghai, China
| | - Jiaheng Shi
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yiming Ma
- Department of General Practice, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yanglei Li
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qingshang Li
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiaona Hu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Jie Chen
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Shanghai Institute of Geriatrics and Gerontology, Shanghai, China
| | - Zhijun Bao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
- Shanghai Institute of Geriatrics and Gerontology, Shanghai, China.
| |
Collapse
|
29
|
Abbaszadeh M, Hosseinpanah F, Tohidi M, Karimpour Reyhan S, Mahdavi M, Valizadeh M. Sex-Specific Impact of Metabolic Dysfunction-Associated Fatty Liver Disease on Incident Cardiovascular Diseases and Mortality. Endocrinol Diabetes Metab 2025; 8:e70035. [PMID: 40140729 PMCID: PMC11946537 DOI: 10.1002/edm2.70035] [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: 09/07/2024] [Revised: 12/30/2024] [Accepted: 02/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND AND AIMS Considering recent revisions in the nomenclature for fatty liver disease, alongside limited data on sex-specific differences in its cardiovascular/mortality outcomes, this study aims to investigate the prevalence and impact of metabolic-associated fatty liver disease (MAFLD) on cardiovascular disease (CVD) and mortality in men and women over a 12-year follow-up period. METHODS In this large population-based cohort study, 7101 individuals aged ≥ 30 were enrolled. The prevalence of MAFLD was investigated in both genders. After excluding individuals with a history of previous CVD, 6331 participants were followed up for CVD and mortality over 12 years. Steatosis was defined as fatty liver index (FLI) ≥ 60. Multivariate-adjusted hazard ratios (HRs) were calculated for CVD and mortality. RESULTS The prevalence of MAFLD was 43.2%, higher in men (46.5%) than women (40.6%). Men with MAFLD (47.7 ± 12.1) were younger than women (52.2 ± 11.1). In the 12-year follow-up of 6331 individuals, multivariable-adjusted CVD HRs for MAFLD were 1.36 (1.10-1.67) in men and 1.48 (1.16-1.88) in women. Adjusted mortality HRs were 1.17 (0.86-1.59) and 1.38 (1.00-1.91) in men and women, respectively. Among patients with MAFLD, a subgroup with diabetes faced the highest hazard for CVD and mortality. CONCLUSION This study found that MAFLD is more common in men at a younger age. Despite the higher prevalence in men, women with MAFLD face a greater risk of cardiovascular events and mortality. Findings highlight the importance of gender-specific considerations in primary prevention programmes for MAFLD-related cardiovascular disease and mortality.
Collapse
Affiliation(s)
- Mahsa Abbaszadeh
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Farhad Hosseinpanah
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical SciencesTehranIran
| | - Sahar Karimpour Reyhan
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Maryam Mahdavi
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Majid Valizadeh
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| |
Collapse
|
30
|
Chu Y, Yang S, Chen X. Fibroblast growth factor receptor signaling in metabolic dysfunction-associated fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther 2025; 269:108844. [PMID: 40113178 DOI: 10.1016/j.pharmthera.2025.108844] [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: 01/08/2025] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as a significant hepatic manifestation of metabolic syndrome, with its prevalence increasing globally alongside the epidemics of obesity and diabetes. MAFLD represents a continuum of liver damage, spanning from uncomplicated steatosis to metabolic dysfunction-associated steatohepatitis (MASH). This condition can advance to more severe outcomes, including fibrosis and cirrhosis. Fibroblast growth factor receptors (FGFRs) are a family of four receptor tyrosine kinases (FGFR1-4) that interact with both paracrine and endocrine fibroblast growth factors (FGFs). This interaction activates the phosphorylation of tyrosine kinase residues, thereby triggering downstream signaling pathways, including RAS-MAPK, JAK-STAT, PI3K-AKT, and PLCγ. In the context of MAFLD, paracrine FGF-FGFR signaling is predominantly biased toward the development of liver fibrosis and carcinogenesis. In contrast, endocrine FGF-FGFR signaling is primarily biased toward regulating the metabolism of bile acids, carbohydrates, lipids, and phosphate, as well as maintaining the overall balance of energy metabolism in the body. The interplay between these biased signaling pathways significantly influences the progression of MAFLD. This review explores the critical functions of FGFR signaling in MAFLD from three perspectives: first, it examines the primary roles of FGFRs relative to their structure; second, it summarizes FGFR signaling in hepatic lipid metabolism, elucidating mechanisms underlying the occurrence and progression of MAFLD; finally, it highlights recent advancements in drug development aimed at targeting FGFR signaling for the treatment of MAFLD and its associated diseases.
Collapse
Affiliation(s)
- Yi Chu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Su Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
31
|
Younossi ZM, Razavi H, Sherman M, Allen AM, Anstee QM, Cusi K, Friedman SL, Lawitz E, Lazarus JV, Schuppan D, Romero-Gómez M, Schattenberg JM, Vos MB, Wong VWS, Ratziu V, Hompesch M, Sanyal AJ, Loomba R. Addressing the High and Rising Global Burden of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Metabolic Dysfunction-Associated Steatohepatitis (MASH): From the Growing Prevalence to Payors' Perspective. Aliment Pharmacol Ther 2025; 61:1467-1478. [PMID: 39967239 DOI: 10.1111/apt.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/10/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND The continuum of metabolic syndrome encompasses a spectrum of dysfunctions impacting obesity-linked insulin resistance, glucose homeostasis, lipid metabolism and pro-inflammatory immune responses. The global prevalence of metabolic diseases, including diabetes, chronic liver disease, cardiometabolic disease and kidney disease, has surged in recent decades, contributing significantly to population mortality. Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease, is a leading cause of liver disease worldwide. MASLD poses a significant global health challenge with its rising prevalence, placing a substantial burden on healthcare systems, impacts patient well-being and incurs significant economic costs. Addressing MASLD requires a comprehensive understanding of its interconnected factors, including its prevalence, healthcare burden and economic implications. Lack of awareness, imprecise non-invasive diagnostic methods and ineffective preventive interventions are core components of the MASLD-related problem. AIM The aim of this article was to summarise the global burden of MASLD from the payer's perspective. METHODS We carried out a review of the global comprehensive burden of MASLD. These topics led to discussions and insights by an expert panel during the 7th Metabolic Continuum Roundtable meeting, which took place in November 2023. This meeting focused on the burden, patient-reported outcomes and health economics, from payor and societal perspectives, and aimed to identify opportunities for improving patient care, optimise resource allocation and mitigate the overall impact on individuals and society related to MASLD. During the roundtable, an emphasis emerged on the need for greater awareness and strategic deployment of diagnostic, therapeutic and preventative measures to address MASLD effectively. CONCLUSION The global burden of MASLD is high and growing. Prioritising the prevention of metabolic dysregulation and timely therapeutic interventions can yield a holistic strategy to combat MASLD, its progression and potentially lower disease costs. TRIAL REGISTRATION NCT06309992.
Collapse
Affiliation(s)
- Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- The Global NASH Council, Washington, DC, USA
| | - Homie Razavi
- Center for Disease Analysis Foundation, Lafayette, Colorado, USA
| | - Michael Sherman
- RA Capital Management, L.P., Boston, Massachusetts, USA
- Department of Population Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Quentin M Anstee
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Center, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes & Metabolism, University of Florida, Gainesville, Florida, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eric Lawitz
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Jeffrey V Lazarus
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York, New York, USA
| | - Detlef Schuppan
- Mainz University, Mainz, Germany
- Germany & Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Manuel Romero-Gómez
- Department of Medicine, UCM Digestive Diseases, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville (HUVR/CSIC/US), CIBEREHD, ISCIII, University of Seville, Seville, Spain
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Center, Homburg, Germany
| | - Miriam B Vos
- Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Vincent Wai-Sun Wong
- State Key Laboratory of Digestive Disease, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vlad Ratziu
- Sorbonne Université and Pitié-Salpêtrière Hospital Paris, Paris, France
| | | | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology at UC San Diego, MASLD Research Center California, La Jolla, California, USA
| |
Collapse
|
32
|
Luo X, Deng H, Li Q, Zhao M, Zhang Y, Guo J, Wen Y, Chen G, Li J. Bulk transcriptome and single-nucleus RNA sequencing analyses highlight the role of recombination activating 1 in non-alcoholic fatty liver disease. Int J Biol Macromol 2025; 307:141919. [PMID: 40074128 DOI: 10.1016/j.ijbiomac.2025.141919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic condition with an incompletely understood pathogenesis. In this study, five candidate genes-RAG1, CKAP2, CENPK, TYMS, and BUB1-were identified as being associated with NAFLD progression through integrative bioinformatics analyses. A predictive model incorporating these genes demonstrated strong robustness and diagnostic accuracy. Single-nucleus RNA sequencing analysis further revealed that RAG1 plays a potential role in hepatocytes of NAFLD patients. Functional experiments using RNA interference to suppress RAG1 expression in HepG2 cells treated with oleic and palmitic acids showed reduced total glyceride and cholesterol levels, mitigated lipid accumulation, and alterations in pathways related to lipid metabolism, inflammation, and fibrosis. Furthermore, adeno-associated virus-specific knockdown of RAG1 in hepatocytes attenuated hepatic steatosis in high-fat diet-fed mice. These findings suggest that investigating the molecular mechanisms of hub genes like RAG1 may advance our understanding of NAFLD pathogenesis and inform therapeutic development.
Collapse
Affiliation(s)
- Xiaohua Luo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Hongbo Deng
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Qiang Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Miao Zhao
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Science, Central South University, 410078 Changsha, China
| | - Yu Zhang
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junjie Guo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yifan Wen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Guangshun Chen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| | - Jiequn Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
33
|
Cho T, Fukunaga S, Ohzono D, Tanaka H, Minami S, Nakane T, Mukasa M, Yoshinaga S, Nouno R, Takedatsu H, Kawaguchi T. Metabolic dysfunction-associated steatotic liver disease is a risk factor for gallstones: A multicenter cohort study. Hepatol Res 2025; 55:663-674. [PMID: 40317549 DOI: 10.1111/hepr.14170] [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: 11/28/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 05/07/2025]
Abstract
AIM Gallstone formation is associated with metabolic dysfunction. Recently, new definitions of steatotic liver disease (SLD) have been proposed, including metabolic dysfunction-associated SLD (MASLD) and moderate alcohol intake (MetALD). We investigated the effects of MASLD/MetALD on gallstone formation. METHODS This multicenter observational cohort study enrolled 8766 consecutive health-check examinees who underwent abdominal ultrasonography between 2008 and 2021 (total observation period 39,105.9 person-years). All patients were classified into non-SLD, MASLD, or MetALD groups. The effect of MASLD on gallstone development was evaluated using multivariate Cox regression analysis. RESULTS Age, male sex, and MASLD were identified as independent risk factors for gallstone development. MASLD was associated with a significantly higher risk of developing gallstones than non-SLD (hazard ratio [HR] 1.7112; 95% confidence interval [CI] 1.4294-2.0486; p < 0.0001) and MetALD (HR 1.3516, 95% CI 1.0130-1.8033, p = 0.0406). However, the risk of MetALD did not significantly differ between the SLD and non-SLD groups. Hypertension was the only significant independent cardiometabolic risk factor for gallstone development in the MASLD group (HR 1.4350, 95% CI 1.0545-1.9528; p = 0.0216). Random forest analysis and directed acyclic graphs identified hypertension as the most important direct factor affecting gallstone development in patients with MASLD. CONCLUSIONS MASLD was an independent risk factor for gallstone development, whereas MetALD presented a similar risk as non-SLD. Moderate alcohol consumption may reduce the risk of gallstone formation in patients with MASLD. Hypertension may be the most significant cardiometabolic risk factor for gallstone development in patients with MASLD.
Collapse
Affiliation(s)
- Tomonori Cho
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shuhei Fukunaga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Daiki Ohzono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hiroshi Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shinpei Minami
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tomoyuki Nakane
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Michita Mukasa
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shinobu Yoshinaga
- Medical Examination Section, Medical Examination Part Facilities, Public Utility Foundation Saga Prefectural Health Promotion Foundation, Saga, Japan
| | - Ryuichi Nouno
- Department of Gastroenterology, Kumamoto Central Hospital, Kikuchi, Kumamoto, Japan
| | - Hidetoshi Takedatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
34
|
Qi W, Cao X, Chen Y, Chen H, Zhang N, Liu R, Wang W, Liu Q, Zheng S, Li S, Li X, Zao X, Ye Y. JiGuCao capsule formula alleviates metabolic fatty liver disease by regulating the gut-liver axis and lipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156559. [PMID: 40064115 DOI: 10.1016/j.phymed.2025.156559] [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: 10/17/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Metabolic-associated fatty liver disease (MAFLD) is a prevalent chronic liver condition globally, characterized by suboptimal treatment outcomes. Traditional therapies often fail to address the multifaceted pathogenesis of MAFLD, which involves lipid metabolism, inflammation, and gut-liver axis dysregulation. JiGuCao Capsule formula (JCF), a patented Chinese medicine, has demonstrated clinical efficacy in liver disease treatment, indicating its potential as a new therapeutic option for MAFLD. PURPOSE This study aimed to investigate the therapeutic effects and underlying mechanisms of JCF in treating MAFLD, particularly focusing on its impact on liver pathology, intestinal health, and gut microbiota composition. METHODS A MAFLD mouse model was developed by administering a high-fat diet and 5% fructose water for 16 weeks. At week 8, mice exhibited significant steatosis, inflammation, and insulin resistance. Fifty mice were allocated into two groups: the normal diet (ND) group with 19 mice and the high-fat feed diet (HFD) group with 31 mice. Seven mice from each group were sacrificed at week 8 for serological and histopathological assessments. The remaining mice were allocated into ND (n = 6), HFD (n = 6), HFD + JCFL (human equivalent dose,780 mg/kg, n = 6), HFD + JCFH (threefold the human equivalent dose, 2340 mg/kg, n = 6), HFD + Polyene Phosphatidylcholine (PPC) (human equivalent dose,177.84 mg/kg, n = 6) and ND+ JCF (human equivalent dose,780 mg/kg, n = 6) groups. Daily gavage started at week 9. At week 16, after fasting, body weight and liver condition were recorded, and mice were euthanized with pentobarbital sodium. Mouse tissues and feces were collected for histopathological, molecular biological, and multi-omics analyses. RESULTS JCF effectively slowed MAFLD progression in mice by decreasing hepatic lipid accumulation and inflammation. Treatment with JCF significantly reduced hepatic triglycerides and inflammatory markers, including TNF-α and IL-6. JCF enhanced lipid metabolism, repaired the intestinal barrier, and lowered inflammatory cytokines in the intestines, as indicated by reduced serum LPS and restored tight junction proteins expression, such as claudin-1 and occludin. Fecal microbiota analysis indicated that JCF treatment elevated Lactobacillus levels and reduced Colidextribacter levels, correlating with enhanced metabolic profiles. The primary bioactive compounds identified in JCF responsible for these therapeutic effects were betulinic acid, cholic acid, deoxycholic acid, oleanolic acid, and pectolinarigenin. Transcriptomic analysis showed that JCF regulated key pathways involved in lipid metabolism, including the pparγ-cd36 axis and modulation of ox-LDL levels. The results indicate that JCF effectively mitigates MAFLD by influencing the gut-liver axis and lipid metabolism. CONCLUSION JCF alleviates MAFLD by modulating the gut-liver axis and lipid metabolism. Its effects involve improving gut barrier function, regulating microbiota, and targeting the pparγ-cd36 axis. Active compounds like betulinic acid support its therapeutic potential. JCF shows promise as a novel treatment for MAFLD, with further clinical studies needed.
Collapse
Affiliation(s)
- Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xu Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Yue Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Hening Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Ningyi Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Ruijia Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Wei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Qiyao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Size Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China.
| | - Yong'an Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China.
| |
Collapse
|
35
|
Lei K, Chen Y, Wu J, Lin Y, Bai Y, Cao H, Che Q, Guo J, Su Z. Mechanism of liver x receptor alpha in intestine, liver and adipose tissues in metabolic associated fatty liver disease. Int J Biol Macromol 2025; 307:142275. [PMID: 40112983 DOI: 10.1016/j.ijbiomac.2025.142275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Metabolism associated fatty liver disease (MAFLD) has emerged as a growing global health challenge with limited effective treatments. Research on nuclear receptors offers promising new therapeutic avenues for MAFLD. The liver X receptor (LXR) has gained attention for its roles in tumors and metabolic and inflammatory diseases; However, its effects on MAFLD treatment remain a subject of debate. This review explores the therapeutic role of LXRα in MAFLD, focusing on its functions in the intestine, hepatic and adipose tissue, and summarizes recent advancements in LXRα ligands over the past five years. In the intestine, LXRα activation enhances the efflux of non-biliary cholesterol and reduces inflammation in the gut-liver axis by regulating intestinal high-density lipoprotein synthesis and its interaction with lipopolysaccharide. In the liver, LXRα activation facilitates cholesterol transport, influences hepatic lipid synthesis, and exerts anti-inflammatory effects. In adipose tissue, LXRα helps delay MAFLD progression by managing lipid autophagy and insulin resistance. Ligands that modulate LXRα transcriptional activity show considerable promise for MAFLD treatment.
Collapse
Affiliation(s)
- Kaiwen Lei
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Chen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jianxing Wu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yiyu Lin
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
36
|
Byrne CD, Armandi A, Pellegrinelli V, Vidal-Puig A, Bugianesi E. Μetabolic dysfunction-associated steatotic liver disease: a condition of heterogeneous metabolic risk factors, mechanisms and comorbidities requiring holistic treatment. Nat Rev Gastroenterol Hepatol 2025; 22:314-328. [PMID: 39962331 DOI: 10.1038/s41575-025-01045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
Μetabolic dysfunction-associated steatotic liver disease (MASLD) comprises a heterogeneous condition in the presence of steatotic liver. There can be a hierarchy of metabolic risk factors contributing to the severity of metabolic dysfunction and, thereby, the associated risk of both liver and extrahepatic outcomes, but the precise ranking and combination of metabolic syndrome (MetS) traits that convey the highest risk of major adverse liver outcomes and extrahepatic disease complications remains uncertain. Insulin resistance, low-grade inflammation, atherogenic dyslipidaemia and hypertension are key to the mechanisms of liver and extrahepatic complications. The liver is pivotal in MetS progression as it regulates lipoprotein metabolism and secretes substances that affect insulin sensitivity and inflammation. MASLD affects the kidneys, heart and the vascular system, contributing to hypertension and oxidative stress. To address the global health burden of MASLD, intensified by obesity and type 2 diabetes mellitus epidemics, a holistic, multidisciplinary approach is essential. This approach should focus on both liver disease management and cardiometabolic risk factors. This Review examines the link between metabolic dysfunction and liver dysfunction and extrahepatic disease outcomes, the diverse mechanisms in MASLD due to metabolic dysfunction, and a comprehensive, personalized management model for patients with MASLD.
Collapse
Affiliation(s)
- Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vanessa Pellegrinelli
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
37
|
She C, Guo Z, Lin Y, Zhou S, Pang M, Liu J, Cao L, Su L, Sun Y, Fang C, Shao X, Nie S. Acute kidney injury is associated with liver-related events in patients with metabolic dysfunction-associated fatty liver disease. DIABETES & METABOLISM 2025; 51:101639. [PMID: 40101895 DOI: 10.1016/j.diabet.2025.101639] [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: 02/24/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Evidence regarding the role of acute kidney injury (AKI) in long-term development of metabolic dysfunction-associated fatty liver disease (MAFLD) is limited. We aimed to investigate the associations between AKI and liver-related events in patients with MAFLD. METHODS This study involved 50,499 Chinese adults with MAFLD from the China Renal Data System (CRDS) database. We identified AKI using patient-level serum creatinine data according to the Kidney Disease Improving Global Outcomes (KDIGO) criteria. The primary outcome was a composite of liver-related mortality and major adverse liver outcomes. The secondary outcome was an escalation of fibrosis-4 (FIB-4) risk scores. Cox proportional hazard models were performed to assess the association between AKI and the study outcomes. RESULTS The median age of the patients was 59.17 years, with 54.7% being male. There were 3,711 (7.3%) patients who experienced AKI during hospitalization. A total of 1,660 (3.3%) patients experienced composite liver outcome. Patients with AKI during hospitalization had higher risk of composite liver outcomes (adjusted hazard ratio (aHR) 1.83 [95% confidence interval 1.38;2.41] P < 0.001), especially among those with severe AKI (stage 2/3) (aHR 2.36 [1.57;3.54] P < 0.001). Regarding the secondary outcome, AKI was also associated with an increased risk of escalation of FIB-4 risk scores (aHR 1.28 [1.14;1.44] P < 0.001). These associations remained consistent across various subgroups and sensitivity analyses. CONCLUSIONS AKI was significantly associated with an increased risk of liver-related events among patients with MAFLD. These findings suggest that enhanced vigilance toward AKI may be justifiable in MAFLD patients.
Collapse
Affiliation(s)
- Caoxiang She
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Zhixin Guo
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yaduan Lin
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Shiyu Zhou
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Mingzhen Pang
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jiao Liu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Lisha Cao
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Licong Su
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yinfang Sun
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Chuyao Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China
| | - Xian Shao
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| | - Sheng Nie
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
38
|
Dai X, Feng S, Li T. Cold atmospheric plasma control metabolic syndromes via targeting fat mass and obesity-associated protein. Pharmacol Res 2025; 215:107720. [PMID: 40174815 DOI: 10.1016/j.phrs.2025.107720] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Both obesity and metabolic disorders are global medical problems. Driven by prolonged inflammation, obesity increases the risk of developing metabolic syndromes such as fatty liver, diabetes, cardiovascular diseases and cancers. The fat mass and obesity-associated protein (FTO) is an m6A demethylase, elevated activity of which is known to promote the pathogenesis of many metabolic disorders, leading to the establishment of various FTO inhibitors. By combing through intrinsic connections among obesity and the four primary metabolic problems, we attribute their shared pathological cause to prolonged inflammation. By reviewing the roles of FTO in promoting these disorders and the current status of existing FTO inhibitors in treating these syndromes, we underpinned the paramount potential of resolving these clinical issues by targeting FTO and the urgent need of establishing novel FTO inhibitors with maximized efficacy and minimized side effect. Cold atmospheric plasma (CAP) is the fourth state of matter with demonstrated efficacy in treating various diseases associated with chronic inflammation. By introducing the medical characteristics of CAP, we proposed it as a possible solution to unresolved issues of current FTO inhibitors given its anti-inflammation feature and demonstrated clinical safety. We also emphasized the need of intensive investigations in exploring the feasibility of using CAP in treating obesity and associated metabolic syndromes that might function through targeting FTO.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Shuo Feng
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Tian Li
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, 8 Changjiang Avenue, Tianjin 300100, China.
| |
Collapse
|
39
|
Zou Y, Dai Y, Li Z, Lin B, Chen H, Zhuang Z, Li W, Yang Q, Dai D. Modified triglyceride-glucose indices as novel predictors of metabolic dysfunction-associated fatty liver disease in US adolescents: a nationally representative study from NHANES 2017-2020. BMC Gastroenterol 2025; 25:325. [PMID: 40312305 PMCID: PMC12044992 DOI: 10.1186/s12876-025-03915-x] [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: 02/27/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) has become the most prevalent chronic liver condition in adolescents. The triglyceride-glucose (TyG) index, a surrogate marker of insulin resistance, has shown promise in adult MAFLD detection but requires pediatric-specific validation, particularly when combined with anthropometric measures. This study investigated the association between modified TyG indices and MAFLD, and evaluated their predictive value in adolescents. METHODS This cross-sectional study analyzed data from 532 adolescents (12-18 years) in the 2017-2020 National Health and Nutrition Examination Survey (NHANES) with complete records. MAFLD diagnosis was based on transient elastography plus metabolic criteria. The investigators employed multivariate linear regression and restricted cubic splines (RCS) to examine linear and nonlinear relationships between modified TyG indices and CAP values. Subgroup analyses were stratified by obesity status, and sensitivity analyses were performed on the NAFLD cohort (n = 527). Receiver operating characteristic (ROC) curve analysis, using Youden's index, evaluated the predictive performance of TyG indices for MAFLD identification. RESULTS Among 130 MAFLD adolescents (vs 402 controls), modified TyG indices demonstrated significantly stronger associations with CAP in fully adjusted models compared to the original TyG index. TyG-WC showed the highest diagnostic accuracy (AUC = 0.923, 95%CI:0.900-0.947), followed by TyG-BMI (AUC = 0.917) and TyG-WHtR (AUC = 0.915), while the original TyG index performed poorly (AUC = 0.673). Subgroup analyses revealed particularly strong associations in non-obese participants, and sensitivity analyses confirmed result robustness after excluding potential confounders. Optimal cutoff values provided clinically useful screening thresholds, with TyG-WC achieving 94% sensitivity at 665.94. CONCLUSION This study demonstrates that modified TyG indices incorporating anthropometric parameters (particularly TyG-WC) significantly outperform the original TyG index for MAFLD detection in adolescents, with superior diagnostic accuracy (AUC 0.915-0.923). The robust predictive performance maintained in sensitivity analyses and non-obese subgroups supports their clinical utility as simple, non-invasive screening tools for pediatric MAFLD risk stratification.
Collapse
Affiliation(s)
- Yigui Zou
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Yu Dai
- Children's Healthcare and Mental Health Center, Shenzhen Children's Hospital, Shenzhen , Guangdong, 518036, China
| | - Ziyuan Li
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Baixian Lin
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Hu Chen
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Zeling Zhuang
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Wenwen Li
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Qinghua Yang
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China
| | - Dongling Dai
- Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Disease, Endoscopy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
40
|
Spencer-Sandino M, Godoy F, Huidobro L, Alvares D, Cruz F, Marco C, Garrido M, Cabrera D, Arab JP, Arrese M, Barrera F, Ferreccio C. New steatotic liver disease criteria diagnostic performance in an agricultural population in Chile. Ann Hepatol 2025:101919. [PMID: 40318788 DOI: 10.1016/j.aohep.2025.101919] [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: 10/22/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION AND OBJECTIVES This study aims to assess the performance of Steatotic Liver Disease (SLD) criteria in identifying liver steatosis compared to the NAFLD and MAFLD definitions in an agricultural population in Chile. PATIENTS AND METHODS We performed a cross-sectional analysis on the MAUCO cohort, composed of 9,013 individuals aged 38 to 74. Health conditions, socio-demographics, anthropometrics, hepatic ultrasonography, blood pressure, and biological samples were obtained. Participants were classified as NAFLD, MAFLD, or any of the five SLD categories: Metabolic dysfunction-associated steatosis liver disease (MASLD), Metabolic and Alcohol-Associated Liver Disease (MetALD), Alcohol-Associated Liver Disease (ALD), Specific aetiologies, and Cryptogenic. The Framingham cardiovascular risk score and BARD liver fibrosis score were used to assess clinical relevance. RESULTS Liver steatosis was present in 4,082 participants (45%); SLD criteria captured an additional 176 individuals not classified under NAFLD and 103 not included under MAFLD definition. The main SLD subgroups were MASLD (95%), MetALD (1.9%) and ALD (1.3%). Individuals classified in the MetALD and ALD subgroups exhibited more severe liver steatosis and a higher cardiovascular risk. Notably, participants categorized under specific etiologies and cryptogenic subgroups were younger and had a higher risk for liver fibrosis. CONCLUSIONS The study reveals that SLD offers a more inclusive classification to identify high-risk individuals in the Chilean population, capturing cases that could be missed by NAFLD or MAFLD definitions by using the same resources.
Collapse
Affiliation(s)
- Maria Spencer-Sandino
- Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile; Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Franco Godoy
- Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile; Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Laura Huidobro
- Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Preclínicas - Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | | | - Francisco Cruz
- Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Marco
- Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile; Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Garrido
- Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile; Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile; Instituto de Salud Pública de Chile, Santiago, Chile
| | - Daniel Cabrera
- Centro de Investigación e Innovación en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile; Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| | - Juan Pablo Arab
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond 23298, VA, USA; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marco Arrese
- Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Barrera
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catterina Ferreccio
- Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile; Advance Center for Chronic Diseases, ACCDIS, Universidad De Chile and Pontificia Universidad Católica de Chile, Santiago, Chile; Instituto de Salud Pública de Chile, Santiago, Chile.
| |
Collapse
|
41
|
Xie F, Zheng W, Chen J, Yao C, Li C, Tang L, Li P, Tan S. Revisiting the causal impact of lipid traits on metabolic dysfunction-associated fatty liver disease: Insights from a multidimensional plasma lipid profile. J Diabetes Investig 2025; 16:917-928. [PMID: 40052234 PMCID: PMC12057391 DOI: 10.1111/jdi.14413] [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: 10/23/2024] [Revised: 12/21/2024] [Accepted: 01/09/2025] [Indexed: 05/08/2025] Open
Abstract
AIMS Recent advancements in plasma lipidomes genome-wide association studies data have enhanced our understanding of lipid categories, significantly improving risk assessments for metabolic dysfunction-associated fatty liver disease (MAFLD) beyond traditional lipid biomarkers. MATERIALS AND METHODS This study utilized Mendelian randomization (MR) to assess the causal relationships between 179 lipid species across 13 subclasses and MAFLD, primarily using the Wald ratio and IVW methods. Corrections were made using false discovery rate (FDR), supplemented by Bayesian colocalization analysis. RESULTS Elevated levels of genetically predicted phosphatidylcholine (16:0_16:1) [ORWald ratio = 2.638, 95% CI 1.557-4.469, P = 3.11 × 10-4], phosphatidylcholine (16:1_18:0) (ORWald ratio = 2.644, 95% CI 1.559-4.486, P = 3.11 × 10-4), triacylglycerol (46:2) (ORWald ratio = 2.515, 95% CI 1.524-4.153, P = 3.11 × 10-4), and triacylglycerol (48:2) (ORIVW = 1.863, 95% CI 1.300-2.669, P = 6.95 × 10-4) were significantly associated with increased MAFLD risk, with rs1260326 within the GCKR gene playing a crucial role. Colocalization analysis indicated that in significant evidences, the posterior probability for hypothesis 4 was over 80%, identifying rs780093 as a shared causal variant. Additionally, 16 suggestive evidences were identified. CONCLUSION The study confirmed the significant role of specific lipid molecules in influencing MAFLD risk, providing new scientific bases and potential therapeutic targets for future treatment strategies.
Collapse
Affiliation(s)
- Fang Xie
- Department of Integrated TCM and Western MedicineNanjing Hospital Affiliated to Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
- Department of Liver DiseaseJinling Hospital Affiliated to Medical College of Nanjing UniversityNanjingJiangsu ProvinceChina
| | - Wenkai Zheng
- Department of Liver DiseaseJinling Hospital Affiliated to Medical College of Nanjing UniversityNanjingJiangsu ProvinceChina
| | - Jing Chen
- Department of Integrated TCM and Western MedicineNanjing Hospital Affiliated to Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Chuanxia Yao
- Department of Liver DiseaseJinling Hospital Affiliated to Medical College of Nanjing UniversityNanjingJiangsu ProvinceChina
| | - Cong Li
- Department of Liver DiseaseJinling Hospital Affiliated to Medical College of Nanjing UniversityNanjingJiangsu ProvinceChina
| | - Li Tang
- Department of GastroenterologyNanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Ping Li
- Department of Liver DiseaseJinling Hospital Affiliated to Medical College of Nanjing UniversityNanjingJiangsu ProvinceChina
| | - Shanzhong Tan
- Department of Integrated TCM and Western MedicineNanjing Hospital Affiliated to Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| |
Collapse
|
42
|
Liu XR, Yin SC, Chen YT, Lee MH. Metabolic dysfunction-associated steatotic liver disease and its associated health risks. J Chin Med Assoc 2025; 88:343-351. [PMID: 40128159 DOI: 10.1097/jcma.0000000000001230] [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] [Indexed: 03/26/2025] Open
Abstract
This article synthesizes the current knowledge on the epidemiology of metabolic dysfunction-associated steatotic liver disease (MASLD), its associated risks, and its genetic determinants. The findings presented in this article can be used to develop clinical strategies to reduce MASLD's growing global burden. MASLD has become a major global health concern due to increasing rates of obesity, sedentary lifestyles, and metabolic disorders. MASLD is a leading cause of end-stage liver diseases, including cirrhosis and hepatocellular carcinoma (HCC), and MASLD also significantly increases the risk of cardiovascular disease (CVD), thereby exerting dual effects on liver and cardiovascular health. MASLD was once referred to as nonalcoholic fatty liver disease, and this change in nomenclature reflects a growing focus on its metabolic underpinnings, facilitating the more precise diagnosis and clinical management of this disease. Epidemiological studies have demonstrated that the prevalence of MASLD is increasing worldwide, although the prevalence varies across regions and populations. Noninvasive diagnostic tools such as ultrasound and fatty liver indices along with biomarkers such as alanine aminotransferase (ALT) are crucial for early detection and risk stratification. Genetic research has identified key gene variants, including PNPLA3 (rs738409) and TM6SF2 (rs58542926), that influence MASLD susceptibility and progression, and these findings have created opportunities for improving precision medicine with respect to treating MASLD. Research has revealed an association between MASLD and major adverse cardiovascular events and increased mortality, which highlights the importance of integrating cardiovascular risk management into treatment strategies for MASLD. Future research should focus on advancing noninvasive diagnostics, leveraging genetic insights to provide tailored care, and implementing population-specific interventions to address regional variations.
Collapse
Affiliation(s)
- Xia-Rong Liu
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Szu-Ching Yin
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Yi-Ting Chen
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Mei-Hsuan Lee
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
43
|
Abdurrachim D, Lek S, Ong CZL, Wong CK, Zhou Y, Wee A, Soon G, Kendall TJ, Idowu MO, Hendra C, Saigal A, Krishnan R, Chng E, Tai D, Ho G, Forest T, Raji A, Talukdar S, Chin CL, Baumgartner R, Engel SS, Ali AAB, Kleiner DE, Sanyal AJ. Utility of AI digital pathology as an aid for pathologists scoring fibrosis in MASH. J Hepatol 2025; 82:898-908. [PMID: 39612947 DOI: 10.1016/j.jhep.2024.11.032] [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: 07/22/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND & AIMS Intra and inter-pathologist variability poses a significant challenge in metabolic dysfunction-associated steatohepatitis (MASH) biopsy evaluation, leading to suboptimal selection of patients and confounded assessment of histological response in clinical trials. We evaluated the utility of an artificial intelligence (AI) digital pathology (DP) platform to help pathologists improve the reliability of fibrosis staging. METHODS A total of 120 digitized histology slides from two trials (NCT03517540, NCT03912532) were analyzed by four expert hepatopathologists, with and without AI assistance in a randomized, crossover design. We utilized an AI DP platform consisting of unstained second harmonic generation/two photon excitation fluorescence (SHG/TPEF) images and AI quantitative fibrosis (qFibrosis) values. RESULTS AI assistance significantly improved inter-pathologist kappa for fibrosis staging, particularly for early fibrosis (F0-F2), with reduced variance around the median reads. Intra-pathologist kappa was unchanged. AI assistance increased pathologist concordance for identifying clinical trial inclusion cases (F2-F3) from 45% to 71%, exclusion cases (F0/F1/F4) from 38% to 55%, and evaluation of fibrosis response to treatment from 49% to 61%. SHG/TPEF images, qFibrosis continuous values, and qFibrosis stage were considered useful by at least three out of four pathologists in 83%, 55%, and 38% of cases, respectively. In the context of a clinical trial, the increase in inter-pathologist concordance was modeled to result in a ∼25% reduction in the potential need for adjudication as well as a ∼45% increase in the study power for a kappa improvement from ∼0.4 to ∼0.7. CONCLUSIONS The use of AI DP enhances inter-rater reliability of fibrosis staging for MASH. This indicates that the SHG/TPEF-based AI DP tool is useful for assisting pathologists in assessing fibrosis, thereby enhancing clinical trial efficiency and reliability of fibrosis readouts in response to treatments. IMPACT AND IMPLICATIONS Implementing an AI DP platform as a tool for pathologists significantly improved inter-pathologist agreement on fibrosis staging, particularly for early-stage fibrosis (F0-F2), which is critical for clinical trial eligibility. The second harmonic generation imaging technology used in conjunction with AI quantitative scores provided enhanced visualization of fibrosis with an indication of severity along the disease continuum. This led to increased pathologist confidence in fibrosis staging and, therefore, increased pathologist concordance for the classification of clinical trial inclusion/exclusion and evaluation of treatment, compared to a standard scoring method based on traditional stains without AI assistance. Improved pathologist concordance with AI assistance could streamline clinical trial processes, reducing the need for adjudication and enhancing study power, potentially decreasing required sample sizes. Continued exploration of the utility of AI assistance across a broader range of pathologists and in prospective clinical trials will be essential for validating the effectiveness of AI assistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Aileen Wee
- Department of Pathology, National University Hospital, Singapore
| | - Gwyneth Soon
- Department of Pathology, National University Hospital, Singapore
| | - Timothy J Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, United Kingdom
| | - Michael O Idowu
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | - Ashmita Saigal
- Cardiometabolic Diseases, Merck & Co., Inc., South San Francisco, CA, USA
| | - Radha Krishnan
- Global Clinical Trial Organization, MSD (UK) Limited, London, UK
| | | | | | | | - Thomas Forest
- Non-clinical Drug Safety, Merck & Co., Inc., West Point, PA, USA
| | - Annaswamy Raji
- Global Clinical Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Saswata Talukdar
- Cardiometabolic Diseases, Merck & Co., Inc., South San Francisco, CA, USA
| | - Chih-Liang Chin
- Cardiometabolic Diseases, Merck & Co., Inc., South San Francisco, CA, USA
| | - Richard Baumgartner
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, USA
| | - Samuel S Engel
- Global Clinical Development, Merck & Co., Inc., Rahway, NJ, USA
| | | | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, NIH, USA
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School Of Medicine, Richmond, VA, USA
| |
Collapse
|
44
|
Cui Y, Qu Z, Li L, Hu W. Gender difference in the association between serum uric acid and metabolic dysfunction-associated steatotic liver disease in patients with newly diagnosed type 2 diabetes. BMC Gastroenterol 2025; 25:322. [PMID: 40307757 PMCID: PMC12042553 DOI: 10.1186/s12876-025-03917-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
PURPOSE To investigate the relationship between serum uric acid (SUA) levels and metabolic dysfunction-associated steatotic liver disease (MASLD) in newly diagnosed type 2 diabetic patients. METHODS We performed this retrospective research among 1087 inpatients with new-onset type 2 diabetes millitus (T2DM). Data were analyzed according to gender. Then, the populations were stratified according to their body mass index (BMI) levels in men and women, respectively. The physical and biochemical indicators were measured and recorded. The relationship between SUA and MASLD was estimated using logistic regression analysis, and the unadjusted and adjusted odds ratios (ORs) were calculated. RESULTS After adjusting for age, BMI, and other components of the metabolic syndrome, SUA was independently associated with MASLD only in men, but not in women. In addition, for men, the SUA levels were independently associated with MASLD in both non-overweight/obesity and overweight/obesity group. However, for women, the SUA levels were independently related to MASLD in non-overweight/obesity group. There was no association between SUA and MASLD in women with overweight/obesity. CONCLUSION In newly diagnosed type 2 diabetic patients, elevated SUA is an independent predictor for the risk of MASLD in males. In females, the relationship between SUA and MASLD may depend on BMI, with significance only in non-overweight/obese individuals.
Collapse
Affiliation(s)
- Yuliang Cui
- Department of Endocrinology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China
| | - Zhenzhen Qu
- Department of Endocrinology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China
| | - Lingling Li
- Department of Health Management, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China
| | - Wenmei Hu
- Department of Endocrinology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China.
| |
Collapse
|
45
|
Li X, Chen W, Jia Z, Xiao Y, Shi A, Ma X. Mitochondrial Dysfunction as a Pathogenesis and Therapeutic Strategy for Metabolic-Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:4256. [PMID: 40362504 DOI: 10.3390/ijms26094256] [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: 03/27/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) has emerged as a significant public health concern, attributed to its increasing prevalence and correlation with metabolic disorders, including obesity and type 2 diabetes. Recent research has highlighted that mitochondrial dysfunction can result in the accumulation of lipids in non-adipose tissues, as well as increased oxidative stress and inflammation. These factors are crucial in advancing the progression of MASLD. Despite advances in the understanding of MASLD pathophysiology, challenges remain in identifying effective therapeutic strategies targeting mitochondrial dysfunction. This review aims to consolidate current knowledge on how mitochondrial imbalance affects the development and progression of MASLD, while addressing existing research gaps and potential avenues for future research. This review was conducted after a systematic search of comprehensive academic databases such as PubMed, Embase, and Web of Science to gather information on mitochondrial dysfunction as well as mitochondrial-based treatments for MASLD.
Collapse
Affiliation(s)
- Xiangqiong Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Wenling Chen
- The First Clinical College of Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zhuangzhuang Jia
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Yahui Xiao
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Anhua Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Xuan Ma
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| |
Collapse
|
46
|
Ren T, Chen Q, Zhu C. The extrahepatic markers in postmenopausal women with metabolic dysfunction-associated steatotic liver disease: A systematic review. Clin Nutr ESPEN 2025; 68:22-31. [PMID: 40315986 DOI: 10.1016/j.clnesp.2025.04.026] [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: 01/26/2025] [Revised: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent, multifactorial systemic metabolic disorder, now recognized as the most common chronic liver disease globally. Female susceptibility to MASLD varies across menstrual states, influenced by genetic factors, age, menopausal status, and physical activity. Postmenopausal women, experiencing a significant reduction in estrogen, are particularly vulnerable to metabolic imbalances, increasing their risk of MASLD, disease progression, liver fibrosis, insulin resistance, and adverse cardiovascular events compared to premenopausal women and age-matched men. This review systematically synthesizes current research on extrahepatic abnormalities associated with MASLD in postmenopausal women. This review identifies key extrahepatic markers associated with MASLD in postmenopausal women, highlighting gaps in current research and proposing targeted screening and management strategies. (Graphical Abstract).
Collapse
Affiliation(s)
- Tingting Ren
- Department of Infectious Disease, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Qingling Chen
- Department of Infectious Disease, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Chuanlong Zhu
- Department of Infectious Disease, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China; Department of Infectious and Tropical Diseases, The Second Affiliated Hospital, NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China.
| |
Collapse
|
47
|
Yan X, Li J, Wang Q, Xu T, Wu D. Peripheral blood lymphocyte subsets of Chinese adults with liver steatosis. Sci Rep 2025; 15:14970. [PMID: 40301513 PMCID: PMC12041267 DOI: 10.1038/s41598-025-99510-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
To evaluate the effect of liver steatosis on systematic cellular immunity through the changes of peripheral blood lymphocyte subsets, we retrospectively reviewed subjects receiving lymphocyte subtyping during annual medical check-ups. Liver steatosis was detected by abdominal computerized tomography or ultrasound. Immunophenotyping of peripheral blood lymphocytes was analyzed by flow cytometry. Cell counts of lymphocyte subsets were calculated using a dual-platform method. The relationship between lymphocyte subsets and liver steatosis was analyzed using multivariate linear models. Using the database from January 2017 to December 2022, we included 5042 subjects, including 1441 participants with liver steatosis. After adjusting for age, gender, and metabolic dysfunctions, the presence of liver steatosis increased the absolute values of CD19+, CD16+56+, CD3+, CD4+, CD4+CD28+, CD4+CD45RA-, CD4+CD45RA+, CD4+CD45RA+62L+, CD8+, CD8+CD28+, CD8+DR+, and CD8+CD38+ cell. In males, T lymphocyte counts of all T subsets increased in liver steatosis. In females, the significant differences in subsets included increased CD4+CD28+, CD4+CD45RA-, CD8+, and CD8+DR+ cells. Significant decreases were revealed in functional and naïve T cells with aging. Metabolic factors such as hypertension and abnormal glucose metabolism increase CD4+, CD4+CD28+, and CD4+CD45RA subsets. Hyperlipidaemia appears not to affect T cell counts, whereas obesity has some effect on both CD4+ (β = 0.041, 95% CI 0.032-0.051) and CD8+ cells (β = 0.036, 95% CI 0.025-0.048). Liver steatosis potentially affects peripheral blood lymphocyte subsets, while age, gender, and metabolic dysfunctions are also associated with these immune alterations.
Collapse
Affiliation(s)
- Xiaxiao Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiang Wang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tengda Xu
- Department of Health Management, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
48
|
Wu D, Liu J, Guo Z, Wang L, Yao Z, Wu Q, Lu Y, Lv W. Natural bioactive compounds reprogram bile acid metabolism in MAFLD: Multi-target mechanisms and therapeutic implications. Int Immunopharmacol 2025; 157:114708. [PMID: 40306110 DOI: 10.1016/j.intimp.2025.114708] [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: 03/05/2025] [Revised: 04/20/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
Metabolic-associated fatty liver disease (MAFLD) has become an increasingly prevalent liver disorder worldwide, being closely associated with obesity, metabolic syndrome, and insulin resistance. Bile acids (BAs), beyond their traditional role in lipid digestion, play a pivotal part in regulating lipid and glucose metabolism as well as inflammatory responses. Recent investigations have recognized BAs as key factors in the onset and progression of MAFLD, mainly via their interactions with nuclear receptors such as the farnesoid X receptor (FXR) and the G protein-coupled bile acid receptor (TGR5). Additionally, active compounds derived from traditional Chinese medicine (TCM) have shown promising potential in the treatment of MAFLD. This study systematically reviews and analyzes the molecular mechanisms and recent progress in the application of TCM active ingredients for MAFLD treatment, with a focus on their regulation of BAs. These active ingredients, including saponins, flavonoids, polysaccharides, and sterols, exert therapeutic effects through diverse mechanisms, such as modulating BA synthesis and mediating receptor-signaling pathways, and are expected to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dongjie Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Liu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ziwei Guo
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Liang Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Qingjuan Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yanping Lu
- Department of Hepatology, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Shenzhen 518100, China.
| | - Wenliang Lv
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
49
|
Tan W, Deng J, Qi L, Tan Z. The role of hepatic sinusoidal microenvironment in NASH: pathogenesis, animal models, and therapeutic prospects. Front Pharmacol 2025; 16:1467950. [PMID: 40356963 PMCID: PMC12066276 DOI: 10.3389/fphar.2025.1467950] [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: 07/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
The incidence of nonalcoholic steatohepatitis (NASH) is increasing annually, posing a significant threat to human health. NASH is typified by hepatic steatosis, inflammation, and hepatocellular injury, frequently culminating in fibrosis and cirrhosis. Yet, the precise pathogenesis of NASH remains to be fully elucidated. The hepatic sinusoid, which serves as the fundamental structural and functional unit of the liver, is intricately composed of endothelial cells, Kupffer cells, and hepatic stellate cells. Consequently, the homeostasis of the hepatic sinusoidal microenvironment may exert a pivotal influence on the progression and prognosis of NASH. However, the limitations of current NASH animal models have significantly impeded advancements in understanding the disease's pathogenesis and the development of effective therapeutic interventions. In light of these challenges, this review endeavors to delve deeper into the critical role of hepatic sinusoidal microenvironment homeostasis in the pathogenesis of NASH, critically analyze the commonly employed animal models, and comprehensively summarize the most recent and promising developments in drug research and development. It is anticipated that these efforts will collectively expedite the advancement of the field of NASH research and therapeutic innovation.
Collapse
Affiliation(s)
- Wanying Tan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiangting Deng
- Sichuan Academy of Chinese Medicine Sciences, Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Chengdu, Sichuan, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Lingjun Qi
- Affiliated Sichuan Gem Flower Hospital of North Sichuan Medical College, Chengdu, Sichuan, China
| | - Zhenghuai Tan
- Sichuan Academy of Chinese Medicine Sciences, Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Yang H, Jung S, Choi EY. E3 ubiquitin ligase TRIM38 regulates macrophage polarization to reduce hepatic inflammation by interacting with HSPA5. Int Immunopharmacol 2025; 157:114662. [PMID: 40300357 DOI: 10.1016/j.intimp.2025.114662] [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: 01/02/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses pathologies from simple steatosis and steatohepatitis (MASH) to cirrhosis. Hepatic inflammation is a common cause of liver pathogenesis, with macrophage activation as a key indicator of both acute and chronic liver dysfunction. While M1 macrophages promote inflammation and M2 macrophages suppress it, their roles in MASLD are dynamic and shift according to disease stage and liver microenvironment. Tripartite motif (TRIM) family proteins, which possess E3 ubiquitin ligase activity, are involved in various cellular processes, including intracellular signaling, development, apoptosis, protein quality control, innate immunity, autophagy, and carcinogenesis. TRIM38 negatively regulates innate immunity and inflammation triggered by viruses, Toll-like receptor 3 and 4, and tumor necrosis factor α/interleukin-1β signaling; however, its role in liver pathogenesis remains unclear. This study investigates the role of macrophage TRIM38 in metabolic liver disease to identify key targets for controlling inflammation. TRIM38 overexpression suppressed lipopolysaccharide-induced macrophage activation and metabolic stress-induced hepatic lipid accumulation. Mechanistically, TRIM38 interacted with heat shock protein family A member 5 (HSPA5) and stabilized it via K63-dependent ubiquitination. This TRIM38-HSPA5 axis promoted the expression of M2 macrophage markers (arginase 1 and retinoic acid-related orphan receptor α), thereby ameliorating liver steatosis. Single-cell RNA sequencing revealed significant downregulation of TRIM38 expression in the liver macrophages of patients with MASLD and negative regulation of liver inflammation via modulation of macrophage polarization. Hence, macrophage TRIM38 suppresses metabolic liver disease progression via HSPA5-mediated M2 macrophage polarization and provides insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Heeyoung Yang
- Center for Predictive Model Research, Division of Advanced Predictive Research, Korea Institute of Toxicology, Daejeon, Republic of Korea.
| | - Soontag Jung
- Center for Regulatory Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Eun-Yong Choi
- Center for Predictive Model Research, Division of Advanced Predictive Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| |
Collapse
|