151
|
Petta S, Armandi A, Bugianesi E. Impact of PNPLA3 I148M on Clinical Outcomes in Patients With MASLD. Liver Int 2025; 45:e16133. [PMID: 39412170 PMCID: PMC11815615 DOI: 10.1111/liv.16133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a heterogenous clinical and histopathological entity, where multiple metabolic co-factors are intertwined with high interindividual variability. The impact and severity of each factor (including obesity and type 2 diabetes) define a systemic dysmetabolism that can lead to either advanced liver disease and its complication (including hepatocellular carcinoma and clinical events related to portal hypertension) or extrahepatic events: incident cardiovascular disease, chronic kidney disease and extrahepatic cancers. The balance between environmental factors and genetic susceptibility has unique implications in MASLD: the intermittent injury of metabolic co-factors, their fluctuation over time and their specific management, are counterbalanced by the presence of gene variants that can significantly impact the disease at multiple levels. The I148M variant in the PNPLA3 gene is the most investigated genetic susceptibility that induces a more severe steatohepatitis, enhanced fibrogenesis and can shape the incidence of long-term clinical events regardless of, or worsened by, other metabolic risk factors. METHODS AND RESULTS In this review, we will summarise the updated evidence on the natural history of MASLD accounting for classical metabolic risk factors, the role of PNPLA3 in clinical sub-phenotyping (e.g., 'lean MASLD'), impact on disease severity and fibrosis progression, as well as its role for prognostication, alone or in combination with non-invasive tools into polygenic risk scores.
Collapse
Affiliation(s)
- Salvatore Petta
- Sezione di Gastroenterologia, Di.Bi.M.I.SUniversity of PalermoPalermoItaly
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical SciencesUniversity of TurinTurinItaly
| |
Collapse
|
152
|
Luukkonen PK. Hidden metabolic effects of acetyl-CoA carboxylase inhibition. J Hepatol 2025; 82:544-545. [PMID: 39681499 DOI: 10.1016/j.jhep.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Affiliation(s)
- Panu K Luukkonen
- Department of Internal Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Abdominal Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
153
|
Li M, Yu H, Wan S, Hu F, Luo Q, Gong W. The fibrosis investigating navigator in diabetes (FIND): A tool to predict liver fibrosis risk in subjects with diabetes. Diabetes Obes Metab 2025; 27:1184-1197. [PMID: 39638772 PMCID: PMC11802394 DOI: 10.1111/dom.16109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Type 2 diabetes increases the risk of cirrhosis and liver cancer. Noninvasive and early assessment of liver fibrosis is essential. We aimed to develop a score to aid in the initial assessment of liver fibrosis in the diabetic population. METHODS A fibrosis investigating navigator in diabetes (FIND) score was developed and validated in the NHANES dataset (2017-2020). Fibrosis was defined as a liver stiffness measurement (LSM) ≥8.0 kPa. The diagnostic accuracies of FIB-4, NFS, LiverRisk, steatosis-associated fibrosis estimator (SAFE) and metabolic dysfunction-associated fibrosis (MAF-5) were compared. FIND was also externally validated in various liver diseases via biopsy as a reference in an Asian centre between 2016 and 2020. Finally, we examined the prognostic implications of the FIND index utilizing data from the UK Biobank cohort (2006-2010). RESULTS The FIND score model yielded an AUROC of 0.781 for the prediction of an LSM ≥8 kPa in the validation set, which was consistently greater than that of other available models (all p < 0.05). In the whole NHANES dataset, the 85% sensitivity cut-off of 0.16 corresponded to a NPV of 91.9%, whereas the 85% specificity cut-off of 0.31 corresponded to a PPV of 50.6%. FIND displayed overall accuracies similar to those of the other models in staging fibrosis stages, with biopsy used as a reference. In the UK Biobank cohort, FIND >0.31 was associated with an increased risk of all-cause and liver-related mortality in the diabetic population in adjusted models (HR, 1.75; 95% CI, 1.62-1.89; HR, 23.59; 95% CI, 13.67-40.69). CONCLUSIONS In diabetes patients, the novel FIND score performs well in identifying subjects at risk of liver fibrosis and predicting all-cause and liver-related mortality.
Collapse
Affiliation(s)
- Mingkai Li
- Department of Gastroenterology, Shenzhen HospitalSouthern Medical UniversityShenzhenPeople's Republic of China
| | - Hongsheng Yu
- Department of GastroenterologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouPeople's Republic of China
| | - Sizhe Wan
- Department of Gastroenterology, Shenzhen HospitalSouthern Medical UniversityShenzhenPeople's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, Public Health collegeShenzhen University Medical SchoolShenzhenPeople's Republic of China
| | - Qingtian Luo
- Department of Gastroenterology and Endoscopy CenterShenzhen Nanshan People’s Hospital, the 6th Affiliated Hospital of Shenzhen University Medical SchoolShenzhenPeople's Republic of China
| | - Wei Gong
- Department of Gastroenterology, Shenzhen HospitalSouthern Medical UniversityShenzhenPeople's Republic of China
| |
Collapse
|
154
|
Chen VL, Vespasiani‐Gentilucci U. Integrating PNPLA3 into clinical risk prediction. Liver Int 2025; 45:e16103. [PMID: 39282715 PMCID: PMC11815612 DOI: 10.1111/liv.16103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 02/05/2025]
Abstract
The PNPLA3-rs738409-G variant was the first common variant associated with hepatic fat accumulation and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Nevertheless, to date, the clinical translation of this discovery has been minimal because it has not yet been clearly demonstrated where the genetic information may play an independent and additional role in clinical risk prediction. In this mini-review, we will discuss the most relevant evidence regarding the potential integration of the PNPLA3 variant into scores and algorithms for liver disease diagnostics and risk stratification, specifically focusing on MASLD but also extending to liver diseases of other etiologies. The PNPLA3 variant adds little in diagnosing the current state of the disease, whether in terms of presence/absence of metabolic dysfunction-associated steatohepatitis or the stage of fibrosis. While it can play an important role in prediction, allowing for the early definition of risk profiles that enable tailored monitoring and interventions over time, this is most valuable when applied to populations with relatively high pre-test probability of having significant fibrosis based on either non-invasive tests (e.g. Fibrosis-4) or demographics (e.g. diabetes). Indeed, in this context, integrating FIB4 with the PNPLA3 genotype can refine risk stratification, though there is still no evidence that genetic information adds to liver stiffness determined by elastography. Similarly, in patients with known liver cirrhosis, knowing the PNPLA3 genotype can play a role in predicting the risk of hepatocellular carcinoma, while more doubts remain about the risk of decompensation.
Collapse
Affiliation(s)
- Vincent L. Chen
- Division of Gastroenterology and Hepatology, Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Umberto Vespasiani‐Gentilucci
- Research Unit of HepatologyUniversità Campus Bio‐Medico di RomeRomeItaly
- Hepatology and Clinical Medicine UnitFondazione Policlinico Universitario Campus‐Biomedico di RomaRomeItaly
| |
Collapse
|
155
|
Pugliese N, Ravaioli F. Generative Artificial Intelligence in Nutrition: A Revolution in Accessibility and Personalization. J Nutr 2025; 155:667-668. [PMID: 39880170 DOI: 10.1016/j.tjnut.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Affiliation(s)
- Nicola Pugliese
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Rozzano, Milan, Italy; Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Federico Ravaioli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
156
|
Lin S, Dong L, De Nardo W, Leeming MG, Cheng Z, Williamson NA, Watt MJ, Montgomery MK. Long-lasting recombinant HEXA treatment improves hepatic steatosis and glycemic control in mild, but not severe, metabolic dysfunction-associated steatohepatitis. Am J Physiol Endocrinol Metab 2025; 328:E377-E394. [PMID: 39925140 DOI: 10.1152/ajpendo.00359.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/24/2024] [Accepted: 01/12/2025] [Indexed: 02/11/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing at an alarming rate. To date, only one therapy has been provisionally approved for the treatment of MASH and liver fibrosis, and novel strategies are urgently needed. In addition, the frequent coexistence of MASH and type 2 diabetes has further intensified interest in devising comprehensive therapies to simultaneously tackle both diseases. We have recently shown that increasing hepatic and/or circulating levels of hexosaminidase A (HEXA), a lysosomal enzyme that remodels GM2 to GM3 gangliosides within lipid rafts, offers therapeutic benefits for metabolic dysfunction-associated steatotic liver disease (MASLD) and type 2 diabetes. Taking advantage of the MUP-uPA mouse model of MASH, including both wild-type (WT) mice with mild MASH and MUP-uPA mice with severe MASH and fibrosis, we show that biweekly treatment with a long-lasting HEXA-FC analog improves features of MASLD, including hepatic steatosis and hepatocyte ballooning, in mice with mild MASH, as well as glycemic control across both mouse models. Mechanistically, HEXA-FC enhances hepatic fatty acid oxidation and peripheral glucose disposal while not impacting endogenous glucose production. Together, these outcomes suggest that while HEXA-FC treatment may offer therapeutic benefits in mild MASH and insulin resistance, it is ineffective against severe MASH and liver fibrosis.NEW & NOTEWORTHY The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) and type 2 diabetes is increasing. Here, we show that chronic FC-HEXA recombinant protein treatment reduces hepatic lipid accumulation and improves blood glucose control in mice with mild MASH and insulin resistance.
Collapse
Affiliation(s)
- Sihan Lin
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Li Dong
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - William De Nardo
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhili Cheng
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas A Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
157
|
Hoevelmann J, Markwirth P, Tokcan M, Haring B. What's new in heart failure? March 2025. Eur J Heart Fail 2025; 27:409-411. [PMID: 40159398 DOI: 10.1002/ejhf.3639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Julian Hoevelmann
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Philipp Markwirth
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
| | - Mert Tokcan
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
| | - Bernhard Haring
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
158
|
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
|
159
|
Antwi MB, Lefere S, Clarisse D, Koorneef L, Heldens A, Onghena L, Decroix K, Fijalkowska D, Thommis J, Hellemans M, Hoorens A, Geerts A, Devisscher L, De Bosscher K. PPARα-ERRα crosstalk mitigates metabolic dysfunction-associated steatotic liver disease progression. Metabolism 2025; 164:156128. [PMID: 39743041 DOI: 10.1016/j.metabol.2024.156128] [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: 10/15/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD), the most prevalent liver disease worldwide, continues to rise. More effective therapeutic strategies are urgently needed. We investigated how targeting two key nuclear receptors involved in hepatic energy metabolism, peroxisome proliferator-activated receptor alpha (PPARα) and estrogen-related receptor alpha (ERRα), ameliorates MASLD. METHODS The PPARα agonist pemafibrate and/or ERRα inverse agonist C29 were administered in a short- and long-term Western diet plus fructose model, and a diabetic-background streptozotocin-Western diet model (STZ-WD). Liver and adipose tissue morphology, histological samples, serum metabolites, RNA and protein levels were analysed and scanning electron microscopy was performed. In addition, we performed cell-based assays and immunohistochemistry and immunofluorescence stainings with light and super-resolution confocal microscopy of healthy, MASLD and MASH human livers. RESULTS The ligand combinations' efficacy was highlighted by reduced liver steatosis across all mouse models, alongside improvements in body weight, inflammation, and fibrosis in both long-term models. Additionally, tumour formation was prevented in the STZ-WD mice model. Cell-based assays demonstrated that ERRα inhibits PPARα's activity, explaining why ERRα blockage improves inflammatory and lipid metabolism gene profiles and enhances lipid-lowering effects. Complementary RNA sequencing and shotgun proteomics, combined with enrichment analysis, jointly identified downregulated serum amyloid A1/A2 as essential components underlying the combination treatment's effectiveness. MASLD/MASH patient livers showed reduced PPARα and increased ERRα levels supporting disrupted NR crosstalk in the hepatocyte nucleus. CONCLUSION Our study supports that dual nuclear receptor targeting, which simultaneously increases PPARα and diminishes ERRα activity, may represent a viable novel strategy against MASLD. IMPACT AND IMPLICATIONS Our research introduces a novel therapeutic strategy against MASLD by simultaneously increasing PPARα activity while diminishing ERRα activity. With PPARα agonists already tested in phase III clinical trials, ERRα ligands/modulators need further (clinical) development to make our findings applicable to both MASLD patients and physicians.
Collapse
Affiliation(s)
- Milton Boaheng Antwi
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium; Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Sander Lefere
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Lisa Koorneef
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Anneleen Heldens
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Louis Onghena
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Kylian Decroix
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Daria Fijalkowska
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Jonathan Thommis
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Madeleine Hellemans
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Anja Geerts
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Lindsey Devisscher
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Karolien De Bosscher
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium.
| |
Collapse
|
160
|
Abdel Monem MS, Adel A, Abbassi MM, Abdelaziz DH, Hassany M, Raziky ME, Sabry NA. Efficacy and safety of dapagliflozin compared to pioglitazone in diabetic and non-diabetic patients with non-alcoholic steatohepatitis: A randomized clinical trial. Clin Res Hepatol Gastroenterol 2025; 49:102543. [PMID: 39884573 DOI: 10.1016/j.clinre.2025.102543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a serious end-stage spectrum of non-alcoholic fatty liver disease (NAFLD) with associated high risk of hepatic and extrahepatic complications. Several studies showed the significant beneficial effect of dapagliflozin on body composition, hepatic and metabolic parameters on NAFLD/NASH patients. The study aimed to investigate the efficacy and safety of dapagliflozin in both diabetic and non-diabetic biopsy-proven NASH patients; compared to pioglitazone. METHODS This was a four-group, prospective, randomized, parallel, open label study in which 100 biopsy-proven NASH patients were selected, stratified to diabetics and non-diabetics and randomized with 1:1 allocation to either 30 mg pioglitazone or 10 mg dapagliflozin, once daily for 24 weeks. Histological evaluation, anthropometric measures, hepatic, metabolic biochemical markers, fibrosis non-invasive markers, quality of life (QOL) and medications adverse events were examined. RESULTS Dapagliflozin showed a comparable histological effect to pioglitazone in both diabetic and non-diabetic patients (P>0.05). As assessed by transient elastography, it also showed a comparable effect on liver fibrosis grade improvement from baseline in diabetics (P=0.287) versus a significant superiority in non-diabetics (P=0.018). Dapagliflozin showed a significant superiority in all anthropometric measures (P<0.001) and QOL (P<0.05) among both diabetics and non-diabetics. There was a significant interaction between interventions and diabetes status on change from baseline of hepatic and metabolic panel collectively (P=0.023) in favor to dapagliflozin among diabetics. CONCLUSION Compared to pioglitazone, dapagliflozin had a comparable effect histologically, superior effect biochemically among diabetics and superior effect on liver fibrosis, steatosis and insulin resistance among non-diabetics. TRIAL REGISTRATION The study was registered on clinicaltrials.gov, identifier number NCT05254626.
Collapse
Affiliation(s)
- Mona S Abdel Monem
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Abdulmoneim Adel
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maggie M Abbassi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Doaa H Abdelaziz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia/Department of Clinical Pharmacy, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Mohamed Hassany
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maissa El Raziky
- Endemic Medicine and Hepatogastroenterology, Faculty of Medicine, Cairo University, Egypt.
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
161
|
Sakane S, Hikita H. Revisiting the relationship between thyroid function and metabolic dysfunction-associated steatotic liver disease in the era of resmetirom. J Gastroenterol 2025; 60:389-391. [PMID: 39847088 DOI: 10.1007/s00535-025-02214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Affiliation(s)
- Sadatsugu Sakane
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
162
|
Ronzoni L, Pelusi S, Moretti V, Malvestiti F, Eidgah Torghabehei H, Jamialahmadi O, Rondena J, Bianco C, Periti G, Filippo MRD, Romeo S, Prati D, Valenti L. Diagnostic Uptake of Targeted Sequencing in Adults With Steatotic Liver Disease and a Suspected Genetic Contribution. Liver Int 2025; 45:e70010. [PMID: 39945383 PMCID: PMC11822878 DOI: 10.1111/liv.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND AND AIMS In patients with steatotic liver diseases (SLD), genetic factors may account for severe liver involvement despite mild or absence of triggering factors or a strong family history. Aim of this study was to examine the diagnostic uptake of targeted sequencing (TS), covering both coding and non-coding regions, of a broad panel of 82 liver and lipid metabolism genes in patients with unexplained SLD. METHODS We enrolled 49 adult patients with SLD and a suspected genetic contribution. Genetic variants were detected through a customised TS panel, whereas the contribution of common genetic variation to the individual susceptibility to SLD was captured by a polygenic risk score (SLD-PRS). RESULTS A diagnosis of rare Mendelian disorder was established in 11 patients (22%), independently of age or family history. Rare variants possibly contributing to clinical phenotype were detected in additional 29 patients (59%). Increased SLD-PRS values were detected in 17 patients (35%), enabling an increase in diagnostic uptake of 24%, especially in those without a strong family history (p = 0.03). Genetic diagnosis allowed refinement of clinical management in 23 (47%) patients. CONCLUSIONS The diagnostic uptake of TS was 22% for Mendelian disorder and 59% for possible contribution to clinical phenotype in selected adult patients with SLD. Evaluation of common variants, as captured by SLD-PRS, yields complementary information increasing the overall utility of the genetic examination.
Collapse
Grants
- 777377 Innovative Medicines Initiative 2 joint undertaking of European Union's Horizon 2020 research and innovation program and EFPIA European Union (EU) Program Horizon 2020 for the project LITMUS
- Gilead Sciences Inc.
- 101016726-REVEAL The European Union, H2020-ICT-2018-20/H2020-ICT-2020-2 program "Photonics"
- Italian ministry of Research (MUR) PNRR - M4 - C2 "National Center for Gene Therapy and Drugs based on RNA Technology" CN3, Spoke 4
- 101096312 The European Union, HORIZON-MISS-2021-CANCER-02-03 program "Genial"
- RF-2016-02364358 The Italian Ministry of Health (Ministero della Salute), Ricerca Finalizzata 2016
- Italian ministry of Research (MUR) PRIN 2022
- PNRR-MAD-2022-12375656 The Italian Ministry of Health, Ricerca Finalizzata PNRR 2022
- RF-2021-12373889 The Italian Ministry of Health, Ricerca Finalizzata 2021
- PR-0361 Fondazione Patrimonio Ca' Granda, "Liver BIBLE"
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Ricerca Corrente
- Innovative Medicines Initiative 2 joint undertaking of European Union’s Horizon 2020 research and innovation program and EFPIA European Union (EU) Program Horizon 2020 for the project LITMUS
- The European Union, H2020‐ICT‐2018‐20/H2020‐ICT‐2020‐2 program “Photonics”
- Italian ministry of Research (MUR) PNRR ‐ M4 ‐ C2 “National Center for Gene Therapy and Drugs based on RNA Technology” CN3, Spoke 4
- The European Union, HORIZON‐MISS‐2021‐CANCER‐02‐03 program “Genial”
- The Italian Ministry of Health (Ministero della Salute), Ricerca Finalizzata 2016
- Italian ministry of Research (MUR) PRIN 2022
- The Italian Ministry of Health, Ricerca Finalizzata PNRR 2022
- The Italian Ministry of Health, Ricerca Finalizzata 2021
- Fondazione Patrimonio Ca’ Granda, “Liver BIBLE”
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Ricerca Corrente
Collapse
Affiliation(s)
- Luisa Ronzoni
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Serena Pelusi
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Vittoria Moretti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Francesco Malvestiti
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanItaly
| | - Hadi Eidgah Torghabehei
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden
| | - Jessica Rondena
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Cristiana Bianco
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Giulia Periti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Maria Rosaria De Filippo
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
- Clinical Nutrition Unit, Department of Medical and Surgical SciencesUniversity Magna GraeciaCatanzaroItaly
| | - Daniele Prati
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| | - Luca Valenti
- Precisione Medicine Lab, Biological Resource Center and Department of Transfusion MedicineFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanItaly
- Omic Sciences Lab, Scientific DirectionFondazione IRCCS ca' Granda Ospedale Maggiore Policlinico MilanoMilanItaly
| |
Collapse
|
163
|
Cooper KM, Patel AK, Zammitti CA, Murchie E, Colletta A, Devuni D. Statin Therapy is Associated With Lower Risk of Mortality Among Liver Transplant Candidates With Metabolic Dysfunction-associated Steatohepatitis. J Clin Exp Hepatol 2025; 15:102427. [PMID: 39678071 PMCID: PMC11638578 DOI: 10.1016/j.jceh.2024.102427] [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: 08/10/2024] [Accepted: 10/09/2024] [Indexed: 12/17/2024] Open
Abstract
Background Statin therapy is historically underutilized in patients with chronic liver disease. There is increasing evidence to support the use of statins in patients with cirrhosis, though data in decompensated patients are limited. The primary aim of this study was to evaluate the association between statin use and mortality in patients with advanced liver disease, comparing MASH and non-MASH cirrhosis. Methods This single-center retrospective cohort study included patients undergoing liver transplant (LT) evaluation at a large quaternary care center. Patients were categorized by etiology as metabolic dysfunction-associated steatohepatitis (MASH) or non-MASH cirrhosis. Statin use was defined as having an active prescription at the time of LT evaluation. The association between statin use and mortality was evaluated using multivariable Cox proportional hazard regression. Results The study included 623 patients; 24% had MASH cirrhosis and 20% were prescribed a statin. Statin users were older, had a higher BMI, and were more likely to have coronary artery disease. At the end of the study, statin use was associated with lower mortality among MASH patients (16% vs. 35%, P = 0.010) and higher mortality among non-MASH patients (31% vs. 19%, P = 0.066). After controlling for age (HR 1.05, 95% CI: 1.00-1.10, P = 0.039), MELD-Na (HR: 1.07, 95% CI: 1.04-1.11, P < 0.001), BMI (HR: 1.09, 95% CI: 1.05-1.14, P < 0.001), and CAD (HR: 1.20, 95% CI: 0.54-2.69, P = 0.653), statin use conferred a 53% lower risk of death compared with no statin use in patients with MASH cirrhosis (HR: 0.47, 95% CI: 0.22-0.98, P = 0.043). Conclusions Statin use was associated with reduced mortality in patients with decompensated MASH cirrhosis undergoing LT evaluation, but increased mortality in those with non-MASH cirrhosis, particularly those with high-MELD-Na. These findings underscore the importance of reviewing individual patient characteristics and disease etiology when considering the benefits of statin therapy in patients with cirrhosis.
Collapse
Affiliation(s)
| | - Ami K. Patel
- UMass Chan Medical School, Department of Medicine, USA
| | | | - Ellen Murchie
- UMass Chan Medical School, Department of Medicine, USA
| | - Alessandro Colletta
- UMass Chan Medical School, Department of Medicine, USA
- UMass Chan Medical School, Division of Gastroenterology and Hepatology, USA
| | - Deepika Devuni
- UMass Chan Medical School, Department of Medicine, USA
- UMass Chan Medical School, Division of Gastroenterology and Hepatology, USA
| |
Collapse
|
164
|
Alkhouri N, Charlton M, Gray M, Noureddin M. The pleiotropic effects of glucagon-like peptide-1 receptor agonists in patients with metabolic dysfunction-associated steatohepatitis: a review for gastroenterologists. Expert Opin Investig Drugs 2025; 34:169-195. [PMID: 40016997 DOI: 10.1080/13543784.2025.2473062] [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: 09/27/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Glucagon-like peptide-1 receptor agonists (GLP-1RAs), and dual GLP-1/glucose-dependent insulinotropic peptide (GIP) or glucagon receptor agonists have emerged as promising agents to treat metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). Although the beneficial effects of GLP-1RAs on glycemic control and weight are well-established, clinicians may be unfamiliar with other potential benefits of this class. AREAS COVERED We examined the pleiotropic effects of GLP-1RAs and how they relate to gastroenterologists for MASLD/MASH treatment. Our narrative review of English articles included four GLP-1RAs (subcutaneous semaglutide, liraglutide, dulaglutide, and efpeglenatide), a dual GLP-1/GIP agonist (tirzepatide), a dual GLP-1/glucagon receptor agonist (survodutide), MASLD/MASH, related disorders, clinical management, treatment outcomes and landscape. EXPERT OPINION In Phase I - III trials, GLP-1RAs are associated with clinically relevant hepatic improvements including MASH resolution, liver fat reduction, and preventing worsening fibrosis. Effects on cardiometabolic parameters align with type 2 diabetes/obesity Phase III data, comprising substantial improvements in glycemic, weight, and cardiovascular outcomes. Promising data also suggest benefits in common comorbidities, including obstructive sleep apnea, polycystic ovary syndrome, chronic kidney disease, and heart failure with preserved ejection fraction.GLP-1RAs represent a valuable pharmacotherapeutic option for gastroenterologists managing individuals with MASLD/MASH and cardiometabolic comorbid conditions.
Collapse
Affiliation(s)
- Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Phoenix, AZ, USA
| | - Michael Charlton
- Transplant Institute, Center for Liver Diseases, University of Chicago Biological Sciences, Chicago, IL, USA
| | - Meagan Gray
- Division of Gastroenterology and Hepatology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mazen Noureddin
- Houston Methodist Hospital, Houston Research Institute, Houston, TX, USA
| |
Collapse
|
165
|
Vrsaljko N, Radmanic Matotek L, Zidovec-Lepej S, Vince A, Papic N. The Impact of Steatotic Liver Disease on Cytokine and Chemokine Kinetics During Sepsis. Int J Mol Sci 2025; 26:2226. [PMID: 40076848 PMCID: PMC11900930 DOI: 10.3390/ijms26052226] [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: 01/16/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been linked with sepsis outcomes. However, the immune mechanisms by which MASLD aggravates sepsis severity are unknown. This prospective cohort study aimed to analyze serum cytokine and chemokine kinetics in patients with MASLD and community-acquired sepsis. Out of the 124 patients, 68 (55%) were diagnosed with MASLD. There were no differences in age, sex, comorbidities, baseline sepsis severity, or etiology between the groups. Serum concentrations of 27 cytokines and chemokines on admission and day 5 of hospitalization were analyzed using a multiplex bead-based assay. Patients with MASLD had significantly higher serum concentrations of IL17A, IL-23, IL-33, CXCL10 and TGF-β1. Different cytokine kinetics were observed; patients with MASLD had a decrease in IL-10, IL-23, CXCL10 and TGF-β1, and an increase in IL-33, CXCL5 and CXCL1 on day 5. In the non-MASLD group, there was a decrease in IFN-γ, IL-6, IL-23 and CCL20, and an increase in CCL11 and CXCL5. While TGF-β1 significantly increased in non-MASLD, in MASLD, it decreased on day 5. Kinetics of TGF- β1 and CCL11 were associated with mortality in patients with MASLD. In conclusion, MASLD is linked with distinct cytokine and chemokine profiles during sepsis.
Collapse
Affiliation(s)
- Nina Vrsaljko
- Emergency Infectious Diseases Department, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia;
| | - Leona Radmanic Matotek
- Department for Immunological and Molecular Diagnostics, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia; (L.R.M.); (S.Z.-L.)
| | - Snjezana Zidovec-Lepej
- Department for Immunological and Molecular Diagnostics, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia; (L.R.M.); (S.Z.-L.)
| | - Adriana Vince
- Department for Viral Hepatitis, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia;
| | - Neven Papic
- Department for Viral Hepatitis, University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia;
- Department for Infectious Diseases, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
166
|
Deb R, Goswami S, Sengupta N, Baidya A, Khare VR, Datta J, Das M, Ray D. Redefining Liver Fibrosis Risk Assessment in Indians with Type 2 Diabetes: New FIB-4 Score Cutoff for Optimizing Sequential Assessment with Transient Elastography. Indian J Endocrinol Metab 2025; 29:237-241. [PMID: 40416452 PMCID: PMC12101750 DOI: 10.4103/ijem.ijem_457_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/12/2025] [Accepted: 02/27/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Liver fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD) is strongly related to hepatic and extrahepatic outcomes. Clinically Significant Liver Fibrosis (CSLF) screening using FIB-4 score is mandated in all T2D patients. The existing FIB-4 cutoff of 1.3 is derived from Western studies and could differ for Indians. Hence, we aimed to determine the FIB-4 cutoff to rule out Transient Elastography (TE) proven CSLF among Indians with T2D. Methods 551 individuals with T2D underwent laboratory tests for FIB-4 calculation and transient elastography (TE) to detect CSLF defined as Liver Stiffness Measurement (LSM) ≥ 8kPa. The Receiver Operative Characteristic (ROC) curve was used to determine the optimum cutoff value of FIB4 to rule out CSLF. Results 129 (23.4%) of 551 T2D patients in our cohort had CSLF. We found that a FIB-4 of 1.5 rules out CSLF with a sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and Diagnostic Accuracy (DA) of 82.9%, 79.9%, 55.7%, 93.8%, and 80.6%, respectively, compared with a FIB4 of 1.3 which has values of 91.5%, 67.3%, 46.1%, 96.2%, and 72.9%, respectively. Conclusion A FIB-4 cutoff of 1.5 rather than 1.3 is suggested for Indian subjects with T2DM and needs to be validated in further large multicenter prospective studies, preferably with histopathology as the gold standard.
Collapse
Affiliation(s)
- Rajat Deb
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Soumik Goswami
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Nilanjan Sengupta
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Arjun Baidya
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Vibhu R. Khare
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Joydip Datta
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Mousumi Das
- Department of Biochemistry, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Debes Ray
- Department of Biochemistry, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| |
Collapse
|
167
|
Anastasopoulos NA, Barbouti A, Goussia AC, Christodoulou DK, Glantzounis GK. Exploring the Role of Metabolic Hyperferritinaemia (MHF) in Steatotic Liver Disease (SLD) and Hepatocellular Carcinoma (HCC). Cancers (Basel) 2025; 17:842. [PMID: 40075688 PMCID: PMC11899477 DOI: 10.3390/cancers17050842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The increasing prevalence of the spectrum of Steatotic Liver Disease (SLD), including Metabolic-Associated Steatotic Liver Disease (MASLD), Metabolic-Associated Steatohepatitis (MASH), and progression to Cirrhosis and Hepatocellular Carcinoma (HCC) has led to intense research in disease pathophysiology, with many studies focusing on the role of iron. Iron overload, which is often observed in patients with SLD as a part of metabolic hyperferritinaemia (MHF), particularly in the reticuloendothelial system (RES), can exacerbate steatosis. This imbalance in iron distribution, coupled with a high-fat diet, can further promote the progression of SLD by means of oxidative stress triggering inflammation and activating hepatic stellate cells (HSCs), therefore leading to fibrosis and progression of simple steatosis to the more severe MASH. The influence of iron overload in disease progression has also been shown by the complex role of ferroptosis, a type of cell death driven by iron-dependent lipid peroxidation. Ferroptosis depletes the liver's antioxidant capacity, further contributing to the development of MASH, while its role in MASH-related HCC is potentially linked to alternations in the tumour microenvironment, as well as ferroptosis resistance. The iron-rich steatotic hepatic environment becomes prone to hepatocarcinogenesis by activation of several pro-carcinogenic mechanisms including epithelial-to-mesenchymal transition and deactivation of DNA damage repair. Biochemical markers of iron overload and deranged metabolism have been linked to all stages of SLD and its associated HCC in multiple patient cohorts of diverse genetic backgrounds, enhancing our daily clinical understanding of this interaction. Further understanding could lead to enhanced therapies for SLD management and prevention.
Collapse
Affiliation(s)
- Nikolaos-Andreas Anastasopoulos
- HPB Unit, Department of Surgery, University Hospital of Ioannina, 45110 Ioannina, Greece
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Anna C. Goussia
- Department of Pathology, University Hospital of Ioannina, 45110 Ioannina, Greece
| | | | | |
Collapse
|
168
|
Lian LY, Huang CX, Chen QF, Zhou XD. Heart failure with preserved ejection fraction and metabolic dysfunction-associated steatotic liver disease: Twin challenges, one metabolic solution. World J Cardiol 2025; 17:103845. [PMID: 40061276 PMCID: PMC11886398 DOI: 10.4330/wjc.v17.i2.103845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) has exceeded HF with reduced ejection fraction (HFrEF), becoming the most common type of HF. Unlike HFrEF, HFpEF is primarily a chronic low-grade inflammatory process closely associated with metabolic disorders. The coexistence of HFpEF and metabolic dysfunction-associated steatotic liver disease (MASLD) presents significant clinical challenges due to shared metabolic pathophysiology and complex interplay. Management strategies for HFpEF and MASLD remain challenging. Sodium-glucose cotransporter 2 inhibitors have shown benefits in managing both conditions. Additionally, glucagon-like peptide-1 receptor agonists are being actively investigated for their potential benefits, particularly in MASLD. A comprehensive, patient-centered approach that combines metabolic and cardiovascular care is essential for improving outcomes in patients with HFpEF and MASLD, addressing the global metabolic health challenges.
Collapse
Affiliation(s)
- Li-You Lian
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chen-Xiao Huang
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Qin-Fen Chen
- Department of Physical Examination Medical Care Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Xiao-Dong Zhou
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China.
| |
Collapse
|
169
|
Cheng Y, Yang R, Jia Y, Zhou Y, Yao Y, Shen C, Li D, Zeng R, Wan Z, Zhao Q, Jiang L, Liao X. The association of chronic pain, painkiller use, and potential mediators with liver fat content. Sci Rep 2025; 15:6688. [PMID: 39994347 PMCID: PMC11850618 DOI: 10.1038/s41598-025-89496-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Excessive accumulation of liver fat content (LFC) is a pathological manifestation of steatotic liver diseases. This study aims to investigate the relationship between chronic pain and LFC development. In the UK Biobank, chronic pain sites were collected via questionnaire, while LFC was measured by magnetic resonance imaging and quantified by Proton Density Fat Fraction (PDFF). During the median follow-up of 10.5 (4.0-17.8) years, in 39,437 individuals, neck/shoulder, back, stomach/abdominal, knee, and general pain achieved significant arithmetic means difference of 0.02, 0.02, 0.04, 0.02, and 0.15 in PDFF (P < 0.05) using multivariable linear regression models. There was a significant dose-effect for number of pain sites and PDFF (P < 0.001). Additionally, the link between pain sites and PDFF was much stronger in aspirin users than non-users, while steroids had the reverse effect (P for interaction < 0.05). C-reactive protein, sleep, diet, and depression were proved to mediated 8.41%, 13.3%, 6.6%, and 23.0% of the relationship, respectively. In conclusion, there were quantified differences in the relationship between chronic pain and LFC. For chronic pain patients with potential liver health issues, aspirin may be prioritized as an analgesic option due to its potential protective benefits, whereas steroid medications should be avoided.
Collapse
Affiliation(s)
- Yu Cheng
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yiheng Zhou
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Yao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Can Shen
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine, Disaster Medical Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zeng
- Department of Cardiology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi Wan
- Department of Emergency Medicine, Disaster Medical Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China
| | - Lihua Jiang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China.
- Department of health policy and management, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
170
|
Åström H, Takami Lageborn C, Hagström H. Psychosocial risks in metabolic dysfunction-associated steatotic liver disease. Expert Rev Gastroenterol Hepatol 2025:1-18. [PMID: 39953908 DOI: 10.1080/17474124.2025.2468297] [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: 12/30/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) is increasingly becoming more prevalent in the general population. MASLD is more common in persons with low socioeconomic status (SES), yet little is known about the psychosocial challenges associated with this disease, and clinical recommendations on how to approach psychosocial challenges are lacking. AREAS COVERED A PubMed search using the search terms MASLD, psychosocial risks, stigmatization, psychiatric comorbidities (i.e. depression, bipolar disorder, psychosis, attention deficit hyperactivity disorder, and substance abuse), SES, quality of life (QoL), over the past 20 years (2004-2024) was performed. EXPERT OPINION Persons with MASLD often experience psychosocial adversities that may be expressed as lower SES, high prevalence of depression, and reduced QoL. Knowledge gaps remain regarding the association between severe mental disorders (e.g. psychosis and bipolar disorders). Timely detection and treatment of MASLD in persons with psychosocial risks may require attention and cross-field collaboration. Studies on QoL in persons with MASLD differ in methodology which makes formal comparisons difficult. Psychosocial adversity may be a barrier to lifestyle modifications, which remain the cornerstone of MASLD management. Guidelines on how to address psychosocial adversities in a clinical setting are warranted to improve outcomes and decrease further multimorbidity.
Collapse
Affiliation(s)
- Hanne Åström
- 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
| |
Collapse
|
171
|
Seagle HM, Akerele AT, DeCorte JA, Hellwege JN, Breeyear JH, Kim J, Levin M, Khodurksy S, Bress A, Lee K, Meiler J, Gill D, Lee JS, Heberer K, Miller DR, Reaven P, Chang KM, Lynch JA, Khankari NK, Shuey MM, Edwards TL, Vujkovic M. Genomics-Informed Drug Repurposing Strategy Identifies Novel Therapeutic Targets for Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25321035. [PMID: 40034783 PMCID: PMC11875238 DOI: 10.1101/2025.02.18.25321035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identification of drug-repurposing targets with genetic and biological support is an economically and temporally efficient strategy for improving treatment of diseases. We employed a cross-disciplinary approach to identify potential treatments for metabolic dysfunction associated steatotic liver disease (MASLD) using humans as a model organism. We identified 212 putative causal genes associated with MASLD using data from a large multi-ancestry genetic association study, of which 158 (74.5%) are novel. From this set we identified 57 genes that encode for druggable protein targets, and where the effects of increasing genetically predicted gene expression on MASLD risk align with the function of that drug on the protein target. These potential targets were then evaluated for evidence of efficacy using Mendelian randomization, pathway analysis, and protein structural modeling. Using these approaches, we present compelling evidence to suggest activation of FADS1 by icosopent ethyl as well as S1PR2 by fingolimod could be promising therapeutic strategies for MASLD.
Collapse
Affiliation(s)
- Hannah M Seagle
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, Georgia, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexis T Akerele
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- School of Graduate Studies and Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Joseph A DeCorte
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jacklyn N Hellwege
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Joseph H Breeyear
- Biostatistics and Computational Biology Branch, National Institute for Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Jeewoo Kim
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Michael Levin
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Samuel Khodurksy
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Adam Bress
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Kyung Lee
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
| | - Jens Meiler
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Jennifer S Lee
- Stanford University, Stanford, California, United States of America
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Kent Heberer
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Donald R Miller
- VA Center for Medication Safety, Department of Veterans Affairs, Chicago, Illinois, United States of America
- Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, United States of America
| | - Peter Reaven
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Kyong-Mi Chang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | - Julie A Lynch
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Nikhil K Khankari
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Megan M Shuey
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Marijana Vujkovic
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
172
|
Qiu X, Shen S, Jiang N, Lu D, Feng Y, Yang G, Xiang B. Adherence to the planetary health diet index and metabolic dysfunction-associated steatotic liver disease: a cross-sectional study. Front Nutr 2025; 12:1534604. [PMID: 40051965 PMCID: PMC11882404 DOI: 10.3389/fnut.2025.1534604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Backgrounds Adherence to the Planetary Health Diet Index (PHDI) has been shown to benefit both individual health and the planet. However, its impact on Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) remains unclear. This study aimed to investigate the relationship between PHDI adherence and the MASLD risk. Methods We analyzed a cohort of 15,865 adults (aged ≥18 years) using data from the National Health and Nutrition Examination Survey (NHANES, 2005-2018). The PHDI was derived from 24-h dietary assessments and comprised the scores of 15 food groups. Multivariate logistic regression was used to investigate the association between PHDI and MASLD, while restricted cubic spline (RCS) regression and threshold analysis were employed to explore potential non-linear relationship. Subgroup analyses were conducted to assess the influence of various demographic and clinical characteristics on the observed associations. Mediation analysis was performed to evaluate the indirect effect of PHDI on MASLD, and weighted quantile sum (WQS) regression was used to assess the influence of individual PHDI nutrients on MASLD. Results Among the cohort, 6,125 individuals were diagnosed with MASLD. Multivariate logistic regression revealed that a higher quintile of PHDI was significantly associated with reduced MASLD risk in the fully adjusted model (OR = 0.610, 95%CI 0.508-0.733, p < 0.001). Notably, nonlinear relationships between PHDI and MASLD risk were observed through RCS analysis (p = 0.002). Subgroup analyses indicated that PHDI was particularly effective in reducing MASLD risk among females, those with higher education attainment, and those living with a partner. WQS regression identified saturated fatty acids as the most significant factor contributing to MASLD risk (weight = 0.313). Additionally, BMI and waist circumference (81.47 and 87.66%, respectively) partially mediated the association between PHDI and MASLD risk, suggesting that the effect of PHDI on MASLD operates, in part, through its impact on BMI and waist circumference. The association between PHDI and MASLD remained robust across multiple sensitivity analyses. Conclusion Our findings indicate that adherence to PHDI is linked to a lower risk of MASLD, providing crucial insights for strategies aimed at mitigating the MASLD epidemic while simultaneously fostering environmental sustainability.
Collapse
Affiliation(s)
- Xin Qiu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shuang Shen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Nizhen Jiang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Donghong Lu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yifei Feng
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guodong Yang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| |
Collapse
|
173
|
Pugliese N, Bertazzoni A, Hassan C, Schattenberg JM, Aghemo A. Revolutionizing MASLD: How Artificial Intelligence Is Shaping the Future of Liver Care. Cancers (Basel) 2025; 17:722. [PMID: 40075570 PMCID: PMC11899536 DOI: 10.3390/cancers17050722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as a leading cause of chronic liver disease. In recent years, artificial intelligence (AI) has attracted significant attention in healthcare, particularly in diagnostics, patient management, and drug development, demonstrating immense potential for application and implementation. In the field of MASLD, substantial research has explored the application of AI in various areas, including patient counseling, improved patient stratification, enhanced diagnostic accuracy, drug development, and prognosis prediction. However, the integration of AI in hepatology is not without challenges. Key issues include data management and privacy, algorithmic bias, and the risk of AI-generated inaccuracies, commonly referred to as "hallucinations". This review aims to provide a comprehensive overview of the applications of AI in hepatology, with a focus on MASLD, highlighting both its transformative potential and its inherent limitations.
Collapse
Affiliation(s)
- Nicola Pugliese
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy; (N.P.); (A.B.); (C.H.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Arianna Bertazzoni
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy; (N.P.); (A.B.); (C.H.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy; (N.P.); (A.B.); (C.H.)
- Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| | - Jörn M. Schattenberg
- Department of Internal Medicine II, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Alessio Aghemo
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, MI, Italy; (N.P.); (A.B.); (C.H.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy
| |
Collapse
|
174
|
Åsberg A, Lian IA, Løfblad L, Mikkelsen G. Personalized risk assessment of advanced liver fibrosis in patients with MASLD. J Hepatol 2025:S0168-8278(25)00093-5. [PMID: 39983835 DOI: 10.1016/j.jhep.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/23/2025]
Affiliation(s)
- Arne Åsberg
- Department of Clinical Chemistry, St. Olav's Hospital, POB 3250 Torgarden, 7006 Trondheim, Norway.
| | - Ingrid Alsos Lian
- Department of Clinical Chemistry, St. Olav's Hospital, POB 3250 Torgarden, 7006 Trondheim, Norway and Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, POB 8900, 7491 Trondheim, Norway
| | - Lena Løfblad
- Department of Clinical Chemistry, St. Olav's Hospital, POB 3250 Torgarden, 7006 Trondheim, Norway and Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, POB 8900, 7491 Trondheim, Norway
| | - Gustav Mikkelsen
- Department of Clinical Chemistry, St. Olav's Hospital, POB 3250 Torgarden, 7006 Trondheim, Norway and Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, POB 8900, 7491 Trondheim, Norway
| |
Collapse
|
175
|
Roeren M, Grip ET, Skröder H, Shang Y, Hagström H. Potential issues with implementation of EASL-EASD-EASO guidelines on management of MASLD in patients with type 2 diabetes in Sweden. J Hepatol 2025:S0168-8278(25)00097-2. [PMID: 39983842 DOI: 10.1016/j.jhep.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Affiliation(s)
- Merle Roeren
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Emilie Toresson Grip
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Quantify Research, Stockholm, Sweden
| | - Helena Skröder
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Quantify Research, Stockholm, Sweden
| | - Ying Shang
- 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 Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
176
|
Shi D, Tan Q, Zhang Y, Qi X, Xu X, Xu G, Bai R, Deng J, Chen M, Jiang T, Mei Y. Serum uric acid trajectories and risk of metabolic dysfunction-associated steatotic liver disease in China: a 2019-2021 cohort health survey. BMC Public Health 2025; 25:653. [PMID: 39962427 PMCID: PMC11834244 DOI: 10.1186/s12889-024-21214-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/26/2024] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE To investigate the associations between serum uric acid (UA) trajectories and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in large cohort survey 2019-2021. METHODS This cohort health survey included 11,644 participants without MASLD before January 1, 2021. Among them, 5578 (47.90%) were men and 6066 (52.10%) were women. The group-based trajectory model method was applied to identify serum UA trajectories from January 1, 2019, to December 30, 2021. New-onset MASLD events in 2021 were treated as outcomes. A logistic regression model was used to assess associations between UA trajectories and incidence of MASLD. RESULTS Four distinct serum UA trajectories among both sexes were identified: "low-stable" trajectory 1 (n = 783 men; n = 1143 women), "moderate-moderate increasing" trajectory 2 (n = 2794 men; n = 3266 women), "moderate high-moderate increasing" trajectory 3 (n = 1660 men; n = 1464 women), and "high-increasing" trajectory 4 (n = 341 men; n = 193 women). During the 3-year follow-up period, 840 (15.06%) men and 408 (6.72%) women developed MASLD, respectively. Compared with the trajectory 1 group, the trajectory 4 group had the highest risk (odds ration [OR] 2.99 [95% confidence interval {CI} 1.70, 5.24] for men; OR 2.37 [95% CI 1.04, 5.33] for women), followed by the trajectory 3 (OR 2.23 [95% CI 1.52, 3.30] for men; OR 2.37 [95% CI 1.45, 3.92] for women) and trajectory 2 (OR 1.43 [95% CI 1.07, 1.94] for men; OR 1.37 [95% CI 0.93, 2.03] for women) groups. CONCLUSIONS High serum UA trajectories were independent risk factors for MASLD in both sexes, which is critical for informing prevention and treatment strategies in public health initiatives and clinical practice.
Collapse
Affiliation(s)
- Dan Shi
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Qilong Tan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
- School of Public Health, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yong Zhang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoya Qi
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoyang Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Guoqiong Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Ruixue Bai
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Jing Deng
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Mengxue Chen
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Tao Jiang
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| | - Ying Mei
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| |
Collapse
|
177
|
Amiri F, Moludi J, Jouybari TA, Ghasemi M, Sharifi M, Mahaki B, Soleimani D. Relationship between dietary inflammatory index and metabolic dysfunction associated steatotic liver disease in children. Sci Rep 2025; 15:5081. [PMID: 39934274 PMCID: PMC11814066 DOI: 10.1038/s41598-025-89690-x] [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: 09/25/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Emerging research suggests a connection between dietary habits and metabolic dysfunction associated steatotic liver disease (MASLD). We aimed to assess the relationship between the Dietary Inflammatory Index (DII) and the severity of MASLD. This cross-sectional study involved 125 participants aged 7-18 with MASLD. Dietary intakes were assessed using a validated 147-item food frequency questionnaire. The DII was applied to measure diet-induced inflammation, categorizing diets into anti-inflammatory (DII < -1) and inflammatory diets (DII > + 1). To evaluate hepatic steatosis and fibrosis, transabdominal ultrasonography and the fibrosis-4 (FIB-4) index were utilized. After adjusting for potential confounding factors, individuals on inflammatory diets exhibited a significantly higher odds ratio (OR) for experiencing more severe steatosis (OR: 4.11; 95% CI: 1.08-15.71) compared to those on anti-inflammatory diets. Furthermore, each unit increase in DII scores was linked to a 2.6-fold increase in the odds of more severe steatosis (OR: 2.61; 95% CI: 1.28-5.32) and a 0.006 increase in FIB-4 scores (β: 0.006; 95% CI: 0.003-0.011). In conclusion, modifying dietary intakes to lower DII scores may be a beneficial approach to improving clinical outcomes in pediatric MASLD, as anti-inflammatory diets are associated with reduced severity of hepatic steatosis and fibrosis.
Collapse
Affiliation(s)
- Fateme Amiri
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jalal Moludi
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Touraj Ahmadi Jouybari
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahmoud Ghasemi
- Department of Pediatrics, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Sharifi
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behzad Mahaki
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Davood Soleimani
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
178
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
179
|
Younossi ZM, Paik JM, Henry L, Pollock RF, Stepanova M, Nader F. Economic evaluation of non-invasive test pathways for high-risk metabolic dysfunction-associated steatotic liver disease (MASLD) in the United Kingdom (UK). Ann Hepatol 2025; 30:101789. [PMID: 39929473 DOI: 10.1016/j.aohep.2025.101789] [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: 09/10/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025]
Abstract
INTRODUCTION AND OBJECTIVES Non-invasive tests (NITs) identifying high-risk MASLD in primary care is suggested but, these strategies cost-effectiveness remain uncertain in the United Kingdom (UK). MATERIALS AND METHODS A cost-utility/budget impact model was developed for cost-effectiveness evaluation of two screening strategies (1) FIB-4 followed by Enhanced Liver Fibrosis (ELF) (FIB-4/ELF); (2) FIB-4 followed by Transient Elastography (FIB-4/TE) compared to standard of care (SoC). A cohort of primary care MASLD patients with an advanced fibrosis prevalence of 4.20 % was simulated. A decision tree classified patients as true positives, false positives, true negatives, or false negatives based on NIT diagnostic accuracy, followed by a 3-year Markov model to estimate costs and quality-adjusted life years (QALYs). The model included 11 health states: MASLD, fibrosis stages (F0-F3), cirrhosis, decompensated cirrhosis, liver transplant, and death. Costs came from the National Tariff, National Schedule of Costs and Personal Social Services Research Unit. RESULTS SoC had a false diagnosis rate of 36.26 %, while FIB-4 with ELF or TE reduced false positive rates to 23.20 % and 20.91 %, respectively. Compared to 112,807 unnecessary hepatology referrals under SoC, FIB-4/ELF or FIB-4/TE reduced unnecessary referrals by 38,031 (33.71 %) and 45,767 (40.57 %), respectively. Both strategies demonstrated cost-effectiveness relative to SoC with total cost per patient of GBP 983.37 for FIB-4/TE, GBP 993.15 for FIB-4/ELF compared to SoC, GBP 1,014.15. CONCLUSIONS Sequential NIT screening strategies, combining FIB-4 with ELF or TE, are cost-saving, reduce unnecessary hepatology referrals, and offer an efficient (improve outcomes and reduce healthcare costs) approach for managing high-risk MASLD in UK primary care.
Collapse
Affiliation(s)
- Zobair M Younossi
- The Global NASH Council, Washington DC, USA; Beatty Liver and Obesity Research, Inova Health System, Falls Church, VA, USA.
| | - James M Paik
- The Global NASH Council, Washington DC, USA; Beatty Liver and Obesity Research, Inova Health System, Falls Church, VA, USA
| | - Linda Henry
- The Global NASH Council, Washington DC, USA; Beatty Liver and Obesity Research, Inova Health System, Falls Church, VA, USA
| | | | - Maria Stepanova
- The Global NASH Council, Washington DC, USA; Beatty Liver and Obesity Research, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Diseases, Washington DC, USA
| | - Fatema Nader
- The Global NASH Council, Washington DC, USA; Beatty Liver and Obesity Research, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Diseases, Washington DC, USA
| |
Collapse
|
180
|
Lau PP, Wei CY, Lin MR, Chou WH, Wan YJY, Chang WC. Genome-wide association study of the fatty liver index in the Taiwanese population reveals shared and population-specific genetic risk factors across ethnicities. Cell Biosci 2025; 15:19. [PMID: 39923089 PMCID: PMC11807309 DOI: 10.1186/s13578-025-01346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/08/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Although the incidence of fatty liver disease (FLD) is increasing worldwide, the genetic basis of this disease is not fully understood. This study uses the fatty liver index (FLI) to identify and compare genetic variants associated with FLD in Taiwanese and European populations. RESULTS In this study, a total of 145,356 Taiwan Biobank participants were included in the discovery analysis. Subjects with elevated FLI were found to have a significantly greater risk of developing FLD, as confirmed by imaging data (OR: 4.43; 95% CI: 3.88-5.06). Through genome-wide association studies (GWAS), we identified 6 variants previously associated with nonalcoholic fatty liver disease (NAFLD) and validated 50 shared risk variants located in ZPR1 and FTO between the Taiwanese and European populations. Conditional analysis of 423 significant variants from FLI-defined FLD further revealed 16 independent variants within 14 genes. Pathway analysis of GWAS significant genes revealed that lipid metabolism and the peroxisome proliferator-activated receptor (PPAR) signaling pathway are causes of hepatic fat accumulation. CONCLUSION This study identified six independent NAFLD-associated variants in GCKR, LPL, TRIB1AL, and FTO and emphasized ZPR1 and FTO as shared risk genes for FLI-defined FLD in both Taiwanese and European populations. These findings support the utility of the FLI for FLD prediction, provide new genetic insights, and reveal the common genetic pathways of FLD across two ethnic groups. This research offers a valuable framework for advancing personalized medicine and therapeutic strategies for FLD.
Collapse
Affiliation(s)
- Pei Pei Lau
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chun-Yu Wei
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, Taiwan
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Jui Yvonne Wan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, Taiwan.
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
181
|
Cazac-Panaite GD, Lăcătușu CM, Grigorescu ED, Foșălău AB, Onofriescu A, Mihai BM. Innovative Drugs First Implemented in Type 2 Diabetes Mellitus and Obesity and Their Effects on Metabolic Dysfunction-Associated Steatohepatitis (MASH)-Related Fibrosis and Cirrhosis. J Clin Med 2025; 14:1042. [PMID: 40004572 PMCID: PMC11857078 DOI: 10.3390/jcm14041042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a progressive liver disease frequently associated with metabolic disorders such as type 2 diabetes mellitus (T2DM) and obesity, has the potential to progress symptomatically to liver cirrhosis and, in some cases, hepatocellular carcinoma. Hence, an urgent need arises to identify and approve new therapeutic options to improve patient outcomes. Research efforts have focused on either developing dedicated molecules or repurposing drugs already approved for other conditions, such as metabolic diseases. Among the latter, antidiabetic and anti-obesity agents have received the most extensive attention, with pivotal trial results anticipated shortly. However, the primary focus underlying successful regulatory approvals is demonstrating a substantial efficacy in improving liver fibrosis and preventing or ameliorating cirrhosis, the key advanced outcomes within MASLD progression. Besides liver steatosis, the ideal therapeutic candidate should reduce inflammation and fibrosis effectively. Although some agents have shown promise in lowering MASLD-related parameters, evidence of their impact on fibrosis and cirrhosis remains limited. This review aims to evaluate whether antidiabetic and anti-obesity drugs can be safely and effectively used in MASLD-related advanced fibrosis or cirrhosis in patients with T2DM. Our paper discusses the molecules closest to regulatory approval and the expectation that they can address the unmet needs of this increasingly prevalent disease.
Collapse
Affiliation(s)
- Georgiana-Diana Cazac-Panaite
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cristina-Mihaela Lăcătușu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Elena-Daniela Grigorescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Adina-Bianca Foșălău
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Alina Onofriescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan-Mircea Mihai
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-D.C.-P.); (E.-D.G.); (A.-B.F.); (A.O.); (B.-M.M.)
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| |
Collapse
|
182
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [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: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
183
|
Wu X, Pan T, Fang Z, Hui T, Yu X, Liu C, Guo Z, Liu C. Identification of EGR1 as a Key Diagnostic Biomarker in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Through Machine Learning and Immune Analysis. J Inflamm Res 2025; 18:1639-1656. [PMID: 39925925 PMCID: PMC11806694 DOI: 10.2147/jir.s499396] [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: 10/05/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
Background Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), as a common chronic liver condition globally, is experiencing an increasing incidence rate which poses significant health risks. Despite this, the detailed mechanisms underlying the disease's onset and progression remain poorly understood. In this study, we aim to identify effective diagnostic biomarkers for MASLD using microarray data combined with machine learning techniques, which will aid in further understanding the pathogenesis of MASLD. Methods We collected six datasets from the Gene Expression Omnibus (GEO) database, using five of them as training sets and one as a validation set. We employed three machine learning methods-LASSO, SVM, and Random Forest (RF)-to identify hub genes associated with MASLD. These genes were further validated using the external dataset GSE164760. Additionally, functional enrichment analysis, immune infiltration analysis, and immune function analysis were conducted. A TF-miRNA-mRNA network was constructed, and single-cell RNA sequencing was used to determine the distribution of key genes within key cell clusters. Finally, the expression of the key genes was further validated using the palmitic acid-induced AML-12 cell line and the MCD mouse model. Results In this study, through differential gene expression (DEGs) analysis and machine learning techniques, we successfully identified 10 hub genes. Among these, the key gene EGR1 was validated and screened using an external dataset, with an area under the curve (AUC) of 0.882. Enrichment analyses and immune infiltration assessments revealed multiple pathways involving EGR1 in the pathogenesis and progression of MASLD, showing significant correlations with various immune cells. Furthermore, additional cellular experiments and animal model validations confirmed that the expression trends of EGR1 are highly consistent with our analytical findings. Conclusion Our research has confirmed EGR1 as a key gene in MASLD, providing novel insights into the disease's pathogenesis and identifying new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Xuanlin Wu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tao Pan
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Titi Hui
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoxiao Yu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zihao Guo
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
184
|
Mo H, Yue P, Li Q, Tan Y, Yan X, Liu X, Xu Y, Luo Y, Palihati S, Yi C, Zhang H, Yuan M, Yang B. The role of liver sinusoidal endothelial cells in metabolic dysfunction-associated steatotic liver diseases and liver cancer: mechanisms and potential therapies. Angiogenesis 2025; 28:14. [PMID: 39899173 DOI: 10.1007/s10456-025-09969-5] [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: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
Liver sinusoidal endothelial cells (LSECs), with their unique morphology and function, have garnered increasing attention in chronic liver disease research. This review summarizes the critical roles of LSECs under physiological conditions and in two representative chronic liver diseases: metabolic dysfunction-associated steatotic liver disease (MASLD) and liver cancer. Under physiological conditions, LSECs act as selective barriers, regulating substance exchange and hepatic blood flow. Interestingly, LSECs exhibit contrasting roles at different stages of disease progression: in the early stages, they actively resist disease advancement and help restore sinusoidal homeostasis; whereas in later stages, they contribute to disease worsening. During this transition, LSECs undergo capillarization, lose their characteristic markers, and become dysfunctional. As the disease progresses, LSECs closely interact with hepatocytes, hepatic stellate cells, various immune cells, and tumor cells, driving processes such as steatosis, inflammation, fibrosis, angiogenesis, and carcinogenesis. Consequently, targeting LSECs represents a promising therapeutic strategy for chronic liver diseases. Relevant therapeutic targets and potential drugs are summarized in this review.
Collapse
Affiliation(s)
- Hanjun Mo
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Pengfei Yue
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Qiaoqi Li
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yinxi Tan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Xinran Yan
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyue Liu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Medical Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Chengdu, 610075, Sichuan, China
| | - Suruiya Palihati
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Cheng Yi
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, China.
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, 610041, China.
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| | - Biao Yang
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
185
|
Ginès P, Guha IN, Fabrellas N, Allen AM, Angeli P, Serra-Burriel M. Liver Health: An Emerging Concept. Gastroenterology 2025:S0016-5085(25)00336-1. [PMID: 39909131 DOI: 10.1053/j.gastro.2024.12.039] [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: 09/13/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 02/07/2025]
Affiliation(s)
- Pere Ginès
- Liver Unit Hospital Clínic, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Centro de Investigación En Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Indra Neil Guha
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; University of Nottingham, Nottingham, United Kingdom
| | - Núria Fabrellas
- Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Centro de Investigación En Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain; Faculty of Nursing, University of Barcelona, Barcelona, Spain
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Miquel Serra-Burriel
- Epidemiology, Statistics, and Prevention Institute, University of Zurich, Zurich, Switzerland
| |
Collapse
|
186
|
Martín-Grau M, Casanova P, Moreno-Morcillo L, Morales JM, Marrachelli VG, Monleón D. Microbiota Co-Metabolism Alterations Precede Changes in the Host Metabolism in the Early Stages of Diet-Induced MASLD in Wistar Rats. Int J Mol Sci 2025; 26:1288. [PMID: 39941056 PMCID: PMC11818068 DOI: 10.3390/ijms26031288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) affects around 30% of the global population. The sexual dimorphism and gut microbiota play an important role in the early development of MASLD. The main objective of this research was to investigate metabolic changes during the early subclinical MASLD progression, for identifying the sequence of events and evaluating the impact of sexual dimorphism and the microbiota on the initial stages of MASLD development. Male and female Wistar rats 18 weeks old were randomly divided into different groups and fed a chow diet or a 45% high-fat diet for 21 weeks. Every three weeks, samples of serum, urine, and faeces were collected and studied by metabolomics. Furthermore, the liver was analysed at the endpoint. In addition, the gut microbiota was analysed from faecal samples over time using 16S rRNA gene-targeted group-specific primers. Our results revealed that three weeks on an HFD reduced the bacterial diversity in the faecal microbiota of Wistar rats, accompanied by changes in the faecal and urine metabolome. The HFD-induced alterations in microbiota-related co-metabolites in the liver, blood, urine, and faeces indicate a significant role of host-microbiota co-metabolism changes in the early stages of MASLD. In this study, we provide a comprehensive longitudinal analysis, detailing the sequence of events in the early development of MASLD. Our findings suggest that alterations in the gut microbiota diversity and co-metabolism occur before changes in host metabolism in the early onset of liver steatosis, a subclinical phase of MASLD.
Collapse
Affiliation(s)
- María Martín-Grau
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Pilar Casanova
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Laura Moreno-Morcillo
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - José Manuel Morales
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Vannina G. Marrachelli
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Departament de Fisiologia, Universitat de València, 46010 Valencia, Spain
| | - Daniel Monleón
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| |
Collapse
|
187
|
Lindfors A, Strandberg R, Hagström H. Screening for advanced liver fibrosis due to metabolic dysfunction-associated steatotic liver disease alongside retina scanning in people with type 2 diabetes: a cross-sectional study. Lancet Gastroenterol Hepatol 2025; 10:125-137. [PMID: 39675369 DOI: 10.1016/s2468-1253(24)00313-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND International guidelines suggest screening for advanced fibrosis due to metabolic dysfunction-associated steatotic liver disease in people with type 2 diabetes, but how to implement these guidelines in clinical care remains unclear. We hypothesise that examination with VCTE could be implemented simultaneously with retina scanning with a high acceptance rate in people with type 2 diabetes. METHODS In this cross-sectional study, we offered VCTE to people with type 2 diabetes referred to routine retina scanning in a large retina scanning facility in Stockholm, Sweden. We excluded people with type 1 diabetes, currently pregnant, with known liver disease, reporting high alcohol consumption, who did not speak Swedish, or younger than 18 years. Between Nov 6, 2020, and June 20, 2023, we conducted surveys with included participants and collected data from medical records on diabetes retinopathy, sex, and VCTE measurements. Increased liver stiffness was defined as at least 8·0 kPa, and possible advanced fibrosis as more than 12·0 kPa. Presence of metabolic dysfunction-associated steatotic liver disease was defined as a controlled attenuation parameter (CAP) value of 280 dB/m or higher. Participants with a liver stiffness measurement of at least 8·0 kPa or those with unreliable measurements were subsequently referred for a secondary evaluation at a liver specialist, including a follow-up liver stiffness measurement with VCTE. The primary outcome was the proportion of eligible people approached for screening who accepted. Secondary outcomes were the prevalence of elevated liver stiffness (≥8·0 kPa or >12·0 kPa), presence of metabolic dysfunction-associated steatotic liver disease, and the proportion of elevated liver stiffness readings at the first VCTE examination that were not elevated in the secondary evaluation with a liver specialist. Secondary outcomes were assessed in all participants who accepted screening, except false positives, which were assessed only in participants who had a second examination. FINDINGS 1301 participants were eligible to undergo assessment with VCTE, which was accepted by 1005 (77·2%). 973 (96·8%) participants had complete measurements, of whom 504 (51·8%) had CAP values of 280 dB/m or higher, indicating metabolic dysfunction-associated steatotic liver disease. Of 977 participants with reliable liver stiffness measurements, 154 (15·8%) had values of at least 8·0 kPa, suggestive of liver fibrosis, and 49 (5·0%) had values higher than 12·0 kPa, indicating possible advanced fibrosis. However, upon reassessment with a second VCTE after referral, 56 (45·2%) of 124 individuals had values less than 8·0 kPa. 74 (7·4%) of 1005 participants had a final liver stiffness of at least 8·0 kPa; 29 (2·9%) had values greater than 12·0 kPa. INTERPRETATION Simultaneous examination with VCTE alongside retina scanning had a high acceptance rate among people with type 2 diabetes and could be a strategy for case-finding of people with fibrosis due to metabolic dysfunction-associated steatotic liver disease. However, a high proportion of participants in our study with elevated liver stiffness measurement at the screening visit did not have an elevated liver stiffness measurement at secondary evaluation, suggesting false-positive findings were common. FUNDING Gilead Sciences, Pfizer, Region Stockholm, Åke Wiberg Foundation, and Bengt Ihre Foundation.
Collapse
Affiliation(s)
- Andrea Lindfors
- Division of Hepatology, Department of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Hannes Hagström
- Division of Hepatology, Department of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
188
|
Elgretli W, Shengir M, Sasson S, Ramanakumar AV, Cinque F, Ballestreros LER, Deschenes M, Wong P, Chen T, Kronfli N, Saeed S, Keeshan A, Tandon S, Cooper C, Sebastiani G. Association of MASLD Phenotypes With Liver Fibrosis in Hepatitis C: The Role of Cardiometabolic Risk Factors. J Viral Hepat 2025; 32:e70004. [PMID: 39868661 PMCID: PMC11771651 DOI: 10.1111/jvh.70004] [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: 10/22/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/28/2025]
Abstract
Steatotic liver disease is prevalent among people with hepatitis C virus (HCV). The new definition of metabolic dysfunction-associated steatotic liver disease (MASLD) emphasises the metabolic drivers of steatosis and recognises its frequent coexistence with other chronic liver diseases, including HCV. We aimed to evaluate the association of coexisting MASLD and HCV with liver fibrosis. Individuals with HCV who underwent transient elastography (TE) with associated controlled attenuation parameter (CAP) were included from two clinical centres. MASLD and significant liver fibrosis were defined as the presence of steatosis (CAP ≥ 275 dB/m) with at least one cardiometabolic risk factor, and liver stiffness measurement (LSM) ≥ 7.1 kPa measured by TE, respectively. Associated cofactors of significant liver fibrosis were determined using stepwise regression and cross-validation by LASSO models to select confounders. Among 590 participants, 31% were diagnosed with MASLD. The prevalence of significant liver fibrosis was the highest among people with MASLD (58%) followed by HCV-related steatosis (45%) and the non-steatosis group (39%). After adjusting for potential confounders, MASLD was associated with significant liver fibrosis (adjusted odds ratio [aOR] 2.29, 95% confidence interval [CI] 1.07-4.87). Furthermore, specific MASLD phenotypes including diabetes, hypertension and overweight were associated with significant liver fibrosis, with aORs of 4.76 (95% CI 2.16-10.49), 3.44 (95% CI 1.77-6.68) and 2.54 (95% CI 1.27-5.07), respectively. In conclusion, MASLD is associated with liver fibrosis in people with HCV, specifically the diabetes, overweight and hypertensive phenotypes. Beyond pursuing a virological cure, healthcare providers should prioritise managing metabolic conditions, particularly diabetes, hypertension and obesity.
Collapse
Affiliation(s)
- Wesal Elgretli
- Division of Experimental MedicineMcGill UniversityMontrealQuebecCanada
| | - Mohamed Shengir
- Division of Experimental MedicineMcGill UniversityMontrealQuebecCanada
| | - Solomon Sasson
- Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | - Felice Cinque
- Department of PathophysiologyTransplantation University of MilanMilanItaly
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Luz Esther Ramos Ballestreros
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Marc Deschenes
- Division of Gastroenterology and Hepatology, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Phil Wong
- Division of Gastroenterology and Hepatology, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Tianyan Chen
- Division of Gastroenterology and Hepatology, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Nadine Kronfli
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
- Centre for Outcomes Research and EvaluationResearch Institute of the McGill University Health CentreMontrealQuebecCanada
| | - Sahar Saeed
- Public Health SciencesQueen's UniversityKingstonOntarioCanada
| | - Alexa Keeshan
- Division of Infectious Diseases, Department of MedicineOttawa Hospital Research Institute, The Ottawa HospitalOttawaOntarioCanada
| | - Saniya Tandon
- Division of Infectious Diseases, Department of MedicineOttawa Hospital Research Institute, The Ottawa HospitalOttawaOntarioCanada
| | - Curtis Cooper
- Division of Infectious Diseases, Department of MedicineOttawa Hospital Research Institute, The Ottawa HospitalOttawaOntarioCanada
| | - Giada Sebastiani
- Division of Experimental MedicineMcGill UniversityMontrealQuebecCanada
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
- Division of Gastroenterology and Hepatology, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| |
Collapse
|
189
|
Zheng Y, Sun Y, Ren W, Duan R, Li S, Chen M, Qin H, Ying M, Ren J. Factors Associated with Nonalcoholic Fatty Liver Disease in a Non-Overweight/Obese and Overweight/Obese Chinese Population at Risk for Metabolic Syndrome: A Cross-Sectional Multicenter Study. Metab Syndr Relat Disord 2025; 23:41-52. [PMID: 39311687 DOI: 10.1089/met.2024.0168] [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: 01/31/2025] Open
Abstract
Background: To investigate the association of demographic, clinical, and metabolic factors with nonalcoholic fatty liver disease (NAFLD) in a non-overweight/obese and overweight/obese Chinese population at risk for metabolic syndrome. Patients and Method: A cross-sectional multicenter study was conducted using convenience sampling from eight selected counties/cities in Zhejiang, China, between May 2021 and September 2022. Demographics, epidemiological, anthropometric, and clinical characteristics were obtained from a questionnaire. Least absolute shrinkage and selection operator (LASSO)-logistic regression analysis was used to identify the variables associated with NAFLD. Receiver operating characteristic (ROC) curve analysis and decision curve analysis (DCA) were performed to evaluate the diagnostic value and clinical utility of the variables and models. Results: A total of 1739 patients were enrolled in the final analysis, 345 (19.8%) were non-overweight/obese and 1394 (80.2%) were overweight/obese participants. There were 114 (33.0%) and 1094 (78.5%) patients who met the criteria for NAFLD in the non-overweight/obese participants and the overweight/obese participants respectively. Older age, current smoking, higher triglyceride (TG) levels, higher AST levels, higher albumin levels, lower insulin levels, and higher controlled attenuation parameter (CAP) scores were associated with NAFLD in both non-overweight/obese and overweight/obese participants. The combination of TG+CAP scores had strong predictive values for NAFLD, especially in non-overweight/obese (Area Under Curve = 0.812, 95% confidence interval: 0.764-0.863). DCA showed a superior net benefit of the TG+CAP score over other variables or models, suggesting a better clinical utility in identifying NAFLD. Conclusions: More stringent lipid management strategies remain essential, and the convenience and efficacy of transient elastography for liver steatosis should be recognized, especially in the non-overweight/obese population.
Collapse
Affiliation(s)
- Yang Zheng
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Allergy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yujing Sun
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen Ren
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruoshu Duan
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Li
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingmin Chen
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongli Qin
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meike Ying
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Ren
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
190
|
Deslande M, Puig-Castellvi F, Castro-Dionicio I, Pacheco-Tapia R, Raverdy V, Caiazzo R, Lassailly G, Leloire A, Andrikopoulos P, Kahoul Y, Zaïbi N, Toussaint B, Oger F, Gambardella N, Lefebvre P, Derhourhi M, Amanzougarene S, Staels B, Pattou F, Froguel P, Bonnefond A, Dumas ME. Intrahepatic levels of microbiome-derived hippurate associates with improved metabolic dysfunction-associated steatotic liver disease. Mol Metab 2025; 92:102090. [PMID: 39746606 PMCID: PMC11772989 DOI: 10.1016/j.molmet.2024.102090] [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: 08/21/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterised by lipid accumulation in the liver and is often associated with obesity and type 2 diabetes. The gut microbiome recently emerged as a significant player in liver metabolism and health. Hippurate, a host-microbial co-metabolite has been associated with human gut microbial gene richness and with metabolic health. However, its role on liver metabolism and homeostasis is poorly understood. METHODS We characterised liver biospies from 318 patients with obesity using RNAseq and metabolomics in liver and plasma to derive associations among hepatic hippurate, hepatic gene expression and MASLD and phenotypes. To test a potential beneficial role for hippurate in hepatic insulin resistance, we profile the metabolome of (IHH) using ultra-high-performance liquid chromatography coupled to high-resolution tandem mass spectrometry (UHPLC-MS/MS), and characterised intracellular triglyceride accumulation and glucose internalisation after a 24 h insulin exposure. RESULTS We first report significant associations among MASLD traits, plasma and hepatic hippurate. Further analysis of the hepatic transcriptome shows that liver and plasma hippurate are inversely associated with MASLD, implicating lipid metabolism and regulation of inflammatory responses pathways. Hippurate treatment inhibits lipid accumulation and rescues insulin resistance induced by 24-hour chronic insulin in IHH. Hippurate also improves hepatocyte metabolic profiles by increasing the abundance of metabolites involved in energy homeostasis that are depleted by chronic insulin treatment while decreasing those involved in inflammation. CONCLUSIONS Altogether, our results further highlight hippurate as a mechanistic marker of metabolic health, by its ability to improve metabolic homeostasis as a postbiotic candidate.
Collapse
Affiliation(s)
- Maxime Deslande
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Francesc Puig-Castellvi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Inés Castro-Dionicio
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Romina Pacheco-Tapia
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Violeta Raverdy
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Robert Caiazzo
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Guillaume Lassailly
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Audrey Leloire
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Petros Andrikopoulos
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Yasmina Kahoul
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nawel Zaïbi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bénédicte Toussaint
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Frédérik Oger
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nicolas Gambardella
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Philippe Lefebvre
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Mehdi Derhourhi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Souhila Amanzougarene
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bart Staels
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - François Pattou
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Philippe Froguel
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Amélie Bonnefond
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Marc-Emmanuel Dumas
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom; The Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, H3A 0G1, Canada.
| |
Collapse
|
191
|
Raikhelson K, Okovityi S, Abdurakhmanov D. New nomenclature for fatty liver disease: Problems of localization in the regions and the position of Russian Scientific Liver Society. J Hepatol 2025; 82:e83-e84. [PMID: 39454691 DOI: 10.1016/j.jhep.2024.10.023] [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: 09/25/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Affiliation(s)
- Karina Raikhelson
- Saint-Petersburg State University 199034, Saint Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, 197022, Saint Petersburg, Russia
| | - Sergey Okovityi
- Saint-Petersburg State University 199034, Saint Petersburg, Russia; Saint Petersburg State Chemical Pharmaceutical University, 197376, Saint Petersburg, Russia
| | - Dzhamal Abdurakhmanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048, Moscow, Russia.
| |
Collapse
|
192
|
Armstrong MJ, Okanoue T, Sundby Palle M, Sejling AS, Tawfik M, Roden M. Similar weight loss with semaglutide regardless of diabetes and cardiometabolic risk parameters in individuals with metabolic dysfunction-associated steatotic liver disease: Post hoc analysis of three randomised controlled trials. Diabetes Obes Metab 2025; 27:710-718. [PMID: 39609879 DOI: 10.1111/dom.16065] [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: 08/22/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
AIMS Weight loss mediated by glucagon-like peptide-1 (GLP-1) analogues is lower in patients with type 2 diabetes versus those without. Type 2 diabetes and obesity are risk factors for metabolic dysfunction-associated steatotic liver disease (MASLD) and associated steatohepatitis (MASH). We evaluated weight changes in adults with MASLD/MASH with or without type 2 diabetes receiving the GLP-1 analogue semaglutide. MATERIALS AND METHODS This was a post hoc analysis of data from three 48-72-week randomised trials investigating the effect of semaglutide versus placebo in adults with MASLD (NCT03357380) or biopsy-confirmed MASH (NCT02970942 and NCT03987451). Pooled data for semaglutide (0.4 mg once daily and 2.4 mg once weekly [n = 163]) and placebo (n = 137) were analysed at 1 year. Weight changes were analysed by type 2 diabetes status (type 2 diabetes [n = 209], pre-type 2 diabetes [n = 51] and no diabetes [n = 40]) and by other cardiometabolic risk parameters using analysis of covariance and Spearman's rank correlations. RESULTS The overall mean weight change was -11.1 kg (-11.7%) and -0.7 kg (-0.6%) with semaglutide and placebo, respectively. While numerically higher for people without type 2 diabetes, estimated treatment differences with semaglutide versus placebo were similar overall for people with type 2 diabetes (-10.2 kg; -10.8%), pre-type 2 diabetes (-9.8 kg; -10.2%) and no diabetes (-11.6 kg; -13.1%). Differences between groups were not statistically significant (p > 0.50 for all). Baseline fasting plasma glucose, glycated haemoglobin, insulin levels, insulin resistance and lipids did not correlate with weight change. CONCLUSIONS People with MASLD/MASH had similar semaglutide-mediated weight loss regardless of type 2 diabetes status and other cardiometabolic risk parameters.
Collapse
Affiliation(s)
- Matthew J Armstrong
- Liver Unit, Queen Elizabeth University Hospital, Birmingham, UK
- National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Osaka, Japan
| | | | | | | | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| |
Collapse
|
193
|
Malandris K, Korakas E, Sarakapina A, Kalopitas G, Iatridi F, Liakos A, Bekiari E, Giouleme O, Tzatzagou G, Karagiannis T, Paschos P, Vasilakou D, Lambadiari V, Tzamou E, Daravigkas D, Sinakos E, Tsapas A. Accuracy of Controlled Attenuation Parameter for Liver Steatosis in High-Risk Patients for MASLD Using MRI-Proton Density Fat Fraction as Reference Standard. Dig Dis Sci 2025; 70:814-824. [PMID: 39708259 DOI: 10.1007/s10620-024-08799-7] [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: 07/11/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
AIM Controlled attenuation parameter (CAP) enables the noninvasive diagnosis of liver steatosis. Magnetic resonance imaging proton density fat fraction (MRI-PDFF) is increasingly used over biopsy for the assessment of steatosis in patients at risk for metabolic dysfunction-associated steatotic liver disease (MASLD). We assessed the accuracy of CAP for liver steatosis defined as MRI-PDFF ≥ 5%. METHODS We performed a cross-sectional, diagnostic accuracy study. We prospectively recruited consecutive adult participants with type 2 diabetes and body mass index (BMI) ≥ 25 kg/m2, who underwent CAP and MRI-PDFF within two weeks. RESULTS We included 113 participants. The area under the receiver operating characteristic (AUROC) of CAP for MRI-PDFF ≥ 5% was 0.82 [95% confidence interval (CI) 0.74-0.89]. CAP thresholds for ruling-out (sensitivity > 90%) and ruling-in (specificity > 90%) liver steatosis were below 249 and over 328 dB/m respectively. The AUROC of CAP for the detection of MRI-PDFF ≥ 10% was 0.81 (0.73-0.88). CAP thresholds for ruling-out and ruling-in MRI-PDFF ≥ 10% were below 271 and over 345 dB/m respectively. CAP measurements with an interquartile range (IQR) < 30 dB/m improved the detection of higher steatosis grades. CONCLUSION CAP has acceptable accuracy for diagnosing MRI-PDFF defined steatosis. Values below 249 dB/m can be used to rule-out liver steatosis, while values over 328 dB/m can set the diagnosis. An IQR < 30 dB/m might improve the accuracy of CAP for higher steatosis grades. CLINICAL TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Konstantinos Malandris
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece.
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| | - Emmanouil Korakas
- Second Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Sarakapina
- First Medical Department, "Papageorgiou" Hospital, Thessaloniki, Greece
| | - Georgios Kalopitas
- First Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fotini Iatridi
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Aris Liakos
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Eleni Bekiari
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Olga Giouleme
- Second Propedeutic Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Thomas Karagiannis
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Paschalis Paschos
- First Medical Department, "Papageorgiou" Hospital, Thessaloniki, Greece
| | - Despoina Vasilakou
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, Athens, Greece
| | - Elli Tzamou
- Affidea Diagnostic Center, Thessaloniki, Greece
| | | | - Emmanouil Sinakos
- Fourth Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Tsapas
- Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Clinical Research and Evidence-Based Medicine Unit, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642, Thessaloniki, Greece
- Harris Manchester College, University of Oxford, Oxford, UK
| |
Collapse
|
194
|
Stefanakis K, Mingrone G, George J, Mantzoros CS. Accurate non-invasive detection of MASH with fibrosis F2-F3 using a lightweight machine learning model with minimal clinical and metabolomic variables. Metabolism 2025; 163:156082. [PMID: 39566717 DOI: 10.1016/j.metabol.2024.156082] [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: 10/31/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND There are no known non-invasive tests (NITs) designed for accurately detecting metabolic dysfunction-associated steatohepatitis (MASH) with liver fibrosis stages F2-F3, excluding cirrhosis-the FDA-defined range for prescribing Resmetirom and other drugs in clinical trials. We aimed to validate and re-optimize known NITs, and most importantly to develop new machine learning (ML)-based NITs to accurately detect MASH F2-F3. METHODS Clinical and metabolomic data were collected from 443 patients across three countries and two clinic types (metabolic surgery, gastroenterology/hepatology) covering the entire spectrum of biopsy-proven MASH, including cirrhosis and healthy controls. Three novel types of ML models were developed using a categorical gradient boosting machine pipeline under a classic 4:1 split and a secondary independent validation analysis. These were compared with twenty-three biomarker, imaging, and algorithm-based NITs with both known and re-optimized cutoffs for MASH F2-F3. RESULTS The NAFLD (Non-Alcoholic Fatty Liver Disease) Fibrosis Score (NFS) at a - 1.455 cutoff attained an area under the receiver operating characteristic curve (AUC) of 0.59, the highest sensitivity (90.9 %), and a negative predictive value (NPV) of 87.2 %. FIB-4 risk stratification followed by elastography (8 kPa) had the best specificity (86.9 %) and positive predictive value (PPV) (63.3 %), with an AUC of 0.57. NFS followed by elastography improved the PPV to 65.3 % and AUC to 0.62. Re-optimized FibroScan-AST (FAST) at a 0.22 cutoff had the highest PPV (69.1 %). ML models using aminotransferases, metabolic syndrome components, BMI, and 3-ureidopropionate achieved an AUC of 0.89, which further increased to 0.91 following hyperparameter optimization and the addition of alpha-ketoglutarate. These new ML models outperformed all other NITs and displayed accuracy, sensitivity, specificity, PPV, and NPV up to 91.2 %, 85.3 %, 97.0 %, 92.4 %, and 90.7 % respectively. The models were reproduced and validated in a secondary sensitivity analysis, that used one of the cohorts as feature selection/training, and the rest as independent validation, likewise outperforming all other applicable NITs. CONCLUSIONS We report for the first time the diagnostic characteristics of non-invasive, metabolomics-based biomarker models to detect MASH with fibrosis F2-F3 required for Resmetirom treatment and inclusion in ongoing phase-III trials. These models may be used alone or in combination with other NITs to accurately determine treatment eligibility.
Collapse
Affiliation(s)
- Konstantinos Stefanakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | | | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA
| |
Collapse
|
195
|
Kokkorakis M, Folkertsma P, Forte JC, Wolffenbuttel BHR, van Dam S, Mantzoros CS. GDF-15 improves the predictive capacity of steatotic liver disease non-invasive tests for incident morbidity and mortality risk for cardio-renal-metabolic diseases and malignancies. Metabolism 2025; 163:156047. [PMID: 39396641 DOI: 10.1016/j.metabol.2024.156047] [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: 08/30/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND & AIMS Noninvasive tools (NITs) are currently used to stratify the risk of having or developing hepatic steatosis or fibrosis. Their performance and a proteomic-enabled improvement in forecasting long-term cardio-renal-metabolic morbidity, malignancies, as well as cause-specific and all-cause mortality, are lacking. Therefore, the performance of established NITs needs to be investigated in identifying cardio-renal-metabolic morbidity, malignancies, cause-specific and overall mortality and improve their performance with novel, proteomic-enabled NITs, including growth differentiation factor 15 (GDF-15), allowing multipurpose utilization. METHODS 502,359 UK Biobank participants free of the study outcomes at baseline with a 14-year median follow-up were grouped into three categories: a) general population, b) potentially metabolic dysfunction-associated steatotic liver disease (MASLD) population, c) individuals with type 2 diabetes mellitus. The investigated NITs include Aspartate aminotransferase to Platelet Ratio Index (APRI), Fibrosis 4 Index (FIB-4), Fatty Liver Index (FLI), Hepatic Steatosis Index (HSI), Lipid Accumulation Product (LAP), and metabolic dysfunction-associated fibrosis (MAF-5) score. RESULTS Adding GDF-15 to the existing NITs led to significantly increased prognostic performance compared to the traditional NITs in almost all instances, reaching substantially high C-indices, ranging between 0.601 and 0.808, with an overall >0.2 improvement in C-index. Overall, with the GDF-15 enhanced NITs, up to more than seven times fewer individuals need to be screened to identify more incident cases of adverse outcomes compared to the traditional NITs. The cumulative incidence of all outcomes, based on the continuous value percentiles of NITs, is increasing exponentially in the upper quintile of the GDF-15 enhanced NITs. CONCLUSIONS The herein-developed GDF-15 enhanced indices demonstrate higher screening effectiveness and significantly improved prognostic abilities, which are reduced to practice through an easy-to-use web-based calculator tool (https://clinicalpredictor.shinyapps.io/multimorbidity-mortality-risk/).
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Pytrik Folkertsma
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - José Castela Forte
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sipko van Dam
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA
| |
Collapse
|
196
|
Sohn W, Lee YS, Kim SS, Kim JH, Jin YJ, Kim GA, Sung PS, Yoo JJ, Chang Y, Lee EJ, Lee HW, Choi M, Yu SJ, Jung YK, Jang BK. KASL clinical practice guidelines for the management of metabolic dysfunction-associated steatotic liver disease 2025. Clin Mol Hepatol 2025; 31:S1-S31. [PMID: 39967303 PMCID: PMC11925433 DOI: 10.3350/cmh.2025.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Won Sohn
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Soon Sun Kim
- Department of Internal Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Jung Hee Kim
- Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Hwaseong, Korea
| | - Young-Joo Jin
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Gi-Ae Kim
- Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Korea
| | - Pil Soo Sung
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Young Chang
- Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Eun Joo Lee
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Won Lee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Miyoung Choi
- Clinical Evidence Research, National Evidence-Based Healthcare Collaborating Agency, Seoul, Korea
| | - Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Young Kul Jung
- Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Byoung Kuk Jang
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
197
|
Vali Y, van Dijk A, Lee J, Boursier J, Ratziu V, Yunis C, Schattenberg JM, Valenti L, Gomez MR, Schuppan D, Petta S, Allison M, Hartman ML, Porthan K, Dufour J, Bugianesi E, Gastadelli A, Derdak Z, Fournier‐Poizat C, Shumbayawonda E, Kalutkiewicz M, Yki‐Jarvinen H, Ekstedt M, Geier A, Trylesinski A, Francque S, Brass C, Pavlides M, Holleboom AG, Nieuwdorp M, Anstee QM, Bossuyt PM, the LITMUS investigators. Precision in Liver Diagnosis: Varied Accuracy Across Subgroups and the Need for Variable Thresholds in Diagnosis of MASLD. Liver Int 2025; 45:e16240. [PMID: 39865358 PMCID: PMC11771619 DOI: 10.1111/liv.16240] [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] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND AND AIMS The performance of non-invasive liver tests (NITs) is known to vary across settings and subgroups. We systematically evaluated whether the performance of three NITs in detecting advanced fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) varies with age, sex, body mass index (BMI), type 2 diabetes mellitus (T2DM) status or liver enzymes. METHODS Data from 586 adult LITMUS Metacohort participants with histologically characterised MASLD were included. The diagnostic performance of the Fibrosis-4 Index (FIB-4), enhanced liver fibrosis (ELF) and vibration-controlled transient elastography liver stiffness measurement (VCTE LSM) was evaluated. Performance was expressed as the area under the receiver operating characteristics curve (AUC). Thresholds for detecting advanced fibrosis (≥F3) were calculated for each NIT for fixed (high) sensitivity, specificity and predictive values. RESULTS Differences in AUC between all subgroups were small and statistically not significant, indicating comparable performance in detecting ≥F3, irrespective of these clinical factors. However, different thresholds were needed to achieve the same level of accuracy with each test. For example, for a fixed sensitivity and specificity, the thresholds for all three NITs were higher in patients with T2DM. Effects for sex, age and liver enzymes were less pronounced. CONCLUSIONS Performance of the selected NITs in detecting advanced liver fibrosis does not vary substantially with clinical characteristics. However, different thresholds have to be selected to achieve the same sensitivity, specificity and predictive values in the respective subgroups. Large prospective studies are called for to study NIT accuracy considering multiple patient characteristics.
Collapse
Affiliation(s)
- Yasaman Vali
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Anne‐Marieke van Dijk
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Jenny Lee
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Jerome Boursier
- Laboratoire HIFIH, UPRES EA 3859, SFR ICAT 4208Université d'AngersAngersFrance
- Service d'Hépato‐Gastroentérologie et Oncologie DigestiveCentre Hospitalier Universitaire d'AngersAngersFrance
| | - Vlad Ratziu
- Assistance Publique‐Hôpitaux de Paris, Hôpital Pitié SalpêtrièreICAN (Institute of Cardiometabolism and Nutrition), Sorbonne UniversityParisFrance
| | - Carla Yunis
- Pfizer Research and Development, Pfizer IncLake MaryFloridaUSA
| | - Jörn M. Schattenberg
- Department of Internal Medine IISaarland University Medical CenterHomburgGermany
- Saarland UniversitySaarbrückenGermany
| | - Luca Valenti
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanoItaly
- Precision MedicineBiological Resource Center Unit, Fondazione IRCCS Ca' Granda PoliclinicoMilanoItaly
| | - Manuel Romero Gomez
- Digestive Diseases UnitHospital Universitario Virgen del RocíoSevillaSpain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd)Instituto de Biomedicina de SevillaSevillaSpain
- Universidad de SevillaSevillaSpain
| | - Detlef Schuppan
- Department of Internal Medine IISaarland University Medical CenterHomburgGermany
- Institute of Translational ImmunologyUniversity Medical Center MainzMainzGermany
- Division of GastroenterologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Salvatore Petta
- Sezione di Gastroenterologia e Epatologia, PROMONISE DepartmentUniversità di PalermoPalermoItaly
| | - Mike Allison
- Liver Unit, Department of Medicine, Cambridge NIHR Biomedical Research CentreCambridge University NHS Foundation TrustCambridgeUK
| | - Mark L. Hartman
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndianaUSA
| | - Kimmo Porthan
- Department of MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | | | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastro‐Hepatology, A.O. Città della Salute e della Scienza di TorinoUniversity of TurinTurinItaly
| | | | - Zoltan Derdak
- GI DDU, Takeda Pharmaceuticals Company Ltd.CambridgeMassachusettsUSA
| | | | | | | | - Hannele Yki‐Jarvinen
- Department of MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Mattias Ekstedt
- Department of Health, Medicine and Caring SciencesLinköping UniversityLinköpingSweden
| | - Andreas Geier
- Division of Hepatology, Department Medicine IIWurzburg University HospitalWurzburgGermany
| | | | - Sven Francque
- Department of Gastroenterology Hepatology, and Laboratory of Experimental Medicine and Paediatrics, Antwerp University HospitalUniversity of AntwerpAntwerpBelgium
| | - Clifford Brass
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Michael Pavlides
- Radcliffe Department of Medicine and Oxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Adriaan G. Holleboom
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Quentin M. Anstee
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Newcastle NIHR Biomedical Research CentreNewcastle upon Tyne Hospitals NHS TrustNewcastle upon TyneUK
| | - Patrick M. Bossuyt
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | | |
Collapse
|
198
|
Macias J, Frias M, Pineda JA, Corona‐Mata D, Corma‐Gomez A, Rivero‐Juarez A, Santos M, García‐Deltoro M, Rivero A, Ricart‐Olmos C, Gonzalez‐Serna A, Real LM. Impact of Nonalcoholic Fatty Liver Disease on the Survival of People Living With HIV. Aliment Pharmacol Ther 2025; 61:550-557. [PMID: 39604811 PMCID: PMC11707644 DOI: 10.1111/apt.18413] [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: 11/23/2023] [Revised: 12/18/2023] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is an increasing concern for people living with HIV (PLWH). However, information on the impact of NAFLD on the prognosis of PLWH is very scarce. AIMS To investigate the influence of NAFLD on the overall and liver-related mortality in PLWH. METHODS PLWH followed in three Spanish centres were included in a prospective cohort at the date of the first transient elastography evaluation. Survival data were recorded, and the causes of death were centrally monitored. The risk of all-cause death and liver-related death was evaluated by applying time-to-event analyses. RESULTS A total of 2151 PLWH were included in the cohort and followed for a median (Q1-Q3) of 7.3 (3.5-10.4) years. There were 174 (8.1%) deaths. The probability of overall death and liver-related death was associated with liver stiffness measurement (LSM) and with FibroScan-AST (FAST) score. Among 844 PLWH with potential for NALFD, LSM was independently associated with all-cause mortality (adjusted hazard ratio [AHR], by 1 kPa increase: 1.06; 95% confidence interval [95% CI]: 1.04-1.08; p < 0.001). In a separate model and after adjustment, FAST score ≥ 0.67 was related to survival (AHR: 1.87; 95% CI: 1.40-2.50; p < 0.001). The AUROC (95% CI) of the models were based on LSM, 0.812 (0.739-0.885); and FAST, 0.825 (0.753-0.897) (p = 0.386). CONCLUSIONS For PLWH, advanced liver fibrosis increases the risk of overall death and liver-related death. LSM and the FAST score are similar predictors of survival for PLWH with potential for NAFLD.
Collapse
Affiliation(s)
- Juan Macias
- Unit of Infectious Diseases and MicrobiologyHospital Universitario Virgen de ValmeSevillaSpain
- Medicine DepartmentUniversity of SevillaSevilleSpain
- Instituto de Biomedicina de Sevilla (IBiS)SevillaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
| | - Mario Frias
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC)Universidad de Córdoba (UCO)CordobaSpain
| | - Juan Antonio Pineda
- Medicine DepartmentUniversity of SevillaSevilleSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
| | - Diana Corona‐Mata
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC)Universidad de Córdoba (UCO)CordobaSpain
| | - Anais Corma‐Gomez
- Unit of Infectious Diseases and MicrobiologyHospital Universitario Virgen de ValmeSevillaSpain
- Instituto de Biomedicina de Sevilla (IBiS)SevillaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
| | - Antonio Rivero‐Juarez
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC)Universidad de Córdoba (UCO)CordobaSpain
| | - Marta Santos
- Unit of Infectious Diseases and MicrobiologyHospital Universitario Virgen de ValmeSevillaSpain
- Instituto de Biomedicina de Sevilla (IBiS)SevillaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
| | | | - Antonio Rivero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Unit of Infectious Diseases, Hospital Universitario Reina Sofia, Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC)Universidad de Córdoba (UCO)CordobaSpain
| | - Carmen Ricart‐Olmos
- Unit of Infectious DiseasesHospital General Universitario de ValenciaValenciaSpain
| | - Alejandro Gonzalez‐Serna
- Unit of Infectious Diseases and MicrobiologyHospital Universitario Virgen de ValmeSevillaSpain
- Instituto de Biomedicina de Sevilla (IBiS)SevillaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Departamento de Fisiología, Facultad de FarmaciaUniversidad de SevillaSevillaSpain
| | - Luis Miguel Real
- Unit of Infectious Diseases and MicrobiologyHospital Universitario Virgen de ValmeSevillaSpain
- Instituto de Biomedicina de Sevilla (IBiS)SevillaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC)MadridSpain
- Department of Surgery, Biochemistry and Immunology, School of MedicineUniversity of MálagaMálagaSpain
| |
Collapse
|
199
|
Wang J, Zhao J, Zhong Y, He C, Hu F. Healthy Lifestyle and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Study of the Efficacy of Fatty Liver Regression. Clin Transl Gastroenterol 2025; 16:e00806. [PMID: 39729093 PMCID: PMC11845202 DOI: 10.14309/ctg.0000000000000806] [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: 09/13/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
INTRODUCTION Obesity is the primary cause of metabolic dysfunction-associated steatotic liver disease (MASLD). Healthy lifestyle management has potential value in the treatment of MASLD. METHODS A total of 150 patients with MASLD diagnosed at the Health Management Center of our hospital were enrolled and randomly divided into a traditional treatment (control group, n = 75) and a healthy lifestyle group (observation group, n = 75). All patients underwent a 3-month intervention. Data on general information, body composition, glucose metabolism, lipid metabolism, and inflammatory factors were analyzed. RESULTS The difference in the change in fatty liver grade was statistically significant ( P < 0.05). There were statistically significant differences in treatment efficiency for physical conditions ( P < 0.05), including body fat mass, body mass index, body weight, waist circumference, and waist-to-hip ratio. In addition, there were statistically significant differences in treatment efficiency for scales such as the Diet Rating Scale, Emotional Stress Scale, and Global Physical Activity Questionnaire ( P < 0.05). Differences in treatment efficiency for body fat parameters, including percentage of body fat, visceral fat area, aspartate aminotransferase, and diastolic blood pressure, were also statistically significant ( P < 0.05). After treatment, statistically significant differences were observed in interferon-γ, insulin, low-density lipoprotein cholesterol, triglycerides, and tumor necrosis factor-α ( P < 0.05). DISCUSSION Our study indicates that a healthy lifestyle can effectively promote the reduction of fatty liver grade in patients with MASLD, demonstrating positive effects in improving lipid metabolism and inflammatory responses in these patients.
Collapse
Affiliation(s)
- Jingwei Wang
- Health Management Center, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Jinli Zhao
- School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Yueyuan Zhong
- Health Management Center, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Chengyue He
- School of Marxism, Hengyang Normal University, Hengyang, China
| | - Fen Hu
- Health Management Center, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| |
Collapse
|
200
|
Younossi ZM, Kalligeros M, Henry L. Epidemiology of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S32-S50. [PMID: 39159948 PMCID: PMC11925440 DOI: 10.3350/cmh.2024.0431] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024] Open
Abstract
As the rates of obesity and type 2 diabetes (T2D) continue to increase globally, so does the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD). Currently, 38% of all adults and 7-14% of children and adolescents have MASLD. By 2040, the MASLD prevalence rate for adults is projected to increase to more than 55%. Although MASLD does not always develop into progressive liver disease, it has become the top indication for liver transplant in the United States for women and those with hepatocellular carcinoma (HCC). Nonetheless, the most common cause of mortality among patients with MASLD remains cardiovascular disease. In addition to liver outcomes (cirrhosis and HCC), MASLD is associated with an increased risk of developing de novo T2D, chronic kidney disease, sarcopenia, and extrahepatic cancers. Furthermore, MASLD is associated with decreased health-related quality of life, decreased work productivity, fatigue, increased healthcare resource utilization, and a substantial economic burden. Similar to other metabolic diseases, lifestyle interventions such as a heathy diet and increased physical activity remain the cornerstone of managing these patients. Although several obesity and T2D drugs are available to treat co-morbid disease, resmetirom is the only MASH-targeted medication for patients with stage 2-3 fibrosis that has approved by the Food and Drug Administration for use in the United States. This review discusses MASLD epidemiology and its related risk factors and outcomes and demonstrates that without further global initiatives, MASLD incidence could continue to increase.
Collapse
Affiliation(s)
- Zobair M. Younossi
- The Global NASH Council, Washington DC, USA
- Beatty Liver and Obesity Program, Inova Health System, Falls Church, VA, USA
- Center for Outcomes Research in Liver Disease, Washington DC, USA
| | - Markos Kalligeros
- Beth Israel Deaconess Medical Center, Harvard University, Cambridge, MA, USA
| | - Linda Henry
- The Global NASH Council, Washington DC, USA
- Beatty Liver and Obesity Program, Inova Health System, Falls Church, VA, USA
- Center for Outcomes Research in Liver Disease, Washington DC, USA
| |
Collapse
|