351
|
Zhang Y, Ju F, Yan L, Shen X, Guo S, Yu M, Cao Y, Wang W. Elevated Porcupine Disrupts Lipid Metabolism and Promotes Inflammatory Response in MASLD. Liver Int 2025; 45:e16130. [PMID: 39403838 DOI: 10.1111/liv.16130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 03/11/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) presents a high incidence globally and is a major cause of cirrhosis and hepatocellular carcinoma, lacking of efficient interventions. Patients with MASLD exhibit exceeded serum levels of palmitic acid (PA). However, the association between PA and MASLD remains obscure. METHODS Gene expression omnibus dataset analysis, western blotting, mRNA-sequencing, RT-qPCR, a click chemistry-immunoprecipitation-immunofluorescence system, ELISA, lipid extraction and UHPLC-MS/MS analysis, CyTOF mass cytometry, gene knockdown via lentivirus-mediated shRNA, and high-fat methionine and choline-deficient diet-fed WT and db/db mice models were used to reveal the expression and functions of Porcupine in MASLD development both in vitro and in vivo. RESULTS Our findings show that PA, as a crucial substrate for protein palmitoylation, induced the expression of palmitoyltransferase Porcupine in a time-dependent manner. This induction was closely associated with dysregulated lipid metabolism and stimulated inflammatory response observed in vitro. Porcupine protein levels were significantly increased in liver tissues from both MASLD mice models, which was predominantly localised in lipid droplet-rich hepatocytes. Pharmacological inhibition of Porcupine by Wnt974 markedly ameliorated the aberrant lipid accumulation and inflammatory response in mouse livers. Furthermore, increased Porcupine positively correlated with CD36 at protein levels, and its inhibition or knockdown decreased CD36 protein levels via mechanisms irrelevant to transcriptional regulation, but primarily dependent on protein palmitoylation. CONCLUSIONS The current study reveals that PA-induced Porcupine disrupts lipid metabolism and promotes inflammatory response during MASLD development, which can be ameliorated by the Porcupine inhibitor Wnt974. Therefore, Porcupine may be a potential pharmacological target for the treatment of MASLD.
Collapse
Affiliation(s)
- Yalin Zhang
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fengyu Ju
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Li Yan
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xin Shen
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shiqing Guo
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Muchen Yu
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yujia Cao
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wenhui Wang
- Department of Pharmacology and School of Basic Medicine Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
352
|
Zhao M, Han X, Fan H, Liang C, Wang H, Zhang X, Zhao S, Guo C, Liu Z, Zhang T. Metabolic Dysfunction-Associated Steatotic Liver Disease Increases the Risk of Severe Infection: A Population-Based Cohort Study. Liver Int 2025; 45:e16136. [PMID: 39422294 DOI: 10.1111/liv.16136] [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: 07/03/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is linked to various intrahepatic and extrahepatic diseases, but its association with severe infectious disease remains to be investigated. METHODS We analysed data from the Shanghai Suburban Adult Cohort and Biobank, encompassing participants enrolled in 2016 and 2017 with available abdominal ultrasonography data, and followed them up until December 2022 (median follow-up = 5.71 years). We categorised the participants into the MASLD group and those without steatotic liver disease (non-SLD). Multivariable-adjusted Cox regression was used to estimate hazard ratios (HR) for severe infections in patients with MASLD compared to the non-SLD group. Cumulative incidences were calculated while accounting for competing risks (non-infection-related deaths). Mediation analyses were performed to explore the roles of cardiometabolic risk factors in the association between MASLD and severe infections. RESULTS Among the 33 072 eligible participants (mean age 56.37 years; 38.20% male), 11 908 (36.01%) were diagnosed with MASLD at baseline. Severe infections occurred in 912 (7.66%) MASLD patients and 1258 (5.94%) non-SLD. The rate of severe infections per 1000 person-years was higher in MASLD patients (13.58) than in comparators (10.48) (fully adjusted HR 1.18, 95% CI 1.07-1.30). The most frequent infections in MASLD were respiratory (7.25/1000 person-years) and urinary tract infections (2.61/1000 person-years). The 5-year cumulative incidence of severe infections was 6.79% (95% CI 6.36-7.26) in MASLD and 5.08% (95% CI 4.79-5.38) in comparators. Cardiometabolic risk factors, including waist circumference, triglycerides and HbA1C, partially mediate the association between MASLD and severe infections. CONCLUSIONS Patients with MASLD were at significantly higher risk of incident severe infections compared to the non-SLD group. Future studies are needed to elucidate the mechanisms linking MASLD to severe infections.
Collapse
Affiliation(s)
- Ming Zhao
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Xinyu Han
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Hong Fan
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Chenyu Liang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Chengnan Guo
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- Yiwu Research Institute, Fudan University, Yiwu, China
| |
Collapse
|
353
|
Cathcart J, Barrett R, Bowness JS, Mukhopadhya A, Lynch R, Dillon JF. Accuracy of Non-Invasive Imaging Techniques for the Diagnosis of MASH in Patients With MASLD: A Systematic Review. Liver Int 2025; 45:e16127. [PMID: 39400428 PMCID: PMC11891385 DOI: 10.1111/liv.16127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/14/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health problem. The secondary stage in MASLD is steatohepatitis (MASH), the co-existence of steatosis and inflammation, a leading cause of progression to fibrosis and mortality. MASH resolution alone improves survival. Currently, MASH diagnosis is via liver biopsy. This study sought to evaluate the accuracy of imaging-based tests for MASH diagnosis, which offer a non-invasive method of diagnosis. METHODS Eight academic literature databases were searched and references of previous systematic reviews and included papers were checked for additional papers. Liver biopsy was used for reference standard. RESULTS We report on 69 imaging-based studies. There were 31 studies on MRI, 27 on ultrasound, five on CT, 13 on transient elastography, eight on controlled attenuation parameter (CAP) and two on scintigraphy. The pathological definition of MASH was inconsistent, making it difficult to compare studies. 55/69 studies (79.71%) were deemed high-risk of bias as they had no preset thresholds and no validation. The two largest groups of imaging papers were on MRI and ultrasound. AUROCs were up to 0.93 for MRE, 0.90 for MRI, 1.0 for magnetic resonance spectroscopy (MRS) and 0.94 for ultrasound-based studies. CONCLUSIONS Our study found that the most promising imaging tools are MRI techniques or ultrasound-based scores and confirmed there is potential to utilise these for MASH diagnosis. However, many publications are single studies without independent prospective validation. Without this, there is no clear imaging tool or score currently available that is reliably tested to diagnose MASH.
Collapse
Affiliation(s)
- Jennifer Cathcart
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
- Gastroenterology DepartmentAberdeen Royal InfirmaryAberdeenUK
| | - Rachael Barrett
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| | - James S. Bowness
- University College London Hospitals NHS Foundation TrustLondonUK
- Department of Targeting InterventionUniversity College LondonLondonUK
| | | | - Ruairi Lynch
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| | - John F. Dillon
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
354
|
Li X, Jin S, Wang D, Wu Y, Tang X, Liu Y, Yao T, Han S, Sun L, Wang Y, Hou SX. Accumulation of Damaging Lipids in the Arf1-Ablated Neurons Promotes Neurodegeneration through Releasing mtDNA and Activating Inflammatory Pathways in Microglia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414260. [PMID: 40019378 PMCID: PMC12021055 DOI: 10.1002/advs.202414260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/02/2025] [Indexed: 03/01/2025]
Abstract
Lipid metabolism disorders in both neurons and glial cells have been found in neurodegenerative (ND) patients and animal models. However, the pathological connection between lipid droplets and NDs remains poorly understood. The recent work has highlighted the utility of a neuron-specific Arf1-knockout mouse model and corresponding cells for elucidating the nexus between lipid metabolism disorders and amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS). In this study, it is found that Arf1 deficiency first induced surplus fatty acid synthesis through the AKT-mTORC1-SREBP1-FASN axis, which further triggered endoplasmic reticulum (ER)-mitochondrial stress cascade via calcium flux. The organelle stress cascade further caused mitochondrial DNA (mtDNA) to be released into cytoplasm. Concurrently, the FASN-driven fatty acid synthesis in the Arf1-deficient neurons might also induce accumulation of sphingolipids in lysosomes that caused dysfunction of autophagy and lysosomes, which further promoted lysosomal stress and mitochondria-derived extracellular vesicles (MDEVs) release. The released MDEVs carried mtDNA into microglia to activate the inflammatory pathways and neurodegeneration. The studies on neuronal lipid droplets (LDs) and recent studies of microglial LDs suggest a unified pathological function of LDs in NDs: activating the inflammatory pathways in microglia. This finding potentially provides new therapeutic strategies for NDs.
Collapse
Affiliation(s)
- Xu Li
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Shuhan Jin
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Danke Wang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Ying Wu
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Xiaoyu Tang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Yufan Liu
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Tiange Yao
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid SurfacesDepartment of Chemical BiologyCollege of Chemistry and Chemical EngineeringThe Key Laboratory for Chemical Biology of Fujian ProvinceThe MOE Key Laboratory of Spectrochemical Analysis & InstrumentationInnovation Center for Cell Signalling NetworkXiamen UniversityXiamen361005China
| | - Lin Sun
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Yuetong Wang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| | - Steven X. Hou
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyChildren's HospitalZhongshan HospitalFudan UniversityShanghai200438China
| |
Collapse
|
355
|
Seko Y, Lin H, Wong VWS, Okanoue T. Impact of PNPLA3 in Lean Individuals and in Cryptogenic Steatotic Liver Disease. Liver Int 2025; 45:e16164. [PMID: 39540675 DOI: 10.1111/liv.16164] [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: 06/28/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Although metabolic dysfunction-associated steatotic liver disease (MASLD) is strongly associated with obesity, around 20% of individuals with hepatic steatosis may nonetheless have normal body mass index, a condition often referred to as lean nonalcoholic fatty liver disease (NAFLD). Under the new nomenclature and definition of MASLD, lean NAFLD can be further divided into lean MASLD (when there is one or more cardiometabolic risk factors) and cryptogenic steatotic liver disease (when there is no cardiometabolic risk factor). RESULTS Current studies suggest that the at-risk PNPLA3 rs738409 variant is more common among individuals with lean NAFLD than their overweight and obese counterparts. However, even in this group, cardiometabolic risk factors are often required for the development of hepatic steatosis and liver injury. In the general population, PNPLA3 gene polymorphism is associated with an increased risk of MASLD, more severe liver histology (i.e., the presence of steatohepatitis and fibrosis) and future development of hepatocellular carcinoma and cirrhotic complications. Emerging data also suggest that individuals carrying the PNPLA3 GG genotype might have a greater reduction in hepatic steatosis and liver enzymes with lifestyle intervention and metabolic treatments, such as glucagon-like peptide-1 receptor agonists. CONCLUSION Studies have not specifically examined the impact of PNPLA3 in lean individuals.
Collapse
Affiliation(s)
- Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Huapeng Lin
- Department of Gastroenterology and Hepatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Medical Data Analytics Center, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| |
Collapse
|
356
|
Konings LAM, Miguelañez-Matute L, Boeren AMP, van de Luitgaarden IAT, Dirksmeier F, de Knegt RJ, Tushuizen ME, Grobbee DE, Holleboom AG, Cabezas MC. Pharmacological treatment options for metabolic dysfunction-associated steatotic liver disease in patients with type 2 diabetes mellitus: A systematic review. Eur J Clin Invest 2025; 55:e70003. [PMID: 39937036 DOI: 10.1111/eci.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is closely related to type 2 diabetes mellitus (T2DM) through a common root in insulin resistance. The more severe stage, metabolic dysfunction-associated steatohepatitis (MASH), increases the risk for cardiovascular complications, liver cirrhosis and hepatocellular carcinoma. Several trials investigating established antidiabetic-drugs in patients with T2DM and MASLD have yielded promising results. Therefore, we aimed to systematically review the effect of T2DM-drug treatment on MALSD parameters. METHODS Medical databases were searched until January 2025 for controlled trials in patients with T2DM and MASLD/MASH. Studies that evaluated the effect of T2DM-medication on the severity of MASLD/MASH in T2DM patients were included. The quality of the studies was assessed by three independent reviewers using a set of Cochrane risk-of-bias tools. RESULTS Of 1748 references, 117 studies fulfilled the inclusion-criteria and were assessed for eligibility in full-text. Fifty-two articles were included. Data included a total of 64.708 patients and study populations ranged from 9 to 50.742. Heterogeneity in study-design and analysis hampered the comparability of the results. Most evidence was present for GLP-1 receptor agonists, SGLT2-inhibitors and PPAR-γ-agonists for regression of liver fibrosis and MASH. CONCLUSION Studies on the value of T2DM-drug treatment in the improvement of MASLD vary significantly in study design, size and quality. GLP-1 receptor agonists, PPAR-γ-agonists, SGLT2-inhibitors may all be preferred pharmacological interventions for patients with MASLD/MASH and T2DM. Newer agents like dual GLP-1/GIP or triple GLP-1/GIP/Glucagon agonists will likely play an important role in the treatment of MASLD/MASH in the near future.
Collapse
Affiliation(s)
- Laura A M Konings
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
- Department of Internal Medicine and Endocrinology, Erasmus MC, Rotterdam, the Netherlands
| | | | - Anna M P Boeren
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
| | | | - Femme Dirksmeier
- Department of Gastroenterology and Hepatology, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
| | - Rob J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, the Netherlands
| | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, LUMC, Leiden, the Netherlands
| | | | | | - Manuel Castro Cabezas
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
- Department of Internal Medicine and Endocrinology, Erasmus MC, Rotterdam, the Netherlands
- Julius Clinical, Zeist, the Netherlands
| |
Collapse
|
357
|
Schneider KM, Schneider CV. Editorial: Lipidomics in MASLD and MetALD-Authors' Reply. Aliment Pharmacol Ther 2025; 61:1410-1411. [PMID: 39991947 DOI: 10.1111/apt.70058] [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: 02/16/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Affiliation(s)
- Kai Markus Schneider
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Carolin V Schneider
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
358
|
Tan KCB. What is new in metabolic dysfunction-associated steatotic liver disease? J Diabetes Investig 2025; 16:581-583. [PMID: 39891535 PMCID: PMC11970290 DOI: 10.1111/jdi.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
The change in nomenclature from nonalcoholic fatty liver disease to metabolic dysfunction associated-steatotic liver disease has emphasized the importance of metabolic abnormalities in this liver disorder. New pharmacological therapy has recently become available and resmetirom, a thyroid hormone receptor agonist, has received approval for the treatment of non-cirrhotic metabolic dysfunction-associated steatohepatitis and significant liver fibrosis.
Collapse
Affiliation(s)
- Kathryn CB Tan
- Department of Medicine, School of Clinical MedicineThe University of Hong KongHong KongHong Kong
| |
Collapse
|
359
|
Ferrarese A, Hurtado Díaz de León I, Tapper EB, Burra P. Sexual health and function in liver disease. Hepatol Commun 2025; 9:e0691. [PMID: 40178496 PMCID: PMC11970893 DOI: 10.1097/hc9.0000000000000691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Sex is a central aspect of human life and is significantly impacted by chronic illness. Cirrhosis, due to its unique pathophysiology and the side effects of common therapies, serves as a paradigmatic example, being associated with very high rates of sexual dysfunction in both men and women. Liver transplantation can modify certain hormonal and pathophysiological aspects related to sexual dysfunction, but complete recovery occurs in only a relatively small percentage of patients. This review examines the pathophysiology, epidemiology, and management of sexual and reproductive dysfunction in patients with cirrhosis and those undergoing liver transplantation. It provides a framework for understanding the sources of dysfunction, tools for identifying it in clinical settings, and interventions to improve sexual health and functioning in these patients.
Collapse
Affiliation(s)
- Alberto Ferrarese
- Multivisceral Transplant Unit, Gastroenterology, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Ivonne Hurtado Díaz de León
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Elliot B. Tapper
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrizia Burra
- Multivisceral Transplant Unit, Gastroenterology, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| |
Collapse
|
360
|
Boeckmans J, Widman L, Shang Y, Strandberg R, Wester A, Schattenberg JM, Hagström H. Risk of hepatic decompensation or HCC is similar in patients with ALD- and MASLD-cirrhosis: A population-based cohort study. Eur J Intern Med 2025; 134:104-113. [PMID: 39952814 DOI: 10.1016/j.ejim.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND It is unclear if the risk of hepatic decompensation or hepatocellular carcinoma (HCC) differs between patients with compensated alcohol-related liver disease (ALD)- and metabolic dysfunction-associated steatotic liver disease (MASLD)-cirrhosis. We investigated the risk to develop hepatic decompensation or HCC based on ALD or MASLD as the underlying etiology of cirrhosis. METHODS All patients with a new diagnosis in hospital-based outpatient care of ALD- or MASLD-cirrhosis in Sweden between 2002 and 2020 were identified using national registers. Hepatic decompensation was analyzed as a composite outcome with HCC. Cox regression was employed to compare rates of hepatic decompensation or HCC, and subsequent death. RESULTS 1660 patients with ALD-cirrhosis and 943 patients with MASLD-cirrhosis were identified. The median ages were 64 years (IQR 57-70) and 69 years (IQR 62-75) in patients with ALD- and MASLD-cirrhosis, respectively. Patients with ALD-cirrhosis consisted of 69.4 % males, compared to 47.6 % males in the MASLD-cirrhosis group. 581 (35 %) patients with ALD-cirrhosis and 284 (30 %) patients with MASLD-cirrhosis developed hepatic decompensation or HCC (median follow-up time: 25 months), resulting in an adjusted hazard ratio of 1.12 (ALD- vs. MASLD-cirrhosis, 95 %-confidence interval=0.88-1.41). The adjusted risk of mortality afterwards was lower in patients with ALD-cirrhosis compared to patients with MASLD-cirrhosis (adjusted hazard ratio 0.62, 95 %-confidence interval=0.39-0.97). CONCLUSIONS The risk of hepatic decompensation or HCC is comparable in patients with ALD- and MASLD-cirrhosis, but the risk of mortality after a decompensation event or HCC tends to be higher in patients with MASLD-cirrhosis.
Collapse
Affiliation(s)
- Joost Boeckmans
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; In Vitro Liver Disease Modelling team - Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Linnea Widman
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Ying Shang
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Rickard Strandberg
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Axel Wester
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Jörn M Schattenberg
- Department of Medicine II, University Medical Center Homburg, Homburg and Saarland University, Saarbrücken, Germany.
| | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
361
|
Arab JP, Díaz LA, Rehm J, Im G, Arrese M, Kamath PS, Lucey MR, Mellinger J, Thiele M, Thursz M, Bataller R, Burton R, Chokshi S, Francque SM, Krag A, Lackner C, Lee BP, Liangpunsakul S, MacClain C, Mandrekar P, Mitchell MC, Morgan MY, Morgan TR, Pose E, Shah VH, Shawcross D, Sheron N, Singal AK, Stefanescu H, Terrault N, Trépo E, Moreno C, Louvet A, Mathurin P. Metabolic dysfunction and alcohol-related liver disease (MetALD): Position statement by an expert panel on alcohol-related liver disease. J Hepatol 2025; 82:744-756. [PMID: 39608457 DOI: 10.1016/j.jhep.2024.11.028] [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: 07/25/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Abstract
In this position statement, we explore the intricate relationship between alcohol intake and metabolic dysfunction in the context of the 2023 nomenclature update for steatotic liver disease (SLD). Recent and lifetime alcohol use should be accurately assessed in all patients with SLD to facilitate classification of alcohol use in grams of alcohol per week. Alcohol biomarkers (i.e., phosphatidylethanol), use of validated questionnaires (i.e. AUDIT-C [alcohol use disorders identification test consumption]), and collateral information from friends and relatives could help facilitate differentiation between alcohol-related liver disease (ALD) per se and liver disease with both metabolic and alcohol-related components (MetALD). Heavy alcohol use can contribute to cardiometabolic risk factors such as high blood pressure, hypertriglyceridaemia, and hyperglycaemia. As a result, caution should be exercised in the application of only one metabolic dysfunction criterion to diagnose MASLD, as suggested in the 2023 nomenclature document, particularly in individuals exceeding weekly alcohol use thresholds of 140 g for women and 210 g for men. This is particularly important in those individuals with isolated high blood pressure, hypertriglyceridaemia, or hyperglycaemia, where the disease process may be driven by alcohol itself. Additionally, metabolic dysfunction and alcohol use should be reassessed over time, especially after periods of change in risk factor exposure. This approach could ensure a more accurate prognosis and effective management of SLD, addressing both metabolic and alcohol-related factors.
Collapse
Affiliation(s)
- Juan Pablo Arab
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Luis Antonio Díaz
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile; MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
| | - Jürgen Rehm
- Institute for Mental Health Policy Research, Campbell Family Mental Health Research Institute, PAHO/WHO Collaborating Centre, Centre for Addiction and Mental Health, Toronto, Canada
| | - Gene Im
- Division of Liver Diseases, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marco Arrese
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patrick S Kamath
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Michael R Lucey
- Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jessica Mellinger
- Department of Internal Medicine, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Maja Thiele
- Odense Liver Research Centre, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Mark Thursz
- Department of Metabolism, Digestion & Reproduction, Imperial College London, UK
| | - Ramon Bataller
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Robyn Burton
- Institute for Social Marketing and Health. University of Stirling, UK
| | - Shilpa Chokshi
- Institute of Hepatology Foundation for Liver Research London UK; School of Immunology and Microbial Sciences King's College London, London, UK
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Aleksander Krag
- Odense Liver Research Centre, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Carolin Lackner
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Brian P Lee
- Division of Gastrointestinal and Liver Diseases, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology, Department of Internal Medicine, and Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Craig MacClain
- Department of Gastroenterology and Hepatology, University of Louisville, Louisville, KY, USA
| | - Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mack C Mitchell
- Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marsha Y Morgan
- UCL Institute for Liver & Digestive Health, Division of Medicine, Royal Free Campus, University College London, London, UK
| | - Timothy R Morgan
- VA Long Beach Healthcare System - Gastroenterology Section, Long Beach, CA, USA
| | - Elisa Pose
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Nick Sheron
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, Kings College London, UK
| | - Ashwani K Singal
- Department of Gastroenterology and Hepatology, University of Louisville, Louisville, KY, USA
| | - Horia Stefanescu
- Liver Unit, Regional Institute of Gastroenterology and Hepatology "Octavian Fodor," University of Medicine and Pharmacy "Iuliu Hatieganu," Cluj-Napoca, Romania
| | - Norah Terrault
- Division of Gastrointestinal and Liver Diseases, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Christophe Moreno
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandre Louvet
- CHRU de Lille, Hôpital Claude Huriez, Rue M. Polonovski CS 70001, 59 037 Lille Cedex, France
| | - Philippe Mathurin
- CHRU de Lille, Hôpital Claude Huriez, Rue M. Polonovski CS 70001, 59 037 Lille Cedex, France.
| |
Collapse
|
362
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [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: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
363
|
Peng Y, Wang P, Liu F, Wang X, Si C, Gong J, Zhou H, Gu J, Qin A, Song W, Song F. Metabolic dysfunction-associated steatotic liver disease and cancer risk: A cohort study. Diabetes Obes Metab 2025; 27:1940-1949. [PMID: 39781580 DOI: 10.1111/dom.16186] [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: 11/08/2024] [Revised: 12/19/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Fatty liver disease may be associated with increased risks of intrahepatic and extrahepatic cancers. Our objective was to investigate associations between new subcategories of steatotic liver disease (SLD) recently proposed by nomenclature consensus group and cancer risk. METHODS A total of 283 238 participants from the UK Biobank were included. Based on information on cardiometabolic factors, alcohol consumption and the specific aetiology of SLD, individuals were categorized into four groups: no SLD (n = 170 885), metabolic dysfunction-associated steatotic liver disease (MASLD, n = 74 510), MASLD with increased alcohol intake (MetALD, n = 23 320) and other SLD (n = 6718). Outcomes were overall incident cancer (n = 39 352) and 21 site-specific cancers. The Cox proportional hazards model was used to estimate relationships between subcategories of SLD and cardiometabolic factors in MASLD with cancer risk. Population attributable risk (PAR) of cancer associated with SLD was estimated. RESULTS MASLD was the most prevalent SLD in the general population. All SLD subcategories were associated with elevated risks of overall cancer, digestive system cancers (except gastric cancer) and breast cancer (HRs 1.079-4.663). Additionally, MASLD was associated with increased risks of renal cancer, endometrial cancer and Hodgkin lymphoma. Compared to MetALD and other SLDs, MASLD has a higher PAR% for the majority of aforementioned cancers. This could be largely explained by its common metabolic abnormalities, dominantly characterized by overweight/obesity and elevated blood pressure, concomitant with hyperglycaemia and hyperlipidaemia. CONCLUSIONS All subcategories of SLD, particularly MASLD with multiple metabolic abnormalities, were associated with increased risks of multiple cancers, providing a new perspective for cancer prevention.
Collapse
Affiliation(s)
- Yu Peng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Peng Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fubin Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xixuan Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Changyu Si
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jianxiao Gong
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Huijun Zhou
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jiale Gu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ailing Qin
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Weijie Song
- Department of Laboratory Animal Center, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
364
|
Mikołajczyk-Stecyna J, Zuk E, Chmurzynska A, Blatkiewicz M, Jopek K, Rucinski M. Exposure to a choline-deficient diet during pregnancy and lactation alters the liver transcriptome profile in offspring of dams with fatty liver. Clin Nutr ESPEN 2025; 66:9-23. [PMID: 39800134 DOI: 10.1016/j.clnesp.2025.01.028] [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: 10/28/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS The developmental origin of health and disease hypothesis shows that early adverse exposures can have lifelong health effects. Thus, the aim of this study was to analyze the impact of choline intake during pregnancy and/or lactation on gene expression profiles in the liver of 24-day-old male rat offspring from dams with non-alcoholic fatty liver disease (NAFLD). METHODS Phenotypic characteristic, histological examination and global transcriptome pattern of liver tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global gene expression profile was analyzed by using microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time polymerase chain reaction (qPCR). RESULTS The histological examination of rat liver sections indicated alternations typical for fatty liver in all analyzed groups with increased progression among groups deprived of choline. Choline deficiency in the maternal diet was associated with changes in body mass and composition but not with biochemical marker levels, except for the high density lipoprotein fraction of cholesterol (HDL). Enhanced expression of genes involved in oxidative stress, cell proliferation, activation of catabolic processes related to hepatocyte dysfunction and cell membrane composition were simultaneously observed in all choline-deficient groups. CONCLUSIONS An adequate amount of choline in the diet of a mother with fatty liver during pregnancy and/or lactation can regulate gene expression in the offspring's liver and contribute to a milder stage of the disease in the progeny. Moreover, proper choline supply during the postpartum period is as crucial as during the prenatal period.
Collapse
Affiliation(s)
- Joanna Mikołajczyk-Stecyna
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland.
| | - Ewelina Zuk
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Agata Chmurzynska
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | | | - Karol Jopek
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Marcin Rucinski
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| |
Collapse
|
365
|
He Y, Hu M, Miao X, Xu F, Deng J, Song Z, Li M, Ming Y, Leng S. Dynamic Status of Systemic Immune Inflammation Index Is Associated With Metabolic Dysfunction-Associated Steatotic Liver Disease: An Evidence From a Ten-Year Prospective Longitudinal Cohort Study. J Inflamm Res 2025; 18:4595-4606. [PMID: 40191090 PMCID: PMC11972002 DOI: 10.2147/jir.s509814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Objective Previous research studies have linked the systemic immune inflammation index (SII), derived from a complete blood count, to metabolic dysfunction-associated steatotic liver disease (MASLD). However, evidence on the relationship between longitudinal changes in SII and MASLD remains limited. This study aimed to explore distinct SII trajectories and their association with MASLD incidence. Methods A longitudinal study analyzed 25,600 individuals who underwent periodic health assessments at a Dalian City hospital between 2014 and 2023. MASLD was diagnosed via ultrasound. The SII was calculated using the formula SII = (platelet count × neutrophil count) / lymphocyte count. Group-based trajectory modeling was used to identify SII trajectories, and restricted cubic spline (RCS) analysis was employed to assesse the dose-response relationship. Stratified analyses and sensitivity analyses were also conducted. Results Three SII trajectories were identified: "low stable" (50.6%), "moderate stable" (35.1%), and "high stable" (8.9%). After adjustments, the hazard ratios (HR) for MASLD incidence were 1.118 (95% CI: 1.057-1.182, P<0.001) for the "moderate stable" group and 1.284 (95% CI: 1.172-1.408, P<0.001) for the "high stable" group. These associations persisted after adjusting for lifestyle factors. A significant non-linear relationship between SII and MASLD risk was found in both the overall population and among different genders. Subgroup and sensitivity analyses consistently confirmed these findings. Conclusion Elevated SII levels are significantly associated with an increased risk of MASLD, particularly among individuals under 45 and women. Regular SII monitoring may improve risk stratification and facilitate targeted prevention strategies for those at higher risk of MASLD.
Collapse
Affiliation(s)
- Yangxuan He
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Manling Hu
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Xinlei Miao
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Fei Xu
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| | - Jiayi Deng
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Ziping Song
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Meng Li
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| | - Yunxiang Ming
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Department of Gastroenterology,the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Song Leng
- Health Management Center, the Second Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- School of Public Health. Dalian Medical University, Dalian, People’s Republic of China
| |
Collapse
|
366
|
Vaz K, Kemp W, Majeed A, Lubel J, Magliano DJ, Glenister KM, Bourke L, Simmons D, Roberts SK. MAFLD but not MASLD increases risk of all-cause mortality in regional Australia, with components of metabolic syndrome exacerbating factors: 20 year longitudinal, cohort study. Hepatol Int 2025; 19:384-394. [PMID: 39673677 DOI: 10.1007/s12072-024-10748-5] [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: 06/26/2024] [Accepted: 11/03/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS Controversy remains whether the mortality risk in people with fatty liver disease (FLD) including metabolic-(dysfunction) associated steatotic liver disease (MASLD) and metabolic-(dysfunction) associated fatty liver disease (MAFLD) is higher than observed in those without FLD. We aimed to determine the mortality rate and mortality rate ratio (MRR) for these FLDs. METHODS The study population was a randomly selected cohort of community-dwelling adults in regional Victoria, Australia between 2001 and 2003 with sufficient data evaluable for Fatty Liver Index and determination on alcohol consumption. MASLD and MAFLD were diagnosed by established criteria. The primary outcome was overall mortality and main secondary outcome was major adverse liver outcomes (MALO) (i.e., decompensated liver disease, primary liver cancer and liver-related death). Non-fatal and fatal outcomes were captured via data linkage to hospital admission, cancer registry, and death registries. MRR was calculated with non-FLD participants as the comparator. RESULTS 1444 (99.3%) and 1324 (91.1%) individuals from a total of 1454 were included in the final MAFLD and MASLD analyses. The median follow-up was 19.7 years (IQR 19.1-20.1) and there were 298 deaths. The MRR for MAFLD and MASLD was 1.39 (95% CI 1.10-1.76) and 1.25 (95% CI 0.96-1.61), respectively. MAFLD persisted as a risk factor for all-cause death on multivariable models correcting for lifestyle and socioeconomic variables, but not when adjusted for metabolic risk factors. MALOs were increased in MAFLD [incidence rate ratio (IRR) 3.03, 95% CI 1.22-8.18] and MASLD (IRR 2.80, 95% CI 1.05-7.90). Metabolic risk factors increased the risk of overall mortality and MALO, and cancer (34.3-34.6%) and cardiovascular disease (30.1-33.7%) were the most common cause of death in FLD. CONCLUSION In this population-based longitudinal study, MAFLD but not MASLD increases the risk of overall mortality, with metabolic syndrome components key risk factors increasing risk of death.
Collapse
Affiliation(s)
- Karl Vaz
- Department of Gastroenterology and Hepatology, Alfred Health, Ground Floor Alfred Centre,55 Commercial Road, Melbourne, VIC, 3004, Australia.
- Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - William Kemp
- Department of Gastroenterology and Hepatology, Alfred Health, Ground Floor Alfred Centre,55 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ammar Majeed
- Department of Gastroenterology and Hepatology, Alfred Health, Ground Floor Alfred Centre,55 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - John Lubel
- Department of Gastroenterology and Hepatology, Alfred Health, Ground Floor Alfred Centre,55 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Dianna J Magliano
- Diabetes and Population Health, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kristen M Glenister
- Department of Rural Health, University of Melbourne, Parkville, VIC, Australia
| | - Lisa Bourke
- Department of Rural Health, University of Melbourne, Parkville, VIC, Australia
| | - David Simmons
- Department of Rural Health, University of Melbourne, Parkville, VIC, Australia
- Macarthur Clinical School, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Alfred Health, Ground Floor Alfred Centre,55 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
367
|
Dong W, Gao X, Guan F, Pan J, Chen W, Zhang L, Zhang L. Establishment and characterization of liver-specific Apoa4-Cre and Cyp2c11-Cre rat models in juvenile and adult stages. Animal Model Exp Med 2025; 8:718-727. [PMID: 39916324 PMCID: PMC12008442 DOI: 10.1002/ame2.12504] [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: 04/08/2024] [Accepted: 10/04/2024] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Liver diseases are a major contributor to both morbidity and mortality. Conditional knockout animals are always produced through crossing floxed animals with a tissue-specific Cre animal. The use of floxed rat resource has rapidly increased, but the liver-specific Cre rat lines for studying liver diseases and interested genes are limited, especially in a spatially and temporally restricted manner. METHODS RNA sequencing and real-time polymerase chain reaction (PCR) were used to screen and confirm the presence of liver-specific genes. Apoa4-Cre rats and Cyp2c11-Cre rats were produced by CRISPR/Cas9 knockin. Rosa26-imCherry rats were employed to hybridize with the Cre rats to obtain the Apoa4-Cre/Rosa26-imCherry and Cyp2c11-Cre/Rosa26-imCherry rats. The temporal and spatial patterns of Cre expression were determined by the observation of red fluorescence on tissue sections. Hematoxylin-eosin stain was used to evaluate the liver histopathologic changes. The blood biochemical analysis of several liver enzymes and liver lipid profile was performed to evaluate the liver function of Cre rats. RESULTS Apoa4 and Cyp2c11 were identified as two liver-specific genes. Apoa4-Cre and Cyp2c11-Cre rats were produced and hybridized with Rosa26-imCherry rats. The red fluorescence indicated that the Cre recombinases were specially expressed in the juvenile and adult liver and not in other organs of two hybridized rats. All the blood biochemical parameters except low-density lipoprotein (LDL) did not change significantly in the Cre rats. No histological alterations were detected in the livers of the Cre rats. CONCLUSIONS Liver-specific Apoa4-Cre and Cyp2c11-Cre rats have been established successfully and could be used to study gene knockout, specifically in juvenile and adult liver.
Collapse
Affiliation(s)
- Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Xiang Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Feifei Guan
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Jirong Pan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Wei Chen
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Li Zhang
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine CollegeChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
368
|
Li Y, Pan T, Wang Y, Wang G, Wang F. The predictive value of triglyceride-glucose-high density lipoprotein-body mass index (TGH-BMI) for different degrees of hepatic steatosis and liver fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD). Clin Nutr ESPEN 2025; 66:290-301. [PMID: 39863255 DOI: 10.1016/j.clnesp.2025.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 12/22/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND & AIMS The triglyceride-glucose index (TyG) and triglyceride-glucose body mass index (TyG-BMI) have been identified as potential predictive factors for metabolic dysfunction-associated steatotic liver disease (MASLD). However, they do not include high density lipoprotein (HDL-C), which is closely related to lipid metabolism. Furthermore, there is a lack of comprehensive and longitudinal data to determine the cut-off points for different degrees of hepatic steatosis and liver fibrosis in MASLD. This study aimed to investigate the predictive capability of triglyceride-glucose-high density lipoprotein-body mass index (TGH-BMI) in determining hepatic steatosis and liver fibrosis in MASLD, as well as to establish the predictive cut-off points. METHODS We analyzed the relationships of TGH-BMI (TGH-BMI = ln [TG (mg/dL) ∗FBG (mg/dL)/HDL-C (mg/dL)] ∗ BMI (kg/m2)) with different degrees of hepatic steatosis and fibrosis in 35,114 participants who underwent health check-ups. A total of 2262 subjects without MASLD were selected for the analysis of cumulative hazard of hepatic steatosis and liver fibrosis in TGH-BMI dichotomous groups over a follow-up period of 1001 days. RESULTS Controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) demonstrated a consistent upward trend as TGH-BMI increased across quartile groups, as determined by One-way analysis of variance (P < 0.001). TGH-BMI and CAP, LSM exhibit distinct curve-like relationships between males and females when utilizing smoothing functions and conducting threshold effect analysis (P < 0.05). In males, prior to the inflection point at TGH-BMI = 177.733, there was a significant increase of 0.807 in CAP for every 1 unit increase in TGH-BMI (P < 0.05), after the inflection point, there was still an increase of 0.417 in CAP for every 1 unit increase in TGH-BMI (P < 0.05); There was no significant correlation between LSM and TGH-BMI before the first inflection point at TGH-BMI = 131.689 (P > 0.05) and after the second inflection point at TGH-BMI = 253.268 (P > 0.05). Between the first and the second inflection, LSM showed an increase of 0.015 for every 1 unit increase in TGH-BMI (P < 0.05). In females, before the inflection point at TGH-BMI = 94.686, there was a significant increase of 0.272 in CAP for every 1 unit increase in TGH-BMI (P < 0.05), after the inflection point, there was a notable change as CAP increased by 0.806 for every 1 unit increase in TGH-BMI (P < 0.05). There was no significant correlation between LSM and TGH-BMI before the inflection point at TGH-BMI = 118.098 (P > 0.05), after the inflection point, LSM showed an increase of 0.017 for every 1 unit increase in TGH-BMI (P < 0.05). Notably, TGH-BMI has been shown to be a strong predictor for the severity of hepatic steatosis and liver fibrosis in MASLD. The Area Under Curves (AUCs) for hepatic steatosis, moderate or above hepatic steatosis, severe hepatic steatosis and liver fibrosis in males were 0.845, 0.846, 0.882 and 0.668 respectively, the AUCs for hepatic steatosis, moderate or above hepatic steatosis, severe hepatic steatosis and liver fibrosis in females were 0.855, 0.895, 0.939 and 0.705 respectively (P < 0.05). In individuals without MASLD, the cumulative hazard of hepatic steatosis was found to be strongly associated with the dichotomy of increased TGH-BMI (TGH-BMID2: Hazard Ratio (HR) = 2.412, 95 % Confidence interval (CI): 2.0164-2.9071, P < 0.0001), while the same is true in liver fibrosis (TGH-BMID2: HR = 1.454, 95 % CI: 1.0633-1.9883, P = 0.0191). CONCLUSIONS The TGH-BMI demonstrates a strong predictive value for hepatic steatosis and liver fibrosis, with significantly different cut-off points for men and women. Therefore, it is important to consider the potential need for gender-specific cut-off points for triglyceride, glucose, high density lipoprotein and body mass index in clinical practice. In individuals without MASLD, a higher TGH-BMI is associated with an increased risk of developing MASLD in the future.
Collapse
Affiliation(s)
- Ying Li
- Department of Endocrinology, Hefei City First People's Hospital, Hefei 230001, Anhui, China
| | - Tianrong Pan
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Yue Wang
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Guojuan Wang
- Department of Endocrinology, Hefei City First People's Hospital, Hefei 230001, Anhui, China
| | - Fang Wang
- Department of the Health Management Center, The First Affiliated Hospital of USTC: Anhui Provincial Hospital, Hefei 230001, Anhui, China.
| |
Collapse
|
369
|
Cannella R, Agnello F, Porrello G, Spinello AU, Infantino G, Pennisi G, Cabibi D, Petta S, Bartolotta TV. Performance of ultrasound-guided attenuation parameter and 2D shear wave elastography in patients with metabolic dysfunction-associated steatotic liver disease. Eur Radiol 2025; 35:2339-2350. [PMID: 39373742 PMCID: PMC11914239 DOI: 10.1007/s00330-024-11076-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: 12/29/2023] [Revised: 07/06/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE To assess the performance and the reproducibility of ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) in patients with biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS This study included consecutive adult patients with MASLD who underwent ultrasound with UGAP, 2D-SWE and percutaneous liver biopsy. The median values of 12 consecutive UGAP measurements were acquired by two independent radiologists (R1 and R2). Hepatic steatosis was graded by liver biopsy as: (0) < 5%; (1) 5-33%; (2) > 33-66%; (3) > 66%. Areas under the curve (AUCs) were calculated to determine the diagnostic performance. Inter- and intra-observer reliability was assessed with intraclass correlation coefficient (ICC). RESULTS A hundred patients (median age 55.0 years old) with MASLD were prospectively enrolled. At histopathology, 70 and 42 patients had grade ≥ 2 and 3 steatosis, respectively. Median UGAP was 0.78 dB/cm/MHz (IQR/Med: 5.55%). For the diagnosis of grade ≥ 2 steatosis, the AUCs of UGAP were 0.828 (95% CI: 0.739, 0.896) for R1 and 0.779 (95% CI: 0.685, 0.856) for R2. The inter- and intra-operator reliability of UGAP were excellent, with an ICC of 0.92 (95% CI: 0.87-0.95) and 0.95 (95% CI: 0.92-0.96), respectively. The median liver stiffness was 6.76 kPa (IQR/Med: 16.30%). For the diagnosis of advanced fibrosis, 2D-SWE had an AUC of 0.862 (95% CI: 0.757, 0.934), and the optimal cutoff value was > 6.75 kPa with a sensitivity of 80.6% and a specificity of 75.7%. CONCLUSION UGAP and 2D-SWE provide a good performance for the staging of steatosis and fibrosis in patients with MASLD with an excellent intra-operator reliability of UGAP. KEY POINTS Question How well do ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) perform for quantifying hepatic steatosis and fibrosis? Findings UGAP had a maximum AUC of 0.828 for the diagnosis of grade ≥ 2 steatosis, and 2D-SWE had an AUC of 0.862 for diagnosing advanced fibrosis. Clinical relevance UGAP and 2D-SWE allow rapid, reproducible, and accurate quantification of hepatic steatosis and fibrosis that can be used for the noninvasive assessment of patients with metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy.
| | - Francesco Agnello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giorgia Porrello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Alessandro Umberto Spinello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giuseppe Infantino
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Grazia Pennisi
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Daniela Cabibi
- Unit of Anatomic Pathology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| |
Collapse
|
370
|
Zhou XD, Yip TCF, Huang DQ, Muthiah MD, Noureddin M, Zheng MH. Vibration-controlled transient elastography in shaping the epidemiology and management of steatotic liver disease: Editorial on "Current burden of steatotic liver disease and fibrosis among adults in the United States, 2017-2023". Clin Mol Hepatol 2025; 31:620-624. [PMID: 39722613 PMCID: PMC12016648 DOI: 10.3350/cmh.2024.1131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Xiao-Dong Zhou
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Terry Cheuk-Fung Yip
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Daniel Q Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
| | - Mark Dhinesh Muthiah
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
| | - Mazen Noureddin
- Division of Digestive and Liver Diseases, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
371
|
Feng G, Mózes FE, Ji D, Treeprasertsuk S, Okanoue T, Shima T, Liang H, Tsochatzis E, Chen J, Schattenberg JM, Labenz C, Mahadeva S, Chan WK, Chi X, Delamarre A, de Lédinghen V, Petta S, Bugianesi E, Hagström H, Boursier J, Calleja JL, Goh GBB, Gallego-Durán R, Sanyal AJ, Fan JG, Castéra L, Lai M, Harrison SA, Romero-Gomez M, Kim SU, Zhu Y, Ooi G, Shi J, Yoneda M, Nakajima A, Zhang J, Lupsor-Platon M, Zhong B, Cobbold JFL, Ye CY, Eddowes PJ, Newsome P, Li J, George J, He F, Song MJ, Tang H, Fan Y, Jia J, Xu L, Lin S, Li Y, Lu Z, Nan Y, Niu J, Yan X, Zhou Y, Liu C, Deng H, Ye Q, Zeng QL, Li L, Wang J, Yang S, Lin H, Lee HW, Yip TCF, Fournier-Poizat C, Wong GLH, Pennisi G, Armandi A, Liu WY, Shang Y, de Saint-Loup M, Llop E, Teh KKJ, Lara-Romero C, Asgharpour A, Mahgoub S, Chan MSW, Canivet CM, Ji F, Xin Y, Chai J, Dong Z, Targher G, Byrne CD, He N, Mi M, Ye F, Wong VWS, Pavlides M, Zheng MH. acFibroMASH Index for the Diagnosis of Fibrotic MASH and Prediction of Liver-related Events: An International Multicenter Study. Clin Gastroenterol Hepatol 2025; 23:785-796. [PMID: 39362618 DOI: 10.1016/j.cgh.2024.07.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) and fibrotic MASH are significant health challenges. This multi-national study aimed to validate the acMASH index (including serum creatinine and aspartate aminotransferase concentrations) for MASH diagnosis and develop a new index (acFibroMASH) for non-invasively identifying fibrotic MASH and exploring its predictive value for liver-related events (LREs). METHODS We analyzed data from 3004 individuals with biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD) across 29 Chinese and 9 international cohorts to validate the acMASH index and develop the acFibroMASH index. Additionally, we utilized the independent external data from a multi-national cohort of 9034 patients with MASLD to examine associations between the acFibroMASH index and the risk of LREs. RESULTS In the pooled global cohort, the acMASH index identified MASH with an area under the receiver operating characteristic curve (AUROC) of 0.802 (95% confidence interval [CI], 0.786-0.818). The acFibroMASH index (including the acMASH index plus liver stiffness measurement) accurately identified fibrotic MASH with an AUROC of 0.808 in the derivation cohort and 0.800 in the validation cohort. Notably, the AUROC for the acFibroMASH index was 0.835 (95% CI, 0.786-0.882), superior to that of the FAST score at 0.750 (95% CI, 0.693-0.800; P < .01) in predicting the 5-year risk of LREs. Patients with acFibroMASH >0.39 had a higher risk of LREs than those with acFibroMASH <0.15 (adjusted hazard ratio, 11.23; 95% CI, 3.98-31.66). CONCLUSIONS This multi-ethnic study validates the acMASH index as a reliable, noninvasive test for identifying MASH. The newly proposed acFibroMASH index is a reliable test for identifying fibrotic MASH and predicting the risk of LREs.
Collapse
Affiliation(s)
- Gong Feng
- Xi'an Medical University, Xi'an, China; Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ferenc E Mózes
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dong Ji
- Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Sombat Treeprasertsuk
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and Thai Red Cross, Bangkok, Thailand
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Huiqing Liang
- Hepatology Unit, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian, China
| | - Emmanuel Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, United Kingdom
| | - Jinjun Chen
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jörn M Schattenberg
- Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Rhineland-Palatinate, Germany; Department of Medicine II, University Medical Center Homburg, Homburg and University of the Saarland, Saarbrücken, Germany
| | - Christian Labenz
- Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Rhineland-Palatinate, Germany; Department of Medicine II, University Medical Center Homburg, Homburg and University of the Saarland, Saarbrücken, Germany
| | - Sanjiv Mahadeva
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Malaysia
| | - Wah Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Malaysia
| | - Xiaoling Chi
- Department of Hepatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Adèle Delamarre
- Centre d'Investigation de la Fibrose Hépatique, Hôpital Haut-Lévêque, Bordeaux University Hospital, Pessac, and INSERM U1312, Bordeaux University, Bordeaux, France
| | - Victor de Lédinghen
- Centre d'Investigation de la Fibrose Hépatique, Hôpital Haut-Lévêque, Bordeaux University Hospital, Pessac, and INSERM U1312, Bordeaux University, Bordeaux, France
| | - Salvatore Petta
- Sezione di Gastroenterologia, Di.Bi.M.I.S., University of Palermo, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Jérôme Boursier
- Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France; HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - José Luis Calleja
- Department of Gastroenterology and Hepatology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Rocio Gallego-Durán
- Digestive Diseases Unit and CIBERehd, Virgen Del Rocío University Hospital, Seville, Spain
| | - Arun J Sanyal
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Department of Internal Medicine, VCU School of Medicine, Richmond, Virginia
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Laurent Castéra
- Université Paris Cité, UMR1149 (CRI), INSERM, Paris, France; Service d'Hépatologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), Clichy, France
| | - Michelle Lai
- Division of Gastroenterology & Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Stephen A Harrison
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Pinnacle Clinical Research, San Antonio, Texas
| | - Manuel Romero-Gomez
- Digestive Diseases Unit and CIBERehd, Virgen Del Rocío University Hospital, Seville, Spain
| | - Seung Up Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yongfen Zhu
- Department of Hepatology and Infection, Sir Run Run Shaw Hospital, Affiliated with School of Medicine, Zhejiang University, Hangzhou, China
| | - Geraldine Ooi
- Centre for Obesity Research and Education, Department of Surgery, Monash University, Melbourne, Australia
| | - Junping Shi
- Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Jing Zhang
- The Third Unit, Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Monica Lupsor-Platon
- Department of Medical Imaging, Iuliu Hatieganu, University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology "Prof. Dr. Octavian Fodor," Cluj-Napoca, Romania
| | - Bihui Zhong
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jeremy F L Cobbold
- Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - Chun-Yan Ye
- Institute for the Study of Liver Diseases, The Third People's Hospital of Changzhou, Changzhou, Jiangsu Province, China
| | - Peter J Eddowes
- National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
| | - Philip Newsome
- National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, United Kingdom; Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom; Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, New South Wales, Australia
| | - Fangping He
- Department of Hepatobiliary Pancreatic Surgery, Eighth Hospital Affiliated to SunYat-sen University, Futian, Guangdong Province, China
| | - Myeong Jun Song
- Division of Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liang Xu
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Su Lin
- Department of Hepatology, Hepatology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yiling Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhonghua Lu
- Clinical Laboratory Center, The Fifth People's Hospital of Wuxi, Wuxi, Jiangsu, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Hebei Medical University Third Hospital, Shijiazhuang, China
| | - Junqi Niu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| | - Xuebing Yan
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Deng
- Department of lnfectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Ye
- Department of Hepatology of The Third Central Hospital of Tianjin, Tianjin, China
| | - Qing-Lei Zeng
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Li
- Department of lnfectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Wang
- Department of Hepatobiliary Diseases, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Song Yang
- Center of Hepatology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Huapeng Lin
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Hye Won Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Terry Cheuk-Fung Yip
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Grace Lai-Hung Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Grazia Pennisi
- Sezione di Gastroenterologia, Di.Bi.M.I.S., University of Palermo, Italy
| | - Angelo Armandi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Wen-Yue Liu
- Department of Endocrinology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Shang
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marc de Saint-Loup
- Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France
| | - Elba Llop
- Department of Gastroenterology and Hepatology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Kevin Kim Jun Teh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Carmen Lara-Romero
- Digestive Diseases Unit and CIBERehd, Virgen Del Rocío University Hospital, Seville, Spain
| | - Amon Asgharpour
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Department of Internal Medicine, VCU School of Medicine, Richmond, Virginia
| | - Sara Mahgoub
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, United Kingdom
| | | | - Clemence M Canivet
- Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France; HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - Fanpu Ji
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongning Xin
- Department of Infectious Diseases, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jin Chai
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zhiyong Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy; Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Na He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Man Mi
- Xi'an Medical University, Xi'an, China
| | - Feng Ye
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| | - Michael Pavlides
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom.
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
372
|
Chen J, Hu J, Zhuang J, Li Z, Peng S, Huang X, Zhuang J. Diagnostic Value of Serum Cytokeratin 18 for the Staging of Liver Inflammation and Fibrosis: A Meta-Analysis. J Clin Lab Anal 2025; 39:e70034. [PMID: 40202219 PMCID: PMC12019703 DOI: 10.1002/jcla.70034] [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: 12/20/2024] [Revised: 03/15/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND AND AIMS Accurate assessment of liver inflammation and fibrosis is of vital importance in the clinical management of patients with liver diseases. Our aim is to conduct a meta-analysis to evaluate the diagnostic accuracy of serum cytokeratin 18 (CK18) for staging of liver inflammation and fibrosis against a liver biopsy in adults. METHODS We systematically searched articles from eight electronic databases. Two authors independently selected included studies, extracted data, and assessed quality. In our meta-analysis, we used the random-effects meta-analysis model. Publication bias, sensitivity analysis, heterogeneity analysis, and post-test probability were used in this meta study. RESULTS A total of 20 studies with 2235 patients were initially found by the search strategies. The pooled sensitivity, specificity, and area under the curve (AUC) of the summary receiver operating characteristic curve were 0.56, 0.81, and 0.810 for significant fibrosis; 0.64, 0.76, and 0.785 for advanced fibrosis; 0.53, 0.76, and 0.830 for cirrhosis; and 0.68, 0.73, and 0.786 for significant inflammation, respectively. High heterogeneity was observed in our meta-analysis because of factors such as the proportion of males, total number, and antigens of CK-18. CONCLUSION Serum CK18 had moderate diagnostic value (AUC > 0.7) in different stages of liver fibrosis and significant inflammation, offering a complementary approach to other non-invasive indicators such as serological biomarkers and imaging techniques. Future research should focus on elucidating the role of CK18 in the occurrence and progression of hepatitis and liver fibrosis, particularly in liver diseases with diverse etiologies.
Collapse
Affiliation(s)
- Jinwen Chen
- Department of Laboratory MedicineGuangdong Provincial Hospital of Chinese Medicine, Zhuhai HospitalZhuhaiChina
| | - Jian Hu
- Department of Laboratory MedicineGuangdong Provincial Hospital of Chinese Medicine, Zhuhai HospitalZhuhaiChina
| | - Jialin Zhuang
- School of AutomationGuangdong University of TechnologyGuangzhouChina
| | - Zhong Li
- Zhuhai Seesheen Medical Technology Company LimitedZhuhaiChina
| | - Se Peng
- Department of Laboratory MedicineGuangdong Provincial Hospital of Chinese Medicine, Zhuhai HospitalZhuhaiChina
| | - Xiaoting Huang
- Medical Research CenterHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenChina
| | - Jialing Zhuang
- Department of Laboratory MedicineGuangdong Provincial Hospital of Chinese Medicine, Zhuhai HospitalZhuhaiChina
| |
Collapse
|
373
|
Hanamatsu H, Suda G, Ohara M, Ogawa K, Tamaki N, Hikita H, Haga H, Maekawa S, Sugiyama M, Kakisaka T, Nakai M, Sho T, Miura N, Kurosaki M, Asahina Y, Taketomi A, Ueno Y, Takehara T, Nishikaze T, Furukawa JI, Sakamoto N. Elevated A2F bisect N-glycans of serum IgA reflect progression of liver fibrosis in patients with MASLD. J Gastroenterol 2025; 60:456-468. [PMID: 39849179 PMCID: PMC11922979 DOI: 10.1007/s00535-024-02206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/25/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Advanced liver fibrosis in cases of metabolic dysfunction-associated steatotic liver disease (MASLD) leads to cirrhosis and hepatocellular carcinoma. The current gold standard for liver fibrosis is invasive liver biopsy. Therefore, a less invasive biomarker that accurately reflects the stage of liver fibrosis is highly desirable. METHODS This study enrolled 269 patients with liver biopsy-proven MASLD. Patients were divided into three groups (F0/1 (n = 41/85), F2 (n = 47), and F3/4 (n = 72/24)) according to fibrosis stage. We performed serum N-glycomics and identified glycan biomarker for fibrosis stage. Moreover, we explored the carrier proteins and developed a sandwich ELISA to measure N-glycosylation changes of carrier protein. RESULTS Comprehensive N-glycomic analysis revealed significant changes in the expression of A2F bisect and its precursors as fibrosis progressed. The sum of neutral N-glycans carrying bisecting GlcNAc and core Fuc (neutral sum) had a better diagnostic performance to evaluate advanced liver fibrosis (AUC = 0.804) than conventional parameters (FIB4 index, aspartate aminotransferase-to-alanine aminotransferase ratio (AAR), and serum level of Mac-2-binding protein glycol isomer (M2BPGi). The combination of the neutral sum and FIB4 index enhanced diagnostic performance (AUC = 0.840). IgM, IgA, and complement C3 were identified as carrier proteins with A2F bisect N-glycan. A sandwich ELISA based on N-glycans carrying bisecting GlcNAc and IgA showed similar diagnostic performance than the neutral sum. CONCLUSIONS A2F bisect N-glycan and its precursors are promising candidate biomarkers for advanced fibrosis in MASLD patients. Analysis of these glycan alterations on IgA may have the potential to serve as a novel ELISA diagnostic tool for MASLD in routine clinical practice. CLINICAL TRIAL NUMBER UMIN000030720.
Collapse
Affiliation(s)
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Masatsugu Ohara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Koji Ogawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Nobuharu Tamaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Haga
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Shinya Maekawa
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Masaya Sugiyama
- Department of Viral Pathogenesis and Controls, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tatsuhiko Kakisaka
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Masato Nakai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Takuya Sho
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Nobuaki Miura
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Aichi, Japan
| | - Masayuki Kurosaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Nishikaze
- Solutions COE, Analytical and Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Jun-Ichi Furukawa
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Aichi, Japan.
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
374
|
Liukkonen V, Semenova M, Hyvärinen K, Lauronen J, Partanen J, Arola J, Nordin A, Färkkilä M, Åberg F. Genetic Risk Factors for Steatotic Liver Disease After Liver Transplantation. Liver Int 2025; 45:e70067. [PMID: 40087975 DOI: 10.1111/liv.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND The role of genetic risk factors for steatotic liver disease (SLD) is intriguing in liver transplantation (LT), as both donor and recipient genetic factors may play a role. There are only a few small-scale studies published so far. METHODS We analysed the incidence and risk factors for post-LT SLD and the impact of 56 SLD-associated genetic variants in 595 donor-recipient pairs with liver biopsy available ≥ 6 months after LT. We evaluated whether the polygenic risk score (PRS-5) improves the ability to predict post-LT SLD in addition to non-genetic risk factors. RESULTS SLD after LT was diagnosed in 34.5% of patients during a median 7.6-year follow-up. In multivariate analysis including non-genetic risk factors, donor PNPLA3 rs738409-G (HR for SLD: C/G 1.34, p = 0.051, G/G 2.25, p = 0.004), donor HSD17B13 rs72613567-TA (HR for SLD: TA/T 0.68, p = 0.02 TA/TA HR 0.50, p = 0.10) and recipient UCP2 rs695366-G (HR for SLD: A/G 0.63, p = 0.002, G/G HR 0.50, p = 0.04) appeared as the most important genetic risk factors for post-LT SLD. The addition of PRS-5 to a multivariate regression model (including non-genetic risk factors) improved the predictive ability for SLD only modestly (AUC 0.78 to 0.80). CONCLUSIONS Various genetic variants contribute to post-LT SLD with separate variants among recipients and donors, with donor PNPLA3 rs738409-G as the most significant risk allele. Still, donor and recipient genotyping provide only modest additional value for individual risk stratification over phenotype data, highlighting the role of modifiable risk factors.
Collapse
Affiliation(s)
- Ville Liukkonen
- North Karelia Central Hospital, Gastroenterology, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Maria Semenova
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Kati Hyvärinen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Jouni Lauronen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Johanna Arola
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, Helsinki University Hospital, Diagnostic Centre, Helsinki, Finland
| | - Arno Nordin
- Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Martti Färkkilä
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fredrik Åberg
- Transplantation and Liver Surgery, Helsinki University Hospital, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| |
Collapse
|
375
|
Han Y, Yao M, Zhao H, Han X, Di H, Xie T, Wu J, Wang Y, Zhang Y, Zeng X. Exploration of the Interrelationship Between Serum Uric Acid, Gout, and Cardiac, Renal, and Metabolic Conditions in Middle Aged and Older People. J Am Heart Assoc 2025; 14:e038723. [PMID: 40145269 DOI: 10.1161/jaha.124.038723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Cardiac, renal, and metabolic (CRM) conditions are major causes of morbidity and mortality globally. This study aims to explore the relationship between serum uric acid (SUA), hyperuricemia, gout, and CRM conditions in middle-aged and elderly populations. METHODS Sample 1 included participants from CHARLS (China Health and Retirement Longitudinal Study, n=9341), and Sample 2 from NHANES (National Health and Nutrition Examination Survey, unweighted n=17 913; weighted n=115 646 390). Ordinal logistic regression, Cox regression, and restricted cubic spline analyses were used to assess the relationship between SUA, hyperuricemia, gout, and CRM conditions. A 2-sample Mendelian randomization analysis was conducted to explore causal associations between SUA and CRM conditions. RESULTS In both samples, SUA, hyperuricemia, and gout were positively correlated with the risk of CRM conditions. Among participants with 3 or ≥1 CRM condition(s), SUA, asymptomatic hyperuricemia, and gout with poorly controlled hyperuricemia showed significant positive associations with all-cause mortality, whereas these associations were not observed in patients with gout with normal SUA levels. The restricted cubic spline analysis revealed a positive relationship between SUA levels and the risk of all-cause mortality in participants with ≥1 CRM condition(s), demonstrating a nonlinear dose-response relationship across both samples (P for nonlinearity <0.05). Mendelian randomization analysis indicated that SUA was causally associated with cardiovascular disease, chronic kidney disease, and diabetes. CONCLUSIONS Hyperuricemia and gout are strong predictors of increased prevalence and mortality of CRM conditions, emphasizing the importance of managing hyperuricemia and gout in these patients.
Collapse
Affiliation(s)
- Yingdong Han
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Menghui Yao
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - He Zhao
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Xinxin Han
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Hong Di
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Tiange Xie
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Juan Wu
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Yibo Wang
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Yun Zhang
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| | - Xuejun Zeng
- Department of family medicine & Division of General Internal Medicine, Department of internal medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital) Beijing China
| |
Collapse
|
376
|
Zhu B, Wei R, Li X, Bi Q. Targeting CCL5 Attenuates Fibrosis via Activation of PI3k/Akt Signaling Axis After Glaucoma Filtration Surgery. Curr Eye Res 2025; 50:394-404. [PMID: 39618346 DOI: 10.1080/02713683.2024.2432399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 01/02/2025]
Abstract
PURPOSE Glaucoma filtration surgery (GFS) stands as a paramount clinical intervention for glaucoma. Nonetheless, the prevalent cause of GFS failure is filtration bleb scarring, and the role of inflammation and immune response in contributing to fibrosis remains elusive. METHODS The study employed 30 female Sprague-Dawley rats (8 weeks old, 200-250 g) to assess the anti-scarring impact of the Chemokine (C-C motif) receptor 5 (CCR5)-Chemokine (C-C motif) ligand 5 (CCL5) antibody after GFS. Additionally, anti-fibrotic effects on HConFs were examined, creating an intra-operative inflammatory response using damaged-HConFs supernatant medium (DHSM). In vitro and in vivo validation aimed to elucidate the potential anti-fibrotic molecular mechanism of the CCR5-CCL5 antibody. RESULTS The CCR5-CCL5 antibody effectively prolonged filtration bleb duration and enhanced the functionality of the filtered bleb. Improved postoperative intraocular pressure values (IOP) and morphological images were observed in the CCR5-CCL5 antibody-treated group. Histochemical staining and cellular experiments confirmed the antifibrotic function of the CCR5-CCL5 antibody. Notably, M2-type macrophage polarization was reduced in the CCR5-CCL5 antibody-treated model. CCL5-induced fibrosis in HConFs was mediated through the PI3K/Akt signaling pathway. Consistently, inhibition of PI3K/Akt significantly attenuated the profibrotic effects of CCR5-CCL5. Mechanistically, the CCL5 antibody exerts its antifibrotic effect by targeting CCR5 on HConFs, leading to the inhibition of the PI3K/Akt mechanism. CONCLUSIONS This study unveils that CCR5-CCL5 promotes fibrosis in GFS through inflammatory stimulation of HConFs and enhanced activation of the PI3K/Akt signaling pathway. The findings suggest that intraoperative CCR5-CCL5 antibody treatment could serve as a cost-effective therapeutic agent or a useful adjuvant in preventing ocular bleb scar formation.
Collapse
Affiliation(s)
- Baixue Zhu
- Yulin Hospital of Traditional Chinese Medicine, Yulin, Shaanxi, China
| | - Ran Wei
- Zhuhai People's Hospital, Zhuhai, Guangdong, China
| | - Xinying Li
- The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingyun Bi
- The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
377
|
Migni A, Bartolini D, Marcantonini G, Sardella R, Rende M, Tognoloni A, Ceccarini MR, Galli F. Melatonin Repairs the Lipidome of Human Hepatocytes Exposed to Cd and Free Fatty Acid-Induced Lipotoxicity. J Pineal Res 2025; 77:e70047. [PMID: 40193217 PMCID: PMC11975211 DOI: 10.1111/jpi.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Hepatocyte lipotoxicity is central to the aetiology of nonalcoholic fatty liver disease (NAFLD), a leading cause of liver failure and transplantation worldwide. Long-lasting toxic pollutants have increasingly been considered as environmental risk factors of NAFLD. These include cadmium (Cd), a metal that synergizes with other cellular toxicants and metabolic stimuli to induce fat build-up and lipotoxicity. Recent studies demonstrated that melatonin (MLT) holds great potential as repairing agent in this form of hepatocyte lipotoxicity. In this study, the molecular hints of this MLT effect were investigated by lipidomics analysis in undifferentiated HepaRG cells, a human pre-hepatocyte cell line, exposed to Cd toxicity either alone or combined with prototypical free fatty acids (FFA), namely the saturated species palmitic acid and the monounsaturated oleic acid (OA and PA, respectively), to simulate the cellular lipotoxicity conditions of fatty liver disease. Cd exposure synergized with FFAs to induce cellular steatosis, and PA produced higher levels of lipotoxicity compared to OA by leading to increased levels of H2O2 production and apoptotic death. These effects were associated with changes of the cellular lipidome, which approximate those of NAFLD liver, with differentially expressed lipids in different classes that included triacylglycerols (TG), di- and mono-acylglycerols, phospholipids (PL), sphingolipids, acylcarnitines and FA; characteristic differences were observed in all these classes comparing the combinations of Cd exposure with PA or OA treatments. MLT significantly reduced the effects of either individual or combinatorial treatments of Cd and FFAs on lipotoxicity hallmarks, also repairing most of the alterations of the cellular lipidome, including those of the chain length and number of double bonds of acyl residues esterified to TG and PL classes. These findings and their bioinformatics interpretation suggest a role for the earliest acyl elongase and desaturase steps of FA metabolism in this repairing effect of MLT; biochemistry studies validated such interpretation identifying a specific role for SCD1 activity. This lipidomics study shed light on the cytoprotective mechanism of MLT in Cd and FFA-induced hepatocyte lipotoxicity, highlighting a repairing effect of this molecule on the cellular lipidome, which may hold therapeutic potential in fatty liver diseases.
Collapse
Affiliation(s)
- Anna Migni
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | - Desirée Bartolini
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | | | - Roccaldo Sardella
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | - Mario Rende
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Alessia Tognoloni
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| | | | - Francesco Galli
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| |
Collapse
|
378
|
Parekh Z, Xiao J, Mani A, Evans Q, Phung C, Barba HA, Xie B, Sidebottom AM, Sundararajan A, Lin H, Ramaswamy R, Dao D, Gonnah R, Yehia M, Hariprasad SM, D'Souza M, Sulakhe D, Chang EB, Skondra D. Fecal Microbial Profiles and Short-Chain Fatty Acid/Bile Acid Metabolomics in Patients With Age-Related Macular Degeneration: A Pilot Study. Invest Ophthalmol Vis Sci 2025; 66:21. [PMID: 40202735 PMCID: PMC11993127 DOI: 10.1167/iovs.66.4.21] [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: 04/11/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Age-related macular degeneration (AMD) is a multifactorial disease, and studies have implicated the role of gut microbiota in its pathogenesis. However, characterization of microbiome dysbiosis and associated microbial-derived metabolomic profiles across AMD stages remains unknown. In this pilot study, we explored how gut microbiome composition and gut-derived metabolites differ in AMD. Methods Our pilot study analyzed fasted stool samples that were collected from 22 patients at a tertiary academic center. Subjects were classified as control, intermediate AMD, or advanced AMD based on clinical presentation. 16S rRNA amplicon sequencing and standard chromatography-mass spectrometry methods were used to identify bacterial taxonomy composition and abundance of short-chain fatty acids (SCFAs) and bile acids (BAs), respectively. Genetic testing was used to investigate the frequency of 14 high-risk single nucleotide polymorphisms (SNPs) associated with AMD in the AMD cohort. Results Forty-three differentially abundant genera were present among the control, intermediate, and advanced groups. Taxa with known roles in immunologic pathways, such as Desulfovibrionales (q = 0.10) and Terrisporobacter (q = 1.16e-03), were in greater abundance in advanced AMD patients compared to intermediate. Advanced AMD patients had decreased abundance of 12 SCFAs, including acetate (P = 0.002), butyrate (P = 0.04), and propionate (P = 0.01), along with 12 BAs, including taurocholic acid (P = 0.02) and tauroursodeoxycholic acid (P = 0.04). Frequencies of high-risk SNPs were not significantly different between the intermediate and advanced AMD groups. Conclusions This pilot study identifies distinct gut microbiome compositions and metabolomic profiles associated with AMD and its stages, providing preliminary evidence of a potential link between gut microbiota and AMD pathogenesis. To validate these findings and elucidate the underlying mechanisms, future research with larger cohorts and more comprehensive sampling is strongly recommended.
Collapse
Affiliation(s)
- Zaid Parekh
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Jason Xiao
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Amir Mani
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Quadis Evans
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Christopher Phung
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Hugo A. Barba
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Bingqing Xie
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Ashley M. Sidebottom
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Anitha Sundararajan
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Huaiying Lin
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Ramanujam Ramaswamy
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - David Dao
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Reem Gonnah
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Madeleine Yehia
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Seenu M. Hariprasad
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Mark D'Souza
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dinanath Sulakhe
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Eugene B. Chang
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
379
|
Liu Q, Liu Y, Feng H, Zhang N, Yang Z. High salt diet causally increases metabolic dysfunction-associated steatotic liver disease risk: A bidirectional mendelian randomization study. Nutr Res 2025; 136:94-104. [PMID: 40184888 DOI: 10.1016/j.nutres.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver disorder associated with metabolic and lifestyle factors, affecting approximately 25% of the global population. Although high salt intake has been implicated as a potential dietary risk factor, its causal relationship with MASLD remains uncertain. We hypothesized that genetic liability to higher salt intake causally increases the risk of MASLD. To address this, bidirectional Mendelian Randomization (MR) analysis was performed to evaluate the causal relationship between "salt added to food" and MASLD. Genetic variants were used as instrumental variables across large-scale genome-wide association study datasets from the UK Biobank and multiple MASLD cohorts. The inverse variance weighting method served as the primary analytical approach, with sensitivity analyses, including MR-Egger and MR-PRESSO, to evaluate pleiotropy and heterogeneity. Forward MR analysis demonstrated a significant association between "salt added to food" and increased MASLD risk across three MASLD datasets: odds ratio (OR) = 1.538, 95% confidence interval (CI): 1.145-2.067, P = .004; OR = 1.787, 95% CI: 1.247-2.561, P = .002; and OR = 2.094, 95% CI: 1.274-3.442, P = .004. Sensitivity analyses indicated low heterogeneity and no evidence of pleiotropy. Reverse MR analysis did not demonstrate a causal effect of MASLD on "salt added to food". These findings provide robust genetic evidence that "salt added to food" is a causal risk factor for MASLD, emphasizing the importance of dietary salt reduction in MASLD prevention strategies. This study supports public health recommendations advocating reduced salt intake to promote liver health and prevent MASLD.
Collapse
Affiliation(s)
- Qi Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China; Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery of Chinese People's Liberation Army, Beijing, China; Medical School of Chinese People's Liberation Army, Beijing, China.
| | - Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming, China.
| | - Hui Feng
- Department of Zhantansi Outpatient, Jingzhong Medical District of Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Ning Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China; Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery of Chinese People's Liberation Army, Beijing, China.
| | - Zhanyu Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China; Institute of Hepatobiliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery of Chinese People's Liberation Army, Beijing, China; Medical School of Chinese People's Liberation Army, Beijing, China.
| |
Collapse
|
380
|
Song K, Kwon YJ, Lee E, Lee HS, Youn YH, Baik SJ, Lee H, Kim JY, Choi Y, Chae HW. Bioelectrical impedance analysis parameters are superior to liver enzymes in predicting metabolic dysfunction-associated steatotic liver disease in young adults. Intern Emerg Med 2025; 20:785-795. [PMID: 40205114 DOI: 10.1007/s11739-025-03880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/22/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND/AIMS The predictive value of bioelectrical impedance analysis (BIA) parameters and liver enzymes for metabolic dysfunction-associated steatotic liver disease (MASLD) in young adults is unclear. We aimed to investigate the usefulness of BIA parameters and liver enzymes in MASLD screening in young adults. METHODS Overall, 2,647 participants aged 19-30 years were evaluated by sex. Logistic regression analyses were performed with MASLD as the dependent variable. Receiver operating characteristic (ROC) curves were used to compare the predictive ability for MASLD. RESULTS In males, percentage body fat (PBF), total body fat (TBF), visceral fat area (VFA), abdominal subcutaneous fat (ASF), alanine aminotransferase (ALT), and elevated ALT levels were positively associated with MASLD, whereas skeletal muscle index (SMI) and AST/ALT were negatively associated after adjusting for age, body mass index, and moderate physical activity. In females, PBF, TBF, ASF, AST, ALT, low SMI (LSMI), and elevated ALT levels were positively associated with MASLD, whereas SMI and AST/ALT were negatively associated. The areas under the ROC curves (AUCs) of PBF, TBF, VFA, ASF, SMI, AST, ALT, and AST/ALT were 0.818, 0.837, 0.834, 0.837, 0.818, 0.653, 0.759, and 0.790 respectively, in males. In females, the corresponding values were 0.915, 0.939, 0.934, 0.940, 0.915, 0.620, 0.799, and 0.849 respectively. The AUC of LSMI was significantly higher than that of increased ALT levels in females. CONCLUSIONS BIA parameters and ALT levels are useful for predicting MASLD in young adults. BIA parameters have superior predictive ability than liver enzymes.
Collapse
Affiliation(s)
- Kyungchul Song
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-Si, Republic of Korea
| | - Eunju Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Youn
- Department of Healthcare Research Team, Health Promotion Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su Jung Baik
- Department of Healthcare Research Team, Health Promotion Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hana Lee
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Young Kim
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngha Choi
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Wook Chae
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
381
|
Jaan A, Najim MS, Farooq U, Dhawan A, Nawaz H, Jahagirdar V, Ali H, Ahlawat S. Influence of Obesity Class on Clinical Outcomes in Alcoholic Hepatitis: A National Cohort Study of Mortality, Complications, and Resource Use. JGH Open 2025; 9:e70166. [PMID: 40236938 PMCID: PMC11998181 DOI: 10.1002/jgh3.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Background & Aims Alcoholic hepatitis (AH) is a severe manifestation of alcoholic liver disease with high morbidity and mortality. This study used the 2016-2020 National Readmission Database to investigate how obesity influences AH outcomes. Methods Adult hospitalizations were categorized as those without obesity, Class 1 obesity (BMI 30-34.9), Class 2 obesity (BMI 35-39.9), or Class 3 obesity (BMI ≥ 40). We compared mortality, complications, and resource utilization across these groups using regression models. Results Among 82 367 AH admissions, 4.09% had Class 1 obesity, 2.73% had Class 2 obesity, and 4.02% had Class 3 obesity. After adjusting for confounders, Class 3 obesity was associated with higher odds of mortality (Odds ratio OR = 1.74; 95% CI: 1.40-2.17; p < 0.01), septic shock (OR = 2.27; 95% CI: 1.60-3.22; p < 0.01), hepatic encephalopathy (OR = 2.53; 95% CI: 1.15-5.56; p = 0.02), and intensive care unit (ICU) admission (OR = 1.93; 95% CI: 1.57-2.36; p < 0.01). All obesity classes had increased associations with hepatorenal syndrome. No significant differences emerged for spontaneous bacterial peritonitis or variceal bleeding. Resource utilization rose with increasing obesity severity, with Class 3 obesity having a 1.84-day longer adjusted length of stay (p < 0.01) and an additional $20 174 in total hospitalization charges (p < 0.01) compared with hospitalizations without obesity. Conclusions Class 3 obesity conferred the greatest burden of mortality, complications, and healthcare costs among hospitalizations with AH. Further research is warranted to clarify the intricate interplay between obesity and AH.
Collapse
Affiliation(s)
- Ali Jaan
- Division of Internal MedicineRochester General HospitalRochesterUSA
| | | | - Umer Farooq
- Division of GastroenterologySaint Louis UniversityMissouriUSA
| | - Ashish Dhawan
- Division of Internal MedicineGian Sagar Medical College and HospitalPatiala districtIndia
| | - Hassan Nawaz
- Division of Internal MedicineNishtar Medical UniversityMultanPakistan
| | - Vinay Jahagirdar
- Division of GastroenterologyVirginia Commonwealth UniversityRichmondUSA
| | - Hassam Ali
- Division of GastroenterologyEast Carolina University/ECU Health Medical CenterGreenvilleUSA
| | - Sushil Ahlawat
- Division of GastroenterologySUNY Downstate Health Sciences UniversityBrooklynUSA
| |
Collapse
|
382
|
Lu F, Liu J, She B, Yang H, Ji F, Zhang L. Global Trends and Inequalities of Liver Complications Related to Metabolic Dysfunction-Associated Steatotic Liver Disease: An Analysis From 1990 to 2021. Liver Int 2025; 45:e16120. [PMID: 39387341 PMCID: PMC11891383 DOI: 10.1111/liv.16120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease is a significant driver of the increasing global burden of chronic liver disease. This study aimed to describe the temporal trends and inequalities of liver complications related to metabolic dysfunction-associated steatotic liver disease (LC-MASLD) by geographical region, age and sex during 1990-2021. METHODS Global Burden of Diseases Study 2021 data were analysed to assess LC-MASLD incidence, prevalence, mortality and disability-adjusted life years (DALYs). Temporal trends during 1990-2021 were measured by 'estimated annual percentage change' (EAPC). Inequalities of LC-MASLD burden across countries were evaluated by the slope index of inequality (SII) and the relative concentration index (RCI). RESULTS During 1990-2021, LC-MASLD rose annually by 0.73% in incidence and prevalence, 0.19% in mortality and 0.16% in DALYs. In 2021, the Middle East and North Africa had the highest incidence and prevalence and Andean and Central Latin America had the highest mortality and DALY rates. While LC-MASLD incidence was earliest in the 15-19 age group, both prevalence and DALY rates peaked at 75-79 years for both sexes. Inequalities in mortality and DALYs by countries' socioeconomic development index increased during 1990-2021, demonstrated by a decline in SII from -0.09 to -0.56 per 100 000 for mortality and from 1.41 to -7.74 per 100 000 for DALYs. RCI demonstrated similar findings. CONCLUSION The LC-MASLD burden is increasing globally, especially in economically disadvantaged countries, with widening disease inequalities during 1990-2021. Effective prevention and subregional interventions are crucial, with a specific focus on resource optimisation for disadvantaged populations.
Collapse
Affiliation(s)
- Fang Lu
- China‐Australia Joint Research Center for Infectious Diseases, School of Public HealthXi'an Jiaotong University Health Science CenterXi'anShaanxiPeople's Republic of China
| | - Jinli Liu
- China‐Australia Joint Research Center for Infectious Diseases, School of Public HealthXi'an Jiaotong University Health Science CenterXi'anShaanxiPeople's Republic of China
| | - Bingyang She
- China‐Australia Joint Research Center for Infectious Diseases, School of Public HealthXi'an Jiaotong University Health Science CenterXi'anShaanxiPeople's Republic of China
| | - Hailin Yang
- China‐Australia Joint Research Center for Infectious Diseases, School of Public HealthXi'an Jiaotong University Health Science CenterXi'anShaanxiPeople's Republic of China
| | - Fanpu Ji
- Department of Infectious DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Lei Zhang
- China‐Australia Joint Research Center for Infectious Diseases, School of Public HealthXi'an Jiaotong University Health Science CenterXi'anShaanxiPeople's Republic of China
- Melbourne Sexual Health CentreAlfred HealthMelbourneAustralia
- Central Clinical School, Faculty of MedicineMonash UniversityMelbourneAustralia
| |
Collapse
|
383
|
Meijnikman AS, Fondevila MF, Arrese M, Kisseleva T, Bataller R, Schnabl B. Towards more consistent models and consensual terminology in preclinical research for steatotic liver disease. J Hepatol 2025; 82:760-766. [PMID: 39581500 DOI: 10.1016/j.jhep.2024.11.025] [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: 06/28/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Steatotic liver disease (SLD) is one of the most prevalent liver conditions globally and a leading cause of liver transplantation, yet therapeutic advances have not kept pace with its major impact on global morbidity and mortality. This underscores the critical importance of developing and refining relevant preclinical animal models. However, preclinical research has faced significant challenges, with concerns about the translational validity of animal models, as findings often fail to accurately reflect human disease. With the recent adoption of new nomenclature for SLD in humans, questions have arisen about how to integrate these changes into preclinical models. Here, we offer suggestions on how to improve preclinical models, including the incorporation of factors such as diet, alcohol, and other metabolic stressors, to better replicate the complexity of human disease. While implementing these improvements presents practical challenges, doing so is essential for enhancing the translational relevance and reproducibility of animal studies, and advancing therapeutic discoveries. Furthermore, we address the persisting inconsistency in terminology used in animal studies and propose clinically meaningful terms that can be applied consistently to preclinical research.
Collapse
Affiliation(s)
- Abraham S Meijnikman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 BK Amsterdam, the Netherlands
| | - Marcos F Fondevila
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Ramon Bataller
- Liver Unit, Hospital Clinic. Institut d'Investigacions Biomediques August Pi i Sunyer (IDI-BAPS). Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
384
|
Gursan A, de Graaf RA, Thomas MA, Prompers JJ, De Feyter HM. Deuterium MRS for In Vivo Measurement of Lipogenesis in the Liver. NMR IN BIOMEDICINE 2025; 38:e70014. [PMID: 39994887 DOI: 10.1002/nbm.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Hepatic de novo lipogenesis (DNL) plays a key role in the pathogenesis of several metabolic diseases that affect the liver. In humans, the detection of deuterium (2H) in triglycerides from very low density lipoprotein collected from blood after administration of deuterated water (D2O) is commonly used as an indirect estimate of hepatic DNL. Here, we tested in rats (1) the feasibility to detect 2H-labeling directly in liver lipids in vivo by using noninvasive 2H MRS and (2) to what extent these results correlated with the gold standard measurement of DNL in excised liver tissue. To increase hepatic DNL, half of the animals (n = 4) underwent a 7-week dietary intervention in which fructose was provided in drinking water. Deuterium MRS data were acquired from a single voxel placed in the liver. In vivo 2H MRS data showed 2H-labeling in the combined peak of methyl and methylene resonances after 1 week of administrati NBM_70014 on of 5% D2O as drinking water. DNL was calculated using 1H and 2H NMR data acquired from extracted lipids of excised liver tissue. The 2H lipid level measured in vivo correlated with the ex vivo estimates of hepatic DNL (r = 0.81, p = 0.016). These results demonstrate the feasibility of direct detection of deuterium labeling in liver lipids using localized 2H MRS in vivo and indicate the potential of this approach to measure hepatic DNL. These initial observations provide a basis for the method to be translated and to develop noninvasive, quantitative measurements of hepatic DNL in humans.
Collapse
Affiliation(s)
- Ayhan Gursan
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robin A de Graaf
- Department of Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University, New Haven, Connecticut, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Monique A Thomas
- Department of Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University, New Haven, Connecticut, USA
| | - Jeanine J Prompers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
- Departments of Human Biology and Imaging, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Henk M De Feyter
- Department of Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University, New Haven, Connecticut, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
385
|
Li Z, Guo H, He H, Wang S, Pei S, Xie L. The relationship between smoking cessation history and significant liver fibrosis among the population with metabolic dysfunction-associated steatotic liver disease in the United States. PLoS One 2025; 20:e0320573. [PMID: 40168280 PMCID: PMC11960941 DOI: 10.1371/journal.pone.0320573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Smoking was identified as a risk factor for the development of liver fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, the association between smoking cessation history and the development of liver fibrosis remains unclear. This study was intended to analyze the association between smoking cessation history and significant liver fibrosis in adult MASLD participants in the United States. METHODS This study utilized data from 2643 patients with MASLD from the National Health and Nutrition Examination Survey (NHANES). Significant liver fibrosis was detected based on transient elastography measurements. According to the smoking questionnaire data, patients were categorized as non-smokers, ex-smokers and current smokers. A multivariate logistic regression analysis, adjusted for weights, was performed to investigate the relationship between smoking cessation history and the presence of significant liver fibrosis in participants with MASLD. RESULTS A total of 2643 patients with MASLD were included in this study. Compared with non-smokers, ex-smokers had a slightly elevated risk of developing significant liver fibrosis (OR: 1.07, 95% CI: 1.02-1.13). Specifically, a positive correlation was observed between patients who quit smoking for < 20 years and significant liver fibrosis (OR: 1.07, 95% CI: 1.01-1.15). Furthermore, MASLD patients who started regularly smoking at an age of ≤ 20 years (OR: 1.09, 95% CI: 1.02-1.16) and had a smoking duration of ≥ 10 years before quitting (OR: 1.10, 95% CI: 1.02-1.18) were also highly correlated with an increased likelihood of developing significant liver fibrosis. CONCLUSIONS This study revealed that individuals with MASLD who have ceased smoking exhibit an elevated risk for significant liver fibrosis when compared to those who never smoked. It is highly emphasized that MASLD patients who quit smoking for < 20 years, started regularly smoking at an age of ≤ 20 years, and had a smoking duration of ≥ 10 years before quitting should be extremely vigilant regarding the risk of significant liver fibrosis.
Collapse
Affiliation(s)
- Zhongtao Li
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hao Guo
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Hongyu He
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Shu Wang
- Department of Urology Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shufen Pei
- Department of Otolaryngology-Head and Neck Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liang Xie
- Department of General Surgery (Wenhua Road Campus), The Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Hepatobiliary Pancreatic and Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
386
|
Jiao W, Zhou H, Zhang J, Yuan Y, Wei J, Gong X, Sun Y, Sang L, Yu M. Mesoporous silica nanoparticles as sensitizers: A novel approach to enhancing shear wave elastography in liver stiffness measurement. BIOMATERIALS ADVANCES 2025; 169:214171. [PMID: 39764903 DOI: 10.1016/j.bioadv.2024.214171] [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: 08/12/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025]
Abstract
PURPOSE The objective of this study is to elucidate the sensitizing effect of mesoporous silica nanoparticles (MSNs) on shear wave elastography (SWE) and to investigate the potential application of MSNs as a sensitizer to enhance the sensitivity of SWE in the diagnosis of metabolic-associated steatohepatitis (MASH). MATERIALS AND METHODS The in vitro gelatin models with varying ratios were assessed using SWE to identify the gelatin ratio that most closely approximates with human liver stiffness. Following the characterization of the dispersion properties of MSNs, in vitro models incorporating MSNs of different particle sizes were developed. The variations in shear wave velocity (SWV) within these models were measured and subjected to statistical analysis using SWE. The biocompatibility of the MSNs was evaluated, and the MSN solution was subsequently administered into a MASH animal model. The sensitizing effect of SWE on rat liver was then analyzed statistically. RESULTS The in vitro model demonstrated that MSNs with smaller particle sizes (100 nm and 200 nm) facilitated the propagation of SWV, thereby enhancing the sensitivity of SWE (P < 0.05). Additionally, the cell viability and hemolysis ratio of 100 nm MSNs were superior to those of 200 nm MSNs (P < 0.05). In vivo animal model experiments indicated that 100 nm fluorescence-modified MSNs could penetrate the MASH liver and elevate the liver stiffness value as measured by SWE. CONCLUSION MSNs have the potential to enhance the sensitivity of SWE in the diagnosis of MASH. This approach offers novel insights for improving the efficacy of SWE in clinical diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Weijie Jiao
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Huihui Zhou
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Jun Zhang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Yuan Yuan
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, No.169 Changle West Rd, 710032 Xi'an, China
| | - Junci Wei
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Xue Gong
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Yuanyuan Sun
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Lin Sang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China
| | - Ming Yu
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No.127 Changle West Rd, 710032 Xi'an, Shaanxi, China.
| |
Collapse
|
387
|
Daniels SJ, Nelander K, Eriksson J, Jermutus L, Saillard J, Oyesola S, Tavaglione F, Arrese M, Ladrón de Guevara AL, Vespasiani-Gentilucci U, Alkhouri N, Blau JE. Design and rationale for a global novel non-invasive screening observational study using genetics and non-invasive methodologies to identify at-risk MASLD participants: The ALIGN study. Contemp Clin Trials Commun 2025; 44:101437. [PMID: 39916681 PMCID: PMC11800087 DOI: 10.1016/j.conctc.2025.101437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common chronic liver disease that is heterogenous in nature with various drivers and modifiers such as metabolic dysfunction and genetic factors. MASLD and the progressive subtype, metabolic dysfunction-associated steatohepatitis (MASH) represent the most rapidly increasing cause of liver-related mortality. There are limited treatment options for patients living with MASLD and MASH, various treatments with an array of different targets are under investigation and one therapeutic has been approved since the initiation of this study. Clinical trials investigating treatments for MASLD and MASH are associated with a high screen failure rate, driven largely by the regulatory required histological inclusion criteria for clinical trial eligibility. Other available clinically utilized biomarkers, typically referred to as non-invasive tests (NITs), can assess both the presence of steatosis and the severity of liver fibrosis in patients with MASLD and MASH in the clinic but are not yet approved over histological changes as endpoints for pivotal trials. However, the use of NITs have been demonstrated to increase the likelihood of meeting clinical trial entry criteria. All-Liver Interventional Global Network (ALIGN) is the first described multi-centre global observational screening study aimed at identifying individuals with a high likelihood of MASLD/MASH interested in participating in therapeutic clinical trials using non-invasive methodologies and genetic testing. This study represents a valuable prototype for industry and academic groups looking to evaluate large populations for MASH eligibility and interest in clinical trial participation.
Collapse
Affiliation(s)
| | - Karin Nelander
- CVRM Biometrics, Late CVRM, AstraZeneca, Gothenburg, Sweden
| | - John Eriksson
- CVRM Biometrics, Late CVRM, AstraZeneca, Gothenburg, Sweden
| | - Lutz Jermutus
- Research, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jelena Saillard
- Clinical Operations CVRM, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Stephanie Oyesola
- Clinical Operations CVRM, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Federica Tavaglione
- Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Umberto Vespasiani-Gentilucci
- Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico and Università Campus Bio-Medico di Roma, Rome, Italy
| | - Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ, USA
| | - Jenny E. Blau
- Early Clinical Development, Early CVRM, AstraZeneca, Gaithersburg, USA
| |
Collapse
|
388
|
Lam BP, Bartholomew J, Bau S, Gilles H, Keller A, Moore A, Nader K, Richards L, Henry L, Younossi ZM. Focused Recommendations for the Management of Metabolic Dysfunction-Associated Steatohepatitis (MASH) by Advanced Practice Providers in the United States. J Clin Gastroenterol 2025; 59:298-309. [PMID: 39889206 DOI: 10.1097/mcg.0000000000002140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) has become the dominant cause of liver disease in the United States. With the growing burden of this disease in gastroenterology practices, the identification and treatment of those at risk of developing adverse outcomes (cirrhosis, hepatocellular carcinoma, or liver-related death) has become urgent. In recent years, the development of noninvasive tests (NITs) to identify "at-risk MASH" patients have provided cost-effective algorithms to identify these patients. Although treatment has historically been limited to lifestyle modification, recent FDA approval of resmetirom for noncirrhosis MASH with stages 2 and 3 fibrosis has provided a new opportunity in the United States to provide these patients with novel treatment options. Other new effective treatment regimens are on the horizon. Given that gastroenterology and hepatology practices in the United States heavily rely on advanced practice providers (APPs) to manage patients with MASLD, the APP Committee of the Global NASH/MASH Council has curated the essentials of day-to-day MASH management for our busy gastrohepatology providers and their APP colleagues. The goal of this document is to equip and mobilize more GI providers with the requisite competencies for the management of at-risk MASH, given the rapidly evolving MASH treatment landscape.
Collapse
Affiliation(s)
- Brian P Lam
- The Global NASH Council
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church
| | | | - Sherona Bau
- The Global NASH Council
- The University of California, Los Angeles, the Pfleger Liver Institute, LA
| | - HoChong Gilles
- The Global NASH Council
- Central Virginia Veterans Affairs Health Care System, Richmond
| | - Andrea Keller
- The Global NASH Council
- MedStar Georgetown Transplant Institute, Fairfax, VA
- MedStar Georgetown University Hospital, Washington, DC
| | - Ann Moore
- The Global NASH Council
- Arizona Liver Health, Chandler, AZ
| | - Khalil Nader
- The Global NASH Council
- GW Medicine, The George Washington University, Washington, DC
| | - Lisa Richards
- The Global NASH Council
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Linda Henry
- The Global NASH Council
- Center for Outcomes Research in Liver Disease
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church
| | - Zobair M Younossi
- The Global NASH Council
- Center for Outcomes Research in Liver Disease
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church
| |
Collapse
|
389
|
Corma-Gómez A, Corona-Mata D, Martín-Carmona J, Galindo MJ, Camacho A, Martín-Sierra C, Gallo-Marín M, Rincón P, Perez-Valero I, Pérez-García M, Carrasco-Dorado A, Pineda JA, Rivero-Juárez A, Rivero A, Real LM, Macías J. FibroScan-AST Score vs Liver Stiffness for the Prediction of Liver Events After HCV Cure. Open Forum Infect Dis 2025; 12:ofae628. [PMID: 40201720 PMCID: PMC11977108 DOI: 10.1093/ofid/ofae628] [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: 07/15/2024] [Accepted: 10/12/2024] [Indexed: 04/10/2025] Open
Abstract
Background Liver stiffness (LS) predicts liver complication occurrence in patients with hepatitis C virus (HCV) infection after sustained virological response (SVR). The FibroScan-AST (FAST) score, which includes aspartate aminotransferase (AST) and controlled attenuation parameter (CAP; measured by FibroScan), may improve the prediction ability of isolated LS. Our aim was to compare the predictive capacity of LS vs FAST in this setting. Methods Multicenter cohort study including individuals with HIV/HCV coinfection or HCV monoinfection from Spain if they had (1) LS ≥9.5 kPa pretreatment, (2) SVR with a direct-acting antiviral (DAA)-based regimen, and (3) LS and CAP measurement at SVR. Fatty liver disease (FLD) was defined as CAP ≥248 dB/m. The primary outcome was the occurrence of a liver complication (decompensation or hepatocellular carcinoma [HCC]) after SVR. Results Three hundred patients were included; 213 (71%) had HIV. At SVR, 131 (44%) had FLD. The FAST score was <0.35 in 182 (61%), 0.35-0.67 in 79 (27%), and >0.67 in 34 (12%) patients. After a median (Q1-Q3) follow-up of 73 (53-83) months, 36 (12%) liver complications (15 [5%] HCC) occurred. LS was independently associated with an increased risk of developing liver complications (sub-hazard ratio [sHR], 1.06; 95% CI, 1.04-1.08; P < .001). In a separate model, FAST ≥0.35 was also independently associated with greater risk of liver complications (sHR, 8.12; 95% CI, 3.11-21.17; P < .001). The area under the receiver operating characteristics curve of the model based on LS was 0.83 (95% CI, 0.76-0.91), and that of the model based on FAST was 0.80 (95% CI, 0.72-0.88; P = .158). Conclusions The FAST score predicts the development of liver events after SVR but does not improve the predictive capacity of LS alone at this time point.
Collapse
Affiliation(s)
- Anaïs Corma-Gómez
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Diana Corona-Mata
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Jésica Martín-Carmona
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Sevilla (US), Sevilla, Spain
| | - María José Galindo
- Unit of Infectious Diseases, Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Angela Camacho
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Carmen Martín-Sierra
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
| | - Marina Gallo-Marín
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Pilar Rincón
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
| | - Ignacio Perez-Valero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Margarita Pérez-García
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
| | - Angela Carrasco-Dorado
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Juan A Pineda
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Sevilla (US), Sevilla, Spain
| | - Antonio Rivero-Juárez
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Antonio Rivero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Virology and Zoonoses Research Group, Unit of Infectious Diseases, Hospital Universitario Reina Sofía, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Universidad de Córdoba (UCO), Córdoba, Spain
| | - Luis M Real
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Sevilla (US), Sevilla, Spain
| | - Juan Macías
- Grupo de Virología Clínica e ITS Cinical Virology and STIs Group, Unit of Infectious Diseases and Microbiology, de Hospital Universitario Virgen de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Sevilla (US), Sevilla, Spain
| |
Collapse
|
390
|
Yun B, Park H, Lee J, Kim BK, Yoon JH. Statin use and liver-related prognosis among patients with MASLD. JHEP Rep 2025; 7:101313. [PMID: 40124167 PMCID: PMC11929059 DOI: 10.1016/j.jhepr.2024.101313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 03/25/2025] Open
Abstract
Background & Aims Metabolic dysfunction-associated steatotic liver disease (MASLD) is a highly prevalent liver condition. We investigated whether statin use reduces liver-related events (LREs) risk among patients with MASLD or MASLD with increased alcohol intake (MetALD). Methods This nationwide cohort study included individuals aged ≥40 years with MASLD/MetALD undergoing health examinations between 2012 and 2013. The primary outcome was LREs; hepatocellular carcinoma (HCC), decompensated liver cirrhosis (DLC), and liver-related mortality. Secondary outcomes included HCC, DLC, and steatotic liver disease (SLD) regression, all-cause mortality, cardiovascular diseases (CVD)-related mortality, and liver-related mortality, respectively. Multivariable Cox regression was performed to estimate the risk of LREs associated with statin use. Results Among 516,575 individuals (median follow-up: 10.1 years), statin users experienced significantly lower LRE rates (1.6%) compared with non-users (2.0%, p <0.001). Multivariable Cox regression analysis revealed that statin use was associated with reduced risks of LREs (adjusted hazard ratio [aHR] 0.64, 95% CI 0.61-0.68), HCC (aHR 0.52, 95% CI 0.47-0.58), DLC (aHR 0.58, 95% CI 0.52-0.65), all-cause mortality (aHR 0.81, 95% CI 0.78-0.84), CVD-related mortality (aHR 0.87, 95% CI 0.80-0.95), and liver-related mortality (aHR 0.51, 95% CI 0.46-0.57). Furthermore, statin use was associated with SLD regression (aHR 1.18, 95% CI 1.15-1.21). Stratified analyses consistently demonstrated risk reductions across all subgroups, particularly in patients with elevated alanine aminotransferase levels. Sensitivity analyses confirmed the robustness of these associations. Conclusions Statins are significantly associated with reduced LRE risk in patients with MASLD, especially among those with elevated alanine aminotransferase levels, suggesting a viable preventive strategy for such population. Impact and implications Our study provides critical evidence supporting the role of statins in reducing liver-related events in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), a condition with significant global health impact. These findings are particularly relevant for clinicians managing high-risk patients with MASLD, especially those with elevated alanine aminotransferase levels, as they highlight the potential for statins to mitigate both liver and cardiovascular risks. By demonstrating the robustness of these results through comprehensive sensitivity and stratified analyses, our research underscores the importance of integrating statin therapy into the management of MASLD. This has practical implications for physicians, researchers, and policymakers in developing guidelines and preventive strategies to improve long-term liver and cardiovascular outcomes.
Collapse
Affiliation(s)
- Byungyoon Yun
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- The Institute for Occupational Health, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Innovation in Digital Healthcare, Yonsei University Health System, Seoul, Republic of Korea
| | - Heejoo Park
- Department of Public Health, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Jian Lee
- Department of Public Health, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Jin-Ha Yoon
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- The Institute for Occupational Health, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
391
|
Zhang Q, Xu F, Wang Z, Liu S, Zhu S, Zhang S, Wu S. Long-Term Risk of Inflammatory Bowel Disease With MASLD: A Large-Scale Prospective Cohort Study in UK Biobank. J Gastroenterol Hepatol 2025; 40:855-865. [PMID: 39828371 DOI: 10.1111/jgh.16880] [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: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Similar worsening epidemics globally have been showed in newly coined metabolic dysfunction-associated steatotic liver disease (MASLD) and inflammatory bowel disease (IBD). We aimed to investigate the prospective association of MASLD, MASLD types, and cardiometabolic risk factors (CMRFs) with long-term risk of incident IBD in a large-scale population cohort. METHODS Participants free of IBD at enrollment from UK Biobank were included. Baseline MASLD was measured by fatty liver index together with at least one CMRF, based on the latest AASLD/EASL criteria. MASLD type was classified as pure MASLD and MetALD (MASLD with increased alcohol intake). Primary outcome was incident IBD, including ulcerative colitis (UC) and Crohn's disease (CD). Multivariable Cox regression was conducted to examine the related associations. RESULTS Overall, 403 520 participants (aged 56.2 ± 8.1 years, 45.6% males) were included. Of whom, 151 578 (37.6%) were considered as MASLD at baseline. During a median of 13.0 years' follow-up, 2398 IBD cases were identified. Compared with normal population, individuals with MASLD showed significant higher associations of incident IBD (HR = 1.39, 95% CI: 1.21-1.60), UC (HR = 1.34, 95% CI: 1.13-1.58), and CD (HR = 1.51, 95% CI: 1.20-1.89). Meanwhile, results were consistent when assessing pure MASLD (HR = 1.43, 95% CI: 1.23-1.66) and MetALD (HR = 1.46, 95% CI: 1.15-1.86). The excess risk of incident IBD was more evident with the increase of CMRFs numbers (ptrend < 0.001). CONCLUSION MASLD, either pure MASLD or MetALD, and a combination of different CMRFs are all associated with increased risk of IBD, including both UC and CD. Additionally, there is greater risk of incident IBD as the number of CMRFs increase.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Fang Xu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Zuyao Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shanshan Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Diseases Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| |
Collapse
|
392
|
Dixon ED, Claudel T, Nardo AD, Riva A, Fuchs CD, Mlitz V, Busslinger G, Scharnagl H, Stojakovic T, Senéca J, Hinteregger H, Grabner GF, Kratky D, Verkade H, Zimmermann R, Haemmerle G, Trauner M. Inhibition of ATGL alleviates MASH via impaired PPARα signalling that favours hydrophilic bile acid composition in mice. J Hepatol 2025; 82:658-675. [PMID: 39357546 DOI: 10.1016/j.jhep.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND & AIMS Adipose triglyceride lipase (ATGL) is an attractive therapeutic target in insulin resistance and metabolic dysfunction-associated steatotic liver disease (MASLD). This study investigated the effects of pharmacological ATGL inhibition on the development of metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis in mice. METHODS Streptozotocin-injected male mice were fed a high-fat diet to induce MASH. Mice receiving the ATGL inhibitor atglistatin (ATGLi) were compared to controls using liver histology, lipidomics, metabolomics, 16s rRNA, and RNA sequencing. Human ileal organoids, HepG2 cells, and Caco2 cells treated with the human ATGL inhibitor NG-497, HepG2 ATGL knockdown cells, gel-shift, and luciferase assays were analysed for mechanistic insights. We validated the benefits of ATGLi on steatohepatitis and fibrosis in a low-methionine choline-deficient mouse model. RESULTS ATGLi improved serum liver enzymes, hepatic lipid content, and histological liver injury. Mechanistically, ATGLi attenuated PPARα signalling, favouring hydrophilic bile acid (BA) synthesis with increased Cyp7a1, Cyp27a1, Cyp2c70, and reduced Cyp8b1 expression. Additionally, reduced intestinal Cd36 and Abca1, along with increased Abcg5 expression, were consistent with reduced levels of hepatic triacylglycerol species containing polyunsaturated fatty acids, like linoleic acid, as well as reduced cholesterol levels in the liver and plasma. Similar changes in gene expression associated with PPARα signalling and intestinal lipid transport were observed in ileal organoids treated with NG-497. Furthermore, HepG2 ATGL knockdown cells revealed reduced expression of PPARα target genes and upregulation of genes involved in hydrophilic BA synthesis, consistent with reduced PPARα binding and luciferase activity in the presence of the ATGL inhibitors. CONCLUSIONS Inhibition of ATGL attenuates PPARα signalling, translating into hydrophilic BA composition, interfering with dietary lipid absorption, and improving metabolic disturbances. Validation with NG-497 opens a new therapeutic perspective for MASLD. IMPACT AND IMPLICATIONS Despite the recent approval of drugs novel mechanistic insights and pathophysiology-oriented therapeutic options for MASLD (metabolic dysfunction-associated steatotic liver disease) are still urgently needed. Herein, we show that pharmacological inhibition of ATGL, the key enzyme in lipid hydrolysis, using atglistatin (ATGLi), improves MASH (metabolic dysfunction-associated steatohepatitis), fibrosis, and key features of metabolic dysfunction in mouse models of MASH and liver fibrosis. Mechanistically, we demonstrated that attenuation of PPARα signalling in the liver and gut favours hydrophilic bile acid composition, ultimately interfering with dietary lipid absorption. One of the drawbacks of ATGLi is its lack of efficacy against human ATGL, thus limiting its clinical applicability. Against this backdrop, we could show that ATGL inhibition using the human inhibitor NG-497 in human primary ileum-derived organoids, Caco2 cells, and HepG2 cells translated into therapeutic mechanisms similar to ATGLi. Collectively, these findings reveal a possible new avenue for MASLD treatment.
Collapse
Affiliation(s)
- Emmanuel Dauda Dixon
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alexander Daniel Nardo
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alessandra Riva
- Chair of Nutrition and Immunology, School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Claudia Daniela Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Veronika Mlitz
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Georg Busslinger
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Tatjana Stojakovic
- Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Austria
| | - Joana Senéca
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria; Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Helga Hinteregger
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Gernot F Grabner
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Dagmar Kratky
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Henkjan Verkade
- Department of Paediatrics, University Medical Centre Groningen, Groningen, Netherlands
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
393
|
Koch V, Gotta J, Chernyak V, Cengiz D, Torgashov K, Eichler K, Vilgrain V, Martin SS, Ziegengeist NS, Konrad P, Booz C, Yel I, D'Angelo T, Mahmoudi S, Scholtz J, Bernatz S, Alizadeh LS, Cimprich M, Solim LA, Thalhammer A, Gruber‐Rouh T, Hammerstingl RM, Zeuzem S, Finkelmeier F, Pathil‐Warth A, Onay M, Kinzler MN, Darwish O, Annio G, Taylor SA, Wild P, Dahmer I, Herrmann E, Almansour H, Vogl TJ, Gruenewald LD, Sinkus R. Biomechanical Assessment of Liver Integrity: Prospective Evaluation of Mechanical Versus Acoustic MR Elastography. J Magn Reson Imaging 2025; 61:1890-1904. [PMID: 39165139 PMCID: PMC11896941 DOI: 10.1002/jmri.29560] [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: 05/24/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Magnetic resonance elastography (MRE) can quantify tissue biomechanics noninvasively, including pathological hepatic states like metabolic dysfunction-associated steatohepatitis. PURPOSE To compare the performance of 2D/3D-MRE using the gravitational (GT) transducer concept with the current commercial acoustic (AC) solution utilizing a 2D-MRE approach. Additionally, quality index markers (QIs) were proposed to identify image pixels with sufficient quality for reliably estimating tissue biomechanics. STUDY TYPE Prospective. POPULATION One hundred seventy participants with suspected or confirmed liver disease (median age, 57 years [interquartile range (IQR), 46-65]; 66 females), and 11 healthy volunteers (median age, 31 years [IQR, 27-34]; 5 females). FIELD STRENGTH/SEQUENCE Participants were scanned twice at 1.5 T and 60 Hz vibration frequency: first, using AC-MRE (2D-MRE, spin-echo EPI sequence, 11 seconds breath-hold), and second, using GT-MRE (2D- and 3D-MRE, gradient-echo sequence, 14 seconds breath-hold). ASSESSMENT Image analysis was performed by four independent radiologists and one biomedical engineer. Additionally, superimposed analytic plane shear waves of known wavelength and attenuation at fixed shear modulus were used to propose pertinent QIs. STATISTICAL TESTS Spearman's correlation coefficient (r) was applied to assess the correlation between modalities. Interreader reproducibility was evaluated using Bland-Altman bias and reproducibility coefficients. P-values <0.05 were considered statistically significant. RESULTS Liver stiffness quantified via GT-2D/3D correlated well with AC-2D (r ≥ 0.89 [95% CI: 0.85-0.92]) and histopathological grading (r ≥ 0.84 [95% CI: 0.72-0.91]), demonstrating excellent agreement in Bland-Altman plots and between readers (κ ≥ 0.86 [95% CI: 0.81-0.91]). However, GT-2D showed a bias in overestimating stiffness compared to GT-3D. Proposed QIs enabled the identification of pixels deviating beyond 10% from true stiffness based on a combination of total wave amplitude, temporal sinusoidal nonlinearity, and wave signal-to-noise ratio for GT-3D. CONCLUSION GT-MRE represents an alternative to AC-MRE for noninvasive liver tissue characterization. Both GT-2D and 3D approaches correlated strongly with the established commercial approach, offering advanced capabilities in abdominal imaging compared to AC-MRE. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Jennifer Gotta
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Victoria Chernyak
- Department of RadiologyMemorial Sloan Kettering Cancer CenterNew York CityNew YorkUSA
| | - Duygu Cengiz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Katerina Torgashov
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Katrin Eichler
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Valérie Vilgrain
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, UMR 1149 INSERM, Université de ParisParisFrance
- Department of RadiologyBeaujon University Hospital Paris Nord, AP‐HPClichyFrance
| | - Simon S. Martin
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Nicole S. Ziegengeist
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Paul Konrad
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Christian Booz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Ibrahim Yel
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Tommaso D'Angelo
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Scherwin Mahmoudi
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Jan‐Erik Scholtz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Simon Bernatz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Leona S. Alizadeh
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Marina Cimprich
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Levent A. Solim
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Axel Thalhammer
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Tatjana Gruber‐Rouh
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Renate M. Hammerstingl
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Stefan Zeuzem
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Fabian Finkelmeier
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Anita Pathil‐Warth
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Melis Onay
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Maximilian N. Kinzler
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Omar Darwish
- School of Biomedical Engineering and Imaging Sciences, King's College LondonLondonUK
| | - Giacomo Annio
- Laboratory of Translational Vascular Sciences, U1148, INSERM, Université de ParisParisFrance
| | - Stuart A. Taylor
- Radiology DepartmentUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Peter Wild
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Iulia Dahmer
- Institute of Biostatistics and Mathematical Modeling, Faculty of Medicine, Goethe University FrankfurtFrankfurt am MainGermany
| | - Eva Herrmann
- Institute of Biostatistics and Mathematical Modeling, Faculty of Medicine, Goethe University FrankfurtFrankfurt am MainGermany
| | - Haidara Almansour
- Department of Diagnostic and Interventional RadiologyEberhard‐Karls University TuebingenTuebingenGermany
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Leon D. Gruenewald
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Ralph Sinkus
- School of Biomedical Engineering and Imaging Sciences, King's College LondonLondonUK
- Laboratory of Translational Vascular Sciences, U1148, INSERM, Université de ParisParisFrance
| |
Collapse
|
394
|
Wu W, Guo Y, Li Q, Jia C. Exploring the potential of large language models in identifying metabolic dysfunction-associated steatotic liver disease: A comparative study of non-invasive tests and artificial intelligence-generated responses. Liver Int 2025; 45:e16112. [PMID: 39526465 DOI: 10.1111/liv.16112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS This study sought to assess the capabilities of large language models (LLMs) in identifying clinically significant metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS We included individuals from NHANES 2017-2018. The validity and reliability of MASLD diagnosis by GPT-3.5 and GPT-4 were quantitatively examined and compared with those of the Fatty Liver Index (FLI) and United States FLI (USFLI). A receiver operating characteristic curve was conducted to assess the accuracy of MASLD diagnosis via different scoring systems. Additionally, GPT-4V's potential in clinical diagnosis using ultrasound images from MASLD patients was evaluated to provide assessments of LLM capabilities in both textual and visual data interpretation. RESULTS GPT-4 demonstrated comparable performance in MASLD diagnosis to FLI and USFLI with the AUROC values of .831 (95% CI .796-.867), .817 (95% CI .797-.837) and .827 (95% CI .807-.848), respectively. GPT-4 exhibited a trend of enhanced accuracy, clinical relevance and efficiency compared to GPT-3.5 based on clinician evaluation. Additionally, Pearson's r values between GPT-4 and FLI, as well as USFLI, were .718 and .695, respectively, indicating robust and moderate correlations. Moreover, GPT-4V showed potential in understanding characteristics from hepatic ultrasound imaging but exhibited limited interpretive accuracy in diagnosing MASLD compared to skilled radiologists. CONCLUSIONS GPT-4 achieved performance comparable to traditional risk scores in diagnosing MASLD and exhibited improved convenience, versatility and the capacity to offer user-friendly outputs. The integration of GPT-4V highlights the capacities of LLMs in handling both textual and visual medical data, reinforcing their expansive utility in healthcare practice.
Collapse
Affiliation(s)
- Wanying Wu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuhu Guo
- Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Qi Li
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Congzhuo Jia
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
395
|
Qu J. Advancing Noninvasive Diagnostics for Hepatic Steatosis in MASLD: The Pivotal Role of UDFF. Acad Radiol 2025; 32:1958-1960. [PMID: 39971700 DOI: 10.1016/j.acra.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Affiliation(s)
- Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China.
| |
Collapse
|
396
|
Ito H, Tsugami E, I C, Miura S, Matsumoto S, Inoue H, Antoku S, Yamasaki T, Mori T, Togane M. Benefits and disadvantages of combination therapy with imeglimin and metformin in patients with type 2 diabetes. Expert Opin Pharmacother 2025; 26:773-781. [PMID: 40151916 DOI: 10.1080/14656566.2025.2486354] [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: 12/14/2024] [Accepted: 03/26/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND We retrospectively examined the benefits and disadvantages of adding imeglimin to metformin therapy in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS Ninety-four patients with type 2 diabetes who had been administered imeglimin were included in the safety analysis set (SAS). Sixty-four patients who had been treated with imeglimin for over 6 months were included in the full analysis set (FAS). The primary outcome was the change in HbA1c levels in the FAS. The secondary outcomes were gastrointestinal adverse events (AEs) in the SAS and changes in body weight and FIB-4 in theFAS. RESULTS In the SAS, gastrointestinal AEs occurred in 15 of 40 (38%) metformin users and 6 of 54 (11%) non-users. In the FAS, HbA1c levels were significantly reduced in both metformin users (n = 27, baseline, 8.5 ± 1.1%;6 months, 7.7 ± 1.2%) and non-users (n = 37, baseline, 8.0 ± 0.9%; 6 months,7.4 ± 0.9%). While body weight (from 72.0 ± 20.5 kg to 70.9 ± 20.8 kg) and FIB-4 (from 1.27 ± 0.57 to 1.17 ± 0.49) significantly decreased in metformin users, they did not significantly differ in non-users. CONCLUSIONS Adding imeglimin to metformin therapy demonstrated favorable reductions in HbA1c, body weight, and FIB-4 in patients with type 2 diabetes, although it was associated with the incidence of gastrointestinal AEs. TRIAL REGISTRATION UMIN000055241.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Emiko Tsugami
- Department of Pharmacy, Edogawa Hospital, Tokyo, Japan
| | - Chiaki I
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Shun Miura
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Suzuko Matsumoto
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Hideyuki Inoue
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Shinichi Antoku
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Tomoko Yamasaki
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Toshiko Mori
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Michiko Togane
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| |
Collapse
|
397
|
Elkattawy HA, Alsemeh AE, Ali LS, Ahmed MM, Eltaweel AM, Shaikh FM, Behiry A, Hassan AES, Sabir DK, Elsherbini DMA, Ali SK, Zakari MO, Mojaddidi MA, Ali EK, Elbastawisy YM, Hadhoud S. Decorin alleviates non-alcoholic fatty liver disease in rats with polycystic ovary syndrome. Tissue Cell 2025; 93:102689. [PMID: 39705869 DOI: 10.1016/j.tice.2024.102689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Endocrine multisystem defect polycystic ovary syndrome (PCOS) causes hyperandrogenism and infertility. Half of PCOS women have (non-alcoholic fatty liver disease) NAFLD, which increases metabolic disease risk. We tested decorin's effect on NAFLD and related processes in PCOS. NAFLD+PCOS, PCOS+decorin, and control rats were studied. Decorin was evaluated on NAFLD/PCOS rats. Test group rats received HF for eight weeks to generate NAFLD. The rats got 1 mg/kg letrozole orally daily for 21 days to diagnosis PCOS. Afterward, rats got injectable decorin for 14 days. Body weight, liver weight, liver coefficient Abdominal Circumference (AC) and body mass index (BMI) were determined. Blood triglycerides (TG), total cholesterol, LDL-c, AST, and glucose were measured. The insulin, testosterone, estrogen, LH, and FSH were measured by ELISA. GPx, SOD, MDA, TNF-, and Caspase-3 liver immunohistochemistry were evaluated. NAFLD liver tissues in PCOS models showed biochemical and histological alterations. NAFLD+PCOS raised BMI, AC, liver weight, and coefficient. Blood glucose, insulin resistance, TG, ALT, and AST increased. Lipid abnormalities (TG, cholesterol, LDL-c, and HDL-c), oxidative stress markers (MDA, SOD, and GPx), and liver dysfunction were found. Low serum E2 and high T supported PCO. Decorin reduced model rat BMI, liver weight, coefficient, insulin resistance, TG, ALT, and AST. It reduced liver inflammation, improved liver extract lipids, and normalized MDA, SOD, and GPx. In the model group, decorin lowered serum T, E2, LH, caspase 3, and TNF-alpha. Decorating improved NAFLD/PCOS group liver histology and function. Decorin reduces hepatosteatosis by reducing liver inflammation, oxidative stress, and dyslipidemia.
Collapse
Affiliation(s)
- Hany A Elkattawy
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia.
| | - Amira Ebrahim Alsemeh
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Lashin Saad Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, P.O. Box 19328, Amman, Jordan; Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Mona Mostafa Ahmed
- Pathology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Asmaa Monir Eltaweel
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt; College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh 11481, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia.
| | - Farha M Shaikh
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia.
| | - Ahmed Behiry
- Department of Tropical Medicine and Endemic Diseases, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Ahmed El-Sayed Hassan
- Medical Physiology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt; Department of Basic Medical Sciences, College of Medicine, Sulaiman Al-Rajhi University, Bukayriah 51941, Saudi Arabia.
| | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Dalia Mahmoud Abdelmonem Elsherbini
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Sahar K Ali
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Madaniah Omar Zakari
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Medina, Saudi Arabia.
| | - Moaz Abdullah Mojaddidi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Medina, Saudi Arabia.
| | - Ehab Kamal Ali
- Department of Anatomy and Embryology, Faculty of Medicine, AL-Azhar University, New Damietta, Egypt.
| | - Yasser M Elbastawisy
- Department of Basic Medical Sciences, College of Medicine, Al-Rayan Colleges, Al-Madinah, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Shimaa Hadhoud
- Medical Physiology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| |
Collapse
|
398
|
Guan L, Zhu Y, Wang J. A Commentary on 'Steatotic Liver Disease in Younger Adults Is Associated With Altered Gut Microbiology'. Liver Int 2025; 45:e70070. [PMID: 40105377 DOI: 10.1111/liv.70070] [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: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Affiliation(s)
- Liying Guan
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Ying Zhu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Juntao Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
399
|
Ryu T, Chang Y, Yoo JJ, Lee SH, Jeong SW, Kim SG, Kim YS, Kim HS, Yang K, Jang JY. Glucosamine supplementation attenuates progression of metabolic dysfunction-associated steatotic liver disease and related comorbidities. Clin Nutr 2025; 47:119-128. [PMID: 40020645 DOI: 10.1016/j.clnu.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND & AIMS This study examines the impact of glucosamine on the progression and outcomes of metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction and alcohol-associated liver disease (MetALD) using a large scale cohort. METHODS Present study utilized inverse probability of treatment weighting (IPTW) to adjust for confounders in this cohort study. Participants were classified based on glucosamine use, and primary and secondary outcomes included all-cause mortality, liver cirrhosis, cardiovascular disease, cerebrovascular disease, and chronic kidney disease (CKD) incidences. Cox proportional hazards models were used to assess hazard ratios and 95 % confidence intervals. RESULTS We found that glucosamine significantly reduces all-cause mortality in MASLD and MetALD cohorts after IPTW adjustment (P < 0.001). Additionally, glucosamine use was associated with lower liver cirrhosis incidence in MASLD both before (P = 0.003) and after IPTW adjustment (P = 0.046). Glucosamine also decreased cardiovascular disease risk in MASLD (P < 0.001) and MetALD (P = 0.037) cohorts, though it showed no significant impact on cerebrovascular disease incidence. Furthermore, glucosamine use was associated with a significantly lower incidence of CKD in the MASLD cohort (P = 0.034) and the entire cohort (P = 0.030), but not in the No steatotic liver disease cohort or MetALD cohort. CONCLUSION The findings suggest that glucosamine could be a beneficial supplementary therapy for managing steatotic liver diseases, particularly for patients at high risk for cardiovascular and renal complications. Further clinical trials are required to validate these potential benefits.
Collapse
Affiliation(s)
- Tom Ryu
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| | - Young Chang
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| | - Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea
| | - Sae Hwan Lee
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Soung Won Jeong
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| | - Sang Gyune Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea
| | - Young Seok Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea
| | - Hong Soo Kim
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Keungmo Yang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.
| | - Jae Young Jang
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea.
| |
Collapse
|
400
|
Zhang J, Sun Z, Xu L, Wang Y, Wang Y, Dong B. Unraveling the link between metabolic dysfunction-associated steatotic liver disease and osteoporosis: a bridging function of gut microbiota. Front Endocrinol (Lausanne) 2025; 16:1543003. [PMID: 40235664 PMCID: PMC11997446 DOI: 10.3389/fendo.2025.1543003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
This review examines the strong association between metabolic dysfunction-associated steatotic liver disease (MASLD) and osteoporosis (OP), with a particular focus on the role of gut microbiota in linking these two disorders. Both MASLD and OP are closely linked to metabolic syndrome, and their pathogenesis involves multiple factors, such as inflammatory response, insulin resistance, altered intestinal permeability, and estrogen deficiency. Dysregulation of gut microbiota not only affects hepatic fat accumulation and bone metabolism disorders through metabolites, such as short-chain fatty acids, but also exacerbates systemic chronic inflammation by impairing the intestinal barrier function, thus accelerating the progression of both diseases. This article summarizes recent studies that highlight the central role of gut microbiota as a co-morbid factor in MASLD and OP, offering new perspectives for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhen Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|