1
|
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
|
2
|
Franczak E, Kugler BA, Salathe SF, Allen JA, Sardiu ME, McCoin CS, Hevener AL, Morris EM, Thyfault JP. Loss of ovarian function prevents exercise-induced activation of hepatic mitophagic flux. Am J Physiol Endocrinol Metab 2025; 328:E869-E884. [PMID: 40293097 DOI: 10.1152/ajpendo.00107.2025] [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: 03/10/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Exercise effectively treats metabolic dysfunction-associated steatotic liver disease (MASLD) by enhancing hepatic mitochondria energy metabolism. However, the efficiency of exercise in treating MASLD in postmenopausal women may be reduced. Previously, we showed acute treadmill exercise activates hepatic mitophagy, the selective degradation of low-functioning mitochondria. Mitophagic flux is differentially regulated in female mice compared with males, possibly by estrogen. Here, we tested if loss of ovarian function via ovariectomy (OVX), which reduces estrogen, drives MASLD, and compromised hepatic mitochondrial energetics, would blunt activation of hepatic mitophagy induced by exercise. Following OVX, 12- to 15-wk-old female mice were placed on a low-fat diet (LFD) or high-fat diet (HFD) for 4 wk to induce MASLD, after which half of the mice performed a single acute bout of treadmill exercise to exhaustion or remained sedentary. Two hours post exercise, isolated hepatic mitochondria were examined via Western blotting and proteomics for accumulation of known mitophagy proteins. After exercise, reduced basal mitophagic flux in LFD-fed OVX was restored to levels found in sham mice. However, exercise possessed blunted capacity to promote mitochondrial recruitment of DRP1 (regulator of fission) and accumulation mitophagy-associated proteins (E3-ubiquitin ligase, ubiquitin, autophagy adaptor proteins, and autophagosome cargo receptors) in OVX versus sham mice on HFD. Mitochondrial H2O2 production, which putatively activates mitophagy, was elevated following exercise in all conditions except OVX + HFD. In summary, OVX reduces mitophagic flux, blunting the stimulatory effects of exercise on these factors. The impaired regulation of mitophagy following the cessation of ovarian function likely contributes to the pathogenesis of MASLD post menopause.NEW & NOTEWORTHY Loss of ovarian function reduces hepatic mitochondrial respiratory capacity, but mechanisms are unknown. Here, we leverage exercise-induced hepatic mitophagy activation to determine if loss of ovarian function impairs mitochondrial quality control mechanisms. Our data reveal that loss of ovarian function reduces both ubiquitin-mediated hepatic mitophagy and mitochondrial recruitment of Drp1 (mitochondrial fission protein) following acute exercise. These impairments to hepatic mitophagy coincided with alterations in hepatic mitochondrial respiratory capacity and mitochondrial-derived H2O2 production.
Collapse
Affiliation(s)
- Edziu Franczak
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Benjamin A Kugler
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Sebastian F Salathe
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Julie A Allen
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Mihaela E Sardiu
- Department of Biostatistics and Data Science, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, California, United States
- Department of Medicine, VA Greater Los Angeles Healthcare System GRECC, Los Angeles, California, United States
| | - E Matthew Morris
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
- KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
3
|
Wang X, Leng S. Serum uric acid may be a mediator of risk factors in metabolic dysfunction associated steatotic liver disease. Scand J Gastroenterol 2025; 60:581-587. [PMID: 40255082 DOI: 10.1080/00365521.2025.2490994] [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: 08/16/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND In recent years, we have witnessed a sharp growth of metabolic dysfunction associated steatotic liver disease (MASLD). How to achieve early prevention of MASLD is imminent. We aimed to determine the dietary pattern and other factors influencing MASLD, to explore whether it mediated by serum uric acid (SUA) or not. METHODS A total of 4,038 adults attending the Health Management Center of the Second Affiliated Hospital of Dalian Medical University between October 2018 and May 2019 were surveyed using a questionnaire and underwent physical measurements. Structural equation model (SEM) was used to verify the risk factors and determine the path affecting MASLD. RESULTS A total of 3,589 participants were studied. The standardized prevalence of MASLD was 47.8%. Three dietary patterns were identified using factor analysis. The SEM showed that SUA was positively associated with MASLD (standardization coefficient 0.285). In rank order, sex, SUA, dyslipidemia, age, a high-protein diet, and a fast-food diet were risk factors for MASLD. Sex, dyslipidemia, and a fast-food diet had positive and indirect effects on NAFLD through SUA, while age and a high-protein diet had negative and indirect effects on MAFLD through SUA. CONCLUSIONS SUA may be an important mediator of the risk of MASLD. People with abnormal SUA, especially men, should limit their intake of fried and cured foods and desserts; have their blood lipid profiles monitored carefully; appropriately increase their consumption of high-quality protein sources, such as eggs, milk, and beans; and be aware of their MASLD risk.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Disease Prevention and Hospital Infection Control, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Song Leng
- Health Management Center, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Zhang X, Nguyen MH. Metabolic dysfunction-associated steatotic liver disease: A sexually dimorphic disease and breast and gynecological cancer. Metabolism 2025; 167:156190. [PMID: 40081614 DOI: 10.1016/j.metabol.2025.156190] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become a global public health and economic burden worldwide in the past few decades. Epidemiological studies have shown that MASLD is a multisystem disease that is associated not only with liver-related complications but also with an increased risk of developing extrahepatic cancers. MASLD is a sexually dimorphic disease with sex hormones playing an important role in the development and progression of MASLD, especially by the levels and ratios of circulating estrogens and androgens. MASLD is associated with hormone-sensitive cancers including breast and gynecological cancer. The risk of breast and gynecological cancer is elevated in individuals with MASLD driven by shared metabolic risk factors including obesity and insulin resistance. Multiple potential mechanisms underline these associations including metabolic dysfunction, gut dysbiosis, chronic inflammation and dysregulated release of hepatokines. However, the effect of hormone therapy including hormone replacement therapy and anti-estrogen treatment on MASLD and female-specific cancers remains debatable at this time. This synopsis will review the associations between MASLD and breast and gynecological cancer, their underlying mechanisms, implications of hormonal therapies, and their future directions.
Collapse
Affiliation(s)
- Xinrong Zhang
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States; Department of Epidemiology and Population Health, Stanford University Medical Center, Palo Alto, CA, United States; Stanford Cancer Institute, Stanford University Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
5
|
Wang C, Ma H, Yang H, Nie Q, Zhu L, Yin J, Zhou L. Sex differences in the association between total energy intake and all-cause mortality among patients with metabolic dysfunction-associated steatotic liver disease. Sci Rep 2025; 15:19176. [PMID: 40450061 DOI: 10.1038/s41598-025-04121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 05/26/2025] [Indexed: 06/03/2025] Open
Abstract
The long-term association between total energy intake and clinical outcomes in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) remains unclear. The present study aimed to explore the associations between total energy intake and mortality in MASLD patients and to evaluate whether the associations differ by sex. A total of 2,466 MASLD patients from the Third National Health and Nutrition Examination Survey was included. Total energy intake was assessed using 24-hour dietary recall. Mortality was ascertained by linkage to National Death Index records through 31 December 2019. Multivariable Cox proportional hazards model was used to estimate the association between total energy intake and mortality. In MASLD patients, both low and high total energy intake were significantly associated with elevated risk of all-cause mortality. Compared with moderate total energy intake (2,000-3,000 kcal/day for males and 1,600-2,400 kcal/day for females), the HRs (95% CIs) for low and high total energy intake were 1.27 (1.05-1.53; P = 0.01) and 1.40 (1.03-1.92; P = 0.03), respectively. A significant interaction was demonstrated between sex and total energy intake for all-cause mortality (P value for interaction = 0.03). In males, both low and high total energy intake were significantly associated with elevated risk of all-cause mortality. The HRs (95% CIs) for low and high total energy intake were 1.35 (1.02-1.80; P = 0.04) and 1.54 (1.05-2.28; P = 0.03), respectively. However, no significant association was observed in females. The HRs (95% CIs) for low and high total energy intake were 1.14 (0.86-1.52; P = 0.34) and 1.14 (0.81-1.61; P = 0.46), respectively. These findings provide novel evidence supporting sex-specific dietary guidelines for MASLD, with particular emphasis on maintaining moderate energy intake to mitigate mortality risk in male patients.
Collapse
Affiliation(s)
- Changsi Wang
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Hongfei Ma
- Wuhan Centers for Disease Prevention and Control, Wuhan, China
| | - Hongfang Yang
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Qi Nie
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Lin Zhu
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Jiawei Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Nutrition Innovation Platform-Sichuan and Chongqing, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Li Zhou
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China.
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Chen L, Sui G, Wu J, Li N, Zhang Z, Du Y, Lü M, Yan X, Pan G, Jia L. Untargeted metabolomics and transcriptomics joint analysis of the effects of polystyrene nanoplastics on lipid metabolism in the mouse liver. Lipids Health Dis 2025; 24:195. [PMID: 40442648 PMCID: PMC12121128 DOI: 10.1186/s12944-025-02613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Micro/nanoplastics (MNPs), as emerging environmental pollutants, are widely present in environments that are essential for human survival. They exist in vast quantities and possess stable properties, making them challenging to manage. Some reports indicated that there is a positive correlation between the production of MNPs and the incidence of obesity. The liver serves as both the central hub for lipid metabolism and a prime target for MNPs toxicity. These studies revealed that MNPs can lead to increased hepatic lipid accumulation, suggesting that they may be potential obesogens. However, the specific metabolic changes and possible mechanisms involved remain to be elucidated. METHODS This study focuses on the impact of nanoplastics (NPs) on liver lipid metabolism, using C57BL/6J mice (hereinafter referred to as C57 mice) as the research subjects, and exposing them to 100 nm NPs at 1000 µg/L continuously for 12 weeks. RESULTS The study revealed that (1) NPs led to nondietary weight gain together with an increase in fat volume and mass in mice. (2) NPs significantly increased serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels, with notable differences between groups. Notably, NPs exposure induced opposing effects on serum lipid profiles, elevating high-density lipoprotein cholesterol (HDL-C) concentrations while suppressing triglyceride (TG) levels, though intergroup differences failed to reach statistical significance. (3) NPs caused multiple inflammatory responses in the liver, with significant lipid deposition. (4) Untargeted metabolomics analysis indicated that NPs exposure led to significant alterations in various lipid metabolites, particularly glycerophospholipids. Additionally, transcriptomics reveals that differentially expressed genes (DEGs) triggered by NPs exposure are predominantly involved in metabolic routes including lipid metabolism and cytochrome P450 (CYP). Taken together, these findings suggested that alterations in lipid metabolism resulting from NPs exposure may involve arachidonic acid metabolism. Phosphatidylcholine (PC) could be the key substance, and the CYP gene family (Cyp2c23, Cyp2c40) might be the critical genes regulating liver lipid metabolism during NPs exposure. CONCLUSIONS This study has demonstrated that NPs exposure induced obesity and hepatic lipid accumulation in male mice independently of food intake. The integrated omics data identified dysregulated PC metabolism and CYP gene family expression, suggesting their involvement in arachidonic acid-associated pathways. These findings provided preliminary mechanistic clues linking NP exposure to hepatic lipid metabolism dysregulation and helped to elucidate the adverse effects of NPs on liver lipid metabolism.
Collapse
Affiliation(s)
- Lijuan Chen
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Guoyuan Sui
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Jin Wu
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Ning Li
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Zhe Zhang
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Ying Du
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Meijun Lü
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Xiaorui Yan
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| | - Guowei Pan
- China Medical University, Shenyang, Liaoning, 110122, China.
| | - Lianqun Jia
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China.
| |
Collapse
|
7
|
Yu H, Wang D, Yan Y, Jiao L, Zhang J, Gu Y, Zhang S, Zhang Q, Liu L, Meng G, Wu H, Wu X, Zhu D, Fu L, Chen Y, Wang D, Wang Y, Geng H, Sun S, Wang X, Jia Q, Song K, Zheng Y, Yu M, Chen YM, Niu K. Dietary manganese intake is positively associated with metabolic dysfunction-associated steatotic liver disease: a multicohort study. Eur J Nutr 2025; 64:188. [PMID: 40419737 DOI: 10.1007/s00394-025-03708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/30/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND AND AIMS Manganese (Mn) is an essential nutrient that plays a crucial role in maintaining normal physiological functions of the human body. However, overexposure to Mn often leads to adverse health outcomes and contributes to the development of a variety of diseases. Several studies have explored the relationship between dietary Mn and metabolic dysfunction-associated steatotic liver disease (MASLD) risk. Two UK Biobank (UKB)-based studies suggested that Mn, as a key nutrient, may be associated with a reduced risk of MASLD. Another study found an association between the dietary antioxidant index and the onset of non-alcoholic fatty liver disease (NAFLD), highlighting the importance of nutritional factors (including Mn) in liver health. However, the relationship between dietary Mn intake and MASLD in the Chinese population remains unexplored, and further research is needed to elucidate its underlying mechanisms. METHODS This prospective multi-cohort study had 1,137 adults from the Guangzhou Nutrition and Health Study (GNHS) cohort and 17,649 people from the Tianjin Chronic Low-grade Systemic Inflammation and Health (TCLSIH) cohort. We measured dietary intake using a validated and standardized food frequency questionnaire. Annual abdominal ultrasound was used to diagnose new-onset MASLD. We used multivariable Cox regression models to assess the relationship between dietary Mn intake and the risk of MASLD. RESULTS In the TCLSIH cohort, 3640 MASLD cases were observed with a follow-up time of 60,190 person-years. After taking into account possible confounding factors, the multivariable HRs (95% CIs) for MASLD across the quartiles of dietary Mn intake in males were 1.00 (reference), 1.08 (0.96-1.21), 1.12 (0.99-1.26), and 1.16 (1.02-1.31), with a P for trend = 0.02; for females, the HRs (95% CIs) for MASLD across the quartiles of dietary Mn intake were 1.00, 1.11 (0.95-1.31), 1.08 (0.91-1.28), and 0.97 (0.81-1.16), with a P for trend = 0.58. After adjustment for proteins, lipids, and carbohydrates, the HRs (95% CIs) for MASLD across the quartiles of dietary Mn intake in males were 1.00 (reference), 1.11 (0.97-1.26), 1.14 (1.00-1.31), and 1.16 (1.00-1.34), with a P for trend = 0.045. For females, the HRs (95% CIs) for MASLD across the quartiles of dietary Mn intake were 1.00 (reference), 1.08 (0.91-1.30), 1.06 (0.88-1.27), and 0.93 (0.76-1.13), with a P for trend = 0.39. In the GNHS cohort, 624 MASLD cases were observed with a follow-up time of 6454 person-years. After adjusting for relevant confounders, the HRs (95% CIs) for males comparing T3 versus T1 of dietary Mn intake were 1.04 (0.65-1.60); the HRs (95% CIs) for females comparing T3 versus T1 of dietary Mn intake were 1.00 (0.78-1.29). CONCLUSION In males, higher dietary Mn intake is associated with a higher incidence of MASLD.
Collapse
Affiliation(s)
- Hao Yu
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Di Wang
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Lirui Jiao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Jinjin Zhang
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Yeqing Gu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Baidi Road 238, Tianjin, 300192, China.
| | - Shunming Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Qing Zhang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Ge Meng
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Hongmei Wu
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Xuehui Wu
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Dandan Zhu
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Liyuan Fu
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yinxiao Chen
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Dongli Wang
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Yaxiao Wang
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Hao Geng
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
- Tianjin Health Management and Promotion Institute, Tianjin, China
| | - Shaomei Sun
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing Wang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiyu Jia
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kun Song
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunliang Zheng
- Tianjin Institute of Modern Health Technology, Tianjin, China
| | - Ming Yu
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Yu-Ming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Kaijun Niu
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
- School of Public Health, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
- Tianjin Health Management and Promotion Institute, Tianjin, China.
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
8
|
Yang Q, Zhang Y, Zhang H, Yang Z, Feng Y, Ye B, Gong P, Qian G, Li D. Advanced N-glycoproteomics and proteomics approach revealed sexually dimorphic molecular signatures in primary mouse hepatocyte. Anal Bioanal Chem 2025:10.1007/s00216-025-05912-1. [PMID: 40410350 DOI: 10.1007/s00216-025-05912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Sexual dimorphism plays a critical role in disease pathophysiology, but the subtlety and complexity of these differences, along with a lack of precise comparative methods, hinder the advancement of precision medicine and drug development. This limitation is particularly evident in metabolic dysfunction-associated steatotic liver disease (MASLD), where sex-specific molecular mechanisms remain insufficiently understood. To address this gap, we employed an advanced integrative N-glycoproteomics and proteomics approach to systematically analyze sex-biased molecular signatures in primary mouse hepatocytes (PMHs) under healthy and MASLD conditions. Our analysis identified 280 sex-biased proteins and 39 sex-biased N-glycosites, and KEGG enrichment revealed that female-biased molecules were primarily involved in lipid metabolism, while male-biased molecules were associated with inflammation and cytoskeletal remodeling. A combined dataset of 302 sex-biased molecules was further analyzed using protein-protein interaction (PPI) analysis and Rc value calculations, resulting in the identification of 21 hub proteins and 2 hub N-glycosites as MASLD-associated sex-biased signatures. Notably, MASLD amplified proteomic sex differences while attenuating them in N-glycosylation. Western blot validation of key signatures, including female-biased MVK and male-biased LGALS3, highlighted distinct molecular adaptations between the sexes in MASLD progression. Our study introduced an advanced analytical framework for high-resolution comparative molecular profiling by integrating N-glycoproteomics with proteomics, providing valuable insights into sex-biased molecular signatures, enhancing preclinical model development, and advancing sex-specific therapeutic strategies in MASLD research and broader biological systems.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yong Zhang
- Department of Nephrology, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanruyu Feng
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Ninth People's Hospital of Zhengzhou, Zhengzhou, 45000, China
| | - Bengui Ye
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Medical College of Tibet University, Lhasa, 850002, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, China
| | - Guangsheng Qian
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Dapeng Li
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Zhang M, Yuan Y, Wang C, Huang Y, Fan M, Li X, Qin Z. Aggregate index of systemic inflammation tied to increased fatty liver disease risk: insights from NHANES data. BMC Gastroenterol 2025; 25:399. [PMID: 40410700 PMCID: PMC12101034 DOI: 10.1186/s12876-025-03998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Fatty liver disease (FLD), characterized by hepatic lipid accumulation, impairs quality of life and can progress to cirrhosis and hepatocellular carcinoma, imposing a healthcare burden. This study investigates the association between the aggregate index of systemic inflammation (AISI) and FLD prevalence, evaluating AISI's potential as an early biomarker for risk assessment. METHODS Data were obtained from the National Health and Nutrition Examination Survey (NHANES) database, which encompasses the years 2017 through 2020. Participants were chosen based on the availability of controlled attenuation parameter (CAP) scores derived from transient elastography (TE), a technique utilized for assessing liver steatosis. The formula employed to compute the AISI is as follows: AISI = N × P × M / L, where N, P, M, and L refer to neutrophils, platelets, monocytes, and lymphocytes, respectively. Additionally, demographic, socioeconomic, dietary, and health-related information was gathered. Logistic regression models were utilized to pinpoint risk factors associated with FLD, and a nomogram was created to forecast FLD risk. RESULTS Of the 3,961 participants, 2,377 (60.0%) were diagnosed with FLD based on a CAP score ≥ 248 dB/m. Elevated AISI was significantly associated with FLD (P = 0.021). Other significant risk factors included sex, age, BMI, race, marital status, hypertension, and diabetes. The nomogram demonstrated excellent discriminatory performance with an AUC of 0.814 (95% CI: 0.800, 0.827) and good calibration. CONCLUSION This study reveals a significant, independent association between elevated AISI and increased FLD risk in the U.S. population, even after adjusting for confounders. AISI demonstrated good discriminative performance for FLD, but its effect size suggests it should supplement, not replace, existing clinical risk assessment tools. AISI, a cost-effective biomarker, holds potential for enhancing FLD screening, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Meng Zhang
- Guangxi International Zhuang Medicine Hospital (Affiliated International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine), Nanning, 530200, China
| | - Yuan Yuan
- School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Chenglong Wang
- Guangxi International Zhuang Medicine Hospital (Affiliated International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine), Nanning, 530200, China
| | - You Huang
- Institute of Chinese Medicine, Zhuang and Yao Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Mingli Fan
- Institute of Chinese Medicine, Zhuang and Yao Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Xiangling Li
- Institute of Chinese Medicine, Zhuang and Yao Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China.
| | - Zujie Qin
- Guangxi International Zhuang Medicine Hospital (Affiliated International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine), Nanning, 530200, China.
| |
Collapse
|
10
|
Luo X, Wang K, Jiang C. Gut microbial enzymes and metabolic dysfunction-associated steatohepatitis: Function, mechanism, and therapeutic prospects. Cell Host Microbe 2025:S1931-3128(25)00153-2. [PMID: 40425014 DOI: 10.1016/j.chom.2025.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent liver disease worldwide. The liver communicates with the intestine, in large part through the gut microbiota. Microbial enzymes are key mediators that affect the progression of MASLD and the more severe metabolic dysfunction-associated steatohepatitis (MASH). These enzymes contribute to the metabolism or biosynthesis of steroids, fatty acids, amino acids, ethanol, choline, and intestinal hormones that contribute to disease progression. Additionally, dysbiosis and functional alterations in the microbiota compromise the intestinal barrier, increasing its permeability to bacterial metabolites and liver exposure to microbial-associated molecular patterns (MAMPs), thereby exacerbating liver inflammation and fibrosis. Furthermore, functional alterations in the gut microbiota can modulate intestinal signaling pathways through metabolites or gut hormones, subsequently affecting hepatic metabolism. A deeper understanding of the roles of the gut microbiota and microbial enzymes in MASH will facilitate the development of personalized treatments targeting specific gut microbes or functional enzymes.
Collapse
Affiliation(s)
- Xi Luo
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
11
|
Huynh J, Hoque AR, Reddy SSK. "Diagnosis Frequency and Associated Factors of Non-Alcoholic Fatty Liver Disease among U.S Hospitalized Adults in Urban vs. Rural populations from 2007 - 2019: An Emerging Public Health Crisis". Endocr Pract 2025:S1530-891X(25)00891-2. [PMID: 40409609 DOI: 10.1016/j.eprac.2025.05.740] [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] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVES To describe and understand differences between U.S. rural and urban populations with respect to outcomes of hospitalization and related epidemiology of Non-alcoholic fatty liver disease (NAFLD). METHODS We analyzed data from the Healthcare Cost and Utilization Project (HCUP) National Inpatient Sample (NIS) from 2007 to 2019, identifying 847,165 NAFLD cases, of which 370,131 met inclusion criteria. Statistical analyses included Pearson's chi-square, independent samples t-tests, Mann-Whitney U tests, and multivariate logistic regression models to examine factors associated with NAFLD. RESULTS Hospitalizations due to NAFLD significantly increased over time from 2007 - 2019 with urban cases constituting 84.9% while rural cases represented 15.1%. Differences in demographics, hospital characteristics, insurance, income, and outcomes were significant between the two groups. Multivariate analysis showed higher odds of NAFLD diagnosis in fringe metro areas (adjusted odds ratio [a.O.R.]=1.074, 95% CI=1.044-1.105), medium metro counties ((a.O. R.=1.146, 95% CI=1.114-1.179), small metro counties (a.O. R.=1.182, 95% CI=1.140-1.226), and rural regions (a.O. R.=1.279, 95% CI=1.233-1.327) compared to central metro areas. NAFLD was more prevalent in females, those aged 35-49 or 50-64 years, and White patients, particularly among those with diabetes, metabolic syndrome, and obesity. CONCLUSION The increasing prevalence of NAFLD suggests a strong association with metabolic and cardiovascular diseases. With increasing closure of rural hospitals, we may see more rural patients with NAFLD admitted to urban centers. Early detection and diagnosis should help prevent long-term complications of NAFLD.
Collapse
Affiliation(s)
- Judy Huynh
- Central Michigan University College of Medicine, 1280 S. East Campus Drive Mount Pleasant, MI 48859.
| | - Asef Raiyan Hoque
- Central Michigan University College of Medicine, 1280 S. East Campus Drive Mount Pleasant, MI 48859.
| | - S Sethu K Reddy
- Central Michigan University College of Medicine, 1280 S. East Campus Drive Mount Pleasant, MI 48859.
| |
Collapse
|
12
|
Wegermann K, Chouairi F, Karachaliou GS, Ahlers C, Au S, Miller K, Biering-Sørensen T, Abdelmalek MF, Diehl AM, Moylan CA, Fudim M. Incident heart failure is common and underrecognized in patients with biopsy-proven metabolic dysfunction-associated steatotic liver disease. Eur J Heart Fail 2025. [PMID: 40389356 DOI: 10.1002/ejhf.3697] [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: 12/12/2024] [Revised: 04/02/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is associated with heart failure (HF), independent of shared risk factors. Our aim was to describe the incidence of HF in patients with biopsy-proven MASLD. METHODS AND RESULTS We followed patients with biopsy-proven MASLD from the prospective Duke NAFLD Biorepository and Clinical Database from liver biopsy (2007-2013) until death or 5 January 2023. Clinical and echocardiographic data were abstracted via manual chart review. Incident HF was defined as one of the following: (1) hospitalization for HF, (2) medical record diagnosis of HF, (3) ≥1 sign/symptom of HF and elevated natriuretic peptide, or (4) diastolic dysfunction on transthoracic echocardiography with ≥1 sign/symptom of HF. Univariable and multivariable logistic regression models were evaluated. Overall, 570 patients with biopsy-proven MASLD were included. The mean age was 49.5 years, 42.5% were male and 87.0% were non-Hispanic White. Ten patients (1.8%) had baseline HF, leaving 560 patients to assess for incident HF. Over a median follow up of 4009 days (11.0 years) (interquartile range 2270-4672 days), 100 (17.9%) patients developed incident HF while 268 (47.9%) met criteria for HF suspicion. In a multivariable model, increasing age (odds ratio [OR] 1.05, 95% confidence interval [CI] 1.02-1.08, p < 0.001) and female sex (OR 1.85, 95% CI 1.12-3.04, p = 0.02) were associated with incident HF. CONCLUSIONS We found a high incidence of HF in patients with biopsy-proven MASLD. Despite nearly half of patients having suspected HF, very few carried a chart diagnosis. Screening for HF in high-risk patients and establishment of formal care pathways to address early HF may reduce morbidity and mortality.
Collapse
Affiliation(s)
- Kara Wegermann
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Fouad Chouairi
- Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Georgia Sofia Karachaliou
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Carolyn Ahlers
- Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Sandra Au
- Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Kaela Miller
- Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Cynthia A Moylan
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
- Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Marat Fudim
- Division of Cardiology, Department of Medicine, Duke University Health System, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
13
|
Gao X, Chen T, Zhou F, Sun Y, Zhang J, Li X, Zhao W, Li Y, Shi Y, Niu K, Wang Y, Zhang Y, Zhang W. The association between different insulin resistance surrogates and all-cause mortality and cardiovascular mortality in patients with metabolic dysfunction-associated steatotic liver disease. Cardiovasc Diabetol 2025; 24:200. [PMID: 40346671 PMCID: PMC12065324 DOI: 10.1186/s12933-025-02758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/26/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is closely associated with insulin resistance (IR). However, the prognostic value of different alternative IR surrogates in patients with MASLD remains unclear. This study aimed to evaluate the association between various IR indices and all-cause mortality and cardiovascular mortality in MASLD patients. METHODS A total of 8,753 adults aged ≥ 20 years with MASLD from the National Health and Nutrition Examination Survey (NHANES, 2003-2018) were included, and their mortality data were obtained from the National Death Index (NDI). Insulin resistance surrogates [including the triglyceride-glucose (TyG) index, TyG-body mass index (TyG-BMI), TyG-waist circumference index, TyG-waist-to-height ratio index, and Homeostatic Model Assessment for IR] were stratified into quartiles. Cox proportional hazards models, receiver operating characteristic (ROC) curve analysis, restricted cubic spline (RCS), mediation analyses, and subgroup analyses were used to explore the associations between these indices and all-cause mortality as well as cardiovascular mortality in MASLD patients. RESULTS During a median follow-up of 98 months, 1,234 deaths were observed, including 409 cardiovascular disease (CVD)-related deaths. In the fully adjusted model, higher quartiles of TyG-related indices were significantly associated with an increased risk of all-cause mortality in MASLD patients. Furthermore, the TyG-BMI index was associated with both all-cause mortality and CVD mortality [all-cause mortality: HR (95% CI) 2.84 (1.73-4.67), P < 0.001; CVD mortality: HR (95% CI) 5.32 (2.26-12.49), P < 0.001]. The RCS analyses indicated a U-shaped relationship between TyG-BMI and mortality, with a threshold value of 270.49. Subgroup analyses demonstrated that TyG-related indices had stronger associations with mortality in elderly MASLD patients. CONCLUSIONS Our findings highlight the prognostic value of IR indices, particularly TyG-BMI index, in predicting all-cause mortality and CVD mortality in MASLD patients.
Collapse
Affiliation(s)
- Xin Gao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Tianyi Chen
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Feilong Zhou
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yanmei Sun
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Jiaqi Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Weijie Zhao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yunxin Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yanlong Shi
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Kaiyi Niu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yizhu Wang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Jiangjiayuan 121, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shangdong University, Jinan, Shandong Province, China.
| | - Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
14
|
Yang J, Zhou Z. Analysis and study of risk factors related to the progression of non-alcoholic fatty liver disease: A retrospective cohort study. PLoS One 2025; 20:e0322990. [PMID: 40333922 PMCID: PMC12057958 DOI: 10.1371/journal.pone.0322990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is becoming increasingly prevalent worldwide. This study aimed to analyze the risk factors associated with NAFLD progression by collecting and evaluating clinical data of NAFLD patients, providing a scientific basis for its prevention and treatment. METHODS Clinical data of NAFLD patients from June 2015 to June 2016 were retrospectively collected, including gender, age, alanine aminotransferase (ALT), aspartate aminotransferase(AST), alkaline phosphatase(ALP),γ-glutamyltranspeptidase (GGT), triglycerides (TG), total cholesterol (TC), high-density lipoprotein (HDL), low-density lipoprotein (LDL), fasting blood glucose (FBG), and visceral fat area (VFA). All patients were stratified by gender and age, and logistic regression analysis was used to explore the risk factors for NAFLD disease progression. RESULTS ALT, TG, FBG, and VFA were identified as independent risk factors for NAFLD progression. Stratified analysis showed that in male patients, ALT, TG, and VFA were independent risk factors, whereas in female patients, TG, FBG, and VFA were identified as independent risk factors. Age-stratified analysis revealed that ALT, TG, and VFA were significant risk factors for progression in young and middle-aged patients. At the same time, age, ALT, TG, and FBG were substantial in elderly patients. CONCLUSION Different risk factors should be closely monitored in sex- and age-specific populations to prevent NAFLD progression effectively.
Collapse
Affiliation(s)
- JunRan Yang
- Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Zhenhua Zhou
- Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| |
Collapse
|
15
|
Abbaszadeh M, Hosseinpanah F, Tohidi M, Karimpour Reyhan S, Mahdavi M, Valizadeh M. Sex-Specific Impact of Metabolic Dysfunction-Associated Fatty Liver Disease on Incident Cardiovascular Diseases and Mortality. Endocrinol Diabetes Metab 2025; 8:e70035. [PMID: 40140729 PMCID: PMC11946537 DOI: 10.1002/edm2.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/30/2024] [Accepted: 02/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND AND AIMS Considering recent revisions in the nomenclature for fatty liver disease, alongside limited data on sex-specific differences in its cardiovascular/mortality outcomes, this study aims to investigate the prevalence and impact of metabolic-associated fatty liver disease (MAFLD) on cardiovascular disease (CVD) and mortality in men and women over a 12-year follow-up period. METHODS In this large population-based cohort study, 7101 individuals aged ≥ 30 were enrolled. The prevalence of MAFLD was investigated in both genders. After excluding individuals with a history of previous CVD, 6331 participants were followed up for CVD and mortality over 12 years. Steatosis was defined as fatty liver index (FLI) ≥ 60. Multivariate-adjusted hazard ratios (HRs) were calculated for CVD and mortality. RESULTS The prevalence of MAFLD was 43.2%, higher in men (46.5%) than women (40.6%). Men with MAFLD (47.7 ± 12.1) were younger than women (52.2 ± 11.1). In the 12-year follow-up of 6331 individuals, multivariable-adjusted CVD HRs for MAFLD were 1.36 (1.10-1.67) in men and 1.48 (1.16-1.88) in women. Adjusted mortality HRs were 1.17 (0.86-1.59) and 1.38 (1.00-1.91) in men and women, respectively. Among patients with MAFLD, a subgroup with diabetes faced the highest hazard for CVD and mortality. CONCLUSION This study found that MAFLD is more common in men at a younger age. Despite the higher prevalence in men, women with MAFLD face a greater risk of cardiovascular events and mortality. Findings highlight the importance of gender-specific considerations in primary prevention programmes for MAFLD-related cardiovascular disease and mortality.
Collapse
Affiliation(s)
- Mahsa Abbaszadeh
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Farhad Hosseinpanah
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical SciencesTehranIran
| | - Sahar Karimpour Reyhan
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Maryam Mahdavi
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Majid Valizadeh
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| |
Collapse
|
16
|
Huang P, Rodriguez-Matos FJ, Qi J, Trehan R, Myojin Y, Zhu XB, Greten TF, Ma C. Hepatic immune environment differences among common mouse strains in models of MASH and liver cancer. JHEP Rep 2025; 7:101380. [PMID: 40342632 PMCID: PMC12060451 DOI: 10.1016/j.jhepr.2025.101380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 05/11/2025] Open
Abstract
Background & Aims Inbred mouse strains are critical tools for studying immune regulation of metabolic dysfunction-associated steatohepatitis (MASH) and hepatocellular carcinoma (HCC). Here, we profiled mouse strain-associated hepatic immune differences, and performed mice-human cross-species immune comparisons. Methods Immune landscapes of C57BL/6, BALB/c, and FVB/N mice were compared under healthy, MASH, or HCC state using high-dimensional spectral flow cytometry (n = 4 per condition). MASH was induced by feeding methionine- and choline-deficient or Western diet + carbon tetrachloride. HCC was caused by hydrodynamic plasmid injection of MYC/sg-p53. Public mouse and human scRNA-seq datasets were used for validation and cross-species comparisons. Results In healthy mice, liver CD4+ T (24% vs. 14% vs. 34%, p <0.05) and B cells (36.5% vs. 35% vs.18%, p <0.05) varied the most among three strains. C57BL/6 mice showed TH1 dominance, whereas BALB/c and FVB/N mice had TH2 dominance (log[TH1:TH2] = 0.17, -0.31, -0.17). In MASH mice, expansion of liver myeloid cells and innate lymphocytes were commonly found, but changes of B cells (log(fold-change) = -0.38, -0.28, -0.58, p <0.05) and T subsets (e.g. CD4+ T log(fold-change) = -0.21, -0.07, -0.15, p <0.05) varied greatly among strains. MYC/sg-p53 HCC induced a consistent expansion of liver Tregs and CD8+ T cells (p <0.05), but differential shifts of liver immune landscape were seen among strains. The flow cytometry data was supported by public scRNA-seq datasets matching C57BL/6 background. Further cross-species comparison in MASH condition confirmed shared changes of adaptive lymphocytes between mice and humans. In two MASH models, BALB/c or C57BL/6 mice were more consistent to recapture loss of CD4+ T or B cells, respectively (p <0.05). Conclusions Substantial liver immune differences exist among common mouse strains. Mice can recapitulate certain human liver immune changes with strain variations. Impact and implications Our immune cell profiling study revealed that the liver immune environment can be quite different among common mouse strains both under healthy and pathologic states, such as steatohepatitis or neoplastic processes. Our results serve as a data resource for studies investigating liver immunology and provide valuable insights for the design of studies on various immune cells in the livers of mice.
Collapse
Affiliation(s)
- Patrick Huang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francisco J. Rodriguez-Matos
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Qi
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rajiv Trehan
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuta Myojin
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiao Bin Zhu
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
| | - Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
17
|
Mj VB, Eh TM, Jm ZM, Em BC, U OM, Ej UG, Td SG, DA OA, Mj VJ. Sex-specific differences in NAFLD development: effect of a high-sucrose diet on biochemical, histological, and genetic markers in C57bl/6N mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025; 35:1237-1250. [PMID: 39107980 DOI: 10.1080/09603123.2024.2386113] [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: 03/22/2024] [Accepted: 07/25/2024] [Indexed: 01/03/2025]
Abstract
Sucrose intake is a potential risk factor for non-alcoholic fatty liver disease (NAFLD). Individual characteristics such as sex, play arole in the biological variation of the disease, potentially related to genetic regulation. This research evaluated sex differences in biochemical, histopathological, and gene expression responses associated with NAFLD in C57bl/6N mice on a high sucrose diet. Female and male mice were assigned to control or high sucrose diets (50% sucrose solution) for 20 weeks. After sacrifice, blood and hepatic tissue were collected for analysis. Female mice revealed moderate-to-high NAFLD, whereas male mice showed mild-to-moderate NAFLD. Sex-specific variations were observed in Cd36 gene expression, an upregulation in females compared with the male group, and Adipor1 gene expression showed significant downregulation in the female group in response to high sucrose diet compared with the control group. These findings highlight the importance of considering gender disparities in the treatment and management of NAFLD.
Collapse
Affiliation(s)
- Vega Burgueño Mj
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Torres Montoya Eh
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Zazueta-Moreno Jm
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Barron-Cabrera Em
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Osuna-Martínez U
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Urías-García Ej
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Salinas-Garza Td
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Ochoa-Acosta DA
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Vergara-Jiménez Mj
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| |
Collapse
|
18
|
Gu X, Chen W, Xia HM, Du LJ, Wang YH, Gao SY, He ZY, Cai JY, Hu X, Zhang XX, Yang LJ, Pan LY, Li J, Li YQ, Gu XJ, Yang B. Inverse association of healthy diet scores with non-alcoholic fatty liver disease among Chinese patients with type 2 diabetes mellitus. Int J Food Sci Nutr 2025:1-10. [PMID: 40300836 DOI: 10.1080/09637486.2025.2499042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 02/20/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025]
Abstract
We aimed to investigate the association of healthy diet scores (HDS), comprising major components (fruits and vegetables, soybean, fish, and sugar-sweetened beverages), with non-alcoholic fatty liver disease (NAFLD) in patients with type 2 diabetes mellitus (T2DM). In this cross-sectional study of 2,404 T2DM individuals aged 35-70 years, individuals with higher HDS (≥3 components) had a lower odds of NAFLD (adjusted odds ratio [OR]: 0.64; 95% confidence interval [CI]: 0.48, 0.84) and lower fatty liver index (FLI) levels (β: -4.70; 95% CI: -7.61, -1.79). Each one-component increase in HDS was associated with a 14% reduction in the odds of NAFLD (OR: 0.86; 95% CI: 0.75, 0.98) and a 1.95-unit reduction in FLI levels (β: -1.95; 95% CI: -3.21, -0.70). These results suggest that adherence to a higher HDS pattern may be protective against NAFLD in T2DM.
Collapse
Affiliation(s)
- Xiao Gu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Wei Chen
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, China
- Insitute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hui-Min Xia
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin-Jia Du
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu-Hua Wang
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Si-Yu Gao
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Ying He
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Yao Cai
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiang Hu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xing-Xing Zhang
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Juan Yang
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin-Yu Pan
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Li
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying-Qian Li
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue-Jiang Gu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bo Yang
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, China
- Insitute of Lipids Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Miao MY, Han WW, Lyu JQ, Liu ZY, Jiang W, Zhang Z, Zhu Z, Qin LQ, Chen GC. Female reproductive factors and metabolic dysfunction-associated steatotic liver disease: an integrated analysis of population cohort, liver imaging, and genetic data. Am J Obstet Gynecol 2025:S0002-9378(25)00212-1. [PMID: 40204176 DOI: 10.1016/j.ajog.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Although premenopausal women are at lower risk for metabolic dysfunction-associated steatotic liver disease than men within the same age group, the sex advantage becomes minimal after menopause, suggesting a role for female hormones. OBJECTIVE This study aimed to elucidate the role of reproductive factors in the development of female metabolic dysfunction-associated steatotic liver disease, using an integrated analysis. STUDY DESIGN Up to 269,607 women without metabolic dysfunction-associated steatotic liver disease during baseline recruitment (2006-2010) of the UK Biobank were included. Among these, 21,017 women did not develop metabolic dysfunction-associated steatotic liver disease across the follow-up period (through December 2021) and had the measure of liver proton density fat fraction quantified by magnetic resonance imaging since 2014. Multivariable Cox regression models were applied to assess the prospective relationships of various reproductive factors with incident metabolic dysfunction-associated steatotic liver disease. Multiple linear regression models were used to explore the relationships of reproductive factors with liver proton density fat fraction. A 2-sample Mendelian randomization analysis was conducted to investigate causality in the relationships of certain reproductive factors with metabolic dysfunction-associated steatotic liver disease. RESULTS During a median of 12.65 years of follow-up, 3077 incident cases of metabolic dysfunction-associated steatotic liver disease were identified. Early menarche, a greater number of live births, younger age at first live birth, and oral contraceptives or hormone replacement therapy initiated at a young age were associated with an elevated risk of metabolic dysfunction-associated steatotic liver disease and higher levels of liver proton density fat fraction. Several other reproductive factors (ie, a greater number of miscarriages, surgical menopause at a young age, and prolonged use of hormone replacement therapy) were associated with incident metabolic dysfunction-associated steatotic liver disease but not with liver proton density fat fraction. In the Mendelian randomization analysis, genetically determined earlier age at menarche and younger age at first live birth were substantially associated with increased risk of metabolic dysfunction-associated steatotic liver disease. CONCLUSION Several reproductive factors were associated with the risk of and histologic features of metabolic dysfunction-associated steatotic liver disease, supporting the role of female hormones in the pathogenesis of metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Meng-Yuan Miao
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Wen-Wen Han
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Jie-Qiong Lyu
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Zhong-Yue Liu
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Wei Jiang
- Department of Hepatopancreatobiliary Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Zheng Zhang
- Centre of Child Health Management, Children's Hospital of Soochow University, Suzhou, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Li-Qiang Qin
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Guo-Chong Chen
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
20
|
McCary A, Sheu YS, Chesbrough K, Jonas MC. Improved Liver Fibrosis Regression After Direct-Acting Antiviral Therapy in Hepatitis C Patients: A Comparison of Patients With and Without MASLD. Clin Ther 2025:S0149-2918(25)00087-6. [PMID: 40287335 DOI: 10.1016/j.clinthera.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE Chronic Hepatitis C (CHC) often results in liver fibrosis. Therefore, an important benefit of CHC treatment with direct-acting antiviral (DAA) medication is liver fibrosis regression. However, it is unclear how concurrent liver steatosis affects fibrosis regression following DAA therapy. Recent guidelines have defined liver steatosis associated with metabolic syndrome as metabolic dysfunction-associated steatotic liver disease (MASLD). We sought to examine the association of MASLD with the fibrosis regression benefits of DAA treatment for CHC. METHODS We conducted an observational retrospective analysis using electronic health records of patients aged 18-65 who completed DAA therapy for CHC from 2016 through 2022. FIB-4 scores were calculated during three time periods: just prior to DAA initiation, within 6 months post-DAA completion, and within 6-12 months post-DAA completion. These scores categorized liver fibrosis as high risk (>3.25), intermediate risk (1.45-3.25), or low risk (<1.45). An ordinal logistic regression model assessed the degree of fibrosis regression across these periods in CHC patients with and without MASLD. FINDINGS We identified 845 patients with CHC who received DAA therapy, of whom 225 met MASLD criteria. Both CHC patients with and without MASLD exhibited a decrease in FIB-4 category (coefficient = -0.361, P < 0.001) within the year following DAA therapy. The reduction in FIB-4 category post-treatment was more pronounced in the MASLD group compared to the non-MASLD group, as evidenced by a significant interaction between group and time period (coefficient = -0.439, P = 0.004). IMPLICATIONS In our cohort, MASLD was associated with greater liver fibrosis regression in the year following DAA therapy for CHC. This suggests that the concurrent presence of MASLD is not associated with diminished fibrosis regression from DAA therapy. Additional research is needed to determine the exact mechanism responsible for DAA-associated fibrosis regression in patients with MASLD.
Collapse
Affiliation(s)
- Alexis McCary
- Department of Gastroenterology, Mid-Atlantic Permanente Medical Group, Upper Marlboro, Maryland; Mid-Atlantic Permanente Research Institute, Mid-Atlantic Permanente Medical Group, Washington, District of Columbia.
| | - Yi-Shin Sheu
- Mid-Atlantic Permanente Research Institute, Mid-Atlantic Permanente Medical Group, Washington, District of Columbia
| | - Karen Chesbrough
- Mid-Atlantic Permanente Research Institute, Mid-Atlantic Permanente Medical Group, Washington, District of Columbia
| | - M Cabell Jonas
- Mid-Atlantic Permanente Research Institute, Mid-Atlantic Permanente Medical Group, Washington, District of Columbia
| |
Collapse
|
21
|
Satari S, Mota INR, Silva ACL, Brito HO, Oliveira PA, Gil da Costa RM, Medeiros R. Hallmarks of Cancer Cachexia: Sexual Dimorphism in Related Pathways. Int J Mol Sci 2025; 26:3952. [PMID: 40362192 PMCID: PMC12071346 DOI: 10.3390/ijms26093952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer-associated cachexia (CAC), also known as wasting syndrome, is a systemic condition that affects multiple tissues and organs via a variety of metabolic pathways. Systemic inflammation, progressive weight loss, depletion of adipose tissue, and skeletal muscle impairment are some of the hallmark features of cachexia. Despite various studies on the clinical features of CAC, the complexity of the syndrome continues to pose significant challenges in clinical practice, leading to late diagnoses and the absence of a standardised treatment. Men and women respond differently to CAC, which may be prompted by the pre-existing physiologic sex differences. This review presents the sexual dimorphism associated with the hallmark pathways involved in CAC. A comprehensive understanding of sexual dimorphism in these pathways could drive research on cachexia to prioritise the inclusion of more females in related studies in order to achieve personalised sex-based therapeutic approaches and, consequently, enhance treatment efficacy and better patient outcomes.
Collapse
Affiliation(s)
- Setareh Satari
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- The Institute of Public Health, University of Porto (ISPUP), Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Inês N. R. Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Ana Carolina Leão Silva
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Haissa Oliveira Brito
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Rui Miguel Gil da Costa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Biomedical Research Center, Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal
- ECO-European Cancer Organization, 1040 Brussels, Belgium
- Research Department of the Portuguese League Against Cancer—Regional Nucleus of the North (Liga Portuguesa Contra o Cancro—Núcleo Regional do Norte), 4200-172 Porto, Portugal
| |
Collapse
|
22
|
Landgraf A, Okada J, Horton M, Liu L, Solomon S, Qiu Y, Kurland IJ, Sidoli S, Pessin JE, Shinoda K. Widespread discordance between mRNA expression, protein abundance and de novo lipogenesis activity in hepatocytes during the fed-starvation transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.15.649020. [PMID: 40376090 PMCID: PMC12080948 DOI: 10.1101/2025.04.15.649020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The mammalian liver plays a critical role in maintaining metabolic homeostasis during fasting and feeding. Liver function is further shaped by sex dimorphism and zonation of hepatocytes. To explore how these factors interact, we performed deep RNA-sequencing and label-free proteomics on periportal and pericentral hepatocytes isolated from male and female mice under fed and starved conditions. We developed a classification system to assess protein-mRNA relationship and found that gene products (mRNA or protein) for most zonation markers showed strong concordance between mRNA and protein. Although classical growth hormone regulated sex-biased gene products also exhibited concordance, ∼60% of sex-biased gene products showed protein-level enrichment without corresponding mRNA differences. In contrast, transition between feeding and starvation triggered widespread changes in mRNA expression without significantly affecting protein levels. In particular, key lipogenic mRNAs (e.g. Acly , Acaca , and Fasn ) were dramatically induced by feeding, but their corresponding proteins (ACLY, ACC1, and FAS) showed little to no change even as functional de novo lipogenic activity increased ∼28-fold in the fed state. To facilitate further exploration of these findings, we developed Discorda ( https://shinoda-lab.shinyapps.io/discorda/ ), a web database for interactive analysis. Our findings reinforce the principle that mRNA changes do not reliably predict corresponding protein levels (and vice versa), particularly in the context of sex and acute metabolic regulation of hepatocytes, and that de novo lipogenesis activity can be completely uncoupled from changes in protein expression.
Collapse
|
23
|
Hosseini MS, Barjesteh F, Azedi F, Alipourfard I, Rezaei Z, Bahreini E. Comparative analysis of β-Estradiol and testosterone on lipid droplet accumulation, and regulatory protein expression in palmitate/oleate-induced fatty HepG2 cells. BMC Gastroenterol 2025; 25:263. [PMID: 40240962 PMCID: PMC12004869 DOI: 10.1186/s12876-025-03863-6] [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: 12/22/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
INTRODUCTION The higher prevalence of non-alcoholic fatty liver disease (NAFLD) in men than women before menopause and the reduced difference post-menopause suggest that sex hormones may influence liver lipid accumulation. This study compared the effects of sex hormones on lipid droplet (LD) accumulation in palmitate/ oleate-treated HepG2 cells. METHODS The MTT method was used to determine effective doses of palmitic and oleic acids in HepG2 cells, followed by a combined dose for inducing LD formation. Changes in LD content after treatment with various doses of β-estradiol and testosterone were evaluated qualitatively and semi-quantitatively using Oil Red O staining and light microscopy. The effects of these hormones on gene expression related to LD formation and lipogenesis, including PLIN2, ATGL, CGI-58, and CIDEB, were assessed using quantitative PCR. RESULTS Treatment of HepG2 cells with palmitate and oleate increased LD accumulation and the expression of PLIN2 and CIDE while elevating ATGL expression without affecting CGI-58. With no significant difference, both β-estradiol and testosterone significantly reduced LD accumulation in steatotic HepG2 cells. Gene analysis indicated that both hormones decreased PLIN2 and increased CGI-58 expression. Testosterone did not affect CIDE, while β-estradiol reduced it at low doses. Combined treatment showed no significant changes in gene expression compared to individual hormone effects, but LD accumulation was synergistically reduced. CONCLUSION This study demonstrates that β-estradiol and testosterone significantly modulate LD content and the expression of key regulatory genes in HepG2 cells, with β-estradiol showing a somewhat dominant role in enhancing lipid turnover and mitigating lipid accumulation.
Collapse
Affiliation(s)
- Motahare-Sadat Hosseini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Barjesteh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Rezaei
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Park SW, Ning H, Carnethon MR, VanWagner LB. Cardiovascular Health Trajectories and Prevalent Metabolic Dysfunction-Associated Steatotic Liver Disease in Midlife: The CARDIA Study. J Am Heart Assoc 2025; 14:e037948. [PMID: 40194968 DOI: 10.1161/jaha.124.037948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/19/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Metabolic-dysfunction associated steatotic liver disease (MASLD) is associated with prevalent cardiovascular disease. More favorable cardiovascular health (CVH) profiles are associated with a lower prevalence of MASLD in cross-sectional studies. The relationship between long-term CVH patterns and MASLD prevalence in midlife remains unknown. METHODS AND RESULTS Participants (aged 18-30 years at baseline) of the CARDIA (Coronary Artery Risk Development in Young Adults) study who had individual CVH components measured at 7 examinations over 20 years and liver fat assessed by noncontrast computed tomography at year 25 follow-up were included. CVH score was defined using published American Heart Association definitions. Group-based trajectory modeling was used to identify CVH trajectories. MASLD was defined as liver attenuation of ≤51 Hounsfield units with at least 1 metabolic risk factor after excluding other causes of liver fat. Logistic regression was used to examine associations of CVH trajectory groups and MASLD prevalence. At baseline, 39% of 2529 participants had high and 5% had low CVH, respectively. MASLD prevalence at year 25 was 23% (n=587). Five distinct CVH trajectories were identified. Between the 2 groups that started at similar CVH scores, those whose CVH declined over time had a higher prevalence of MASLD at year 25 (7.0% in high-stable versus 23.0% high-decreasing; 24.4% in moderate-stable versus 35.7% in moderate-decreasing). Lower and decreasing trajectories were associated with higher year-25 MASLD prevalence compared with the high-stable trajectory. CONCLUSIONS Achieving and maintaining high CVH scores starting in young adulthood lowers the risk of prevalent MASLD in midlife.
Collapse
Affiliation(s)
- Seong W Park
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL USA
| | - Hongyan Ning
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL USA
| | - Mercedes R Carnethon
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL USA
| | - Lisa B VanWagner
- Department of Medicine, Division of Digestive and Liver Diseases University of Texas Southwestern Medical Center Dallas TX USA
| |
Collapse
|
25
|
Huang L, Luo Y, Zhang L, Wu M, Hu L. Machine learning-based disease risk stratification and prediction of metabolic dysfunction-associated fatty liver disease using vibration-controlled transient elastography: Result from NHANES 2021-2023. BMC Gastroenterol 2025; 25:255. [PMID: 40229697 PMCID: PMC11998142 DOI: 10.1186/s12876-025-03850-x] [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: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a common chronic liver disease and represents a significant public health issue. Nevertheless, current risk stratification methods remain inadequate. The study aimed to use machine learning in the identification of significant features and the development of a predictive model to determine its usefulness in discrimination of MAFLD's risk stratification (low, moderate, and high) in adults. METHODS The data of the 2021-2023 NHANES database were analyzed. Vibration-controlled transient elastography measurements, including controlled attenuation parameter for the evaluation of steatosis and liver stiffness for the evaluation of fibrosis, were used for risk stratification. The participants were grouped into low-risk, moderate-risk, and high-risk groups based on specific criteria. Feature selection was conducted through Least Absolute Shrinkage and Selection Operator (LASSO) regression and random forest classification. RESULTS A total of 4,227 participants were included in the study. There were 16 significant predictors identified by LASSO regression, among which the top 10 predictors were demographic (age, gender, race, hypertension history), clinical (body mass index, waist circumference, hemoglobin, glycohemoglobin, lymphocyte count), and education level. The area under the receiver operating characteristic curve (AUC) of the random forest model in the validation set was 0.80, and the individual AUC was 0.83, 0.66 and 0.79 for the low-, moderate-, and high-risk groups, respectively. CONCLUSION Our machine learning model has excellent performance in stratification of risk for MAFLD with readily available clinical and demographic parameters. This model could be employed as a valuable screening tool to refer high-risk patients for further hepatological evaluation.
Collapse
Affiliation(s)
- Liqiong Huang
- Department of Ultrasound, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Sichuan Province, No. 18 Wanxiang North Road, High Tech Zone, Chengdu, China
| | - Yu Luo
- Department of Ultrasound, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Sichuan Province, No. 18 Wanxiang North Road, High Tech Zone, Chengdu, China
| | - Li Zhang
- Department of Ultrasound, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Sichuan Province, No. 18 Wanxiang North Road, High Tech Zone, Chengdu, China
| | - Mengqi Wu
- Department of Ultrasound, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Sichuan Province, No. 18 Wanxiang North Road, High Tech Zone, Chengdu, China
| | - Lirong Hu
- Department of Ultrasound, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Sichuan Province, No. 18 Wanxiang North Road, High Tech Zone, Chengdu, China.
| |
Collapse
|
26
|
Oral A, Solmaz I, Koca N, Topaloglu US, Demir I, Dundar A, Kirik A, Basci OK, Sen H, Binnetoglu E, Okuroglu N, Aydin A, Kaya ZI, Yildiz H, Acet A, Tazegul G, Sozel H, Ozudogru O, Issever K, Yaylacı S, Korkmaz UB, Oflas ND, Küçük C, Konur K, Ayaz T, Isiklar A, Arac E, Sumbul HE, Ozturk HA, Govez AB, Durmus YU, Onmez A, Serin SO, Yalcin N, Ertinmaz A, Guven AT, Kok M, Sahinturk Y, Uyar S. Obesity-Related Disorders in Türkiye: A Multi Center, Retrospective, Cross-Sectional Analysis from the OBREDI-TR Study. J Clin Med 2025; 14:2680. [PMID: 40283509 PMCID: PMC12028298 DOI: 10.3390/jcm14082680] [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: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: Obesity is a significant public health concern, as it is associated with the development of numerous chronic diseases. The prevalence of obesity and attendant diseases has been increasing over recent years. This study attempted to ascertain the frequency of chronic diseases in obese patients in Türkiye for the first time on this scale. Methods: A retrospective study was conducted, with patients admitted to the internal medicine outpatient clinics or obesity centers between December 2023 and December 2024 included in this study. Participants were recruited from seven regions, 20 provinces, and 28 centers, and the inclusion criteria were met by those aged 18 years and over with a body mass index (BMI) of 30 kg per square meter (kg/m2) or above. Their status, with respect to chronic diseases, and their anthropometric parameters were documented. Results: The total number of patients was 10,121, with a mean age of 45.2 ± 13.92. Of these, 7222 (71.35%) were female. The prevalence of type 2 diabetes mellitus (T2DM), hypertension (HT), dyslipidemia (DL), coronary artery disease (CAD), obstructive pulmonary disease (OPD), obstructive sleep apnea syndrome (OSAS), and fatty liver disease (FLD) was found to be 35.01%, 78.19%, 12.37%, 10.32%, 5.88%, and 75.12%, respectively. A subsequent analysis of the prevalence of these diseases by region revealed a statistically significant variation between regions (p < 0.001 for all regions). Conclusions: This study represents a substantial contribution to the existing body of knowledge in this field, particularly with regard to the identification of the current chronic disease rate of obese patients in Türkiye.
Collapse
Affiliation(s)
- Alihan Oral
- Department of Internal Medicine, Faculty of Medicine, Biruni University, Halkalı Street No. 99, 34295 İstanbul, Türkiye;
| | - Ihsan Solmaz
- Department of Internal Medicine, Diyarbakir Gazi Yasargil Education Research Hospital, 21070 Diyarbakir, Türkiye;
| | - Nizameddin Koca
- Department of Internal Medicine, Health Sciences University Bursa Health Application and Research Center, Bursa City Hospital, 16250 Bursa, Türkiye; (N.K.); (N.Y.); (A.E.)
| | | | - Ismail Demir
- Department of Internal Medicine, Bozyaka Education Research Hospital, 35170 Izmir, Türkiye;
| | - Ahmet Dundar
- Department of Internal Medicine, Mardin Savur Prof. Dr. Aziz Sancar State Hospital, 47860 Savur, Türkiye;
| | - Ali Kirik
- Department of Internal Medicine, Faculty of Medicine, Balikesir University, Altieylül, 10145 Balikesir, Türkiye; (A.K.); (O.K.B.); (H.S.)
| | - Ozge Kama Basci
- Department of Internal Medicine, Faculty of Medicine, Balikesir University, Altieylül, 10145 Balikesir, Türkiye; (A.K.); (O.K.B.); (H.S.)
| | - Hacer Sen
- Department of Internal Medicine, Faculty of Medicine, Balikesir University, Altieylül, 10145 Balikesir, Türkiye; (A.K.); (O.K.B.); (H.S.)
| | - Emine Binnetoglu
- Department of Internal Medicine, Corlu Vatan Hospital, 59860 Corlu, Türkiye;
| | - Nalan Okuroglu
- Department of Internal Medicine, Fatih Sultan Mehmet Education Research Hospital, 34752 Istanbul, Türkiye;
| | - Ahmet Aydin
- Department of Internal Medicine, Faculty of Medicine, Medipol University, Bagcilar, 34214 Istanbul, Türkiye;
| | - Zeynep Irmak Kaya
- Department of Internal Medicine, Health Sciences University Eskisehir Health Application and Research Center, Eskisehir City Hospital, 26080 Eskisehir, Türkiye;
| | - Hamit Yildiz
- Department of Internal Medicine, Faculty of Medicine, Gaziantep University, 27600 Sehitkamil, Türkiye;
| | - Aycan Acet
- Department of Internal Medicine, Faculty of Medicine, Kutahya Health Sciences University, 43020 Kutahya, Türkiye;
| | - Gokhan Tazegul
- Department of Internal Medicine, Faculty of Medicine, Marmara University, 34854 Istanbul, Türkiye;
| | - Hasan Sozel
- Department of Internal Medicine, Faculty of Medicine, Akdeniz University, 07100 Antalya, Türkiye;
| | - Osman Ozudogru
- Department of Internal Medicine, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Türkiye;
| | - Kubilay Issever
- Department of Internal Medicine, Giresun University Education Research Hospital, 28100 Giresun, Türkiye;
| | - Selcuk Yaylacı
- Department of Internal Medicine, Faculty of Medicine, Sakarya University, 54100 Sakarya, Türkiye;
| | - Ugur Bayram Korkmaz
- Department of Internal Medicine, Izmir Katip Celebi Education Research Hospital, 35360 Izmir, Türkiye;
| | - Nur Duzen Oflas
- Department of Internal Medicine, Faculty of Medicine, Van Yuzuncu Yil University, 54100 Van, Türkiye;
| | | | - Kamil Konur
- Department of Internal Medicine, Faculty of Medicine, Recep Tayyip Erdogan University, 53020 Rize, Türkiye
| | - Teslime Ayaz
- Department of Internal Medicine, Bakircay University Cigli Education Research Hospital, 36610 Izmir, Türkiye;
| | - Aysun Isiklar
- Department of Internal Medicine, Acibadem Atasehir Hospital, Atasehir, 34642 Istanbul, Türkiye;
| | - Esref Arac
- Department of Internal Medicine, Faculty of Medicine, Dicle University, 21010 Diyarbakir, Türkiye;
| | - Hilmi Erdem Sumbul
- Department of Internal Medicine, Health Sciences University Adana Health Application and Research Center, Adana City Hospital, 01230 Adana, Türkiye; (H.E.S.); (H.A.O.); (A.B.G.); (Y.U.D.)
| | - Huseyin Ali Ozturk
- Department of Internal Medicine, Health Sciences University Adana Health Application and Research Center, Adana City Hospital, 01230 Adana, Türkiye; (H.E.S.); (H.A.O.); (A.B.G.); (Y.U.D.)
| | - Ali Burak Govez
- Department of Internal Medicine, Health Sciences University Adana Health Application and Research Center, Adana City Hospital, 01230 Adana, Türkiye; (H.E.S.); (H.A.O.); (A.B.G.); (Y.U.D.)
| | - Yusuf Usame Durmus
- Department of Internal Medicine, Health Sciences University Adana Health Application and Research Center, Adana City Hospital, 01230 Adana, Türkiye; (H.E.S.); (H.A.O.); (A.B.G.); (Y.U.D.)
| | - Atilla Onmez
- Department of Internal Medicine, Faculty of Medicine, Duzce University, 81000 Duzce, Türkiye;
| | - Sibel Ocak Serin
- Department of Internal Medicine, Umraniye Education Research Hospital, Umraniye, 34764 Istanbul, Türkiye;
| | - Nazif Yalcin
- Department of Internal Medicine, Health Sciences University Bursa Health Application and Research Center, Bursa City Hospital, 16250 Bursa, Türkiye; (N.K.); (N.Y.); (A.E.)
| | - Aysegul Ertinmaz
- Department of Internal Medicine, Health Sciences University Bursa Health Application and Research Center, Bursa City Hospital, 16250 Bursa, Türkiye; (N.K.); (N.Y.); (A.E.)
| | - Alper Tuna Guven
- Department of Internal Medicine, Faculty of Medicine, Baskent University, 06790 Ankara, Türkiye;
| | - Mehmet Kok
- Department of Internal Medicine, Antalya Education Research Hospital, 07080 Antalya, Türkiye; (M.K.); (Y.S.); (S.U.)
| | - Yasin Sahinturk
- Department of Internal Medicine, Antalya Education Research Hospital, 07080 Antalya, Türkiye; (M.K.); (Y.S.); (S.U.)
| | - Seyit Uyar
- Department of Internal Medicine, Antalya Education Research Hospital, 07080 Antalya, Türkiye; (M.K.); (Y.S.); (S.U.)
| |
Collapse
|
27
|
Yuan M, Shi M, Yang H, Ashraf S, Iqbal S, Turkez H, Boren J, Zhang C, Uhlén M, Altay O, Mardinoglu A. Targeting PKLR in liver diseases. Trends Endocrinol Metab 2025:S1043-2760(25)00054-2. [PMID: 40221236 DOI: 10.1016/j.tem.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
Pyruvate kinase is a key regulator in hepatic glucose metabolism, encoded by the gene pyruvate kinase liver/red blood cells (PKLR). Systems biology-based approaches, including metabolic and gene co-expression networks analyses, as well as genome-wide association studies (GWAS), have led to the identification of PKLR as a pivotal gene influencing liver metabolism in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) and hepatocellular carcinoma (HCC). Here, we review the critical role of PKLR in MASLD and HCC progression and examine the effects of PKLR modulation both in vitro and in vivo. We also discuss the development of therapeutic strategies for patients with MASLD and HCC by modulating PKLR, highlighting its promising future in a broader range of liver diseases.
Collapse
Affiliation(s)
- Meng Yuan
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden
| | - Mengnan Shi
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden
| | - Hong Yang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden
| | - Sajda Ashraf
- Trustlife Laboratories, Drug Research & Development Center, 34774, Istanbul, Turkey
| | - Shazia Iqbal
- Trustlife Laboratories, Drug Research & Development Center, 34774, Istanbul, Turkey
| | - Hasan Turkez
- Medical Biology Department, Faculty of Medicine, Atatürk University, Erzurum TR-25240, Turkey
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden; Institute of Liver Studies, King's College London, London, SE5 8AF, UK
| | - Mathias Uhlén
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden
| | - Ozlem Altay
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-17165, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
28
|
Emanuele F, Biondo M, Tomasello L, Arnaldi G, Guarnotta V. Ketogenic Diet in Steatotic Liver Disease: A Metabolic Approach to Hepatic Health. Nutrients 2025; 17:1269. [PMID: 40219026 PMCID: PMC11990071 DOI: 10.3390/nu17071269] [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: 03/05/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major cause of chronic liver dysfunction worldwide, characterized by hepatic steatosis that may progress to nonalcoholic steatohepatitis and cirrhosis. Owing to its strong association with metabolic disorders, current management focuses on weight reduction via lifestyle modifications. Recently, the very-low-calorie ketogenic diet (VLCKD) has emerged as a promising intervention due to its potential for rapid weight loss and reduction in liver fat. This review aims to evaluate the clinical evidence regarding the impact of ketogenic diets on hepatic steatosis. We conducted an extensive MEDLINE literature search in databases including PubMed, Scopus, and Web of Science up to December 2024. Studies assessing the effects of ketogenic or low-carbohydrate high-fat diets on liver fat, evaluated by imaging, histology, or biochemical markers, were included. The analysis indicates that ketogenic diets significantly reduce hepatic fat content and improve metabolic parameters, including insulin sensitivity and liver enzyme levels. Evidence further suggests that substituting saturated fats with unsaturated fats or replacing carbohydrates with proteins may enhance these benefits. However, considerable variability exists among studies and long-term data remain limited. Although short-term outcomes are encouraging, potential adverse effects such as dyslipidaemia, gastrointestinal disturbances, and transient 'keto flu' symptoms require careful clinical monitoring. Future research should focus on elucidating underlying mechanisms, optimizing dietary composition, and assessing long-term safety to establish ketogenic diets as a robust strategy for managing MASLD.
Collapse
Affiliation(s)
- Fabrizio Emanuele
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (F.E.); (L.T.); (G.A.); (V.G.)
| | - Mattia Biondo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Laura Tomasello
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (F.E.); (L.T.); (G.A.); (V.G.)
| | - Giorgio Arnaldi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (F.E.); (L.T.); (G.A.); (V.G.)
| | - Valentina Guarnotta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (F.E.); (L.T.); (G.A.); (V.G.)
| |
Collapse
|
29
|
Zhou Y, Chen D, Zhu W, Liang Z, Zhao L, Zeng H, Wu L, Ye X, Ao C, Diao K. Causal Relationships between Iron Status and Nonalcoholic Fatty Liver Disease: Two-Sample, Multivariable, and Two-Step Mendelian Randomization. INT J VITAM NUTR RES 2025; 95:26773. [PMID: 40298154 DOI: 10.31083/ijvnr26773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/08/2024] [Accepted: 01/20/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) was clinically documented to be accompanied by iron homeostasis imbalances, however, the causal relationship between them remains unclear. Therefore, this study aimed to examine the relationship between iron homeostasis indicators (serum iron, ferritin, transferrin, total iron binding capacity (TIBC), and transferrin saturation (TSAT)) and NAFLD risk. METHODS We applied two-sample Mendelian randomization (MR) to assess the effects of genetic liability to iron homeostasis indicators (N = 43,220-246,139) on NAFLD risk (N = 377,988) in individuals of European ancestry. Reverse direction MR, multivariable MR, and two-step MR were performed to estimate reverse association, causal effects independent of smoking or drinking, and the mediating effect of lipid metabolism, respectively. Smoking and drinking as confounders were considered confounders. RESULTS Genetically predicted serum iron, ferritin, and TSAT were significantly associated with a higher risk of NAFLD (odds ratio (OR): 1.286, 95% confidence interval (CI): 1.075-1.539; p = 0.0059; OR: 1.260, 95% CI: 1.050-1.500, p = 0.0195; and OR: 1.223, 95% CI: 1.067-1.402; p = 0.0039, respectively). Reverse direction MR analysis suggested that genetic liability to NAFLD had no significant causal effect on iron homeostasis. Sex-specific MR exhibited a stronger effect size for the association of elevated ferritin with NAFLD risk in males (OR: 1.723, 95% CI: 1.338-2.219; p = 2.48 × 10-5). Two-step MR revealed that elevated triglycerides (TGs) mediated approximately 3%-5% of the observed effect of serum iron and TSAT on NAFLD risk, while decreased low-density lipoprotein cholesterol (LDL-C) mediated 9%-10%. CONCLUSION Genetic liability to iron status imbalance may causally affect NAFLD. This evidence may support the clinical treatment of NAFLD in the target population.
Collapse
Affiliation(s)
- Yi Zhou
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Dongze Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, 100142 Beijing, China
| | - Weilin Zhu
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Zhisheng Liang
- Department of Global Health, School of Public Health, Peking University, 100091 Beijing, China
| | - Liang Zhao
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Huatang Zeng
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Liqun Wu
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Xin Ye
- Institute for Global Public Policy, Fudan University, 200433 Shanghai, China
- LSE-Fudan Research Center for Global Public Policy, Fudan University, 200433 Shanghai, China
| | - Chaoqun Ao
- Shenzhen Health Development Research and Data Management Center, 518028 Shenzhen, China
| | - Kaichuan Diao
- Shenzhen Center for Chronic Disease Control, 518038 Shenzhen, Guangdong, China
| |
Collapse
|
30
|
Milani I, Chinucci M, Leonetti F, Capoccia D. MASLD: Prevalence, Mechanisms, and Sex-Based Therapies in Postmenopausal Women. Biomedicines 2025; 13:855. [PMID: 40299427 PMCID: PMC12024897 DOI: 10.3390/biomedicines13040855] [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: 03/08/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease influenced by genetic, lifestyle, and environmental factors. While MASLD is more prevalent in men, women are at increased risk after menopause, highlighting the critical pathogenetic role of sex hormones. The complex interplay between estrogen deficiency, visceral fat accumulation, metabolic syndrome (MetS), and inflammation accelerates disease progression, increases cardiovascular (CV) risk, and triggers a cycle of worsening adiposity, metabolic dysfunction, and psychological problems, including eating disorders. Weight loss in postmenopausal women can significantly improve both metabolic and psychological outcomes, helping to prevent MASLD and related conditions. This review examines the prevalence of MASLD, its comorbidities (type 2 diabetes T2D, CV, mental disorders), pathogenetic mechanisms, and pharmacological treatment with GLP-1 receptor agonists (GLP1-RAs), with a focus on postmenopausal women. Given the use of GLP1-RAs in the treatment of obesity and T2D in MASLD patients, and the increase in MetS and MASLD after menopause, this review analyzes the potential of a stable GLP-1-estrogen conjugate as a therapeutic approach in this subgroup. By combining the synergistic effects of both hormones, this dual agonist has been shown to increase food intake and food reward suppression, resulting in greater weight loss and improved insulin sensitivity, glucose, and lipid metabolism. Therefore, we hypothesize that this pharmacotherapy may provide more targeted therapeutic benefits than either hormone alone by protecting the liver, β-cells, and overall metabolic health. As these effects are only supported by preclinical data, this review highlights the critical need for future research to evaluate and confirm the mechanisms and efficacy in clinical settings, particularly in postmenopausal women.
Collapse
Affiliation(s)
- Ilaria Milani
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (M.C.); (F.L.); (D.C.)
| | | | | | | |
Collapse
|
31
|
Esposito M, Buono R, Angeli P, Girardi P, Di Pascoli M. Cardiometabolic risk factors and clinical course of liver cirrhosis. Dig Liver Dis 2025; 57:869-876. [PMID: 39672771 DOI: 10.1016/j.dld.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND The global prevalence of Metabolic Dysfunction-Associated Liver Disease is dramatically increasing with the diffusion of cardiometabolic risk factors (CMRFs). The aim of the present study was to assess the natural course of liver cirrhosis, in terms of decompensation, development of hepatocellular carcinoma and mortality, in relation to the presence of CMRFs (type 2 diabetes mellitus, obesity, arterial hypertension, low HDL levels, hypertriglyceridemia). PATIENTS 667 patients with liver cirrhosis (50 with CMRFs and without non-metabolic aetiological factors, 167 with non-metabolic aetiological factors and without CMRFs, and 450 with both non-metabolic aetiological factors and at least one CMRF) followed at the University and General Hospital of Padua, Italy, from 1998 to 2022, were included. RESULTS No difference in the occurrence of cirrhosis decompensating events and development of hepatocellular carcinoma was observed, whereas patients in the metabolic or mixed group had 4-3-fold higher all-cause mortality and significantly lower 3-years survival compared to patients in the non-metabolic group, despite a better liver function at enrolment. Hypertriglyceridemia and low HDL levels were the less prevalent CMRFs, but those associated with the highest risk of cirrhosis decompensation. Hypertriglyceridemia was also associated with an increased risk of mortality. Arterial hypertension was associated with a reduced risk of cirrhosis decompensation, but a higher risk of mortality. CONCLUSION Compared to patients without CMRFs, those with CMRFs had similar rates of liver cirrhosis decompensation but higher overall mortality. Hypertriglyceridemia was associated with a high risk of both liver decompensation and death.
Collapse
Affiliation(s)
- Michele Esposito
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Raffaele Buono
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paolo Girardi
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari, University of Venice, Italy
| | - Marco Di Pascoli
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine - DIMED, University of Padova, Padova, Italy.
| |
Collapse
|
32
|
Bagheri Lankarani K, Jamalinia M, Zare F, Heydari ST, Ardekani A, Lonardo A. Liver-Kidney-Metabolic Health, Sex, and Menopause Impact Total Scores and Monovessel vs. Multivessel Coronary Artery Calcification. Adv Ther 2025; 42:1729-1744. [PMID: 39951214 DOI: 10.1007/s12325-025-03121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/17/2025] [Indexed: 03/02/2025]
Abstract
INTRODUCTION Liver-kidney-metabolic health (LKMH) depends on complex interactions between metabolic dysfunction-associated steatotic liver disease (MASLD), chronic kidney disease (CKD), sex, and reproductive status. This study evaluates in a holistic manner how LKMH, sex, and menopause influence coronary artery calcification (CAC) burden. METHODS Patients without previous cardiovascular disease were prospectively recruited. Liver fat was assessed via ultrasonography and categorized as mild or moderate-to-severe. CKD was classified using estimated glomerular filtration rate (eGFR). CAC burden was quantified as 0, 1-299, ≥ 300, single-vessel, or multivessel with coronary computed tomography. Stepwise backward multinomial logistic regression was applied for analysis. RESULTS A total of 446 patients (59.2% female, average age 52.9 years) were included. Moderate-to-severe MASLD was independently associated with an increased risk of CAC 1-299 [OR 2.30 (1.21-4.36)], CAC ≥ 300 [OR 4.93 (1.46-16.59)], and single-vessel CAC [OR 2.03 (1.03-4.00)]. Mild MASLD [OR 2.47 (1.20-4.21)], moderate-to-severe MASLD [OR 3.74 (1.76-7.93)], and CKD stage 2 [OR 2.27 (1.26-4.08)] were independently associated with increased multivessel CAC risk. Liver fat content showed a dose-response association with CAC burden. Subgroup analysis revealed that MASLD and CKD increased CAC risk in male but not female patients, with menopause significantly modifying LKMH's effect. CONCLUSION LKMH's impact on CAC burden is significantly influenced by liver fat content, eGFR, sex, and menopause, suggesting that MASLD, CKD, sex, and reproductive status should be integrated into CAC risk prediction models.
Collapse
Affiliation(s)
- Kamran Bagheri Lankarani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohamad Jamalinia
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Fatemeh Zare
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Taghi Heydari
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ardekani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amedeo Lonardo
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| |
Collapse
|
33
|
Zhao Y, Gao L, Chen J, Wei J, Lin G, Hu K, Zhao W, Wei W, Huang W, Gao L, Yuan A, Qian K, Chen AF, Pu J. Remote limb ischemic conditioning alleviates steatohepatitis via extracellular vesicle-mediated muscle-liver crosstalk. Cell Metab 2025; 37:886-902.e7. [PMID: 40118054 DOI: 10.1016/j.cmet.2025.02.009] [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: 02/15/2024] [Revised: 08/30/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is an advanced form of liver disease with adverse outcomes. Manipulating interorgan communication is considered a promising strategy for managing metabolic disease, including steatohepatitis. Here, we report that remote limb ischemic conditioning (RIC), a clinically validated therapy for distant organ protection by transient muscle ischemia, significantly alleviated steatohepatitis in different mouse models. The beneficial effect of limb ischemic conditioning was mediated by muscle-to-liver transfer of small extracellular vesicles (sEVs) and their cargo microRNAs, leading to elevation of miR-181d-5p in the liver. Hepatic miR-181d-5p overexpression faithfully mirrored the molecular and histological benefits of limb ischemic conditioning by suppressing nuclear receptor 4A3 (NR4A3). Furthermore, circulating EVs from human volunteers undergoing limb ischemic conditioning improved steatohepatitis and transcriptomic perturbations in primary human hepatocytes and animal models. Our data underscore the translational potential of limb ischemic conditioning for steatohepatitis management and extend our understanding of muscle-liver crosstalk.
Collapse
Affiliation(s)
- Yichao Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Jianqing Chen
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Jingze Wei
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Guanqiao Lin
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewei Hu
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Wubin Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Huang
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingchen Gao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ancai Yuan
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Graduate School of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
34
|
He Y, Hu M, Miao X, Xu F, Deng J, Song Z, Li M, Ming Y, Leng S. Dynamic Status of Systemic Immune Inflammation Index Is Associated With Metabolic Dysfunction-Associated Steatotic Liver Disease: An Evidence From a Ten-Year Prospective Longitudinal Cohort Study. J Inflamm Res 2025; 18:4595-4606. [PMID: 40191090 PMCID: PMC11972002 DOI: 10.2147/jir.s509814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Objective Previous research studies have linked the systemic immune inflammation index (SII), derived from a complete blood count, to metabolic dysfunction-associated steatotic liver disease (MASLD). However, evidence on the relationship between longitudinal changes in SII and MASLD remains limited. This study aimed to explore distinct SII trajectories and their association with MASLD incidence. Methods A longitudinal study analyzed 25,600 individuals who underwent periodic health assessments at a Dalian City hospital between 2014 and 2023. MASLD was diagnosed via ultrasound. The SII was calculated using the formula SII = (platelet count × neutrophil count) / lymphocyte count. Group-based trajectory modeling was used to identify SII trajectories, and restricted cubic spline (RCS) analysis was employed to assesse the dose-response relationship. Stratified analyses and sensitivity analyses were also conducted. Results Three SII trajectories were identified: "low stable" (50.6%), "moderate stable" (35.1%), and "high stable" (8.9%). After adjustments, the hazard ratios (HR) for MASLD incidence were 1.118 (95% CI: 1.057-1.182, P<0.001) for the "moderate stable" group and 1.284 (95% CI: 1.172-1.408, P<0.001) for the "high stable" group. These associations persisted after adjusting for lifestyle factors. A significant non-linear relationship between SII and MASLD risk was found in both the overall population and among different genders. Subgroup and sensitivity analyses consistently confirmed these findings. Conclusion Elevated SII levels are significantly associated with an increased risk of MASLD, particularly among individuals under 45 and women. Regular SII monitoring may improve risk stratification and facilitate targeted prevention strategies for those at higher risk of MASLD.
Collapse
Affiliation(s)
- Yangxuan He
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Manling Hu
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Xinlei Miao
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Fei Xu
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| | - Jiayi Deng
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Ziping Song
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Meng Li
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| | - Yunxiang Ming
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Song Leng
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| |
Collapse
|
35
|
Li D, Meng K, Liu G, Wen Z, Han Y, Liu W, Xu X, Song L, Cai H, Yang P. Lactiplantibacillus plantarum FRT4 protects against fatty liver hemorrhage syndrome: regulating gut microbiota and FoxO/TLR-4/NF-κB signaling pathway in laying hens. MICROBIOME 2025; 13:88. [PMID: 40158133 PMCID: PMC11954192 DOI: 10.1186/s40168-025-02083-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/08/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Fatty liver hemorrhage syndrome (FLHS) has become one of the major factors leading to the death of laying hen in caged egg production. FLHS is commonly associated with lipid peroxidation, hepatocyte injury, decreased antioxidant capacity, and inflammation. However, there are limited evidences regarding the preventive effect of Lactiplantibacillus plantarum on FLHS in laying hens and its mechanisms. Our previous results showed that Lp. plantarum FRT4 alleviated FLHS by regulating lipid metabolism, but did not focus on its antioxidant and anti-inflammatory functions and mechanisms. Therefore, this study aimed to investigate the preventive mechanisms of Lp. plantarum FRT4 in alleviating FLHS, with a focus on its role in antioxidant activity and inflammation regulation. RESULTS Supplementation with Lp. plantarum FRT4 enhanced the levels of T-AOC, T-SOD, and GSH-Px, while reducing the levels of TNF-α, IL-1β, IL-8, and NLRP3 in the liver and ovary of laying hens. Additionally, Lp. plantarum FRT4 upregulated the mRNA expressions of SOD1, SOD2, CAT, and GPX1, downregulated the mRNA expressions of pro-inflammatory factors IL-1β, IL-6, and NLRP3, and upregulated the mRNA expressions of anti-inflammatory factors IL-4 and IL-10. Lp. plantarum FRT4 improved the structure and metabolic functions of gut microbiota, and regulated the relative abundances of dominant phyla (Bacteroidetes, Firmicute, and Proteobacteria) and genera (Prevotella and Alistipes). Additionally, it influenced key KEGG pathways, including tryptophan metabolism, amino sugar and nucleotide sugar metabolism, insulin signaling pathway, FoxO signaling pathway. Spearman analysis revealed that the abundance of microbiota at different taxonomic levels was closely related to antioxidant enzymes and inflammatory factors. Furthermore, Lp. plantarum FRT4 modulated the mRNA expressions of related factors in the FoxO/TLR-4/NF-κB signaling pathway by regulating gut microbiota. Moreover, the levels of E2, FSH, and VTG were significantly increased in the ovary after Lp. plantarum FRT4 intervention. CONCLUSIONS Lp. plantarum FRT4 effectively ameliorates FLHS in laying hens. This efficacy is attributed to its antioxidant and anti-inflammatory properties, which are mediated by modulating the structure and function of gut microbiota, and further intervening in the FoxO/TLR-4/NF-κB signaling pathway. These actions enhance hepatic and ovarian function and increase estrogen levels. Video Abstract.
Collapse
Affiliation(s)
- Daojie Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Kun Meng
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Guohua Liu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhiguo Wen
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yunsheng Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Weiwei Liu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xin Xu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Liye Song
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hongying Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Peilong Yang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
36
|
Li X, Wang S, Zhang Z, Deng W, Zhang Y, Cao W, Wei X, Gao Z, Yao L, Wang S, Yi W, Xie Y, Li M. Exploring the severity and risk factors of non-alcoholic fatty liver disease using the SAF scoring system. Front Med (Lausanne) 2025; 12:1510679. [PMID: 40224639 PMCID: PMC11985763 DOI: 10.3389/fmed.2025.1510679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Objective The steatosis, activity, and fibrosis (SAF) score is a histological scoring system developed by the European Association for the Study of the Liver to evaluate liver biopsy samples in cases of non-alcoholic fatty liver disease (NAFLD). Based on histopathological results and SAF scores, NAFLD patients were categorized into mild, moderate, and severe groups. We compared the differences between these groups and identified the risk factors influencing lesion severity. Methods We gathered data from 539 NAFLD patients who underwent percutaneous liver biopsy confirmation at Beijing Ditan Hospital between January 2018 and December 2022. All biopsies were graded according to the SAF scoring system, and the severity of the disease was classified as mild, moderate, or severe. We compared the differences in gender, age, BMI, history of diabetes, history of hypertension, aspartate aminotransferase (AST), alanine aminotransferase (ALT), serum cholesterol levels, and other factors among NAFLD patients with varying degrees of disease severity. Additionally, we explored the risk factors that influenced the severity of lesions. Results A total of 539 patients were enrolled in this study, with ages ranging from 6 to 79 years. Among them, there were 325 men and 214 women in an average age of 39 ± 13 years. The patients were divided into three groups based on disease severity: mild NAFLD group (162 cases), moderate NAFLD group (210 cases), and severe NAFLD group (167 cases). The results showed significant differences between the three groups in terms of age composition, high-calorie diet, family history of hypertension, ALT, AST, GGT, total bile acids, cholinesterase, glycosylated albumin, blood glucose, uric acid, type III procollagen, serum human laminin, liver stiffness, and hepatic steatosis. Conclusion BMI, uric acid, AST, type III procollagen, liver stiffness, and hepatic steatosis play critical roles in the progression of NAFLD and contribute to high pathological SAF scores in NAFLD patients.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shiyu Wang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziyu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wen Deng
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yaqin Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Weihua Cao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xin Wei
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zixuan Gao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Linmei Yao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shuojie Wang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wei Yi
- Department of Gynecology and Obstetrics, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yao Xie
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- HBV Infection, Clinical Cure and Immunology Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Department of Hepatology Division, Peking University Ditan Teaching Hospital, Beijing, China
| | - Minghui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- HBV Infection, Clinical Cure and Immunology Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Department of Hepatology Division, Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
37
|
Wang DH, He DW, Lv TT, Zhang XK, Li ZJ, Wang ZY. Estrogen receptor α suppresses hepatocellular carcinoma by restricting M2 macrophage infiltration through the YAP-CCL2 axis. BMC Cancer 2025; 25:550. [PMID: 40148834 PMCID: PMC11948847 DOI: 10.1186/s12885-025-13676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, with significant differences in incidence and outcomes between men and women. Estrogen receptor alpha (ERα) expression is associated with sex-based differences and poor prognostic outcomes in HCC. However, the detailed function of ERα in the tumor microenvironment of HCC remains unclear. METHODS Bioinformatics analysis of differentially expressed genes in HCC samples was performed from publicly available databases, and ERα was selected. The function of ERα was examined in the cell experiments. A co-culture system was built to study function of ERα-treated liver cells on macrophages in vitro. The precise mechanism was determined using quantitative real-time PCR, western blotting, immunohistochemistry, mass spectrometry, co-immunoprecipitation, and dual-luciferase reporter assay. RESULTS ERα played an important role in the pathogenesis of sexual dimorphism in HCC. ERα mainly acted on macrophages in the tumor microenvironment (TME) of HCC and reduced M2 macrophage infiltration through CCL2. By acting on NF2 and 14-3-3theta, ERα enhanced YAP phosphorylation and attenuated the nuclear translocation of YAP, thereby suppressing CCL2 expression. It also acted as a transcription factor that regulated CCL2 expression at the transcriptional level. CONCLUSION ERα/YAP/CCL2 signaling reduced M2 macrophages infiltration to inhibit HCC progression, revealing the effect of ERα in cancer cells on immune cells in HCC microenvironment.
Collapse
Affiliation(s)
- De-Hua Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
- Division of Liver Disease, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, Hebei, 050023, P. R. China
| | - Dong-Wei He
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Ting-Ting Lv
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Xiao-Kuan Zhang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Zi-Jie Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China
| | - Zhi-Yu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, P. R. China.
- , 12, Jiankang Road, Chang'an District, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
38
|
Tsutsumi T, Kawaguchi T, Fujii H, Kamada Y, Suzuki Y, Sawada K, Tatsuta M, Maeshiro T, Tobita H, Akahane T, Hasebe C, Kawanaka M, Kessoku T, Eguchi Y, Syokita H, Nakajima A, Kamada T, Yoshiji H, Sakugawa H, Morishita A, Masaki T, Ohmura T, Watanabe T, Yoda Y, Enomoto N, Ono M, Fuyama K, Okada K, Nishimoto N, Ito YM, Takahashi H, Charlton MR, Rinella ME, Sumida Y. Low HDL cholesterol levels in women and hypertriglyceridemia in men: predictors of MASLD onset in individuals without steatosis. J Gastroenterol 2025:10.1007/s00535-025-02242-y. [PMID: 40097845 DOI: 10.1007/s00535-025-02242-y] [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: 01/22/2025] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Individuals with metabolic-associated steatotic liver disease (MASLD) have a worse prognosis compared to patients without steatosis, and its prevalence is increasing. However, detailed risk factors based on obesity and sex remain unclear. We aimed to investigate the impact of cardiometabolic risk factors (CMRFs) on the risk of MASLD in individuals without pre-existing SLD. METHODS SLD was diagnosed by ultrasonography. Non-SLD individuals were followed 65,657 person-years. Incidence rates of MASLD were assessed by Kaplan-Meier analysis. Furthermore, independent factors associated with the development of MASLD were identified using Cox regression analysis, stratified by four groups: obese men, non-obese men, obese women, and non-obese women. RESULTS The overall incidence rate of MASLD was 39.3/1,000 person-years. The cumulative incidence was highest in obese men, followed by obese women, non-obese men, and non-obese women. Two or more CMRFs increased the risk of MASLD in all groups. Low HDL cholesterol level was the strongest independent risk factor in both obese and non-obese women and hypertriglyceridemia for both obese and non-obese men. The impact of these CMRFs was stronger in non-obese individuals. (HR [95% CI]: women non-obese 1.9 [1.5-2.4], obese 1.4 [1.1-1.8]; men non-obese 2.3 [1.9-2.9], obese 1.5 [1.2-2.0]). CONCLUSIONS Multiple CMRFs are important to MASLD development, regardless of sex and obesity. In this Japanese cohort, low HDL cholesterol in women and hypertriglyceridemia in men were the most significant risk factors, especially among the non-obese group. These findings suggest that sex-specific CMRFs may play a role in the development of MASLD.
Collapse
Affiliation(s)
- Tsubasa Tsutsumi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA.
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Hideki Fujii
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abeno, Osaka, 545-8585, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Suzuki
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo-Shi, Yamanashi, 409-3898, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1, Midorigaoka Higashi, Asahikawa-City, Hokkaido, 078-8510, Japan
| | - Miwa Tatsuta
- Department of Gastroenterology, KKR Takamatsu Hospital, 4-18 Tenjinmae, Takamatsu, Kagawa, 760-0018, Japan
| | - Tatsuji Maeshiro
- First Department of Internal Medicine, University of the Ryukyus Hospital, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Tobita
- Department of Hepatology, Shimane University Hospital, 89-1 Enya-Cho, Izumo, Shimane, 693-8501, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Uda City Hospital, 815 Haibarahagihara, Uda, Nara, 633-0253, Japan
| | - Chitomi Hasebe
- Department of Gastroenterology, Japanese Red Cross Asahikawa Hospital, Akebono-1Jo 1Chome 1-1, Asahikawa-City, Hokkaido, 070-0061, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, Kawasaki Medical School, Kawasaki Medical Center, 2-6-1, Nakayamashimo, Kita, Okayama, 700-8505, Japan
| | - Takaomi Kessoku
- Kanagawa Dental University Yokohama Clinic, Yokohama, Kanagawa, Japan
- Department of Palliative Medicine and Gastroenterology, International University Health and Welfare Narita Hospital, 4-3, Kimizunomori 4-Chome, Narita-Shi, Chiba, 286-8686, Japan
- Division of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yuichiro Eguchi
- Loco Medical General Institute, Mikatsukicho Kanada, Ogi-Shi, Saga, 845-0032, Japan
| | - Hayashi Syokita
- Department of Gastroenterology, Northern OKINAWA Medical Center, 1712-3 Umusa, Nago, Okinawa, 905-0006, Japan
| | - Atsushi Nakajima
- Division of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Tomoari Kamada
- Department of General Internal Medicine 2, Kawasaki Medical School, Kawasaki Medical Center, 2-6-1, Nakayamashimo, Kita, Okayama, 700-8505, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Uda City Hospital, 815 Haibarahagihara, Uda, Nara, 633-0253, Japan
| | - Hiroshi Sakugawa
- Department of Gastroenterology, Heartlife Hospital, 208, Iju, Nakagusuku, Nakagami, Okinawa, 901-2492, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Takumi Ohmura
- Department of Health Care, Asahikawa-Kosei General Hospital, 24-111, 1 Jo-Dori, Asahikawa-City, Hokkaido, 078-8211, Japan
| | - Toshio Watanabe
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshioki Yoda
- JA Yamanashi Koseiren Health Care Center, 1-26, Iida 1, Kofu, Yamanashi, 400-0035, Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo-Shi, Yamanashi, 409-3898, Japan
| | - Masafumi Ono
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Kanako Fuyama
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Naoki Nishimoto
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Yoichi M Ito
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Hirokazu Takahashi
- Liver Center, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Michael R Charlton
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA
| | - Mary E Rinella
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, 4-1-26, Akasaka, Minato-Ku, Tokyo, Japan
| |
Collapse
|
39
|
Abu-Rumaileh M, Dhoop S, Pace J, Qapaja T, Martinez ME, Tincopa M, Loomba R. Social Determinants of Health Associated With Metabolic Dysfunction-Associated Steatotic Liver Disease Prevalence and Severity: A Systematic Review and Meta-Analysis. Am J Gastroenterol 2025:00000434-990000000-01643. [PMID: 40094308 DOI: 10.14309/ajg.0000000000003421] [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: 10/21/2024] [Accepted: 01/31/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Social determinants of health (SDOHs) affect disease risk and severity leading to health disparities. SDOH impacting metabolic dysfunction-associated steatotic liver disease (MASLD) prevalence and severity are poorly characterized, and results are conflicting. The aim of this systematic review and meta-analysis was to assess the impact of individual SDOH factors on MASLD burden for adults in the United States. METHODS We searched MEDLINE, Embase, and Cochrane databases per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Studies from January 2010 to May 2024 were included. Quantitative studies of adults in the United States that evaluated SDOH beyond race/ethnicity were included. Outcomes included prevalence of MASLD, metabolic dysfunction-associated steatohepatitis (MASH), MASH-associated advanced fibrosis or cirrhosis, and clinical outcomes. RESULTS We identified 18 studies comprising 547,634 total subjects from 11 unique cohorts. Nine studies evaluated MASLD prevalence, 3 MASH prevalence, 6 MASH-associated advanced fibrosis/cirrhosis prevalence, and 9 clinical outcomes. High-diet quality was the most consistent SDOH factor associated with both MASLD and MASH-associated advanced fibrosis/cirrhosis prevalence (summarized odds ratio of 0.76, P < 0.01, and 0.74, P < 0.01, respectively). Lower income was most consistently associated with risk of clinical outcomes (significant in 3/9 studies). DISCUSSION Diet quality was the most consistent SDOH associated with disease prevalence and severity in MASLD, with other SDOH showing inconsistent associations. Prospective assessments using consensus, validated tools to assess the impact of specific SDOH on MASLD burden in heterogenous patient populations are needed to inform public health interventions.
Collapse
Affiliation(s)
| | - Sudheer Dhoop
- Department of Internal Medicine, University of Toledo, Toledo, Ohio, USA
| | - Jordan Pace
- School of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Thabet Qapaja
- Division of Hospital Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Elena Martinez
- Herbet Wertheim School of Public Health, University of California San Diego, San Diego, California, USA
| | - Monica Tincopa
- MASLD Research Center, Department of Gastroenterology and Hepatology, University of California at San Diego, Lo Jolla, California, USA
| | - Rohit Loomba
- MASLD Research Center, Department of Gastroenterology and Hepatology, University of California at San Diego, Lo Jolla, California, USA
| |
Collapse
|
40
|
Zou H, Xie J, Ma X, Xie Y. The Value of TyG-Related Indices in Evaluating MASLD and Significant Liver Fibrosis in MASLD. Can J Gastroenterol Hepatol 2025; 2025:5871321. [PMID: 40114971 PMCID: PMC11925628 DOI: 10.1155/cjgh/5871321] [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: 06/17/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025] Open
Abstract
Background: Triglyceride glucose (TyG) and its related index (TyG-body mass index, TyG-BMI) are recognized as markers for nonalcoholic fatty liver disease (NAFLD), but their associations with metabolic dysfunction-associated steatotic liver disease (MASLD) and significant liver fibrosis (SLF) risk are less studied. Therefore, this study explores the effectiveness of these indices in assessing MASLD and SLF risk in the U.S. population. Methods: Utilizing data from the National Health and Nutrition Examination Survey (NHANES), a cross-sectional study involving 5520 participants from the general population was performed. This research measured demographic, anthropometric, biochemical, comorbid, and lifestyle characteristics, all of which are considered risk factors for MASLD/SLF. Results: Upon controlling for confounding variables, only the TyG-BMI was found to have a consistent positive association with the risk of MASLD and SLF. Specifically, for each standard deviation increase, the odds ratio (OR) and 95% confidence interval (CI) were 4.44 (3.64-9.26, p for trend < 0.001) for MASLD and 2.48 (2.15-2.87, p for trend < 0.001) for SLF. Significant interactions were identified among age, sex, and the risk of MASLD associated with the TyG-BMI. The TyG-BMI also had a significant threshold effect on the risk of MASLD at a cutoff point of 180.71. Furthermore, the area under the receiver operating characteristic curve (AUC) revealed that the TyG-BMI better predicted the risk of MASLD and SLF (AUC 0.820, 95% CI 0.810-0.831; AUC 0.729, 95% CI 0.703-0.756, respectively). In addition, the integrated discrimination improvement (IDI), decision curve analysis (DCA), and net reclassification index (NRI) also demonstrated the satisfactory predictive ability of the TyG-BMI. Conclusions: Within this large dataset, the TyG-BMI was independently associated with both the MASLD score and the SLF in the MASLD cohort. Its predictive efficacy consistently surpassed that of TyG and other noninvasive models, indicating that TyG-BMI has potential for the early identification of MASLD and SLF risk.
Collapse
Affiliation(s)
- Haoxuan Zou
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiejie Xie
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaopu Ma
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Xie
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Ashraf H, Anushiravani A, Rayatpisheh M, Hamidi Alamdari D, Hossieni A, Kazeminezhad B. Association between oxidative stress and liver fibrosis severity in non-alcoholic fatty liver disease: insights from the pro-oxidant antioxidant balance method in a population from Tehran and Mashhad, Iran. Front Med (Lausanne) 2025; 12:1539605. [PMID: 40144874 PMCID: PMC11936954 DOI: 10.3389/fmed.2025.1539605] [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: 12/04/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background The exact mechanisms of non-alcoholic fatty liver disease (NAFLD), recently redefined as metabolic dysfunction-associated steatotic liver disease (MASLD), remain unclear. However, oxidative stress is recognized as a factor across all stages of NAFLD. The Pro-oxidant Antioxidant Balance (PAB) method is an important clinical tool that provides an assessment of the balance between oxidants and antioxidant. We aimed to explore oxidative stress in NAFLD using the PAB method. Methods Individuals with NAFLD were recruited in 2021. Eligible participants underwent detailed assessments, including liver elastography for fibrosis evaluation. Blood samples (5 mL) were collected to measure serum PAB levels. The METAVIR score, derived from FibroScan measurements of liver stiffness, categorized fibrosis severity from F0 (no fibrosis) to F4 (advanced fibrosis or cirrhosis). Results The study included 102 participants, with a mean age of 50.12 ± 10.03 years. Significant correlations were observed between FibroScan scores and variables such as age, body mass index (BMI), history of chronic diseases, and family history of NAFLD. PAB levels were notably higher in patients with advanced fibrosis (F2 and F3 groups: 86.32 ± 25.53) compared to those in early stages (F0 and F1 groups: 45.36 ± 21.29). Moreover, FibroScan scores showed a significant positive association with PAB values (odds ratio [OR]: 1.07; 95% confidence interval (CI): 1.04, 1.10), even after adjusting for confounding variables (OR: 1.13; 95% CI: 1.07, 1.18). Conclusion Elevated PAB levels were strongly associated with advanced stages of liver fibrosis in NAFLD patients, reflecting increased oxidative stress with disease progression. These results highlight the potential of PAB as a marker for monitoring oxidative stress and disease severity in NAFLD. Nevertheless, further large-scale studies are warranted.
Collapse
Affiliation(s)
- Hami Ashraf
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Innovative Laboratory Assays in Biomedicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Anushiravani
- Digestive Diseases Research Institute, Tehran University of Medical Science, Tehran, Iran
| | - Maryam Rayatpisheh
- Digestive Diseases Research Institute, Tehran University of Medical Science, Tehran, Iran
| | | | - Arianaz Hossieni
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Innovative Laboratory Assays in Biomedicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behrang Kazeminezhad
- Modarres Hospital, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Lai P, Miao G, Zhao Y, Han Y, Li Y, Liu Y, Guo J, Zhang W, Guo X, Xu Y, Zhang L, Chen G, Zhou Z, Mei S, Chen J, Chen J, Xu L, Zhang C, Ding Y, Dou X, Wen S, Lam SM, Shui G, Wang Y, Huang W, Zhao D, Xian X. SR-A3 suppresses AKT activation to protect against MAFLD by inhibiting XIAP-mediated PTEN degradation. Nat Commun 2025; 16:2430. [PMID: 40069146 PMCID: PMC11897346 DOI: 10.1038/s41467-025-57585-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Scavenger receptor class A member 3 (SR-A3) is implicated in metabolic diseases; however, the relationship between SR-A3 and metabolic dysfunction-associated fatty liver disease (MAFLD) has not been documented. Here, we show that hepatic SR-A3 expression is significantly reduced in human and animal models in the context of MAFLD. Genetic inhibition of SR-A3 in hamsters elicits hyperlipidemia, hyperglycemia, insulin resistance, and hepatic steatosis under chow-diet condition, yet escalates in diet-induced MAFLD. Mechanistically, SR-A3 ablation enhances E3 ligase XIAP-mediated proteasomal ubiquitination of PTEN, leading to AKT hyperactivation. By contrast, hepatic overexpression of human SR-A3 is sufficient to attenuate metabolic disorders in WT hamsters fed a high-fat-high-cholesterol diet and ob/ob mice via suppressing the XIAP/PTEN/AKT axis. In parallel, pharmacological intervention by PTEN agonist oroxin B or lipid lowering agent ezetimibe differentially corrects MAFLD in hamsters.
Collapse
Affiliation(s)
- Pingping Lai
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guolin Miao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Yinqi Zhao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yufei Han
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanwei Li
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yiran Liu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenxi Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Yitong Xu
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lianxin Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Gonglie Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zihao Zhou
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Si Mei
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jingxuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jinxuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luzheng Xu
- Peking University Medical and Health Analysis Center, Peking University, Beijing, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shengmei Wen
- NGGT (Suzhou) Biotechnology Co. Ltd, Suzhou, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Lipidall Technologies Company Limited, Changzhou, 213022, Jiangsu Province, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wei Huang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
43
|
Zhang Z, Su V, Wiese CB, Cheng L, Wang D, Cui Y, Kallapur A, Kim J, Wu X, Tran PH, Zhou Z, Casero D, Li W, Hevener AL, Reue K, Sallam T. A genome-wide ATLAS of liver chromatin accessibility reveals that sex dictates diet-induced nucleosome dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.13.623052. [PMID: 40161732 PMCID: PMC11952359 DOI: 10.1101/2024.11.13.623052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The three-dimensional organization of the genome plays an important role in cellular function. Alterations between open and closed chromatin states contributes to DNA binding, collaborative transcriptional activities and informs post-transcriptional processing. The liver orchestrates systemic metabolic control and has the ability to mount a rapid adaptive response to environmental challenges. We interrogated the chromatin architecture in liver under different dietary cues. Using ATAC-seq, we mapped over 120,000 nucleosome peaks, revealing a remarkably preserved hepatic chromatin landscape across feeding conditions. Stringent analysis of nucleosome rearrangements in response to diet revealed that sex is the dominant factor segregating changes in chromatin accessibility. A lipid-rich diet led to a more accessible chromatin confirmation at promoter regions in female mice along with enrichment of promoter binding CCAAT-binding domain proteins. Male liver exhibited stronger binding for nutrient sensing nuclear receptors. Integrative analysis with gene expression corroborated a role for chromatin states in informing functional differences in metabolic traits. We distinguished the impact of gonadal sex and chromosomal sex as determinants of chromatin modulation by diet using the Four Core Genotypes mouse model. Our data provide mechanistic evidence underlying the regulation for the critical sex-dimorphic GWAS gene, Pnpla3 . In summary, we provide a comprehensive epigenetic resource in murine liver that uncovers the complexity of chromatin dynamics in response to diet and sex. Highlights ATAC-Seq, RNA-Seq, and FCG model-integrated analysis unravel sex differences in chromatin accessibility and transcriptome responses to dietary challenges.Lipid-rich diet led to sex-biased chromatin confirmation at promoter regions.Gonadal sex emerged as the most prevalent determinant of the sex bias hepatic chromatin modulation by lipid-rich diets. The critical sex-dimorphic GWAS gene Pnpla3 is suppressed by testosterone, which underlies hepatic differences in expression between the sexes.
Collapse
|
44
|
Yang L, Qiao Y, Zhao M, Xi B. Resolving Metabolic Dysfunction-associated Steatotic Liver Disease Reduces Subclinical Cardiovascular Damage in Chinese Youths. Clin Gastroenterol Hepatol 2025:S1542-3565(25)00156-9. [PMID: 40058748 DOI: 10.1016/j.cgh.2025.01.014] [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: 10/23/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Lili Yang
- Department of Epidemiology, Department of Maternal, Child, and Adolescent Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanan Qiao
- Department of Epidemiology, Department of Maternal, Child, and Adolescent Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Xi
- Department of Epidemiology, Department of Maternal, Child, and Adolescent Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
45
|
Suri S, Singh S, Rapelly SS, Rungta S, Ahmad E. Waist circumference as a predictor for severity of liver fibrosis in non-alcoholic fatty liver disease patients. J Family Med Prim Care 2025; 14:1073-1078. [PMID: 40256066 PMCID: PMC12007795 DOI: 10.4103/jfmpc.jfmpc_1464_24] [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: 08/24/2024] [Revised: 10/15/2024] [Accepted: 11/01/2024] [Indexed: 04/22/2025] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) may in some cases progress to increasing grades of liver fibrosis and eventually cirrhosis. NAFLD patients often succumb to cardiovascular causes. Previous studies have linked visceral fat, a known cardiovascular risk factor, to NAFLD. Visceral fat is best quantified by measuring the waist circumference (WC). This study is aimed to determine the association of waist circumference with severity of liver fibrosis in NAFLD patients. Methods In this cross-sectional study we recruited 82 NAFLD patients diagnosed via ultrasonography. They underwent anthropometric examination followed by transient elastography with Fibro Scan to assess of liver stiffness measure (LSM). A cutoff LSM value of 7kPa was used to indicate significant fibrosis. Among the participants, 40 patients had an LSM value of <7 kPa (insignificant/no fibrosis), while 42 were found to have >7 kPa (significant fibrosis). Biochemical parameters of Lipid profile and Liver enzymes were also analyzed. Statistical Analysis Used SPSS software with Student t-test, Chi-square t-test, ANOVA, and Spearman correlation with 95% CI is used. P <0.05 is considered significant. Results Patients with significant fibrosis had higher mean weight (P < 0.001), BMI (P = 0.009), WC (P = 0.002), and waist-hip ratio (WHR, P = 0.032) compared to those with no fibrosis. However, hip circumference (HC) was not significantly associated between the two groups. In correlation studies, BMI (P = 0.001), weight (P < 0.001), WC (P = 0.001), and HC (P = 0.008) positively correlated with severity of liver fibrosis in NAFLD patients. However, no significant correlation was found with WHR. Conclusion Weight, BMI, and visceral fat indicators like WC and WHR are strongly associated with liver fibrosis severity in NAFLD patients. Notably, weight, BMI, WC, and HC positively correlate with fibrosis severity, while WHR does not. Early diagnosis of fatty liver is crucial to prevent progression to life-threatening conditions like NASH or NASH cirrhosis. Waist circumference could serve as a practical screening tool in primary health care centres for identifying NAFLD patients at risk of fibrosis progression.
Collapse
Affiliation(s)
- Shalini Suri
- Department of Physiology, King George Medical University Lucknow, Uttar Pradesh, India
| | - Shraddha Singh
- Department of Physiology, King George Medical University Lucknow, Uttar Pradesh, India
| | - Sushma Swaraj Rapelly
- Department of Physiology, King George Medical University Lucknow, Uttar Pradesh, India
| | - Sumit Rungta
- Department of Gastromedicine, King George Medical University Lucknow, Uttar Pradesh, India
| | - Ehsan Ahmad
- Department of Physiology, King George Medical University Lucknow, Uttar Pradesh, India
| |
Collapse
|
46
|
Zhang JW, Zhang N, Lyu Y, Zhang XF. Influence of Sex in the Development of Liver Diseases. Semin Liver Dis 2025; 45:15-32. [PMID: 39809453 DOI: 10.1055/a-2516-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The liver is a sexually dimorphic organ. Sex differences in prevalence, progression, prognosis, and treatment prevail in most liver diseases, and the mechanism of how liver diseases act differently among male versus female patients has not been fully elucidated. Biological sex differences in normal physiology and disease arise principally from sex hormones and/or sex chromosomes. Sex hormones contribute to the development and progression of most liver diseases, with estrogen- and androgen-mediated signaling pathways mechanistically involved. In addition, genetic factors in sex chromosomes have recently been found to contribute to the sex disparity of many liver diseases, which might explain, to some extent, the difference in gene expression pattern, immune response, and xenobiotic metabolism between men and women. Although increasing evidence suggests that sex is one of the most important modulators of disease prevalence and outcomes, at present, basic and clinical studies have long been sex unbalanced, with female subjects underestimated. As such, this review focuses on sex disparities of liver diseases and summarizes the current understanding of sex-specific mechanisms, including sex hormones, sex chromosomes, etc. We anticipate that understanding sex-specific pathogenesis will aid in promoting personalized therapies for liver disease among male versus female patients.
Collapse
Affiliation(s)
- Jie-Wen Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yi Lyu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
47
|
Zhang Q, Wang Y, Liu S, Zhu S, Li P, Wu S. Mortality risk associated with MASLD, MASLD type and different cardiometabolic risk factors in IBD patients: A long-term prospective cohort study. Dig Liver Dis 2025; 57:744-752. [PMID: 39581836 DOI: 10.1016/j.dld.2024.11.002] [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: 08/28/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE To examine the mortality risk associated with metabolic dysfunction-associated steatotic liver disease (MASLD), MASLD type, lean/non-lean MASLD and different cardiometabolic risk factors (CMRFs) in patients with inflammatory bowel disease (IBD) based on a long-term prospective cohort. METHODS Prevalent IBD patients at baseline who were free of alcoholic liver disease, cancer and hepatitis B/C virus seropositive were included (N=4622). MASLD, MASLD type [pure MASLD, MASLD with increased alcohol intake (MetALD)], lean/non-lean MASLD and CMRFs at baseline were defined according to the latest criteria proposed by AASLD and EASL. Primary outcome was all-cause mortality. Cox proportional hazard model was used to examine the association. RESULTS Overall, 1,763 (38.1%) were diagnosed with MASLD. During a median of 13.3-year follow-up, 451 all-cause deaths were identified. Compared with IBD-only patients, those with MASLD had a 58% excess risk of mortality (HR=1.58, 95%CI:1.07-2.32). Furthermore, as number of CMRFs increased in MASLD patients, mortality risk was significantly increased (Ptrend=0.005), with a 85% and 83% higher risk in MASLD with 3 CMRFs (HR=1.85, 95%CI:1.20-2.85) and ≥4 CMRFs (HR=1.83, 95%CI:1.16-2.89) versus IBD-only patients. Specifically, similar elevated mortality risk was observed in either pure MASLD (HR= 1.62, 95%CI:1.09-2.43) or MetALD (HR=2.03, 95%CI:1.24-3.32). Moreover, the excess mortality risk was both indicated in lean (HR=3.14, 95%CI:1.57-6.29) and non-lean MASLD (HR=1.67, 95%CI:1.12-2.48). CONCLUSIONS MASLD, either pure MASLD or MetALD, as well as lean/non-lean MASLD, is associated with increased mortality risk in IBD patients, with greater risk as number of cardiometabolic risk factors increased and evidently higher risk in lean MASLD patients.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yutao Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Shanshan Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
48
|
Zhu R, Xu C, Jiang S, Xia J, Wu B, Zhang S, Zhou J, Liu H, Li H, Lou J. Risk factor analysis and predictive model construction of lean MAFLD: a cross-sectional study of a health check-up population in China. Eur J Med Res 2025; 30:137. [PMID: 40001266 PMCID: PMC11863909 DOI: 10.1186/s40001-025-02373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
AIM Cardiovascular disease morbidity and mortality rates are high in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). The objective of this study was to analyze the risk factors and differences between lean MAFLD and overweight MAFLD, and establish and validate a nomogram model for predicting lean MAFLD. METHODS This retrospective cross-sectional study included 4363 participants who underwent annual health checkup at Yuyao from 2019 to 2022. The study population was stratified into three groups: non-MAFLD, lean MAFLD (defined as the presence of fatty liver changes as determined by ultrasound in individuals with a BMI < 25 kg/m2), and overweight MAFLD (BMI ≥ 25.0 kg/m2). Subsequent modeling analysis was conducted in a population that included healthy subjects with < 25 kg/m2 (n = 2104) and subjects with lean MAFLD (n = 849). The study population was randomly split (7:3 ratio) to a training vs. a validation cohort. Risk factors for lean MAFLD was identify by multivariate regression of the training cohort, and used to construct a nomogram to estimate the probability of lean MAFLD. Model performance was examined using the receiver operating characteristic (ROC) curve analysis and k-fold cross-validation (k = 5). Decision curve analysis (DCA) was applied to evaluate the clinical usefulness of the prediction model. RESULTS The multivariate regression analysis indicated that the triglycerides and glucose index (TyG) was the most significant risk factor for lean MAFLD (OR: 4.03, 95% CI 2.806-5.786). The restricted cubic spline curves (RCS) regression model demonstrated that the relationships between systolic pressure (SBP), alanine aminotransferase (ALT), serum urate (UA), total cholesterol (TCHO), triglyceride (TG), triglyceride glucose (TyG) index, high density lipoprotein cholesterol (HDLC), and MAFLD were nonlinear and the cutoff values for lean MAFLD and overweight MAFLD were different. The nomogram was constructed based on seven predictors: glycosylated hemoglobin A1c (HbA1c), serum ferritin (SF), ALT, UA, BMI, TyG index, and age. In the validation cohort, the area under the ROC curve was 0.866 (95% CI 0.842-0.891), with 83.8% sensitivity and 76.6% specificity at the optimal cutoff. The PPV and NPV was 63.3% and 90.8%, respectively. Furthermore, we used fivefold cross-validation and the average area under the ROC curve was 0.866 (Figure S3). The calibration curves for the model's predictions and the actual outcomes were in good agreement. The DCA findings demonstrated that the nomogram model was clinically useful throughout a broad threshold probability range. CONCLUSIONS Lean and overweight MAFLD exhibit distinct metabolic profiles. The nomogram model developed in this study is designed to assist clinicians in the early identification of high-risk individuals with lean MAFLD, including those with a normal BMI but at metabolic risk, as well as those with abnormal blood lipid, glucose, uric acid or transaminase levels. In addition, this model enhances screening efforts in communities and medical screening centers, ultimately ensuring more timely and effective medical services for patients.
Collapse
Affiliation(s)
- Ruya Zhu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Caicai Xu
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China
| | - Suwen Jiang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jianping Xia
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Sijia Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| | - Jianjun Lou
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China.
| |
Collapse
|
49
|
Wang H, Zheng C, Wang P. Exploring the nexus between hypothyroidism and metabolic dysfunction-associated steatotic liver disease: a UK biobank cohort study. Sci Rep 2025; 15:6692. [PMID: 40000892 PMCID: PMC11862249 DOI: 10.1038/s41598-025-91221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterised by lipid deposition in liver cells. The global prevalence of NAFLD has significantly increased from 8.2% in 1990 to 30.2% in 2023, establishing it as a growing public health concern. In recent years, the name NAFLD has been replaced by metabolic dysfunction-associated fatty liver disease (MASLD). Numerous observational studies have investigated the potential association between hypothyroidism and MASLD; however, the findings remain inconsistent. In this context, a systematic analysis was conducted to examine the relationship between hypothyroidism and MASLD using data from a large cohort within the UK Biobank. Utilising prospective data from the UK Biobank, a Cox proportional hazards model supplemented with multiple sensitivity analyses was applied to investigate the association between the incidence of hypothyroidism and the onset of MASLD. In addition, stratified analyses and prognostic assessments were performed to assess potential effect modifiers. To explore the underlying mechanisms, mediation analyses were conducted, along with restricted cubic spline regression, to examine potential non-linear relationships and mediation effects within this association. The study found that after fully adjusting for multiple covariates, the risk of MASLD in hypothyroidism patients was 1.711 times that of non-hypothyroidism patients (95% CI 1.560-1.877, P < 0.001). Both subtypes of hypothyroidism, namely non-surgical related hypothyroidism (NSRH) and surgical related hypothyroidism (SRH), were associated with a markedly elevated risk of MASLD onset. For NSRH, the risk is increased by 1.710 times (95% CI 1.557-1.878, P < 0.001), and for SRH, the risk is increased by 1.763 times (95% CI 1.344-2.313, P < 0.001). Stratified analysis revealed an interaction effect between gender and BMI in relation to the risk of MASLD among individuals with NSRH. Mediation analysis revealed the critical role of specific biomarkers in elucidating the relationship between hypothyroidism and MASLD. Notably, red cell distribution width, C-reactive protein, HbA1c, and total protein were identified as significant mediators in this association. Patients with hypothyroidism exhibit a significantly increased risk of developing MASLD, with inflammatory and metabolic markers playing a mediating role in this association. These findings suggest that individuals with hypothyroidism, particularly those with elevated levels of inflammatory markers, may be at heightened risk for MASLD. As such, enhanced clinical monitoring of liver function in these patients is recommended to facilitate early detection and intervention.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun, China
| | - Changlin Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Peisong Wang
- Thyroid Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
50
|
Manjarrés L, Xavier A, González L, Garrido C, Zacconi FC, Rivera K, Parra L, Phinikaridou A, Besa C, Andia ME. Sex differences in the relationship between body composition and MASLD progression in a murine model of metabolic syndrome. iScience 2025; 28:111863. [PMID: 39991541 PMCID: PMC11847041 DOI: 10.1016/j.isci.2025.111863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/30/2024] [Accepted: 01/03/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) progression exhibits significant sex differences, with males generally developing more severe disease. This study used an endothelial nitric oxide synthase knockout (eNOS KO) murine model to investigate sex-specific MASLD progression under a Western diet intervention. Magnetic resonance imaging (MRI) assessed body composition and liver and skeletal muscle fat fraction, revealing greater visceral fat, liver volume, and liver-to-muscle fat ratios in males. Dimensionality reduction and clustering analyses identified distinct sex-specific MASLD phenotypes and progression patterns. Histological evaluations confirmed greater liver damage in males, evidenced by higher MAFLD Activity Scores. These findings highlight the critical role of sex as a biological variable in MASLD pathology and emphasize the influence of body composition and fat distribution on disease progression. The study underscores the utility of advanced imaging and analytical techniques for refining non-invasive diagnostics and guiding sex-specific interventions, paving the way for personalized MASLD management strategies.
Collapse
Affiliation(s)
- Laura Manjarrés
- PhD Program in Medical Sciences, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
| | - Aline Xavier
- Faculty of Engineering, Universidad de Santiago de Chile, Santiago, Chile
| | - Leticia González
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
- Biomedical Imaging Center and Radiology Department, School of Medicine. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Garrido
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
- Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Flavia C. Zacconi
- Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine Rivera
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
| | - Laura Parra
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
| | - Alkystis Phinikaridou
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Cecilia Besa
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
- Biomedical Imaging Center and Radiology Department, School of Medicine. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo E. Andia
- Millennium Institute for Intelligent Healthcare Engineering, i-Health, Santiago, Chile
- Biomedical Imaging Center and Radiology Department, School of Medicine. Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|