1
|
Krag A, Torp N, Israelsen M. Letter to the Editor: Alcohol a confounding factor in the assessment of resmetirom therapy. Hepatology 2025; 81:E162-E163. [PMID: 39960323 DOI: 10.1097/hep.0000000000001266] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 05/20/2025]
Affiliation(s)
- Aleksander Krag
- Department of Gastroenterology and Hepatology, Centre for Liver Research, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Nikolaj Torp
- Department of Gastroenterology and Hepatology, Centre for Liver Research, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Israelsen
- Department of Gastroenterology and Hepatology, Centre for Liver Research, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
2
|
Zoncapè M, Tsochatzis EA. Liver Fibrosis Testing in Patients With Type 2 Diabetes: The Time Is Now. Diabetes Care 2025; 48:871-873. [PMID: 40392997 DOI: 10.2337/dci25-0008] [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: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/22/2025]
Affiliation(s)
- Mirko Zoncapè
- Sheila Sherlock Liver Centre, Royal Free Hospital, London, U.K
- UCL Institute of Liver and Digestive Health, University College London, London, U.K
| | - Emmanuel A Tsochatzis
- Sheila Sherlock Liver Centre, Royal Free Hospital, London, U.K
- UCL Institute of Liver and Digestive Health, University College London, London, U.K
| |
Collapse
|
3
|
Zhao Y, Bo Y, Zu J, Xing Z, Yang Z, Zhang Y, Deng Y, Liu Y, Zhang L, Yuan X, Wang Y, Henry L, Ji F, Nguyen MH. Global Burden of Chronic Liver Disease and Temporal Trends: A Population-Based Analysis From 1990 to 2021 With Projections to 2050. Liver Int 2025; 45:e70155. [PMID: 40421876 DOI: 10.1111/liv.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/30/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND AND AIMS Globally, the aetiology and epidemiology of chronic liver disease (CLD) are undergoing significant changes. We aimed to investigate the updated global burden of CLD, evaluate the cross-country inequalities, and provide 2050 predictions. METHODS Using the Global Burden of Disease Study (GBD) 2021 data resources, we analysed and forecasted CLD prevalence, incidence, and related death from 1990-2021 to 2050, respectively. We calculated average annual percent change (AAPC) by joinpoint regression model and quantified inequalities according to World Health Organisation-recommended health equity standards. RESULTS In 2021, the number of prevalent, incident CLD and related deaths globally were 1.7 billion (95% uncertainty interval (UI): 1.6-1.8), 58.4 million (95% UI: 54.2-62.8) and 1.4 million (95% UI: 1.3-1.5), respectively. During 1990-2021, the age-standardised incidence rate (ASIR) increased, especially in those aged 15-49 (AAPC: 0.49%; 95% confidence interval [CI]: 0.45%-0.53%), in Europe (AAPC: 0.41%; 95% CI: 0.41%-0.42%) and the Americas (AAPC: 0.41%; 95% CI: 0.39%-0.42%), whereas the age-standardised death rate (ASDR) decreased globally (AAPC: -1.26%; 95% CI: -1.35% [-1.17%]) and across subgroups. During 1990-2021, the ASIR of metabolic dysfunction-associated steatotic liver disease (MASLD) increased the most in those aged 15-49 (AAPC: 0.72%; 95% CI: 0.67%-0.77%) and in the Western Pacific region (AAPC: 0.73%; 95% CI: 0.59%-0.86%). Socio-demographic index (SDI)-related inequalities decreased for the age-standardised prevalence rate (ASPR) and ASIR of CLD but increased for ASDR, placing a disproportionately heavier burden on low-SDI countries. From 2022 to 2050, the ASIR of CLD is projected to increase (AAPC: 0.20%; 95% CI: 0.19%-0.20%), but the ASDR is projected to decline (AAPC: -1.91%; 95% CI: -1.96% [-1.85%]). CONCLUSIONS This study's findings highlight targeted interventions for CLD disparities, focusing on MASLD management, the younger population (15-49 years), and socio-demographic inequalities.
Collapse
Affiliation(s)
- Yunyu Zhao
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yajing Bo
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Zu
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, China
| | - Zixuan Xing
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhanpeng Yang
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, China
| | - Yue Zhang
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, China
| | - Yujiao Deng
- Division of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Liu
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lanting Zhang
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Yuan
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan Wang
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Linda Henry
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University Medical Center, Palo Alto, California, USA
| | - Fanpu Ji
- Department of Hepatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi, China
- Global Health Institute, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center of Infectious Diseases, Xi'an, China
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University Medical Center, Palo Alto, California, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
| |
Collapse
|
4
|
Díaz LA, Tavaglione F, Mittal N, Bettencourt R, Amangurbanova M, Johnson A, Marti-Aguado D, Tincopa M, Loomba R, Khan-Riches A, Madamba E, Siddiqi H, Richards L, Sirlin CB, Ajmera V, Loomba R. Noninvasive pathway for stratifying fibrosis in suspected metabolic dysfunction and alcohol-associated liver disease (MetALD). Hepatol Commun 2025; 9:e0718. [PMID: 40377491 PMCID: PMC12088636 DOI: 10.1097/hc9.0000000000000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/13/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Metabolic dysfunction and alcohol-associated liver disease (MetALD) may increase liver fibrosis progression, but data on screening are scarce. We aimed to assess the performance of noninvasive tests (NITs) for detecting significant fibrosis in individuals with suspected MetALD. METHODS This is a cross-sectional study of prospectively enrolled adults identified as overweight or obese. We included adults with suspected MetALD defined by ≥1 of 5 cardiometabolic criteria and self-reported alcohol use within MetALD ranges or lower self-reported alcohol use but with phosphatidylethanol (PEth) levels ≥25 ng/mL. Clinical assessment included contemporaneous magnetic resonance elastography (MRE) and vibration-controlled transient elastography (VCTE). Significant fibrosis was defined as MRE ≥3.14 kPa (or VCTE ≥7.6 kPa if MRE was missing). Analyses included AUROCs. RESULTS Among 617 individuals screened, we identified 97 (15.7%) with suspected MetALD. The mean age was 50.6±12.8 years, 67% were men, the mean body mass index was 31.4±6.5 kg/m2, 12.4% had diabetes, and 8% had significant fibrosis. Fibrosis-4 ≥1.3 demonstrated good performance for significant fibrosis (AUROC: 0.78, 95% CI: 0.58-0.98, sensitivity 80%, specificity 76%, positive predictive value 17%, and negative predictive value 98%). VCTE ≥8 kPa also had good performance (AUROC: 0.85, 95% CI: 0.66-1.00, sensitivity 80%, specificity 91%, positive predictive value 36%, and negative predictive value 99%). A stepwise approach using fibrosis-4 followed by VCTE yielded a low false negative rate (2% misclassified as low risk). CONCLUSIONS A clinical care algorithm utilizing a stepwise approach with fibrosis-4 and VCTE shows adequate performance in detecting significant fibrosis in individuals with suspected MetALD.
Collapse
Affiliation(s)
- Luis Antonio Díaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Federica Tavaglione
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Nikita Mittal
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Ricki Bettencourt
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Maral Amangurbanova
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Amy Johnson
- Liver Unit, Department of Medicine, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - David Marti-Aguado
- Digestive Disease Department, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Monica Tincopa
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Ria Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Asma Khan-Riches
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Egbert Madamba
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Harris Siddiqi
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Lisa Richards
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Claude B. Sirlin
- Liver Imaging Group, Department of Radiology, University of California at San Diego, La Jolla, California, USA
| | - Veeral Ajmera
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, California, USA
- School of Public Health, University of California at San Diego, La Jolla, California, USA
| |
Collapse
|
5
|
Shu R, Tian S, Qu W, Yang J, Shi W, Li X, Zou T, Jiang C, Zhang Y, Yang Z, Tian H, Yang H, Fu J, She ZG, Li H, Zhang XJ. Hepatoprotective drug screening identifies daclatasvir a promising therapeutic candidate for MASLD by targeting PLIN2. J Lipid Res 2025:100835. [PMID: 40449734 DOI: 10.1016/j.jlr.2025.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 05/22/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) has become global health challenges with limited therapeutic strategy. Here, the present study aims to identify promising drug candidates for MASH and to clarify their pharmacological mechanism. By an extensive screening of FDA-approved hepatoprotective medicines using a PA/OA-stimulated hepatocytes model, we identified daclatasvir showing potent anti-MASH capacity against hepatic steatosis deposition and inflammatory response. The hepatoprotective benefits of daclatasvir was further validated in MASH mouse models, induced by a high-fat high-cholesterol (HFHC) diet for 16 weeks or a methionine-choline-deficient (MCD) diet for 4 weeks, as supported by markedly improved histopathological characteristics, serum biochemical level, and transcriptomic analyses. Using the molecular docking assay followed by isothermal titration calorimetry confirmation, we identified that daclatasvir functions as a new perilipin-2 (PLIN2) inhibitor by interrupting its stability. In specific, PLIN2 subjected to MARCH6-mediated protein degradation in a K11-type ubiquitination. Daclatasvir can directly bind to PLIN2 and enhance its interaction with MARCH6, leading to markedly strengthened PLIN2 ubiquitinational degradation and the subsequent decline in lipid droplet disintegration and lipotoxicity. The specific mutation at the binding amino acid sites of PLIN2 with daclatasvir largely abolished the anti-MASH benefit of daclatasvir. In conclusion, the findings of our present study for the first time identified the anti-HCV drug daclatasvir as a novel and potent PLIN2 protein degradant for MASH protection.
Collapse
Affiliation(s)
- Rui Shu
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Song Tian
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Weiyi Qu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinjie Yang
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China; Qujing Medical College Basic Medical Department, Qujing, China
| | - Wei Shi
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Xinyan Li
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Toujun Zou
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Changjin Jiang
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Yuxuan Zhang
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Zifeng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Han Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hailong Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Jiajun Fu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Zhi-Gang She
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongliang Li
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Xiao-Jing Zhang
- Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute; School of Pharmacy, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
6
|
Wang JP, Wang JY, Sun PQ, Wang XW, Yuan ZT, Cao Q, Pan SM, Jiang YY. Association between weight fluctuation and the risk of metabolic dysfunction-associated steatotic liver disease. World J Hepatol 2025; 17:103852. [DOI: 10.4254/wjh.v17.i5.103852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/24/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND The global incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) has increased in recent years. It has already been demonstrated that exercise and weight change are associated with the occurrence of MASLD; however, the association between weight fluctuation caused by different exercise intensities and the risk of MASLD remains to be studied.
AIM To investigate the impact of weight fluctuation and physical activity intensity on the risk of MASLD prevalence.
METHODS Data from the National Health and Nutrition Examination Survey database including five cycles from 2009 to 2018 were analyzed. The model included variables such as age, sex, and poverty income ratio. Weighted multivariate logistic regression was used to examine the influence of different weight fluctuation patterns within the two time intervals on the prevalence of MASLD. Nonparametric restricted cubic spline curves were used to analyze the non-linear relationship between net weight change and MASLD prevalence.
RESULTS Among 3183 MASLD cases, the risk of MASLD increased with age for individuals transitioning from non-obese to obese or maintaining obesity, with odds ratio (OR) changing from 8.91 (95%CI: 7.40–10.88) and 11.87 (95%CI: 9.65–14.60) at 10 years before baseline to 9.58 (95%CI: 8.08–11.37) and 12.51 (95%CI: 9.33-16.78) at 25 years. Stable obesity correlated with age-dependent MASLD prevalence escalation, whereas increased physical activity attenuated MASLD risk in this group, with an OR changing from 13.64 (95%CI: 10.59–17.57) to 6.42 (95%CI: 4.24–9.72). Further analysis of the net weight changes revealed a paradoxical risk elevation with intensified physical activity during different time periods.
CONCLUSION The risk of MASLD increases in individuals transitioning from non-obese to obese or maintaining obesity. High-intensity physical activity is beneficial for MASLD among individuals with stable obesity.
Collapse
Affiliation(s)
- Jin-Ping Wang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Jia-Yang Wang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Pei-Qi Sun
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Xue-Wei Wang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Ze-Ting Yuan
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Qin Cao
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Shu-Ming Pan
- Department of Emergency, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yuan-Ye Jiang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| |
Collapse
|
7
|
Lian K, Fan Q, Sheng S, Zhang K, Sun X, Kan C, Pan R, Guo Z. Metabolic Dysfunction-Associated Steatotic Liver Disease and Chronic Kidney Disease: Unraveling Connections and Advancing Therapies. BRATISL MED J 2025. [DOI: 10.1007/s44411-025-00189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 06/02/2025]
|
8
|
Chen X, Wan Z, Wu L, Cai X, Si H, Liu X, Zhao Q, Xu F, Deng H. CiR-EIS alleviates metabolic dysfunction-associated steatohepatitis by modulating macrophage polarization involving the miR-548m/IGF1 axis. Clin Nutr 2025; 50:104-116. [PMID: 40409233 DOI: 10.1016/j.clnu.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/22/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is a significant public health concern, with macrophage phenotypes implicated in its progression. Although extensive inflammation-suppressing circular RNA (ciR-EIS, formerly named as hsa_circ_0008882) has been implicated in inflammation regulation, its role in macrophage polarization within the context of MASH remains unexplored. This study aimed to clarify the effect of ciR-EIS on macrophage polarization in MASH. METHODS Immunofluorescence-fluorescence in situ hybridization was used to evaluate the localization of ciR-EIS in human liver sections. THP-1-derived macrophages (TDMs) were utilized to study ciR-EIS functions in vitro. Flow cytometry and RT-qPCR were employed to evaluate macrophage polarization after transfection. Bodipy assay was used to measure lipid buildup in HepG2 cells. Immunohistochemistry was used to confirm liver insulin-like growth factor 1 (IGF1) expression. Retrospective clinical records were analyzed to examine the association between cir-EIS, IGF1, and MASH. RESULTS CiR-EIS was downregulated in patients with MASH and colocalized with the macrophage marker CD68. CiR-EIS and mitochondrially encoded NADH dehydrogenase 5 (MT-ND5) were downregulated in M1 macrophages and upregulated in M2 macrophages. TDM-derived supernatants overexpressed ciR-EIS, significantly reducing HepG2 lipid deposition and inhibiting LX2 proliferation. Overexpression of ciR-EIS in TDMs significantly inhibited M1 macrophage markers CD86, interleukin-1 beta, and tumor necrosis factor-alpha while enhancing M2 macrophage markers CD163 and CD206. CiR-EIS regulated macrophage polarization in a manner involving the miR-548m/IGF1 axis. Serum IGF1 levels were positively correlated with ciR-EIS, and both of them were notably reduced in patients with MASH and inversely correlated with APRI and FIB-4 scores. CONCLUSIONS CiR-EIS regulates macrophage polarization in a manner involving the miR-548/IGF1 axis, thereby reducing hepatocyte lipid accumulation and stellate cell proliferation in MASH. It demonstrates potential as a diagnostic marker and therapeutic target for MASH.
Collapse
Affiliation(s)
- Xiaoman Chen
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhiping Wan
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Lili Wu
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiang Cai
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hang Si
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoquan Liu
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qiyi Zhao
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Fen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Hong Deng
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
9
|
Stefanakis K, George J, Mantzoros CS. Non-invasive diagnosis and prognosis of MASH with fibrosis F2-F3: In need for a tailored, accessible, and affordable solution for the 21st century public health epidemic. Metabolism 2025; 169:156296. [PMID: 40355078 DOI: 10.1016/j.metabol.2025.156296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Konstantinos Stefanakis
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, and Department of Gastroenterology & Hepatology, Westmead Hospital and Sydney West Local Health District, Sydney Medical School, Australia
| | - Christos S Mantzoros
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Urlandini L, Leonetti AE, Conforti F, Perri A, Lofaro D, Antonucci G, Mandalà M, Bossio S, Di Agostino S, Rago V. Calorie-restriction treatment mitigates the aging in rat liver model. Biogerontology 2025; 26:108. [PMID: 40332584 PMCID: PMC12058891 DOI: 10.1007/s10522-025-10245-8] [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: 12/08/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
The aging process promotes progressive impairment of homeostasis and the increase of the risk of disease and death. A major hallmark of the aging process is the systemic chronic inflammation which strongly contributes to the onset of aging-related diseases. In the liver, the aging condition drives the hepatocytes to develop a metabolic dysfunction-associated steatosis. Caloric restriction (CR) is a remarkable strategy to delay biological aging, occurring through several mechanisms. In this study we aimed to explore, employing an in vivo rat model, the impact of CR on aging-mediated liver inflammation markers. The experiments were performed on 14 male Sprague-Dawley rats (24 months old). At 18 months old, rats were allocated into two groups: the normal diet (ND) group was continued ad libitum diet, and the CR regimen group was fed a diet of the same chow restricted to 60% of the intake. All animals were sacrificed at 24 months old. Compared to the ND group, morphological examination of the liver revealed a lower level of fibrosis in the CR group, concomitantly with a reduced expression of key fibrotic markers, such as collagen I, fibronectin, and αSMA. Furthermore, CR improved the liver oxidative balance, as showed by the increased expression of two scavenging enzymes, SOD1/SOD. Moreover, we reported concomitant reduction of NLRP3 inflammasome signalling. Interestingly, CR significantly improved the signalling of key members of the nutrition-sensitizing affected by aging, AMPK/SIRT1/LKB1. Collectively our findings support the evidence on the metabolic benefits of CR about aging-related liver inflammation, by inducing a morphological improvement that mirrors the decrease in the expression of inflammatory molecular markers.
Collapse
Affiliation(s)
- L Urlandini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - A E Leonetti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - F Conforti
- Pathology Unit, Annunziata Hospital, 87100, Cosenza, Italy
| | - A Perri
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - D Lofaro
- University of Calabria, Department of Mathematics and Computer Science, Rende, 87036, Cosenza, Italy
| | - G Antonucci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - M Mandalà
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - S Bossio
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - S Di Agostino
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy.
| | - V Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
11
|
Zhu X, Yin H, Han J, Zhang X, Han Q, Sun W, Liu Y, Tao W, Liu X, Wang G, Li L. Association Between Uric Acid to HDL-C Ratio and Metabolic Dysfunction-Associated Steatotic Liver Disease in Type 2 Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2025; 18:1459-1466. [PMID: 40356713 PMCID: PMC12067459 DOI: 10.2147/dmso.s520688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Background Patients with type 2 diabetes mellitus (T2DM) exhibit an elevated risk of developing metabolic dysfunction-associated steatotic liver disease (MASLD). The uric acid to high-density lipoprotein cholesterol ratio (UHR) has emerged as a novel metabolic biomarker implicated in MASLD pathogenesis. This study aimed to evaluate the association between UHR and MASLD in a T2DM population. Methods In this cross-sectional study, we analyzed clinical data from 1081 T2DM patients (464 without MASLD, 617 with MASLD). Physiological and biochemical parameters were collected and analyzed. UHR was calculated as [uric acid (mg/dL)/HDL-C (mg/dL)] × 100%. Univariate and multivariate logistic regression analyses were performed to examine the association between UHR and MASLD. Results T2DM patients with MASLD had significantly higher UHR levels than those without MASLD (12.12[9.06-16.83] vs 10.36[7.65-14.08], p<0.001). UHR showed a strong positive correlation with TG/HDL (r =0.673, p < 0.001), moderate correlations with TG (r = 0.516, p < 0.001) and TC/HDL (r =0.548, p < 0.001), weak but significant associations with BMI (r = 0.330), WHR (r = 0.289), HOMA-IR (r = 0.121), ALT (r = 0.123), and GGT (r = 0.267) (all p < 0.05). Multivariate logistic regression showed that elevated UHR levels were significantly associated with increased MASLD risk (adjusted OR = 1.057, 95% CI: 1.016-1.100, p = 0.006), after adjusting for age, diabetes duration, BMI, blood pressure, and biochemical confounders. Conclusion Elevated UHR is independently associated with MASLD in T2DM patients, suggesting its clinical relevance in MASLD screening among this high-risk population.
Collapse
Affiliation(s)
- Xiangyun Zhu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Han Yin
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Jing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xiaoyan Zhang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Qing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Yijun Liu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Wenxuan Tao
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xinliang Liu
- Department of Endocrinology, Lianyungang Affiliated Hospital of Nanjing Medical University, Lianyungang, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Xuyi People’s Hospital of Clinical College of Yangzhou University, Huai’an, Jiangsu, 211700, People’s Republic of China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| |
Collapse
|
12
|
Trapp HM, Machado-Júnior PAB, Pimentel SK. INCIDENTAL HEPATIC STEATOSIS IDENTIFIED ON ULTRASOUND IN PATIENTS UNDERGOING CHOLECYSTECTOMY: HIGH PREVALENCE AND INSUFFICIENT INVESTIGATIVE AND CLINICAL MANAGEMENT. ARQUIVOS DE GASTROENTEROLOGIA 2025; 62:e24118. [PMID: 40332311 PMCID: PMC12052268 DOI: 10.1590/s0004-2803.24612024-118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/07/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Steatotic liver disease (SLD) affects about 1 billion people globally, making its proper management essential to prevent progression to more severe stages. OBJECTIVE The aim of this study was to evaluate medical management concerning hepatic steatosis incidentally identified by ultrasound in patients undergoing elective cholecystectomy. METHODS This observational, cross-sectional, and retrospective study included patients aged 18 years or older who underwent elective cholecystectomy at Hospital do Trabalhador, in Curitiba/PR, between 2018 and 2022. Patients with external ultrasound reports or incomplete data in their medical records were excluded. Medical records, laboratory tests, and ultrasound reports were analyzed to evaluate the prevalence of steatosis in these patients. RESULTS The study sample consisted of 355 patients, and 103 (29.01%) of them presented steatosis on ultrasound. Older age (P=0.0022), male sex (P=0.03009), higher body mass index (P<0.001), obesity (P<0.001), hypertension (P<0.001), dyslipidemia (P=0.0072), and elevated levels of oxaloacetic and pyruvic aminotransferases (P=0.02112) were associated with the presence of this finding. No action was taken regarding the presence of steatosis in 60.19% of patients. Approximately 39.81% had the finding recorded in their medical records, 6.80% received lifestyle change counseling, and 4.85% were investigated for the stage of steatosis. CONCLUSION A significant prevalence of hepatic steatosis was incidentally identified in the ultrasound of patients undergoing cholecystectomy. However, the approach to this finding was insufficient, highlighting the need for substantial improvements on its management and investigation.
Collapse
Affiliation(s)
- Heloísa Mello Trapp
- Universidade Federal do Paraná, Faculdade de Medicina, Curitiba, PR, Brasil
- Hospital do Trabalhador, Serviço de Cirurgia Geral, Curitiba, PR, Brasil
| | | | - Silvania Klug Pimentel
- Universidade Federal do Paraná, Faculdade de Medicina, Curitiba, PR, Brasil
- Hospital do Trabalhador, Serviço de Cirurgia Geral, Curitiba, PR, Brasil
| |
Collapse
|
13
|
Manolis AA, Manolis TA, Vouliotis A, Manolis AS. Metabolic dysfunction-associated steatotic liver disease and the cardiovascular system. Trends Cardiovasc Med 2025; 35:258-265. [PMID: 39848507 DOI: 10.1016/j.tcm.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty-liver disease, is an important and rising health issue with a link with atherosclerotic cardiovascular (CV) disease (CVD), affecting ∼25-30 % of the adults in the general population; in patients with diabetes, its prevalence culminates to ∼70 %; its evolutive form, nonalcoholic steatohepatitis, is estimated to be the main cause of liver transplantation in the future. MASLD is a multisystem disease that affects, besides the liver, extra-hepatic organs and regulatory pathways; it raises the risk of type 2 diabetes mellitus (T2D), CVD, and chronic kidney disease; the disease may also progress to hepatocellular carcinoma. Its diagnosis requires hepatic steatosis and at least one cardiometabolic risk factor and the exclusion of both significant alcohol consumption and other competing causes of chronic liver disease. Beyond CV events, associated metabolic comorbidities comprise obesity (∼50 %), T2D (∼20 %), hyperlipidemia (∼70 %), hypertension (∼40 %), and metabolic syndrome (∼40 %). Among the various clinical events, CV events mostly determine prognosis as they are the leading cause of death in these patients. Regarding management, statins exert beneficial effects in improving liver injury; silybin, derived from Silybum marianum, has some protective effects; lifestyle modification, such as weight loss, dietary changes, physical exercise, and abstention from alcohol use combined with optimal management of comorbidities are most helpful. Bariatric surgery may be an option in persons with MASLD and obesity. Adults with non-cirrhotic MASLD and significant liver fibrosis may be candidates for targeted treatment with resmetirom, which has histological efficacy on steatohepatitis and fibrosis with an acceptable safety and tolerability profile, whereas, no MASLD-targeted pharmacotherapy can be beneficial in the cirrhotic stage, whereby other measures may include metabolic drugs, nutritional counseling, surveillance for portal hypertension and hepatocellular carcinoma, and finally, liver transplantation in decompensated cirrhosis.
Collapse
Affiliation(s)
| | - Theodora A Manolis
- Department of Psychiatry, Aiginiteio University Hospital. Athens, Greece
| | | | - Antonis S Manolis
- Department of Cardiology, Euroclinic Hospital, Athens, Greece; First Department of Cardiology, Athens University School of Medicine, Athens, Greece.
| |
Collapse
|
14
|
Ayares G, Diaz LA, Idalsoaga F, Alkhouri N, Noureddin M, Bataller R, Loomba R, Arab JP, Arrese M. MetALD: New Perspectives on an Old Overlooked Disease. Liver Int 2025; 45:e70017. [PMID: 40179033 PMCID: PMC11967760 DOI: 10.1111/liv.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 04/05/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-associated liver disease (ALD) are the major contributors to the liver disease burden globally. The rise in these conditions is linked to obesity, type 2 diabetes, metabolic syndrome and increased alcohol consumption. MASLD and ALD share risk factors, pathophysiology and histological features but differ in their thresholds for alcohol use, and the ALD definition does not require the presence of metabolic dysfunction. A recent multi-society consensus overhauled the nomenclature of liver steatosis and introduced the term MetALD to describe patients with metabolic dysfunction who drink more than those with MASLD and less than those with ALD. This new terminology aims to enhance the understanding and management of liver disease but poses challenges, such as the need to accurately measure alcohol consumption in research and clinical practice settings. Recent studies show that MetALD has significant implications for patient management, as it is associated with increased mortality risks and more severe liver outcomes compared to MASLD alone. MetALD patients face increased risks of liver disease progression, cancer and cardiovascular disease. The diagnosis of MetALD involves the adequate quantification of alcohol use through standardised questionnaires and/or biomarkers as well as proper assessment of liver disease stage and progression risk using non-invasive tools including serologic markers, imaging, elastography techniques and genetic testing. Effective management requires addressing both metabolic and alcohol-related factors to improve outcomes. This review intends to provide a comprehensive overview of MetALD, covering pathogenesis, potential diagnostic approaches, management strategies and emerging therapies.
Collapse
Affiliation(s)
- Gustavo Ayares
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Escuela de Medicina, Universidad Finis TerraeSantiagoChile
| | - Luis Antonio Diaz
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- MASLD Research Center, Division of Gastroenterology and HepatologyUniversity of California San DiegoCaliforniaUSA
| | - Francisco Idalsoaga
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Division of Gastroenterology Department of MedicineSchulich School of Medicine, Western University & London Health Sciences CentreLondonOntarioCanada
| | - Naim Alkhouri
- Department of HepatologyArizona Liver HealthChandlerArizonaUSA
| | | | - Ramon Bataller
- Liver UnitHospital Clinic and Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and HepatologyUniversity of California San DiegoCaliforniaUSA
| | - Juan Pablo Arab
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal MedicineVirginia Commonwealth University School of MedicineVirginiaUSA
| | - Marco Arrese
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
15
|
Li W, Qian Y, Cai X, He Y, Meng X, Zhang L. Therapeutic intervention with anti-TNF alleviates colonic and hepatic toxicity induced by perfluorooctanoic acid (PFOA). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118125. [PMID: 40220356 DOI: 10.1016/j.ecoenv.2025.118125] [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: 11/25/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Perfluorooctanoic acid (PFOA) is a prevalent and chemically stable environmental contaminant. Our preliminary data suggest that chronic exposure to PFOA induces colonic damage in mice that resembles inflammatory bowel disease (IBD). Anti-TNF therapies are commonly used in the clinical management of IBD. Building upon our previous findings, we administered anti-TNF treatment to mice exposed to PFOA. Our results show that anti-TNF therapy significantly reduces the colonic inflammatory response, activation of the NLR family pyrin domain containing 3 (NLRP) inflammasome, and apoptosis induced by PFOA. Additionally, anti-TNF treatment restores intestinal barrier integrity, which is disrupted by PFOA exposure, and enhances the regenerative capacity of the colon by promoting intestinal stem cell function. Furthermore, anti-TNF therapy effectively mitigates hepatic inflammation, liver dysfunction, lipid metabolism disturbances, NLRP3 inflammasome activation, and apoptosis in the liver triggered by PFOA. In conclusion, our study provides compelling evidence that anti-TNF therapy can alleviate both colonic and hepatic injuries induced by PFOA exposure. This research expands our understanding of environmental toxin-induced diseases and offers potential therapeutic strategies for managing PFOA-related disorders in the future.
Collapse
Affiliation(s)
- Wei Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Yongjing Qian
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Xiaojing Cai
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Yu He
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, No.9 Chongwen Road, Suzhou, 215000, Jiangsu, China
| | - Xiannan Meng
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Ling Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China; National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
16
|
Cunneely OP, Roberts A, Fargue S, Knight J, Assimos DG, Wood KD. Metabolic dysfunction associated steatotic liver and kidney stones: what is going on? Curr Opin Nephrol Hypertens 2025; 34:247-253. [PMID: 39882643 DOI: 10.1097/mnh.0000000000001062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
PURPOSE OF REVIEW Metabolic dysfunction associated steatotic liver disease (MASLD) is increasing throughout the world, affecting nearly one in three individuals. Kidney stone disease, which is also increasing, is associated with MASLD. Common risk factors for both, including obesity, diabetes, dyslipidemia, hypertension, and metabolic syndrome, are likely drivers of this association. We present here a review of the associations and possible interconnections between these two common disease processes. RECENT FINDINGS Epidemiological studies are discordant regarding the impact of sex on this association and on the impact of MASLD on incident stone risk. The nature of kidney stones is rarely taken into account.A favorable milieu for uric acid kidney stone formation may be created by a lower urine pH resulting from defective ammonium production associated with insulin resistance, common in MASLD.Endogenous oxalate synthesis, a major risk factor for calcium oxalate kidney stones, may be increased in MASLD via decline in the activity of enzymes involved in the detoxification of glyoxylate, the immediate precursor of oxalate. SUMMARY The nature of kidney stones associated with MASLD and factors driving this association remain to be elucidated. Potential mechanisms identified underlying this include an increase in the risk factors for both uric acid and calcium oxalate kidney stones.
Collapse
Affiliation(s)
- Owen P Cunneely
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | | | |
Collapse
|
17
|
Gu Y, Guo C, Liu Z, Zhang Y, Han X, Zhang X, Zhao S, Wang H, Zhang T. The trend in incidence of non-alcoholic fatty liver disease and its impact on cirrhosis and liver cancer: An analysis from Global Burden of Disease 2021. Public Health 2025; 242:79-86. [PMID: 40037155 DOI: 10.1016/j.puhe.2025.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
OBJECTIVES We aimed to recognize the burden of NAFLD and support public health policy development for its prevention and management. STUDY DESIGN A cross-sectional analysis of GBD 2021 results was conducted. METHODS We collected incidence data on NAFLD from 1990 to 2021 using Global Burden of Disease Study in 2021. Estimated annual percentage changes (EAPCs) in NAFLD age standardized incidence rate (ASR) were calculated to quantify the temporal trends in NAFLD ASR. Bayesian age-period-cohort models were constructed to project NAFLD incidence rates and cases up to 2050. Additionally, we assessed the percentage of cirrhosis and liver cancer attributable to NAFLD. RESULTS Globally, the newly-occurred cases of NAFLD increased by 94.49 % from 24, 856, 159 in 1990 to 48, 353, 272 in 2021. The case number will further increase to 78,602,984 in 2050, and ASR will increase from 5.93 per 1000 in 2021 to 7.26 per 1000 in 2050. The most pronounced increases were observed in young people and men. In 2021, NAFLD accounted for 82.7 % of cirrhosis and other chronic liver diseases and 8.0 % of liver cancer cases. CONCLUSIONS From 1990 to 2021, the incidence of NAFLD has been continuously increasing and is expected to continue rising until 2050. The increases in young people and men highlight their priority in future schedules. The rising proportions of cirrhosis and liver cancer caused by NAFLD further underscore the serious health risks.
Collapse
Affiliation(s)
- Yu Gu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Chengnan Guo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Zhenqiu Liu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, 200032, China
| | - Yujiao Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Xinyu Han
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, China; Yiwu Research Institute, Fudan University, Yiwu, 200032, China.
| |
Collapse
|
18
|
Wang W, Gao X, Niu W, Yin J, He K. Targeting Metabolism: Innovative Therapies for MASLD Unveiled. Int J Mol Sci 2025; 26:4077. [PMID: 40362316 PMCID: PMC12071536 DOI: 10.3390/ijms26094077] [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: 02/22/2025] [Revised: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The recent introduction of the term metabolic-dysfunction-associated steatotic liver disease (MASLD) has highlighted the critical role of metabolism in the disease's pathophysiology. This innovative nomenclature signifies a shift from the previous designation of non-alcoholic fatty liver disease (NAFLD), emphasizing the condition's progressive nature. Simultaneously, MASLD has become one of the most prevalent liver diseases worldwide, highlighting the urgent need for research to elucidate its etiology and develop effective treatment strategies. This review examines and delineates the revised definition of MASLD, exploring its epidemiology and the pathological changes occurring at various stages of the disease. Additionally, it identifies metabolically relevant targets within MASLD and provides a summary of the latest metabolically targeted drugs under development, including those in clinical and some preclinical stages. The review finishes with a look ahead to the future of targeted therapy for MASLD, with the goal of summarizing and providing fresh ideas and insights.
Collapse
Affiliation(s)
- Weixin Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Xin Gao
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wentong Niu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Jinping Yin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130041, China;
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| |
Collapse
|
19
|
Zhang Y, Wang J, Yang S, Kou H, Liu P. Tanshinone IIA alleviate atherosclerosis and hepatic steatosis via down-regulation of MAPKs/NF-κB signaling pathway. Int Immunopharmacol 2025; 152:114465. [PMID: 40090083 DOI: 10.1016/j.intimp.2025.114465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/09/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVES Tanshinone IIA (Tan IIA) exhibits therapeutic potential for atherosclerosis (AS) and hepatic steatosis (HS). The study aims to explore the mechanisms underlying the anti-atherosclerosis and anti-hepatic steatosis effects of Tan IIA. METHODS The LDLR-/-mice were divided into control, model, low/high Tan IIA and atorvastatin group, which fed with High-fat diet to build NAFLD-associated AS model, then administrated with 0.9 % saline, Tan IIA or atorvastatin. RAW264.7 cells divided into control, LPS, LPS plus low/high Tan IIA and LPS plus Tan IIA plus JNK activator group. The different goups' pathological changes visualized with H&E, Oil Red O and Immunofluorescence staining. The therapeutic effect of Tan IIA was reflected by lipids metabolism changes, hepatic indexes, inflammation levels. ELISA, RT-qPCR and Western blot assay were used to determine the inflammatory factors and upstream proteins. Molecular docking was used to reconfirm the importance of genes studied and locate the specific gene will study. RESULTS Tan IIA alleviated LDLR-/-mice AS and HS by reducing AS plaque area, lowering serum &liver lipid levels (TC, TG), improving liver function (AST, ALT). Tan IIA decreased serum inflammation levels (IL-1β, IL-6, TNF-α) and aorta & liver inflammatory-related cytokines levels (iNOS, VCAM-1, IL-6) and inhibited the phosphorylation of aorta & liver protein ERK1/2, JNK, p38 and NF-κB p65, which were validated in the LPS-stimulated macrophages supernatant and cells. CONCLUSIONS The study indicated that Tan IIA can alleviate atherosclerosis and hepatic steatosis via down-regulating MAPKs/NF-κB signaling pathway. This provides a potential therapeutic strategy for the co-existing situation of atherosclerosis and hepatic steatosis.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiarou Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuo Yang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haixin Kou
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
20
|
Schaefer B, Zoller H. Diving deep: Finding pathogenic intronic variants and making them actionable. J Hepatol 2025:S0168-8278(25)00240-5. [PMID: 40246151 DOI: 10.1016/j.jhep.2025.04.013] [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: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Affiliation(s)
- Benedikt Schaefer
- Medical University of Innsbruck, Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Heinz Zoller
- Medical University of Innsbruck, Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Anichstrasse 35, A-6020 Innsbruck, Austria; Medical University of Innsbruck, Christian Doppler Laboratory for Iron and Phosphate Biology, Anichstrasse 35, A-6020 Innsbruck, Austria.
| |
Collapse
|
21
|
Israelsen M, Thorhauge KH, Andersen P, Johansen S, Schnefeld HL, Bech KT, Hansen JK, Torp N, Jensen EL, Hansen ED, Spedtsberg IZ, Villesen IF, Hansen CD, Hansen T, Lindvig KP, Thiele M, Krag A. Steatotic Liver Disease Classification is Dynamic, Affecting Clinical Trial Eligibility and Subclass-Specific Treatments. Clin Gastroenterol Hepatol 2025:S1542-3565(25)00243-5. [PMID: 40204204 DOI: 10.1016/j.cgh.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND & AIMS Steatotic liver disease (SLD) includes the subclasses metabolic-dysfunction-associated steatotic liver disease, metabolic and alcohol-related liver disease, and alcohol-related liver disease. We investigated the robustness of the SLD diagnoses after 2 years. METHODS We performed a prospective cohort study among individuals from the general population and individuals at risk of SLD. Participants were classified according to the diagnostic criteria of the SLD nomenclature at baseline and after 2 years. RESULTS We included 1042 participants, mean age was 57 ± 10 years, 663 were male (64%) and 379 (36%) female, and 70 (7%) had advanced fibrosis. At baseline, 595 (57%) had SLD, hereof 371 (62%) met the criteria for metabolic-dysfunction-associated steatotic liver disease, 140 (24%) for metabolic and alcohol-related liver disease, and 83 (14%) for alcohol-related liver disease, and 1 (<1%) was classified as cryptogenic SLD. Median time between the baseline and follow-up visit was 25 months (interquartile range, 24-31). Overall, 399 (38%) of the 1042 participants changed SLD classification from baseline to follow-up. In the group without SLD at baseline, 25% had SLD at follow-up. In comparison, 38% with metabolic-dysfunction-associated steatotic liver disease at baseline were classified differently at follow-up, whereas 62% and 69% of participants with baseline metabolic and alcohol-related liver disease and alcohol-related liver disease were classified differently at follow-up. The primary reasons for changing classification were that individuals no longer met the criteria for having SLD or had altered their alcohol intake. CONCLUSIONS SLD and the subclassification hereof is highly dynamic, especially driven by changes in alcohol use and hepatic steatosis. This affects eligibility for clinical trials and clinical management of patients with SLD.
Collapse
Affiliation(s)
- Mads Israelsen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Katrine Holtz Thorhauge
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Peter Andersen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Stine Johansen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Helle Lindholm Schnefeld
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Katrine Tolstrup Bech
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Johanne Kragh Hansen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Nikolaj Torp
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ellen Lyngbeck Jensen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Emil Deleuran Hansen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ida Ziegler Spedtsberg
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ida Falk Villesen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Camilla Dalby Hansen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Prier Lindvig
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Maja Thiele
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Aleksander Krag
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
22
|
Krag A, Torp N, Younossi ZM, Israelsen M. Reporting discrepancy of alcohol intake affecting estimated prevalence of MetALD and ALD. Lancet Gastroenterol Hepatol 2025; 10:282-284. [PMID: 39855240 DOI: 10.1016/s2468-1253(24)00427-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Affiliation(s)
- Aleksander Krag
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense 5000, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Nikolaj Torp
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense 5000, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Zobair M Younossi
- Global NASH Council, Washington, DC, USA; Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Mads Israelsen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense 5000, Denmark; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
Zhan C, Peng C, Wei H, Wei K, Ou Y, Zhang Z. Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases. Int J Mol Sci 2025; 26:2778. [PMID: 40141420 PMCID: PMC11943347 DOI: 10.3390/ijms26062778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
γδT cells, a distinct group of T lymphocytes, serve as a link between innate and adaptive immune responses. They are pivotal in the pathogenesis of various liver disorders, such as viral hepatitis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), liver fibrosis, autoimmune liver diseases, and hepatocellular carcinoma (HCC). Despite their importance, the functional diversity and regulatory mechanisms of γδT cells remain incompletely understood. Recent advances in high-throughput single-cell sequencing and spatial transcriptomics have revealed significant heterogeneity among γδT cell subsets, particularly Vδ1+ and Vδ2+, which exhibit distinct immunological roles. Vδ1+ T cells are mainly tissue-resident and contribute to tumor immunity and chronic inflammation, while Vδ2+ T cells, predominantly found in peripheral blood, play roles in systemic immune surveillance but may undergo dysfunction in chronic liver diseases. Additionally, γδT17 cells exacerbate inflammation in NAFLD and ALD, whereas IFN-γ-secreting γδT cells contribute to antiviral and antifibrotic responses. These discoveries have laid the foundation for the creation of innovative solutions. γδT cell-based immunotherapeutic approaches, such as adoptive cell transfer, immune checkpoint inhibition, and strategies targeting metabolic pathways. Future research should focus on harnessing γδT cells' therapeutic potential through targeted interventions, offering promising prospects for precision immunotherapy in liver diseases.
Collapse
Affiliation(s)
- Chenjie Zhan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Chunxiu Peng
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Huaxiu Wei
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Ke Wei
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Yangzhi Ou
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Zhiyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
- Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, New Brunswick, NJ 08901-8554, USA
| |
Collapse
|
24
|
Trizzino M, Gaudiano R, Arena DM, Pipitò L, Gioè C, Cascio A. Switching to Bictegravir/Emtricitabine/Tenofovir Alafenamide Fumarate Regimen and Its Effect on Liver Steatosis Assessed by Fibroscan. Viruses 2025; 17:440. [PMID: 40143367 PMCID: PMC11945801 DOI: 10.3390/v17030440] [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: 03/04/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Antiretroviral therapy has transformed HIV infection from a fatal disease to a chronic and manageable condition, but increasing health issues beyond acquired immunodeficiency syndrome, such as metabolic, liver, and cardiovascular diseases, have been observed. Furthermore, the increasing prevalence of HIV strains resistant to older antiretroviral regimens has necessitated a re-evaluation of treatment strategies. METHODS We performed a retrospective, observational study to evaluate the long-term outcomes of an antiretroviral switch from a non-nucleoside reverse transcriptase inhibitor-based to bictegravir-based regimen; this study aimed to assess the impact of this antiretroviral switch on treatment adherence, the safety profile, and virologic outcomes. The secondary objectives were to analyze the changes in lipid, kidney function, liver function, and anthropometric parameters after switching. RESULTS A total of 25 patients were included in this analysis; virologic suppression was maintained over time, with 100% of patients demonstrating undetectable viral loads at 6, 12, 24, and 36 months. In parallel, a significant increase in CD4+ cell count was observed after switching. No significant differences were observed compared to the previous therapy regarding anthropometric parameters or laboratory parameters. However, a significant reduction in liver steatosis, as assessed by Fibroscan, was observed. CONCLUSIONS bictegravir-based regimens are a valid therapeutic option for people living with HIV, particularly for those with metabolic comorbidities.
Collapse
Affiliation(s)
- Marcello Trizzino
- Infectious and Tropical Disease Unit and Sicilian Regional Reference Center for the Fight Against AIDS, AOU Policlinico “P. Giaccone”, 90127 Palermo, Italy;
| | - Roberta Gaudiano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (D.M.A.); (L.P.)
| | - Dalila Mimì Arena
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (D.M.A.); (L.P.)
| | - Luca Pipitò
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (D.M.A.); (L.P.)
| | - Claudia Gioè
- Infectious and Tropical Disease Unit and Sicilian Regional Reference Center for the Fight Against AIDS, AOU Policlinico “P. Giaccone”, 90127 Palermo, Italy;
| | - Antonio Cascio
- Infectious and Tropical Disease Unit and Sicilian Regional Reference Center for the Fight Against AIDS, AOU Policlinico “P. Giaccone”, 90127 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (D.M.A.); (L.P.)
- Palermo Fast-Track City, Casa dei Diritti, Via Libertà 45, 90143 Palermo, Italy
| |
Collapse
|
25
|
Wei R, Liu W, Yuan C, Zhang C, Sang Z, Ma Q. Editorial: Preventing and treating liver diseases: medicinal and food plants, their metabolites as potential options. Front Pharmacol 2025; 16:1577547. [PMID: 40170722 PMCID: PMC11959060 DOI: 10.3389/fphar.2025.1577547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Affiliation(s)
- Rongrui Wei
- College of Medicine and Health Science & School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Wenmin Liu
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanchang, China
| | - Chunsu Yuan
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, Committee On Clinical Pharmacology and Pharmacogenomics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States
| | - Chunlei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhipei Sang
- School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Qinge Ma
- College of Medicine and Health Science & School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
26
|
Åberg F, Männistö V. Prediction of major liver-related events in the population using prognostic models. Gastroenterol Rep (Oxf) 2025; 13:goaf028. [PMID: 40093587 PMCID: PMC11908767 DOI: 10.1093/gastro/goaf028] [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: 12/20/2024] [Revised: 01/17/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Liver disease poses a significant global health burden, with steatotic liver disease related to metabolic dysfunction and/or alcohol use being the most prevalent type. Current risk stratification strategies emphasize detecting advanced fibrosis as a surrogate marker for liver-related events (LREs), such as hospitalization, liver cancer, or death. However, fibrosis alone does not adequately predict imminent outcomes, particularly in fast-progressing individuals without advanced fibrosis at evaluation. This underscores the need for models designed specifically to predict LREs, enabling timely interventions. The Chronic Liver Disease (CLivD) risk score, the dynamic aspartate aminotransferase-to-alanine aminotransferase ratio (dAAR), and the Cirrhosis Outcome Risk Estimator (CORE) were explicitly developed to predict LRE risk rather than detect fibrosis. Derived from general population cohorts, these models incorporate either standard liver enzymes (dAAR and CORE) or risk factors (CLivD), enabling broad application in primary care and population-based settings. They directly estimate the risk of future LREs, improving on traditional fibrosis-focused approaches. Conversely, widely used models like the Fibrosis-4 index and newer ones, such as the LiverRisk and LiverPRO scores, were initially developed to detect significant/advanced fibrosis or liver stiffness. While not designed for LRE prediction, they have later been analyzed for this purpose. Integrating fibrosis screening with LRE-focused models like CLivD, dAAR, and CORE can help healthcare systems adopt proactive, preventive care. This approach emphasizes identifying individuals at imminent risk of severe outcomes, potentially ensuring better resource allocation and personalized interventions.
Collapse
Affiliation(s)
- Fredrik Åberg
- Transplantation and Liver Surgery Unit, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ville Männistö
- School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
27
|
Feng J, MengHuan L, TingTing Y, XueJie Y, HaiNing G. Research progress on AMPK in the pathogenesis and treatment of MASLD. Front Immunol 2025; 16:1558041. [PMID: 40134423 PMCID: PMC11932893 DOI: 10.3389/fimmu.2025.1558041] [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: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as non-alcoholic fatty liver disease, NAFLD) has become one of the most prevalent chronic liver diseases worldwide, with its incidence continuously rising alongside the epidemic of metabolic disorders. AMP-activated protein kinase (AMPK), as a key regulator of cellular energy metabolism, influences multiple pathological processes associated with MASLD. This review systematically summarizes the regulatory roles of AMPK in lipid metabolism, inflammatory response, cell apoptosis, and fibrosis. Additionally, it discusses the latest developments of AMPK activators from preclinical to clinical studies, while analyzing the major challenges currently faced and potential strategies for resolution. A deeper understanding of AMPK regulatory mechanisms will contribute to the development of more effective therapeutic approaches for MASLD.
Collapse
Affiliation(s)
- Jiang Feng
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Li MengHuan
- School of Physical Education, Liaoning Normal University, Dalian, China
| | - Yao TingTing
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yi XueJie
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Gao HaiNing
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
28
|
Kozłowska A. Clinical Insights into Non-Alcoholic Fatty Liver Disease and the Therapeutic Potential of Flavonoids: An Update. Nutrients 2025; 17:956. [PMID: 40289935 PMCID: PMC11944923 DOI: 10.3390/nu17060956] [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: 02/21/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered a significant global health issue related to serious metabolic disorders. However, effective pharmacological treatments are still limited. Flavonoids, a wide group of polyphenol substances, exert anti-inflammatory and lipid-lowering effects in preclinical data. Thus, implementing these research findings in clinical practice could significantly help manage NAFLD and its consequences. This narrative review assesses the therapeutic potential of flavonoids in managing NAFLD. The research collected randomized controlled trials (RCTs) and meta-analyses of RCTs from the past five years concerning the impact of flavonoids on NAFLD. A total of 20 studies were selected according to predetermined inclusion criteria, comprising thirteen randomized controlled trials (RCTs) and seven meta-analyses. The research underscores the beneficial effects of flavonoids in the management of NAFLD through the enhancement of lipid metabolism, the reduction in hepatic steatosis, and the provision of anti-inflammatory actions. Clinical trials demonstrate that interventions rich in flavonoids, including quercetin, epigallocatechin gallate, naringenin, and isoflavones, substantially reduce liver fat content and enhance liver enzyme profiles, with certain compounds exhibiting superior efficacy in particular subgroups, such as older adults and females. Nonetheless, whereas these therapies significantly diminish hepatic steatosis, their effect on fibrosis is constrained. To sum up, flavonoids exhibit significant potential as supplementary treatments for NAFLD by enhancing liver function, lipid metabolism, and inflammation. Additional extensive controlled clinical trials are necessary to create uniform treatment methods and ascertain their long-term therapeutic advantages.
Collapse
Affiliation(s)
- Aleksandra Kozłowska
- Department of Social Medicine and Public Health, Medical University of Warsaw, 02-106 Warsaw, Poland
| |
Collapse
|
29
|
Lian YE, Wang Y, Yang Y, Chen J. Weight-adjusted waist circumference index with hepatic steatosis and fibrosis in adult females: a cross-sectional, nationally representative study (NHANES 2017-2020). BMC Gastroenterol 2025; 25:137. [PMID: 40045243 PMCID: PMC11884151 DOI: 10.1186/s12876-025-03706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Obesity is detrimental to liver health. Weight-adjusted waist circumference (WWI) is a new indicator of obesity that is superior to body mass index (BMI) and waist circumference (WC) in predicting obesity. There are limited studies on the relationship between Metabolic Associated Fatty Liver Disease (MASLD) and WWI. Therefore, this study aimed to investigate the association between WWI, Controlled Attenuation Parameters (CAP), and Liver Stiffness Measurement (LSM), with special attention to gender differences. METHODS This cross-sectional study included participants from the 2017 to 2020 National Health and Nutrition Examination Survey (NHANES). The study used multiple linear regression models, smoothed curves, and threshold effects analyses to describe the relationships between variables. Multiple regression analyses were used to examine the associations between the four obesity indicators and CAP and LSM. Subject work characteristics (ROC) curves were used to assess the predictive value of WWI and other traditional obesity indicators for hepatic steatosis and liver fibrosis, and predictive power was assessed by area under the curve (AUC). RESULTS The study involved 6713 participants, including 3072 men (46%) and 3641 women (54%). The results showed that among female participants, higher WWI was associated with hepatic steatosis (OR = 1.71, 95% CI: 1.43, 2.04; P < 0.0001) and hepatic fibrosis (OR = 2.11, 95% CI: 1.58, 2.84; P < 0.0001). Smoothed curve fitting of WWI versus CAP showed a statistically significant positive correlation between WWI in male and female participants There was a statistically significant positive correlation with CAP for both male and female participants. The same significant non-linear relationship was found between WWI and LSM, with no significant difference between males and females. WWI was also a good predictor of hepatic steatosis compared to other obesity indicators and was more pronounced in male participants (AUC = 0.8224). Whereas in the comparison of WWI with LSM, wBMI was a better predictor in female participants (AUC = 0.7751). CONCLUSIONS Based on this study, WWI was significantly associated with the risk of hepatic steatosis and hepatic fibrosis in women, suggesting the potential of WWI as a screening tool. Due to the cross-sectional design, causality cannot be inferred. Longitudinal studies are needed to validate our findings.
Collapse
Affiliation(s)
- Yu-E Lian
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China
| | - Yixuan Wang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China
| | - Yinyin Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, 730050, China
| | - Jiayu Chen
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, 730050, China.
| |
Collapse
|
30
|
Wang S, Zhang R, Guo P, Yang H, Liu Y, Zhu H. Association of prebiotic/probiotic intake with MASLD: evidence from NHANES and randomized controlled trials in the context of prediction, prevention, and a personalized medicine framework. EPMA J 2025; 16:183-197. [PMID: 39991098 PMCID: PMC11842653 DOI: 10.1007/s13167-025-00398-4] [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: 09/07/2024] [Accepted: 01/13/2025] [Indexed: 02/25/2025]
Abstract
Objective Metabolic-associated fatty liver disease (MASLD) is a growing global health concern. From the standpoint of preventive and personalized medicine, understanding the early determinants and modifiable risk factors is essential for targeted prevention and personalized treatment strategies. This study aimed to evaluate the specific association between probiotics/prebiotics and the occurrence of MASLD, contributing to the development of innovative preventive measures and personalized therapeutic approaches. Methods Data were obtained from the National Health and Nutrition Examination Survey (NHANES) from 2001 to 2018. The study employed logistic regression analysis to examine the relation between MASLD and probiotics/prebiotics. The efficacy of various MASLD predictive models was assessed using receiver operating characteristic (ROC) curves. A meta-analysis was conducted by searching databases up to 4 May 2024. The analysis included randomized controlled trials of liver function in patients with MASLD or nonalcoholic steatohepatitis treated with probiotics, prebiotics, or yogurt for a minimum of 6 months. Results A total of 5014 adults from NHANES were included in this study, with a weighted prevalence of MASLD observed at 24.47%. MASLD adults who consumed both probiotics and prebiotics exhibited a reduced risk of MASLD (OR = 0.71, 95% CI: 0.53 to 0.94). The use of probiotics/prebiotics can enhance the simplicity and practicality of the model. Model 1, adjusted for sex, BMI, race, and HEI-2015, achieved an area under the curve (AUC) of 0.8544, while Model 2, adjusted for sex, BMI, race, and prebiotics/probiotics use, showed a similar AUC of 0.8537. The comparison between the two models revealed no statistically significant difference (0.8544 vs. 0.8537; 95% CI: - 0.0010 to 0.0025; Z = 0.8332; p = 0.4047). Subgroup analysis of the NHANES data revealed that individuals aged 40 and older benefit from consuming probiotics or prebiotics. Furthermore, the meta-analysis demonstrated that probiotic or prebiotic interventions resulted in significant improvements in biochemical markers, including alanine aminotransferase, aspartate aminotransferase, low-density lipoprotein cholesterol, and triglycerides. Conclusions The consumption of probiotics/prebiotics has been linked to a reduced risk of developing MASLD in adults. Integrating probiotics/prebiotics into early intervention and personalized treatment plans may facilitate targeted prevention and management of MASLD, promoting a more individualized approach to disease prevention and care. Supplementary information The online version contains supplementary material available at 10.1007/s13167-025-00398-4.
Collapse
Affiliation(s)
- Senlin Wang
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, No. 19 Yangshi Road, Chengdu, Sichuan 610031 China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, College of Medicine, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan China
| | - Ruimin Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Peisen Guo
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, No. 19 Yangshi Road, Chengdu, Sichuan 610031 China
| | - Huawu Yang
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, No. 19 Yangshi Road, Chengdu, Sichuan 610031 China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Yanjun Liu
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, No. 19 Yangshi Road, Chengdu, Sichuan 610031 China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, College of Medicine, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan China
| | - Hongmei Zhu
- The Center of Obesity and Metabolic Diseases, Department of General Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, No. 19 Yangshi Road, Chengdu, Sichuan 610031 China
- Medical Research Center, The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
31
|
Kartalis N, Grigoriadis A. Photon-counting CT: The New Kid on the Block for Liver Fat Quantification. Radiology 2025; 314:e250480. [PMID: 40100017 DOI: 10.1148/radiol.250480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Affiliation(s)
- Nikolaos Kartalis
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, and Department of Radiology Huddinge, Karolinska University Hospital, O-huset 42, 14186 Stockholm, Sweden
| | - Aristeidis Grigoriadis
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, and Department of Radiology Huddinge, Karolinska University Hospital, O-huset 42, 14186 Stockholm, Sweden
| |
Collapse
|
32
|
Huang YL, Sun C, Wang Y, Cheng J, Wang SW, Wei L, Lu XY, Cheng R, Wang M, Fan JG, Dong Y. Ultrasound-guided attenuation parameter for identifying metabolic dysfunction-associated steatotic liver disease: a prospective study. Ultrasonography 2025; 44:134-144. [PMID: 39935289 PMCID: PMC11938800 DOI: 10.14366/usg.24204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 02/13/2025] Open
Abstract
PURPOSE This study assessed the performance of the ultrasound-guided attenuation parameter (UGAP) in diagnosing and grading hepatic steatosis in patients with metabolic dysfunctionassociated steatotic liver disease (MASLD). Magnetic resonance imaging proton density fat fraction (MRI-PDFF) served as the reference standard. METHODS Patients with hepatic steatosis were enrolled in this prospective study and underwent UGAP measurements. MRI-PDFF values of ≥5%, ≥15%, and ≥25% were used as references for the diagnosis of steatosis grades ≥S1, ≥S2, and S3, respectively. Spearman correlation coefficients and area under the receiver operating characteristic curves (AUCs) were calculated. RESULTS Between July 2023 and June 2024, the study included 88 patients (median age, 40 years; interquartile range [IQR], 36 to 46 years), of whom 54.5% (48/88) were men and 45.5% (40/88) were women. Steatosis grades exhibited the following distribution: 22.7% (20/88) had S0, 50.0% (44/88) had S1, 21.6% (19/88) had S2, and 5.7% (5/88) had S3. The success rate for UGAP measurements was 100%. The median UGAP value was 0.74 dB/cm/MHz (IQR, 0.65 to 0.82 dB/ cm/MHz), and UGAP values were positively correlated with MRI-PDFF (r=0.77, P<0.001). The AUCs of UGAP for the diagnoses of ≥S1, ≥S2, and S3 steatosis were 0.91, 0.90, and 0.88, respectively. In the subgroup analysis, 98.4% (60/61) of patients had valid controlled attenuation parameter (CAP) values. UGAP measurements were positively correlated with CAP values (r=0.65, P<0.001). CONCLUSION Using MRI-PDFF as the reference standard, UGAP demonstrates good diagnostic performance in the detection and grading of hepatic steatosis in patients with MASLD.
Collapse
Affiliation(s)
- Yun-Lin Huang
- Department of Ultrasound, Zhongshan Hospital Fudan University, Shanghai, China
| | - Chao Sun
- Center for Fatty Liver Disease, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Wang
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Cheng
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Wen Wang
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wei
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiu-Yun Lu
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Cheng
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Wang
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Gao Fan
- Center for Fatty Liver Disease, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yi Dong
- Department of Ultrasound, Zhongshan Hospital Fudan University, Shanghai, China
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Ronzoni L, Pelusi S, Moretti V, Malvestiti F, Eidgah Torghabehei H, Jamialahmadi O, Rondena J, Bianco C, Periti G, Filippo MRD, Romeo S, Prati D, Valenti L. Diagnostic Uptake of Targeted Sequencing in Adults With Steatotic Liver Disease and a Suspected Genetic Contribution. Liver Int 2025; 45:e70010. [PMID: 39945383 PMCID: PMC11822878 DOI: 10.1111/liv.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND AND AIMS In patients with steatotic liver diseases (SLD), genetic factors may account for severe liver involvement despite mild or absence of triggering factors or a strong family history. Aim of this study was to examine the diagnostic uptake of targeted sequencing (TS), covering both coding and non-coding regions, of a broad panel of 82 liver and lipid metabolism genes in patients with unexplained SLD. METHODS We enrolled 49 adult patients with SLD and a suspected genetic contribution. Genetic variants were detected through a customised TS panel, whereas the contribution of common genetic variation to the individual susceptibility to SLD was captured by a polygenic risk score (SLD-PRS). RESULTS A diagnosis of rare Mendelian disorder was established in 11 patients (22%), independently of age or family history. Rare variants possibly contributing to clinical phenotype were detected in additional 29 patients (59%). Increased SLD-PRS values were detected in 17 patients (35%), enabling an increase in diagnostic uptake of 24%, especially in those without a strong family history (p = 0.03). Genetic diagnosis allowed refinement of clinical management in 23 (47%) patients. CONCLUSIONS The diagnostic uptake of TS was 22% for Mendelian disorder and 59% for possible contribution to clinical phenotype in selected adult patients with SLD. Evaluation of common variants, as captured by SLD-PRS, yields complementary information increasing the overall utility of the genetic examination.
Collapse
Grants
- 777377 Innovative Medicines Initiative 2 joint undertaking of European Union's Horizon 2020 research and innovation program and EFPIA European Union (EU) Program Horizon 2020 for the project LITMUS
- Gilead Sciences Inc.
- 101016726-REVEAL The European Union, H2020-ICT-2018-20/H2020-ICT-2020-2 program "Photonics"
- Italian ministry of Research (MUR) PNRR - M4 - C2 "National Center for Gene Therapy and Drugs based on RNA Technology" CN3, Spoke 4
- 101096312 The European Union, HORIZON-MISS-2021-CANCER-02-03 program "Genial"
- RF-2016-02364358 The Italian Ministry of Health (Ministero della Salute), Ricerca Finalizzata 2016
- Italian ministry of Research (MUR) PRIN 2022
- PNRR-MAD-2022-12375656 The Italian Ministry of Health, Ricerca Finalizzata PNRR 2022
- RF-2021-12373889 The Italian Ministry of Health, Ricerca Finalizzata 2021
- PR-0361 Fondazione Patrimonio Ca' Granda, "Liver BIBLE"
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Ricerca Corrente
- Innovative Medicines Initiative 2 joint undertaking of European Union’s Horizon 2020 research and innovation program and EFPIA European Union (EU) Program Horizon 2020 for the project LITMUS
- The European Union, H2020‐ICT‐2018‐20/H2020‐ICT‐2020‐2 program “Photonics”
- Italian ministry of Research (MUR) PNRR ‐ M4 ‐ C2 “National Center for Gene Therapy and Drugs based on RNA Technology” CN3, Spoke 4
- The European Union, HORIZON‐MISS‐2021‐CANCER‐02‐03 program “Genial”
- The Italian Ministry of Health (Ministero della Salute), Ricerca Finalizzata 2016
- Italian ministry of Research (MUR) PRIN 2022
- The Italian Ministry of Health, Ricerca Finalizzata PNRR 2022
- The Italian Ministry of Health, Ricerca Finalizzata 2021
- Fondazione Patrimonio Ca’ Granda, “Liver BIBLE”
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Ricerca Corrente
Collapse
Affiliation(s)
- Luisa Ronzoni
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Serena Pelusi
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Vittoria Moretti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Francesco Malvestiti
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanItaly
| | - Hadi Eidgah Torghabehei
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden
| | - Jessica Rondena
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Cristiana Bianco
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Giulia Periti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Maria Rosaria De Filippo
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
- Clinical Nutrition Unit, Department of Medical and Surgical SciencesUniversity Magna GraeciaCatanzaroItaly
| | - Daniele Prati
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Luca Valenti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanItaly
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| |
Collapse
|
34
|
Wu Y, Zheng G, Zhang F, Li W. Association of high-sensitivity C-reactive protein with hepatic fibrosis in patients with metabolic dysfunction-associated steatotic liver disease. Front Immunol 2025; 16:1544917. [PMID: 39995674 PMCID: PMC11847791 DOI: 10.3389/fimmu.2025.1544917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
Objective This study aimed to investigate the association between high-sensitivity C-reactive protein (hsCRP) levels and hepatic fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) and assess its predictive efficacy. Methods The study included 1,477 participants from the United States and 1,531 from China diagnosed with MASLD. Liver stiffness measurement (LSM) and controlled attenuation parameter (CAP) were assessed by vibration-controlled transient elastography (VCTE) to evaluate the presence and degree of hepatic fibrosis and steatosis. The relationship between hsCRP levels and hepatic fibrosis in MASLD patients was examined using multivariable-adjusted and restricted cubic spline (RCS) models. Additionally, subgroup analyses were conducted to investigate the potential heterogeneity among different characteristic subgroups. Results The results demonstrated a significant correlation between elevated hsCRP levels and an increased risk of significant fibrosis, advanced fibrosis, and cirrhosis in the US cohort of MASLD patients (OR 2.22, 1.69, and 2.85, respectively; all P <0.05). The results of the Chinese cohort were consistent with those of the US cohort, and there was a significant and positive correlation between hsCRP levels and the risk of hepatic fibrosis in patients with MASLD (OR 2.53, 3.85, and 3.78, respectively, all P <0.001). The RCS analysis revealed a significant non-linear relationship between hsCRP levels and the degree of hepatic fibrosis, with disparate inflection point values observed across different cohorts (approximately 9 mg/L in the US cohort and 4 mg/L in the Chinese cohort). The impact of hsCRP levels on the risk of hepatic fibrosis varied across different subgroups with distinct characteristics. Conclusion The present study demonstrated a significant correlation between hsCRP levels and the degree of hepatic fibrosis in patients with MASLD, with notable dose-response relationships and subgroup differences.
Collapse
Affiliation(s)
- Yunfei Wu
- Department of Pathology, Changzhou Third People’s Hospital, Changzhou, China
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
| | - Guojun Zheng
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
- Clinical Laboratory, Changzhou Third People’s Hospital, Changzhou, China
| | - Fan Zhang
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
- Department of Endocrinology, Changzhou Third People’s Hospital, Changzhou, China
- Department of Clinical Nutrition, Changzhou Third People’s Hospital, Changzhou, China
| | - Wenjian Li
- Changzhou Clinical College, Xuzhou Medical University, Changzhou, China
- Department of Urology, Changzhou Third People’s Hospital, Changzhou, China
| |
Collapse
|
35
|
Tian ZF, Hu RY, Wang Z, Wang YJ, Li W. Molecular mechanisms behind the inhibitory effects of ginsenoside Rg3 on hepatic fibrosis: a review. Arch Toxicol 2025; 99:541-561. [PMID: 39729114 DOI: 10.1007/s00204-024-03941-w] [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: 08/27/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Hepatitis is a chronic inflammatory liver disease and an important cause of liver fibrosis, which can progress to cirrhosis and even hepatocellular carcinoma if left untreated. However, liver fibrosis is a reversible disease, so finding new intervention targets and molecular markers is the key to preventing and treating liver fibrosis. Ginseng, the roots of Panax ginseng C. A. Meyer, is a precious Traditional Chinese Medicines with high medicinal value and is known as the "king of all herbs", and its active ingredient, ginsenoside Rg3 is a rare saponin and a new class of drug, one of the most thoroughly and extensively studied in a large number of studies. Ginsenoside Rg3 is an active ingredient extracted from ginseng that possesses a variety of biological activities, including anti-inflammatory, antioxidant, and anti-fibrotic effects. Several studies have suggested that ginsenoside Rg3 may help reduce hepatic inflammation and oxidative stress, thereby slowing the progression of liver fibrosis. Ginsenoside Rg3 may have some therapeutic effects on liver fibrosis, and the underlying molecular mechanisms behind these effects are attributed to cellular autophagy, apoptosis, and anti-inflammation, as well as the modulation of antioxidant activity and multiple signaling pathways. The molecular mechanisms behind the inhibitory effect of ginsenoside Rg3 on hepatic fibrosis are reviewed, with a view to providing reference for related studies.
Collapse
Affiliation(s)
- Zhao-Feng Tian
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Rui-Yi Hu
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
| | - Ya-Jun Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
36
|
Pasta A, Formisano E, Calabrese F, Marabotto E, Furnari M, Bodini G, Torres MCP, Pisciotta L, Giannini EG, Zentilin P. From Dysbiosis to Hepatic Inflammation: A Narrative Review on the Diet-Microbiota-Liver Axis in Steatotic Liver Disease. Microorganisms 2025; 13:241. [PMID: 40005608 PMCID: PMC11857840 DOI: 10.3390/microorganisms13020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The gut microbiota has emerged as a critical player in metabolic and liver health, with its influence extending to the pathogenesis and progression of steatotic liver diseases. This review delves into the gut-liver axis, a dynamic communication network linking the gut microbiome and liver through metabolic, immunological, and inflammatory pathways. Dysbiosis, characterized by altered microbial composition, contributes significantly to the development of hepatic steatosis, inflammation, and fibrosis via mechanisms such as gut barrier dysfunction, microbial metabolite production, and systemic inflammation. Dietary patterns, including the Mediterranean diet, are highlighted for their role in modulating the gut microbiota, improving gut-liver axis integrity, and attenuating liver injury. Additionally, emerging microbiota-based interventions, such as fecal microbiota transplantation and bacteriophage therapy, show promise as therapeutic strategies for steatotic liver disease. However, challenges such as population heterogeneity, methodological variability, and knowledge gaps hinder the translational application of current findings. Addressing these barriers through standardized approaches and integrative research will pave the way for microbiota-targeted therapies to mitigate the global burden of steatotic liver disease.
Collapse
Affiliation(s)
- Andrea Pasta
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
| | - Elena Formisano
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesco Calabrese
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Manuele Furnari
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giorgia Bodini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Maria Corina Plaz Torres
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Livia Pisciotta
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Edoardo Giovanni Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Patrizia Zentilin
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
37
|
Galvanin C, Buch S, Nahon P, Trépo E. PNPLA3 in Alcohol-Related Liver Disease. Liver Int 2025; 45:e16211. [PMID: 39679853 DOI: 10.1111/liv.16211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
The discovery of PNPLA3 as a genetic risk factor for liver disease has transformed our understanding of the pathogenesis of alcohol-related liver disease (ALD). The recent reclassification of fatty liver disease as steatotic liver disease (SLD), introducing metabolic dysfunction and alcohol-related liver disease (MetALD), has highlighted how genetic and environmental factors synergistically drive liver damage. The PNPLA3 rs738409 variant stands as a paradigmatic example of gene-environment interaction, where its effect on liver disease is dramatically amplified by alcohol consumption, obesity and type 2 diabetes. Understanding these interactions has revealed novel pathogenic mechanisms. The robust genetic evidence and a growing understanding of molecular mechanisms have made PNPLA3 an attractive therapeutic target. Several compounds targeting PNPLA3 are now in clinical development. While initial trials have focused on metabolic dysfunction-associated SLD, the recognition that almost all individuals with excessive alcohol consumption have metabolic comorbidities provides an unprecedented opportunity to evaluate these genetically informed therapies in MetALD. In this review, we examine the role of PNPLA3 in ALD, focusing on gene-environment interactions and therapeutic implications in the context of the new disease classification framework.
Collapse
Affiliation(s)
- Clélia Galvanin
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Stephan Buch
- Department of Medicine I, Dresden University Hospital, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Pierre Nahon
- Service d'Hépatologie, AP-HP Avicenne, Bobigny, France
- Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Team «Functional Genomics of Solid Tumors», Paris, France
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
38
|
Yang W, Pan Q, Li Q, Zhou S, Cao X. A simple, reliable and easily generalizable cell-based assay for screening potential drugs that inhibit lipid accumulation. Curr Res Toxicol 2024; 8:100213. [PMID: 39834517 PMCID: PMC11743863 DOI: 10.1016/j.crtox.2024.100213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Ectopic lipid deposition in the hepatocyte plays an important role in the development of nonalcoholic fatty liver disease (NAFLD), which has become one of the most common causes of chronic liver disease worldwide yet no approved drugs are currently available. In this study, a cell-based method was developed to screen potential drugs with low toxicity that inhibit lipid accumulation. In the same 96-well plate, cytotoxicity was measured using CCK8 assay, followed by lipid content detection using BODIPY 493/503 via fluorometry assay, a lipid droplet-specific fluorescent dye commonly used in microscopy and flow cytometry, but not previously reported in fluorometry. Lipid content was normalized to DAPI staining to control for cell number. The results of this assay were highly consistent with the fluorescence microscopy, with significantly lower intra-group variability in detecting lipid accumulation induced by free fatty acids in Huh7 cells. Validation was conducted using 10 well documented steatotic compounds and 5 negative controls, all of which were correctly identified by the assay. In addition, the inhibitory effect of ML261, a well-known inhibitor of hepatic lipid droplets formation, was also confirmed by the assay both in AML12 cells and Hepa1-6 cells. To our knowledge, this study is the first to quantify lipid droplets using BODIPY 493/503 by fluorometry assay, and to demonstrate that CCK8 does not interfere with subsequent BODIPY 493/503 staining, both of which will reduce the cost and increase the efficiency. In conclusion, the method is simple, reliable, efficient and does not rely on expensive instruments, making it an easily generalizable approach to identify potential drug candidates for NAFLD treatment.
Collapse
Affiliation(s)
- Weili Yang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Qiuyue Pan
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Qi Li
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Sirui Zhou
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xi Cao
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
39
|
Weiskirchen R, Lonardo A. PNPLA3 as a driver of steatotic liver disease: navigating from pathobiology to the clinics via epidemiology. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2024; 8:355-77. [DOI: 10.20517/jtgg.2024.70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Steatotic liver disease (SLD), particularly metabolic dysfunction-associated SLD, represents a significant public health concern worldwide. Among the various factors implicated in the development and progression of this condition, the patatin-like phospholipase domain-containing protein 3 (PNPLA3 ) gene has emerged as a critical player. Variants of PNPLA3 are associated with altered lipid metabolism, leading to increased hepatic fat accumulation and subsequent inflammation and fibrosis. Understanding the role of PNPLA3 not only enhances our comprehension of the pathomechanisms driving SLD but also informs potential therapeutic strategies. The molecular mechanisms through which PNPLA3 variants contribute to lipid dysregulation and hepatocyte injury in SLD are critically discussed in the present review article. We extensively analyze clinical cohorts and population-based studies underpinning the association between PNPLA3 polymorphisms and the risk of developing SLD, and its liver-related and protean extrahepatic outcomes, in concert with other risk modifiers, notably including age, sex, and ethnicity in adults and children. We also discuss the increasingly recognized role played by the PNPLA3 gene in liver transplantation, autoimmune hepatitis, and acquired immunodeficiency syndrome. Finally, we examine the clinical implications of PNPLA3 diagnostics regarding risk stratification and targeted therapies for patients affected by SLD in the context of precision medicine approaches.
Collapse
|