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
|
Hao X, Song H, Su X, Li J, Ye Y, Wang C, Xu X, Pang G, Liu W, Li Z, Luo T. Prophylactic effects of nutrition, dietary strategies, exercise, lifestyle and environment on nonalcoholic fatty liver disease. Ann Med 2025; 57:2464223. [PMID: 39943720 PMCID: PMC11827040 DOI: 10.1080/07853890.2025.2464223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease and its prevalence has risen sharply. However, whether nutrition, dietary strategies, exercise, lifestyle and environment have preventive value for NAFLD remains unclear. METHODS Through searching 4 databases (PubMed, Web of Science, Embase and the Cochrane Library) from inception to January 2025, we selected studies about nutrition, dietary strategies, exercise, lifestyle and environment in the prevention of NAFLD and conducted a narrative review on this topic. RESULTS Reasonable nutrient intake encompassing macronutrients and micronutrients have an independent protective relationship with NAFLD. Besides, proper dietary strategies including mediterranean diet, intermittent fasting diet, ketogenic diet, and dietary approaches to stop hypertension diet have their inhibitory effects on the developmental process of NAFLD. Moreover, right exercises including walking, jogging, bicycling, and swimming are recommended for the prevention of NAFLD because they could effectively reduce weight, which is an important risk factor for NAFLD, and improve liver function. In addition, embracing a healthy lifestyle including reducing sedentary behavior, not smoking, sleeping well and brushing teeth regularly is integral since it not only could reduce the risk of NAFLD but also significantly contribute to overall prevention and control. Finally, the environment, including the social and natural environments, plays a potential role in NAFLD prevention. CONCLUSION Nutrition, dietary strategies, exercise, lifestyle and environment play an important role in the prevention of NAFLD. Moreover, this review offers comprehensive prevention recommendations for people at high risk of NAFLD.
Collapse
Affiliation(s)
- Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Hao Song
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Xin Su
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Jian Li
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Youbao Ye
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Cailiu Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Xiao Xu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Guanglong Pang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Wenxiu Liu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Zihan Li
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Tian Luo
- The Institute for Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
3
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Gui Z, Chen X, Wang D, Chen Z, Liu S, Yu G, Jiang Y, Duan H, Pan D, Lin X, Liu L, Wan H, Shen J. Inflammatory and metabolic markers mediate the association of hepatic steatosis and fibrosis with 10-year ASCVD risk. Ann Med 2025; 57:2486594. [PMID: 40189927 PMCID: PMC11980196 DOI: 10.1080/07853890.2025.2486594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND AIMS Liver steatosis and fibrosis increase the predicted 10-year atherosclerotic cardiovascular disease (ASCVD) risk, though the roles of chronic inflammation and metabolic dysregulation remain unclear. This cross-sectional study quantitatively assesses this association and evaluates the mediating effects of metabolic dysregulation and chronic inflammation. METHODS In this study, we enrolled 6110 adults from ten communities in Canton, China. Hepatic steatosis and fibrosis were assessed using vibration-controlled transient elastography (VCTE) through controlled attenuation parameter (CAP) and liver stiffness measurement (LSM), while predicted 10-year ASCVD risk was calculated using the China-PAR project model. Associations between CAP/LSM values and predicted 10-year ASCVD risk were analyzed. Mediation analysis quantified the effects of high-sensitivity C-reactive protein (hs-CRP), homeostasis model assessment of insulin resistance (HOMA-IR), remnant cholesterol (RC), and non-high-density lipoprotein cholesterol (non-HDL-C). The main statistical methods used included logistic regression, restricted cubic splines (RCS) analysis, interaction calculations, and mediation analysis to examine the relationships and mediators. RESULTS The study population had a mean age of 50.1 years (SD = 9.7), with 3927 females (64.3%) and 2183 males (35.7%). Additionally, 808 participants (13.2%) had type 2 diabetes, and 1911 participants (31.3%) had hypertension. Compared to the first CAP quartile (Q1), higher CAP quartiles showed increased odds ratios (OR) for predicted moderate to high 10-year ASCVD risk: 1.14 (0.89, 1.45), 1.37 (1.08, 1.73), and 2.44 (1.93, 3.10). Mediation analysis showed hs-CRP and HOMA-IR mediated CAP's link to ASCVD risk, with mediation proportions of 15.40% and 27.37%. RC and non-HDL-C mediated this association at 7.12% and 6.26%. Among patients with hepatic steatosis (CAP ≥ 248 dB/m), LSM Q4 participants had a significantly higher predicted 10-year ASCVD risk than those in LSM Q1 (OR 2.22, [1.52, 3.25]), with hs-CRP and HOMA-IR mediating 2.62% and 13.75%, respectively. CONCLUSION Liver steatosis and fibrosis were associated with the increased predicted ASCVD risk, with mediation effects from hs-CRP, HOMA-IR, RC, and non-HDL-C.
Collapse
Affiliation(s)
- Zihao Gui
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Xingying Chen
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Dongmei Wang
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Zhi Chen
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Siyang Liu
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Genfeng Yu
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Yuqi Jiang
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Hualin Duan
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Daoyan Pan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People’s Republic of China
| | - Xu Lin
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Lan Liu
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Heng Wan
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, Guangdong, China
| |
Collapse
|
5
|
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
|
6
|
Qian Z, Chen S, Liao X, Xie J, Xu Y, Zhong H, Ou L, Zuo X, Xu X, Peng J, Wu J, Cai S. Decreased intestinal abundance of Akkermansia muciniphila is associated with metabolic disorders among people living with HIV. Ann Med 2025; 57:2474730. [PMID: 40052450 DOI: 10.1080/07853890.2025.2474730] [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: 10/30/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/12/2025] Open
Abstract
BACKGROUND Previous studies have shown changes in gut microbiota after human immunodeficiency virus (HIV) infection, but there is limited research linking the gut microbiota of people living with HIV (PLWHIV) to metabolic diseases. METHODS A total of 103 PLWHIV were followed for 48 weeks of anti-retroviral therapy (ART), with demographic and clinical data collected. Gut microbiome analysis was conducted using metagenomic sequencing of fecal samples from 12 individuals. Nonalcoholic fatty liver disease (NAFLD) was diagnosed based on controlled attenuation parameter (CAP) values of 238 dB/m from liver fibro-scans. Participants were divided based on the presence of metabolic disorders, including NAFLD, overweight, and hyperlipidemia. Akkermansia abundance in stool samples was measured using RT-qPCR, and Pearson correlation and logistic regression were applied for analysis. RESULTS Metagenomic sequencing revealed a significant decline in gut Akkermansia abundance in PLWHIV with NAFLD. STAMP analysis of public datasets confirmed this decline after HIV infection, while KEGG pathway analysis identified enrichment of metabolism-related genes. A prospective cohort study with 103 PLWHIV followed for 48 weeks validated these findings. Akkermansia abundance was significantly lower in participants with NAFLD, overweight, and hyperlipidemia at baseline, and it emerged as an independent predictor of NAFLD and overweight. Negative correlations were observed between Akkermansia abundance and both CAP values and body mass index (BMI) at baseline and at week 48. At the 48-week follow-up, Akkermansia remained a predictive marker for NAFLD. CONCLUSIONS Akkermansia abundance was reduced in PLWHIV with metabolic disorders and served as a predictive biomarker for NAFLD progression over 48 weeks of ART.
Collapse
Affiliation(s)
- Zhe Qian
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suling Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyang Liao
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Jingfang Xie
- Department of Geriatrics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Yuyuan Xu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huiqun Zhong
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lang Ou
- Department of hepatobiliary surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Xiang Zuo
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuwen Xu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Wu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shaohang Cai
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Li Y, Cong S, Chen R, Tang J, Zhai L, Liu Y. Kaili sour soup in alleviation of hepatic steatosis in rats via lycopene route: an experimental study. Ann Med 2025; 57:2479585. [PMID: 40257305 PMCID: PMC12013139 DOI: 10.1080/07853890.2025.2479585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver diseases, with a range of manifestations, such as hepatic steatosis. Our previous study showed that Kaili Sour Soup (KSS) significantly attenuated hepatic steatosis in rats. This study explored the main components of KSS and the mechanisms by which it exerts its protective effects against NAFLD. METHODS Twenty-four 6-week-old male Sprague-Dowley (SD) rats were randomly assigned to three treatments: feeding a normal standard diet, a high-fat diet, or a high-fat diet plus gavage KSS. The effects of KSS treatment on hepatic lipid accumulation were assessed using biochemical, histological, and molecular experiments. The amounts of KSS ingredients were measured using biochemical assays. Network pharmacology analyses were performed to identify the hub genes of KSS targets and enriched pathways. CCK-8 assay was used to determine the effect of free fatty acids (FFA), lycopene, and estrogen on HepG2 viability. Quantitative Real-Time polymerase chain reaction (qRT-PCR) and Western blot assays were performed to determine the effect of KSS or lycopene on estrogen signaling and expression of lipid metabolism-related molecules. Statistical analyses were performed using GraphPad Prism and SPSS. RESULTS KSS alleviated fat deposition in rat liver tissue and affected the expression of hepatic lipid synthesis, catabolism, and oxidative molecules. Lycopene was identified as the ingredient with the highest amount in KSS. Network pharmacology analyses showed that the hub genes were enriched in the estrogen signaling pathway. Cellular experiments showed that lycopene increased the expression of Estrogen Receptor α (ERα), Carnitine palmitoyltransferase 1 A (CPT1A), Peroxisome proliferator-activated receptor α (PPARα) (all p < 0.01), and Hormone sensitive lipase (HSL) (p < 0.05), and reduced the expression of lipid metabolism-related factors 1c(SREBP-1c) (p < 0.01), Acetyl-CoA carboxylase 1 (ACC) and Lipoprotein lipase (LPL) (all p < 0.05). CONCLUSIONS KSS ameliorated abnormal lipid metabolism in patients with NAFLD. Lycopene was the major component of KSS, and it affected estrogen signaling and the expression of lipid metabolism molecules. In short, both KSS and LYC could change lipid metabolism by lowering lipid accumulation and raising lipolysis.
Collapse
Affiliation(s)
- Yi Li
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Third People’s Hospital of Guizhou Province, Laboratory Department, Guiyang, Guizhou Province, China
| | - Shuo Cong
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Rui Chen
- Acupuncture and Moxibustion Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Juan Tang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Liqiong Zhai
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yongmei Liu
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou Province, China
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
8
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [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: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
9
|
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
|
10
|
Zhang D, Wang Q, Li D, Chen S, Chen J, Zhu X, Bai F. Gut microbiome composition and metabolic activity in metabolic-associated fatty liver disease. Virulence 2025; 16:2482158. [PMID: 40122128 PMCID: PMC11959907 DOI: 10.1080/21505594.2025.2482158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/24/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Metabolic Associated Fatty Liver Disease (MAFLD) impacts approximately 25% of the global population. Between April 2023 and July 2023, 60 patients with MAFLD, along with 60 age, ethnicity, and sex-matched healthy controls (HCs), were enrolled from the Inner Mongolia Autonomous Region, China. Analysis of gut microbiota composition and plasma metabolic profiles was conducted using metagenome sequencing and LC-MS. LEfSe analysis identified five pivotal species: Eubacterium rectale, Dialister invisus, Pseudoruminococcus massiliensis, GGB3278 SGB4328, and Ruminococcaceae bacteria. In subgroup analysis, Eubacterium rectale tended to increase by more than 2 times and more than double in the non-obese MAFLD group, and MAFLD with moderate hepatic steatosis (HS), respectively. Plasma samples identified 172 metabolites mainly composed of fatty acid metabolites such as propionic acid and butyric acid analogues. Ruminococcaceae bacteria have a strong positive correlation with β-alanine, uric acid, and L-valine. Pseudoruminococcus massiliensis has a strong positive correlation with β-alanine. Combinations of phenomics and metabolomics yielded the highest accuracy (AUC = 0.97) in the MAFLD diagnosis. Combinations of phenomics and metagenomics yielded the highest accuracy (AUC = 0.94) in the prediction of the MAFLD HS progress. Increases in Eubacterium rectale and decreases in Dialister invisus seem to be indicative of MAFLD patients. Eubacterium rectale may predict HS degree of MAFLD and play an important role in the development of non-obese MAFLD. Eubacterium rectale can generate more propionic acid and butyric acid analogues to absorb energy and increase lipid synthesis and ultimately cause MAFLD.
Collapse
Affiliation(s)
- Daya Zhang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Qi Wang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Da Li
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Shiju Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Jinrun Chen
- Department of Gastroenterology, Otog Front Banner People 's Hospital, Otog Front Banner, China
| | - Xuli Zhu
- Department of Gastroenterology, Otog Front Banner People 's Hospital, Otog Front Banner, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
11
|
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
|
12
|
Santos-Sánchez G, Cruz-Chamorro I. Plant-derived bioactive peptides and protein hydrolysates for managing MAFLD: A systematic review of in vivo effects. Food Chem 2025; 481:143956. [PMID: 40147387 DOI: 10.1016/j.foodchem.2025.143956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) represents a growing health concern worldwide. Among the pursuit of therapeutic interventions, interest in natural bioactive compounds has intensified because of their potential hepatoprotective effects. This systematic review aims to evaluate the impact of plant-derived hydrolysates and peptides on MAFLD through the current literatures, encompassing their mechanisms of action. Key outcomes evaluated included changes in liver enzymes, liver lipid content, inflammation markers, and histopathological improvements. Preliminary findings suggest a potential beneficial effect of plant-derived hydrolysates and peptides on the improvement of MAFLD-related parameters, with mechanisms implicating antioxidant, anti-inflammatory, and lipid-lowering properties. This review highlights emerging evidence supporting the potential therapeutic role of plant-derived hydrolysates and peptides in the management of MAFLD. However, more well-designed clinical trials with larger sample sizes and longer durations are warranted to elucidate their efficacy, optimal dose, and long-term safety.
Collapse
Affiliation(s)
- Guillermo Santos-Sánchez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), 28049 Madrid, Spain.
| | - Ivan Cruz-Chamorro
- Facultad de Enfermería, Universidad de Castilla-La Mancha, 02071 Albacete, Spain.
| |
Collapse
|
13
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [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: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
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
|
15
|
Martínez-Sánchez FD, Corredor-Nassar MJ, Feria-Agudelo SM, Paz-Zarza VM, Martinez-Perez C, Diaz-Jarquin A, Manzo-Santana F, Sánchez-Gómez VA, Rosales-Padron A, Baca-García M, Mejía-Ramírez J, García-Juárez I, Higuera-de la Tijera F, Pérez-Hernandez JL, Barranco-Fragoso B, Méndez-Sánchez N, Córdova-Gallardo J. Factors Associated With Advanced Liver Fibrosis in a Population With Type 2 Diabetes: A Multicentric Study in Mexico City. J Clin Exp Hepatol 2025; 15:102536. [PMID: 40226389 PMCID: PMC11982025 DOI: 10.1016/j.jceh.2025.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/23/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major cause of chronic liver disease, primarily due to insulin resistance and type 2 diabetes (T2D). Despite the strong link between T2D and MASLD, identifying and treating liver fibrosis in T2D patients is still poor. This study aimed to identify the factors related to advanced liver fibrosis in T2D patients. METHODS This retrospective observational study used medical records from four centers in Mexico City from 2018 to 2023. The study included 2000 patients with T2D. Liver fibrosis was evaluated using the Fibrosis-4 (FIB-4) index, and insulin resistance was assessed using the estimated glucose disposal rate (eGDR). RESULTS The mean age of the patients was 58.9 years, with 63.7% being women. The median duration of T2D was 7 years, and the mean HbA1c was 7.63%. Overall, 20.4% had advanced liver fibrosis. The multivariate logistic regression analysis showed that diabetes duration >10 years {odds ratio (OR) = 2.105 (95% confidence interval [CI] 1.321-3.355)}, fasting glucose >126 mg/dL (OR = 1.568 [95% CI 1.085-2.265]), and microalbuminuria >300 mg/24 h (OR = 2.007 [95% CI 1.134-3.552]) were associated with advanced liver fibrosis. Conversely, the eGDR (OR = 0.805 [95% CI 0.703-0.888]), statins (OR = 0.111 [95% CI 0.073-0.168]), and pioglitazone (OR = 0.082 [95% CI 0.010-0.672]) were inversely associated. CONCLUSION Longer diabetes duration, insulin resistance, and microalbuminuria are independently linked to advanced liver fibrosis in T2D patients. Statins and pioglitazone may protect against liver fibrosis. Enhanced screening and management strategies targeting these factors could slow fibrosis progression and reduce the global burden of MASLD.
Collapse
Affiliation(s)
- Froylan D. Martínez-Sánchez
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Escolar 411A, Copilco Universidad, Coyoacán, 04360 Ciudad de México, Mexico
- Department of Internal Medicine, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
- Department of Hepatology, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Maria J. Corredor-Nassar
- Department of Internal Medicine, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Sandra M. Feria-Agudelo
- Department of Internal Medicine, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Victor M. Paz-Zarza
- Department of Internal Medicine, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Carolina Martinez-Perez
- Department of Hepatology, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Alejandra Diaz-Jarquin
- Department of Internal Medicine, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Fátima Manzo-Santana
- Department of Gastroneterology, Instituto Nacional de Ciencias Medicas y Nutricion “Salvador Zubiran”, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Victor A. Sánchez-Gómez
- Department of Gastroneterology, Instituto Nacional de Ciencias Medicas y Nutricion “Salvador Zubiran”, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Alondra Rosales-Padron
- Department of Gastroneterology, Instituto Nacional de Ciencias Medicas y Nutricion “Salvador Zubiran”, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Mónica Baca-García
- Department of Gastroneterology and Hepatology, Hospital General de Mexico “Dr. Eduardo Liceaga”, Dr. Balmis 148, Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Jessica Mejía-Ramírez
- Department of Gastroneterology and Hepatology, Hospital General de Mexico “Dr. Eduardo Liceaga”, Dr. Balmis 148, Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Ignacio García-Juárez
- Department of Gastroneterology, Instituto Nacional de Ciencias Medicas y Nutricion “Salvador Zubiran”, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| | - Fatima Higuera-de la Tijera
- Department of Gastroneterology and Hepatology, Hospital General de Mexico “Dr. Eduardo Liceaga”, Dr. Balmis 148, Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Jose L. Pérez-Hernandez
- Department of Gastroneterology and Hepatology, Hospital General de Mexico “Dr. Eduardo Liceaga”, Dr. Balmis 148, Doctores, Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Beatriz Barranco-Fragoso
- Department of Gastroneterology, Centro Medico Nacional 20 de Noviembre, ISSSTE, Félix Cuevas 540, Col del Valle Sur, Benito Juárez, 03104 Ciudad de México, Mexico
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Tlalpan, 14050, Ciudad de México, Mexico
| | - Jacqueline Córdova-Gallardo
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Escolar 411A, Copilco Universidad, Coyoacán, 04360 Ciudad de México, Mexico
- Department of Hepatology, Hospital General “Dr. Manuel Gea González”, Calz. de Tlalpan 4800, Belisario Domínguez Secc 16, Tlalpan, 14080 Ciudad de México, Mexico
| |
Collapse
|
16
|
Wu Z, Zhang M, Yan MK, Li C, Pan G, Xuan L. Synthesis and biological evaluation of amino-conjugated bile acid derivatives against non-alcoholic steatohepatitis. Bioorg Med Chem Lett 2025; 122:130210. [PMID: 40139332 DOI: 10.1016/j.bmcl.2025.130210] [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: 12/19/2024] [Revised: 02/24/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Non-alcoholic steatohepatitis (NASH) is emerging as a rapidly growing health concern. Bile acids (BAs) function as endocrine signaling molecules and exhibit therapeutic potential for NASH. To develop safer and more effective BA derivatives for NASH treatment, 25 amino acid-conjugated bile acid derivatives were designed and synthesized based on the pharmacological properties of the leading compound A17. The anti-lipid accumulation, anti-inflammatory and anti-fibrosis activities of these compounds were evaluated, and their structure-activity relationships were elucidated. Notably, compound C04 exhibited superior in vitro activity compared to obeticholic acid and demonstrated enhanced efficacy in improving both NASH and fibrosis in preclinical murine models via oral administration. These findings suggest that C04 is a promising candidate for NASH treatment and warrants further investigation.
Collapse
Affiliation(s)
- Zhitao Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China; State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China
| | - Mingge Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Meng Kun Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Chenyue Li
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Guoyu Pan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China; State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| | - Lijiang Xuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China; State Key Laboratory of Drug Research, Shanghai Institute of Material Medica, Chinese Academy of Sciences,501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
17
|
Saha T, Mehrotra S, Gupta P, Kumar A. Exosomal miRNA combined with anti-inflammatory hyaluronic acid-based 3D bioprinted hepatic patch promotes metabolic reprogramming in NAFLD-mediated fibrosis. Biomaterials 2025; 318:123140. [PMID: 39892017 DOI: 10.1016/j.biomaterials.2025.123140] [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: 09/05/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex metabolic disorder, where the underlying molecular mechanisms are mostly not well-understood and therefore, warrants the need for therapeutic interventions targeting several metabolic pathways as a unified response. Of late, promising outcomes have been observed with mesenchymal stem cell-derived exosomes. However, reduced bioavailability due to systemic delivery and the need for repeated fresh isolation hinders their feasibility for clinical applications. In this regard, an 'off-the-shelf' 3D bioprinted hyaluronic acid-based hepatic patch to deliver encapsulated exosomes alone/or with hepatocytes (as dual-therapy) is developed as a holistic approach for ameliorating the disease condition and promoting tissue regeneration. The bioprinted hepatic patch demonstrated sustained and localized release of exosomes (∼82 % in 21 days), and healthy liver tissue-like mechanical properties while being biocompatible and biodegradable. Assessment in NAFLD rat models displayed alleviation of the altered biochemical parameters such as fat deposition, deranged liver functions, disrupted lipid, glucose, and insulin metabolism along with a reduction in localized inflammation, and associated liver fibrosis. The study suggests that a synergistic effect between the miRNA population of released exosomes, cell therapy, and the bioprinted matrix materials is crucial in targeting multiple complex metabolic pathways associated with the severity of the disease.
Collapse
Affiliation(s)
- Triya Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| | - Purva Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| |
Collapse
|
18
|
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
|
19
|
van Kleef LA, Pustjens J, Janssen HLA, Brouwer WP. Diagnostic Accuracy of the LiverRisk Score to Detect Increased Liver Stiffness Among a United States General Population and Subgroups. J Clin Exp Hepatol 2025; 15:102512. [PMID: 40093506 PMCID: PMC11908561 DOI: 10.1016/j.jceh.2025.102512] [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: 02/02/2025] [Indexed: 03/19/2025] Open
Abstract
Background The LiverRisk score (LRS) has recently been proposed to predict liver fibrosis and future development of liver-related outcomes in the general population. Here, we performed an external validation of this score. Methods We used data from National Health and Nutrition Examination Survey 2017-2020, a United States population-based cohort to assess the diagnostic accuracy of the LRS to detect a liver stiffness measurement (LSM) ≥8 and ≥12 kPa. Performance was tested among the entire general population and clinically relevant subgroups. Results The cohort comprised 7,025 participants (aged 49 [33-63], 49% male), and 9.7% had an LSM ≥8 and 3.2% had an LSM ≥12 kPa. The area under the receiver characteristic operator curve (AUC) in the overall population was 0.73 (95% confidence interval [CI] :0.71-0.75) and 0.78 (95% CI: 0.74-0.81) to detect an LSM ≥8 and ≥ 12 kPa, respectively, significantly outperforming the fibrosis 4 index (FIB-4) but not the nonalcoholic fatty liver disease fibrosis score, steatosis-associated fibrosis estimator (SAFE), or metabolic dysfunction-associated fibrosis 5 (MAF-5). Performance was consistent among most subgroups, but AUC levels to detect an LSM ≥8 kPa decreased to <0.70 among participants aged 18-40 or 60-80 years, blacks, and individuals with diabetes or liver steatosis. The LRS categorized 80.5% as very low risk, 17.7% as low risk, and 1.8% as at risk, prevalence of an LSM ≥8 in these groups was 6.3%, 20.8%, and 50.5%, respectively. The sensitivity to detect an LSM ≥8 kPa was 47.3% in the overall population (but dropped to 21.3% for individuals aged 18-40 years) despite applying the lowest cut-off, which should yield the highest sensitivity. Conclusion The LRS score is a promising new tool to predict liver fibrosis; however, its diagnostic accuracy attenuates especially among patients aged 18-40 or 60-80 years. The overall sensitivity was only 47.3% at the lowest LRS cut-off. Further studies assessing cost-benefit ratios according to the LRS compared to FIB-4 and other risk scores such as MAF-5 and SAFE are required to determine its usefulness in referral strategies.
Collapse
Affiliation(s)
- Laurens A van Kleef
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Jesse Pustjens
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Canada
| | - Willem P Brouwer
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Xiong X, Du Y, Liu P, Li X, Lai X, Miao H, Ning B. Unveiling EIF5A2: A multifaceted player in cellular regulation, tumorigenesis and drug resistance. Eur J Pharmacol 2025; 997:177596. [PMID: 40194645 DOI: 10.1016/j.ejphar.2025.177596] [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: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
The eukaryotic initiation factor 5A2 gene (EIF5A2) is a highly conserved and multifunctional gene that significantly influences various cellular processes, including translation elongation, RNA binding, ribosome binding, protein binding and post-translational modifications. Overexpression of EIF5A2 is frequently observed in multiple cancers, where it functions as an oncoprotein. Additionally, EIF5A2 is implicated in drug resistance through the regulation of various molecular pathways. In the review, we describe the structure and functions of EIF5A2 in normal cells and its role in tumorigenesis. We also elucidate the molecular mechanisms associated with EIF5A2 in the context of tumorigenesis and drug resistance. We propose that the biological roles of EIF5A2 in regulating diverse cellular processes and tumorigenesis are clinically significant and warrant further investigation.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China; Guangzhou Institute of Burn Clinical Medicine, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Yanli Du
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Peng Liu
- Departments of Burn and Plastic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xinye Li
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xudong Lai
- Department of infectious disease, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Haixiong Miao
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| | - Bo Ning
- Department of Neurosurgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| |
Collapse
|
24
|
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
|
25
|
Tang Y, Fan J, Hou X, Wu H, Zhang J, Wu J, Wang Y, Zhang Z, Lu B, Zheng J. Metabolic dysfunction-associated steatotic liver disease and increased risk of atrial fibrillation in the elderly: A longitudinal cohort study. IJC HEART & VASCULATURE 2025; 58:101676. [PMID: 40255886 PMCID: PMC12008591 DOI: 10.1016/j.ijcha.2025.101676] [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: 01/05/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/22/2025]
Abstract
Background Emerging evidence suggests a link between metabolic dysfunction-associated steatotic liver disease (MASLD) and cardiac arrhythmia. This study aims to investigate the potential relationship between MASLD and atrial fibrillation (AF). Methods This retrospective cohort study included 8511 participants (age > 65 years) without a history of cardiovascular diseases, cancer, or severe kidney dysfunction. MASLD was diagnosed using hepatic ultrasound in the presence of at least one cardiometabolic risk factor. Poisson regression models were employed to estimate the relative risk (RR) of AF, adjusting for potential confounders. Results Participants were categorized into MASLD (n = 3,926) and non-MASLD (n = 4,585) groups. During a mean follow-up period of 3.65 ± 1.20 years, 307 participants with MASLD developed AF, however, the number in the non-MASLD group was 144 (incidence rate 7.82 % vs. 3.14 %). After adjusting for multiple cardiovascular risk factors, MASLD was associated with increased risk of AF (RR = 1.55, 95 %, confidence interval (CI): 1.12-2.13). Positive correlations were observed between age, body mass index (BMI), systolic and diastolic blood pressure, low-density lipoprotein levels, and AF risk. Subgroup analysis revealed a stronger association between MASLD and AF in participants with BMI < 24 kg/m2 (P < 0.01). Conclusion This study highlights a significant association between MASLD and an increased risk of developing AF. The elevated risk in patients with MASLD may involve mechanisms extending beyond traditional cardiometabolic factors, particularly in individuals with lower BMI. Further experimental research is warranted to elucidate the underlying pathways linking MASLD and AF.
Collapse
Affiliation(s)
- Yehua Tang
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jianling Fan
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xingyun Hou
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Honghong Wu
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jiaqi Zhang
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jia Wu
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yifan Wang
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Zhiyu Zhang
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Bin Lu
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Jiaoyang Zheng
- Health Management Centre, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| |
Collapse
|
26
|
Taheri E, Ehtesham H, Safdari R, Hormati A. A scientometric analysis and visualization of research on fatty liver diseases in Iran from 2003 to 2023. J Diabetes Metab Disord 2025; 24:103. [PMID: 40224531 PMCID: PMC11992304 DOI: 10.1007/s40200-025-01606-8] [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: 01/14/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025]
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide and is now a major global health threat. Due to the mechanistic links between these conditions, along with the rising prevalence of fatty liver disease, diabetes, and obesity, MAFLD (metabolic-associatedfatty liver diseases) was introduced as a new terminology and then updated to MASLD (Metabolic Dysfunction-associated Steatotic Liver Disease. These changes reflect a growing recognition of the importance of fatty liver and its associated health risks. Methods This scientometric study analyzed publications on "fatty liver diseases" (FLD) indexed in Scopus from 2003 to 2023 in Iran. Online Analysis Platforms and VOSviewer were used to assess publication trends and identify research hotspots. Results We retrieved 1,857 English articles on fatty liver diseases published between 2003 and 2023. The Journal of Hepatitis Monthly was notable for Iranian publications on fatty liver disease. Most publications were original articles and Dr. Sahebkar AH was the most prolific author. Iranian researchers primarily collaborated with scholars from the United States. The leading institution in terms of productivity was Tehran University of Medical Sciences. Conclusion This study reveals an increasing trend in both the number of Iranian publications and the citations of articles in the field of FLD. We believe this study can serve as a roadmap for future research and policy development on fatty liver diseases, which are a significant public health concern in Iran. Additionally, new strategies are needed to foster multi-disciplinary research and enhance international cooperation. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-025-01606-8.
Collapse
Affiliation(s)
- Ehsaneh Taheri
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamideh Ehtesham
- Department of Health Information Technology, Birjand University of Medical Sciences, Birjand, Iran
| | - Reza Safdari
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hormati
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Han Y, Zhang Y, Chen J, Jiang S, Zheng Y, Xu Y, Li Y, Kong J, Yu X, Du H. Iron overload exacerbates metabolic dysfunction-associated steatohepatitis via the microbiota-gut-liver axis through lipopolysaccharide-mediated Akr1b8 activation. Free Radic Biol Med 2025; 233:196-208. [PMID: 40157463 DOI: 10.1016/j.freeradbiomed.2025.03.039] [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: 02/11/2025] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Iron homeostatic is closely linked to the development of metabolic dysfunction-associated steatohepatitis (MASH). However, the underlying mechanisms remain poorly understood. HFE knockout (KO) mice were used to generate mild iron-overload models. MASH was induced by feeding mice a methionine- and choline-deficient (MCD) diet for 4 weeks. Iron overload significantly exacerbated the pathologies of MCD-induced MASH, including liver injury, hepatic lipid accumulation, inflammation, and fibrosis. Additionally, iron overload reshaped the composition of gut microbiota, and fecal microbiota transplantation assay proved that gut microbiota from iron-overload mice contributed to hepatic lipid accumulation in control mice. Furthermore, iron overload-induced dysbacteriosis altered the metabolite profiles, reducing short-chain fatty acid levels and increasing lipopolysaccharide (LPS) levels. Notably, elevated LPS levels upregulated the expression of aldo-keto reductase family 1 member B8 (Akr1b8), which accelerated lipid accumulation and inflammation in hepatocytes. Above results indicated that iron overload promoted MASH progression through the microbiota-gut-liver axis, mediated by LPS-induced activation of Akr1b8. These findings highlight the critical role of iron homeostasis and gut microbiota in MASH pathogenesis.
Collapse
Affiliation(s)
- Yu Han
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhui Zhang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianjun Chen
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shouchuan Jiang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi Zheng
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yecheng Xu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yunqin Li
- Analysis Center of Agrobiology and Environmental Science, Zhejiang University, Hangzhou, 310058, China
| | - Jingxia Kong
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China
| | - Xin Yu
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, China.
| | - Huahua Du
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China; Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, 310016, China.
| |
Collapse
|
30
|
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
|
31
|
Sardar MA, Abbasian S, Moghavemi H, Karabi M. HIIT may ameliorate inter-organ crosstalk between liver and hypothalamus of HFD-induced MAFLD rats; A two-phase study to investigate the effect of exercise intensity as a stressor. Brain Res 2025; 1856:149591. [PMID: 40120709 DOI: 10.1016/j.brainres.2025.149591] [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: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Previous studies demonstrate that GDF15 and its related signaling activators may be affected by exercise training, leading to the suppression of inflammatory factors and the promotion of immune-metabolic balance. Therefore, the purpose of the study was to evaluate the effect of high-intensity interval training (HIIT) on amelioration of inter-organ crosstalk between liver and hypothalamus of the high-fat diet (HFD)-induced metabolic dysfunction-associated fatty liver disease (MAFLD) rats in a two-phase study. In this regard, rats were initially divided into two groups, the normal diet-inactive (NS) and the HFD groups. HFD course lasted 12 weeks to induce MAFLD in the latter group. After ensuring the induction of MAFLD, 25 rats were divided into three groups: the HFD-inactive group (HS), the HFD-HIIT group (HH), as well as the HFD-aerobic group (HA). The training interventions were consistently applied over a period of eight weeks, five days a week, with each session lasting 40-60 min, and the duration of the whole research was 21 weeks. The results of this study displayed that HIIT intervention promotes hypothalamic Gdf15 gene expression and there were similar alterations in genes expression of Foxo1 and Akt2. Moreover, our results confirmed that HIIT ameliorated hypothalamic NFKB gene expression and there was a similar trend in genes expression of Tnfa and Il1b following both HIIT as well as aerobic training protocols. Taking these findings together, it is concluded that interventions, particularly exercise training, uniquely contribute to the reduction of hypothalamic-associated inflammatory responses that result in prolonged and chronic increases in GDF15.
Collapse
Affiliation(s)
- Mohammad Ali Sardar
- Department of General Courses, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sadegh Abbasian
- Department of Physical Education, Farhangian University, P.O. Box 14665-889, Tehran, Iran.
| | - Hamid Moghavemi
- Department of Exercise Physiology, Faculty of Sport Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mina Karabi
- Department of Sport Sciences, Khavaran Institute of Higher Education, Mashhad, Iran
| |
Collapse
|
32
|
Ren Q, Tan Y, Zhang G, Dai Y, Yang L, Wu Y, He H, Chen J. Efficacy of Hypoglycemic Agents in Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD): A Systematic Review and Network Meta-Analysis. J Evid Based Med 2025; 18:e70021. [PMID: 40229658 DOI: 10.1111/jebm.70021] [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: 01/23/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025]
Abstract
AIMS Metabolic dysfunction associated steatotic liver disease (MASLD) is a universal hepatic disease, and many recent randomized clinical trials (RCTs) have explored whether hypoglycemic agents may be beneficial for its treatment. This study aimed to assess the relative effectiveness of each hypoglycemic agent for MASLD. METHODS China National Knowledge Infrastructure(CNKI), WanFang, Weipu, PubMed, Embase, The Cochrane Library, and Web of Science Core Collection were searched for RCTs on the efficacy of hypoglycemic agents in MASLD published up to December 31, 2024. All statistical analyses were performed using R version 4.3.3. The network meta-analysis was conducted using Bayesian statistical methods. RESULTS A total of 26 hypoglycemic agents for treating MASLD in 37 studies with 2406 participants were included. Empagliflozin was most effective in improving liver stiffness measurement (LSM), whereas liraglutide showed significant benefits in body weight, body mass index (BMI), and waist circumference. Both sodium-glucose co-transporter 2 (SGLT-2) inhibitors (e.g., empagliflozin) and glucagon-like peptide-1 (GLP-1) receptor agonists (e.g., liraglutide) improved liver enzymes (alanine aminotransferase [ALT], aspartate aminotransferase [AST], gamma-glutamyltransferase [GGT]), glucose metabolism (fasting plasma glucose [FPG], and homeostasis model assessment of insulin resistance [HOMA-IR]), and lipid profiles. Pioglitazone had limited benefits in these outcomes. Secondary outcomes such as inflammatory markers and fibrosis showed minimal changes. CONCLUSIONS Several hypoglycemic agents can improve laboratory and imaging indicators in adult patients with MASLD. Liraglutide is more effective than other agents, whereas empagliflozin emerged as the most effective for reducing LSM. However, different agents have different effects on the indicators; therefore, the relevant agents must be selected according to the specific patient condition.
Collapse
Affiliation(s)
- Qiao Ren
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Tan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Guixiang Zhang
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhao Dai
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Lidan Yang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
| | - Yunmo Wu
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - He He
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Department of Laboratory Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jie Chen
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
Tizianel I, Madinelli A, Crimì F, Barbot M, Censi S, Sabbadin C, Ceccato F. Prevalence of Metabolic-Associated Steatotic Liver Disease in Patients With Primary Aldosteronism. Clin Endocrinol (Oxf) 2025; 102:618-625. [PMID: 40079488 PMCID: PMC12046541 DOI: 10.1111/cen.15231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
OBJECTIVE To assess the prevalence of metabolic associated steatotic liver disease (MASLD) in patients with primary aldosteronism (PA) compared to benign adrenal adenomas, and to evaluate the impact of hormonal excess in inducing MASLD. DESIGN Single-centre retrospective study. METHODS Hepatic steatosis was assessed by liver/spleen (L/S) ratio from unenhanced abdomen computed tomography images (reference value < 1.1) in a cohort of 41 patients with PA without cortisol cosecretion, 20 unilateral (uPA) and 21 bilateral (BPA), 50 with nonfunctioning adrenal incidentalomas (NF-AI), 48 with mild autonomous cortisol secretion (MACS) and 10 with adrenal Cushing Syndrome (CS). RESULTS Hepatic steatosis was increased in patients with PA at diagnosis: L/S ratio was lower in PA than NF-AI (1.1 vs. 1.25, p < 0.001) and MACS (1.1 vs. 1.21, p 0.007), but was similar to adrenal CS (1.1 vs. 1.15, p = 0.147). A improvement in L/S ratio after medical or surgical treatment was observed in PA patients, resulting in reduced liver steatosis. MASLD prevalence was higher in PA compared to MACS (49% vs. 25%, p < 0.05) and NF-AI (49% vs. 14%, p < 0.001), but similar to CS (49% vs. 45%, p = 0.61). uPA patients had higher MASLD prevalence compared to BPA group 71% (53%-89%) versus 25% (7%-43%). CONCLUSIONS Prevalence of MASLD was increased in PA (higher in uPA than BPA) compared to MACS and NFAI, and similar to adrenal CS.
Collapse
Affiliation(s)
- Irene Tizianel
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| | - Alberto Madinelli
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| | - Filippo Crimì
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Institute of Radiology, University‐Hospital of PadovaPaduaItaly
| | - Mattia Barbot
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| | - Simona Censi
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| | - Chiara Sabbadin
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| | - Filippo Ceccato
- Department of Medicine DIMEDEndocrine UnitPaduaItaly
- Endocrine Unit, University‐Hospital of PadovaPaduaItaly
| |
Collapse
|
36
|
Taheri E, Yilmaz Y, Ghorat F, Moslem A, Zali MR. Association of diet quality scores with risk of metabolic-associated fatty liver disease in Iranian population: a nested case-control study. J Diabetes Metab Disord 2025; 24:46. [PMID: 39816985 PMCID: PMC11729581 DOI: 10.1007/s40200-024-01544-x] [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: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025]
Abstract
Background and aim A healthy diet has been recommended for non-alcoholic fatty liver disease (NAFLD). We aim to investigate the associations of diet quality indices with the risk of developingmetabolic-associated fatty liver disease (MAFLD). Methods We conducted this nested case-control study by recruiting 968 cases with MAFLD and 964 controls from the participants of the baseline phase of the Sabzevar Persian Cohort Study (SPCS). MAFLD was defined as having a fatty liver index ≥ 60 plus at least one of the following: overweight or obese, Type II diabetes mellitus, or evidence of metabolic dysregulation. Healthy Eating Index-2015 (HEI-2015) and Alternative Healthy Eating Index-2010 (AHEI-2010) were calculated from a validated food frequency questionnaire. We estimated the associations of HEI-2015 and AHEI-2010 with MAFLD risk using multivariable logistic regression. Results Among those in the highest relative to the lowest quintile of HEI-2015 and AHEI-2010, the multivariable-adjusted odds ratios (OR) were 0.45 (95% CI [confidence interval] 0.29-0.69; P trend = 0.002) and 0.55 (95% CI 0.35-0.85; P trend = 0.04), respectively. Conclusion The results of our study suggest that there is a significant associationbetween adherence to a healthy diet, indicated by a higher score of HEI or AHEI, and a reduced likelihood of developingMAFLD. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-024-01544-x.
Collapse
Affiliation(s)
- Ehsaneh Taheri
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
| | - Fereshteh Ghorat
- Non-communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Alireza Moslem
- Department of Anesthesiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Huang C, Luo Y, Liu Y, Liu J, Chen Y, Zeng B, Liao X, Liu Y, Wang X. DNA hypermethylation-induced suppression of ALKBH5 is required for folic acid to alleviate hepatic lipid deposition by enhancing autophagy in an ATG12-dependent manner. J Nutr Biochem 2025; 140:109870. [PMID: 39993647 DOI: 10.1016/j.jnutbio.2025.109870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/08/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when too much fat builds up in the liver. As a growing worldwide epidemic, NAFLD is strongly linked with multiple metabolic diseases including obesity, insulin resistance, and dyslipidemia. However, very few effective treatments are currently available. Folate, an essential B-group vitamin with important biological functions including DNA and RNA methylation regulation, has been shown to have a protective effect against NAFLD with its underlying mechanism remains largely unclear. Here, we show that administration of folic acid significantly improves glucose tolerance, insulin sensitivity, and dyslipidemia in high-fat diet (HFD) fed mice. Moreover, folic acid treatment significantly inhibits lipid deposition in hepatocytes both in vivo and in vitro. Mechanically, folic acid reduces the expression of m6A demethylase AlkB homolog 5 (ALKHB5) via promoter DNA hypermethylation. Decreased ALKBH5 causes increased m6A modification and increased expression of ATG12 in a demethylase activity-dependent manner, thereby promoting autophagy and preventing hepatic steatosis. Inhibition of ATG12 induced by overexpression of ALKBH5 could impair autophagy and the inhibitory effect of folic acid on lipid accumulation in hepatocytes. Together, these findings provide novel insights into understanding the protective role of folic acid in the treatment of NAFLD and suggest that folic acid may be a potential agent for combating NAFLD.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China.
| |
Collapse
|
38
|
Jin H, Zhong X, Zhang C, Wu Y, Sun J, Wang X, Wang Z, Zhu J, Jiang Y, Du X, Zhang Z, Zhang D, Sun G. CD73 promotes the immunoregulatory functions of hepatic Tregs through enzymatic and nonenzymatic pathways in MASLD development. Mol Metab 2025; 96:102131. [PMID: 40139441 PMCID: PMC12002957 DOI: 10.1016/j.molmet.2025.102131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading chronic liver disease characterized by chronic inflammation. Regulatory T cells (Tregs) highly express CD73 and play a critical role in modulating the immune response. However, the roles and mechanisms by which CD73 modulates Tregs in MASLD are still unknown. A choline-deficient high-fat diet (CDHFD) or methionine/choline-deficient diet (MCD) was used to establish a MASLD model. We found that CD73 expression was upregulated in Tregs via the FFA-mediated p38/GATA2 signaling pathway. Cd73 KO promoted MASLD progression, accompanied by decreased Treg viability and activity. Compared with Cd73 KO Tregs, adoptively transferred WT Tregs exhibited increased Treg activity and provided greater protection against hepatic inflammatory responses in MASLD. This immune protection is mediated by CD73 via both enzymatic and nonenzymatic pathways, degrading AMP into ADO to increase Treg function and block DR5-TRAIL-mediated cell death signaling. These findings suggest a potential immunotherapeutic approach for MASLD treatment and highlight its possible relevance for clinical application.
Collapse
Affiliation(s)
- Hua Jin
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xinjie Zhong
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Chunpan Zhang
- Department of Infectious Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yongle Wu
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jie Sun
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiyu Wang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zeyu Wang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jingjing Zhu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuan Jiang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaonan Du
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zihan Zhang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dong Zhang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing 100069, China
| | - Guangyong Sun
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
39
|
Fu Y, Hu P, Hu Y, Fang Y, Zhou Y, Shi Y, Yang K, Fu T, Li W, Gritskevitch ER, Jin L, Lyu J, Zhao Q. Hepatocyte-specific RAP1B deficiency ameliorates high-fat diet-induced obesity and liver inflammation in mice. Diabetes Obes Metab 2025; 27:3036-3049. [PMID: 40083059 DOI: 10.1111/dom.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
AIM This study investigated the role of RAP1B in hepatic lipid metabolism and its implications in obesity and associated metabolic disorders, focusing on the molecular mechanisms through which RAP1B influences lipid accumulation, inflammation and oxidative stress in liver tissues and hepatocyte cell lines. MATERIALS AND METHODS Liver-specific RAP1B-knockout (LKO) and overexpression (OE) mice were generated and fed a high-fat diet for 18 weeks to evaluate systemic and hepatic metabolic changes. Comprehensive metabolic phenotyping included measurements of body weight, body fat content, activity levels, energy expenditure (EE), respiratory exchange ratio (RER), glucose tolerance test and insulin tolerance test. RAP1B-knockdown AML12 hepatocytes were used for in vitro studies. Comprehensive transcriptome and metabolome analyses identified differentially expressed genes and key metabolic shifts. Biochemical and histological analyses were performed to assess lipid accumulation, oxidative stress and inflammatory markers. RESULTS We found that LKO mice exhibited significant reductions in body weight, fat pad size and liver mass, along with decreased hepatic lipid accumulation due to enhanced lipid breakdown. These mice demonstrated improved glucose tolerance and insulin sensitivity without changes in food intake. Liver histology showed reduced F4/80-positive macrophage infiltration, indicating decreased inflammatory cell recruitment. Additionally, markers of oxidative stress were significantly lower, and molecular analysis revealed downregulation of the MAPK(p38) and NF-κB signaling pathways, further supporting an anti-inflammatory hepatic environment. In contrast, OE mice showed increased liver weight, aggravated hepatic lipid accumulation driven by enhanced lipogenesis, worsened insulin resistance and elevated inflammation. CONCLUSIONS This study highlights RAP1B's pivotal role in hepatic metabolism and positions it as a potential therapeutic target for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Yinxu Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
| | - Pingyi Hu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yanyang Hu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yu Fang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yaping Zhou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yu Shi
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiqiang Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ting Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Weijia Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- International Sakharov Environmental Institute, Belarusian State University, Minsk, Republic of Belarus
| | | | - Liqin Jin
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jianxin Lyu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qiongya Zhao
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
40
|
Syanda AM, Georgantaki D, Awsaf M, Molokhia M, Rashid ST. Liver Disease and Prevalence of Liver Transplantation in Adults With ZZ Alpha-1 Antitrypsin Deficiency-A Meta-Analysis. LIVER INTERNATIONAL COMMUNICATIONS 2025; 6:e70013. [PMID: 40248356 PMCID: PMC12001869 DOI: 10.1002/lci2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 04/19/2025]
Abstract
Alpha-1 antitrypsin deficiency (A1ATD) is an inherited metabolic disorder caused by a mutation (ZZ) in the SERPINA1 gene. Carriers are predisposed to liver and lung pathology. The severity of A1ATD-associated liver disease is highly variable, necessitating further characterisation. This study aims to investigate the risk and extent of liver disease and the prevalence of liver transplantation in ZZ A1ATD patients. Several established databases, including Ovid, EBSCO, PubMed, and Cochrane Library, were searched from inception to May 12, 2024. Data were pooled using a random effects model, and study weight was calculated using the inverse variance method. Crude odds ratios (cOR) were calculated using participants with the MM genotype as the comparator. The study was registered in PROSPERO (CRD42022335666). Of the 4420 studies identified, 45 studies and 8638 A1ATD patients (38.8% female) were included. ZZ A1ATD patients demonstrate an increased risk of liver diseases compared to controls, including steatosis (crude odds ratio (cOR): 1.52 [95% CI: 1.21, 1.91]), fibrosis (cOR: 9.85 [95% CI: 5.70, 17.03]), cirrhosis (cOR: 10.43 [95% CI: 5.51, 19.73]), and liver cancers (cOR: 14.12 [95% CI: 6.50, 30.66]). The prevalence of liver transplantation is considerable, with rates reaching 5% [95% CI: 0.00, 12.34]. Our findings confirm the substantial burden of liver disease in ZZ A1ATD patients, including subclinical manifestations such as steatosis and fibrosis that may remain undetected. Given the lack of approved treatments for A1ATD-associated liver disease, prioritising the development of novel therapies to stop or reverse liver disease is essential.
Collapse
Affiliation(s)
- Adam M. Syanda
- Department of Metabolism, Digestion and Reproduction, Faculty of MedicineImperial College LondonLondonUK
- Department of Population Health Sciences, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Dimitra Georgantaki
- Department of Metabolism, Digestion and Reproduction, Faculty of MedicineImperial College LondonLondonUK
| | - Muhammad Awsaf
- Department of Metabolism, Digestion and Reproduction, Faculty of MedicineImperial College LondonLondonUK
| | - Mariam Molokhia
- Department of Population Health Sciences, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - S. Tamir Rashid
- Department of Metabolism, Digestion and Reproduction, Faculty of MedicineImperial College LondonLondonUK
| |
Collapse
|
41
|
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
|
42
|
Choullamy T, Kaadi L, Bezdikian A, Hachem S, Hachem K. Liver Fat Quantification With Ultrasound: The Influence of the Size of the Region of Interest on Attenuation Coefficient. Ultrasound Q 2025; 41:e00712. [PMID: 40173292 DOI: 10.1097/ruq.0000000000000712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
ABSTRACT Noninvasive assessment of liver fat content is crucial due to the high global prevalence of nonalcoholic fatty liver disease. Algorithms based on ultrasound (US) attenuation coefficient (AC) for estimating liver fat content are commercially available, but a lack of consensus exists regarding the best estimation protocol. The aim of our study was to evaluate the influence of the size of the region of interest (ROI) on the US AC.A prospective study was conducted. An abdominal US was done for 86 outpatients. A sampling box was positioned within the liver parenchyma, approximately 2 cm beneath the liver capsule with a ROI, measuring about 2 × 4 cm and then 4 × 5 cm, precisely placed at the center of this sampling box. Five readings of the AC were captured, and the average of these measurements was employed to assess the severity of hepatic steatosisA statistically significant difference between AC with 2 different ROI sizes was shown (P < 0.001) with AC values with 2 × 4 cm ROI were higher than those obtained with 4 × 5 cm ROI (AC mean 0.668 VS 0.653). However, the agreement between AC values obtained with 2 different ROI sizes was excellent (correlation coefficient 0.941)An ROI size dependence is observed in the measurement of AC in the liver. A standardized acquisition protocol with a fixed size of the ROI needs to be developed to minimize differences in AC measurements and to assess changes in serial measurements reliably.
Collapse
Affiliation(s)
- Theresia Choullamy
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| | - Lea Kaadi
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| | - Aren Bezdikian
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | - Samir Hachem
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | - Kamal Hachem
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| |
Collapse
|
43
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
44
|
Zhang H, Feng S, Song S, Zhao Q, Gao Y, Zhang T. First evidence in the association of phenolic endocrine-disrupting chemicals with secondary non-alcoholic fatty liver disease: A case-control study in South China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126086. [PMID: 40118363 DOI: 10.1016/j.envpol.2025.126086] [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: 11/11/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
The presence of phenolic endocrine disrupting chemicals (EDCs) in patients with secondary non-alcoholic fatty liver disease (S-NAFLD) and their associations with S-NAFLD incidence have not been previously documented. In this study, serum concentrations of 32 phenolic EDCs, including parabens, benzophenone-type UV-filters, bisphenols, and bisphenol A diglycidyl ether derivatives, were detected in patients with S-NAFLD as well as healthy population from South China. These target EDCs were ubiquitous in serum samples from both cohorts. Interestingly, significantly higher (p < 0.05) serum levels of most analytes were detected in individuals with S-NAFLD compared to those in the healthy population. Through multiple modeling analyses, we observed that parabens and bisphenols mixtures were positively associated with S-NAFLD incidence. A list of high-risk EDCs for S-NAFLD-related diseases was identified, including propyl paraben (PrP), butyl paraben (BuP), bisphenol A (BPA), and bisphenol AP (BPAP). Furthermore, significant positive correlations were found between the serum levels of these high-risk analytes and liver clinic indices. To the best of our knowledge, this study firstly examined the serum levels of multiple phenolic EDCs in patients with S-NAFLD, aiming to provide novel insights into high-risk EDCs associated with S-NAFLD incidence and their associations with clinic liver indices.
Collapse
Affiliation(s)
- Henglin Zhang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Shuai Feng
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Shiming Song
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Qing Zhao
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yanxia Gao
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Tao Zhang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
45
|
Tian C, Deng S, Zhang Z, Zheng K, Wei L. Bifidobacterium bifidum 1007478 derived indole-3-lactic acid alleviates NASH via an aromatic hydrocarbon receptor-dependent pathway in zebrafish. Life Sci 2025; 369:123557. [PMID: 40074143 DOI: 10.1016/j.lfs.2025.123557] [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: 08/23/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
AIMS This study investigates the potential of Bifidobacterium bifidum 1007478 (BB478) and its metabolite indole-3-lactic acid (ILA) in alleviating non-alcoholic steatohepatitis (NASH) induced by a high-fat diet (HFD) and fructose exposure. MATERIALS AND METHODS A zebrafish model of NASH was established by exposure to HFD and fructose. BB478 was administered, and the effects on liver lipid accumulation, oxidative stress, and inflammation were assessed. ILA production by BB478 was confirmed, and its impact on hepatic lipogenesis and inflammatory pathways was evaluated. The involvement of the aromatic hydrocarbon receptor (AhR) was also examined using an AhR inhibitor. KEY FINDINGS BB478 supplementation inhibited lipid accumulation in the liver, reduced triglycerides (TG) and total cholesterol (TC), and mitigated oxidative stress, as evidenced by lower levels of reactive oxygen species (ROS) and malondialdehyde (MDA). ILA, produced by BB478, could alleviate the hepatic damage and fat deposition in liver. Mechanistically, it suppressed hepatic lipogenesis by downregulating lipogenesis-related genes, including sterol response element binding protein 1 (SREBP1) and fatty acid synthase (FASN). ILA also inhibited the expression of pro-inflammatory cytokines to suppress inflammation. The therapeutic effects of ILA were reversed by the AhR inhibitor, indicating that ILA's actions are AhR-dependent. SIGNIFICANCE These findings reveal the potential of ILA, produced by Bifidobacterium bifidum, as a therapeutic agent for NASH. The mechanistic insights into AhR-mediated effects provide a foundation for further exploration of ILA as a novel approach for managing liver diseases.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China
| | - Zhao Zhang
- Research and Development Centre, GuangDong Longseek Testing Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Kangdi Zheng
- Research and Development Centre, GuangDong Longseek Testing Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
46
|
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
|
47
|
Yan JH, Hao PL, Zeng SM, Liang CX, Chen Y, Chen KW, Qu WQ, Zeng X, Zhong Z, Zhang XZ. Injectable multifunctional hydrogels for adipose tissue remodeling to treat obesity and alleviate metabolic syndrome. J Control Release 2025; 381:113585. [PMID: 40032012 DOI: 10.1016/j.jconrel.2025.113585] [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: 06/24/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Dysfunction of adipose tissue in obese individuals is associated with metabolic imbalances. Current treatments focus on reducing calorie intake and increasing exercise but have unsatisfactory efficacy and significant side effects. Here, a subcutaneously in situ formed therapeutic hydrogel (RL lip@gel) that can simultaneously modulate the inflammatory environment of adipose tissue and induce adipocyte browning is developed to combat obesity. The alginate solution encapsulates the anti-inflammatory natural flavonoid luteolin and rosiglitazone-loaded liposomes (RL lip), which can be formed as an in situ hydrogel through crosslinking with divalent metal ions in adipose tissue following subcutaneous injection. RL lip@gel not only reprograms M1-like adipose tissue macrophages into an M2-like phenotype but diminishes the expression of proinflammatory cytokine, thereby decreasing systemic glucose resistance and lipid storage. Meanwhile, rosiglitazone embedded within the adipose tissue promotes the browning of white adipocytes, and boosts energy expenditure, with further enhancement from luteolin, as demonstrated by decreased white adipose tissue masses. This localized intervention not only ensures the efficacy of combating obesity systemically but enhances overall metabolism while minimizing side effects.
Collapse
Affiliation(s)
- Jian-Hua Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Pei-Ling Hao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Si-Min Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Chun-Xiao Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China
| | - Ke-Wei Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Wen-Qiang Qu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China; Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
48
|
Yang J, Yang Y, Tan X, Du H, Zhou Z, Chen L, Tian X, Zheng G, Hu J, Zhang C, Qiu Z. Unlocking the potential of the ACE2/Ang-(1-7)/Mas Axis in liver diseases: From molecular mechanisms to translational applications. Diabetes Obes Metab 2025. [PMID: 40344459 DOI: 10.1111/dom.16435] [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: 01/25/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Over the past two decades, the identification of new functions within the renin-angiotensin system (RAS) has extended beyond its traditional roles, with the emergence of the angiotensin-converting enzyme 2 (ACE2)/Ang-(1-7)/Mas axis being particularly significant. This axis is hypothesized to balance or modulate the effects of the traditional ACE/Ang II/AT1 axis in various physiological and pathological contexts. ACE2, a membrane-bound carboxypeptidase and an ancient homologue of ACE converts Angiotensin II (Ang II) into Angiotensin 1-7 (Ang-(1-7)). The Mas receptor is a G-protein-coupled receptor that specifically binds Ang-(1-7). Recent research has increasingly focused on the local expression of RAS in different tissues. Ang-(1-7) produces a variety of biological effects by binding to the Mas receptor, including anti-inflammatory, antioxidant, anti-apoptotic and anti-fibrotic actions, thereby influencing a range of mechanisms in the heart, kidneys, brain and other tissues. Preclinical animal model studies indicate that manipulating the protective RAS can significantly alter the progression of multiple liver diseases. Hepatic overexpression of ACE2 or administration of Ang-(1-7) and its analogues has been shown to be therapeutically effective against drug-induced liver injury, metabolic-associated fatty liver disease, liver fibrosis and hepatocellular carcinoma progression. These effects are achieved through various pathways, including the regulation of lipid metabolism, inhibition of epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) production, as well as suppression of aerobic glycolysis. In current clinical trials, while recombinant human ACE2 (Rh-ACE2) has demonstrated safety and good tolerance in most studies, research on the relevance of activating the ACE2/Ang-(1-7) axis in the mechanisms and evolution of human diseases remains in its early stages. Therefore, further elucidation of the complex interactions between the classical and counter-regulatory RAS axes in clinical settings is crucial. This review will summarize the roles of selective activation of the ACE2/Ang-(1-7)/Mas axis, with a focus on its mechanisms in the treatment of liver diseases. Additionally, we will discuss the safety concerns regarding selective activation of the ACE2/Ang-(1-7)/Mas axis in clinical applications and the challenges of tissue-specific activation of this axis, providing effective therapeutic strategies for targeted activation of the hepatic ACE2/Ang-(1-7)/Mas axis in clinical practice.
Collapse
Affiliation(s)
- Jun Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Xianxiang Tian
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Guohua Zheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Shizhen Laboratory, Wuhan, People's Republic of China
| |
Collapse
|
49
|
Colussi C, Baillargeon JP, Ngueta G. Examining the relationship between persistent organic pollutants and metabolic-associated fatty liver disease: a cross-sectional analysis using NHANES data. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025:10.1007/s11356-025-36493-2. [PMID: 40342089 DOI: 10.1007/s11356-025-36493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Recently, the term Metabolic-Associated Fatty Liver Disease (MAFLD) has been adopted to better reflect the underlying pathology and association with metabolic issues. Beyond dietary factors and physical activity, previous studies have suggested that persistent organic pollutants (POPs) may contribute to the etiology of MAFLD; however, this disease can also develop at very low POP exposure levels, making it challenging to discern their specific effect. This study aims to investigate the potential link between exposure to POPs and the prevalence of MAFLD. Data from the National Health and Nutrition Examination Survey (NHANES) was utilized for this cross-sectional study. Participants were categorized based on their MAFLD status and levels of various POPs measured in their blood serum. Cox regression to estimate adjusted prevalence ratio (aPR) of MAFLD was used. Hazard Index (HI), Proportion of Maximum Scaling (POMS), and Toxicity Burden Index (TBI) were applied to assess exposure to mixtures. A total of 4,224 participants were included, 47 (33-65) years, 53.0% were women, and 50.1% had MAFLD. No significant sex differences were observed in the main analysis regarding the association between individual POPs and MAFLD prevalence. However, sensitivity analyses revealed an inverse relationship between certain POPs and MAFLD prevalence, particularly in women. Higher levels of specific PCBs were associated with a lower prevalence of MAFLD in women. This study highlighted the effects of individual pollutants, mixtures, and sex-specific differences. The combined use of HI, POMS, and TBI provided a more detailed risk assessment. Findings suggest that biological sex and metabolic stressors play significant roles in how POPs influence MAFLD, warranting further investigation into mechanisms and health outcomes in different exposure ranges.
Collapse
Affiliation(s)
- Carlina Colussi
- Université de Sherbrooke, Faculté de Médecine Et Des Sciences de La Sante, Sherbrooke, Canada.
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada.
- CHU de Sherbrooke Research Center, DOCC (Diabetes, Obesity and Cardiovascular Complications), Endocrinology, CRCHUS - Hôpital Fleurimont, 3001, 12eme Avenue Nord, Sherbrooke, Canada.
| | - Jean-Patrice Baillargeon
- Université de Sherbrooke, Faculté de Médecine Et Des Sciences de La Sante, Sherbrooke, Canada
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
- CHU de Sherbrooke Research Center, DOCC (Diabetes, Obesity and Cardiovascular Complications), Endocrinology, CRCHUS - Hôpital Fleurimont, 3001, 12eme Avenue Nord, Sherbrooke, Canada
| | - Gérard Ngueta
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
- Department of Community Health Sciences, Faculty of Medicine & Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
50
|
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
|