51
|
Eslam M, Fan JG, Yu ML, Wong VWS, Cua IH, Liu CJ, Tanwandee T, Gani R, Seto WK, Alam S, Young DY, Hamid S, Zheng MH, Kawaguchi T, Chan WK, Payawal D, Tan SS, Goh GBB, Strasser SI, Viet HD, Kao JH, Kim W, Kim SU, Keating SE, Yilmaz Y, Kamani L, Wang CC, Fouad Y, Abbas Z, Treeprasertsuk S, Thanapirom K, Al Mahtab M, Lkhagvaa U, Baatarkhuu O, Choudhury AK, Stedman CAM, Chowdhury A, Dokmeci AK, Wang FS, Lin HC, Huang JF, Howell J, Jia J, Alboraie M, Roberts SK, Yoneda M, Ghazinian H, Mirijanyan A, Nan Y, Lesmana CRA, Adams LA, Shiha G, Kumar M, Örmeci N, Wei L, Lau G, Omata M, Sarin SK, George J. The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2025; 19:261-301. [PMID: 40016576 DOI: 10.1007/s12072-024-10774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/28/2024] [Indexed: 03/01/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) affects over one-fourth of the global adult population and is the leading cause of liver disease worldwide. To address this, the Asian Pacific Association for the Study of the Liver (APASL) has created clinical practice guidelines focused on MAFLD. The guidelines cover various aspects of the disease, such as its epidemiology, diagnosis, screening, assessment, and treatment. The guidelines aim to advance clinical practice, knowledge, and research on MAFLD, particularly in special groups. The guidelines are designed to advance clinical practice, to provide evidence-based recommendations to assist healthcare stakeholders in decision-making and to improve patient care and disease awareness. The guidelines take into account the burden of clinical management for the healthcare sector.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of MedicineSchool of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, Kaohsiung Medical University, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Global City, Philippines
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal MedicineHepatitis Research CenterGraduate Institute of Clinical Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tawesak Tanwandee
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rino Gani
- Department of Internal Medicine, Hepatobiliary Division, Dr. Cipto Mangunkusumo National General Hospital, Universitas Indonesia, Pangeran Diponegoro Road No. 71St, Central Jakarta, 10430, Indonesia
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Dan Yock Young
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Saeed Hamid
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Payawal
- Department of Medicine, Cardinal Santos Medical Center, Mandaluyong, Philippines
| | - Soek-Siam Tan
- Department of Hepatology, Selayang Hospital, Batu Caves, Malaysia
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Hang Dao Viet
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Jia-Horng Kao
- Graduate Institute of Clinical MedicineDepartment of Internal MedicineHepatitis Research CenterDepartment of Medical Research, National Taiwan University College of Medicine, National Taiwan University, National Taiwan University Hospital, 1 Chang-Te Street, 10002, Taipei, Taiwan
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Chia-Chi Wang
- Buddhist Tzu Chi Medical Foundation and School of Medicine, Taipei Tzu Chi Hospital, Tzu Chi University, Taipei, Taiwan
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Cairo, Egypt
| | - Zaigham Abbas
- Department of Hepatogastroenterology, Dr.Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | | | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Undram Lkhagvaa
- Department of Health Policy, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oidov Baatarkhuu
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ashok Kumar Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| | | | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - A Kadir Dokmeci
- Department of Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Institute of Clinical Medicine, School of Medicine, Taipei Veterans General Hospital, National Yang-Ming Chiao Tung University, No. 201, Section 2, Shipai RdNo. 155, Section 2, Linong St, Beitou District, Taipei City, 112, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jess Howell
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Clayton, VIC, 3008, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3165, Australia
| | - Jidong Jia
- Liver Research Center, Beijing Key Laboratory of Translational Medicine On Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, 11884, Egypt
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Central Clinical School, The Alfred, Monash University, Melbourne, Australia
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Aram Mirijanyan
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Leon A Adams
- Medical School, Faculty of Medicine and Health Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansoura, Egypt
| | - Manoj Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Necati Örmeci
- Department of Gastroenterohepatology, Istanbul Health and Technology University, Istanbul, Turkey
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - George Lau
- Humanity and Health Medical Group, Humanity and Health Clinical Trial Center, Hong Kong SAR, China
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
52
|
Youcheng L, Xun W, Zhufeng C. Association between nonalcoholic fatty liver disease and erectile dysfunction among American Adults from the National Health and Nutrition Examination Survey: a cross-sectional study. Int J Impot Res 2025; 37:320-328. [PMID: 38783042 DOI: 10.1038/s41443-024-00914-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 05/01/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a pressing public health concern. NAFLD is recognized as a disease with systemic involvement. Erectile dysfunction is a prevalent condition among men. The study examined the relationship between NAFLD, assessed via U.S. Fatty Liver Index (USFLI), and erectile dysfunction. The study used cross-sectional data from the National Health and Nutrition Examination Survey conducted between 2001 and 2004 to examine the health of those over 20 years of age, collecting details on their erectile dysfunction, USFLI, and several other essential variables. A USFLI score equal to or exceeding 30 was chosen to diagnose NAFLD, while a USFLI score below 10 was utilized to exclude the presence of fatty liver. There were 3763 participants, with 29.1% (1095/3763) who experienced erectile dysfunction. After accounting for all potential covariates, USFLI was positively associated with erectile dysfunction (OR, 1.02; 95% CI, 1.02 ~ 1.03; P < 0.001). Compared with individuals with Q1 (USFLI < 10), the adjusted odds ratio values for USFLI and erectile dysfunction in Q2 (10 ≤ USFLI < 30) and Q3 (USFLI ≥ 30, NAFLD) were 1.84 (95% CI: 1.46 ~ 2.32, p < 0.001) and 2.18 (95% CI: 1.66 ~ 2.87, p < 0.001), respectively. The association USFLI and erectile dysfunction exhibited an L-shaped curve (nonlinear, P = 0.014). The odds ratio value of developing erectile dysfunction was 1.03 (95% CI: 1.021 ~ 1.04, P < 0.001) in participants with USFLI < 50.18. This study identified a positive correlation between USFLI and erectile dysfunction within the adult American population. Our findings imply that NAFLD might constitute an independent risk factor for erectile dysfunction.
Collapse
Affiliation(s)
- Lin Youcheng
- Shengli Clinical Medical College Surgical Teaching Office, Fujian Medical University, Fuzhou, China
- Department of Urology, Urology and Nephrology Medical Center, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Wu Xun
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Chen Zhufeng
- Department of Infectious diseases, Fujian Provincial Hospital South Branch, Fuzhou, China.
- Shengli Clinical Medical College Internal Medicine Teaching Office, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
53
|
Yang Y, Chen X, Zhang H, Yang G, Zhu X, Si X, Chen F, Zhao Y, Jin F, Lu J. The correlation between the polymorphism of lysolecithin acyltransferase (MBOAT7) rs641738 and liver fibrosis. Per Med 2025; 22:113-119. [PMID: 40055064 DOI: 10.1080/17410541.2025.2476379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE We aimed to explore the relationship between the MBOAT7 rs641738 gene polymorphism and liver fibrosis and inflammation. METHODS A total of 214 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) were allocated into the mild-to-moderate and severe liver fibrosis groups based on liver fibrosis degree. The genotypes at the MBOAT7 rs641738 locus were evaluated. Differences in clinical and biochemical indicators, as well as the genotype and allele frequency distributions of the MBOAT7 rs641738 polymorphism, were analyzed across groups with varying degrees of liver fibrosis. Additionally, the clinical and biochemical differences among patients with different genotypes were examined. RESULTS Significant differences were observed in the distribution of CC, CT, and TT genotypes, as well as C and T allele frequencies at the MBOAT7 rs641738 locus, between patients with mild-to-moderate and severe fibrosis. Carriers of the CT + TT genotype had a higher risk of developing severe liver fibrosis compared to those with the CC genotype (OR > 1). Furthermore, CT + TT carriers had higher levels of inflammatory cytokines and more severe fibrosis than CC genotype carriers (all p < 0.05). CONCLUSION The MBOAT7 rs641738 gene polymorphism is associated with the severity of liver fibrosis and inflammation.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiang Chen
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Huiqin Zhang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Gang Yang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiaoyun Zhu
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiujing Si
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Feilong Chen
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yan Zhao
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Feng Jin
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Juanjuan Lu
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
54
|
Li Y, Pan T, Wang Y, Wang G, Wang F. The predictive value of triglyceride-glucose-high density lipoprotein-body mass index (TGH-BMI) for different degrees of hepatic steatosis and liver fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD). Clin Nutr ESPEN 2025; 66:290-301. [PMID: 39863255 DOI: 10.1016/j.clnesp.2025.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 12/22/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND & AIMS The triglyceride-glucose index (TyG) and triglyceride-glucose body mass index (TyG-BMI) have been identified as potential predictive factors for metabolic dysfunction-associated steatotic liver disease (MASLD). However, they do not include high density lipoprotein (HDL-C), which is closely related to lipid metabolism. Furthermore, there is a lack of comprehensive and longitudinal data to determine the cut-off points for different degrees of hepatic steatosis and liver fibrosis in MASLD. This study aimed to investigate the predictive capability of triglyceride-glucose-high density lipoprotein-body mass index (TGH-BMI) in determining hepatic steatosis and liver fibrosis in MASLD, as well as to establish the predictive cut-off points. METHODS We analyzed the relationships of TGH-BMI (TGH-BMI = ln [TG (mg/dL) ∗FBG (mg/dL)/HDL-C (mg/dL)] ∗ BMI (kg/m2)) with different degrees of hepatic steatosis and fibrosis in 35,114 participants who underwent health check-ups. A total of 2262 subjects without MASLD were selected for the analysis of cumulative hazard of hepatic steatosis and liver fibrosis in TGH-BMI dichotomous groups over a follow-up period of 1001 days. RESULTS Controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) demonstrated a consistent upward trend as TGH-BMI increased across quartile groups, as determined by One-way analysis of variance (P < 0.001). TGH-BMI and CAP, LSM exhibit distinct curve-like relationships between males and females when utilizing smoothing functions and conducting threshold effect analysis (P < 0.05). In males, prior to the inflection point at TGH-BMI = 177.733, there was a significant increase of 0.807 in CAP for every 1 unit increase in TGH-BMI (P < 0.05), after the inflection point, there was still an increase of 0.417 in CAP for every 1 unit increase in TGH-BMI (P < 0.05); There was no significant correlation between LSM and TGH-BMI before the first inflection point at TGH-BMI = 131.689 (P > 0.05) and after the second inflection point at TGH-BMI = 253.268 (P > 0.05). Between the first and the second inflection, LSM showed an increase of 0.015 for every 1 unit increase in TGH-BMI (P < 0.05). In females, before the inflection point at TGH-BMI = 94.686, there was a significant increase of 0.272 in CAP for every 1 unit increase in TGH-BMI (P < 0.05), after the inflection point, there was a notable change as CAP increased by 0.806 for every 1 unit increase in TGH-BMI (P < 0.05). There was no significant correlation between LSM and TGH-BMI before the inflection point at TGH-BMI = 118.098 (P > 0.05), after the inflection point, LSM showed an increase of 0.017 for every 1 unit increase in TGH-BMI (P < 0.05). Notably, TGH-BMI has been shown to be a strong predictor for the severity of hepatic steatosis and liver fibrosis in MASLD. The Area Under Curves (AUCs) for hepatic steatosis, moderate or above hepatic steatosis, severe hepatic steatosis and liver fibrosis in males were 0.845, 0.846, 0.882 and 0.668 respectively, the AUCs for hepatic steatosis, moderate or above hepatic steatosis, severe hepatic steatosis and liver fibrosis in females were 0.855, 0.895, 0.939 and 0.705 respectively (P < 0.05). In individuals without MASLD, the cumulative hazard of hepatic steatosis was found to be strongly associated with the dichotomy of increased TGH-BMI (TGH-BMID2: Hazard Ratio (HR) = 2.412, 95 % Confidence interval (CI): 2.0164-2.9071, P < 0.0001), while the same is true in liver fibrosis (TGH-BMID2: HR = 1.454, 95 % CI: 1.0633-1.9883, P = 0.0191). CONCLUSIONS The TGH-BMI demonstrates a strong predictive value for hepatic steatosis and liver fibrosis, with significantly different cut-off points for men and women. Therefore, it is important to consider the potential need for gender-specific cut-off points for triglyceride, glucose, high density lipoprotein and body mass index in clinical practice. In individuals without MASLD, a higher TGH-BMI is associated with an increased risk of developing MASLD in the future.
Collapse
Affiliation(s)
- Ying Li
- Department of Endocrinology, Hefei City First People's Hospital, Hefei 230001, Anhui, China
| | - Tianrong Pan
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Yue Wang
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Guojuan Wang
- Department of Endocrinology, Hefei City First People's Hospital, Hefei 230001, Anhui, China
| | - Fang Wang
- Department of the Health Management Center, The First Affiliated Hospital of USTC: Anhui Provincial Hospital, Hefei 230001, Anhui, China.
| |
Collapse
|
55
|
Yao Y, Hong Q, Ding S, Cui J, Li W, Zhang J, Sun Y, Yu Y, Yu M, Zhang C, Chen L, Jiang J, Hu Y. An umbrella review of meta-analyses on the effects of microbial therapy in metabolic dysfunction-associated steatotic liver disease. Clin Nutr 2025; 47:1-13. [PMID: 39978229 DOI: 10.1016/j.clnu.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Current pharmacological treatments for metabolic dysfunction-associated steatotic liver disease (MASLD) are often accompanied by adverse side effects. Consequently, probiotics, prebiotics, and synbiotics, which are bioactive compounds from fermented foods and offer fewer side effects, have garnered significant attention as alternative therapeutic strategies. OBJECTIVE This study aims to assess the efficacy of microbial therapies-probiotics, prebiotics, and synbiotics-in managing MASLD and to identify the optimal treatment modality for various clinical indicators through a comprehensive umbrella review of meta-analyses. METHODS A thorough literature search was conducted across PubMed, Web of Science, EMBASE, Cochrane Library, and Scopus to identify 23 meta-analyses over 18,999 MASLD patients as of November 2024. RESULTS The findings indicate that microbial treatments positively influence levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), homeostasis model assessment of insulin resistance (HOMA-IR), insulin, tumour necrosis factor-alpha (TNF-α), C-reactive protein (CRP), and body mass index (BMI) in MASLD patients. Notably, probiotics were most effective in reducing TC, ALT, AST, GGT, insulin, TNF-α, and BMI; prebiotics were most effective in reducing TG; and synbiotics were most effective in reducing LDL-C, HOMA-IR, and CRP. CONCLUSION Our study provides robust evidence for microbial treatments of MASLD, enabling targeted interventions for different indicators.
Collapse
Affiliation(s)
- Yuanyue Yao
- College of Biological and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, China
| | - Siqi Ding
- College of Biological and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Jie Cui
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Wenhui Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Jian Zhang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Ye Sun
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Yiyang Yu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Mingzhou Yu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, 214122, Wuxi, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Lianmin Chen
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China; Nanjing Medical University, Nanjing, 21100, China
| | - Jinchi Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China.
| | - Yonghong Hu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
56
|
Li Z, Guo H, He H, Wang S, Pei S, Xie L. The relationship between smoking cessation history and significant liver fibrosis among the population with metabolic dysfunction-associated steatotic liver disease in the United States. PLoS One 2025; 20:e0320573. [PMID: 40168280 PMCID: PMC11960941 DOI: 10.1371/journal.pone.0320573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Smoking was identified as a risk factor for the development of liver fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, the association between smoking cessation history and the development of liver fibrosis remains unclear. This study was intended to analyze the association between smoking cessation history and significant liver fibrosis in adult MASLD participants in the United States. METHODS This study utilized data from 2643 patients with MASLD from the National Health and Nutrition Examination Survey (NHANES). Significant liver fibrosis was detected based on transient elastography measurements. According to the smoking questionnaire data, patients were categorized as non-smokers, ex-smokers and current smokers. A multivariate logistic regression analysis, adjusted for weights, was performed to investigate the relationship between smoking cessation history and the presence of significant liver fibrosis in participants with MASLD. RESULTS A total of 2643 patients with MASLD were included in this study. Compared with non-smokers, ex-smokers had a slightly elevated risk of developing significant liver fibrosis (OR: 1.07, 95% CI: 1.02-1.13). Specifically, a positive correlation was observed between patients who quit smoking for < 20 years and significant liver fibrosis (OR: 1.07, 95% CI: 1.01-1.15). Furthermore, MASLD patients who started regularly smoking at an age of ≤ 20 years (OR: 1.09, 95% CI: 1.02-1.16) and had a smoking duration of ≥ 10 years before quitting (OR: 1.10, 95% CI: 1.02-1.18) were also highly correlated with an increased likelihood of developing significant liver fibrosis. CONCLUSIONS This study revealed that individuals with MASLD who have ceased smoking exhibit an elevated risk for significant liver fibrosis when compared to those who never smoked. It is highly emphasized that MASLD patients who quit smoking for < 20 years, started regularly smoking at an age of ≤ 20 years, and had a smoking duration of ≥ 10 years before quitting should be extremely vigilant regarding the risk of significant liver fibrosis.
Collapse
Affiliation(s)
- Zhongtao Li
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hao Guo
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hongyu He
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Shu Wang
- Department of Urology Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shufen Pei
- Department of Otolaryngology-Head and Neck Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liang Xie
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
57
|
Zhang F, Yue K, Sun S, Lu S, Jia G, Zha Y, Zhang S, Chou CJ, Liao C, Li X, Duan Y. Targeting Histone Deacetylase 11 with a Highly Selective Inhibitor for the Treatment of MASLD. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412903. [PMID: 39976110 PMCID: PMC12005767 DOI: 10.1002/advs.202412903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the most prevalent chronic liver disorder globally. Due to its intricate pathogenesis and the current lack of efficacious pharmacological interventions, there is a pressing need to discover novel therapeutic targets and agents for MASLD treatment. Herein, it is found that histone deacetylase 11 (HDAC11), a subtype of HDAC family, is markedly overexpressed in both in vitro and in vivo models of MASLD. Furthermore, the knockdown of HDAC11 is observed to mitigate lipid accumulation in hepatic cells. A highly selective HDAC11 inhibitor, B6, which exhibits favorable pharmacokinetic property and liver distribution, is further designed and synthesized. Integrating RNA-seq data with in vivo and in vitro experiments, B6 is found to inhibit de novo lipogenesis (DNL) and promote fatty acid oxidation, thus mitigating hepatic lipid accumulation and pathological symptoms in MASLD mice. Further omics analysis and experiments reveal that B6 enhances the phosphorylation of AMPKα1 at Thr172 through the inhibition of HDAC11, consequently modulating DNL and fatty acid oxidation in the liver. In summary, this study identifies HDAC11 as a potential therapeutic target in MASLD and reports the discovery of a highly selective HDAC11 inhibitor with favorable drug-like properties for the treatment of MASLD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Kairui Yue
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Simin Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Shengyuan Lu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Geng Jia
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Yang Zha
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - C. James Chou
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South CarolinaCharlestonSC29425USA
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Xiaoyang Li
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
- Marine Biomedical Research Institute of QingdaoQingdaoShandong266071China
| | - Yajun Duan
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
58
|
Ma P, Ou H, Cai J, Zhang Y, Ou Y. DRD2-Mediated AMPK Ubiquitination Regulates the Occurrence of Hepatic Steatosis. Liver Int 2025; 45:e70053. [PMID: 40052721 DOI: 10.1111/liv.70053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND & AIMS G protein-coupled receptors (GPCRs) are important potential drug targets for the treatment of metabolic disorders. The D2 dopamine receptor (DRD2), a GPCR receptor, is a member of the dopamine receptor family. However, the role of DRD2 in regulating lipid metabolism, especially in hepatic steatosis, is unclear. METHODS Eight-week male mice were fed HFHC/MCD to induce the MASH model. AAV2/8 containing the TBG promoter was used to knock down and overexpress DRD2 in mouse liver. Co-immunoprecipitation, Western lotting, immunofluorescence, and immunohistochemistry were used to investigate the mechanisms and screen DRD2 antagonists. RESULTS The study found that activation of PKC leads to the elevation and internalisation of DRD2 in a high-fat environment. Knockdown of DRD2 in mouse liver can effectively interfere with the progression of MASH, while overexpression of DRD2 significantly aggravates the process of MASH. The study on the mechanism of DRD2 regulating lipid metabolism found that the internalisation of DRD2 could lead to dephosphorylation of pAKT (T308) by binding to β-arrestin2 and pAKT, thereby inducing ubiquitin-dependent degradation of AMPK and exacerbating steatosis. L-741626, a DRD2 antagonist, was found to interfere with the internalisation of DRD2 in a high-fat environment. It has been shown that L-741626 can treat MASH by regulating the AKT-AMPK signalling axis in vitro and in vivo. CONCLUSIONS In conclusion, this study demonstrated that internalisation of DRD2 in a high-fat environment aggravated MASH progression through the AKT-AMPK signalling axis. Furthermore, L-741626, as a DRD2 antagonist, has the potential to treat MASH.
Collapse
Affiliation(s)
- Peng Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hao Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junze Cai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yuanli Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
59
|
He Z, Zhao Y, Tang H. Serum manganese and its association with non-alcoholic fatty liver disease: findings from NHANES. Front Nutr 2025; 12:1527207. [PMID: 40225343 PMCID: PMC11985439 DOI: 10.3389/fnut.2025.1527207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Objective This study examines the link between serum manganese (Mn) levels and non-alcoholic fatty liver disease (NAFLD), with a focus on gender differences. Methods Utilizing data from the NHANES 2017-2018, we included participants aged 18 and older, excluding those without ultrasonic liver assessment, serum Mn data, or with hepatitis or significant alcohol use. The final analysis comprised 4,294 individuals, with 2,708 in the NAFLD group and 1,586 in the non-NAFLD group. Serum Mn was quantified via inductively coupled plasma mass spectrometry. We compared demographic and health-related variables between groups using appropriate statistical tests and categorized participants into quartiles based on Mn levels. Multivariate logistic regression and spline regression analyses were conducted to evaluate the association between serum Mn and NAFLD risk by gender. Results Serum Mn was significantly elevated in the NAFLD group compared to non-NAFLD individuals (9.06 vs. 9.33 μg/L, Z = 2.815, p = 0.005). After adjustments, males in the third Mn quartile showed a higher NAFLD risk (OR = 1.575; 95% CI: 1.193-2.087), while females in the fourth quartile also had increased risk (OR = 1.725; 95% CI: 1.313-2.269), both compared to the first quartile (p < 0.01). A positive dose-response relationship was found for both genders (P for trend <0.01), with nonlinear associations in males (P for nonlinearity <0.01) and linear associations in females (P for nonlinearity = 0.818). Significant interactions with ethnicity in males and hypertension in females were also noted. Conclusion Higher serum Mn levels are significantly associated with increased NAFLD risk in both genders, highlighting the need for gender-specific considerations in future studies and clinical practices.
Collapse
Affiliation(s)
- Zipeng He
- Department of Ultrasound Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanrui Zhao
- Department of Radiology, The First Hospital of Fangshan Distict, Beijing, China
| | - Hua Tang
- Department of Ultrasound Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
60
|
Du K, Umbaugh DS, Wang L, Jun JH, Dutta RK, Oh SH, Ren N, Zhang Q, Ko DC, Ferreira A, Hill J, Gao G, Pullen SS, Jain V, Gregory S, Abdelmalek MF, Diehl AM. Targeting senescent hepatocytes for treatment of metabolic dysfunction-associated steatotic liver disease and multi-organ dysfunction. Nat Commun 2025; 16:3038. [PMID: 40155379 PMCID: PMC11953480 DOI: 10.1038/s41467-025-57616-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] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/23/2025] [Indexed: 04/01/2025] Open
Abstract
Senescent hepatocytes accumulate in metabolic dysfunction-associated steatotic liver disease (MASLD) and are linked to worse clinical outcomes. However, their heterogeneity and lack of specific markers have made them difficult to target therapeutically. Here, we define a senescent hepatocyte gene signature (SHGS) using in vitro and in vivo models and show that it tracks with MASLD progression/regression across mouse models and large human cohorts. Single-nucleus RNA-sequencing and functional studies reveal that SHGS+ hepatocytes originate from p21+ cells, lose key liver functions and release factors that drive disease progression. One such factor, GDF15, increases in circulation alongside SHGS+ burden and disease progression. Through chemical screening, we identify senolytics that selectively eliminate SHGS+ hepatocytes and improve MASLD in male mice. Notably, SHGS enrichment also correlates with dysfunction in other organs. These findings establish SHGS+ hepatocytes as key drivers of MASLD and highlight a potential therapeutic strategy for targeting senescent cells in liver disease and beyond.
Collapse
Affiliation(s)
- Kuo Du
- Department of Medicine, Duke University, Durham, NC, USA.
| | | | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ji Hye Jun
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rajesh K Dutta
- Department of Medicine, Duke University, Durham, NC, USA
| | - Seh Hoon Oh
- Department of Medicine, Duke University, Durham, NC, USA
| | - Niansheng Ren
- Department of Medicine, Duke University, Durham, NC, USA
| | - Qiaojuan Zhang
- Department of Neurology, Duke University, Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Ana Ferreira
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Jon Hill
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Guannan Gao
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Simon Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | | | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
61
|
Zhang Y, Yang J, Min J, Huang S, Li Y, Liu S. The emerging role of E3 ubiquitin ligases and deubiquitinases in metabolic dysfunction-associated steatotic liver disease. J Transl Med 2025; 23:368. [PMID: 40133964 PMCID: PMC11938720 DOI: 10.1186/s12967-025-06255-2] [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: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, with a prevalence as high as 32.4%. MASLD encompasses a spectrum of liver pathologies, ranging from steatosis to metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and, in some cases, progression to end-stage liver disease (cirrhosis and hepatocellular carcinoma). A comprehensive understanding of the pathogenesis of this highly prevalent liver disease may facilitate the identification of novel targets for the development of improved therapies. E3 ubiquitin ligases and deubiquitinases (DUBs) are key regulatory components of the ubiquitin‒proteasome system (UPS), which plays a pivotal role in maintaining intracellular protein homeostasis. Emerging evidence implicates that aberrant expression of E3 ligases and DUBs is involved in the progression of MASLD. Here, we review abnormalities in E3 ligases and DUBs by (1) discussing their targets, mechanisms, and functions in MASLD; (2) summarizing pharmacological interventions targeting these enzymes in preclinical and clinical studies; and (3) addressing challenges and future therapeutic strategies. This review synthesizes current evidence to highlight the development of novel therapeutic strategies based on the UPS for MASLD and progressive liver disease.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiahui Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shan Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Yuchen Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China.
| |
Collapse
|
62
|
Münte E, Hartmann P. The Role of Short-Chain Fatty Acids in Metabolic Dysfunction-Associated Steatotic Liver Disease and Other Metabolic Diseases. Biomolecules 2025; 15:469. [PMID: 40305160 PMCID: PMC12025087 DOI: 10.3390/biom15040469] [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: 02/04/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
With its increasing prevalence, metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as a major global public health concern over the past few decades. Growing evidence has proposed the microbiota-derived metabolites short-chain fatty acids (SCFAs) as a potential factor in the pathophysiology of MASLD and related metabolic conditions, such as obesity and type 2 diabetes mellitus (T2DM). By influencing key pathways involved in energy homeostasis, insulin sensitivity, and inflammation, SCFAs play an important role in gut microbiota composition, intestinal barrier function, immune modulation, and direct metabolic signaling. Furthermore, recent animal and human studies on therapeutic strategies targeting SCFAs demonstrate their potential for treating these metabolic disorders.
Collapse
Affiliation(s)
- Eliane Münte
- Department of Pediatrics, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
- Division of Gastroenterology, Hepatology & Nutrition, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| |
Collapse
|
63
|
Liao X, Yu S, Wang L, Zhang R, Yu K. Elevated red blood cell folate levels are associated with metabolic dysfunction-associated steatotic liver disease: results from NHANES 2017-2020. Front Physiol 2025; 16:1494863. [PMID: 40182691 PMCID: PMC11965589 DOI: 10.3389/fphys.2025.1494863] [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: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. However, the role of folate in MASLD remains controversial. This study aimed to investigate the association between two folate indicators [serum folate and red blood cell (RBC) folate] and MASLD prevalence using data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES). Methods A total of 3,879 participants without liver disease or significant alcohol consumption were included in the final analysis. Hepatic steatosis was assessed via transient elastography, with MASLD defined as a controlled attenuation parameter (CAP) ≥285 dB/m and the presence of at least one cardiometabolic risk factor. Logistic regression and generalized additive models (GAMs) were used to evaluate associations between folate levels and MASLD, with subgroup analyses stratified by age, gender, and body mass index (BMI). Results After full adjustment for confounders, RBC folate exhibited a significant positive association with MASLD (OR = 1.111 and 95% CI: 1.015-1.216 per 1-unit increase). In contrast, serum folate showed a transient negative association in minimally adjusted models (OR = 0.869 and 95% CI: 0.802-0.941), which disappeared after further adjustments. Subgroup analyses confirmed that age, gender, and BMI did not modify the RBC folate-MASLD relationship. Discussion These findings suggest that elevated RBC folate levels are independently associated with MASLD prevalence, whereas serum folate may lack clinical relevance due to susceptibility to confounding factors. RBC folate, as a stable biomarker of long-term folate status, may serve as a superior indicator for investigating folate-MASLD associations.
Collapse
Affiliation(s)
- Xin Liao
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Song Yu
- Department of General Medicine, the Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People’s Hospital, Chengdu, Sichuan, China
| | - Lin Wang
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Ruyue Zhang
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Ke Yu
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
64
|
Liu S, Li F, Cai Y, Sun L, Ren L, Yin M, Cui H, Pan Y, Gang X, Wang G. Gout drives metabolic dysfunction-associated steatotic liver disease through gut microbiota and inflammatory mediators. Sci Rep 2025; 15:9395. [PMID: 40102566 PMCID: PMC11920238 DOI: 10.1038/s41598-025-94118-7] [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: 11/25/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
This study explores the relationship between gout and metabolic dysfunction-associated steatotic liver disease (MASLD), two metabolic conditions linked to worsening health outcomes. While hyperuricemia's association with MASLD is established, the specific connection between gout and MASLD remains less explored. Using data from the UK Biobank, the study employs COX proportional hazard models, multi-state survival analysis, and Mendelian randomization to assess the independent and mutual risks of gout and MASLD. Findings indicate a mutual risk increase: male gout patients, those younger than 60, and those with high BMI are particularly susceptible to MASLD, while female MASLD patients are at heightened risk for gout. Shared risk factors for both conditions include high BMI, hypertension, diabetes, and hyperuricemia. The study further identifies a bidirectional causal link, with gout leading to MASLD, mediated by gut microbiota Ruminococcaceae and proteins like IL-2 and GDF11, implicating specific metabolic pathways. The findings highlight a clinical and mechanistic correlation, emphasizing the need for targeted interventions to address these overlapping metabolic pathways in future treatments.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Fan Li
- Department of Hepatobiliary and Pancreatic Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yunjia Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Mengsha Yin
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Huijuan Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yujie Pan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
65
|
Cheng B, Su X, He J, Gu Y, Chen M, Wei Y, Yi Y, Chen P, Lin X, Li T, Xu C, Liu Q, Li B. A Mendelian randomization study reveals a causal association between NASH and the risk of atrial fibrillation. Front Endocrinol (Lausanne) 2025; 16:1390259. [PMID: 40171195 PMCID: PMC11958169 DOI: 10.3389/fendo.2025.1390259] [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: 02/23/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background Epidemiological evidence suggests that non-alcoholic fatty liver disease (NAFLD) may increase the risk of atrial fibrillation (AF). However, the findings are inconsistent, and the causality remains to be established. Methods We conducted two-step, two-sample Mendelian randomization (MR) analysis to assess the association between genetically predicted NAFLD (i.e. chronically elevated serum alanine aminotransferase levels [cALT], imaging-based and biopsy-confirmed NAFLD) and AF. Subsequently, we further performed Mendelian randomization to investigate the causal relationship between non-alcoholic steatohepatitis (NASH), a subtype of NAFLD, and AF. The inverse variance weighted (IVW) method was used as the primary approach to reveal the potential causation between the exposure and outcome. Results There was no significant causal association between NAFLD diagnosed based on cALT, confirmed by imaging, or verified by biopsy, and an increased risk of atrial fibrillation. Furthermore, the results of the IVW method revealed a positive causal effect of NASH on AF (OR=1.113, 95% CI=1.025-1.209, P = 0.011). In the reverse analysis, however, no evidence supported a significant genetic association between AF and NASH (OR=0.974, 95% CI=0.934-1.016, P = 0.214). Conclusion A causal relationship existed between NASH and the risk of AF. However, no significant genetic association has been observed between NAFLD and AF risk. This suggests that managing the progression of NAFLD may hold potential value in preventing the onset of AF.
Collapse
Affiliation(s)
- Biwei Cheng
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xuekang Su
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jue He
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yanghui Gu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Mingtai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yi Wei
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yumeng Yi
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peiying Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaojuan Lin
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Tao Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Chong Xu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Qiang Liu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Biao Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
66
|
Li C, Cheng D, Ren H, Zhang T. Unraveling the gut microbiota's role in PCOS: a new frontier in metabolic health. Front Endocrinol (Lausanne) 2025; 16:1529703. [PMID: 40171188 PMCID: PMC11958223 DOI: 10.3389/fendo.2025.1529703] [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: 11/17/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder affecting reproductive-age women, characterized primarily by hyperandrogenism, ovulatory dysfunction, and metabolic abnormalities. In recent years, the gut microbiota has garnered widespread attention for its potential role as a key regulator of host metabolism in the pathogenesis of PCOS. Studies have shown that PCOS patients exhibit dysbiosis in their gut microbiota, characterized by reduced microbial diversity, an imbalance in the ratio of Firmicutes to Bacteroidetes, changes in the abundance of specific taxa, and abnormal levels of metabolic products. These alterations may exacerbate metabolic dysfunction in PCOS through multiple mechanisms, including influencing host energy metabolism, disrupting lipid and bile acid metabolism, and inducing chronic inflammation. Addressing gut dysbiosis through the modulation of patients' microbiomes-such the use of, prebiotics, fecal microbiota transplantation, and optimizing diet lifestyle-may offer strategies for improving metabolic abnormalities and alleviating clinical symptoms in PCOS. Additionally, the gut microbiome promises as a potential marker, aiding in the precise diagnosis and personalization of PCOS. Although our current understanding of how the gut microbiota influences PCOS is still limited, research is needed to explore the causal relationships and mechanisms involved, providing a more reliable theoretical basis for clinical. This review aims summarize the research progress on the relationship between gut microbiota and PCOS, and to suggest future directions to promote the development of prevention and treatment strategies for PCOS.
Collapse
Affiliation(s)
- Caihong Li
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Dongkai Cheng
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Haiqin Ren
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang, China
| |
Collapse
|
67
|
Bahrami G, Sajadimajd S, Kazemi F, Yarmohammadi F, Miraghaee SS. An oligosaccharide isolated from Rosa canina ameliorates lipid profile and liver damage in MASLD modeled rabbits: in vivo and in silico studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04027-9. [PMID: 40095053 DOI: 10.1007/s00210-025-04027-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is designated as the main hepatic evidence of metabolic syndrome. Over the past few decades, the use of herbal medications in the treatment of MASLD has been increasingly studied due to evidence of their potential therapeutic mechanisms, wide availability, and lower side effects. This study aimed to scrutinize the effect of an oligosaccharide isolated from Rosa canina on high-cholesterol diet-induced MASLD rabbits. Twenty-four New Zealand rabbits were categorized into three groups (eight rabbits in each group). The first group received only the standard diet. Others received 2% cholesterol for 120 days. Then, the rabbits were treated with 20 mg/kg of an isolated oligosaccharide daily by gavage for 60 days or 6 mg/kg of simvastatin as a standard. After 14-16 h of starvation, blood samples were collected to measure lipid profile and liver enzymes. In addition, histological sampling of the liver and thoracic aorta was done. Cholesterol and triglyceride were significantly decreased in the oligosaccharide-treated group (p < 0.05). Also, the activity of alanine aminotransferase, aspartate aminotransferase, and creatine phosphokinase decreased significantly. Lactate dehydrogenase activity was also decreased. Based on histopathological studies, the treatment with an isolated oligosaccharide prevented atherosclerotic changes and decreased liver injury. Our data suggest that an oligosaccharide isolated from Rosa canina possesses hypolipidemic and hepatoprotective activities which will be beneficial in MASLD.
Collapse
Affiliation(s)
- Gholamreza Bahrami
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| | - Farnoush Kazemi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Shahram Miraghaee
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
68
|
Zhang Y, Wang H, Xu C, Ye X, Nan Y, Hu X, Fan J, Wang X, Ju D. Rubicon siRNA-encapsulated liver-targeting nanoliposome is a promising therapeutic for non-alcoholic fatty liver disease. Int J Pharm 2025; 672:125291. [PMID: 39880145 DOI: 10.1016/j.ijpharm.2025.125291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/01/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic liver disorder worldwide, and effective therapeutic strategies for its treatment remains limited. In this article, we introduced Glipo-siRubi, a hepatocytes-targeting RNA interference (RNAi) nanoliposome for suppression of Rubicon expression, aiming to achieve precise regulation of autophagy in NAFLD. Autophagy activation induced by Rubicon suppression resulted in reduced endoplasmic reticulum stress and intracellular lipid accumulation in vitro. Moreover, Glipo-siRubi administration exhibited remarkable therapeutic efficacy, characterized by decreased liver lipid accumulation, ameliorated histopathology and improved insulin sensitivity in mice with western diet, indicating its notable potential against NAFLD. By inducing autophagy activation, the hepatocytes-targeting Glipo-siRubi provided a promising method for NAFLD treatment, addressing the limitations of current approaches. Our study highlighted the significance of Rubicon-specific suppression in NAFLD treatment, offering a specific, safe, and efficient approach to mitigate NAFLD.
Collapse
Affiliation(s)
- Yuting Zhang
- Minhang Hospital Fudan University Shanghai China; Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| | - Hanqi Wang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| | - Caili Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| | - Xiaomiao Ye
- Minhang Hospital Fudan University Shanghai China.
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| | - Xiaozhi Hu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| | - Jiajun Fan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China; Shanghai Hailu Biological Technology Co., Ltd, Shanghai 201200 China.
| | - Xuebin Wang
- Department of Pharmacy, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Dianwen Ju
- Minhang Hospital Fudan University Shanghai China; Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics Fudan University School of Pharmacy Shanghai China.
| |
Collapse
|
69
|
Jiang S, Zhang F, Yang H, Han X, Mao J, Zheng G, Fan Y. Estimated sdLDL-C as a biomarker of hepatic steatosis severity in MASLD: a retrospective study. BMC Gastroenterol 2025; 25:168. [PMID: 40082781 PMCID: PMC11907928 DOI: 10.1186/s12876-025-03759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. However, there is a lack of cost-effective and accurate biomarkers to assess the degree of hepatic steatosis. Estimated small dense low-density lipoprotein cholesterol (EsdLDL-C), a calculated value derived from triglyceride (TG) and low-density lipoprotein cholesterol (LDL-C) levels, has emerged as a potential indicator. This study aimed to explore the relationship between EsdLDL-C and the severity of hepatic steatosis. METHODS This single-center retrospective study estimated and directly measured small dense low-density lipoprotein cholesterol (sdLDL-C) in 1,969 patients who underwent serum lipid testing at Changzhou Third People's Hospital between January and July 2024. Among these, 461 patients diagnosed with MASLD were included in the study. These patients were further classified into mild (Mil) and moderate-to-severe (Mod-Sev) groups based on controlled attenuation parameter (CAP) values to explore the relationship between EsdLDL-C and the severity of hepatic steatosis. RESULTS The correlation coefficient (R) between EsdLDL-C and DsdLDL-C was 0.837, with a bias of 0.223. Both EsdLDL-C (OR 1.095, 95% CI 1.029-1.180) and visceral fat area (VFA) (OR 1.019, 95% CI 1.010-1.028) were identified as independent risk factors for Mod-Sev steatosis compared to the Mil group. After adjusting for all confounders, patients with MASLD had a 1.155-fold increased risk of developing Mod-Sev hepatic steatosis for each unit increase in EsdLDL-C. Furthermore, EsdLDL-C demonstrated good predictive value for Mod-Sev steatosis in MASLD patients, with an area under the curve (AUC) of 0.825 (95% CI 0.784-0.867). CONCLUSIONS EsdLDL-C may serve as a practical and cost-effective biomarker for identifying high-risk MASLD patients. TRIAL REGISTRATION The retrospective study was approved by the Ethics Committee of Changzhou Third People's Hospital (02 A-A20230015), and a waiver of informed consent was agreed to, as the data were obtained from medical records, and a waiver of informed consent would not have affected the participants.
Collapse
Affiliation(s)
- Shuo Jiang
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China
| | - Fan Zhang
- Department of Endocrinology, Changzhou Third People's Hospital, Changzhou, Jiangsu, China
| | - Hui Yang
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China
| | - Xue Han
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China
| | - Jieru Mao
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China
| | - Guojun Zheng
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China.
| | - Yan Fan
- Center of Medical Laboratory, Changzhou Third People's Hospital, Changzhou, Jiangsu, China.
| |
Collapse
|
70
|
Mou Y, Tang Y, Zheng X, Liu X, Wu X, Wang H, Zeng J, Rao Q, Ben-David Y, Li Y, Huang L. Unraveling the molecular mechanisms of Fufangduzhong formula in alleviating high-fat diet-induced non-alcoholic fatty liver disease in mice. Front Pharmacol 2025; 16:1542143. [PMID: 40144651 PMCID: PMC11936930 DOI: 10.3389/fphar.2025.1542143] [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: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease, characterized by hepatic lipid accumulation. The Fufangduzhong formula (FFDZ) is a traditional Chinese medicine (TCM) formulation composed of Eucommia ulmoides Oliv., Leonurus artemisia (Lour.) S. Y. Hu, Prunella vulgaris Linn, Uncariarhynchophylla (Miq.) Miq. ex Havil., and Scutellaria baicalensis Georgi. It has demonstrated hepatoprotective effects and the ability to reduce lipid accumulation. However, its mechanisms against NAFLD remain unclear. Methods UPLC-MS/MS was used to identify FFDZ metabolites. C57BL/6J mice were fed a high-fat diet (HFD) supplemented with or without FFDZ (HFD+L, 0.45 g/kg/d; HFD+H, 0.9 g/kg/d) for 12 weeks. Biochemical indicators and histopathological observations were utilized to assess the extent of metabolic homeostasis disorder and hepatic steatosis. An analysis of differentially expressed genes and regulated signaling pathways was conducted using hepatic transcriptomics. Metabolomics analysis was performed to investigate the significantly changed endogenous metabolites associated with NAFLD in mice serum using UPLC-Q-TOF/MS. Western blot was employed to detect proteins involved in the lipid metabolism-related signaling pathways. Oleic acid-induced hepatic steatosis was used to examine the lipid-lowering effect of FFDZ-containing serum in vitro. Results A total of eight active metabolites were identified from the FFDZ formula and FFDZ-containing serum through UPLC-MS/MS analysis. FFDZ reduced body weight, liver weight, and levels of inflammatory cytokines, and it ameliorated hepatic steatosis, serum lipid profiles, insulin sensitivity, and glucose tolerance in mice with HFD-induced NAFLD. Transcriptomics revealed that FFDZ modulated the lipid metabolism-related pathways, including the PPAR signaling pathway, Fatty acid metabolism, and AMPK signaling pathway. Meanwhile, Western blot analysis indicated that FFDZ downregulated the expression of lipid synthesis-related proteins (Srebp-1c, Acly, Scd-1, Fasn, Acaca, and Cd36) and upregulated the fatty acid oxidation-related proteins (p-Ampk, Ppar-α, and Cpt-1). Furthermore, metabolomics identified FFDZ-mediated reversal of phospholipid dysregulation (PC, PE, LPC, LPE). Additionally, FFDZ-containing serum remarkedly reduced OA-induced lipid accumulation in HepG2 cells. Conclusion The present results demonstrate that FFDZ exerts anti-NAFLD effects by enhancing glucose tolerance and insulin sensitivity, as well as regulating the Ampk signaling pathway to ameliorate lipid metabolism disorder, lipotoxicity, hepatic steatosis, and inflammatory responses.
Collapse
Affiliation(s)
- Yu Mou
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yao Tang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Xiuyan Zheng
- Guizhou Institute of Integrated Agriculture Development, Guiyang, China
| | - Xiang Liu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Xuemei Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Hongji Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jie Zeng
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qing Rao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yaacov Ben-David
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- School of Basic Medical, Guizhou Medical University, Guiyang, China
| | - Yanmei Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Lei Huang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
71
|
Ivashkin VT, Drapkina OM, Maevskaya MV, Raikhelson KL, Okovityi SV, Zharkova MS, Grechishnikova VR, Abdulganieva DI, Alekseenko SA, Ardatskaya MD, Bakulin IG, Bakulina NV, Bogomolov PO, Breder VV, Vinnitskaya EV, Geyvandova NI, Golovanova EV, Grinevich VB, Doshchitsin VL, Dudinskaya EN, Ershova EV, Kodzoeva KB, Kozlova IV, Komshilova KA, Konev YV, Korochanskaya NV, Kotovskaya YV, Kravchuk YA, Loranskaya ID, Maev IV, Martynov AI, Mekhtiev SN, Mishina EE, Nadinskaia MY, Nikitin IG, Osipenko MF, Ostroumova OD, Pavlov CS, Pogosova NV, Radchenko VG, Roytberg GE, Saifutdinov RG, Samsonov AA, Seliverstov PV, Sitkin SI, Tarasova LV, Tarzimanova AI, Tkacheva ON, Tkachenko EI, Troshina EA, Turkina SV, Uspenskiy YP, Fominykh YA, Khlynova OV, Tsyganova YV, Shamkhalova MS, Sharkhun OO, Shestakova MV. Clinical Guidelines of the Russian Society for the Study of the Liver, Russian Gastroenterological Association, Russian Society for the Prevention of Non-Communicable Diseases, Russian Association of Endocrinologists, Russian Scientific Medical Society of Therapists, National Society of Preventive Cardiology, Russian Association of Gerontologists and Geriatricians on Non-Alcoholic Fatty Liver Disease. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2025; 35:94-152. [DOI: 10.22416/1382-4376-2025-35-1-94-152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2025]
Abstract
Aim. The clinical guidelines are intended to provide information support for making decisions by gastroenterologists, general practitioners and internists that will improve the quality of medical care for patients with non-alcoholic fatty liver disease, taking into account the latest clinical data and principles of evidence-based medicine. Key points. Clinical guidelines contain information about current views on etiology, risk factors and pathogenesis of nonalcoholic fatty liver disease, peculiarities of its clinical course. Also given recommendations provide information on current methods of laboratory and instrumental diagnostics, invasive and non-invasive tools for nonalcoholic fatty liver disease and its clinical phenotypes assessment, approaches to its treatment, considering the presence of comorbidities, features of dispensary monitoring and prophylaxis. The information is illustrated with algorithms of differential diagnosis and physician's actions. In addition, there is information for the patient and criteria for assessing the quality of medical care. Conclusion. Awareness of specialists in the issues of diagnosis, treatment and follow-up of patients with nonalcoholic fatty liver disease contributes to the timely diagnosis and initiation of treatment, which in the long term will significantly affect their prognosis and quality of life.
Collapse
Affiliation(s)
- V. T. Ivashkin
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| | - M. V. Maevskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - K. L. Raikhelson
- Saint Petersburg State University;
Academician I.P. Pavlov First Saint Petersburg State Medical University
| | - S. V. Okovityi
- Saint Petersburg State Chemical Pharmaceutical University
| | - M. S. Zharkova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | | - M. D. Ardatskaya
- Central State Medical Academy of the Department of Presidential Affairs
| | - I. G. Bakulin
- North-Western State Medical University named after I.I. Mechnikov
| | - N. V. Bakulina
- North-Western State Medical University named after I.I. Mechnikov
| | - P. O. Bogomolov
- Russian University of Medicine;
Moscow Regional Research Clinical Institute
| | - V. V. Breder
- National Medical Research Center of Oncology named after N.N. Blokhin
| | | | | | | | | | | | | | | | - K. B. Kodzoeva
- National Medical Research Center for Transplantology and Artificial Organs named after Academician V.I. Shumakov
| | - I. V. Kozlova
- Saratov State Medical University named after V.I. Razumovsky
| | | | | | | | | | | | | | | | | | - S. N. Mekhtiev
- Academician I.P. Pavlov First Saint Petersburg State Medical University
| | | | - M. Yu. Nadinskaia
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. G. Nikitin
- N.I. Pirogov Russian National Research Medical University;
National Medical Research Center “Treatment and Rehabilitation Center”
| | | | | | - Ch. S. Pavlov
- I.M. Sechenov First Moscow State Medical University (Sechenov University);
Moscow Multidisciplinary Scientific and Clinical Center named after S.P. Botkin
| | - N. V. Pogosova
- National Medical Research Center of Cardiology named after Academician E.I. Chazov
| | | | - G. E. Roytberg
- N.I. Pirogov Russian National Research Medical University
| | - R. G. Saifutdinov
- Kazan State Medical Academy — Branch Campus of the Russian Medical Academy of Continuous Professional Education
| | | | | | - S. I. Sitkin
- North-Western State Medical University named after I.I. Mechnikov;
V.A. Almazov National Medical Research Center
| | | | - A. I. Tarzimanova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - O. N. Tkacheva
- N.I. Pirogov Russian National Research Medical University
| | | | | | | | - Yu. P. Uspenskiy
- Academician I.P. Pavlov First Saint Petersburg State Medical University;
Saint Petersburg State Pediatric Medical University
| | - Yu. A. Fominykh
- V.A. Almazov National Medical Research Center; Saint Petersburg State Pediatric Medical University
| | - O. V. Khlynova
- Perm State Medical University named after Academician E.A. Wagner
| | | | | | - O. O. Sharkhun
- N.I. Pirogov Russian National Research Medical University
| | | |
Collapse
|
72
|
Nguyen MT, Lian A, Guilford FT, Venketaraman V. A Literature Review of Glutathione Therapy in Ameliorating Hepatic Dysfunction in Non-Alcoholic Fatty Liver Disease. Biomedicines 2025; 13:644. [PMID: 40149620 PMCID: PMC11940638 DOI: 10.3390/biomedicines13030644] [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: 01/13/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global cause of liver dysfunction. This spectrum of hepatic disorders can progress to severe conditions, such as non-alcoholic steatohepatitis (NASH) and cirrhosis, due to oxidative stress and sustained cellular injury. With limited pharmacological options, glutathione (GSH), a key antioxidant, has shown promising potential in reducing oxidative stress, maintaining redox balance, and improving liver function. This literature review examines studies from 2014-2024 exploring GSH therapy in NAFLD patients. Eligible studies assessed GSH as the primary intervention for NAFLD in human subjects, reporting outcomes such as liver function or oxidative stress markers. Randomized clinical trials (RCTs) were eligible, while combination therapy studies were included if GSH's effect could be isolated. Exclusions applied to non-NAFLD studies, animal/in vitro models, and non-GSH antioxidant interventions. Analysis of three studies (totaling 109 participants) demonstrated consistent improvements in alanine transaminase (ALT) levels and reductions in oxidative stress markers like 8-hydroxy-2-deoxyguanosine (8-OHdG). However, small sample sizes and inconsistent protocols limit generalizability. Further large-scale RCTs are required to confirm GSH's efficacy, determine optimal dosing, and assess long-term effects. This literature review highlights GSH's potential as a novel NAFLD therapeutic strategy while emphasizing the need for further studies to refine its clinical application.
Collapse
Affiliation(s)
- Michelle Thuy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| |
Collapse
|
73
|
Xu Y, He P, He B, Chen Z. Bioactive flavonoids metabolites in citrus species: their potential health benefits and medical potentials. Front Pharmacol 2025; 16:1552171. [PMID: 40098613 PMCID: PMC11911525 DOI: 10.3389/fphar.2025.1552171] [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: 12/27/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Citrus flavonoids are naturally occurring phytochemicals widely present in the peels and pulps of citrus fruits. They exhibit a wide range of biological activities, including antioxidant, anti-inflammatory, hypoglycemic, lipid-lowering, antimicrobial, and gut-protective effects. These metabolites show great potential in improving metabolic syndromes such as diabetes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases. Additionally, citrus flavonoids have demonstrated significant effects in inhibiting pancreatic lipase activity, regulating lipid metabolism, and enhancing intestinal barrier function. Advances in extraction and purification techniques have further promoted their applications in the fields of food, medicine, and functional materials. This review systematically summarizes the types, bioactivities, and mechanisms of action of citrus flavonoids, providing scientific evidence for their research and development.
Collapse
Affiliation(s)
- Yuqian Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Pan He
- Sichuan Provincial Women's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, China
| | - Beihui He
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zheng Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
74
|
Marjot T. New evidence of cross-disease communication between heart and liver. J Hepatol 2025; 82:541-543. [PMID: 39721919 DOI: 10.1016/j.jhep.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Thomas Marjot
- Oxford Centre for Diabetes Endocrinology and Metabolism (OCDEM), Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, UK; Translational Gastroenterology and Liver Unit (TGLU), Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, UK.
| |
Collapse
|
75
|
Zhang S, You M, Shen Y, Zhao X, He X, Liu J, Ma N. Improving fatty liver hemorrhagic syndrome in laying hens through gut microbiota and oxylipin metabolism by Bacteroides fragilis: A potential involvement of arachidonic acid. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:182-199. [PMID: 39967692 PMCID: PMC11834063 DOI: 10.1016/j.aninu.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 08/02/2024] [Accepted: 08/18/2024] [Indexed: 02/20/2025]
Abstract
Bacteroides fragilis (B. fragilis), a crucial commensal bacterium within the gut, has shown connections with hepatic lipid metabolism and inflammation regulation. Nonetheless, the role of B. fragilis in the progression of fatty liver hemorrhagic syndrome (FLHS) remains unknown. This study aims to explore the ameliorative effects of B. fragilis on FLHS in laying hens, as well as its underlying mechanisms. This is the first study to employ a chicken FLHS model, combining microbiomics and oxylipin metabolomics to investigate the mechanism of action of intestinal symbiotic bacteria. Exp. 1: 40 laying hens at 25 weeks old were randomly divided into five treatment groups (eight replicates per group and one hen per replicate), including the control group (basal diet), the high-energy and low-protein (HELP) group, and the HELP group with three different levels (108, 109, and 1010 CFU) of B. fragilis. Exp. 2: 18 chickens at 25 weeks old were randomly divided into three treatment groups (six replicates per group and one hen per replicate) including the control group (basal diet), the model group (HELP diet), and the arachidonic acid (AA) group (HELP diet with 0.3% AA). The experiment period of Exp. 1 and Exp. 2 were 8 weeks. B. fragilis significantly improved body weight of seventh week (P = 0.006), liver lipid degeneration, blood lipid levels (triglycerides, cholesterol, and low-density lipoprotein cholesterol; P < 0.05), and liver function (alanine aminotransferase and aminotransferase; P < 0.05) in laying hens. B. fragilis downregulated the expression of lipid synthesis-related genes fatty acid synthase, acetyl-CoA carboxylase, and liver X receptor α, and inflammation-related genes tumor necrosis factor α, interleukin (IL)-1β, IL-6, and IL-8 in the liver of FLHS-affected hens (P < 0.05), while upregulating the expression of lipid oxidation-related genes carnitine palmitoyl transferase-1, peroxisome proliferator activated receptor (PPAR) α, and PPARγ (P < 0.05). The in-depth analysis indicated alterations in oxylipin pathways triggered by B. fragilis, as evidenced by changes in the expression of pivotal genes arachidonate 15-lipoxygenase, arachidonate 5-lipoxygenase (P < 0.05), subsequently causing modifications in relevant metabolites. This included a decrease in pro-inflammatory substances such as 15-oxoETE (P = 0.004), accompanied by an increase in AA (P = 0.008). B. fragilis regulated the homeostasis of intestinal flora by increasing the abundance of Bacteroides and decreasing the abundance of Succinatimonas and Faecalicoccus (P < 0.05). The integrated analysis revealed a robust positive correlation between Bacteroides abundance and AA levels (P = 0.007). This relationship was corroborated through in vitro experiments. Subsequently, the beneficial effect of AA in mitigating FLHS was confirmed in laying hens with FLHS, further supported by reverse transcription-polymerase chain reaction analysis demonstrating gene expression patterns akin to B. fragilis intervention. This study demonstrated that B. fragilis exerts an anti-FLHS effect through modulation of oxylipin metabolism and gut microbiota stability, with a pivotal role played by AA.
Collapse
Affiliation(s)
- Shaobo Zhang
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Manhua You
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Youming Shen
- Research Institute of Pomology, Chinese Academy of Agricultural Sciences, Xingcheng 125100, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Xin He
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Juxiang Liu
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Ning Ma
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
76
|
Theel WB, de Jong VD, Castro Cabezas M, Grobbee DE, Jukema JW, Trompet S. Risk of cardiovascular disease in elderly subjects with obesity and liver fibrosis and the potential benefit of statin treatment. Eur J Clin Invest 2025; 55:e14368. [PMID: 39636216 PMCID: PMC11810556 DOI: 10.1111/eci.14368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Liver fibrosis progression is influenced by older age and cardiometabolic risk factors such as obesity and is associated with an increased risk of cardiovascular events. While statins may protect against cardiovascular complications, their effects in elderly individuals with obesity and liver fibrosis are unclear. METHOD The PROspective Study of Pravastatin in the Elderly at Risk (PROSPER) database was used to evaluate the effect of pravastatin on major adverse cardiovascular events in an elderly population (>70 years). Subjects were categorized by BMI: lean (<25 kg/m2), overweight (25-29.9 kg/m2) and obese (≥30 kg/m2). Liver fibrosis was assessed using the FIB-4 index: low risk (<2.0), intermediate risk (2.0-2.66) and high risk (≥2.67). Time-to-event data were analysed using the Cox proportional hazards model, adjusted for confounders and compared the placebo and pravastatin groups. RESULTS A total of 5.804 subjects were included. In the placebo group, the highest risk group (high FIB-4 and obesity) had a significantly higher hazard ratio for (non-)fatal stroke (HR 2.74; 95% CI 1.19-6.29) compared to the low FIB-4, lean BMI group. This risk disappeared in the same pravastatin group. Pravastatin did not affect other cardiovascular endpoints. All-cause mortality was significantly higher in subjects with lean weight and high FIB-4 on placebo (HR 1.88; 95% CI 1.14-3.11), but not on pravastatin (HR .58; 95% CI .28-1.20). CONCLUSION Elderly individuals with obesity and liver fibrosis are at higher risk for (non-)fatal stroke, which is reduced with pravastatin. Pravastatin also potentially lowers all-cause mortality in subjects with lean weight and liver fibrosis.
Collapse
Affiliation(s)
- Willy B. Theel
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Obesity Center CGGRotterdamThe Netherlands
| | - Vivian D. de Jong
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Manuel Castro Cabezas
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Julius ClinicalZeistThe Netherlands
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Diederick E. Grobbee
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Johan W. Jukema
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Stella Trompet
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Internal Medicine, Section of Gerontology & GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
77
|
Vujasinovic M, Ebrahimi F, Roelstraete B, Bergman D, Sun J, Sadr‐Azodi O, Löhr J, Ludvigsson JF. Metabolic Dysfunction-Associated Steatotic Liver Disease and Pancreatic Disease-A Population-Based Nationwide Cohort and Sibling-Controlled Study. United European Gastroenterol J 2025; 13:247-256. [PMID: 39868838 PMCID: PMC11975618 DOI: 10.1002/ueg2.12761] [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: 09/24/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) has been linked to pancreatic diseases, but evidence from population-based studies with liver histology is lacking. AIMS AND METHODS In this population-based cohort including all Swedish adults (n = 8563) with biopsy-proven MASLD, we aimed to investigate incidences of pancreatic diseases compared with matched reference individuals from the general population (n = 38,858) and full siblings (n = 6696). Using Cox proportional hazard models, we calculated multivariable adjusted hazard ratios (aHRs) and confidence intervals (CIs). RESULTS We documented 359 incidents of pancreatic diseases in MASLD patients and 880 events in matched reference individuals, resulting in an incidence rate difference of 1.54 (95% CI, 1.25-1.84). The relative risk of pancreatic disease was highest in the first two years after MASLD diagnosis (aHR, 2.19 [95% CI, 1.92-2.50), but remained statistically significant increased even up to ten years [aHR, 1.60 (95% CI, 1.38-1.85)]. The most common pancreatic disease in individuals with MASLD was acute non-biliary pancreatitis (1.44 vs. 0.44 events/1000 PY), followed by chronic pancreatitis (0.54 vs. 0.12/1000 PY) and pancreatic cancer (0.88 vs. 0.47/1000 PY). We documented 130 versus 344 pancreas-related deaths among individuals with MASLD and their matched comparators, yielding an absolute risk difference of 0.51/1000 PY and an aHR of 2.41 (95%CI = 1.95-2.97). The findings were consistent in sibling-controlled analyses with an aHR of 2.21 (95%CI = 1.69-2.90). CONCLUSIONS MASLD was associated with significantly higher rates of acute and chronic pancreatitis of predominantly non-biliary origin, as well as an increased risk of pancreatic cancer and pancreas-related mortality.
Collapse
Affiliation(s)
- Miroslav Vujasinovic
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Department of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Fahim Ebrahimi
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of Gastroenterology and HepatologyUniversity Digestive Health Care Center Basel—ClarunisBaselSwitzerland
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - David Bergman
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Jiangwei Sun
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Omid Sadr‐Azodi
- Department of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
- Unit of Upper Gastrointestinal SurgerySaint Göran's HospitalStockholmSweden
- Centre for Clinical Research SörmlandUppsala UniversityEskilstunaSweden
| | - J.‐Matthias Löhr
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Department of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Celiac Disease CenterDepartment of MedicineColumbia University Medical CenterNew YorkNew YorkUSA
- Department of PediatricsÖrebro University HospitalÖrebro UniversityÖrebroSweden
| |
Collapse
|
78
|
Gu S, Zheng Y, Chen C, Liu J, Wang Y, Wang J, Li Y. Research progress on the molecular mechanisms of Saikosaponin D in various diseases (Review). Int J Mol Med 2025; 55:37. [PMID: 39717942 PMCID: PMC11722148 DOI: 10.3892/ijmm.2024.5478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
Bupleurum, a Traditional Chinese Medicine (TCM) herb, is widely used in China and other Asian countries to manage chronic liver inflammation and viral hepatitis. Saikosaponin D (SSD), a triterpenoid saponin extracted from Bupleurum, exhibits extensive pharmacological properties, including anti‑inflammatory, antioxidant, anti‑apoptotic, anti‑fibrotic and anti‑cancer effects, making it a therapeutic candidate for numerous diseases. Clarifying the targets and molecular mechanisms underlying TCM compounds is essential for scientifically validating TCM's therapeutic roles in disease prevention and treatment, as well as for identifying novel therapeutic targets and lead compounds. This analysis comprehensively examines SSD's mechanisms across various conditions, such as myocardial injury, pulmonary diseases, hepatic disorders, renal pathologies, neurological disorders, diabetes and cancer. In addition, challenges and potential solutions encountered in SSD research are addressed. SSD is posited as a promising monomer for multifaceted therapeutic applications and this article aims to enhance researchers' understanding of the current landscape of SSD studies, offering strategic insights to guide future investigations.
Collapse
Affiliation(s)
- Simin Gu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yiyuan Zheng
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Chong Chen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jing Liu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yanping Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Junmin Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
79
|
Ye M, He Y, Xia Y, Zhong Z, Kong X, Zhou Y, Xia W, Wang W, Fan H, Chen L, Wu X, Li Q. Association Between Serum Zinc and Non-Alcoholic Fatty Liver Disease and Advanced Liver Fibrosis: NHANES 2011-2016. Biol Trace Elem Res 2025; 203:1305-1316. [PMID: 38861177 DOI: 10.1007/s12011-024-04261-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: 03/21/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Limited and inconclusive evidence exists regarding the correlation between serum zinc levels and non-alcoholic fatty liver disease (NAFLD) and advanced fibrosis. The objective of this cross-sectional study was to investigate the association between serum zinc concentration and both NAFLD and advanced liver fibrosis among the United States (US) adults. 3398 subjects from National Health and Nutrition Examination Survey (NHANES) 2011-2016 were included. Serum zinc concentration was measured by inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS). NAFLD was diagnosed with Hepatic Steatosis Index (HSI), and advanced fibrosis risk was assessed by NAFLD Fibrosis Score (NFS). Weighted logistic regression and restricted cubic splines (RCS) were used to examine the association between serum zinc concentration and NAFLD and advanced fibrosis. Linear trend tests were conducted by incorporating the median of serum zinc quartiles as a continuous variable in the models. We employed sensitivity analysis and subgroup analysis to enhance the robustness of our results. The results from the RCS regression revealed no evident nonlinear relationship between serum zinc concentration and the presence of NAFLD and advanced fibrosis (p-nonlinear > 0.05). Compared with those in the lowest quartile (Q1) of serum zinc concentrations, the odds ratios (95% confidence intervals) of NAFLD were 1.49 (0.89,2.49) in Q2, 0.99 (0.68,1.45) in Q3, and 2.00 (1.40,2.86) in Q4 (p-trend = 0.002). Similarly, the odds ratios (95% confidence intervals) for advanced fibrosis in Q2-4 compared to Q1 were 0.86 (0.50,1.47), 0.60 (0.26,1.39), and 0.41 (0.21,0.77), respectively (p-trend = 0.006). Subgroup analyses and sensitivity analyses reinforce the same conclusion. The investigation revealed a positive linear relationship between serum zinc concentrations and the probability of developing NAFLD. Conversely, an inverse correlation was observed between serum zinc concentrations and the incidence of advanced liver fibrosis among individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Miaomin Ye
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yin Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Ziyi Zhong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yunting Zhou
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wenqing Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Weiping Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Huan Fan
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Lu Chen
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaohui Wu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
80
|
Sola-Sevilla N, Garmendia-Berges M, Mera-Delgado MC, Puerta E. Context-dependent role of sirtuin 2 in inflammation. Neural Regen Res 2025; 20:682-694. [PMID: 38886935 PMCID: PMC11433891 DOI: 10.4103/nrr.nrr-d-23-02063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/09/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Sirtuin 2 is a member of the sirtuin family nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, known for its regulatory role in different processes, including inflammation. In this context, sirtuin 2 has been involved in the modulation of key inflammatory signaling pathways and transcription factors by deacetylating specific targets, such as nuclear factor κB and nucleotide-binding oligomerization domain-leucine-rich-repeat and pyrin domain-containing protein 3 (NLRP3). However, whether sirtuin 2-mediated pathways induce a pro- or an anti-inflammatory response remains controversial. Sirtuin 2 has been implicated in promoting inflammation in conditions such as asthma and neurodegenerative diseases, suggesting that its inhibition in these conditions could be a potential therapeutic strategy. Conversely, arthritis and type 2 diabetes mellitus studies suggest that sirtuin 2 is essential at the peripheral level and, thus, its inhibition in these pathologies would not be recommended. Overall, the precise role of sirtuin 2 in inflammation appears to be context-dependent, and further investigation is needed to determine the specific molecular mechanisms and downstream targets through which sirtuin 2 influences inflammatory processes in various tissues and pathological conditions. The present review explores the involvement of sirtuin 2 in the inflammation associated with different pathologies to elucidate whether its pharmacological modulation could serve as an effective strategy for treating this prevalent symptom across various diseases.
Collapse
Affiliation(s)
- Noemí Sola-Sevilla
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maider Garmendia-Berges
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - MCarmen Mera-Delgado
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| |
Collapse
|
81
|
Ebeid A, Mokhtar F, Martinez-Lebron V, Park S, Degann S, Payano J, Vahora Z, Gray S, Johnson L, El-Maouche D, Abutaleb A. Use of noninvasive fibrosis calculators in an urban diabetes center suggests a large burden of undetected advanced liver disease. BMC Endocr Disord 2025; 25:53. [PMID: 40011894 PMCID: PMC11866707 DOI: 10.1186/s12902-025-01881-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction associated steatotic liver disease (MASLD) is prevalent in up to 60% of patients with type 2 diabetes mellitus (T2DM). T2DM accelerates the risk of hepatic fibrosis and hepatocellular carcinoma in patients with MASLD. Our goal in this study was to identify patients with suspected MASLD and hepatic fibrosis in a large T2DM clinic by using noninvasive fibrosis scoring systems. METHODS We conducted a retrospective study of patients with T2DM seen by our endocrinologists at the Medical Faculty Associates (MFA) Diabetes Center in Washington, DC, from November 1, 2021, until November 1, 2022. We included all subjects who were over 18 years old with a hemoglobin A1c (HbA1c) of 6.5 or higher. Patients with a history of significant alcohol consumption, decompensated cirrhosis, previous bariatric surgery, or prior chronic liver disease were excluded from the study. We identified patients at risk for hepatic fibrosis by using the Fibrosis-4 (FIB-4) Index, NAFLD Fibrosis Score (NFS) and AST to Platelet Ratio Index (APRI) when lab values were available. RESULTS A total of 1,411 patients were evaluated for T2DM by an endocrinology provider during the one-year period. Out of these, 336 patients met one or more of the exclusion criteria, leaving a total of 1075 patients included in the analysis. The majority were African American (n = 582, 54%), 261 were Caucasian (24.3%), and 85 were Hispanic (7.9%). Most patients were females (n = 675, 62.7%). The mean HbA1c was 8.1 ± 2.3. 643 patients (59.8%) were insulin dependent. Based on FIB-4 scores, we found that 35 (3.9%) patients had a score of > 2.67 associated with advanced fibrosis and 257 (29%) patients with scores of 1.3-2.67 had moderate fibrosis. Using the NFS calculator, there were 281 (28%) patients with values of > 0.675 consistent with F3-F4 disease. 715 (71.8%) patients with values of < 0.675 consistent with F0-F2 fibrosis. A total of 6(< 1%) patients met criteria for advanced fibrosis by APRI scoring. CONCLUSION In our urban Diabetes Center, utilizing the NFS calculator may detect many patients with advanced liver disease. Further research is needed to ensure the internal validity of the non-invasive tests in predicting liver fibrosis and to correlate these findings with transient elastography and other imaging evidence of fatty liver disease. CLINICAL TRIAL NUMBER Non-applicable.
Collapse
Affiliation(s)
- Ahmed Ebeid
- The George Washington University, Washington, DC, USA
| | - Fatma Mokhtar
- The George Washington University, Washington, DC, USA
| | | | - Susie Park
- Department of Medicine, The George Washington University Hospital, Washington, DC, USA
| | - Seta Degann
- Department of Medicine, The George Washington University Hospital, Washington, DC, USA
| | - Jeremy Payano
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Zahid Vahora
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Stephen Gray
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Lynt Johnson
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Diala El-Maouche
- Department of Endocrinology, The George Washington University Medical Faculty Associates, Washington, DC, USA
| | - Ameer Abutaleb
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA.
| |
Collapse
|
82
|
Jian Y, Li Y, Zhou Y, Mu W. Pollutants in Microenvironmental Cellular Interactions During Liver Inflammation Cancer Transition and the Application of Multi-Omics Analysis. TOXICS 2025; 13:163. [PMID: 40137490 PMCID: PMC11945810 DOI: 10.3390/toxics13030163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
This study categorizes pollutant-induced inflammation-cancer transition into three stages: non-alcoholic fatty liver disease (NAFLD), liver fibrosis, and hepatocellular carcinoma (HCC). It systematically reveals the temporal heterogeneity of pollutant-induced liver damage. The findings indicate that pollutants not only directly damage hepatocytes but also modulate key cells in the immune microenvironment, such as hepatic stellate cells (HSCs) and Kupffer cells, thereby amplifying inflammatory and fibrotic responses, ultimately accelerating the progression of HCC. Mechanistically, in the early stage (NAFLD), pollutants primarily cause hepatocyte injury through oxidative stress and lipid metabolism dysregulation. During the fibrosis stage, pollutants promote liver fibrosis by inducing extracellular matrix accumulation, while in the HCC stage, they drive tumorigenesis via activation of the Wnt/β-catenin pathway and p53 inactivation. Through multi-omics analyses, this study identifies critical pathogenic molecules and signaling pathways regulated by pollutants, providing new insights into their pathogenic mechanisms, potential biomarkers, and therapeutic targets. These findings offer valuable guidance for the development of diagnostic and therapeutic strategies for liver diseases and the formulation of environmental health risk prevention measures.
Collapse
Affiliation(s)
| | | | | | - Wei Mu
- School of Public Health, Center for Single-Cell Omics, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.J.); (Y.L.); (Y.Z.)
| |
Collapse
|
83
|
Vizioli G, Nicoletti A, Feliciani D, Funaro B, Zileri Dal Verme L, Ponziani FR, Zocco MA, Gasbarrini A, Gabrielli M. Immunotherapy and MASLD-Related HCC: Should We Reconsider the Role of Etiology in the Therapeutic Approach to HCC? APPLIED SCIENCES 2025; 15:2279. [DOI: 10.3390/app15052279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2025]
Abstract
Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancers and typically arises in the context of chronic liver disease. With the increasing prevalence of metabolic disorders, metabolic dysfunction-associated steatotic liver disease (MASLD) has become the leading cause of chronic liver disease and the most rapidly increasing cause of HCC. The role of dysfunctional innate and adaptive immune responses in the development and progression of HCC is well-established, prompting numerous trials to evaluate the efficacy of immune checkpoint inhibitors (ICIs) in targeting tumor cells. These trials have yielded promising results, and ICIs, in combination with anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, are now approved as first-line therapy for patients with metastatic or unresectable HCC, irrespective of the underlying liver disease. Notably, MASLD itself is characterized by immune system dysfunction, as metabolic inflammation plays a central role in its onset and progression. However, clinical studies and post-hoc analyses suggest that immunotherapy may be less effective in MASLD-associated HCC compared to viral-related HCC. This emerging evidence raises the question of whether the underlying liver disease influences the therapeutic response to ICIs in HCC. It may be time to consider tailoring therapeutic strategies for HCC based on the specific etiological, histological, and genotypical subgroups.
Collapse
Affiliation(s)
- Giuseppina Vizioli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alberto Nicoletti
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniela Feliciani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Barbara Funaro
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Zileri Dal Verme
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Assunta Zocco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maurizio Gabrielli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
84
|
Jin L, Cui Y, Zhang Q, Xiong Y, Zheng X, Ye J, Zhang A, Xiao Z, Zhuang Z, Liang G, Hu X, Luo W, Zhu W. The natural product-derived JM-9 alleviates high-fat diet-induced fatty liver in mice by targeting MD2. Int Immunopharmacol 2025; 148:114053. [PMID: 39827672 DOI: 10.1016/j.intimp.2025.114053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), is gradually emerging as one of the most prevalent liver diseases worldwide. Previous research demonstrated the involvement of myeloid differentiation factor 2 (MD2), a co-receptor of TLR4, as a key mediator in MASLD pathogenesis. The current study identifies JM-9 as a novel MD2 inhibitor, and focuses on evaluating its potential therapeutic effects in mitigating MASLD progression. METHODS Drug affinity responsive target stability (DARTS) assay and surface plasmon resonance assay were utilized to evaluate the MD2-targeting specificity of JM-9. In vitro, hepatocytes and macrophages were stimulated with palmitic acid (PA) followed by JM-9 treatment. In vivo, a high-fat diet (HFD)-induced MASLD model was established and subjected to JM-9 administration during the last 2 months. RESULTS JM-9 directly bound the Phe76 residue of MD2 to disrupt the PA-induced MD2/TLR4 complex formation, thus further restoring AMPK phosphorylation and inhibiting NF-κB activation to reducing lipid accumulation and inflammation, respectively. In the HFD-mediated MASLD mouse model, JM-9 alleviated the binding between MD2 and TLR4 in the liver and counteracted hepatic TBK1 and p65 activation and AMPK suppression, thereby mitigating liver inflammation, steatosis, and fibrosis. CONCLUSION JM-9, as a novel MD2 inhibitor, holds potential as a viable treatment option for MASLD by playing a dual role in regulating both lipid metabolism and inflammation response.
Collapse
Affiliation(s)
- Leiming Jin
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Yaqian Cui
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Qianhui Zhang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Yongqiang Xiong
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiangsheng Zheng
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiaxi Ye
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Anqi Zhang
- Department of Anesthesiology and Operation Room, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhongxiang Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| | - Zaishou Zhuang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Guang Liang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Xiang Hu
- Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Wu Luo
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| | - Weiwei Zhu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China.
| |
Collapse
|
85
|
Huang X, Wu M, Huang B, Zhang Y. Gastrointestinal adverse events associated with GLP-1 receptor agonists in metabolic dysfunction-associated steatotic liver disease (MASLD): a systematic review and meta-analysis. Front Med (Lausanne) 2025; 12:1509947. [PMID: 40051726 PMCID: PMC11882565 DOI: 10.3389/fmed.2025.1509947] [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: 11/18/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease, a prevalent chronic liver condition, can cause severe complications like hepatitis, cirrhosis, and hepatocellular carcinoma. In recent years, glucagon-like peptide-1 receptor agonists (GLP - 1RA) have shown unique therapeutic advantages and may become a preferred treatment for it. This meta-analysis aims to systematically examine GLP-1RA associated adverse events, providing a basis for guiding patient clinical management. Methods We conducted a search for randomized controlled trials (RCTs) investigating the therapeutic effects of GLP-1RA in the treatment of metabolic dysfunction-associated steatotic liver disease across four databases: PubMed, Embase, Web of Science, and Cochrane Library. The search period extended from the inception of each database until December 2023. Information pertaining to various adverse events was collected as outcome measures. Statistical analysis of the results and assessment of bias risk were conducted utilizing Review Manager (version 5.4.1) software. Results An analysis of 10 studies encompassing 960 participants revealed a significantly higher overall incidence of adverse events in the GLP-1RA group compared to the control group (OR: 2.40 [1.10, 5.26], P = 0.03). Subgroup analysis based on treatment duration demonstrated a higher rate of adverse events in the GLP-1RA group during follow-ups of less than 30 weeks (P = 0.0005, OR: 3.58 [1.75, 7.32]), but no statistical difference was observed between the two groups in follow-ups exceeding 30 weeks. There was no statistically significant difference between the two groups in adverse events leading to discontinuation (P = 0.29, OR: 1.47 [0.72, 2.98]). However, a notable difference was observed in gastrointestinal adverse events (P < 0.00001, OR: 4.83 [3.36, 6.95]). Conclusion GLP-1RA exhibits an overall higher incidence of adverse events in the treatment of metabolic dysfunction-associated steatotic liver disease, particularly in the gastrointestinal domain. Short-term use of GLP-1RA may be associated with a greater occurrence of adverse events, underscoring the importance of educating patients on preventive measures and establishing tolerance. However, there was no statistically significant difference between the two groups in severe adverse events and adverse events leading to discontinuation, confirming the safety profile of GLP-1RA application.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Miaohui Wu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Baoliang Huang
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yi Zhang
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
86
|
Liu T, Sui M, Tian M, Wu N, Zhao S, Wang Y, Yang Y, Ma S, Jiao D, Wang L, Feng Y, Zhang Y, Qin C, Liu C, Qi J, Zhu Q. Sulfonated albumin from hepatocytes accelerates liver fibrosis in nonalcoholic fatty liver disease through endoplasmic reticulum stress. Free Radic Biol Med 2025; 228:150-162. [PMID: 39743026 DOI: 10.1016/j.freeradbiomed.2024.12.055] [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: 09/19/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Posttranslational modifications (PTM) of albumin occur in liver diseases; however, little is known about the source and function of sulfonated albumin, a significant modification of albumin occurring in nonalcoholic fatty liver disease (NAFLD). We aimed to investigate the mechanism underlying sulfonated albumin production and its role in the progression of NAFLD-related liver fibrosis. METHODS Serum samples from healthy controls and patients with NAFLD were used to measure the proportion of sulfonated albumin. Mice models with NAFLD fed with high-fat diet (HFD) and methionine choline-deficient diet (MCD) were constructed. RNA sequencing, KEGG analysis, and GSEA were used to explore the mechanism of sulfonated albumin production and its mechanism of activating hepatic stellate cells (HSCs) and promoting the progression of liver fibrosis in NAFLD. RESULTS Sulfonated albumin levels increased significantly in both human and mouse NAFLD serum samples. In vivo studies in mice have shown that the intraperitoneal injection of sulfonated albumin promotes inflammation, hepatic steatosis, and liver fibrosis in NAFLD. In addition, autophagy has been verified as a key mechanism in the regulation of sulfonated albumin production. We also demonstrated that reactive oxygen species (ROS) production depends on the accumulation of damaged mitochondria and affects the production of sulfonated albumin under the regulation of autophagy. Hepatocyte-derived sulfonated albumin activates HSCs through the GAL3 receptor, thereby activating the endoplasmic reticulum (ER) stress pathway and promoting profibrotic activation of HSCs. CONCLUSIONS Our study demonstrated that sulfonated albumin activated HSCs through GAL3, thereby accelerating NAFLD-related liver fibrosis. Serum sulfonated albumin may be a potential diagnostic marker for liver fibrosis and an important target for the treatment of NAFLD-related liver fibrosis.
Collapse
Affiliation(s)
- Tiantian Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Minghao Sui
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Miaomiao Tian
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Nijin Wu
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Songbo Zhao
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yingchun Wang
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yinuo Yang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shujun Ma
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Deyan Jiao
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Le Wang
- Department of Health Care Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuemin Feng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yahui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Chengyong Qin
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Chenxi Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Jianni Qi
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Qiang Zhu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
87
|
Romaniuk-Drapała A, Kosicka-Noworzyń K, Sheng YH, Yohn C, Brunetti L, Kagan L. Evaluation of reference genes for qPCR in human liver and kidney tissue from individuals with obesity. Sci Rep 2025; 15:5347. [PMID: 39948154 PMCID: PMC11825690 DOI: 10.1038/s41598-025-87911-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: 10/07/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Given the obesity epidemic and the prevalence of comorbidities, there is an ongoing need to understand the health consequences of this disease state better. Understanding gene expression signals will facilitate the identification of mechanisms of kidney and liver dysfunction/disease often present in individuals with obesity. qPCR is the standard method for studying changes in relative gene expression. Reference genes (RGs) are obligatory for accurately normalizing mRNA transcript levels across samples. Despite the prevalence of qPCR, the reliability of the data is often compromised because RGs are still used without validation or have proven to be unstable in different tissues and various diseases. In this study, we validated seven reference genes (ACTB, B2M, RPLP0, HPRT1, GAPDH, 18S rRNA, and PPIA) using human liver tissue from 15 lean individuals and 17 individuals with a BMI ≥ 25 and human kidney tissue from 13 lean individuals and 15 individuals with a BMI ≥ 25. Cross-validation of expression stability was performed using the RefFinder platform with four algorithms: NormFinder, BestKeeper, geNorm, and the comparative ΔCt method. In obesity-related studies, the most suitable reference genes in gene expression studies are RPLP0 and HPRT1 in human kidney tissue and RPLP0 and GAPDH in the liver.
Collapse
Affiliation(s)
- Aleksandra Romaniuk-Drapała
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan, 60-806, Poland.
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| | - Katarzyna Kosicka-Noworzyń
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan, 60-806, Poland
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Yi-Hua Sheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Christine Yohn
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Luigi Brunetti
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
- Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
88
|
You B, Chen Z. Association of blood manganese and selenium levels with hepatic steatosis among adolescents: a nationwide cross-sectional analysis. Front Pediatr 2025; 13:1522219. [PMID: 40007873 PMCID: PMC11850372 DOI: 10.3389/fped.2025.1522219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Objective This study aimed to investigate the association between blood manganese and selenium levels and hepatic steatosis among adolescents, using data from the National Health and Nutrition Examination Survey (NHANES) 2017-2023. Methods A cross-sectional analysis was conducted using data from 2,459 adolescents (aged 12-19 years) with complete data on liver ultrasound transient elastography, blood manganese, and selenium levels. Hepatic steatosis was defined as a controlled attenuation parameter (CAP) score of ≥248 dB/m, a measure of liver steatosis, which is a primary characteristic and a less severe stage of hepatic steatosis, assessed by vibration-controlled transient elastography (VCTE). Multivariate logistic regression models were used to assess the associations between blood manganese and selenium levels and hepatic steatosis, while restricted cubic splines (RCS) were employed to examine the dose-response relationships. Results The mean age of the participants was 15.37 years, with 52.22% boy. Higher blood manganese and selenium levels were significantly associated with an increased prevalence of hepatic steatosis. In the fully adjusted model, adolescents in the highest quartile of blood manganese had more than twice the odds of hepatic steatosis compared to those in the lowest quartile (OR = 2.41, 95% CI: 1.55-3.75, P < 0.01). Similarly, the highest quartile of blood selenium was associated with a 57% increase in hepatic steatosis prevalence compared to the lowest quartile (OR = 1.57, 95% CI: 1.19-2.08, P < 0.01). RCS analysis confirmed a linear association between both blood manganese and selenium levels and hepatic steatosis prevalence. Subgroup analyses did not reveal statistically significant interactions by age, sex, or obesity status, although associations appeared stronger in younger adolescents. Conclusion Elevated blood manganese and selenium levels are associated with a higher prevalence of hepatic steatosis in adolescents. These findings suggest a potential role of trace elements in the development of hepatic steatosis, highlighting the need for further research to better understand the underlying mechanisms involved in liver fat accumulation in this population.
Collapse
Affiliation(s)
- Bin You
- Department of Pediatrics, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatrics, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
- Department of Pediatrics, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, Zhejiang, China
- Department of Pediatrics, The Third Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| | - Zhiyuan Chen
- Department of Pediatrics, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatrics, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
- Department of Pediatrics, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, Zhejiang, China
- Department of Pediatrics, The Third Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| |
Collapse
|
89
|
Lei Y, Tao S, Yang Y, Xie F, Xie W. Association between prognostic nutritional index and all-cause mortality and cardiovascular disease mortality in American adults with non-alcoholic fatty liver disease. Front Nutr 2025; 12:1526801. [PMID: 39996009 PMCID: PMC11847695 DOI: 10.3389/fnut.2025.1526801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background The current research was to investigate the relationship between prognostic nutritional index (PNI) and mortality, with a focus on all-cause and cardiovascular disease (CVD) mortality, for those with non-alcoholic fatty liver disease (NAFLD). Methods Data from 20,142 patients who participated in the National Health and Nutrition Examination Survey (NHANES), which was carried out between 2005 and 2014, were included in this research. To examine the relationship between PNI and both all-cause and cardiovascular mortality, we employed weighted Cox regression models with multiple variables. Kaplan-Meier survival curves were utilized to visualize the survival distribution across different levels of PNI. The non-linear association between PNI and mortality was addressed through penalized spline smoothing. Subgroup analyses were conducted to examine the potential influence of relevant clinical variables on the relationship between PNI and mortality. The precision of PNI in forecasting the outcome of survival was assessed as well using time-dependent receiver operating characteristic curve (ROC) analysis. Results Kaplan-Meier analysis linked higher PNI to significantly reduced all-cause and CVD mortality. Multivariable Cox models demonstrated that increasing PNI consistently lowered mortality risks. With a threshold value of 50.5, the link between PNI and mortality showed a non-linear pattern after adjusting for confounding factors. Subgroup analyses confirmed robust associations, particularly in race, education, BMI, and fibrosis. Time-dependent ROC analysis highlighted the strong predictive performance of PNI across various time points. Conclusion PNI played a significant role as an effective predictor of prognosis in individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Yuqing Lei
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Shaohong Tao
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yubo Yang
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, Foshan, Guangdong, China
| | - Fang Xie
- Department of Liver Disease, Jinling Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu, China
| | - Weining Xie
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong, China
| |
Collapse
|
90
|
Zeng T, Li F, Yang M, Wu Y, Cui W, Mou H, Luo X. Feasibility of Serum Galectin-1 as a Diagnostic Biomarker for Metabolic Dysfunction-Associated Steatotic Liver Disease: A Study on a Segment of the Chinese Population Using Convenience Sampling. Biomedicines 2025; 13:425. [PMID: 40002838 PMCID: PMC11853191 DOI: 10.3390/biomedicines13020425] [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: 12/20/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is commonly considered as a hepatic manifestation of metabolic syndrome, posing considerable public health and economic challenges due to its high prevalence. This study investigates the diagnostic potential of serum galectin-1 levels in MASLD patients. Methods: A total of 128 participants were analyzed for this study, comprising 68 healthy controls and 60 MASLD patients. The hepatic steatosis index (HSI) and fatty liver index (FLI) were calculated to evaluate the liver steatosis. Serum galectin-1 levels were measured using an enzyme-linked immunosorbent assay. We additionally conducted a comparative analysis of galectin-1 mRNA and protein expression levels in the liver tissue between the mouse models of MASLD, including ob/ob mice (n = 6), high-fat diet-fed C57 mice (n = 6), and the control group (n = 6). Results: Average serum galectin-1 levels significantly differed between groups, with lower values in the controls (p < 0.01). The frequency of MASLD increased with higher quartiles of galectin-1 levels (p < 0.01). The correlation analysis showed a positive relationship between serum galectin-1 and both HSI and FLI (p < 0.01). The multivariate logistic regression indicated that elevated galectin-1 was associated with an increased risk of MASLD (p < 0.01), yielding an area under the receiver operating characteristic curve for predicting MASLD at 0.745 (95% CI: 0.662-0.829). Hepatic galectin-1 levels were also elevated in the MASLD mouse model at both transcript and protein levels (p < 0.01). Conclusions: Serum galectin-1 can be used as a potential biomarker to help diagnose MASLD.
Collapse
Affiliation(s)
- Ting Zeng
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Fang Li
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Min Yang
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Yao Wu
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Wei Cui
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| | - Huaming Mou
- Department of Cardiovascular Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| | - Xiaohe Luo
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| |
Collapse
|
91
|
Havranek B, Loh R, Torre B, Redfield R, Halegoua-DeMarzio D. Glucagon-like peptide-1 receptor agonists improve metabolic dysfunction-associated steatotic liver disease outcomes. Sci Rep 2025; 15:4947. [PMID: 39930071 PMCID: PMC11811119 DOI: 10.1038/s41598-025-89408-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of chronic liver disease and is associated with significant cardiovascular morbidity and mortality. This study aims to investigate the association of glucagon-like peptide-1 (GLP-1) agonists with major cardiovascular events, clinically significant portal hypertension events, and all-cause mortality in patients with MASLD. A large, population-based retrospective cohort study was conducted using the TriNetX platform, which provided real-time access to electronic health records of 634,265 adult patients with MASLD/MASH. Propensity score matching (PSM) was employed to create two cohorts: A GLP-1 agonists group and a control group without GLP-1 agonists usage. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using Cox proportional hazards models along with Kaplan-Meier survival analyses to estimate outcomes at the end of 1, 3, 5, and 7 years. After PSM, 6,243 patients were included in each group. The GLP-1 agonist group had significantly lower risk of heart failure (at 7 years, HR, 0.721; 95% Cl, 0.593-0.876), composite cardiovascular events (at years 7, HR, 0.594; 95% Cl, 0.475-0.745), clinically significant portal hypertension events (at 7 years, HR, 0.463; 95% Cl, 0.348-0.611), and all-cause mortality (at 7 years, HR, 0.303; 95% Cl, 0.239-0.385). These results were consistent at 1-, 3-, 5-, and 7-years post index event. GLP-1 agonists usage in patients with MASLD is associated with reduced risk of major cardiovascular events, clinically significant portal hypertension, and all-cause mortality. These findings highlight the potential of GLP-1 agonists in MASLD/MASH management, warranting further prospective studies.
Collapse
Affiliation(s)
- Brandon Havranek
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Rebecca Loh
- Department of Gastroenterology & Hepatology, Thomas Jefferson University Hospital, 132 S 10th St, Suite 480, Philadelphia, PA, 19107, USA
| | - Beatriz Torre
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, 19107, USA
| | - Rachel Redfield
- Department of Gastroenterology & Hepatology, Thomas Jefferson University Hospital, 132 S 10th St, Suite 480, Philadelphia, PA, 19107, USA
| | - Dina Halegoua-DeMarzio
- Department of Gastroenterology & Hepatology, Thomas Jefferson University Hospital, 132 S 10th St, Suite 480, Philadelphia, PA, 19107, USA.
| |
Collapse
|
92
|
Byrne CD, Dore GJ. Metabolic dysfunction-associated steatotic liver disease a multisystem disease: Assessing the cost effectiveness of pharmacotherapies. J Hepatol 2025:S0168-8278(25)00075-3. [PMID: 39938652 DOI: 10.1016/j.jhep.2025.02.001] [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: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/14/2025]
Affiliation(s)
- Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK.
| | - Gregory J Dore
- Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
93
|
da Silva Pereira ENG, Franco RLC, Santos RDCD, Daliry A. Statins and non-alcoholic fatty liver disease: A concise review. Biomed Pharmacother 2025; 183:117805. [PMID: 39755024 DOI: 10.1016/j.biopha.2024.117805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common hepatic manifestation of metabolic syndrome affecting 20-30 % of the adult population worldwide. This disease, which includes simple steatosis and non-alcoholic steatohepatitis, poses a significant risk for cardiovascular and metabolic diseases. Lifestyle modifications are crucial in the treatment of NAFLD; however, patient adherence remains challenging. As there is no specific treatment, drug repositioning is being researched as an alternative strategy. Statins, which are known for their cholesterol-lowering effects, are considered potential interventions for NAFLD. This review aimed to present the current understanding of the effects of statins on liver physiology in the context of NAFLD. The pathophysiology of NAFLD includes steatosis, inflammation, and fibrosis, which are exacerbated by dyslipidemia and insulin resistance. Statins, which inhibit 3-hydroxy-3-methylglutaryl-CoA reductase, have pleiotropic effects beyond cholesterol-lowering and affect pathways related to inflammation, fibrogenesis, oxidative stress, and microcirculation. Although clinical guidelines support the use of statins for dyslipidemia in patients with NAFLD, more studies are needed to demonstrate their efficacy in liver disease. This comprehensive review serves as a foundation for future studies on the therapeutic potential of statins in NAFLD.
Collapse
Affiliation(s)
| | - Rafaela Luiza Costa Franco
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Rafaele Dantas Cruz Dos Santos
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Anissa Daliry
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
94
|
He Y, Ye M, Xia Y, Zhong Z, Li Q. Antioxidants and the risk of metabolic dysfunction-associated steatotic liver disease: results of National Health and Nutrition Examination Survey and two-sample Mendelian randomization analyses. Eur J Gastroenterol Hepatol 2025; 37:230-239. [PMID: 39621882 DOI: 10.1097/meg.0000000000002898] [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: 12/22/2024]
Abstract
BACKGROUND The link between antioxidants and metabolic dysfunction-associated steatotic liver disease (MASLD) is a topic of considerable discussion in the field of observational studies, with the exact causal connections still being unclear. METHODS In this investigation, a cohort consisting of 17 061 participants from the National Health and Nutrition Examination Surveys was studied. Initially, a cross-sectional analysis was carried out to examine the relationship between the CDAI and MASLD. Further, Mendelian randomization (MR) was utilized to assess the possible causal links between antioxidant levels in the bloodstream and MASLD. RESULTS The association between the CDAI and MASLD was found to be significant in the fully adjusted logistic regression model, showing an OR of 0.95 [95% confidence interval (CI): 0.94-0.97; P < 0.001]. The use of restricted cubic spline regression revealed no significant nonlinear association between the CDAI and the occurrence of MASLD ( Pnonlinearity = 0.321). Additionally, MR findings did not suggest any causal connections between circulating levels of various antioxidants and MASLD. These antioxidants included vitamin A (retinol) (IVW: OR: 0.67, 95% CI: 0.33-1.36, P = 0.272), vitamin C (ascorbate) (IVW: OR: 0.61, 95% CI: 0.34-1.09, P = 0.094), vitamin E (α-tocopherol) (IVW: OR: 0.55, 95% CI: 0.13-2.25, P = 0.407), vitamin E (γ-tocopherol) (IVW: OR: 0.89, 95% CI: 0.36-2.23, P = 0.806), zinc (IVW: OR: 0.95, 95% CI: 0.82-1.09, P = 0.449), selenium (IVW: OR: 0.98, 95% CI: 0.84-1.16, P = 0.855), and carotene (IVW: OR: 0.80, 95% CI: 0.36-1.81, P = 0.596). CONCLUSION The findings highlight a significant negative linear relationship between CDAI and MASLD prevalence in the observational component of the study. However, the MR analysis did not indicate any causal effects of circulating antioxidant levels on MASLD.
Collapse
Affiliation(s)
- Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
95
|
Luo J, Yang Y, Liu H, Tan Z, Chen C, Li W, Yang R. Ellagic acid alleviates high-fructose diet-induced non-alcoholic fatty liver disease by modulating liver metabolic profiles and gut microbiota. Int J Food Sci Nutr 2025; 76:47-61. [PMID: 39627026 DOI: 10.1080/09637486.2024.2435849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 02/12/2025]
Abstract
This study integrated analyses of gut microbiota and metabolomics to investigate the impact of ellagic acid (EA) on non-alcoholic fatty liver disease (NAFLD). Compared to the high-fructose diet (HFruD) group, the EA group exhibited reduced body weight and fat mass, alongside improvements in blood glucose and lipid metabolism. Liver metabolomics analysis revealed that EA increased the abundance of metabolites in pathways related to unsaturated fatty acids, amino acids and bile acids. Furthermore, EA induced alterations in the composition and structure of gut microbiota, notably decreasing bacterial genera enriched by HFruD while promoting beneficial bacteria such as Faecalibaculum. Correlation analysis demonstrated significant associations among NAFLD markers, gut microbiota and liver metabolites influenced by EA. This study provides new insights into the anti-NAFLD effects of EA, suggesting EA as a promising nutraceutical for improving NAFLD.
Collapse
Affiliation(s)
- Jinxin Luo
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yuzhe Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Hui Liu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Zhaolun Tan
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Chunlian Chen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Wu Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Ruili Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
96
|
Yamashita S, Takase H, Kawakatsu N, Hayashi K, Kin F, Isogaki T, Dohi Y. Fibrosis-4 index is closely associated with future development of hypertension in the Japanese general population. Hypertens Res 2025; 48:796-804. [PMID: 39639128 DOI: 10.1038/s41440-024-02028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024]
Abstract
The fibrosis-4 index, a noninvasive method for evaluating liver fibrosis, is closely associated with cardiovascular events. In this study, we tested the hypothesis that the fibrosis-4 index is associated with new-onset hypertension in the general population. A total of 15,502 individuals (51.0 ± 13.2 years) who participated in our health checkup program were screened. Participants with hypertension were excluded, and the remaining 8719 normotensive participants (48.4 ± 12.6 years) were followed up (median 1739 days) with the endpoint of the new onset of hypertension. During follow-up, 1750 participants (39.0 per 1000 person-years) developed hypertension. In Kaplan-Meier analysis, where participants were divided into three groups according to the fibrosis-4 index at baseline (low, <1.30; intermediate, 1.30-2.67; high, ≥2.67), the risk of hypertension increased with increasing fibrosis-4 index (low, 33.8; intermediate, 55.2; high, 69.4 per 1000 person-years). Multivariate Cox hazard regression analysis revealed that the log-transformed fibrosis-4 index was independently associated with the development of new hypertension (hazard ratio 4.279, 95% confidence interval 3.318-5.518). These results suggest that the fibrosis-4 index is a useful tool to evaluate a risk of developing hypertension in the general population. Hypertension and liver fibrosis may share a common basis.
Collapse
Affiliation(s)
- Sumiyo Yamashita
- Department of Cardiology, Nagoya City University Mirai Kousei Hospital, Nagoya, Japan
| | - Hiroyuki Takase
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan.
| | - Naomi Kawakatsu
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Kazusa Hayashi
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Fumihiko Kin
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Takeru Isogaki
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Yasuaki Dohi
- Division of Internal Medicine, Faculty of Rehabilitation Sciences, Nagoya Gakuin University, Nagoya, Japan
| |
Collapse
|
97
|
Zhang J, Ran S, Wei S, Tian F, Chen L, Yang Z, Chen G, Lin H. Associations of MAFLD subtypes and air pollutants with multi-system morbidity and all-cause mortality: A prospective cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117893. [PMID: 39955868 PMCID: PMC11860302 DOI: 10.1016/j.ecoenv.2025.117893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) and air pollution are both significant health concerns. However, their combined effects on multi-system morbidity and all-cause mortality remain poorly understood. METHODS We analyzed data from 434,417 UK Biobank participants, categorizing them into four groups: non-MAFLD, MAFLD-diabetes, MAFLD-lean, and MAFLD-overweight/obesity. To evaluate the long-term effects of air pollution exposure, we used time-varying Cox proportional hazard models to assess four air pollutants: particulate matter with an aerodynamic diameter < 2.5 μm (PM2.5), PM10, nitrogen dioxide (NO2), and nitrogen oxides (NOx). We examined the associations between these air pollutants, MAFLD subtypes, and their joint impact on multi-system morbidity and all-cause mortality. Furthermore, we explored the additive and multiplicative interactions between air pollutants and MAFLD subtypes. RESULTS At baseline, 15,325 participants were classified as MAFLD-diabetes, 3341 as MAFLD-lean, and 140,934 as MAFLD-overweight/obesity. Among these groups, MAFLD-diabetes was most strongly associated with adverse outcomes compared to other subtypes. Air pollution exposure had a synergistic effect on cirrhosis risk across all MAFLD subtypes, with the most pronounced effects observed for PM2.5 [relative excess risk due to interaction (RERI): 2.10 (0.94, 3.26)] and NO2 [RERI:1.85 (0.67, 3.04)] in MAFLD-lean group. Positive additive and multiplicative interactions between air pollutants and MAFLD subtypes were also observed for coronary artery disease (CAD), with the exception of nitrogen oxide in the MAFLD-lean group. Additionally, only the MAFLD-diabetes demonstrated significant positive additive interactions with all four air pollutants in relation to chronic kidney disease (CKD). CONCLUSIONS This study highlights the distinct impacts of MAFLD subtypes on multi-system morbidity and all-cause mortality, underscoring the critical need for targeted prevention and treatment strategies, particularly for individuals with MAFLD-diabetes. Our findings reveal significant additive and synergistic effects of air pollution exposure on the risks of cirrhosis, CAD, and CKD among MAFLD patients.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shanshan Ran
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shengtao Wei
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Fei Tian
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lan Chen
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zijun Yang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ge Chen
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
98
|
Basaly V, Bhattacharya A, Guo GL. Insights of direct and indirect regulation of PXR through phosphorylation in fatty liver disease. Mol Pharmacol 2025; 107:100014. [PMID: 40023513 DOI: 10.1016/j.molpha.2024.100014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 03/04/2025] Open
Abstract
The pregnane X receptor (PXR), a ligand-activated nuclear receptor, regulates the transcription of several genes that encode many enzymes and transporters related to drug metabolism. PXR also performs an important role as a physiological sensor in the modulation of endobiotic metabolism for hormones, bile acids, cholesterol, fatty acids, and glucose. Dysregulation of these PXR-mediated pathways is implicated in the progression of metabolic dysfunction-associated steatohepatitis (MASH), contributing to the complex interplay of factors involved in chronic liver disease development and exacerbation affecting millions worldwide. This review highlights the current knowledge of PXR expression and its role in endobiotic metabolism related to MASH development, which is associated with diverse causes and dire outcomes. This review focuses on elucidating the molecular pathways associated with PXR activation directly or indirectly and PXR interaction with other regulatory factors. Although there is still much to comprehend about the intricate details of these pathways, the conclusion is drawn that PXR exerts a crucial role in the pathological and physiological pathways of hepatic cellular processes, which holds promise as a potential pharmacological target for exploring novel therapeutic approaches for MASH treatment and/or prevention. SIGNIFICANCE STATEMENT: The pregnane X receptor (PXR) plays a fundamental role in regulating gene expression involved in xenobiotic and endobiotic metabolism. Dysregulation of PXR-mediated pathways is related to the development of metabolic dysfunction-associated steatohepatitis. The ligand-independent pathways regulating PXR hepatic functions through phosphorylation shed light on possible indirect molecular mechanisms and pathways that regulate PXR activity and function. Understanding these pathways may provide insight into new pharmaceutical interventions for metabolic dysfunction-associated steatohepatitis development.
Collapse
Affiliation(s)
- Veronia Basaly
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Anisha Bhattacharya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Rutgers Center for Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey.
| |
Collapse
|
99
|
Mounika N, Mungase SB, Verma S, Kaur S, Deka UJ, Ghosh TS, Adela R. Inflammatory Protein Signatures as Predictive Disease-Specific Markers for Non-Alcoholic Steatohepatitis (NASH). Inflammation 2025; 48:25-41. [PMID: 38676759 DOI: 10.1007/s10753-024-02035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic disease worldwide, consisting of a broad spectrum of diseases such as simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Hepatic inflammation plays a key role in the pathophysiology of NAFLD. Inflammatory mediators such as cytokines and chemokines are considered as contributing factors to NAFLD development and progression. In the present study, we aimed to investigate the inflammatory protein signatures as predictive disease-specific markers for non-alcoholic fatty liver disease (NAFLD). This cross-sectional study included healthy control (n = 64), NAFL (n = 109), and NASH (n = 60) human subjects. Serum concentrations of various cytokines and chemokines were evaluated using sensitive multiplex assays. We used principal component analysis (PCoA) to reveal distinct differences in the levels of cytokines and chemokines between each of the study groups. Further, a random forest classification model was developed to identify the panel of markers that could predict diseases. The protein-protein network analysis was performed to determine the various signaling pathways associated with the disease-specific panel of markers. Serum concentrations of TNF-α, IL-1β, IL-1ra, G-CSF, PDGF-BB, MCP-1, MIP-1a, MIP-1b, RANTES, eotaxin, IL-8 and IP-10 were significantly increased in NASH group as compared to control group. Furthermore, serum concentrations of IL-9 and IL-13 were significantly lower in the NASH group, whereas IL-2 levels were significantly decreased in the NAFL group when compared to the control group. PCoA results demonstrated statistically significant differences in cytokines and chemokines between each of the study groups (PERMANOVA p = 0.001; R2 = 0.102). RANTES, IL-1ra, MIP-1b, IL-2, and G-CSF could differentiate the NAFL group from the controls; G-CSF, IL-1ra, TNF-α, RANTES, and IL-9 could differentiate the NASH group from the controls; and G-CSF, IL-9, IL-13, eotaxin, and TNF- α could differentiate the NASH group from the NAFL group. Our protein-protein network revealed that these markers are involved in cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, TNF, chemokine, JAK/STAT, P13K/Akt, TLR, NOD-like receptor, NF-kB, and adipocytokine signaling pathways which might be responsible for disease pathogenesis. Our study findings revealed a set of distinct cytokine and chemokine markers and they might be considered as biomarkers in distinguishing NASH from NAFL. Future multicentre studies with larger sample size are recommended to determine the potential utility of these panels of markers.
Collapse
Affiliation(s)
- Nadella Mounika
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Suraj Bhausaheb Mungase
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Shivangi Verma
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver & Biliary Science (ILBS), New Delhi-110 070, Vasant Kunj, India
| | - Utpal Jyoti Deka
- Department of Gastroenterology, Downtown Hospital, GS Road, Bormotoria, Guwahati, Assam-781006, India
| | - Tarini Shankar Ghosh
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India.
| |
Collapse
|
100
|
Kaffas AE, Bhatraju KC, Vo-Phamhi JM, Tiyarattanachai T, Antil N, Negrete LM, Kamaya A, Shen L. Development of a Deep Learning Model for Classification of Hepatic Steatosis from Clinical Standard Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:242-249. [PMID: 39537545 DOI: 10.1016/j.ultrasmedbio.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Early detection and monitoring of hepatic steatosis can help establish appropriate preventative measures against progression to more advanced disease. We aimed to develop a deep learning (DL) program for classification of hepatic steatosis from standard-of-care grayscale ultrasound (US) images. METHODS In this single-center retrospective study, we utilized grayscale US images from January 1, 2010, to October 23, 2022, labeled with magnetic resonance imaging (MRI) proton density fat fraction (MRI-PDFF) to develop a DL multi-instance program for differentiating normal (S0) from steatotic liver (S1/2/3) and normal/mild steatosis (S0/1) from moderate/severe steatosis (S2/3). Diagnostic performances were assessed with area under the receiver operating characteristic curves (AUC), sensitivity, specificity and balanced accuracy with 95% confidence interval (CI). RESULTS A total of 403 patients with 403 US exams were included: 171 (42%) were normal (S0: MRI-PDFF <5%), 154 (38%) had mild steatosis (S1: MRI-PDFF 5-17.4%), 29 (7%) had moderate steatosis (S2: MRI-PDFF >17.4%-22.1%) and 49 (12%) had severe steatosis (S3: MRI-PDFF >22.1%). The dataset was split to include 322 patients in train/validation and 81 patients in a holdout test set (kept blind). The S0 versus S1/2/3 model achieved 81.3% (95% CI 72.1-90.5) AUC, 81.1% (70.6-91.6) sensitivity, 71.4% (54.7-88.2) specificity and 76.3% (66.4-86.2) balanced accuracy. The S0/1 versus S2/3 model achieved 95.9% (89-100) AUC, 87.5% (71.3-100) sensitivity, 96.9% (92.7-100) specificity and 92.2% (83.8-100) balanced accuracy. A multi-class model achieved a sensitivity of 71.4% (54.7-88.2) for S0, 67.6% (52.5-82.7) for S1 and 87.5% (71.3-100) for S2/3; specificity for the same model was 81.1% (70.6-91.6) for S0, 77.3% (64.9-89.7) for S1 and 96.9% (92.7-100) for S2/3. CONCLUSION Our DL program offered high sensitivity and accuracy in detecting and categorizing hepatic steatosis from standard-of-care ultrasound.
Collapse
Affiliation(s)
- Ahmed El Kaffas
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Krishna Chaitanya Bhatraju
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jenny M Vo-Phamhi
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thodsawit Tiyarattanachai
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Neha Antil
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lindsey M Negrete
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aya Kamaya
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Luyao Shen
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|