1
|
Kalacun V, Ekart R, Bevc S, Skok P, Hojs R, Vodošek Hojs N. Oxidative stress and inflammation in hemodialysis: a comparison of patients with or without advanced nonalcoholic fatty liver disease (NAFLD). Ren Fail 2025; 47:2455523. [PMID: 39842820 PMCID: PMC11755732 DOI: 10.1080/0886022x.2025.2455523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and chronic kidney disease are global public health issues associated with high morbidity and mortality. Both diseases are also interlinked. Little is known about the meaning of NAFLD in hemodialysis (HD) patients. Therefore, the aim of our study was to investigate the difference in oxidative stress and inflammation in HD patients with or without advanced NAFLD. Seventy-seven HD patients were included (65.14 ± 12.34 years, 59.2% male) and divided according to abdominal ultrasound and two-dimensional shear wave elastography (2D-SWE) measurements into two groups: 1) no NAFLD or no advanced NAFLD (2D-SWE <9 kPa) and 2) advanced NAFLD (2D-SWE ≥9 kPa). Medical history data and blood results were collected. HD patients with advanced NAFLD had significantly higher levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG; p = 0.025), tumor necrosis factor-alpha (TNF-α; p = 0.023), and intercellular adhesion molecule 1 (ICAM-1; p = 0.015) in comparison to HD patients without advanced NAFLD. Interleukin 6 (IL-6) was higher in the advanced NAFLD group, but the difference was of borderline significance (p = 0.054). There was no significant difference in high-sensitivity C-reactive protein (hs-CRP), and vascular cell adhesion molecule 1 (VCAM-1) between groups. In binary logistic regression analysis, advanced NAFLD was significantly associated with 8-OHdG and ICAM-1. In conclusion, higher oxidative stress and inflammation levels are present in HD patients with advanced NAFLD.
Collapse
Affiliation(s)
- Vanja Kalacun
- Department of Gastroenterology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Sebastjan Bevc
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Pavel Skok
- Department of Gastroenterology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Radovan Hojs
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Nina Vodošek Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| |
Collapse
|
2
|
Saha T, Mehrotra S, Gupta P, Kumar A. Exosomal miRNA combined with anti-inflammatory hyaluronic acid-based 3D bioprinted hepatic patch promotes metabolic reprogramming in NAFLD-mediated fibrosis. Biomaterials 2025; 318:123140. [PMID: 39892017 DOI: 10.1016/j.biomaterials.2025.123140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex metabolic disorder, where the underlying molecular mechanisms are mostly not well-understood and therefore, warrants the need for therapeutic interventions targeting several metabolic pathways as a unified response. Of late, promising outcomes have been observed with mesenchymal stem cell-derived exosomes. However, reduced bioavailability due to systemic delivery and the need for repeated fresh isolation hinders their feasibility for clinical applications. In this regard, an 'off-the-shelf' 3D bioprinted hyaluronic acid-based hepatic patch to deliver encapsulated exosomes alone/or with hepatocytes (as dual-therapy) is developed as a holistic approach for ameliorating the disease condition and promoting tissue regeneration. The bioprinted hepatic patch demonstrated sustained and localized release of exosomes (∼82 % in 21 days), and healthy liver tissue-like mechanical properties while being biocompatible and biodegradable. Assessment in NAFLD rat models displayed alleviation of the altered biochemical parameters such as fat deposition, deranged liver functions, disrupted lipid, glucose, and insulin metabolism along with a reduction in localized inflammation, and associated liver fibrosis. The study suggests that a synergistic effect between the miRNA population of released exosomes, cell therapy, and the bioprinted matrix materials is crucial in targeting multiple complex metabolic pathways associated with the severity of the disease.
Collapse
Affiliation(s)
- Triya Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| | - Purva Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| |
Collapse
|
3
|
Marjot T. Positioning the liver at the centre of fructose-associated extrahepatic cancer. J Hepatol 2025; 82:1135-1137. [PMID: 40102073 DOI: 10.1016/j.jhep.2025.02.021] [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: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Affiliation(s)
- Thomas Marjot
- Oxford Centre for Diabetes Endocrinology and Metabolism (OCDEM), Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, UK; Translational Gastroenterology and Liver Unit (TGLU), Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, UK
| |
Collapse
|
4
|
Jönsson C, Ma'ayeh S, Zhang B, Kechagias S, Liljeblad M, Nasr P, Hansson SF, Ekstedt M. Vascular endothelial growth factor A, a potential non-invasive biomarker for metabolic dysfunction-associated steatotic liver disease progression. Clin Biochem 2025; 137:110920. [PMID: 40127834 DOI: 10.1016/j.clinbiochem.2025.110920] [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: 09/09/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
INTRODUCTION AND OBJECTIVES Liver fibrosis is the primary predictor of complications in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, there are currently no non-invasive prognostic tests to stratify patients at risk for hepatic fibrosis progression. This study aimed to explore whether plasma proteins could serve as non-invasive biomarkers for monitoring MASLD disease progression. MATERIALS AND METHODS Blood plasma protein analysis was performed on samples from a long-term follow-up study of patients with MASLD with repeated liver biopsies. Over 1100 proteins covering a broad range of biological processes were analyzed using 13 Olink® Target 96 panels. Protein level changes were compared between the different time points and between patients with or without an increase in the liver fibrosis stage between the two biopsies. RESULTS Increased vascular endothelial growth factor A (VEGFA) plasma levels were significantly associated with liver fibrosis progression in patients with a histologically assessed increase in the fibrosis stage. CONCLUSIONS These findings suggest that the plasma protein VEGFA may be an effective biomarker for monitoring fibrosis progression in patients with MASLD.
Collapse
Affiliation(s)
- Cecilia Jönsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| | | | | | - Stergios Kechagias
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| | - Mathias Liljeblad
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden.
| | - Patrik Nasr
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden; Wallenberg Centre for Molecular Medicine, Linköping University, Sweden.
| | - Sara F Hansson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden.
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| |
Collapse
|
5
|
Marjot T, Armstrong MJ, Stine JG. Skeletal muscle and MASLD: Mechanistic and clinical insights. Hepatol Commun 2025; 9:e0711. [PMID: 40408301 DOI: 10.1097/hc9.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 05/25/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is intrinsically linked with widespread metabolic perturbations, including within skeletal muscle. Indeed, MASLD is associated with a range of skeletal muscle abnormalities, including insulin resistance, myosteatosis, and sarcopenia, which all converge on the liver to drive disease progression and adverse patient outcomes. This review explores the mechanistic links between skeletal muscle and MASLD, including the role of abnormal glycemic control, systemic inflammation, and disordered myokine signaling. In turn, we discuss how intrinsic liver pathology can feed back to further exacerbate poor skeletal muscle health. Given the central importance of skeletal muscle in MASLD pathogenesis, it offers clinicians an opportunity to intervene for therapeutic benefit. We, therefore, summarize the role of nutrition and physical activity on skeletal muscle mass, quality, and metabolic function and discuss the knock-on effect this has on the liver. An awareness of these treatment strategies is particularly important in the era of effective pharmacological and surgical weight loss interventions, which can be associated with the development of sarcopenia. Finally, we highlight a number of promising drug agents in the clinical trial pipeline that specifically target skeletal muscle in an attempt to improve metabolic and physical functioning.
Collapse
Affiliation(s)
- Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, UK
- Translational Gastroenterology and Liver Unit (TGLU), Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Matthew J Armstrong
- Liver Unit, Queen Elizabeth University Hospital Birmingham, Birmingham, UK
- Birmingham NIHR Biomedical Research Centre, University of Birmingham, Birmingham, UK
| | - Jonathan G Stine
- Department of Medicine, Division of Gastroenterology and Hepatology, Penn State Health-Milton S. Hershey Medical Centre, Hershey, Pennsylvania, USA
| |
Collapse
|
6
|
Koutoukidis D, Jebb S, Tomlinson J, Mozes F, Pavlides M, Lacharie M, Saffioti F, Aveyard P, Cobbold J. Severe Dietary Energy Restriction for Compensated Cirrhosis Due to Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomised Controlled Trial. J Cachexia Sarcopenia Muscle 2025; 16:e13783. [PMID: 40275677 PMCID: PMC12022231 DOI: 10.1002/jcsm.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Compensated cirrhosis due to metabolic dysfunction-associated steatotic liver disease (CC-MASLD) increases morbidity and mortality risk but has no aetiology-specific treatment. We investigated the safety and efficacy signals of severe energy restriction. METHODS In this randomised controlled trial, adults with CC-MASLD and obesity in a tertiary hepatology centre were randomised 2:1 to receive one-to-one remote dietetic support with a low-energy (880 kcal/day, 80 g protein/day) total diet replacement programme for 12 weeks and stepped food reintroduction for another 12 weeks or standard of care (SoC). Given the exploratory nature of the study, three pre-defined co-primary outcomes were used to assess safety and efficacy signals: severe increases in liver biochemistry, changes in iron-corrected T1, and changes in liver stiffness on magnetic resonance elastography. Changes in liver steatosis on magnetic resonance imaging, physical performance based on the physical performance test and liver frailty index, and changes in fat-free mass were secondary outcomes. Magnetic resonance outcomes were assessed blind. RESULTS Between February 2022 and September 2023, 17 participants (36% female, median [IQR] age 58 [7.5] years) were randomised to SoC (n = 6) or intervention (n = 11). The trial stopped earlier than planned due to slow recruitment rate. 91% and 94% of participants completed the intervention and attended the 24-week follow-up, respectively. Compared with the SoC, the between-group weight change in the intervention was -11.9 kg (95% CI: -17.2, -6.6, p < 0.001) at 24 weeks. Liver biochemistry markers (alanine transaminase, aspartate transaminase, and total bilirubin) were stable in everyone throughout the trial. Iron-corrected T1 and steatosis significantly reduced (-149.9 ms [95% CI -258.1, -41.7, p = 0.01] and -6% [95% CI -11.3, -0.6, p = 0.03], respectively). There were no between-group differences in changes in liver stiffness (0.2 kPa [95% CI -1.1, 1.6]), the physical performance test (1.5 points [95% CI -1.9 to 4.9], p = 0.70) or the liver frailty index (0 [95% CI -0.6 to 0.6], p = 0.97). Compared with SoC, absolute fat-free mass reduced (-3.2 kg [95% CI -6 to -0.3], p = 0.04) but relative fat-free mass as percentage of total body weight increased (5.4% [95% CI 0.5 to 10.3], p = 0.046). No participant met the pre-defined safety criteria for enhanced observation or intervention discontinuation. There was no between-group differences in changes in cardiovascular markers and no evidence of hepatic decompensation or serious adverse events. CONCLUSIONS Severe energy restriction appears a safe option to achieve significant weight loss and reduce liver fat without adverse effects in people with CC-MASLD. A larger study is needed to confirm these findings. CLINICAL TRIAL REGISTRATION ISRCTN13053035, prospectively registered, overall study status: closed.
Collapse
Affiliation(s)
- Dimitrios A. Koutoukidis
- Nuffield Department of Primary Care Health SciencesUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreOxfordUK
| | - Susan A. Jebb
- Nuffield Department of Primary Care Health SciencesUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreOxfordUK
| | - Jeremy W. Tomlinson
- NIHR Oxford Biomedical Research CentreOxfordUK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Ferenc E. Mozes
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Michael Pavlides
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
- Department of Gastroenterology and HepatologyJohn Radcliffe Hospital, Oxford University Hospitals NHS Foundation TrustOxfordUK
| | - Miriam Lacharie
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Francesca Saffioti
- Department of Gastroenterology and HepatologyJohn Radcliffe Hospital, Oxford University Hospitals NHS Foundation TrustOxfordUK
| | - Paul Aveyard
- Nuffield Department of Primary Care Health SciencesUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreOxfordUK
| | - Jeremy F. Cobbold
- NIHR Oxford Biomedical Research CentreOxfordUK
- Department of Gastroenterology and HepatologyJohn Radcliffe Hospital, Oxford University Hospitals NHS Foundation TrustOxfordUK
| |
Collapse
|
7
|
de Jong VD, Alings M, Bruha R, Cortez-Pinto H, Dedoussis GV, Doukas M, Francque S, Fournier-Poizat C, Gastaldelli A, Hankemeier T, Holleboom AG, Miele L, Moreno C, Muris JWM, Ratziu V, Romero-Gomez M, Schattenberg JM, Serfaty L, Stefan DC, Tushuizen ME, Verheij J, Willemse J, Franco OH, Grobbee DE, Castro Cabezas M, GRIPonMASH consortium. Global research initiative for patient screening on MASH (GRIPonMASH) protocol: rationale and design of a prospective multicentre study. BMJ Open 2025; 15:e092731. [PMID: 40447415 PMCID: PMC12164639 DOI: 10.1136/bmjopen-2024-092731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 05/12/2025] [Indexed: 06/16/2025] Open
Abstract
INTRODUCTION The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) may be as high as 38% in the adult population with potential serious complications, multiple comorbidities and a high socioeconomic burden. However, there is a general lack of awareness and knowledge about MASLD and its progressive stages (metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis). Therefore, MASLD is still far underdiagnosed. The 'Global Research Initiative for Patient Screening on MASH' (GRIPonMASH) consortium focuses on this unmet public health need. GRIPonMASH will help (primary) healthcare providers to implement a patient care pathway, as recommended by multiple scientific societies, to identify patients at risk of severe MASLD and to raise awareness. Furthermore, GRIPonMASH will contribute to a better understanding of the pathophysiology of MASLD and improved identification of diagnostic and prognostic markers to detect individuals at risk. METHODS This is a prospective multicentre observational study in which 10 000 high-risk patients (type 2 diabetes mellitus, obesity, metabolic syndrome or hypertension) will be screened in 10 European countries using at least two non-invasive tests (Fibrosis-4 index and FibroScan). Blood samples and liver biopsy material will be collected and biobanked, and multiomics analyses will be conducted. ETHICS AND DISSEMINATION The study will be conducted in compliance with this protocol and applicable national and international regulatory requirements. The study initiation package is submitted at the local level. The study protocol has been approved by local medical ethical committees in all 10 participating countries. Results will be made public and published in scientific, peer-reviewed, international journals and at international conferences. REGISTRATION DETAILS NCT05651724, registration date: 15 Dec 2022.
Collapse
Affiliation(s)
- Vivian D de Jong
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Clinical BV, Zeist, The Netherlands
| | - Marco Alings
- Cardiology, Amphia Hospital, Breda, The Netherlands
| | - Radan Bruha
- General University Hospital and the First Faculty of Medicine, Charles University, Praha, Czech Republic
| | - Helena Cortez-Pinto
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - George V Dedoussis
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, Greece
| | - Michail Doukas
- Pathology, Erasmus MC Universitair Medisch Centrum Rotterdam, Rotterdam, Zuid-Holland, The Netherlands
| | - Sven Francque
- Division of Gastroenterology and Hepatology, University Hospital Antwerp, Edegem, Belgium
| | | | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Thomas Hankemeier
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, Zuid-Holland, The Netherlands
| | - Adriaan G Holleboom
- Department of Vascular Medicine and Internal Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Luca Miele
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Lazio, Italy
- Liver Transplant Unit and Steatotic Liver Disease Clinic, Digestive Disease Center CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Lazio, Italy
| | - Christophe Moreno
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hopital Erasme, Bruxelles, Belgium
| | - Jean W M Muris
- Family Medicine, Maastricht University, Maastricht, The Netherlands
| | - Vlad Ratziu
- ICAN - Institute for Cardiometabolism and Nutrition, Hôpital Pitié Salpêtrière, Sorbonne University, Paris, France
| | - Manuel Romero-Gomez
- UCM Digestive Diseases and SeLiver Group, Virgen del Rocio University Hospital, Institute of Biomedicine of Seville (HUVR/CSIC/US), University of Seville, Sevilla, Andalucía, Spain
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Hospital, Homburg, Saarland, Germany
- Saarland University, Saarbrucken, Germany
| | | | | | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | | | | | - Oscar H Franco
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Diederick E Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Clinical BV, Zeist, The Netherlands
| | - Manuel Castro Cabezas
- Julius Clinical BV, Zeist, The Netherlands
- Department of Internal Medicine, Franciscus Gasthuis en Vlietland, Rotterdam, Zuid-Holland, The Netherlands
- Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
8
|
Horn P, Tacke F. [Diagnostic and therapeutic management of MASLD]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025:10.1007/s00108-025-01896-3. [PMID: 40418344 DOI: 10.1007/s00108-025-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 05/27/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed fatty liver, is closely associated with insulin resistance, obesity and cardiometabolic risk factors. It is the most common cause of liver cirrhosis and hepatocellular carcinoma and is associated with a significantly increased risk of cardiovascular events. With a global prevalence of 25-35%, it represents a growing socioeconomic burden. The diagnosis of MASLD is based on imaging evidence of liver steatosis and clinical criteria of cardiometabolic risk. In the extended diagnostic workup, noninvasive procedures are increasingly replacing liver biopsy for the assessment of liver fibrosis and proper risk stratification. Therapeutic management is based on lifestyle interventions to reduce weight, with studies also demonstrating positive liver effects of glucagon-like peptide‑1 (GLP-1) agonists as well as bariatric surgery. Specific drugs for the treatment of steatohepatitis (MASH) are currently in the approval process.
Collapse
Affiliation(s)
- Paul Horn
- Med. Klinik m. S. Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte, Augustenburger Pl. 1, 13353, Berlin, Deutschland
| | - Frank Tacke
- Med. Klinik m. S. Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte, Augustenburger Pl. 1, 13353, Berlin, Deutschland.
| |
Collapse
|
9
|
De Nardo W, Lee O, Johari Y, Bayliss J, Pensa M, Miotto PM, Keenan SN, Ryan A, Rucinski A, Svinos TM, Ooi GJ, Brown WA, Kemp W, Roberts SK, Parker BL, Montgomery MK, Larance M, Burton PR, Watt MJ. Integrated liver-secreted and plasma proteomics identify a predictive model that stratifies MASH. Cell Rep Med 2025; 6:102085. [PMID: 40250425 DOI: 10.1016/j.xcrm.2025.102085] [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: 11/10/2024] [Revised: 01/30/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Obesity is a major risk factor for metabolic-associated steatotic liver disease (MASLD), which can progress to metabolic-associated steatohepatitis (MASH). There are no validated non-invasive tests to stratify persons with obesity with a greater risk for MASH. Herein, we assess plasma and liver from 266 obese individuals spanning the MASLD spectrum. Ninety-six human livers were precision-cut, and mass spectrometry-based proteomics identifies 3,333 proteins in the liver-secretion medium, of which 107 are differentially secreted in MASH compared with no pathology. The plasma proteome is markedly remodeled in MASH but is not different between patients with steatosis and no pathology. The APASHA model, comprising plasma apolipoprotein F (APOF), proprotein convertase subtilisin/kexin type 9 (PCSK9), afamin (AFM), S100 calcium-binding protein A6 (S100A6), HbA1c, and zinc-alpha-2-glycoprotein (AZGP1), stratifies MASH (area under receiver operating characteristic [AUROC] = 0.88). Our investigations detail the evolution of liver-secreted and plasma proteins with MASLD progression, providing a rich resource defining human liver-secreted proteins and creating a predictive model to stratify patients with obesity at risk of MASH.
Collapse
Affiliation(s)
- William De Nardo
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Olivia Lee
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yazmin Johari
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Bariatric Unit, Department of General Surgery, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Jacqueline Bayliss
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Marcus Pensa
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Paula M Miotto
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Stacey N Keenan
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrew Ryan
- TissuPath, Mount Waverley, VIC 3149, Australia
| | - Amber Rucinski
- Department of Oncology, Bendigo Health, Bendigo, VIC 3550, Australia
| | - Tessa M Svinos
- Department of General Surgery, Barwon Health, Geelong, VIC 3220, Australia
| | - Geraldine J Ooi
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Bariatric Unit, Department of General Surgery, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Wendy A Brown
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Bariatric Unit, Department of General Surgery, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - William Kemp
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, VIC 3181, Australia
| | - Stuart K Roberts
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, VIC 3181, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mark Larance
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Paul R Burton
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Bariatric Unit, Department of General Surgery, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
10
|
Miyake T, Furukawa S, Matsuura B, Yoshida O, Kanamoto A, Miyazaki M, Shiomi A, Nakaguchi H, Okazaki Y, Nakamura Y, Imai Y, Koizumi M, Watanabe T, Yamamoto Y, Koizumi Y, Tokumoto Y, Hirooka M, Kumagi T, Abe M, Hiasa Y. Association Between Serum Remnant Cholesterol Level and Metabolic Dysfunction-associated Steatotic Liver Histology. J Clin Endocrinol Metab 2025; 110:e2064-e2070. [PMID: 39193707 DOI: 10.1210/clinem/dgae597] [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: 03/06/2024] [Revised: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 08/29/2024]
Abstract
CONTEXT Estimated remnant cholesterol (Rem-C) level, a risk factor for cardiovascular disease, is associated with metabolic dysfunction-associated steatotic liver disease (MASLD) diagnosed via ultrasonography. However, the relationship between accurate serum Rem-C level measurements and histological findings of MASLD remains unclear. OBJECTIVE We aimed to elucidate the relationship between accurately measured serum Rem-C levels and histological findings of MASLD. DESIGN Cross-sectional single-center observational study. METHODS We assessed 222 patients (94 men and 128 women; age 20-80) who were diagnosed with MASLD via liver biopsy with available medical history, physical examination, and biochemical measurement data. Serum ester-type cholesterol and free cholesterol contents in the remnant lipoproteins were measured using an enzymatic method. RESULTS Serum Rem-C levels were significantly higher in patients with NAFLD activity score (NAS) 5 to 8, >66% steatosis grade, lobular inflammation with ≥5 foci, and many cells/prominent ballooning cells (a contiguous patch of hepatocytes showing prominent ballooning injury) than in patients with NAS 1 to 4, < 33% steatosis grade, lobular inflammation with <2 foci, and few ballooning cells (several scattered balloon cells), respectively. While univariate analysis revealed no significant association between Rem-C levels and advanced fibrosis, a significant association between Rem-C levels and NAS was evident. This relationship remained significant in multivariate analysis adjusted for confounders. Furthermore, in the analysis by sex, these relationships were significant for men but not for women. CONCLUSION High serum Rem-C levels were associated with high NAS but not with fibrosis stage, particularly in men. Controlling serum Rem-C level may improve MASLD activity.
Collapse
Affiliation(s)
- Teruki Miyake
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Shinya Furukawa
- Health Services Center, Ehime University, 3 Bunkyocho, Matsuyama City, Ehime 790-0826, Japan
| | - Bunzo Matsuura
- Department of Lifestyle-related Medicine and Endocrinology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Ayumi Kanamoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Masumi Miyazaki
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Akihito Shiomi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Hironobu Nakaguchi
- Department of Lifestyle-related Medicine and Endocrinology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yuki Okazaki
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yoshiko Nakamura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yusuke Imai
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Mitsuhito Koizumi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Takao Watanabe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yasunori Yamamoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yohei Koizumi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yoshio Tokumoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Teru Kumagi
- Postgraduate Medical Education Center, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon City, Ehime 791-0295, Japan
| |
Collapse
|
11
|
Wang K, Liu J. Anti-aging protein α-Klotho is potential for reducing comorbidity risk of cardiometabolic diseases in vulnerable populations and enhancing long-term prognosis. Sci Rep 2025; 15:16722. [PMID: 40369033 PMCID: PMC12078659 DOI: 10.1038/s41598-025-01580-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
This study investigated the impact of anti-aging protein α-Klotho on cardiometabolic diseases (CMDs) among middle-aged and elderly population. A total of 11,198 participants aged 40-79 years were included in the National Health and Nutrition Examination Survey (NHANES) spanning 2007-2016. Serum α-Klotho levels were quantified via enzyme-linked immunosorbent assays. CMDs comprised cardiovascular disease (CVD), and four metabolic disorders: type 2 diabetes (T2DM), obesity, chronic kidney disease (CKD), and non-alcoholic fatty liver disease (NAFLD). Weighted logistic regression analysis, subgroup analysis, mediation analysis, restricted cubic splines (RCS), and Cox proportional hazards regression analysis were used. α-Klotho exhibited negative associations with each single CMD except T2DM, and RCS showed U-shape and L-shape dose-response relationships of α-Klotho with risk of T2DM and CKD, respectively. Ordered logistic regression analysis revealed that higher levels of Klotho markedly reduced the cumulative number of metabolic comorbidities complicating CVD (OR 0.56 (0.35, 0.91)). Simple mediation analysis showed CKD may explain up to 20.42% of the association between Klotho and CVD. Notably, α-Klotho's association with cardiometabolic comorbidities was particularly evident among individuals who were widowed/divorced/separated, non-Hispanic Black, lower-income, or less educated, with hypertension, current smokers, lower leisure and commuting physical activity, but higher work-related physical activity. Regarding long-term effects, higher α-Klotho levels were associated with lower all-cause mortality among participants with CMDs, but not among those without CMDs. Higher α-Klotho levels were associated with lower CMD prevalence, particularly in high-risk cardiovascular populations with lower socioeconomic status and unfavorable lifestyles and reduced all-cause mortality risk among CMD patients.
Collapse
Affiliation(s)
- Kai Wang
- Medical School, Southeast University, Nanjing, China
| | - Jianing Liu
- Medical Faculty, Ulm University, Ulm, Germany.
| |
Collapse
|
12
|
Hedayati S, Doustmohammadian A, Jambarsang S, Maadi M, Zamani F, Hosseinzadeh M. Dietary lipophilic index and odds of metabolic dysfunction-associated steatotic liver disease (MASLD): A population-based study. Dig Liver Dis 2025:S1590-8658(25)00312-3. [PMID: 40368699 DOI: 10.1016/j.dld.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (MASLD) is one of the highly prevalent metabolic disorders worldwide The present study aimed to determine the association between a novel dietary lipophilic index (LI) with metabolic profile and MASLD in a population-based study in Amol, Iran. PATIENTS AND METHODS A cross-sectional study was conducted among 2979 Iranian adults within the framework of the Amol cohort study (AmolCS) Dietary assessments were performed using a validated 168-item food frequency questionnaire (FFQ) The dietary fatty acids were determined using the food composition table in Food Data Central of the USDA to indicate the lipophilic index Information about the melting point of fatty acids was obtained from the lipid bank database MASLD was defined as the ultrasound detection of hepatic steatosis that ruled out other causes of hepatic fat accumulation Then, dietary LI and lipophilic load (LL) were calculated using dietary fatty acid intake and melting point Multivariate MASLD The analysis was carried out for all participants stratified by sex and BMI Potential confounders were included in three different adjusted models. RESULTS The results revealed that dietary LI was associated with higher BMI and (WHtR), low physical activity, being female, living in urban residencies, and diabetes After adjustment for potential confounders, age, and energy intake, the odds ratio of MASLD in women was 1.33 (95 % CI: 1.05-1.99, p = 0.048) in the last tertile of dietary LI compared to the first tertile In adjustment by age and energy intake, chronic disease, smoking, physical activity, waist circumference, and residency women have a higher chance of MASLD in the second tertile of dietary LI (OR:1.38 95 % CI: 1.01-1.89) as well as in the third LI (OR:1.39, 95 % CI: 1.02-1.91) compared to the first tertile When the body mass index (BMI) was added to other confounders variables, the odds ratio of MASLD was 1.44 (95 % CI: 1.05-1.99) in the second tertile of LI and 1.41(95 % CI:1.02-1.95) in the third tertiles, Ptrend=0.04 In normal weight participants (BMI< 25), after adjustment for age and energy intake, the odds of MASLD were 86 % higher (CI; 1.07-3.25, Ptrend< 0.03) in the last tertile of LL compared to the first one. CONCLUSIONS This study found that higher levels of dietary fatty acids are associated with 40 % higher odds of MASLD in women Additionally, higher levels of fatty acids in normal-weight individuals were linked to an 86 % higher chance of MASLD It is highly recommended to reduce intake of saturated fatty acids and trans fatty acids, which are associated with a lower risk of MASLD.
Collapse
Affiliation(s)
- Safoora Hedayati
- International campus of shahidsadoughiUniversity of Medical Sciences, Yazd, Iran; Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azam Doustmohammadian
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Jambarsang
- Center for Healthcare Data Modeling, Departments of Biostatistics and Epidemiology, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mansooreh Maadi
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahdieh Hosseinzadeh
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
13
|
Yi H, Zhang Y, Zhou Z, Sun W, Wang Y, Tao W, Yu H, Yao L, Li J, Li L. Diagnostic Performance of Noninvasive Tests for Identifying Advanced Fibrosis in Metabolic Dysfunction-Associated Fatty Liver Disease With Mixed Etiologies. Endocr Pract 2025:S1530-891X(25)00140-5. [PMID: 40334939 DOI: 10.1016/j.eprac.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/10/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVES To assess the performance of fibrosis-4 index (FIB-4), nonalcoholic fatty liver disease fibrosis score (NFS) and aspartate aminotransferase to platelet ratio index (APRI) for advanced fibrosis in metabolic dysfunction-associated fatty liver disease (MAFLD) subgroups categorized by concomitant liver conditions. METHODS We conducted a multicentered study comprising inpatients with type 2 diabetes mellitus and MAFLD. Participants were categorized into 2 groups: MAFLD with pure metabolic etiologies (MAFLD-P) and MAFLD with mixed etiologies (MAFLD-M). Diagnostic performance of FIB-4, NFS, and APRI was assessed by area under the curve (AUC), sensitivity, and specificity. RESULTS This study comprised a total of 1475 participants, with a mean (SD) age of 58.4 (13) years and 835 (56.6%) males. FIB-4 and APRI had higher AUCs for advanced fibrosis in the MAFLD-M group than in the MAFLD-P group (MAFLD-M vs MAFLD-P: FIB-4 0.680 vs 0.591, P = .0442; APRI 0.723 vs 0.631, P = .0363). No significant difference was observed in the AUC of NFS between the 2 subgroups (MAFLD-M 0.572 vs MAFLD-P 0.617; P = .3237). Besides, the sensitivity of FIB-4 (69.6% vs 54.0%; P = .019) and APRI (43.5% vs 26.1%; P = .005) was higher in the MAFLD-M group. However, no significant difference in sensitivity of NFS and specificity of FIB-4, NFS, and APRI was observed between subgroups. CONCLUSIONS In this diagnostic study of the type 2 diabetes mellitus population, FIB-4 and APRI showed better performance for identifying advanced fibrosis in MAFLD with mixed etiologies.
Collapse
Affiliation(s)
- He Yi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Yan Zhang
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ziwei Zhou
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Yifan Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Wenxuan Tao
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Hekai Yu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Liqin Yao
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China.
| | - Jia Li
- Department of Ultrasound, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China.
| |
Collapse
|
14
|
Panzeri D, Laohawetwanit T, Akpinar R, De Carlo C, Belsito V, Terracciano L, Aghemo A, Pugliese N, Chirico G, Inverso D, Calderaro J, Sironi L, Di Tommaso L. Assessing the diagnostic accuracy of ChatGPT-4 in the histopathological evaluation of liver fibrosis in MASH. Hepatol Commun 2025; 9:e0695. [PMID: 40304570 PMCID: PMC12045550 DOI: 10.1097/hc9.0000000000000695] [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: 09/17/2024] [Accepted: 01/26/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Large language models like ChatGPT have demonstrated potential in medical image interpretation, but their efficacy in liver histopathological analysis remains largely unexplored. This study aims to assess ChatGPT-4-vision's diagnostic accuracy, compared to liver pathologists' performance, in evaluating liver fibrosis (stage) in metabolic dysfunction-associated steatohepatitis. METHODS Digitized Sirius Red-stained images for 59 metabolic dysfunction-associated steatohepatitis tissue biopsy specimens were evaluated by ChatGPT-4 and 4 pathologists using the NASH-CRN staging system. Fields of view at increasing magnification levels, extracted by a senior pathologist or randomly selected, were shown to ChatGPT-4, asking for fibrosis staging. The diagnostic accuracy of ChatGPT-4 was compared with pathologists' evaluations and correlated to the collagen proportionate area for additional insights. All cases were further analyzed by an in-context learning approach, where the model learns from exemplary images provided during prompting. RESULTS ChatGPT-4's diagnostic accuracy was 81% when using images selected by a pathologist, while it decreased to 54% with randomly cropped fields of view. By employing an in-context learning approach, the accuracy increased to 88% and 77% for selected and random fields of view, respectively. This method enabled the model to fully and correctly identify the tissue structures characteristic of F4 stages, previously misclassified. The study also highlighted a moderate to strong correlation between ChatGPT-4's fibrosis staging and collagen proportionate area. CONCLUSIONS ChatGPT-4 showed remarkable results with a diagnostic accuracy overlapping those of expert liver pathologists. The in-context learning analysis, applied here for the first time to assess fibrosis deposition in metabolic dysfunction-associated steatohepatitis samples, was crucial in accurately identifying the key features of F4 cases, critical for early therapeutic decision-making. These findings suggest the potential for integrating large language models as supportive tools in diagnostic pathology.
Collapse
Affiliation(s)
- Davide Panzeri
- Department of Physics, University of Milano-Bicocca, Milan, Italy
| | - Thiyaphat Laohawetwanit
- Division of Pathology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Reha Akpinar
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Camilla De Carlo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Vincenzo Belsito
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Luigi Terracciano
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alessio Aghemo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Nicola Pugliese
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Chirico
- Department of Physics, University of Milano-Bicocca, Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Julien Calderaro
- Team «Viruses, Hepatology, Cancer», Institut Mondor de Recherche Biomédicale, INSERM U955, Hôpital, Henri Mondor (AP-HP), Université Paris-Est, Créteil, France
- Department of Pathology, AP-HP, Henri Mondor University Hospital, Créteil, France
| | - Laura Sironi
- Department of Physics, University of Milano-Bicocca, Milan, Italy
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
15
|
Capinha F, Carvalhana S, Cortez-Pinto H. Role of Alcohol in Steatotic Liver Disease: Impact on Patients with Cardiometabolic Risk Factors. Dig Dis Sci 2025; 70:1746-1756. [PMID: 40025309 DOI: 10.1007/s10620-025-08912-4] [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: 07/23/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
The new definition of steatotic liver disease (SLD), as a broader concept, was a step forward in the increasing recognition of the substantial overlap between alcohol and cardiometabolic risk factors (CMRFs), in a continuum way. The spectrum of pathophysiological aspects, ranging from liver steatosis to fibrosis, has similarities in MASLD and ALD. Also, there is now considerable evidence that the association of metabolic dysfunction with increased alcohol consumption impacts on the risk of severe liver disease and prognosis. The new MetALD class, as recently proposed, shows clear differences in prognosis when comparing with MASLD and ALD groups. However, there is room for improvement, such as considering the role of previous alcohol intake, fluctuations of consumption over time, including binge drinking, refinement of alcohol assessment, and better understanding of the role of biomarkers. In summary, SLD is no doubt a significant improvement, but the new classification needs to be dynamic and adapting to patients needing frequent reassessment. Furthermore, it brings opportunities for research on the interaction between alcohol consumption and CMRFs.
Collapse
Affiliation(s)
- Francisco Capinha
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal
| | - Sofia Carvalhana
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal
| | - Helena Cortez-Pinto
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal.
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal.
| |
Collapse
|
16
|
Choque Vargas C, Cáceres F, Landeira G, Perez S, Marchi L, Ruffillo G, Tevez S, Puga-Tejada M, Fassio E. Cardiovascular events and incident diabetes in 220 patients with MASLD according to basal liver fibrosis: a 10-year follow-up historic cohort. Eur J Gastroenterol Hepatol 2025; 37:660-667. [PMID: 39975992 DOI: 10.1097/meg.0000000000002943] [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] [Indexed: 02/21/2025]
Abstract
AIM The aim of this study is to analyze association between liver fibrosis with CVE, incident diabetes, and cirrhosis complications. METHODS Historic cohort of biopsy-proven MASLD patients, divided into two groups: F0-F2 vs F3-F4 fibrosis. Baseline data included metabolic traits and liver function tests. Patients were contacted and scheduled for laboratory analysis and elastography. Endpoints were (a) CVE, defined as any of acute myocardial infarction, coronary stenting, ischemic cardiopathy, and stroke; (b) incident diabetes; (c) cirrhosis complications. Baseline data were collected at the time of liver biopsy, while follow-up data were recovered through personal interview or medical records. A stepwise logistic regression determined predictive variables for each endpoint. RESULTS Study population included 220 patients with median age 53 years, and 145 were women; baseline fibrosis was F0-F2 in 165 patients and F3-F4 in 55 patients; median follow-up was 9.9 years. A higher percentage of F3-F4 patients had CVE (29.4%) than F0-F2 ones (13.1%) (hazard ratio 2.42; 95% CI: 1.26-4.6; P = 0.008). Incident diabetes occurred in 53.3% of F3-F4 and 20.2% of F0-F2 cohort (hazard ratio 3.04; 95% CI: 1.99-4.86; P < 0.001); cirrhosis complications occurred in 9/55 F3-F4 patients and in 1/165 F0-F2 ones (hazard ratio 26.3; 95% CI: 3.3-208.3; P = 0.002). Multivariate analysis confirmed liver fibrosis as an independent predictor of incident diabetes and cirrhosis complications. CVE were associated with baseline diabetes and aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio. CONCLUSION In a cohort of 220 MASLD patients followed for 9.9 years, baseline F3-F4 was associated with incident diabetes and cirrhosis complications. AST/ALT ratio and diabetes were associated with CVE.
Collapse
Affiliation(s)
- Cinthia Choque Vargas
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Francisco Cáceres
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Graciela Landeira
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Soledad Perez
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Laura Marchi
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Gabriela Ruffillo
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Silvina Tevez
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Miguel Puga-Tejada
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
- División de Investigación Médica & Bioestadística, Instituto Ecuatoriano de Enfermedades Digestivas, Guayaquil, Ecuador
| | - Eduardo Fassio
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| |
Collapse
|
17
|
Sun DQ, Shen JQ, Tong XF, Ren YY, Yuan HY, Li YY, Wang XL, Chen SD, Zhu PW, Wang XD, Byrne CD, Targher G, Wei L, Wong VW, Tai D, Sanyal AJ, You H, Zheng MH. Liver fibrosis progression analyzed with AI predicts renal decline. JHEP Rep 2025; 7:101358. [PMID: 40321195 PMCID: PMC12048807 DOI: 10.1016/j.jhepr.2025.101358] [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: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 05/03/2025] Open
Abstract
Background & Aims The relationship between biopsy-proven liver fibrosis progression and renal function decline in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) has not been fully elucidated. We used an automated quantitative liver fibrosis assessment (qFibrosis) technique to investigate the temporal changes in regional liver fibrosis. Methods This retrospective longitudinal study included 68 MASLD patients and their paired formalin-fixed sections of liver biopsies. One hundred eighty-four fibrosis parameters were quantified in five different hepatic regions, including portal tract, peri-portal, zone 2, peri-central and central vein regions, and qFibrosis continuous values were calculated for all samples based on 10 fibrosis parameters using qFibrosis assessment. Liver fibrosis progression (QLF+, n = 18) and regression (QLF-, n = 23) was defined as at least a 20% relative change in qFibrosis over a 23-month follow-up. Renal function decline was assessed by estimated glomerular filtration rate (eGFR) changes. Results The eGFR decline was greater in the QLF+ group (106.53 ± 13.71 ml/min/1.73 m2 vs. 105.28 ± 12.46 ml/min/1.73 m2) than in the QLF- group (110.87 ± 14.58 ml/min/1.73 m2 vs. 114.18 ± 14.81 ml/min/1.73 m2). In addition, liver fibrosis changes in the central vein and pericentral regions were more strongly associated with eGFR decline than in periportal, zone 2 and portal tract regions. We combined these parameters to construct a prediction model, which better differentiated eGFR decline (a cut-off value of qFibrosis combined index = 0.52, p <0.001). Conclusions A decline in renal function is significantly related to liver fibrosis progression in MASLD. Regional qFibrosis assessment may efficiently predict eGFR decline, thus highlighting the importance of assessing renal function in patients with MASLD with worsening liver fibrosis. Impact and implications The study shows that liver fibrosis progression assessed by qFibrosis may be associated with renal function decline, which provides a new perspective for understanding complex pathological processes. A combination of artificial intelligence and digital pathology may earlier and more precisely quantify the progression of regional liver fibrosis, thus better identifying changes in renal function. This opens the possibility of early interventions, which are essential to improve patients' outcomes.
Collapse
Affiliation(s)
- Dan-Qin Sun
- Department of Nephrology, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Wuxi No. 2 People's Hospital, Wuxi, China
| | - Jia-Qi Shen
- Department of Nephrology, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Wuxi No. 2 People's Hospital, Wuxi, China
| | - Xiao-Fei Tong
- Liver Research Center, Beijing Friendship Hospital, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Capital Medical University, Beijing, China
| | | | - Hai-Yang Yuan
- 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
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Yang-Yang Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Sui-Dan Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pei-Wu Zhu
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Dong Wang
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - 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, UK
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Vincent W.S. Wong
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | | | - Arun J. Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Capital Medical University, Beijing, China
| | - 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
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Safarova MS, Weintraub S, Sadaniantz K, Kovell L, Warden BA, Garshick MS, Duell PB, Gianos E. Statin Use in Special Populations for the Prevention of Cardiovascular Disease in Adults. Curr Atheroscler Rep 2025; 27:54. [PMID: 40310600 DOI: 10.1007/s11883-025-01298-8] [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] [Accepted: 04/06/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE OF REVIEW Outcome benefits for HMG-CoA reductase inhibitor (statin) use in the prevention of atherosclerotic cardiovascular disease (ASCVD) are well established and yet, statins remain underutilized with only half of eligible individuals receiving them among certain vulnerable populations. This review critically examines available data to provide a summary of the current evidence for statin use in select populations. RECENT FINDINGS Lipid management can be more complex in patients with chronic kidney disease (CKD), organ transplants, metabolic dysfunction associated with steatotic liver disease (MASLD), and human immunodeficiency virus (HIV). Statins are generally safe and effective to reduce the burden of ASCVD among these highly heterogeneous groups of patients and should be considered with careful attention to their concomitant disease state. Herein, we focus on appropriate statin use in these challenging to treat conditions, their relationship with increased ASCVD risk, and approaches to statin use for ASCVD risk reduction. Although further research is needed to define optimal therapy in select high risk groups for ASCVD prevention, statins are proven to be clinically efficacious, safe, and cost-effective for ASCVD prevention, warranting greater efforts to increase their use.
Collapse
Affiliation(s)
- Maya S Safarova
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Spencer Weintraub
- Northwell Cardiovascular Institute, North Shore University Hospital, Manhasset, NY, USA
| | - Katherine Sadaniantz
- Department of Medicine, Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Lara Kovell
- Department of Medicine, Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Bruce A Warden
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Michael S Garshick
- Division of Cardiology, Department of Medicine and Department of Dermatology, NYU Langone Health, New York, NY, USA
| | - P Barton Duell
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Endocrinology, Diabetes, and Clinical Nutrition, Oregon Health & Science University, Portland, OR, USA
| | - Eugenia Gianos
- Northwell Cardiovascular Institute, Lenox Hill Hospital, New York, NY, USA
| |
Collapse
|
19
|
Khalil SM, de Souza MHG, de Oliveira FD, Sato EDBDS, Meine GC. Efficacy and safety of aldafermin for the treatment of metabolic dysfunction-associated steatohepatitis: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 2025; 49:102579. [PMID: 40147589 DOI: 10.1016/j.clinre.2025.102579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND We aimed to assess the efficacy and safety of Aldafermin in treating patients with biopsy-confirmed metabolic dysfunction-associated steatohepatitis (MASH). METHODS We searched PubMed, Embase, and Cochrane Library for randomized controlled trials (RCTs) comparing Aldafermin to placebo for treating patients with MASH up to December 8, 2024. The risk ratios (RR) with 95 % confidence intervals (CI) were pooled for binary outcomes using a random-effects model. Additionally, we conducted subgroup analysis by fibrosis stage and Aldafermin dosage, and meta-regression analysis assuming the dosage of Aldafermin as a covariate. RESULTS We included 4 RCTs, encompassing 491 patients. Compared to placebo, Aldafermin had a higher probability of MASH resolution without worsening of fibrosis (RR 3.04; 95 %CI 1.12-8.28), composite of fibrosis improvement and MASH resolution (RR 5.86; 95 %CI 1.15-29.94), and reduction ≥30 % in hepatic fat fraction by MRI-PDFF (RR 3.14; 95 %CI 1.44-6.85). There were no significant differences in fibrosis improvement ≥1 stage without worsening of MASH (RR 1.48; 95 %CI 0.93-2.35), and overall AEs (RR 1.02; 95 %CI 0.95-1.11) between the groups. Subgroup analysis by fibrosis stage and Aldafermin dosage showed consistent results, and meta-regression analysis by dosage showed a dose-dependent improvement for the outcome of ≥30 % reduction in hepatic fat fraction by MRI-PDFF. CONCLUSION In conclusion, Aldafermin improved MASH resolution without worsening fibrosis, enhanced the composite of fibrosis improvement and MASH resolution, reduced hepatic fat fraction by MRI-PDFF, and was safe for treating patients with biopsy-confirmed MASH compared to placebo.
Collapse
Affiliation(s)
| | | | | | | | - Gilmara Coelho Meine
- Division of Gastroenterology, Internal Medicine Department, Feevale University, Novo Hamburgo, Brazil.
| |
Collapse
|
20
|
Liu XR, Yin SC, Chen YT, Lee MH. Metabolic dysfunction-associated steatotic liver disease and its associated health risks. J Chin Med Assoc 2025; 88:343-351. [PMID: 40128159 DOI: 10.1097/jcma.0000000000001230] [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] [Indexed: 03/26/2025] Open
Abstract
This article synthesizes the current knowledge on the epidemiology of metabolic dysfunction-associated steatotic liver disease (MASLD), its associated risks, and its genetic determinants. The findings presented in this article can be used to develop clinical strategies to reduce MASLD's growing global burden. MASLD has become a major global health concern due to increasing rates of obesity, sedentary lifestyles, and metabolic disorders. MASLD is a leading cause of end-stage liver diseases, including cirrhosis and hepatocellular carcinoma (HCC), and MASLD also significantly increases the risk of cardiovascular disease (CVD), thereby exerting dual effects on liver and cardiovascular health. MASLD was once referred to as nonalcoholic fatty liver disease, and this change in nomenclature reflects a growing focus on its metabolic underpinnings, facilitating the more precise diagnosis and clinical management of this disease. Epidemiological studies have demonstrated that the prevalence of MASLD is increasing worldwide, although the prevalence varies across regions and populations. Noninvasive diagnostic tools such as ultrasound and fatty liver indices along with biomarkers such as alanine aminotransferase (ALT) are crucial for early detection and risk stratification. Genetic research has identified key gene variants, including PNPLA3 (rs738409) and TM6SF2 (rs58542926), that influence MASLD susceptibility and progression, and these findings have created opportunities for improving precision medicine with respect to treating MASLD. Research has revealed an association between MASLD and major adverse cardiovascular events and increased mortality, which highlights the importance of integrating cardiovascular risk management into treatment strategies for MASLD. Future research should focus on advancing noninvasive diagnostics, leveraging genetic insights to provide tailored care, and implementing population-specific interventions to address regional variations.
Collapse
Affiliation(s)
- Xia-Rong Liu
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Szu-Ching Yin
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Yi-Ting Chen
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Mei-Hsuan Lee
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
21
|
Zhang Z, Zhang W, Liu Z, Ou J, Sun Y, Zhang L, Ji G. Association between systemic inflammation markers and cardiovascular mortality in adults with metabolic dysfunction-associated steatotic liver disease. Nutr Metab Cardiovasc Dis 2025; 35:103781. [PMID: 39643477 DOI: 10.1016/j.numecd.2024.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/20/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIM Identifying metabolic dysfunction-associated steatotic liver disease (MASLD) patients at increased risk of cardiovascular mortality remains an unmet clinical need. We investigated the ability of four systemic inflammation markers to identify cardiovascular mortality risk in MASLD patients. METHODS AND RESULTS This cohort study included 4787 MASLD patients from the National Health and Nutrition Examination Survey (NHANES) from 2005 through 2018. The weighted Cox proportional hazards model was used to assess the relationship between four systemic indicators of inflammation and cardiovascular mortality. During a median (IQR) follow-up of 7.0 (3.8-10.3) years, 567 all-cause mortality and 174 cardiovascular mortality were recorded. Compared to the first quartile of systemic inflammation levels, the HRs of cardiovascular mortality in the fourth quartile were 3.22 (95 % CI 1.83-5.66) for SII, 2.74 (95 % CI 1.32-5.69) for SIRI, 3.69 (95 % CI 1.87-7.28) for NLR, and 1.83 (95 % CI 1.05-3.20) for PLR. For predicting 10-year cardiovascular mortality, SIRI (AUC = 0.70) and NLR (AUC = 0.69) were superior to SII (AUC = 0.60) and PLR (AUC = 0.52). Stratification of MASLD patients based on the optimal cutoff values revealed an HR of 2.67 (95 % CI 1.65-4.32) for cardiovascular mortality with SIRI > 1.23, and an HR of 2.39 (95 % CI 1.51-3.79) with NLR > 2.18. Combining systemic inflammation markers with the Fibrosis-4 Score can provide more accurate prognostic information for MASLD patients. CONCLUSIONS SIRI and NLR outperformed SII and PLR in predicting the risk of cardiovascular mortality, proving to be useful tools for risk stratification in MASLD patients.
Collapse
Affiliation(s)
- Ziqi Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weijie Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhidong Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiayi Ou
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yunhong Sun
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China.
| |
Collapse
|
22
|
Mohanty A, Austad K, Bosch NA, Long MT, Nolen-Doerr E, Walkey AJ, Drainoni ML, Rizo I, Fantasia KL. Assessing Clinician Engagement with a Passive Clinical Decision Support System for Liver Fibrosis Risk Stratification in a Weight Management Clinic. Endocr Pract 2025:S1530-891X(25)00133-8. [PMID: 40288606 DOI: 10.1016/j.eprac.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is common in obesity. Guidelines recommend liver fibrosis risk stratification with tools such as Fibrosis-4 (FIB-4) index, liver stiffness measurement with vibration-controlled transient elastography (VCTE) and/or hepatology referral for elevated FIB-4. Despite recommendations, implementation remains limited. Using mixed methods, we evaluated a three-strategy implementation bundle to improve fibrosis risk stratification-a FIB-4-based electronic health record embedded clinical decision support system (CDSS), educational outreach, and internal facilitation in a weight management clinic. METHODS The primary outcome was penetration: the proportion of patients with elevated FIB-4 completing VCTE or hepatology referral. We compared rates, pre and post activation of implementation bundle using Fischer's exact test. Semi-structured provider interviews, guided by the i-PARIHS framework, assessed acceptability and feasibility three months post-implementation. RESULTS In the pre-activation phase, 880/3933 (22.4%) weight management visits had the necessary labs to calculate automated FIB-4 scores with 128 elevated scores. In the post-activation phase, 2513/4,634 weight management visits (54.2%) had automated FIB-4 scores; with 234 elevated score. Pre-activation, there were no VCTE and 2 hepatology referrals. Post-activation, there were 3 VCTE referrals and 2 hepatology referrals (Fischer's exact test p-value=1.00). Providers cited shared responsibility with primary care, low awareness and trust in risk-stratification tools, workflow challenges, and competing demands as barriers. Educational outreach and facilitation improved CDSS engagement, while technical issues reduced it. CONCLUSION This implementation strategy bundle did not achieve meaningful MASLD fibrosis risk stratification. EHR based CDSS shows promise but requires alignment with provider priorities, seamless workflow integration, and robust technical infrastructure.
Collapse
Affiliation(s)
- Arpan Mohanty
- Section of Gastroenterology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts.
| | - Kirsten Austad
- Department of Family Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts; Evans Center for Implementation and Improvement Sciences (CIIS), Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Nicholas A Bosch
- Evans Center for Implementation and Improvement Sciences (CIIS), Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Section of Pulmonary, Allergy, Critical Care and Sleep, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Michelle T Long
- Medical & Science, Clinical Drug Development, Novo Nordisk A/S, Vandtårnsvej, Søborg, Denmark
| | - Eric Nolen-Doerr
- Section of Endocrinology Diabetes and Nutrition, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Allan J Walkey
- Division of Health Systems Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Mari-Lynn Drainoni
- Evans Center for Implementation and Improvement Sciences (CIIS), Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Section of Infectious Diseases, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Department of Health Law Policy and Management, Boston University School of Public Health, Boston, Massachusetts
| | - Ivania Rizo
- Section of Endocrinology Diabetes and Nutrition, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Kathryn L Fantasia
- Evans Center for Implementation and Improvement Sciences (CIIS), Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Section of Endocrinology Diabetes and Nutrition, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| |
Collapse
|
23
|
Liu M, Jiang L, Yang J, Yao Y, Puyang X, Ge X, Lu J, Zhang L, Yan Y, Shen H, Song C. Development and Validation of a Machine Learning-based Model for Prediction of Liver Fibrosis and MASH. J Clin Gastroenterol 2025:00004836-990000000-00441. [PMID: 40299904 DOI: 10.1097/mcg.0000000000002166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/09/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND AND AIM The development of accurate noninvasive tests to identify individuals with metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis is of great clinical importance. In this study, we aimed to develop 2 noninvasive diagnostic models on the basis of routine clinical and laboratory data, using machine learning, to identify patients with MASH and significant fibrosis (fibrosis stages 2 to 4), respectively. METHODS This analysis included the training (n=456) and the validation (n=105) sets of patients who underwent liver biopsy and laboratory testing for liver disease at 2 hospitals in China. Logistic regression, random forest, support vector machine, and the XGBoost algorithm were used to construct models, respectively. The best diagnostic models for MASH and significant fibrosis were compared with 7 existing noninvasive scoring systems including AAR, AST to platelet ratio index (APRI), BARD score, fibrosis-4 (FIB-4), fibrotic non-alcoholic steatohepatitis (NASH) index (FNI), homeostatic model assessment of insulin resistance (HOMA-IR), and non-alcoholic fatty liver disease fibrosis score (NFS). Performance was estimated by the area under the receiver operating characteristic curve (AUROC). RESULTS The final noninvasive diagnostic model integrated 19 indicators derived from routine clinical and laboratory tests. The XGBoost models exhibited superior performance in MASH and significant fibrosis with an improved AUROC value (MASH, 0.670, 95% CI 0.530-0.811; significant fibrosis, 0.713, 95% CI 0.611-0.815) compared with other noninvasive scoring systems in the validation set. CONCLUSIONS Utilizing machine learning can assist in diagnosing MASH and significant fibrosis based on clinical epidemiological information with good diagnostic performance.
Collapse
Affiliation(s)
| | | | | | - Yao Yao
- School of Public Health, Nanjing Medical University
| | | | | | - Jing Lu
- Department of Epidemiology
- Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | - Ci Song
- Department of Epidemiology
- Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Jiao X, Lai L, Qian Y, Sun B, Yang W. Identification of MEOX1 as a potential target in metabolic dysfunction-associated steatohepatitis-related liver fibrosis. Int J Biol Markers 2025:3936155251335975. [PMID: 40270091 DOI: 10.1177/03936155251335975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe mechanisms underlying the occurrence and progression of metabolic dysfunction-associated steatohepatitis (MASH)-related liver fibrosis remains poorly understood. This study aims to identify key transcription factors involved in the development of liver fibrosis in MASH patients, thereby providing potential targets for drug discovery.MethodsMicroarray data were retrieved from liver biopsy specimens of MASH patients exhibiting varying stages of fibrosis via the Gene Expression Omnibus database. Differentially expressed transcription factors (DETFs) were identified through the application of Weighted Gene Co-expression Network Analysis. A set of in vitro and in vivo experiments were conducted to investigate the role of MEOX1 in MASH-related fibrosis. To delineate the potential mechanisms, the transcriptomic RNA sequencing (RNA-seq), Alphafold, and PyMOL were used.ResultsA total of six DETFs (MEOX1, SOX4, LEF1, SOX9, MYC, and CBX2) were identified as being positively correlated with the progression of MASH-related fibrosis. MEOX1 was increased in mouse model of MASH diet-induced liver fibrosis and hepatic stellate cells (HSCs) stimulated by transforming growth factor-β1. Knockdown of the MEOX1 markedly suppressed the activation, proliferation, and migration of HSCs. RNA-Seq analysis identified serine protease inhibitor family E member 1 (SERPINE1) as the critical target of MEOX1 within HSCs. The protein interaction sites of MEOX1 and SERPINE1 were predicted using Alphafold and PyMOL.ConclusionIn summary, as a pivotal transcription factor, MEOX1 activates HSCs via SERPINE1, thereby promoting liver fibrosis associated with MASH. Inhibition of the MEOX1-SERPINE1 pathway could offer a novel therapeutic avenue for treating MASH-related fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| |
Collapse
|
25
|
Fulda ES, Portas L, Harper C, Preiss D, Bennett D, Doherty A. Association of Daily Steps with Incident Non-Alcoholic Fatty Liver Disease: Evidence from the UK Biobank Cohort. Med Sci Sports Exerc 2025:00005768-990000000-00789. [PMID: 40279651 PMCID: PMC7617666 DOI: 10.1249/mss.0000000000003738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
PURPOSE Low physical activity has been shown to be associated with higher risk of non-alcoholic fatty liver disease (NAFLD). However, the strength and shape of this association are currently uncertain due to a reliance on self-reported physical activity measures. This report aims to investigate the relationship of median daily step count with NAFLD using accelerometer-derived step count from a large prospective cohort study. METHODS The wrist-worn accelerometer sub-study of the UK Biobank (N = ~100,000) was used to characterise median daily step count over a seven-day period. NAFLD cases were ascertained via record linkage with hospital inpatient data and death registers or by using a measure of liver fat from imaging. Cox proportional hazards models were employed to assess the association between step count and NAFLD, adjusting for age, sociodemographic, and lifestyle factors. Mediation analyses were conducted. RESULTS Among 91,031 participants (709,440 person-years of follow-up), there were 762 incident NAFLD cases. Higher step count was log-linearly and inversely associated with risk of NAFLD. A 1000-step increase (representing 10 minutes of walking) was associated with a 12% (95% CI: 10%-14%) lower hazard of NAFLD. When using imaging to identify NAFLD, a 1,000-step increase was associated with a 6% (95% CI: 6%-7%) lower risk. There was evidence for mediation by adiposity, accounting for 39% of the observed association. CONCLUSIONS Daily step count, a modifiable risk factor, is log-linearly and inversely associated with NAFLD. This association was only partially explained by adiposity. These findings from a large cohort study may have important implications for strategies to lower NAFLD risk.
Collapse
Affiliation(s)
- Evelynne S. Fulda
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Laura Portas
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Charlie Harper
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - David Preiss
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| | - Aiden Doherty
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UNITED KINGDOM
| |
Collapse
|
26
|
Denimal D, Ponnaiah M, Phan F, Jeannin AC, Redheuil A, Salem JE, Boussouar S, Paulstephenraj P, Laroche S, Amouyal C, Hartemann A, Foufelle F, Bourron O. Metabolic dysfunction-associated steatotic liver disease (MASLD) biomarkers and progression of lower limb arterial calcification in patients with type 2 diabetes: a prospective cohort study. Cardiovasc Diabetol 2025; 24:176. [PMID: 40269920 PMCID: PMC12020187 DOI: 10.1186/s12933-025-02705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Studies have demonstrated that both lower limb arterial calcification and metabolic dysfunction-associated steatotic liver disease (MASLD) are linked to the development of peripheral artery disease. However, the potential relationship between MASLD biomarkers and progression of lower limb arterial calcification in individuals with type 2 diabetes (T2D) remains unclear. This study aimed to investigate whether the biomarkers of MASLD included in the FibroMax® panels are associated with the progression of lower limb arterial calcification in patients with T2D. METHODS The lower limb arterial calcification score (LLACS) was evaluated through computed tomography at baseline and after an average follow-up of 31.2 ± 3.7 months in a cohort of 150 patients with T2D. We also measured the serum biomarkers included in the FibroMax® panels (SteatoTest®, FibroTest®, NashTest®, ActiTest®). The predictive ability of these biomarkers of MASLD on LLACS progression was assessed through univariate and multivariate linear regression models, principal component regression analysis, as well as machine learning algorithms. RESULTS During the follow-up period, LLACS increased in 127 (85%) of the 150 patients with T2D. In univariate analysis, the annualized change in LLACS was positively and mainly correlated with baseline LLACS (r = 0.860, p < 0.0001), the FibroTest® score (r = 0.304, p = 0.0002), and age (r = 0.275, p = 0.0006), and negatively correlated with glomerular filtration rate (r = - 0.242, p = 0.003). In multivariate analysis, the FibroTest® score remained independently associated with the annualized change in LLACS, after adjusting for baseline LLACS and risk factors for lower extremity artery disease (β coefficient [95% confidence interval]: 988 [284-1692], p = 0.006). This association persisted even after adjustment for variables selected by principal component analysis (β = 1029 [289-1768], p = 0.007). Two advanced machine learning models identified the FibroTest® score as the second most important predictor of annualized change in LLACS, following baseline LLACS. CONCLUSIONS This study represents the first demonstration of an independent relationship between a non-invasive liver fibrosis test and the progression of lower limb arterial calcification in patients with T2D. Beyond its utility in assessing liver fibrosis, the FibroTest® could be a valuable and easy-to-use biomarker for predicting the risk of worsening lower limb arterial calcification. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT02431234.
Collapse
Affiliation(s)
- Damien Denimal
- INSERM U1231, Center for Translational and Molecular Medicine, Dijon, France.
- Department of Clinical Biochemistry, CHU Dijon-Bourgogne, Dijon Bourgogne University Hospital, 2 rue Ducoudray, 21000, Dijon, France.
| | | | - Franck Phan
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
- INSERM UMR_S 1166, Sorbonne University, Team Metabolic Diseases, Diabetes and Co-Morbidities, Paris, France
| | - Anne-Caroline Jeannin
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, Paris, France
- ICT Cardiovascular and Thoracic Imaging Unit, AP-HP, Pitié Salpêtrière University Hospital, Paris, France
| | - Joe-Elie Salem
- Department of Pharmacology, INSERM, AP-HP, CIC-1901, Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Samia Boussouar
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, Paris, France
- ICT Cardiovascular and Thoracic Imaging Unit, AP-HP, Pitié Salpêtrière University Hospital, Paris, France
| | | | - Suzanne Laroche
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
| | - Chloé Amouyal
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
- INSERM UMR_S 1166, Sorbonne University, Team Metabolic Diseases, Diabetes and Co-Morbidities, Paris, France
| | - Agnès Hartemann
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
- INSERM UMR_S 1166, Sorbonne University, Team Metabolic Diseases, Diabetes and Co-Morbidities, Paris, France
| | - Fabienne Foufelle
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- INSERM UMR_S 1166, Sorbonne University, Team Metabolic Diseases, Diabetes and Co-Morbidities, Paris, France
| | - Olivier Bourron
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
- INSERM UMR_S 1166, Sorbonne University, Team Metabolic Diseases, Diabetes and Co-Morbidities, Paris, France
| |
Collapse
|
27
|
Li C, Fang L, Su X, Zhang J, Xiong H, Yu H, Zhu Z, Lin X, Min K, Wu D, Chen Z, Gong J, Xie CM. Macrophage miR-4524a-5p/TBP promotes β-TrCP -TIM3 complex activation and TGFβ release and aggravates NAFLD-associated fibrosis. Cell Death Dis 2025; 16:315. [PMID: 40251185 PMCID: PMC12008196 DOI: 10.1038/s41419-025-07574-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025]
Abstract
Macrophages hold a critical position in maintenance of hepatic homeostasis and in injury and repair processes in acute and chronic liver diseases. TIM3 is a promising protector in MCD-induced steatohepatitis in acute liver injury. However, we recently find TIM3 as a driver of fibrosis in MCD/HFD-induced chronic liver injury. This study aims to explore how macrophage TIM3 drivers NAFLD-associated chronic liver injury as well as identify a subtype of fibrotic patients suitable for anti-TIM3 immunotherapy. Here, we found that TIM3 was highly expressed in liver macrophages in a long-term MCD- or HFD-fed mice with fibrotic NASH. Elevated β-TrCP in macrophages promoted TIM3 polyubiquitination and membrane translocation. The ubiquitinated TIM3 then bound with PI3K and followed by inhibition of mTOR and activation of macrophage M2 polarization and TGF-β release, leading to HSC activation and liver fibrosis. Furthermore, elevated TIM3 was attributed to the transcriptional TBP upregulation and miR-4524a-5p downregulation. Targeting of TIM3 significantly attenuated liver fibrosis in mice. In clinical NASH patients, elevated macrophage TIM3 is positively correlated with TBP expression and negatively associated with miR-4524a-5p. Decreased miR-4524a-5p in plasma was a biomarker for the NASH fibrosis patients suitable for anti-TIM3 therapy. In conclusion, this study reveals that miR-4524a-5p/TBP promotes β-TrCP/TIM3 complex activation in macrophages and aggravates chronic NASH fibrosis, providing miR-4524a-5p as an effective blood biomarker for a subtype of chronic NASH patients with fibrosis suitable for anti-TIM3 treatment.
Collapse
Affiliation(s)
- Chunming Li
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Fang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xingxing Su
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haojun Xiong
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongqiang Yu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhu Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaotong Lin
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ke Min
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Di Wu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyu Chen
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Chuan-Ming Xie
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
28
|
Moon SY, Son M, Kang YW, Koh M, Lee JY, Baek YH. Associations between non-insulin-based insulin resistance surrogate markers and liver-related outcomes in metabolic dysfunction-associated steatotic liver disease: a nationwide cohort study in South Korea. BMC Gastroenterol 2025; 25:274. [PMID: 40251533 PMCID: PMC12008836 DOI: 10.1186/s12876-025-03877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) is a crucial mechanism in the pathogenesis and clinical progression of metabolic dysfunction-associated steatotic liver disease (MASLD). This study aimed to examine the relationship between non-insulin-based IR surrogate markers and the incidence of liver-related outcomes in individuals with MASLD in a nationwide Korean cohort. METHODS A total of 66,334 individuals with MASLD who underwent a health examination between 1 January 2009 and 31 December 2010 were included in the study and followed until 31 December 2019, with a median follow-up period of 9.4 years. Hepatic steatosis was defined as a fatty liver index ≥ 30. The triglyceride-glucose (TyG) index, triglyceride-to-high-density lipoprotein cholesterol (TG/HDL) ratio, and metabolic score of IR (METS-IR) were employed as non-insulin-based IR surrogate markers. The MASLD groups were divided into four groups based on the non-insulin-based IR surrogate markers quartiles. The primary outcome was liver-related outcomes, as a composite of hepatocellular carcinoma and decompensated liver cirrhosis. RESULTS The MASLD group was 64.4% male (average age, 59.0 years). Using the lowest quartile (Q1) of the three non-insulin-based IR surrogate markers as a reference, a decrease in the adjusted hazard ratio for liver-related outcomes was observed from Q2 to Q4: (TyG: Q2 0.90 [95% confidence interval [CI]: 0.79-1.02], Q3 0.80 [95% CI: 0.70-0.91], Q4 0.80 [95% CI: 0.69-0.92]; TG/HDL: Q2 0.85 [95% CI: 0.75-0.97], Q3 0.81 [95% CI: 0.71-0.92], Q4 0.81 [95% CI: 0.71-0.93]; METS-IR: Q2 0.83 [95% CI: 0.73-0.95], Q3 0.80 [95% CI: 0.70-0.91], Q4 0.80 [95% CI: 0.70-0.92]). CONCLUSIONS A lower non-insulin-based IR surrogate marker in the MASLD group may be associated with an increased risk of liver-related outcomes. In the course of monitoring MASLD, metabolic alterations must be meticulously observed.
Collapse
Affiliation(s)
- Sang Yi Moon
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea.
- Department of Data Sciences Convergence, Dong-A University Interdisciplinary Program, Busan, South Korea.
| | - Minkook Son
- Department of Data Sciences Convergence, Dong-A University Interdisciplinary Program, Busan, South Korea
- Department of Physiology, Dong-A University College of Medicine, Busan, South Korea
| | - Yeo Wool Kang
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Myeongseok Koh
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Jong Yoon Lee
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Yang Hyun Baek
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea.
| |
Collapse
|
29
|
Habibi M, Ferguson D, Eichler SJ, Chan MM, Fu C, Pietka TA, Bredemeyer AL, LaPoint A, Shew TM, He M, Liss KHH, Lutkewitte AJ, Cho K, Schilling JD, Patti GJ, Finck BN. A Critical Role for the Mitochondrial Pyruvate Carrier in Hepatic Stellate Cell Activation. Cell Mol Gastroenterol Hepatol 2025:101517. [PMID: 40239806 DOI: 10.1016/j.jcmgh.2025.101517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) are non-parenchymal cells of the liver that produce the extracellular matrix that forms fibrotic lesions in chronic liver disease, including metabolic dysfunction-associated steatohepatitis (MASH). The mitochondrial pyruvate carrier (MPC) catalyzes the transport of pyruvate from the cytosol into the mitochondrial matrix, which is a critical step in pyruvate metabolism. An MPC inhibitor has shown promise as a novel therapeutic for MASH and HSC activation, but a mechanistic understanding of the direct effects of MPC inhibition on HSC activation is lacking. METHODS Stable lines of LX2 cells expressing short hairpin RNA against MPC2 were established and examined in a series of studies to assess HSC metabolism and activation. Mice with conditional, HSC-specific MPC2 deletion were generated and their phenotypes assessed in the context of diets that cause hepatic steatosis, injury, and early-stage fibrosis. RESULTS Genetic suppression of MPC activity markedly decreased expression of markers of HSC activation in vitro. MPC knockdown reduced the abundance of several intermediates of the tricarboxylic acid cycle and attenuated HSC activation by suppressing hypoxia inducible factor-1α signaling. Supplementing alpha-ketoglutarate to replenish the tricarboxylic acid cycle intermediates was sufficient to overcome the effects of MPC inhibition on hypoxia inducible factor-1α and HSC activation. On high-fat diets, mice with HSC-specific MPC deletion exhibited reduced circulating transaminases, numbers of HSCs, and hepatic expression of markers of HSC activation and inflammation compared with wild-type mice. CONCLUSIONS These data suggest that MPC inhibition modulates HSC metabolism to attenuate activation and illuminate mechanisms by which MPC inhibitors could prove therapeutically beneficial for treating MASH.
Collapse
Affiliation(s)
- Mohammad Habibi
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel Ferguson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Sophie J Eichler
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mandy M Chan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Christina Fu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Terri A Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Andrea L Bredemeyer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew LaPoint
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Trevor M Shew
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Kim H H Liss
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri; (4)Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew J Lutkewitte
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin Cho
- Department of Chemistry, Washington University, St. Louis, Missouri
| | - Joel D Schilling
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Gary J Patti
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Chemistry, Washington University, St. Louis, Missouri
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
30
|
Romero-Gómez M, Escalada J, Noguerol M, Pérez A, Carretero J, Crespo J, Mascort JJ, Aguilar I, Tinahones F, Cañones P, Gómez-Huelgas R, de Luis D, Genúa Trullos I, Aller R, Rubio MA. Multidisciplinary clinical practice guideline on the management of metabolic hepatic steatosis. GASTROENTEROLOGIA Y HEPATOLOGIA 2025:502442. [PMID: 40221023 DOI: 10.1016/j.gastrohep.2025.502442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Metabolic hepatic steatosis (MetHS) is a clinically heterogeneous, multisystemic, dynamic, and complex disease, whose progression is one of the main causes of cirrhosis and hepatocarcinoma. This clinical practice guideline aims to respond to its main challenges, both in terms of disease burden and complexity. To this end, recommendations have been proposed to experts through the Delphi method. The consensus was optimal in recommendations regarding type 2 diabetes as a risk factor (1.5.1, 4.5.1), in which cases early detection of MetHS should be carried out (4.5.2). Its results also emphasize the importance of the use of non-invasive tests (FIB-4, NFS, HFS) for the exclusion of significant fibrosis in patients with suspected MetHS (2.3.1, 2.3.3). Diagnosis should be carried out through the sequential combination of non-invasive indices and transient elastography by FibroScan® for its risk stratification (2.3.3). A nearly unanimous consensus was reached regarding the role of early prevention in the impact on the quality of life and survival of patients (5.1.2), as well as on the effectiveness of the Mediterranean diet and physical exercise in relation to the improvement of steatosis, steatohepatitis and fibrosis in MetHS patients (5.2.2) and on the positive results offered by resmiterom and semaglutide in promoting fibrosis regression (5.4.1). Finally, a great consensus has been reached regarding the importance of multidisciplinary management in MetHS, for which it is essential to agree on multidisciplinary protocols for referral between levels in each health area (6.2.1), as well as ensuring that referrals to Hepatology/Digestive and Endocrinology or Internal Medicine services are effective and beneficial to prevent the risk of disease progression (6.2.3, 6.3.1).
Collapse
Affiliation(s)
- Manuel Romero-Gómez
- UGC Aparato Digestivo, Hospital Universitario Virgen del Rocío, Sevilla, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, España; Instituto de Biomedicina de Sevilla (HUVR/CSIC/US), Departamento de Medicina, Universidad de Sevilla, Sevilla, España; Asociación España para el Estudio del Hígado, España.
| | - Javier Escalada
- Clínica Universidad de Navarra, Pamplona, España; Sociedad Española de Endocrinología y Nutrición, España; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, España; Instituto de Investigación en la Salud de Navarra (IdiSNA), Pamplona, España.
| | - Mar Noguerol
- Centro de Salud Universitario Cuzco de Fuenlabrada, Madrid, España; Sociedad Española de Medicina de Familia y Comunitaria, España
| | - Antonio Pérez
- Servicio de Endocrinología y Nutrición, Hospital de la Santa Creu i Sant Pau, Barcelona, España; CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), España; Sociedad Española de Diabetes, España
| | - Juana Carretero
- Hospital Universitario de Badajoz, Badajoz, España; Sociedad Española de Medicina Interna (SEMI), España
| | - Javier Crespo
- Hospital Universitario Marqués de Valdecilla, Santander, España; Sociedad Española de Patología Digestiva, España; Grupo de Investigación Clínica y Traslacional en Enfermedades Digestivas, España; Instituto de Investigación Valdecilla (IDIVAL), Santander, España
| | - Juan J Mascort
- Sociedad Española de Medicina de Familia y Comunitaria, España; Centro de Salud Florida Sud, Institut Català de la Salut, Hospitalet de Llobregat, España
| | - Ignacio Aguilar
- Clínica Universidad de Navarra, Pamplona, España; Sociedad Española de Endocrinología y Nutrición, España; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, España; Instituto de Investigación en la Salud de Navarra (IdiSNA), Pamplona, España
| | - Francisco Tinahones
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, España; Departamento de Endocrinología y Nutrición, Hospital Virgen de la Victoria, Málaga, España; Sociedad Española de Obesidad, España; Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma Bionard, Universidad de Málaga, Málaga, España
| | - Pedro Cañones
- Sociedad Española de Médicos Generales y de Familia, España
| | - Ricardo Gómez-Huelgas
- Sociedad Española de Medicina Interna (SEMI), España; Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Málaga, España; Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, España
| | - Daniel de Luis
- Sociedad Española de Endocrinología y Nutrición, España; Servicio de Endocrinología y Nutrición, Hospital Clínico Universitario de Valladolid, Valladolid, España; Centro de Investigación de Endocrinología y Nutrición, Universidad de Valladolidad, Valladolid, España
| | - Idoia Genúa Trullos
- Servicio de Endocrinología y Nutrición, Hospital de la Santa Creu i Sant Pau, Barcelona, España; CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), España; Sociedad Española de Diabetes, España
| | - Rocío Aller
- Servicio de Endocrinología y Nutrición, Hospital de la Santa Creu i Sant Pau, Barcelona, España; CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), España; Sociedad Española de Diabetes, España; Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Universidad de Valladolid, Valladolid, España; Ciber Enfermedades infecciosas (CIBERINFEC), España
| | - Miguel A Rubio
- Sociedad Española de Endocrinología y Nutrición, España; Hospital Clínico San Carlos, Madrid, España
| |
Collapse
|
31
|
Boulos M, Mousa RS, Jeries N, Simaan E, Alam K, Bulus B, Assy N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. Int J Mol Sci 2025; 26:3448. [PMID: 40244398 PMCID: PMC11989262 DOI: 10.3390/ijms26073448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic syndrome (MetS) and metabolic dysfunction-associated steatotic liver disease (MASLD) are closely related, with rapidly increasing prevalence globally, driving significant public health concerns. Both conditions share common pathophysiological mechanisms such as insulin resistance (IR), adipose tissue dysfunction, oxidative stress, and gut microbiota dysbiosis, which contribute to their co-occurrence and progression. While the clinical implications of this overlap, including increased cardiovascular, renal, and hepatic risk, are well recognized, current diagnostic and therapeutic approaches remain insufficient due to the clinical and individuals' heterogeneity and complexity of these diseases. This review aims to provide an in-depth exploration of the molecular mechanisms linking MetS and MASLD, identify critical gaps in our understanding, and highlight existing challenges in early detection and treatment. Despite advancements in biomarkers and therapeutic interventions, the need for a comprehensive, integrated approach remains. The review also discusses emerging therapies targeting specific pathways, the potential of precision medicine, and the growing role of artificial intelligence in enhancing research and clinical management. Future research is urgently needed to combine multi-omics data, precision medicine, and novel biomarkers to better understand the complex interactions between MetS and MASLD. Collaborative, multidisciplinary efforts are essential to develop more effective diagnostic tools and therapies to address these diseases on a global scale.
Collapse
Affiliation(s)
- Mariana Boulos
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Rabia S. Mousa
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Nizar Jeries
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Elias Simaan
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Klode Alam
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Bulus Bulus
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Nimer Assy
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
32
|
Gao Z, Cao S, Yuan H, Wu JZ, Zou G. Broad antifibrotic activities of AK3280 in pulmonary, hepatic, cardiac, and skin fibrosis animal models. Int Immunopharmacol 2025; 151:114337. [PMID: 40015207 DOI: 10.1016/j.intimp.2025.114337] [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: 12/12/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 03/01/2025]
Abstract
Fibrosis is the pathological outcome of many chronic inflammatory diseases, affecting various human organs. It is a significant contributor to global morbidity and mortality that affects nearly half of the elderly population. Pirfenidone (PFD) and nintedanib are approved by the FDA for treating pulmonary fibrosis, but these treatments are associated with poor tolerability and limited efficacy. Moreover, no antifibrotic drugs are approved for other fibrosis-related diseases, highlighting an urgent unmet medical need for more effective therapies. Here we report the in vivo pharmacological activities of AK3280, a novel, orally bioavailable small molecule designed to enhance pharmacokinetics, antifibrotic activity, and tolerability over PFD. AK3280 demonstrated antifibrotic effects across multiple organs, including the lungs, liver, heart, and skin, in various animal models. These results suggest that AK3280 holds promise as a clinically beneficial antifibrotic therapy for a range of fibrotic diseases, especially pulmonary, hepatic, cardiac, and skin fibrosis.
Collapse
Affiliation(s)
- Zhao Gao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Sushan Cao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Haiqing Yuan
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Jim Zhen Wu
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Gang Zou
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China.
| |
Collapse
|
33
|
Torp N, Israelsen M, Krag A. The steatotic liver disease burden paradox: unravelling the key role of alcohol. Nat Rev Gastroenterol Hepatol 2025; 22:281-292. [PMID: 39639157 DOI: 10.1038/s41575-024-01022-y] [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] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
The classification of steatotic liver disease (SLD) has evolved, incorporating all conditions characterized by hepatic lipid accumulation. SLD represents a continuum of disorders that are shaped by the dynamic factors of alcohol intake and cardiometabolic risk factors. This updated classification has profound implications for both the management and research of SLD, especially with the new distinct category of patients with both metabolic and alcohol-related liver disease. In this Perspective, we highlight the pivotal role of alcohol within the SLD framework. We introduce the 'SLD burden paradox': a concept illustrating the disparity in which metabolic dysfunction-associated steatotic liver disease is more prevalent, yet individuals with SLD and excessive alcohol intake (such as in metabolic and alcohol-related liver disease and in alcohol-related liver disease) account for greater global liver-related morbidity and mortality. We explore strategies to mitigate the effect of SLD on morbidity and mortality, emphasizing the importance of early detection and reducing stigma associated with alcohol intake. Our discussion extends to methods for assessing and monitoring alcohol intake together with the critical role of managing cardiometabolic risk factors in patients across the SLD spectrum. Conclusively, we advocate for a coordinated care framework that adopts a person-centric approach when managing SLD, aiming to improve outcomes and patient care.
Collapse
Affiliation(s)
- Nikolaj Torp
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Israelsen
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
34
|
Leite NC, Villela-Nogueira CA, Santos LV, Cardoso CRL, Salles GF. Prognostic value of changes in vibration-controlled transient elastography parameters for liver, cardiovascular and mortality outcomes in individuals with type 2 diabetes and metabolic dysfunction-associated steatotic liver disease: The Rio de Janeiro type 2 diabetes cohort. Diabetes Obes Metab 2025; 27:2024-2034. [PMID: 39822129 DOI: 10.1111/dom.16195] [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: 11/23/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND/AIMS The prognostic importance of changes in vibration-controlled transient elastography (VCTE) parameters, liver stiffness measurement (LSM), and controlled attenuation parameter (CAP), in individuals with type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD) is unknown. METHODS A prospective cohort of 288 patients underwent 2 VCTE exams at least 2 years apart, and the relative percentage changes in LSM and CAP were calculated. Outcomes were the occurrence of any liver-related events (LREs), cardiovascular events (CVEs), and all-cause mortality. Multivariable Cox analyses, adjusted for liver and cardiometabolic factors, assessed associations between VCTE parameters changes, both as continuous and dichotomical variables (LSM increase >15% and CAP reduction >10%), and outcomes. RESULTS During a median follow-up of 6 years, there were 22 LREs, 28 CVEs, and 37 all-cause deaths. For LREs, baseline LSM was the strongest predictor, but LSM increases added further prognostic value (hazard ratio [HR]: 1.5 [1.0-2.1], 1-SD increment). For CVEs, both LSM increase (HR: 1.7 [1.3-2.3]) and CAP reduction (HR: 1.5 [1.0-2.3], 1-SD decrease) were significant predictors. For all-cause mortality, baseline CAP was a protective predictor. When classified into subgroups based on LSM and CAP changes, the subgroup with both increased LSM and reduced CAP had the highest risks for CVEs (HR:5.3 [1.4-19.6]) and all-cause mortality (HR: 3.4 [1.2-9.6]). The highest risk for LREs was observed in the subgroup with increased LSM without CAP reduction (HR: 3.5 [0.9-12.9]). CONCLUSIONS VCTE parameters changes, LSM increase and CAP reduction, provide prognostic information for adverse liver, cardiovascular, and mortality outcomes in individuals with T2D and MASLD.
Collapse
Affiliation(s)
- Nathalie C Leite
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristiane A Villela-Nogueira
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lorrane V Santos
- School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia R L Cardoso
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gil F Salles
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Loh Jiezhen T, Salomao M, Moreira R, Pai R. Reliability of an Expanded Non-alcoholic Steatohepatitis Grading and Staging System for Assessment of Disease Activity and Fibrosis. Int J Surg Pathol 2025; 33:281-288. [PMID: 39034304 DOI: 10.1177/10668969241260226] [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] [Indexed: 07/23/2024]
Abstract
The nonalcoholic steatohepatitis clinical research network (NASH-CRN) system is commonly used for histologic assessment of disease activity and fibrosis in NASH. Despite this, the system does not fully capture the range of disease activity and fibrosis. As such, an expanded NAS (E-NAS) grading and staging system with a calculated E-NAS index was developed by our group. In this follow up study, we aim to revalidate the E-NAS system and compare its reliability to existing systems. Hematoxylin and eosin and trichrome stained sections from 40 liver biopsies were reviewed digitally by four hepatopathologists and assessed using the NASH-CRN and E-NAS systems as well as a modified Ishak fibrosis stage. The pathologist's gestalt impression of disease activity and fibrosis was scored on a Visual Analogue Scale (VAS), which ranged from 0 (no activity/fibrosis) to 100 (the worst activity/fibrosis ever seen). Inter-rater reliability was assessed, and Spearman correlation coefficients were calculated. The E-NAS index had higher inter-rater agreement versus the NAS score (ICC 0.70 vs 0.61). The inter-rater agreement for ballooning in the E-NAS system was also higher at 0.67 compared to the NAS (ICC 0.60). ICCs for fibrosis were comparable between all the systems assessed (0.78 to 0.88). Finally, the calculated E-NAS index was higher with increasing stage of fibrosis compared to the NAS suggesting that it associates better with fibrosis. In summary, the E-NAS system demonstrates substantial inter-rater reliability as well as improved correlation with disease activity VAS and fibrosis compared to the NAS score.
Collapse
Affiliation(s)
- Tracy Loh Jiezhen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Marcela Salomao
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Roger Moreira
- Department of Pathology and Laboratory Medicine, Mayo Clinic Rochester, Rochester, MN, USA
| | - Rish Pai
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
36
|
Arvanitakis K, Koufakis T, Cholongitas E, Francque S, Germanidis G. Insights into the results of Resmetirom trials: Can a thyroid hormone receptor agonist be the holy grail of MASH therapy? Pharmacol Ther 2025; 268:108811. [PMID: 39938598 DOI: 10.1016/j.pharmthera.2025.108811] [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: 10/26/2024] [Revised: 01/12/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
Despite the heavy individual patient and socioeconomic burden of metabolic dysfunction-associated steatohepatitis (MASH), until recently, no pharmacological therapy for MASH was approved, with available treatment options geared towards associated cardiometabolic risk factors. Accelerated approval of resmetirom, a thyroid hormone receptor-β agonist to be used in conjunction with diet and exercise, marks a significant step forward in the treatment of MASH, offering tempered optimism to healthcare providers and millions of patients around the world for more effective management. Evidence from phase 2 and 3 clinical trials suggests that resmetirom has the potential to alleviate hepatic fibrosis and inflammation and significantly reduce liver lipid content. Notwithstanding this landmark event, the clinical implementation of resmetirom comes with important challenges, for example, ensuring patient access to treatment and demonstrating effects on hard MASH-related outcomes, such as progression to cirrhosis and hepatocellular carcinoma. Additional considerations include the evaluation of co-administration with other hepatoprotective treatments and the assessment of the efficacy in specific MASH sub-phenotypes. Furthermore, the accumulation of real-world data and experience is expected to help answer the remaining questions about the (long-term) effectiveness and safety profile of the drug. The purpose of this article is to provide an updated and critical review of the mechanisms of action, efficacy, and safety of resmetirom based on the latest clinical trials, to define its place within the broader landscape of MASH management, and to highlight current knowledge gaps and opportunities for future research in the field.
Collapse
Affiliation(s)
- Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- Second Propedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital, Medical School of National and Kapodistrian University of Athens, Athens, Greece
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece.
| |
Collapse
|
37
|
Yang Y, Chen X, Zhang H, Yang G, Zhu X, Si X, Chen F, Zhao Y, Jin F, Lu J. The correlation between the polymorphism of lysolecithin acyltransferase (MBOAT7) rs641738 and liver fibrosis. Per Med 2025; 22:113-119. [PMID: 40055064 DOI: 10.1080/17410541.2025.2476379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE We aimed to explore the relationship between the MBOAT7 rs641738 gene polymorphism and liver fibrosis and inflammation. METHODS A total of 214 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) were allocated into the mild-to-moderate and severe liver fibrosis groups based on liver fibrosis degree. The genotypes at the MBOAT7 rs641738 locus were evaluated. Differences in clinical and biochemical indicators, as well as the genotype and allele frequency distributions of the MBOAT7 rs641738 polymorphism, were analyzed across groups with varying degrees of liver fibrosis. Additionally, the clinical and biochemical differences among patients with different genotypes were examined. RESULTS Significant differences were observed in the distribution of CC, CT, and TT genotypes, as well as C and T allele frequencies at the MBOAT7 rs641738 locus, between patients with mild-to-moderate and severe fibrosis. Carriers of the CT + TT genotype had a higher risk of developing severe liver fibrosis compared to those with the CC genotype (OR > 1). Furthermore, CT + TT carriers had higher levels of inflammatory cytokines and more severe fibrosis than CC genotype carriers (all p < 0.05). CONCLUSION The MBOAT7 rs641738 gene polymorphism is associated with the severity of liver fibrosis and inflammation.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiang Chen
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Huiqin Zhang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Gang Yang
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiaoyun Zhu
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiujing Si
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Feilong Chen
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yan Zhao
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Feng Jin
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Juanjuan Lu
- Department of Digestive, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Pavan Sai Kumar Rao D, Patro S, Sharma V, Choudhary A, Desale S, Nath P. Diagnostic Accuracy of Red Cell Distribution Width to Platelet Ratio for the Prediction of Liver Fibrosis in Patients With Chronic Liver Disease From Eastern India. Cureus 2025; 17:e82014. [PMID: 40352011 PMCID: PMC12065511 DOI: 10.7759/cureus.82014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
Background Early diagnosis of liver cirrhosis in patients with chronic liver disease (CLD) can help delay/prevent complications and thereby improve survival. The currently available diagnostic modalities for the non-invasive assessment of hepatic fibrosis, especially FibroScan, are costly and not widely available, whereas various non-invasive scores for the assessment of fibrosis are cumbersome. Hence, we aimed to develop an easy and simple score for predicting cirrhosis in patients from Eastern India suffering from CLD with a better diagnostic accuracy. Methodology This cross-sectional, observational study was conducted between September 2019 and September 2021 in East India. Our study participants were patients who had CLD of etiologies such as alcohol-related liver disease, non-alcoholic fatty liver disease, chronic viral hepatitis B, chronic viral hepatitis C, primary biliary cholangitis, and autoimmune hepatitis, who had undergone FibroScan of the liver. All demographic details were noted, and the patients were subjected to physical examination, followed by hematological as well as biochemical investigations, including liver function tests. Non-invasive scores (such as aspartate aminotransferase (AST) to platelet ratio index (APRI) and Fibrosis-4 score (FIB-4) and red cell distribution width (RDW) to platelet ratio (RPR)) were computed, and their diagnostic accuracy for prediction of advanced fibrosis and cirrhosis were evaluated by receiver operating characteristic curve (ROC curve) analysis with comparison of area under the ROC curves. Pearson correlation and logistic regression analysis were also performed to study the association of these scores with advanced fibrosis and cirrhosis. Results The area under the ROC (AUROC) curve of the APRI score, FIB-4 score, RPR, and RPR × AST for prediction of advanced liver fibrosis was 0.817, 0.799, 0.706, and 0.811, respectively. Similarly, the AUROC of the above scores for the prediction of cirrhosis was 0.889, 0.858, 0.797, and 0.898. However, the product of RPR and AST was superior than APRI and FIB-4 for predicting cirrhosis. An RPR × AST value above the cut-off of 4.818 can help predict liver cirrhosis with 85.7% sensitivity and 85.5% specificity. Pearson correlation and logistic regression analysis also proved the association of these scores with liver fibrosis. Conclusions RPR is a simple, inexpensive, and easily available marker for predicting liver cirrhosis. Nevertheless, the variable RPR × AST can predict liver cirrhosis in patients with CLD with even greater diagnostic accuracy.
Collapse
Affiliation(s)
- D Pavan Sai Kumar Rao
- Department of Medical Gastroenterology, Gleneagles BGS Global Hospitals, Bengaluru, IND
| | - Shubhransu Patro
- General Medicine, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| | - Vibha Sharma
- General Medicine, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| | - Arushi Choudhary
- General Medicine, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| | - Shubham Desale
- General Medicine, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| | - Preetam Nath
- Gastroenterology and Hepatology, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| |
Collapse
|
41
|
Issa G, Shang Y, Strandberg R, Hagström H, Wester A. Cause-specific mortality in 13,099 patients with metabolic dysfunction-associated steatotic liver disease in Sweden. J Hepatol 2025:S0168-8278(25)00156-4. [PMID: 40139508 DOI: 10.1016/j.jhep.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/08/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND & AIMS Data on cause-specific mortality in metabolic dysfunction-associated steatotic liver disease (MASLD) are limited. We aimed to determine the rate and risk of death from different causes in patients with MASLD compared to the general population in Sweden. METHODS In this population-based cohort study, we identified individuals with an ICD-10 code for MASLD in inpatient or specialized outpatient care using Swedish healthcare registers from 2002-2020 (n = 13,099) and matched them with up to 10 controls (median 9) from the general population for age, sex, municipality, and calendar year (n = 118,884). We used Cox regression to estimate adjusted hazard ratios (HRs) and 95% CIs for 11 different primary causes of death. 15-year cumulative incidences of death were calculated while accounting for competing risks. RESULTS In total, 1,628 (12.4%) deaths occurred in patients with MASLD and 9,119 (7.7%) in controls during a median follow-up of 4.7 (IQR 2.0-9.2) and 5.8 years (IQR 2.7-10.5), respectively. MASLD was associated with higher all-cause mortality (HR 1.85, 95% CI 1.74-1.96) and higher rates of all specific causes of death except mental health disorder. The strongest associations were observed for non-hepatocellular carcinoma (HCC) liver-related (HR 26.9, 95% CI 19.4-37.3) and HCC-related (HR 35.0, 95% CI 17.0-72.1) mortality. However, the highest estimated 15-year cumulative incidences of death in patients with MASLD were for non-HCC cancer (7.3%) and cardiovascular disease (7.2%). CONCLUSIONS MASLD was strongly associated with liver- and HCC-related mortality, but the absolute risks of death were highest for non-HCC cancer and cardiovascular disease. Mortality was increased for nearly all causes in patients with MASLD, suggesting that earlier multidisciplinary care is needed to reduce excess mortality. IMPACT AND IMPLICATIONS Previous studies on mortality in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) were either small, restricted to liver-related mortality, relied on liver biopsy to identify patients (thus inducing selection bias), or mainly used data from old cohorts. In a nationwide cohort study of all patients diagnosed with MASLD in inpatient or specialized outpatient care in Sweden between 2002 and 2020, we found a nearly doubled all-cause mortality rate and higher mortality than the general population from a wide range of causes, indicating that earlier multidisciplinary care may be needed to reduce premature mortality in patients with MASLD. The absolute risk estimates of death in our study may be useful for clinicians and policymakers to inform patients about their prognosis and potentially implement clinical or public health strategies to reduce premature mortality.
Collapse
Affiliation(s)
- Gabriel Issa
- 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
| | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - Axel Wester
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
42
|
Truong E, Alnimer L, Gornbein JA, Yang JD, Alkhouri N, Harrison SA, Noureddin M. Agile 3+ and 4 Scores Accurately Predict Major Adverse Liver Outcomes, Liver Transplant, Progression of MELD Score, the Development of Hepatocellular Carcinoma, and Death in NAFLD. Dig Dis Sci 2025:10.1007/s10620-025-08850-1. [PMID: 40126753 DOI: 10.1007/s10620-025-08850-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/04/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND AND AIMS Based on liver stiffness measurement by vibration controlled transient elastography (LSM by VCTE), the Agile 3+ and 4 are novel noninvasive scores that accurately identify advanced fibrosis (≥ F3) and cirrhosis (F4), respectively. We investigated and compared the Agile 3+ and 4 scores' performances in predicting adverse events to LSM alone, FIB-4 and Fibroscan-AST (FAST) score. METHOD This retrospective analysis included NAFLD patients with LSM by VCTE and laboratory testing from a tertiary care center from 2013 to 2022. Adverse events were defined as major adverse liver outcomes (MALO), hepatocellular carcinoma, liver transplant, and death. MALO was defined as ascites, hepatic encephalopathy, or esophageal variceal bleeding. We used the Cox proportional hazard rate model and the Harrell's concordance (C) statistic to compare predictive performances. RESULTS 733 total subjects with median follow-up of 27.0 months were included. Average age was 58.1 years and 32.8% had type 2 diabetes. Average alanine aminotransferase was 46.6 IU/L, aspartate aminotransferase: 34.5 IU/L, albumin: 4.4 g/dL, and platelets: 241.1 × 109/L. Fourteen subjects had 21 adverse outcomes, including 10 MALO, 5 HCC, 4 liver transplants, 3 progression of MELD score, and 6 deaths. Agile 3+ and 4 respectively had the highest C stats of 0.911 (C stat SE 0.028) and 0.909 (C stat SE 0.029) compared to LSM (C stat 0.857, C stat SE 0.045), FIB-4 (C stat 0.843, C stat SE 0.037) or FAST (C stat 0.703, C stat SE 0.085). CONCLUSION The Agile 3+ and 4 scores had the highest likelihood of accurately predicting adverse outcomes including MALO and death compared to LSM alone, FIB-4 or FAST score.
Collapse
Affiliation(s)
- Emily Truong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lynna Alnimer
- Division of Gastroenterology, Henry Ford Providence Hospital, Michigan State University/College of Human Medicine, Southfield, MI, USA
| | - Jeffrey A Gornbein
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Mazen Noureddin
- Houston Methodist Hospital, Houston Research Institute, 1155 Dairy Ashford Suite 200, Houston, TX, 77079, USA.
- Lynda K. and David M. Underwood Center for Digestive Disorders, Department of Medicine, J.C. Walter Jr. Transplant Center, Sherrie & Alan Conover Center for Liver Disease & Transplantation, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, USA.
| |
Collapse
|
43
|
Qiu T, Zhu X, Wu J, Hong W, Hu W, Fang T. Mechanisms of rifaximin inhibition of hepatic fibrosis in mice with metabolic dysfunction associated steatohepatitis through the TLR4/NFκB pathway. Sci Rep 2025; 15:9815. [PMID: 40118973 PMCID: PMC11928543 DOI: 10.1038/s41598-025-92282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) has become a serious public health problem, posing an increasingly dangerous threat to human health owing to its increasing prevalence and accompanying intra- and extrahepatic adverse outcomes. Rifaximin is considered to have therapeutic potential for MASH; however, its efficacy remains controversial. Our study aimed to observe the ameliorative effects of rifaximin and explore its possible mechanisms at the cellular level. 1. 42 male C57BL/6J mice were divided into 3 groups, the CON group and MCD group were fed with normal feed and MCD feed for 12 weeks respectively, and the MCD + RFX group was treated with rifaximin by gavage for 4 weeks on the basis of MCD feed. Hematoxylin-eosin staining, Sirius red staining and immunohistochemical staining were used to observe the histopathological changes of liver and intestine. Differences in liver transaminases, inflammatory factors, fibrosis indexes and intestinal tight junction proteins were compared among the 3 groups of mice. 2. A MASH cell model was constructed by inducing HepG2 cells with free fatty acids to observe the effects of rifaximin on MASH in vitro. In addition, the effects of rifaximin on TLR4/NF-κB signaling pathway were explored by applying TLR4 agonist LPS and TLR4 inhibitor TAK-242. Hepatic histopathology was significantly improved in MASH mice after rifaximin treatment, and their serum alanine aminotransferase and aspartate aminotransferase levels were (72.72 ± 5.68) U/L and (222.8 ± 11.22) U/L, respectively, which were significantly lower than those in the MCD group [(293.3 ± 10.69) U/L and (414.1 ± 36.29) U/L, P < 0.05], and the levels of inflammatory factors and fibrosis indicators were reduced. Rifaximin ameliorated intestinal barrier injury with increased expression of intestinal tight junction protein ZO-1 in the MCD + RFX group of mice, and the concentration of LPS-binding proteins (4.92 ± 0.55 vs. 15.82 ± 1.71, P < 0.05) was lower than that in the MCD group. In the NASH cell model, rifaximin similarly exerted inhibitory effects on its inflammatory factors and TLR4/NF-κB signaling pathway. Application of TLR4 inhibitors weakened the inhibitory effect of rifaximin on MASH. Our study supports rifaximin as a potential treatment for MASH, with potential mechanisms related to improving intestinal barrier integrity and downregulating the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ting Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of General Practice, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Xiaodong Zhu
- Department of Gastroenterology, Quanzhou First Hospital, Quanzhou, China
| | - Jingju Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wenyuan Hong
- Anxi Maternal and Child Health Hospital, Quanzhou, China
| | - Weitao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
44
|
Zhang L, Ma S, Sun R, Xie R, Shen P. Cobalt exposure was associated with the risk of hepatic steatosis and advanced liver fibrosis based on a cross-sectional study from NHANES. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 293:118003. [PMID: 40068554 DOI: 10.1016/j.ecoenv.2025.118003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Although Emerging evidence suggests the association of environmental factors with hepatic steatosis and fibrosis, the relationship between Cobalt exposure and hepatic steatosis and fibrosis was not clear. AIM Our study was aimed to explore the association between blood Cobalt level and hepatic steatosis and advanced liver fibrosis diagnosed by vibration controlled transient elastography (VCTE) in US adults. METHODS This study analyzed data from 3193 individuals participating in the 2017-2018 National Health and Nutrition Examination Surveys. Participants were classified into four groups according to the quartiles of blood cobalt concentration. Liver stiffness and fat content were assessed through vibration-controlled transient elastography (VCTE), including measurements of the controlled attenuation parameter (CAP). The association between blood cobalt levels and the prevalence of hepatic steatosis and advanced liver fibrosis was explored using logistic regression models and stratified subgroup analyses. RESULTS The CAP values showed a significant decline across increasing cobalt quartiles. Participants in the highest quartile had a 41 % lower risk of hepatic steatosis compared to those in the lowest quartile (odds ratio: 0.59, 95 % confidence interval: 0.46-0.76, p < 0.001). However, no significant association existed between blood Cobalt and advanced liver fibrosis. Subgroup analysis revealed that the relationship was consistent across age, gender and body mass index subgroups. CONCLUSIONS This study showed that blood Cobalt level was negatively association with hepatic steatosis. This may be due to increased development from hepatic steatosis to advanced liver fibrosis upon Cobalt exposure.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Geriatric Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huaian 223000, China
| | - Shijie Ma
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huaian 223000, China
| | - Rui Sun
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huaian 223000, China
| | - Rui Xie
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huaian 223000, China.
| | - Peng Shen
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huaian 223000, China.
| |
Collapse
|
45
|
Leszczynska A, Alle T, Kaufmann B, Sung H, Stoess C, Reca A, Kim A, Kim S, Tran C, Oukoloff K, Monti L, Lucero B, Gertsman I, Momper JD, Hartmann P, Feldstein AE, Dohil R, Ballatore C. d 4-Cystamine: A Deuterated Cystamine Derivative with Improved Anti-Inflammatory and Anti-Fibrotic Activities in a Murine Model of Fibrosing Steatohepatitis. ACS Pharmacol Transl Sci 2025; 8:885-898. [PMID: 40109735 PMCID: PMC11915185 DOI: 10.1021/acsptsci.4c00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial chronic disease that can progress to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis, ultimately leading to liver cirrhosis and hepatocellular carcinoma. Oxidative stress is believed to play an important role in the development of MASH. Small aminothiol compounds such as cysteamine and its oxidized precursor, cystamine, are known pleiotropic compounds that exhibit relatively potent antioxidant and other effects. Herein, we evaluate the efficacy of cystamine, as well as two deuterated derivatives, in a choline-deficient, L-amino acid-defined, high-fat-diet (CDAA-HFD) mouse model of rapidly progressing liver fibrosis. Compared to control mice, daily oral administration of isotopically reinforced cystamine derivatives (200 mg/kg) led to a significant reduction of liver fibrosis and inflammation as well as oxidative stress. Moreover, the efficacy of treatment appeared to increase with the deuteration state of cystamine, with the tetradeuterated derivative, d 4 -cystamine, being the most effective. These results indicate that deuterated cystamine derivatives hold promise as potential candidates for the treatment of MASH.
Collapse
Affiliation(s)
- Aleksandra Leszczynska
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Thibault Alle
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Benedikt Kaufmann
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Hana Sung
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Christian Stoess
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Agustina Reca
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Andrea Kim
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Sun Kim
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Chelsea Tran
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Killian Oukoloff
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ludovica Monti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bobby Lucero
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ilya Gertsman
- Clarus Analytical LLC, 8545 Arjons Dr. Suite A, San Diego, California 92126, United States
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Phillipp Hartmann
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Global Drug Discovery, Novo Nordisk, Copenhagen DK-2880, Denmark
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
46
|
Diaz LA, Arab JP, Idalsoaga F, Perelli J, Vega J, Dirchwolf M, Carreño J, Samith B, Valério C, Moreira RO, Acevedo M, Brahm J, Hernández N, Gadano A, Oliveira CP, Arrese M, Castro-Narro G, Pessoa MG. Updated recommendations for the management of metabolic dysfunction-associated steatotic liver disease (MASLD) by the Latin American working group. Ann Hepatol 2025:101903. [PMID: 40089151 DOI: 10.1016/j.aohep.2025.101903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 03/17/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is one of the leading causes of chronic liver disease globally. Based on the 2023 definition, MASLD is characterized by the presence of metabolic dysfunction and limited alcohol consumption (<140 grams/week for women, <210 grams/week for men). Given the significant burden of MASLD in Latin America, this guidance was developed by the Latin American Association for the Study of the Liver (ALEH) Working Group to address key aspects of its clinical assessment and therapeutic strategies. In Latin America, ultrasonography is recommended as the initial screening tool for hepatic steatosis due to its accessibility, while Fibrosis-4 (FIB-4) is preferred for fibrosis risk stratification, with further evaluation using more specific techniques (i.e., vibration-controlled transient elastography or Enhanced Liver Fibrosis [ELF] test). A Mediterranean diet is advised for all MASLD patients, with a target of 7-10% weight loss for those with excess weight. Complete alcohol abstinence is recommended for patients with significant fibrosis, and smoking cessation is encouraged regardless of fibrosis stage. Pharmacological options should be tailored based on the presence of steatohepatitis, liver fibrosis, excess weight, and diabetes, including resmetirom, incretin-based therapies, pioglitazone, and sodium-glucose cotransporter-2 inhibitors. Bariatric surgery may be considered for MASLD patients with obesity unresponsive to lifestyle and medical interventions. Hepatocellular carcinoma screening is advised for all cirrhotic patients, with consideration given to those with advanced fibrosis based on individual risk. Finally, routine cardiovascular risk assessment and proper diabetes prevention and management remain crucial for all patients with MASLD.
Collapse
Affiliation(s)
- Luis Antonio Diaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Francisco Idalsoaga
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University & London Health Sciences Centre, London, Ontario, Canada
| | - Javiera Perelli
- Unidad de Diabetes y Nutrición Clínica, Clínica Universidad de los Andes, Santiago, Chile
| | - Javier Vega
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Javiera Carreño
- Asociación Latinoamericana para el Estudio del Hígado (ALEH), Santiago, Chile
| | - Bárbara Samith
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cynthia Valério
- Instituto Estadual de Diabetes e Endocrinologia Luiz Capriglione, Rio de Janeiro, RJ, Brasil
| | - Rodrigo Oliveira Moreira
- Instituto Estadual de Diabetes e Endocrinologia Luiz Capriglione, Rio de Janeiro, RJ, Brasil; Faculdade de Medicina de Valença, Centro Universitário de Valença, Valença, RJ, Brasil; Faculdade de Medicina, Centro Universitário Presidente Antônio Carlos, Juiz de Fora, MG, Brasil
| | - Mónica Acevedo
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Brahm
- Unidad de Gastroenterología, Clínica Universidad de los Andes, Santiago, Chile
| | - Nelia Hernández
- Asociación Latinoamericana para el Estudio del Hígado (ALEH), Santiago, Chile; Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Adrian Gadano
- Liver Unit, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina; Department of Research, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Claudia P Oliveira
- Gastroenterology Department, Hospital das Clínicas (LIM07) HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Asociación Latinoamericana para el Estudio del Hígado (ALEH), Santiago, Chile
| | - Graciela Castro-Narro
- Asociación Latinoamericana para el Estudio del Hígado (ALEH), Santiago, Chile; Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico; Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Mario G Pessoa
- Asociación Latinoamericana para el Estudio del Hígado (ALEH), Santiago, Chile; Gastroenterology Department, Hospital das Clínicas (LIM07) HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| |
Collapse
|
47
|
Li S, Zhu H, Zhai Q, Hou Y, Yang Y, Lan H, Jiang M, Xuan J. Exploring Mechanisms of Lang Qing Ata in Non-Alcoholic Steatohepatitis Based on Metabolomics, Network Pharmacological Analysis, and Experimental Validation. Drug Des Devel Ther 2025; 19:1681-1701. [PMID: 40098906 PMCID: PMC11911237 DOI: 10.2147/dddt.s503757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
Background Non-alcoholic steatohepatitis (NASH), as a progressive form of Non-alcoholic fatty liver disease (NAFLD), poses a significant threat to human health as a prevalent and common condition, with a lack of safe and effective therapeutic options. However, the therapeutic effects and potential mechanisms of Lang Qing Ata (LQAtta) against NASH remain elusive. Materials and Methods The components of LQAtta were identified using Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry (UHPLC-MS/MS). Subsequently, we employed network construction and analysis approaches within the field of network pharmacology. By integrating known databases and target prediction algorithms, which encompassed database-based target prediction, protein-protein interaction networks, as well as Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, we unveiled the potential key targets and signaling pathways that these bioactive components might engage with. These discoveries were further validated in subsequent mouse models. An HFHC-induced NASH mouse model was used to validate the therapeutic effects and potential mechanisms of LQAtta on NASH. Results From the UHPLC-MS/MS analysis of LQAtta, a total of 1518 chemical components were identified, with 106 of them being absorbed into the bloodstream. Additionally, based on the acquisition of targets from both LQAtta and the NASH database, a total of 160 common targets were screened. KEGG enrichment analysis indicated that LQAtta may alleviate NASH by modulating pathways such as the Toll-like receptor signaling pathway, the NF-κB signaling pathway, and inflammation-related pathways. In vivo experimental results demonstrated that LQAtta could alleviate liver injury, steatosis, and inflammation induced by NASH, thereby slowing down the disease process. Additionally, LQAtta inhibited the expression and phosphorylation of NF-κB protein, playing a role in preventing NASH. Conclusion In this study, the combination of mass spectrometry analysis, network pharmacology, and animal experiments preliminarily elucidated the potential of LQAtta to treat NASH through NF-κB pathways.
Collapse
Affiliation(s)
- Shupei Li
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Hanlong Zhu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Qi Zhai
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Yu Hou
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Ya Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Haifeng Lan
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Mingzuo Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Ji Xuan
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
48
|
Pan LX, Tian W, Huang ZH, Li JR, Su JY, Wang QY, Fan XH, Zhong JH. Identification of a liver fibrosis and disease progression-related transcriptome signature in non-alcoholic fatty liver disease. Int J Biochem Cell Biol 2025; 180:106751. [PMID: 39909111 DOI: 10.1016/j.biocel.2025.106751] [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: 12/18/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD)-related liver fibrosis is closely associated with long-term outcomes of patients. This study aimed to establish a transcriptome signature to distinguish NAFLD patients with mild or advanced fibrosis and to monitor disease progression. Using least absolute shrinkage selection operator regression, we identified a signature of 11 hub genes by performing differential gene expression analysis in six bulk transcriptome profiles in the Gene Expression Omnibus database from liver fibrosis patients with different etiologies. Patients with NAFLD were classified using the 11-hub gene signature. Integrated analysis of signaling pathway enrichment, gene set enrichment, nearest template prediction, infiltration by hepatic stellate cells (HSCs) and pseudotime trajectories was performed on three bulk and one single-cell transcriptomes from NAFLD patients. Molecular features were compared between high-risk and low-risk groups, and associations were explored between hub gene signature expression and activation of HSCs. It was found that the high-risk group was characterized by advanced fibrosis stage, elevated risk for hepatocellular carcinoma, more significant infiltration by activated HSCs, as well as enrichment in signaling pathways related to fibrogenesis and NAFLD progression. Moreover, the 11-hub gene signature at the single-cell transcriptome level correlated with HSCs activation. In vitro experiments were conducted to evaluate the expression levels of hub genes, and IL6 was found to be up-regulated in activated LX-2 cells showing lipid accumulation. Our findings suggest that the 11-hub gene signature can help identify fibrosis stage in patients with NAFLD and detect disease progression. We also suggest that the role of IL6 in HSC activation deserves more investigation in the context of NAFLD.
Collapse
Affiliation(s)
- Li-Xin Pan
- Department of Microbiology, School of Preclinical Medicine, Center for Genomics and Personalized Medicine, Guangxi Medical University, Nanning 530021, China; Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Wei Tian
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China; School of Life Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhi-Hao Huang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jian-Rong Li
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jia-Yong Su
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Qiu-Yan Wang
- School of Life Sciences, Guangxi Medical University, Nanning 530021, China.
| | - Xiao-Hui Fan
- Department of Microbiology, School of Preclinical Medicine, Center for Genomics and Personalized Medicine, Guangxi Medical University, Nanning 530021, China.
| | - Jian-Hong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China; Key Laboratory of Early Prevention and Treatment for Regional High, Frequency Tumors (Guangxi Medical University), Ministry of Education, Nanning 530021, China; Guangxi Key Laboratory of Early Prevention and Treatment for Regional High, Frequency Tumors, Nanning 530021, China.
| |
Collapse
|
49
|
Dong R, Tian T, Luo Z, Chang D, Xue H, Qu S, Wang J, Shen C, Zhang R, Wang J. Cardiometabolic phenotype linked to fibrosis and mortality in metabolic dysfunction-associated steatotic liver disease. Nutr Metab Cardiovasc Dis 2025; 35:103797. [PMID: 39674720 DOI: 10.1016/j.numecd.2024.103797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS Patients with metabolic dysfunction-associated steatotic liver disease (MASLD) often manifest a combination of cardiometabolic risk factors of varying severity. The cardiometabolic phenotypes and their associations with advanced liver fibrosis and all-cause mortality among patients with MASLD warrant further investigation. METHODS AND RESULTS A total of 4209 and 1901 eligible participants were obtained from the National Health and Nutrition Examination Survey and included in the original and replication datasets, respectively. In the original dataset, three distinct and stable cardiometabolic phenotypes were identified using unsupervised cluster analyses, including mild cardiometabolic risk factor (MCMRF) phenotype, overweight combined with high diastolic blood pressure dominated (OCHBP) phenotype, and severe glucose and lipid metabolic dysfunction dominated (SGLMD) phenotype. The above phenotypes were subsequently replicated in the replication dataset, demonstrating similar characteristics. After adjusting for potential covariates, the results of logistic and Cox regression models showed that OCHBP and SGLMD phenotypes were significantly associated with higher odds of advanced liver fibrosis (OCHBP: OR = 4.37, 95 % CI: 1.54-12.35, P = 0.020; SGLMD: OR = 9.66, 95 % CI: 4.76-19.61, P = 0.002) and an increased risk of all-cause mortality (OCHBP: HR = 1.39, 95 % CI: 1.17-1.65, P < 0.001; SGLMD: HR = 2.51, 95 % CI: 1.86-3.40, P < 0.001) compared to the MCMRF phenotype. Moreover, the observed associations remained statistically significant in most subgroups, and a series of sensitivity analyses further confirmed the robustness of these findings. CONCLUSION Three heterogeneous cardiometabolic phenotypes were identified among participants with MASLD, showing significant associations with two critical outcomes. These novel phenotypes may be of great importance to precision medicine in MASLD.
Collapse
Affiliation(s)
- Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ting Tian
- Institute of Nutrition and Food Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Zhenghan Luo
- Department of Infectious Disease Prevention and Control, Huadong Research Institute for Medicine and Biotechniques, Nanjing, China
| | - Dongchun Chang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Hong Xue
- Department of Liver Disease, Third Affiliated Hospital of Nantong University, Nantong, China
| | - Sen Qu
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Jia Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Chao Shen
- Department of Immunization Program, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Ru Zhang
- School of Nursing and Midwifery, Jiangsu College of Nursing, Huaian, China.
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
50
|
Theel WB, de Jong VD, Castro Cabezas M, Grobbee DE, Jukema JW, Trompet S. Risk of cardiovascular disease in elderly subjects with obesity and liver fibrosis and the potential benefit of statin treatment. Eur J Clin Invest 2025; 55:e14368. [PMID: 39636216 PMCID: PMC11810556 DOI: 10.1111/eci.14368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Liver fibrosis progression is influenced by older age and cardiometabolic risk factors such as obesity and is associated with an increased risk of cardiovascular events. While statins may protect against cardiovascular complications, their effects in elderly individuals with obesity and liver fibrosis are unclear. METHOD The PROspective Study of Pravastatin in the Elderly at Risk (PROSPER) database was used to evaluate the effect of pravastatin on major adverse cardiovascular events in an elderly population (>70 years). Subjects were categorized by BMI: lean (<25 kg/m2), overweight (25-29.9 kg/m2) and obese (≥30 kg/m2). Liver fibrosis was assessed using the FIB-4 index: low risk (<2.0), intermediate risk (2.0-2.66) and high risk (≥2.67). Time-to-event data were analysed using the Cox proportional hazards model, adjusted for confounders and compared the placebo and pravastatin groups. RESULTS A total of 5.804 subjects were included. In the placebo group, the highest risk group (high FIB-4 and obesity) had a significantly higher hazard ratio for (non-)fatal stroke (HR 2.74; 95% CI 1.19-6.29) compared to the low FIB-4, lean BMI group. This risk disappeared in the same pravastatin group. Pravastatin did not affect other cardiovascular endpoints. All-cause mortality was significantly higher in subjects with lean weight and high FIB-4 on placebo (HR 1.88; 95% CI 1.14-3.11), but not on pravastatin (HR .58; 95% CI .28-1.20). CONCLUSION Elderly individuals with obesity and liver fibrosis are at higher risk for (non-)fatal stroke, which is reduced with pravastatin. Pravastatin also potentially lowers all-cause mortality in subjects with lean weight and liver fibrosis.
Collapse
Affiliation(s)
- Willy B. Theel
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Obesity Center CGGRotterdamThe Netherlands
| | - Vivian D. de Jong
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Manuel Castro Cabezas
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Julius ClinicalZeistThe Netherlands
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Diederick E. Grobbee
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Johan W. Jukema
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Stella Trompet
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Internal Medicine, Section of Gerontology & GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|