1
|
Hao X, Song H, Su X, Li J, Ye Y, Wang C, Xu X, Pang G, Liu W, Li Z, Luo T. Prophylactic effects of nutrition, dietary strategies, exercise, lifestyle and environment on nonalcoholic fatty liver disease. Ann Med 2025; 57:2464223. [PMID: 39943720 PMCID: PMC11827040 DOI: 10.1080/07853890.2025.2464223] [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: 08/14/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease and its prevalence has risen sharply. However, whether nutrition, dietary strategies, exercise, lifestyle and environment have preventive value for NAFLD remains unclear. METHODS Through searching 4 databases (PubMed, Web of Science, Embase and the Cochrane Library) from inception to January 2025, we selected studies about nutrition, dietary strategies, exercise, lifestyle and environment in the prevention of NAFLD and conducted a narrative review on this topic. RESULTS Reasonable nutrient intake encompassing macronutrients and micronutrients have an independent protective relationship with NAFLD. Besides, proper dietary strategies including mediterranean diet, intermittent fasting diet, ketogenic diet, and dietary approaches to stop hypertension diet have their inhibitory effects on the developmental process of NAFLD. Moreover, right exercises including walking, jogging, bicycling, and swimming are recommended for the prevention of NAFLD because they could effectively reduce weight, which is an important risk factor for NAFLD, and improve liver function. In addition, embracing a healthy lifestyle including reducing sedentary behavior, not smoking, sleeping well and brushing teeth regularly is integral since it not only could reduce the risk of NAFLD but also significantly contribute to overall prevention and control. Finally, the environment, including the social and natural environments, plays a potential role in NAFLD prevention. CONCLUSION Nutrition, dietary strategies, exercise, lifestyle and environment play an important role in the prevention of NAFLD. Moreover, this review offers comprehensive prevention recommendations for people at high risk of NAFLD.
Collapse
Affiliation(s)
- Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Hao Song
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Xin Su
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Jian Li
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Youbao Ye
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Cailiu Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Xiao Xu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Guanglong Pang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Wenxiu Liu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Zihan Li
- Department of clinical medicine, The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Tian Luo
- The Institute for Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
2
|
Choullamy T, Kaadi L, Bezdikian A, Hachem S, Hachem K. Liver Fat Quantification With Ultrasound: The Influence of the Size of the Region of Interest on Attenuation Coefficient. Ultrasound Q 2025; 41:e00712. [PMID: 40173292 DOI: 10.1097/ruq.0000000000000712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
ABSTRACT Noninvasive assessment of liver fat content is crucial due to the high global prevalence of nonalcoholic fatty liver disease. Algorithms based on ultrasound (US) attenuation coefficient (AC) for estimating liver fat content are commercially available, but a lack of consensus exists regarding the best estimation protocol. The aim of our study was to evaluate the influence of the size of the region of interest (ROI) on the US AC.A prospective study was conducted. An abdominal US was done for 86 outpatients. A sampling box was positioned within the liver parenchyma, approximately 2 cm beneath the liver capsule with a ROI, measuring about 2 × 4 cm and then 4 × 5 cm, precisely placed at the center of this sampling box. Five readings of the AC were captured, and the average of these measurements was employed to assess the severity of hepatic steatosisA statistically significant difference between AC with 2 different ROI sizes was shown (P < 0.001) with AC values with 2 × 4 cm ROI were higher than those obtained with 4 × 5 cm ROI (AC mean 0.668 VS 0.653). However, the agreement between AC values obtained with 2 different ROI sizes was excellent (correlation coefficient 0.941)An ROI size dependence is observed in the measurement of AC in the liver. A standardized acquisition protocol with a fixed size of the ROI needs to be developed to minimize differences in AC measurements and to assess changes in serial measurements reliably.
Collapse
Affiliation(s)
- Theresia Choullamy
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| | - Lea Kaadi
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| | - Aren Bezdikian
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | - Samir Hachem
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | - Kamal Hachem
- Medical Imaging Department, Hôtel-Dieu de France, Alfred Naccache Boulevard, Achrafieh, Beirut, Lebanon
| |
Collapse
|
3
|
Abbaszadeh M, Hosseinpanah F, Tohidi M, Karimpour Reyhan S, Mahdavi M, Valizadeh M. Sex-Specific Impact of Metabolic Dysfunction-Associated Fatty Liver Disease on Incident Cardiovascular Diseases and Mortality. Endocrinol Diabetes Metab 2025; 8:e70035. [PMID: 40140729 PMCID: PMC11946537 DOI: 10.1002/edm2.70035] [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: 09/07/2024] [Revised: 12/30/2024] [Accepted: 02/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND AND AIMS Considering recent revisions in the nomenclature for fatty liver disease, alongside limited data on sex-specific differences in its cardiovascular/mortality outcomes, this study aims to investigate the prevalence and impact of metabolic-associated fatty liver disease (MAFLD) on cardiovascular disease (CVD) and mortality in men and women over a 12-year follow-up period. METHODS In this large population-based cohort study, 7101 individuals aged ≥ 30 were enrolled. The prevalence of MAFLD was investigated in both genders. After excluding individuals with a history of previous CVD, 6331 participants were followed up for CVD and mortality over 12 years. Steatosis was defined as fatty liver index (FLI) ≥ 60. Multivariate-adjusted hazard ratios (HRs) were calculated for CVD and mortality. RESULTS The prevalence of MAFLD was 43.2%, higher in men (46.5%) than women (40.6%). Men with MAFLD (47.7 ± 12.1) were younger than women (52.2 ± 11.1). In the 12-year follow-up of 6331 individuals, multivariable-adjusted CVD HRs for MAFLD were 1.36 (1.10-1.67) in men and 1.48 (1.16-1.88) in women. Adjusted mortality HRs were 1.17 (0.86-1.59) and 1.38 (1.00-1.91) in men and women, respectively. Among patients with MAFLD, a subgroup with diabetes faced the highest hazard for CVD and mortality. CONCLUSION This study found that MAFLD is more common in men at a younger age. Despite the higher prevalence in men, women with MAFLD face a greater risk of cardiovascular events and mortality. Findings highlight the importance of gender-specific considerations in primary prevention programmes for MAFLD-related cardiovascular disease and mortality.
Collapse
Affiliation(s)
- Mahsa Abbaszadeh
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Farhad Hosseinpanah
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical SciencesTehranIran
| | - Sahar Karimpour Reyhan
- Endocrinology and Metabolism Research CenterImam Khomeini Hospital Complex, Tehran University of Medical SciencesTehranIran
| | - Maryam Mahdavi
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| | - Majid Valizadeh
- Obesity Research CenterResearch Institute for Endocrine Sciences, Shahid Beheshti University of Medical ScienceTehranIran
| |
Collapse
|
4
|
Kobayashi Y, Yamashita Y, Kimura T, Iwadare T, Okumura T, Wakabayashi SI, Kobayashi H, Sugiura A, Joshita S, Umemura T. Hormone replacement therapy for steatotic liver management after surgical menopause. Clin J Gastroenterol 2025; 18:352-356. [PMID: 39760964 DOI: 10.1007/s12328-024-02090-y] [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: 10/16/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Although steatotic liver onset after natural menopause has been reported, evidence on the clinical course and treatment options for steatotic liver after surgical menopause is scarce. A 34-year-old woman with a history of severe obesity presented to our department with liver dysfunction following total hysterectomy and bilateral oophorectomy. Her serum estradiol level was notably low at 22 pg/mL, and a liver biopsy revealed significant fatty degeneration, lobular inflammation, hepatocyte ballooning, and stage F1 fibrosis. These findings supported a diagnosis of steatotic liver disease following surgical menopause. Subsequent initiation of hormone replacement therapy (HRT) with estrogen led to rapid improvements in liver function and steatotic liver symptoms. Steatotic liver disease should be considered in cases of liver impairment in postoperative menopausal patients, for which HRT represents a promising treatment option.
Collapse
Affiliation(s)
- Yoshiaki Kobayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Yuki Yamashita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan.
| | - Takefumi Kimura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan
| | - Takanobu Iwadare
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Taiki Okumura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Shun-Ichi Wakabayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Hiroyuki Kobayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Ayumi Sugiura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Satoru Joshita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Takeji Umemura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan
| |
Collapse
|
5
|
Liu L, Deng Y, Yang L, Wang M, Lai Y. Comparison of efficacy and safety of pioglitazone and SGLT2 inhibitors in treating Asian patients in MASLD associated with type 2 diabetes: A meta-analysis. J Diabetes Complications 2025; 39:108998. [PMID: 40043473 DOI: 10.1016/j.jdiacomp.2025.108998] [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: 07/03/2024] [Revised: 01/09/2025] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE To comprehensively evaluate the therapeutic efficacy of pioglitazone and SGLT2 inhibitors (SGLT2i) in patients with MASLD and Type 2 Diabetes(T2DM). METHODS Electronic databases, including Web of Science, PubMed, the Cochrane Library, China National Knowledge Internet (CNKI), Wan-Fang Digital Database, and China Science and Technology Journal Database (VIP) were searched from inception to December 2024. Two reviewers independently assessed study eligibility, performed continuous data extraction, and independently evaluated bias risks. Liver ultrasonography, computed tomography (CT), and biochemical indices were utilized to determine the impact of treatment in both groups. Improvement in liver biomarkers and fibrosis as primary outcome indicators; Improvement in body composition, metabolic parameters, glucose parameters, and incidence of adverse effects as a secondary outcome indicator. For continuous variables, mean and standard deviation (SD) were extracted. RevMan 5.4 software was used to systematically analyze the literature, including heterogeneity testing, odds ratios (OR) calculation, and 95 % confidence intervals (CI) for each influencing factor. RESULTS Nine randomly controlled trials with 755 Asian participants were included. Our research showed that SGLT2i was more effective than pioglitazone in improving fibrosis-4 score (SMD 0.41 [95%CI 0.18,0.64] p = 0.005), visceral fat area (SMD 0.34 [95%CI 0.14,0.54] p = 0.0007), BMI (SMD 0.29 [95%CI 0.03,0.56] p = 0.03), and low-density lipoprotein levels (SMD 0.21 [95%CI 0.04,0.38] p = 0.01). In contrast, no statistically significant differences were observed in other outcomes. CONCLUSIONS Our study demonstrated that in patients with MASLD and T2DM, SGLT2i was more effective overall in improving liver fibrosis, blood lipids, liver fat, and body composition in the short term. These findings establish a theoretical basis for safe and rational drug use in clinical practice. Additionally, they may contribute to new insights into the pathogenesis of MASLD and type 2 diabetes and drug discovery and development efforts.
Collapse
Affiliation(s)
- Lingyan Liu
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yongkun Deng
- Department of Medical Protection Center, The 926th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kaiyuan 661600, Yunnan Province, China.
| | - Lijuan Yang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Miaojiao Wang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yong Lai
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali 671000, Yunnan Province, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali 671000, Yunnan Province, China; College of Pharmacy Dali University, Dali 671000, Yunnan Province, China.
| |
Collapse
|
6
|
Younossi ZM, Paik JM, Henry L, Stepanova M, Nader F. Pharmaco-Economic Assessment of Screening Strategies for High-Risk MASLD in Primary Care. Liver Int 2025; 45:e16119. [PMID: 39373093 DOI: 10.1111/liv.16119] [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: 05/30/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND AND AIMS Several scientific associations recommend a sequential combination of non-invasive tests (NITs) to identify high-risk MASLD patients but their cost-effectiveness is unknown. METHODS A cost-utility model was developed to assess the incremental cost-effectiveness ratio (ICER) of recommended screening strategies for patients with clinically suspected MASLD, specifically those with type 2 diabetes (T2D) and obesity with multiple cardiometabolic risk factors which will be initiated in primary care. Six screening strategies were assessed, using either vibration-controlled transient elastography (VCTE) or the enhanced liver fibrosis (ELF) test as a second-line test following an initial Fibrosis-4 (FIB-4) assessment as the first line NIT. The model included treatment effects of resmetirom for metabolic dysfunction-associated steatohepatitis (MASH) patients with F2 or F3 fibrosis. RESULTS All screening strategies for high-risk MASLD in US incurred additional costs compared to no screening, ranging from $13 587 to $14 730 per patient with T2D and $14 274 to $15 661 per patient with obesity. However, screening reduced long-term costs, ranging from $22 150 to $22 279 per patient with T2D and $13 704 to $13 705 per patient with obesity, compared to $24 221 and $14 956 for no screening, respectively. ICERs ranged from $26 913 to $27 884 per QALY for T2D patients and $23 265 to $24 992 per QALY for patients with obesity. While ICERs were influenced by VCTE availability, they remained cost-effective when using ELF as the second-line test. Our findings remain robust across a range of key parameters. CONCLUSIONS Screening for high-risk MASLD is cost-effective according to recent guidelines. Implementing these screening strategies in primary care should be considered.
Collapse
Affiliation(s)
- Zobair M Younossi
- The Global NASH Council, Washington, DC, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - James M Paik
- The Global NASH Council, Washington, DC, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
| | - Linda Henry
- The Global NASH Council, Washington, DC, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Maria Stepanova
- The Global NASH Council, Washington, DC, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, 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 Program, Inova Health System, Falls Church, Virginia, USA
- Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| |
Collapse
|
7
|
Villanueva C, Tripathi D, Bosch J. Preventing the progression of cirrhosis to decompensation and death. Nat Rev Gastroenterol Hepatol 2025; 22:265-280. [PMID: 39870944 DOI: 10.1038/s41575-024-01031-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/29/2025]
Abstract
Two main stages are differentiated in patients with advanced chronic liver disease (ACLD), one compensated (cACLD) with an excellent prognosis, and the other decompensated (dACLD), defined by the appearance of complications (ascites, variceal bleeding and hepatic encephalopathy) and associated with high mortality. Preventing the progression to dACLD might dramatically improve prognosis and reduce the burden of care associated with ACLD. Portal hypertension is a major driver of the transition from cACLD to dACLD, and a portal pressure of ≥10 mmHg defines clinically significant portal hypertension (CSPH) as the threshold from which decompensating events may occur. In recent years, innovative studies have provided evidence supporting new strategies to prevent decompensation in cACLD. These studies have yielded major advances, including the development of noninvasive tests (NITs) to identify patients with CSPH with reasonable confidence, the demonstration that aetiological therapies can prevent disease progression and even achieve regression of cirrhosis, and the finding that non-selective β-blockers can effectively prevent decompensation in patients with cACLD and CSPH, mainly by reducing the risk of ascites, the most frequent decompensating event. Here, we review the evidence supporting new strategies to manage cACLD to prevent decompensation and the caveats for their implementation, from patient selection using NITs to ancillary therapies.
Collapse
Affiliation(s)
- Càndid Villanueva
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Ministerio de Sanidad, Madrid, Spain.
| | - Dhiraj Tripathi
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham Health Partners, Birmingham, UK
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jaume Bosch
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Ministerio de Sanidad, Madrid, Spain
- Department of Visceral Surgery and Medicine (Hepatology), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Konings LAM, Miguelañez-Matute L, Boeren AMP, van de Luitgaarden IAT, Dirksmeier F, de Knegt RJ, Tushuizen ME, Grobbee DE, Holleboom AG, Cabezas MC. Pharmacological treatment options for metabolic dysfunction-associated steatotic liver disease in patients with type 2 diabetes mellitus: A systematic review. Eur J Clin Invest 2025; 55:e70003. [PMID: 39937036 DOI: 10.1111/eci.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is closely related to type 2 diabetes mellitus (T2DM) through a common root in insulin resistance. The more severe stage, metabolic dysfunction-associated steatohepatitis (MASH), increases the risk for cardiovascular complications, liver cirrhosis and hepatocellular carcinoma. Several trials investigating established antidiabetic-drugs in patients with T2DM and MASLD have yielded promising results. Therefore, we aimed to systematically review the effect of T2DM-drug treatment on MALSD parameters. METHODS Medical databases were searched until January 2025 for controlled trials in patients with T2DM and MASLD/MASH. Studies that evaluated the effect of T2DM-medication on the severity of MASLD/MASH in T2DM patients were included. The quality of the studies was assessed by three independent reviewers using a set of Cochrane risk-of-bias tools. RESULTS Of 1748 references, 117 studies fulfilled the inclusion-criteria and were assessed for eligibility in full-text. Fifty-two articles were included. Data included a total of 64.708 patients and study populations ranged from 9 to 50.742. Heterogeneity in study-design and analysis hampered the comparability of the results. Most evidence was present for GLP-1 receptor agonists, SGLT2-inhibitors and PPAR-γ-agonists for regression of liver fibrosis and MASH. CONCLUSION Studies on the value of T2DM-drug treatment in the improvement of MASLD vary significantly in study design, size and quality. GLP-1 receptor agonists, PPAR-γ-agonists, SGLT2-inhibitors may all be preferred pharmacological interventions for patients with MASLD/MASH and T2DM. Newer agents like dual GLP-1/GIP or triple GLP-1/GIP/Glucagon agonists will likely play an important role in the treatment of MASLD/MASH in the near future.
Collapse
Affiliation(s)
- Laura A M Konings
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
- Department of Internal Medicine and Endocrinology, Erasmus MC, Rotterdam, the Netherlands
| | | | - Anna M P Boeren
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
| | | | - Femme Dirksmeier
- Department of Gastroenterology and Hepatology, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
| | - Rob J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, the Netherlands
| | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, LUMC, Leiden, the Netherlands
| | | | | | - Manuel Castro Cabezas
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, the Netherlands
- Department of Internal Medicine and Endocrinology, Erasmus MC, Rotterdam, the Netherlands
- Julius Clinical, Zeist, the Netherlands
| |
Collapse
|
9
|
Cannella R, Agnello F, Porrello G, Spinello AU, Infantino G, Pennisi G, Cabibi D, Petta S, Bartolotta TV. Performance of ultrasound-guided attenuation parameter and 2D shear wave elastography in patients with metabolic dysfunction-associated steatotic liver disease. Eur Radiol 2025; 35:2339-2350. [PMID: 39373742 PMCID: PMC11914239 DOI: 10.1007/s00330-024-11076-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 07/06/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE To assess the performance and the reproducibility of ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) in patients with biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS This study included consecutive adult patients with MASLD who underwent ultrasound with UGAP, 2D-SWE and percutaneous liver biopsy. The median values of 12 consecutive UGAP measurements were acquired by two independent radiologists (R1 and R2). Hepatic steatosis was graded by liver biopsy as: (0) < 5%; (1) 5-33%; (2) > 33-66%; (3) > 66%. Areas under the curve (AUCs) were calculated to determine the diagnostic performance. Inter- and intra-observer reliability was assessed with intraclass correlation coefficient (ICC). RESULTS A hundred patients (median age 55.0 years old) with MASLD were prospectively enrolled. At histopathology, 70 and 42 patients had grade ≥ 2 and 3 steatosis, respectively. Median UGAP was 0.78 dB/cm/MHz (IQR/Med: 5.55%). For the diagnosis of grade ≥ 2 steatosis, the AUCs of UGAP were 0.828 (95% CI: 0.739, 0.896) for R1 and 0.779 (95% CI: 0.685, 0.856) for R2. The inter- and intra-operator reliability of UGAP were excellent, with an ICC of 0.92 (95% CI: 0.87-0.95) and 0.95 (95% CI: 0.92-0.96), respectively. The median liver stiffness was 6.76 kPa (IQR/Med: 16.30%). For the diagnosis of advanced fibrosis, 2D-SWE had an AUC of 0.862 (95% CI: 0.757, 0.934), and the optimal cutoff value was > 6.75 kPa with a sensitivity of 80.6% and a specificity of 75.7%. CONCLUSION UGAP and 2D-SWE provide a good performance for the staging of steatosis and fibrosis in patients with MASLD with an excellent intra-operator reliability of UGAP. KEY POINTS Question How well do ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) perform for quantifying hepatic steatosis and fibrosis? Findings UGAP had a maximum AUC of 0.828 for the diagnosis of grade ≥ 2 steatosis, and 2D-SWE had an AUC of 0.862 for diagnosing advanced fibrosis. Clinical relevance UGAP and 2D-SWE allow rapid, reproducible, and accurate quantification of hepatic steatosis and fibrosis that can be used for the noninvasive assessment of patients with metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy.
| | - Francesco Agnello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giorgia Porrello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Alessandro Umberto Spinello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giuseppe Infantino
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Grazia Pennisi
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Daniela Cabibi
- Unit of Anatomic Pathology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| |
Collapse
|
10
|
Beyer C, Andersson A, Shumbayawonda E, Alkhouri N, Banerjee A, Pandya P, Harisinghani M, Corey K, Dennis A, Pansini M. Quantitative MRI for Monitoring Metabolic Dysfunction-Associated Steatotic Liver Disease: A Test-Retest Repeatability Study. J Magn Reson Imaging 2025; 61:1947-1955. [PMID: 39319470 PMCID: PMC11896917 DOI: 10.1002/jmri.29610] [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: 06/28/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Quantitative magnetic resonance imaging metrics iron-corrected T1 (cT1) and liver fat from proton density fat-fraction (PDFF) are both commonly used as noninvasive biomarkers for metabolic dysfunction-associated steatohepatitis (MASH); however, their repeatability in this population has rarely been characterized. PURPOSE To quantify the variability of cT1 and liver fat fraction from PDFF in patients with biopsy-confirmed metabolic dysfunction-associated steatotic liver disease (MASLD) and MASH. STUDY TYPE Prospective, single center. POPULATION Twenty-one participants (female = 11, mean age 53 ± 24 years) with biopsy-confirmed MASLD, including 6 with MASH and fibrosis ≥2. FIELD STRENGTH/SEQUENCE 3 T; T1 and T2* mapping for the generation of cT1 (shMOLLI: CardioMaps and 2D MDE, T1map-FIESTA and LMS MOST: StarMap, 2D Multi-Echo FSPGR) and magnitude-only PDFF sequence for liver fat quantification (LMS IDEAL: StarMap, 2D Multi-Echo FSPGR). ASSESSMENT T1 mapping and PDFF scans were performed twice on the same day for all participants (N = 21), with an additional scan 2-4 weeks later for MASH patients with fibrosis ≥2 (N = 6). Whole liver segmentation masks were generated semi-automatically and average pixel counts within these masks were used for the calculation of cT1 and liver fat fraction. STATISTICAL TESTS Bland-Altman analysis for repeatability coefficient (RC) and 95% limits of agreement (LOA) and intraclass correlation coefficient (ICC). RESULTS Same-day RC was 32.1 msec (95% LOA: -36.6 to 24.2 msec) for cT1 and 0.6% (95% LOA: -0.5% to 0.7%) for liver fat fraction; the ICCs were 0.98 (0.96-0.99) and 1.0, respectively. Short-term RC was 65.2 msec (95% LOA: -63.8 to 76.5 msec) for cT1 and 2.6% (95% LOA: -2.8% to 3.1%) for liver fat fraction. DATA CONCLUSION In participants with MASLD and MASH, cT1 and liver fat fraction measurements show excellent test-retest repeatability, supporting their use in monitoring MASLD and MASH. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
| | | | | | | | - Ami Banerjee
- University College London Hospitals NHS TrustLondonUK
- Institute of Health InformaticsUniversity College LondonLondonUK
- Barts Health NHS Trust, The Royal London HospitalLondonUK
| | | | | | | | | | - Michele Pansini
- Clinica Di Radiologia EOC, Istituto Di Imaging Della Svizzera Italiana (IIMSI), Ente Ospedaliero CantonaleLuganoSwitzerland
- John Radcliffe Hospital, Oxford University Hospitals NHS Foundation TrustOxfordUK
| |
Collapse
|
11
|
Lefere S, Mosca A, Hudert C, Dupont E, Fitzpatrick E, Kyrana E, Dhawan A, Kalveram L, Pietrobattista A, Geerts A, De Bruyne R. Development and validation of pFIB scores for exclusion of significant liver fibrosis in pediatric MASLD. Hepatology 2025; 81:1276-1287. [PMID: 39028885 DOI: 10.1097/hep.0000000000001016] [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: 04/03/2024] [Accepted: 07/01/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent pediatric liver disease, yet accurate risk scores for referral of children/adolescents with suspected clinically significant liver fibrosis are currently lacking. APPROACH AND RESULTS Clinical and biochemical variables were collected in a prospective cohort of 327 children and adolescents with severe obesity, in whom liver fibrosis was evaluated by transient elastography. Logistic regression was performed to establish continuous (pFIB-c) and simplified (pFIB-6) diagnostic scores that accurately exclude significant (≥F2) fibrosis. Performance for each was compared to established noninvve fibrosis scores. These scores were validated in elastography (n=504) and multiple biopsy-proven MASLD (n=261) cohorts. Patient sex, ethnicity, weight z-score, homeostatic model assessment of insulin resistance index, ALT, and presence of hypertension were included in the scores. The pFIB-c and pFIB-6 exhibited good discriminatory capacity (c-statistic of 0.839 and 0.826), outperforming existing indices. Negative predictive values were >90% for both scores in the derivation and elastography validation cohorts. Performance in the histological cohorts varied (AUROCs for the pFIB-c between 0.710 and 0.770), as the scores were less accurate when applied to populations in tertiary referral centers characterized by a high prevalence of significant fibrosis and high ALT levels. CONCLUSIONS Analyzing several cohorts totaling approximately 1100 children and adolescents, we developed novel risk scores incorporating readily available clinical variables. In accordance with the aim of excluding pediatric MASLD-associated fibrosis, the scores performed better in nonselected cohorts of children and adolescents living with obesity than in patients referred to tertiary liver units.
Collapse
Affiliation(s)
- Sander Lefere
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent, Belgium
| | - Antonella Mosca
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Christian Hudert
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Emer Fitzpatrick
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, United Kingdom
- Department of Gastroenterology, Hepatology and Nutrition, Children's Health Ireland and University College Dublin, Ireland
| | - Eirini Kyrana
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Anil Dhawan
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, United Kingdom
| | - Laura Kalveram
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Pietrobattista
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent, Belgium
| | - Ruth De Bruyne
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Ito H, Tsugami E, I C, Miura S, Matsumoto S, Inoue H, Antoku S, Yamasaki T, Mori T, Togane M. Benefits and disadvantages of combination therapy with imeglimin and metformin in patients with type 2 diabetes. Expert Opin Pharmacother 2025; 26:773-781. [PMID: 40151916 DOI: 10.1080/14656566.2025.2486354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/26/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND We retrospectively examined the benefits and disadvantages of adding imeglimin to metformin therapy in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS Ninety-four patients with type 2 diabetes who had been administered imeglimin were included in the safety analysis set (SAS). Sixty-four patients who had been treated with imeglimin for over 6 months were included in the full analysis set (FAS). The primary outcome was the change in HbA1c levels in the FAS. The secondary outcomes were gastrointestinal adverse events (AEs) in the SAS and changes in body weight and FIB-4 in theFAS. RESULTS In the SAS, gastrointestinal AEs occurred in 15 of 40 (38%) metformin users and 6 of 54 (11%) non-users. In the FAS, HbA1c levels were significantly reduced in both metformin users (n = 27, baseline, 8.5 ± 1.1%;6 months, 7.7 ± 1.2%) and non-users (n = 37, baseline, 8.0 ± 0.9%; 6 months,7.4 ± 0.9%). While body weight (from 72.0 ± 20.5 kg to 70.9 ± 20.8 kg) and FIB-4 (from 1.27 ± 0.57 to 1.17 ± 0.49) significantly decreased in metformin users, they did not significantly differ in non-users. CONCLUSIONS Adding imeglimin to metformin therapy demonstrated favorable reductions in HbA1c, body weight, and FIB-4 in patients with type 2 diabetes, although it was associated with the incidence of gastrointestinal AEs. TRIAL REGISTRATION UMIN000055241.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Emiko Tsugami
- Department of Pharmacy, Edogawa Hospital, Tokyo, Japan
| | - Chiaki I
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Shun Miura
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Suzuko Matsumoto
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Hideyuki Inoue
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Shinichi Antoku
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Tomoko Yamasaki
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Toshiko Mori
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| | - Michiko Togane
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Tokyo, Japan
| |
Collapse
|
13
|
Gleeson D, Bornand R, Brownlee A, Dhaliwal H, Dyson JK, Hails J, Henderson P, Kelly D, Mells GF, Miquel R, Oo YH, Sutton A, Yeoman A, Heneghan MA. British Society of Gastroenterology guidelines for diagnosis and management of autoimmune hepatitis. Gut 2025:gutjnl-2024-333171. [PMID: 40169244 DOI: 10.1136/gutjnl-2024-333171] [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: 06/22/2024] [Accepted: 10/22/2024] [Indexed: 04/03/2025]
Abstract
Autoimmune hepatitis (AIH) is a chronic inflammatory liver disease which, if untreated, often leads to cirrhosis, liver failure and death. The last British Society of Gastroenterology (BSG) guideline for the management of AIH was published in 2011. Since then, our understanding of AIH has advanced in many areas. This update to the previous guideline was commissioned by the BSG and developed by a multidisciplinary group. The aim of this guideline is to review and summarise the current evidence, in order to inform and guide diagnosis and management of patients with AIH and its variant syndromes. The main focus is on AIH in adults, but the guidelines should also be relevant to older children and adolescents.
Collapse
Affiliation(s)
- Dermot Gleeson
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Science, University of Sheffield, Sheffield, UK
| | | | | | - Harpreet Dhaliwal
- Department of Gastroenterology, Manchester Royal Infirmary, Manchester, UK
| | - Jessica K Dyson
- Liver Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Janeane Hails
- Division of Gastroenterology and Hepatology, Addenbrooke's Hospital, Cambridge, UK
| | - Paul Henderson
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Deirdre Kelly
- Birmingham Women's & Children's Hospital, Birmingham, UK
- University of Birmingham, Birmingham, UK
| | - George F Mells
- Division of Gastroenterology and Hepatology, Addenbrooke's Hospital, Cambridge, UK
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Rosa Miquel
- Liver Histopathology Laboratory, Institute of Liver Studies, King's College London, London, UK
| | - Ye H Oo
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- NIHR Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
- Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) centre, Birmingham, UK
| | - Anthea Sutton
- Sheffield Centre for Health and Related Research, The University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
14
|
de la Hoz Gil L, Seijas Martínez-Echevarría V, Lozano Estevan MDC. [Metabolic associated steatotic liver disease (MASLD) and hepatic fibrosis. Score proposal with FIB4+NFS+APRI]. NUTR HOSP 2025. [PMID: 40195792 DOI: 10.20960/nh.05252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
. INTRODUCTION liver fibrosis (FH) is one of the main risks and mortality factors for many patients. Metabolic syndrome, MASLD, alcoholism or hepatotropic viruses are the main causes of the development of FH. Due to the wide range of patients affected, FH screening is the order of the day. Furthermore, it is of enormous nutritional interest given that the only effective treatment today is diet and physical exercise. Metabolic syndrome and MASLD are closely linked. The prevalence of these is increasing and consequently, so is FH. Therefore, finding less invasive diagnostic strategies than liver biopsy (gold standard) is a priority. OBJECTIVE this study analyzes the relationship between age, sex, diabetes mellitus, hypertension, obesity and dyslipidemia with the development of FH. MATERIAL AND METHODS the diagnostic capacity of the main non-invasive markers available today is analyzed, as well as their confounding factors. Finally, a score with FIB4, NFS and APRI is proposed to improve their individual diagnostic capacity. RESULTS the prevalence of FH in the study is 44 %. Diabetes mellitus and hypertension are significant in the development of FH. For their part, FIB4, NFS and APRI show acceptable specificity, although in all of them platelets are an important confounding factor. CONCLUSIONS the score of the three markers improves specificity (89.28 %), leaving fewer indeterminates than any of the indices. Therefore, it improves the diagnostic capacity compared to individual use.
Collapse
|
15
|
Ito H, Someya R, Koyanagi T, I C, Miura S, Matsumoto S, Inoue H, Antoku S, Yamasaki T, Mori T, Togane M. Effect of luseogliflozin on liver fibrosis differs depending on alcohol consumption in patients with type 2 diabetes. J Diabetes Investig 2025. [PMID: 40123308 DOI: 10.1111/jdi.70032] [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: 11/13/2024] [Revised: 02/03/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
AIM Changes in FIB-4 levels after the initiation of luseogliflozin therapy were compared between patients with type 2 diabetes according to the presence or absence of alcohol consumption. METHODS A total of 192 patients with type 2 diabetes who continued luseogliflozin therapy for over 12 months were retrospectively investigated. The primary outcome was the change in FIB-4. The secondary outcomes were changes in HbA1c, body weight, and serum albumin concentration. A current drinker was defined as an individual consuming >20 g ethanol equivalent/day. Patients were classified according to their risk of developing liver fibrosis into the low-risk (FIB-4 < 1.3) and intermediate/high-risk (FIB-4 ≥ 1.3) groups. RESULTS In the low-risk group, while FIB-4 increased dramatically from 0.91 ± 0.30 at the baseline to 1.14 ± 0.34 at 12 months in drinkers (n = 27), non-drinkers (n = 79) showed no significant change (0.87 ± 0.22-0.91 ± 0.26). In the intermediate/high-risk group (n = 63), although the FIB-4 in drinkers (n = 23) showed no significant change (2.18 ± 1.00-2.16 ± 0.93), it significantly decreased from 2.10 ± 0.87 to 1.80 ± 0.68 in non-drinkers (n = 63). In both the low- and intermediate/high-risk groups, HbA1c and body weight significantly decreased in both drinkers and non-drinkers. Serum albumin concentrations significantly increased in both drinkers and non-drinkers in the low-risk group. Although serum albumin concentration did not significantly change in drinkers, it dramatically increased in non-drinkers in the intermediate/high-risk group. CONCLUSIONS HbA1c levels and body weight decreased in patients with type 2 diabetes after initiating luseogliflozin therapy, regardless of drinking habits. However, it is desirable to limit alcohol consumption when considering its effects on liver fibrosis.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Ryota Someya
- Department of Pharmacy, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Tomoko Koyanagi
- Secretarial Section, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Chiaki I
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Shun Miura
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Suzuko Matsumoto
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Hideyuki Inoue
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Shinichi Antoku
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Tomoko Yamasaki
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Toshiko Mori
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| | - Michiko Togane
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa, Tokyo, Japan
| |
Collapse
|
16
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [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: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
17
|
Cheng B, Su X, He J, Gu Y, Chen M, Wei Y, Yi Y, Chen P, Lin X, Li T, Xu C, Liu Q, Li B. A Mendelian randomization study reveals a causal association between NASH and the risk of atrial fibrillation. Front Endocrinol (Lausanne) 2025; 16:1390259. [PMID: 40171195 PMCID: PMC11958169 DOI: 10.3389/fendo.2025.1390259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background Epidemiological evidence suggests that non-alcoholic fatty liver disease (NAFLD) may increase the risk of atrial fibrillation (AF). However, the findings are inconsistent, and the causality remains to be established. Methods We conducted two-step, two-sample Mendelian randomization (MR) analysis to assess the association between genetically predicted NAFLD (i.e. chronically elevated serum alanine aminotransferase levels [cALT], imaging-based and biopsy-confirmed NAFLD) and AF. Subsequently, we further performed Mendelian randomization to investigate the causal relationship between non-alcoholic steatohepatitis (NASH), a subtype of NAFLD, and AF. The inverse variance weighted (IVW) method was used as the primary approach to reveal the potential causation between the exposure and outcome. Results There was no significant causal association between NAFLD diagnosed based on cALT, confirmed by imaging, or verified by biopsy, and an increased risk of atrial fibrillation. Furthermore, the results of the IVW method revealed a positive causal effect of NASH on AF (OR=1.113, 95% CI=1.025-1.209, P = 0.011). In the reverse analysis, however, no evidence supported a significant genetic association between AF and NASH (OR=0.974, 95% CI=0.934-1.016, P = 0.214). Conclusion A causal relationship existed between NASH and the risk of AF. However, no significant genetic association has been observed between NAFLD and AF risk. This suggests that managing the progression of NAFLD may hold potential value in preventing the onset of AF.
Collapse
Affiliation(s)
- Biwei Cheng
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xuekang Su
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jue He
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yanghui Gu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Mingtai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yi Wei
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yumeng Yi
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peiying Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaojuan Lin
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Tao Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Chong Xu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Qiang Liu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Biao Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
18
|
Mohamed AA, Christensen DM, Mohammad M, Gluud LL, Knop FK, Biering-Sørensen T, Torp-Pedersen C, Andersson C, Schou M, Gislason G. The prognostic role of Fibrosis-4 score in heart failure with reduced ejection fraction. Int J Cardiol 2025; 429:133174. [PMID: 40107387 DOI: 10.1016/j.ijcard.2025.133174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/22/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Heart failure with reduced ejection fraction (HFrEF) and metabolic dysfunction-associated steatotic liver disease (MASLD) are both associated with liver fibrosis. HFrEF patients may develop liver fibrosis due to hepatic congestion, MASLD, or a combination of both. The Fibrosis-4 (FIB-4) score calculated using age, aspartate aminotransferase, alanine aminotransferase, and platelet count, serves as a screening tool for advanced liver fibrosis. This study examines the association between the FIB-4 score and all-cause mortality, cardiovascular mortality, and major adverse liver outcomes (MALO) in patients with HFrEF. METHOD AND RESULTS This study included 4523 HFrEF patients from the Danish Heart Failure Registry. Based on FIB-4 score, 25.5 % were low-risk, 45.7 % were indeterminate-risk, and 28.8 % were high-risk for advanced liver fibrosis. After five years, the cumulative incidence of all-cause mortality was 43 % for the high-risk group, 36 % for the indeterminate-risk group, and 23 % for the low-risk group. The indeterminate-risk and high-risk group had an increased hazard ratio (HR) for all-cause mortality (HR 1.33, 95 % confidence interval [CI] 1.16-1.52; HR 1.51, 95 % CI 1.31-1.74) compared to the low-risk group. Similarly, HRs were elevated for cardiovascular mortality (HR 1.61, 95 % CI 1.27-2.05; HR 2.14, 95 % CI 1.67-2.74) and MALO (HR 1.77, 95 % CI 1.01-3.31; HR 2.54, 95 % CI 1.43-4.52). CONCLUSION A high FIB-4 score in patients with HFrEF is associated with increased mortality and MALO.
Collapse
Affiliation(s)
- Abdullahi A Mohamed
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark.
| | - Daniel M Christensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Department of Cardiology, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Milan Mohammad
- Centre for Physical Activity Research, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2200 Copenhagen, Denmark
| | - Lise L Gluud
- Gastro Unit, Copenhagen University Hospital - Hvidovre, Kettegaards Alle 36, 2650, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark; Center for Clinical Metabolic Research, Copenhagen University Hospital - Gentofte, Hospitalsvej 1, 2900 Hellerup, Denmark
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Hospitalsvej 1, 2900 Hellerup, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2200 Copenhagen, Denmark; Department of Public Health, Øster Farimagsgade 5, University of Copenhagen, Denmark
| | - Christian Torp-Pedersen
- Department of Public Health, Øster Farimagsgade 5, University of Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Hillerød, Dyrehavevej 29, Hillerød, Denmark
| | - Charlotte Andersson
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Center for Advanced Heart Disease, Section of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis street, Boston, MA 02115, USA
| | - Morten Schou
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Gunnar Gislason
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Isiklar A, Denizoglu N, Buyukcam F, Ozer Etik D. Does platelet to lymphocyte ratio predict the ultrasound stage in hepatosteatosis? Acta Radiol 2025:2841851251322480. [PMID: 40091566 DOI: 10.1177/02841851251322480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundThe prevalence of hepatosteatosis, or fatty liver disease, has been increasing globally in recent years largely due to increasing rates of obesity, diabetes, and metabolic syndrome.PurposeTo examine the platelet to lymphocyte ratio (PLR) reflection on the hepatosteatosis stage.Material and MethodsWe evaluated healthy individuals who applied to the check-up department in our hospital. The platelet and lymphocyte counts from blood tests, along with upper abdominal ultrasound results obtained as part of routine diagnostic check-ups, results recorded retrospectively, between November 2022 and April 2024.ResultsA total 748 participants were included in the study. All participants were divided in three groups according to hepatosteatosis stages.The PLR levels were highest in the stage 1 hepatosteatosis group. There was statistical significance in PLR levels between stage 1 and 3 hepatosteatosis (P = 0003). In addition, PLR levels were higher in stage 2 than in stage 3, which was also statistically significant (P = 0037).ConclusionThese results could help in early detection and monitoring of disease progression in patients with hepatoteatosis. Lower PLR values (<115.26) in advanced stages might prompt closer monitoring or more aggressive interventions to prevent progression to fibrosis.
Collapse
Affiliation(s)
- Aysun Isiklar
- Department of Internal Medicine, Acibadem Atasehir Hospital, Istanbul, Turkey
| | - Nurper Denizoglu
- Department of Radiology, Acibadem Atasehir Hospital, Istanbul, Turkey
| | - Fatih Buyukcam
- Department of Emergency Medicine, Acibadem Atasehir Hospital, Istanbul, Turkey
| | - Didem Ozer Etik
- Department of Gastroenterology, Acibadem Atasehir Hospital, Istanbul, Turkey
| |
Collapse
|
20
|
Hua S, Zhong W, Sha Y, Ma M, Ge S. Negative association of composite dietary antioxidant index with risk of hepatic fibrosis in individuals underwent cholecystectomy: a cross-sectional study. Sci Rep 2025; 15:9040. [PMID: 40091109 PMCID: PMC11911428 DOI: 10.1038/s41598-025-93782-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: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Cholecystectomy increases the incidence of metabolic associated fatty liver disease and fibrosis. Dietary antioxidants protect the liver from oxidative stress and inhibit the development of metabolic dysfunction-associated steatotic liver disease (MASLD), yet the epidemiological evidence that links antioxidants intake to hepatic steatosis and fibrosis in patients underwent cholecystectomy is not available. Therefore, the present study aimed to investigate the association of composite dietary antioxidant index (CDAI) with the risk of hepatic steatosis and fibrosis in the cholecystectomy population. Data of 773 participants from the National Health and Nutrition Examination Survey (NHANES) 2017-2020 were analyzed. Dietary and supplementary intake of antioxidants were collected to calculate CDAI. The controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) assessed by the vibration controlled transient elastography (VCTE) were used to identify hepatic steatosis and hepatic fibrosis, respectively. Weighted multivariate logistic regression and restricted cubic spline (RCS) regression were conducted to analyze the association. Weighted multivariate logistic regression analysis with full adjustment for confounding variables showed a negative association between CDAI and hepatic fibrosis in individuals underwent Scholecystectomy [OR (95%CI) = 0.87 (0.79, 0.94), P = 0.010], while non-significant association was found between CDAI and MASLD. Moreover, compared to the lowest quartile, the highest quartile of CDAI was associated with a lower risk of hepatic fibrosis [OR (95%CI) = 0.28 (0.13, 0.60), P = 0.007]. The RCS analysis further indicated a linear negative relationship between intake of CDAI, vitamin E, and selenium and the risk of hepatic fibrosis, whereas vitamin A was non-linearly negatively correlated with the risk of hepatic fibrosis (all P overall < 0.001). Our findings suggest that higher antioxidants intake, especially vitamin E, may be associated with a lower risk of hepatic fibrosis among individuals underwent cholecystectomy. Further studies are needed to validate current findings and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Shuyao Hua
- Department of Clinical Nutrition, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Zhong
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanpu Sha
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Mingyang Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China.
| | - Sheng Ge
- Department of Clinical Nutrition, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Clinical Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Li X, Min M, Duan F, Ruan X, Xu L. Biochemical, sex hormonal, and anthropometric predictors of non-alcoholic fatty liver disease in polycystic ovary syndrome. BMC Womens Health 2025; 25:118. [PMID: 40087649 PMCID: PMC11908060 DOI: 10.1186/s12905-025-03648-9] [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: 07/23/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is linked to non-alcoholic fatty liver disease (NAFLD). Biochemical, sex hormonal, and anthropometric indicators have been explored for screening NAFLD in PCOS patients. However, the accuracy of NAFLD screening using these indicators in PCOS patients remains uncertain. This study aimed to identify biochemical, sex hormonal, and anthropometric indicators associated with NAFLD in overweight and obese PCOS patients and assess the diagnostic efficacy of combined indicators. METHODS This cross-sectional study (Clinical trial number ChiCTR1900020986; Registration date January 24th, 2019) involved 87 overweight or obese women with PCOS (mean age 29 ± 4 years). Measurements included anthropometric indices, biochemistry, sex hormone levels, and liver proton density fat fraction (PDFF). Correlation analysis, intergroup comparisons, and logistic regression analysis were used to identify risk factors for NAFLD (PDFF > 5.1%). The receiver operating characteristic curve, area under the curve (AUC), sensitivity, specificity, positive predictive value, and negative predictive value were used to determine cut-off values and evaluate diagnostic accuracy. RESULTS Liver PDFF was 7.69% (3.93%, 14.80%) in overweight and obese PCOS patients, with 67.8% diagnosed with NAFLD. NAFLD was associated with increased body mass index (BMI), abdominal circumference (AC), and triglyceride, total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C), glucose, insulin, and free testosterone (FT) levels, and with decreased high-density lipoprotein-cholesterol (HDL-C) and sex hormone-binding globulin (SHBG) levels (P < 0.05). Risk factors for NAFLD in PCOS included BMI > 26.8 kg/m2, AC > 88.3 cm, triglyceride > 1.57 mmol/L, TC > 4.67 mmol/L, LDL-C > 3.31 mmol/L, glucose > 4.83 mmol/L, insulin > 111.35 pmol/L, FT > 7.6 pg/mL and SHBG < 25 nmol/L (β = 1.411-2.667, P < 0.005). A multi-indicator model including triglycerides, LDL-C, glucose, insulin, and SHBG showed higher diagnostic accuracy (AUC = 0.899, P < 0.001) for screening NAFLD in PCOS patients than single indicators (AUC = 0.667-0.761, P < 0.05). CONCLUSIONS Overweight and obese PCOS patients have higher incidences of liver PDFF and NAFLD. A multi-indicator model including triglycerides > 1.57 mmol/L, LDL-C > 3.31 mmol/L, glucose > 4.83 mmol/L, insulin > 111.35 pmol/L, and SHBG < 25 nmol/L is highly accurate for screening NAFLD in overweight and obese PCOS patients.
Collapse
Affiliation(s)
- Xintong Li
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, PR China
| | - Min Min
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, PR China
- Department of Gynecology, Aviation General Hospital, Beijing, China
| | - Fangfang Duan
- Clinical Epidemiology Research Center, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, PR China.
| | - Li Xu
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, PR China.
| |
Collapse
|
22
|
Agoglia L, Peixoto H, Cardoso AC, Barbosa L, Victer CSXL, Carneiro S, Salles GF, Villela-Nogueira CA, Chindamo MC. Psoriasis and cardiovascular risk: associated and protective factors. An Bras Dermatol 2025:S0365-0596(25)00021-2. [PMID: 40082144 DOI: 10.1016/j.abd.2024.07.013] [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: 06/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Psoriasis (Pso) is an inflammatory skin disease associated with Metabolic Syndrome (MetS), Steatotic Liver Disease (SLD) and cardiovascular risk. However, the effect of anti-inflammatory therapy on cardiovascular risk is uncertain. OBJECTIVES To determine the relationship between anti-inflammatory therapy and subclinical atherosclerosis in individuals with Pso, using the gold standard carotid-femoral Pulse Wave Velocity (cf-PWV) measurement. Additionally, to evaluate the association between cf-PWV, steatosis and Advanced Fibrosis (AF) using Transient Elastography (TE) by Fibroscan®. METHODS Cross-sectional study including Pso patients submitted to cf-PWV and TE. Steatosis was defined as a controlled attenuation parameter ≥ 275 dB/m, AF as liver stiffness measurement ≥ 10 kPa, and increased Aortic Stiffness (AoS) as cf-PWV ≥ 10 m/s. Significant cumulative methotrexate dose was ≥ 1500 mg (MTX1500). Logistic regression analysis evaluated the independent variables associated with increased AoS. RESULTS Eighty patients were included (mean age 56.2 ± 11.5-years, 57.5% female, BMI 28.6 ± 5.3 kg/m2). Prevalences of MetS, diabetes mellitus, dyslipidemia, systemic arterial hypertension, steatosis and AF were 57.5%, 40.0%, 67.5%, 70.0%, 50.0% and 16.3%, respectively. MTX1500 was present in 45%, immunobiological treatment in 33.8%, and cf-PWV ≥ 10 m/s in 21.2%. On logistic regression analysis, age was independently related to cf-PWV ≥ 10 m/s (OR = 1.21; 95% CI 1.06‒1.38; p = 0.003) and MTX1500 was a protective cardiovascular factor (OR = 0.18; 95% CI 0.038‒0.87; p = 0.033). No association was observed between steatosis, AF or immunobiological therapy and cf-PWV ≥10 m/s. STUDY LIMITATIONS Sample size. CONCLUSION In patients with Pso, increased AoS was associated with age, but not with steatosis or AF. A protective cardiovascular effect of MTX was found in a Pso population with a high prevalence of MetS and its components.
Collapse
Affiliation(s)
- Luciana Agoglia
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil; Section of Gastroenterology, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - Helena Peixoto
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil
| | - Ana Carolina Cardoso
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil
| | - Lívia Barbosa
- Dermatology Division, Hospital Federal de Bonsucesso, Rio de Janeiro, RJ, Brazil
| | - Cecília S X L Victer
- Dermatology Unit, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sueli Carneiro
- Dermatology Unit, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gil F Salles
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil
| | - Cristiane A Villela-Nogueira
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil
| | - Maria Chiara Chindamo
- Department of Internal Medicine, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ Brazil
| |
Collapse
|
23
|
Wang J, Bao S, An Q, Li C, Feng J. Roles of extracellular vesicles from different origins in metabolic-associated fatty liver disease: progress and perspectives. Front Immunol 2025; 16:1544012. [PMID: 40129979 PMCID: PMC11930831 DOI: 10.3389/fimmu.2025.1544012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is the most common chronic liver disease worldwide, associated with systemic metabolic dysregulation. It can progress from simple hepatic steatosis (MAFL) to more severe conditions like Metabolic-Associated Steatohepatitis (MASH), fibrosis, cirrhosis, and Hepatocellular Carcinoma (HCC). There is a critical lack of reliable non-invasive diagnostic methods and effective pharmaceutical treatments for MAFLD/MASH, emphasizing the need for further research. Extracellular vesicles (EVs) are nanoscale structures that play important roles in cell signaling by delivering bioactive molecules. However, there is a significant gap in literature regarding the roles of EVs from hosts, plants, and microbiota in MAFLD. This review explores the potential of EVs from various sources-host, plants, and microbiota-as biomarkers, therapeutic agents, drug carriers, and treatment targets for MAFLD. Firstly, the roles of host-derived extracellular vesicles (EVs) in MAFLD, with a focus on cell-type specific EVs and their components-proteins, miRNAs, and lipids-for disease diagnosis and monitoring were discussed. Moreover, it highlighted the therapeutic potential of mesenchymal stem cell (MSC)-derived EVs in reducing lipid accumulation and liver injury, and immune cell-derived EVs in mitigating inflammation and fibrosis. The review also discussed the use of host-derived EVs as drug carriers and therapeutic targets due to their ability to deliver bioactive molecules that impact disease mechanisms. Additionally, it summarized research on plant-derived EVs, which help reduce liver lipid accumulation, inflammation, and enhance gut barrier function in MAFLD. Also, the review explored microbial-derived EVs as novel therapeutic targets, particularly in relation to insulin resistance, liver inflammation, and dysfunction in MAFLD. Overall, by exploring the diverse roles of EVs from host, plant, and microbiota sources in MAFLD, this review offers valuable insights into their potential as non-invasive biomarkers and novel therapeutic strategies, which could pave the way for more effective diagnostic and treatment options for this increasingly prevalent liver disease. Notably, the challenges of translating EVs into clinical practice were also thoroughly discussed, aiming to provide possible directions and strategies for future research.
Collapse
Affiliation(s)
- Jing Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shuoqiang Bao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qi An
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caihong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
24
|
Erceg S, Munjas J, Sopić M, Tomašević R, Mitrović M, Kotur-Stevuljević J, Mamić M, Vujčić S, Klisic A, Ninić A. Expression Analysis of Circulating miR-21, miR-34a and miR-122 and Redox Status Markers in Metabolic Dysfunction-Associated Steatotic Liver Disease Patients with and Without Type 2 Diabetes. Int J Mol Sci 2025; 26:2392. [PMID: 40141039 PMCID: PMC11942408 DOI: 10.3390/ijms26062392] [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: 01/14/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a hepatic form of metabolic syndrome, often co-occurs with type 2 diabetes (T2D) and now affects approximately 30% of the global population. MASLD encompasses conditions from simple steatosis to metabolic dysfunction-associated steatohepatitis, with oxidative stress (OS) driving progression through inflammation. This study analyzes the expression levels of circulating miRNAs and redox status markers in MASLD patients with and without T2D, exploring their potential as disease biomarkers. The expressions of miR-21, miR-34a, and miR-122 were analyzed in the platelet-poor plasma of 147 participants, divided into three groups: MASLD + T2D (48), MASLD (50), and a control group (49). Total oxidant status (TOS), total antioxidant status (TAS), ischemia-modified albumin (IMA), and superoxide anion radical (O2•-) were measured in serum and plasma. Logistic regression showed that miR-21, miR-34a, TOS, TAS, O2•-, and IMA were positive predictors of MASLD, while miR-21 and TAS were negative predictors of T2D in MASLD. Although miR-122 did not show a significant association with either condition, in combination with miR-34a and other markers such as lipid status and liver enzymes, a new significant predictor of MASLD was identified. Circulating miRNAs in combination with redox status markers, lipid status and liver enzymes show potential as MASLD biomarkers.
Collapse
Affiliation(s)
- Sanja Erceg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| | - Ratko Tomašević
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Department of Gastroenterology and Hepatology, Clinic for Internal Medicine, Clinical Hospital Center Zemun, 11080 Belgrade, Serbia
| | - Miloš Mitrović
- Clinical Department for Gastroenterology and Hepatology, University Medical Center Zvezdara, 11120 Belgrade, Serbia;
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| | - Milica Mamić
- Department of Laboratory Diagnostics, Clinical Hospital Center Zemun, 11080 Belgrade, Serbia;
| | - Sanja Vujčić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| | - Aleksandra Klisic
- Faculty of Medicine, University of Montenegro, 81000 Podgorica, Montenegro;
- Center for Laboratory Diagnostics, Primary Health Care Center, 81000 Podgorica, Montenegro
| | - Ana Ninić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (S.E.); (J.M.); (M.S.); (J.K.-S.); (S.V.)
| |
Collapse
|
25
|
Pafili K, Zaharia OP, Strassburger K, Knebel B, Herder C, Huttasch M, Karusheva Y, Kabisch S, Strom A, Nowotny B, Szendroedi J, Roden M. PNPLA3 gene variation modulates diet-induced improvement in liver lipid content in type 2 diabetes. Clin Nutr 2025; 48:6-15. [PMID: 40090039 DOI: 10.1016/j.clnu.2025.02.032] [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: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND&AIMS Lifestyle-induced weight reduction remains crucial for managing type 2 diabetes and steatotic liver disease, but its effectiveness varies. We postulated that the G allele in the rs738409 single nucleotide polymorphism within patatin-like phospholipase domain-containing protein 3 (PNPLA3), which associates with metabolic dysfunction-associated steatotic liver disease, also modulates diet-related metabolic effects. METHODS Participants with type 2 diabetes were randomized to 8-week hypocaloric diets (energy intake: -1,256 kJ/d of, <30 kcal% fat): high in cereal fiber and coffee excluding red meat (HF-RM + C; n = 16), or low in cereal fiber, devoid of coffee, but high in red meat (LF + RM-C; n = 15). Whole-body insulin sensitivity (M value) was assessed using [2H]glucose and hyperinsulinemic-normoglycemic clamps, hepatic lipid content (HCL) and body fat volumes by magnetic resonance spectroscopy/imaging before and after intervention. RESULTS Despite comparable weight loss, HCL decreased more in non-carriers (-65 %) than in G-allele carriers (-36 %) upon HF-RM + C diet (both p < 0.05 vs baseline and between groups), but only among non-carriers (-46 %, p < 0.05 vs baseline) upon LF + RM-C. Upon HF-RM + C diet, increase in insulin sensitivity was not different between carriers (+27 % p = 0.051 from baseline) and non-carriers (+21 %, p = 0.032 from baseline), p > 0.05 for between-group comparison. Upon LF + RM-C diet, both groups equally improved their whole-body insulin sensitivity (+42 % for non-carriers and +37 % for carriers, p < 0.05 vs baseline). Upon HF-RM + C diet, non-carriers decreased circulating interleukin-18 from baseline by -31 %, whereas, upon LF + RM-C diet, non-carriers decreased circulating anti-inflammatory interleukin-1 receptor antagonist levels by 14 % (both p < 0.05 vs baseline). CONCLUSIONS Humans with the PNPLA3 G-allele show modified dietary-induced effects on steatotic liver disease in type 2 diabetes despite body weight reduction. Registration at Clinicaltrials.gov, Identifier number: NCT01409330.
Collapse
Affiliation(s)
- Kalliopi Pafili
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Oana-Patricia Zaharia
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Klaus Strassburger
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Birgit Knebel
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Christian Herder
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Maximilian Huttasch
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Yanislava Karusheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Stefan Kabisch
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Department of Endocrinology and Metabolic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Bettina Nowotny
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; Bayer AG, Research and Development Pharmaceuticals, Aprather Weg 42113 Wuppertal, Germany
| | - Julia Szendroedi
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Department for Internal Medicine I, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
26
|
Huang DQ, Wong VWS, Rinella ME, Boursier J, Lazarus JV, Yki-Järvinen H, Loomba R. Metabolic dysfunction-associated steatotic liver disease in adults. Nat Rev Dis Primers 2025; 11:14. [PMID: 40050362 DOI: 10.1038/s41572-025-00599-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the umbrella term that comprises metabolic dysfunction-associated steatotic liver, or isolated hepatic steatosis, through to metabolic dysfunction-associated steatohepatitis, the progressive necroinflammatory disease form that can progress to fibrosis, cirrhosis and hepatocellular carcinoma. MASLD is estimated to affect more than one-third of adults worldwide. MASLD is closely associated with insulin resistance, obesity, gut microbial dysbiosis and genetic risk factors. The obesity epidemic and the growing prevalence of type 2 diabetes mellitus greatly contribute to the increasing burden of MASLD. The treatment and prevention of major metabolic comorbidities such as type 2 diabetes mellitus and obesity will probably slow the growth of MASLD. In 2023, the field decided on a new nomenclature and agreed on a set of research and action priorities, and in 2024, the US FDA approved the first drug, resmetirom, for the treatment of non-cirrhotic metabolic dysfunction-associated steatohepatitis with moderate to advanced fibrosis. Reliable, validated biomarkers that can replace histology for patient selection and primary end points in MASH trials will greatly accelerate the drug development process. Additionally, noninvasive tests that can reliably determine treatment response or predict response to therapy are warranted. Sustained efforts are required to combat the burden of MASLD by tackling metabolic risk factors, improving risk stratification and linkage to care, and increasing access to therapeutic agents and non-pharmaceutical interventions.
Collapse
Affiliation(s)
- Daniel Q Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | - Vincent W S Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Mary E Rinella
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Jerome Boursier
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Centre Hospitalier Universitaire d'Angers, Angers, France
- Laboratoire HIFIH, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, San Diego, CA, USA.
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California at San Diego, San Diego, CA, USA.
| |
Collapse
|
27
|
Dong L, Lou W, Xu C, Wang J. Naringenin cationic lipid-modified nanoparticles mitigate MASLD progression by modulating lipid homeostasis and gut microbiota. J Nanobiotechnology 2025; 23:168. [PMID: 40038718 DOI: 10.1186/s12951-025-03228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
Naringenin (NAR) possesses various pharmacological activities including antioxidant, anti-inflammatory, and hepatoprotective effects. However, its therapeutic efficacy is limited by its hydrophobic and crystalline nature. This study aimed to investigate the therapeutic potential and molecular mechanisms of NAR efficiently loaded into cationic nanoparticles (NP-NAR) for treating metabolic dysfunction-associated steatotic liver disease (MASLD) in a mouse model. The results demonstrated that NP-NAR effectively ameliorated lipid metabolism dysbiosis, oxidative stress, insulin resistance, and inflammation in MASLD mice. Transcriptomic analysis and molecular data revealed that NP-NAR promoted fatty acid oxidation via activation of the PPAR signaling pathway, reduced hepatic lipid uptake and lipogenesis by inhibiting the expressions of key genes including CD36, ACC, and FASN. Moreover, NP-NAR modulated cholesterol metabolism by inhibiting the classical bile acid synthesis pathway. 16 S rDNA gene sequencing revealed a disbalanced gut microbiota in MASLD mice, whereas NP-NAR treatment statistically reversed the abundance changes of several intestinal bacteria at the phylum and genus levels, which partly contributed to the balance in intestinal metabolite production, including short-chain fatty acids. In conclusion, these findings suggest that NP-NAR may be a promising candidate for the treatment of obesity-associated MASLD, offering new insight into the mechanisms underlying NAR's efficacy against MASLD.
Collapse
Affiliation(s)
- Lu Dong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Wenyong Lou
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Congfei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Juan Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China.
| |
Collapse
|
28
|
Gu Y, Guo C, Liu Z, Zhang Y, Han X, Zhang X, Zhao S, Wang H, Zhang T. The trend in incidence of non-alcoholic fatty liver disease and its impact on cirrhosis and liver cancer: An analysis from Global Burden of Disease 2021. Public Health 2025; 242:79-86. [PMID: 40037155 DOI: 10.1016/j.puhe.2025.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
OBJECTIVES We aimed to recognize the burden of NAFLD and support public health policy development for its prevention and management. STUDY DESIGN A cross-sectional analysis of GBD 2021 results was conducted. METHODS We collected incidence data on NAFLD from 1990 to 2021 using Global Burden of Disease Study in 2021. Estimated annual percentage changes (EAPCs) in NAFLD age standardized incidence rate (ASR) were calculated to quantify the temporal trends in NAFLD ASR. Bayesian age-period-cohort models were constructed to project NAFLD incidence rates and cases up to 2050. Additionally, we assessed the percentage of cirrhosis and liver cancer attributable to NAFLD. RESULTS Globally, the newly-occurred cases of NAFLD increased by 94.49 % from 24, 856, 159 in 1990 to 48, 353, 272 in 2021. The case number will further increase to 78,602,984 in 2050, and ASR will increase from 5.93 per 1000 in 2021 to 7.26 per 1000 in 2050. The most pronounced increases were observed in young people and men. In 2021, NAFLD accounted for 82.7 % of cirrhosis and other chronic liver diseases and 8.0 % of liver cancer cases. CONCLUSIONS From 1990 to 2021, the incidence of NAFLD has been continuously increasing and is expected to continue rising until 2050. The increases in young people and men highlight their priority in future schedules. The rising proportions of cirrhosis and liver cancer caused by NAFLD further underscore the serious health risks.
Collapse
Affiliation(s)
- Yu Gu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Chengnan Guo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Zhenqiu Liu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, 200032, China
| | - Yujiao Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Xinyu Han
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, 200032, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, 225300, China; Yiwu Research Institute, Fudan University, Yiwu, 200032, China.
| |
Collapse
|
29
|
Huang X, Luo M, Zeng Y, Yi J, Lin S, Wang Y, Zheng X, Luo X. Effect of therapeutic lifestyle changes on patients with overweight/obesity and non-alcoholic fatty liver disease: A randomized controlled trial. Am J Med Sci 2025; 369:373-379. [PMID: 39454726 DOI: 10.1016/j.amjms.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Non-alcoholic fatty liver disease is a liver condition that is increasing globally. Unfortunately, there are no successful or approved pharmacological treatments for non-alcoholic fatty liver disease. Hence, this study aimed to investigate the effect of therapeutic lifestyle changes on patients with overweight/obesity and non-alcoholic fatty liver disease. METHODS A prospective, parallel-group, randomized controlled trial was conducted. The patients were randomized into intervention and control groups using tables with random numbers. In the control group, routine health guidance was provided for 3 months, while in the intervention group, diversified lifestyle intervention was provided. The body composition, visceral fat area, abdominal circumference, and body mass index of the control and intervention groups were compared before and after the intervention. Descriptive statistics, paired t-tests, and linear regression models were used for data analysis. RESULTS A total of 115 participants (57 in the intervention group and 58 in the control group) completed the study. The intervention groups had significantly greater high-density lipoprotein cholesterol levels, basal metabolic rate, muscle mass, and questionnaire scores than the control groups (P < 0.05). Furthermore, the intervention participants had lower body mass index, abdominal circumference, triglyceride levels, low-density lipoprotein cholesterol levels, and fatty liver index (P < 0.05). CONCLUSIONS Therapeutic lifestyle changes therapy for non-alcoholic fatty liver disease patients with overweight/obesity can significantly control body mass index, improve blood lipid levels, reduce fatty liver and body fat rates, improve basic metabolism, alleviate disease, and improve quality of life. More research is needed to determine the long-term impact of therapeutic lifestyle changes in high-risk groups.
Collapse
Affiliation(s)
- Xishun Huang
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Meixuan Luo
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - YanYan Zeng
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Jiao Yi
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Sumei Lin
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Yitao Wang
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Xuan Zheng
- Department of health medicine, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China
| | - Xiaohua Luo
- Department of oncology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, PR China.
| |
Collapse
|
30
|
Capinha F, Carvalhana S, Cortez-Pinto H. Role of Alcohol in Steatotic Liver Disease: Impact on Patients with Cardiometabolic Risk Factors. Dig Dis Sci 2025:10.1007/s10620-025-08912-4. [PMID: 40025309 DOI: 10.1007/s10620-025-08912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
The new definition of steatotic liver disease (SLD), as a broader concept, was a step forward in the increasing recognition of the substantial overlap between alcohol and cardiometabolic risk factors (CMRFs), in a continuum way. The spectrum of pathophysiological aspects, ranging from liver steatosis to fibrosis, has similarities in MASLD and ALD. Also, there is now considerable evidence that the association of metabolic dysfunction with increased alcohol consumption impacts on the risk of severe liver disease and prognosis. The new MetALD class, as recently proposed, shows clear differences in prognosis when comparing with MASLD and ALD groups. However, there is room for improvement, such as considering the role of previous alcohol intake, fluctuations of consumption over time, including binge drinking, refinement of alcohol assessment, and better understanding of the role of biomarkers. In summary, SLD is no doubt a significant improvement, but the new classification needs to be dynamic and adapting to patients needing frequent reassessment. Furthermore, it brings opportunities for research on the interaction between alcohol consumption and CMRFs.
Collapse
Affiliation(s)
- Francisco Capinha
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal
| | - Sofia Carvalhana
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal
| | - Helena Cortez-Pinto
- Serviço de Gastrenterologia e Hepatologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal.
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-035, Lisbon, Portugal.
| |
Collapse
|
31
|
Kim D, Shah M, Kim JH, Kim J, Baek YH, Jeong JS, Han SY, Lee YS, Park G, Cho JH, Roh YH, Lee SW, Choi GB, Park JH, Yoo KH, Seong RH, Lee YS, Woo HG. Integrative transcriptomic and genomic analyses unveil the IFI16 variants and expression as MASLD progression markers. Hepatology 2025; 81:962-975. [PMID: 38385945 DOI: 10.1097/hep.0000000000000805] [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: 10/18/2023] [Accepted: 01/05/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a broad and continuous spectrum of liver diseases ranging from fatty liver to steatohepatitis. The intricate interactions of genetic, epigenetic, and environmental factors in the development and progression of MASLD remain elusive. Here, we aimed to achieve an integrative understanding of the genomic and transcriptomic alterations throughout the progression of MASLD. APPROACH AND RESULTS RNA-Seq profiling (n = 146) and whole-exome sequencing (n = 132) of MASLD liver tissue samples identified 3 transcriptomic subtypes (G1-G3) of MASLD, which were characterized by stepwise pathological and molecular progression of the disease. Macrophage-driven inflammatory activities were identified as a key feature for differentiating these subtypes. This subtype-discriminating macrophage interplay was significantly associated with both the expression and genetic variation of the dsDNA sensor IFI16 (rs6940, A>T, T779S), establishing it as a fundamental molecular factor in MASLD progression. The in vitro dsDNA-IFI16 binding experiments and structural modeling revealed that the IFI16 variant exhibited increased stability and stronger dsDNA binding affinity compared to the wild-type. Further downstream investigation suggested that the IFI16 variant exacerbated DNA sensing-mediated inflammatory signals through mitochondrial dysfunction-related signaling of the IFI16-PYCARD-CASP1 pathway. CONCLUSIONS This study unveils a comprehensive understanding of MASLD progression through transcriptomic classification, highlighting the crucial roles of IFI16 variants. Targeting the IFI16-PYCARD-CASP1 pathway may pave the way for the development of novel diagnostics and therapeutics for MASLD.
Collapse
Affiliation(s)
- Doyoon Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea
| | - Masaud Shah
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jang Hyun Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea
| | - JungMo Kim
- Ajou Translational Omics Center (ATOC), Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon, Republic of Korea
| | - Yang-Hyun Baek
- Department of Internal Medicine, Liver Center, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Jin-Sook Jeong
- Pathology and Laboratory Medicine, St Mary's Hospital, Busan, Republic of Korea
| | | | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Gaeul Park
- Division of Rare Cancer, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Jin-Han Cho
- Department of Diagnostic Radiology, Dong-A University Medical Center, Busan, Republic of Korea
| | - Young-Hoon Roh
- Department of Surgery, Dong-A University Medical Center, Busan, Republic of Korea
| | - Sung-Wook Lee
- Department of Internal Medicine, Liver Center, Dong-A University Medical Center, Busan, Republic of Korea
| | - Gi-Bok Choi
- Department of Radiology, On Hospital, Busan, Republic of Korea
| | - Jong Hoon Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Kyung Hyun Yoo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Rho Hyun Seong
- Department of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Yeon-Su Lee
- Division of Rare Cancer, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, Republic of Korea
- Ajou Translational Omics Center (ATOC), Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon, Republic of Korea
| |
Collapse
|
32
|
Dong L, Lou W, Wang J. β-Carotene-loaded cationic nanoparticles ameliorate MASLD via modulating lipid homeostasis and gut microbiome. Food Res Int 2025; 205:115816. [PMID: 40032486 DOI: 10.1016/j.foodres.2025.115816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/19/2025] [Accepted: 01/19/2025] [Indexed: 03/05/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is, increasingly, a major threat to human health, yet without any approved drug. β-carotene (BC) contributes to alleviating several metabolic diseases. However, the bioavailability of BC is hindered by hydrophobicity and environmental sensitivity. Herein, we explore the utilization of cationic lipid-assisted nanoparticles to achieve efficient delivery of BC. In the MASLD model, NP-BC ameliorated the development of metabolic disorders, insulin resistance, inflammatory injury and hepatic steatosis. Transcriptomic analysis showed that NP-BA rectifies various pathways involved in steatosis development by inhibiting the PI3K/AKT/mTOR pathway and PPARγ gene expression. Meanwhile, NP-BC also reshaped the composition of gut microbiota in MASLD mice by reducing the Firmicutes/Bacteroidetes ratio and increasing the abundance of beneficial bacteria. Taken together, our study demonstrates that NP-BC can improve MASLD and may be a promising candidate for treating MASLD.
Collapse
Affiliation(s)
- Lu Dong
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Wenyong Lou
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Juan Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
33
|
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
|
34
|
Zhang X, Lau HCH, Yu J. Pharmacological treatment for metabolic dysfunction-associated steatotic liver disease and related disorders: Current and emerging therapeutic options. Pharmacol Rev 2025; 77:100018. [PMID: 40148030 DOI: 10.1016/j.pharmr.2024.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as nonalcoholic fatty liver disease) is a chronic liver disease affecting over a billion individuals worldwide. MASLD can gradually develop into more severe liver pathologies, including metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and liver malignancy. Notably, although being a global health problem, there are very limited therapeutic options against MASLD and its related diseases. While a thyroid hormone receptor agonist (resmetirom) is recently approved for MASH treatment, other efforts to control these diseases remain unsatisfactory. Given the projected rise in MASLD and MASH incidence, it is urgent to develop novel and effective therapeutic strategies against these prevalent liver diseases. In this article, the pathogenic mechanisms of MASLD and MASH, including insulin resistance, dysregulated nuclear receptor signaling, and genetic risk factors (eg, patatin-like phospholipase domain-containing 3 and hydroxysteroid 17-β dehydrogenase-13), are introduced. Various therapeutic interventions against MASH are then explored, including approved medication (resmetirom), drugs that are currently in clinical trials (eg, glucagon-like peptide 1 receptor agonist, fibroblast growth factor 21 analog, and PPAR agonist), and those failed in previous trials (eg, obeticholic acid and stearoyl-CoA desaturase 1 antagonist). Moreover, given that the role of gut microbes in MASLD is increasingly acknowledged, alterations in the gut microbiota and microbial mechanisms in MASLD development are elucidated. Therapeutic approaches that target the gut microbiota (eg, dietary intervention and probiotics) against MASLD and related diseases are further explored. With better understanding of the multifaceted pathogenic mechanisms, the development of innovative therapeutics that target the root causes of MASLD and MASH is greatly facilitated. The possibility of alleviating MASH and achieving better patient outcomes is within reach. SIGNIFICANCE STATEMENT: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, and it can progress to more severe pathologies, including steatohepatitis, cirrhosis, and liver cancer. Better understanding of the pathogenic mechanisms of these diseases has facilitated the development of innovative therapeutic strategies. Moreover, increasing evidence has illustrated the crucial role of gut microbiota in the pathogenesis of MASLD and related diseases. It may be clinically feasible to target gut microbes to alleviate MASLD in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
35
|
Houttu V, Boulund U, Troelstra M, Csader S, Stols-Gonçalves D, Mak AL, Dijk AMV, Bouts J, Winkelmeijer M, Verdoes X, van den Berg-Faay S, Lek D, Ronteltap T, de Haan F, Jorstad H, Männistö V, Savonen K, Pentikäinen H, Hanhineva K, Babu AF, Panagiotou G, van Delden O, Verheij J, Doukas M, Nederveen A, Schwab U, Grefhorst A, Nieuwdorp M, Holleboom AG. Deep phenotyping of patients with MASLD upon high-intensity interval training. JHEP Rep 2025; 7:101289. [PMID: 40051412 PMCID: PMC11883402 DOI: 10.1016/j.jhepr.2024.101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 03/09/2025] Open
Abstract
Background & Aims Exercise is a key component of lifestyle management in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), but neither its therapeutic effect on the active stage of the disease, that is metabolic dysfunction-associated steatohepatitis (MASH) nor the mediating mechanisms have been characterized. Therefore, we performed multi-omic phenotyping of patients with MASLD-MASH on an exercise program. Methods Fifteen patients with MASLD conducted high-intensity interval training (HIIT) combined with home-based training for 12 weeks. MASLD was evaluated using histology, transient elastography, and multiparametric magnetic resonance imaging (MRI) before and after the intervention. Change in maximal oxygen consumption (VO2max) and MRI-determined liver fat were compared with a control group of patients with MASLD (n = 22). RNA sequencing was performed on liver, muscle, and fat biopsies of patients in the exercise group. Stool was analyzed by shotgun metagenomics and untargeted metabolomics was performed on plasma, urine, adipose, and stool. Results HIIT increased VO2max by 10.1% and improved mitochondrial metabolism in skeletal muscle, indicating improved cardiorespiratory fitness and adherence. VO2max increased significantly in the exercise group compared with controls. Histologically, no reduction in steatosis, MASH, or liver fibrosis was observed; however, transient elastography tended to improve. MRI-determined liver fat did not change in the exercise group compared with controls. HIIT induced changes in mRNA expression of genes related to beiging of adipose tissue and fibrogenesis in liver. In addition, specific gut microbial taxa and metabolites changed. Conclusions HIIT increased cardiorespiratory fitness and induced beneficial gene expression changes in muscle, adipose tissue, and liver, but without translation into histological improvement of MASLD. Longer exercise intervention trials are warranted to validate or refute current recommendations for exercise as a cornerstone treatment for MASLD-MASH. Impact and implications Despite exercise being considered as a key component of lifestyle management for steatotic liver disease, neither the clinical effects nor the mechanisms involved are completely understood. We show that a high-intensity interval training (HIIT) program in 15 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) improved cardiorespiratory fitness, compared with 22 control patients with MASLD who did not participate in an exercise program, however, it did not improve MASLD. HIIT induced a positive effect on fat tissue and muscle metabolism which was accompanied with changes in certain gut bacteria and metabolites in blood and urine. These findings improve our understanding of the effects of exercise on the whole-body metabolism in relation to steatotic liver disease. As such, this study provides a basis for future exercise interventions in patients with MASLD, required to thoroughly test current guideline advice for exercise as a cornerstone treatment for MASLD of all stages. Clinical trial registry Dutch Trial Register (registration number NL7932).
Collapse
Affiliation(s)
- Veera Houttu
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ulrika Boulund
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne Csader
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julia Bouts
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Xanthe Verdoes
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Donne Lek
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ted Ronteltap
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ferdinand de Haan
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Harald Jorstad
- Department of Cardiology, Amsterdam Movement Sciences, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland, and Kuopio University Hospital, Kuopio, Finland
| | - Kai Savonen
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | | | - Kati Hanhineva
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
- Department of Life Technologies, Food Chemistry, and Food Development Unit, University of Turku, Turku, Finland
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research, and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
- Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Otto van Delden
- Department of Interventional Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michial Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ursula Schwab
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Aldo Grefhorst
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan Georgius Holleboom
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Barat M, Ollivier C, Taibi L, Nitsche V, Sogni P, Soyer P, Parlati L, Dohan A, Abdoul H, Revel MP. Standard of care versus standard of care plus Ericksonian hypnosis for percutaneous liver biopsy: Results of a randomized control trial. Diagn Interv Imaging 2025; 106:93-97. [PMID: 39358154 DOI: 10.1016/j.diii.2024.09.009] [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: 09/01/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE The purpose of this study was to compare levels of pain and anxiety during percutaneous ultrasound-guided liver biopsy between patients receiving standard of care and those receiving standard of care plus the support of Ericksonian hypnosis. MATERIALS AND METHODS This prospective, single-center, single-blind, randomized controlled superiority trial included 70 participants. Participants were randomly assigned to either the standard of care group and received oral anxiolytic medications with reassuring conversational support, or to the experimental group, and received Ericksonian hypnosis (i.e., conversational hypnosis) in addition to standard of care. The primary outcome was the level of pain experienced during the biopsy, measured on a 10-point visual analog scale (0 indicating no pain to 10 indicating excruciating pain). Secondary outcomes included anxiety level during the biopsy, pain level within one hour of the biopsy measured using the same 10-point visual analog scale, amount of analgesic medication taken in the 24 h following the biopsy, and patient willingness to undergo another ultrasound-guided percutaneous liver biopsy in the future. RESULTS Thirty-six participants were included in the standard of care group, and 34 were included in the experimental group. The mean score of pain experienced during the biopsy was lower in the experimental group (2.4 ± 1.9 [standard deviation (SD)]) compared to the standard of care group (4.4 ± 2.6 [SD]) (P = 0.001). The level of anxiety experienced during the biopsy was lower in the hypnosis group (2.1 ± 1.8 [SD]) compared to the standard of care group (4.8 ± 2.4 [SD]) (P < 0.001). No significant differences in other secondary outcomes were observed between the two groups. CONCLUSION The addition of Ericksonian hypnosis to standard of care reduces the pain experienced by patients during percutaneous ultrasound-guided percutaneous liver biopsy by comparison with standard of care alone.
Collapse
Affiliation(s)
- Maxime Barat
- Université Paris Cité, Paris 75006, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Génomique et Signalisation des Tumeurs Endocrines, Institute Cochin, INSERM U 1016, CNRS UMR8104, Paris 75014, France.
| | - Camille Ollivier
- URP7323, Pharmacologie et Évaluations des Thérapeutiques chez l'Enfant et la Femme Enceinte, Université Paris Cité, Paris 75014, France; Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Linda Taibi
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Véronique Nitsche
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Philippe Sogni
- Université Paris Cité, Paris 75006, France; Department of Hepatology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Philippe Soyer
- Université Paris Cité, Paris 75006, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Lucia Parlati
- Université Paris Cité, Paris 75006, France; Department of Hepatology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Anthony Dohan
- Université Paris Cité, Paris 75006, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Génomique et Signalisation des Tumeurs Endocrines, Institute Cochin, INSERM U 1016, CNRS UMR8104, Paris 75014, France
| | - Hendy Abdoul
- URP7323, Pharmacologie et Évaluations des Thérapeutiques chez l'Enfant et la Femme Enceinte, Université Paris Cité, Paris 75014, France; Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Marie-Pierre Revel
- Université Paris Cité, Paris 75006, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| |
Collapse
|
37
|
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
|
38
|
Zhan T, Liu JX, Huang M, Chen MT, Tian XR, Yang XL, Tan J, Zou YL, Han Z, Chen W, Tian X, Huang XD. ILF3 inhibits p-AMPK expression to drive non-alcoholic fatty liver disease progression. World J Hepatol 2025; 17:101691. [PMID: 40027551 PMCID: PMC11866148 DOI: 10.4254/wjh.v17.i2.101691] [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: 09/24/2024] [Revised: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a disease of increasing global prevalence and an important risk factor for the development of insulin resistance, type 2 diabetes, non-alcoholic steatohepatitis and hepatocellular carcinoma, but the pathogenesis is not clear. The aim of this study was to explore the role of ILF3 in NAFLD. AIM To investigate the molecular processes through which ILF3 facilitates the advancement of NAFLD by inhibiting the expression of p-AMPK. This exploration seeks to provide new insights into the etiology of NAFLD and evaluate the potential of ILF3 as a diagnostic marker and potential treatment focus for future interventions. METHODS In vitro and in vivo experiments were conducted using HepG2 cells and NAFLD animal models. The effects of ILF3 knockdown on lipid synthesis and triglyceride (TG) secretion were examined by analyzing the expression levels of p-AMPK. Additionally, the roles of ILF3 and the AMPK signaling pathway were verified using techniques such as Western blotting, quantitative reverse transcription PCR, Oil Red O staining, and immunohistochemistry. RESULTS Investigations revealed an increase in ILF3 Levels within both HepG2 cells and animal models of NAFLD, concurrently with a decrease in p-AMPK expression. Knocking down ILF3 activated the AMPK pathway, reducing lipid production and TG secretion in hepatocytes, thereby mitigating the advancement of NAFLD. CONCLUSION ILF3 promotes the evolution of NAFLD by inhibiting the expression of p-AMPK. The knockdown of ILF3 activates the AMPK signaling pathway, alleviating the severity of NAFLD. These findings underscore the function of ILF3 in the pathogenesis of NAFLD and demonstrate its viability as a treatment focus and diagnostic indicator.
Collapse
Affiliation(s)
- Ting Zhan
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Jia-Xi Liu
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Min Huang
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Ming-Tao Chen
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Xiao-Rong Tian
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Xiu-Lin Yang
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Jie Tan
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Yan-Li Zou
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Zheng Han
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Wei Chen
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Xia Tian
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China
| | - Xiao-Dong Huang
- Department of Gastroenterology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan 430000, Hubei Province, China.
| |
Collapse
|
39
|
Pecoraro V, Nascimbeni F, Cuccorese M, Gabrielli F, Fasano T, Trenti T. Diagnostic Accuracy of Golgi Protein 73 (GP73) for Liver Fibrosis Staging in Metabolic Dysfunction-Associated Steatotic Liver Disease: A Scoping Review and Cohort Study. Diagnostics (Basel) 2025; 15:544. [PMID: 40075792 PMCID: PMC11898419 DOI: 10.3390/diagnostics15050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Golgi protein 73 (GP73) is a transmembrane protein expressed by epithelial cells of the bile duct in the normal liver. High serum levels of GP73 have been detected in patients with acute or chronic liver diseases, MASLD, and its measurement has been suggested as a potential biomarker for liver fibrosis staging. We evaluated the utility of GP73 in the diagnosis of MASLD, MASH, and for liver fibrosis staging. Methods: We performed a literature scoping review to map the current evidence about the accuracy of GP73 in patients with MASLD. We searched in Medline and EMBASE for English studies reporting an AUC value of GP73 in diagnosing MASLD and MASH and evaluating GP73 for fibrosis staging. A narrative synthesis of the evidence was conducted. Moreover, we performed an observational study including 84 patients with MASLD, of which 60 were biopsy-confirmed MASH, and different liver fibrosis stages, and 15 healthy controls. Serum GP73 levels were determined using a chemiluminescent assay and reported as mean and standard deviation (SD). Sensitivity (SE), specificity (SP), the area under the receiver operating characteristic (AUROC) curve, and the optimal cut-off value were calculated. Data were considered statistically significant when p < 0.05. Results: Available studies evaluating GP73 in MASLD reported the ability to discriminate MASH from simple steatosis and distinguish patients at different fibrotic stages, but the evidence is still scarce. Our experimental study showed that the serum levels of GP73 were 30 ± 12 ng/mL in MASLD and 32 ± 12 ng/mL in MASH patients and were statistically higher than those of the control group (19 ± 30 ng/mL), increasing from liver fibrosis stage F0 to F4. GP73 levels were significantly higher in patients with significant and advanced fibrosis than controls and no significant fibrosis (p > 0.05). ROC analysis demonstrated that serum GP73 had a good diagnostic potential for MASLD (AUROC 0.85; SE 90%; SP 73%), MASH (AUROC 0.75; SE 82%; SP64%), and significant fibrosis (AUROC 0.7; SE 56%; SP 79%) and was better than other biomarkers for chronic liver diseases. Conclusions: Serum GP73 could support clinicians in the evaluation of patients with MASH and significant fibrosis.
Collapse
Affiliation(s)
- Valentina Pecoraro
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Michela Cuccorese
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Tommaso Fasano
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | |
Collapse
|
40
|
Xia M, Lu Y, Yin F, Cao Z, Yao P, Li H. The external validation of Dallas Steatosis Index among Asian population: a useful tool for metabolic dysfunction-associated steatotic liver disease identification and prevention. J Gastroenterol 2025:10.1007/s00535-025-02220-4. [PMID: 39994040 DOI: 10.1007/s00535-025-02220-4] [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: 05/01/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND The Dallas Steatosis Index (DSI) is a non-invasive tool (NIT) developed to detect the risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in multi-ethnic populations, external validation in Asians has yet to be conducted. Therefore, we evaluated the ability of the DSI with the BMI classification of WPRO (DSI_WPRO) to identify MASLD in the Chinese population. In addition, we investigated the associations between the DSI_WPRO and the risk of MASLD in a longitudinal study. METHODS Baseline data from the Dongfeng-Tongji cohort were collected to investigate the ability of the DSI_WPRO to identify MASLD patients by ROC analysis. Furthermore, multivariate logistic regressions were performed to investigate the associations of the DSI_WPRO and MASLD risks in a 5-year follow-up of the DFTJ cohort study. RESULTS Among a total of 9,376 MASLD participants and 25,974 non-MASLD participants, the area under the curve (AUC) of the DSI_WPRO reached 0.777 after adjusting BMI classification, which is higher than other NITs in this study. In addition, we redefined the risk category and the screening proposal of MASLD in Asians with the DSI_WPRO. We found that the cutoff point of 0 has the best ability to recognize the presence or absence of MASLD. Furthermore, compared with the low DSI_WPRO (DSI_WPRO < 0), OR (95% CIs) of higher DSI_WPRO (DSI_WPRO ≥ 0) was 3.048 (2.827 ~ 3.285) for MASLD. CONCLUSION The DSI is a useful tool for MASLD identification and prevention. After more validation studies, DSI can be generalized in the Asian population.
Collapse
Affiliation(s)
- Mengyang Xia
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yixuan Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feiyang Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqiang Cao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongxia Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
41
|
Zisis M, Chondrogianni ME, Androutsakos T, Rantos I, Oikonomou E, Chatzigeorgiou A, Kassi E. Linking Cardiovascular Disease and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): The Role of Cardiometabolic Drugs in MASLD Treatment. Biomolecules 2025; 15:324. [PMID: 40149860 PMCID: PMC11940321 DOI: 10.3390/biom15030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
The link between cardiovascular disease (CVD) and metabolic dysfunction-associated steatotic liver disease (MASLD) is well-established at both the epidemiological and pathophysiological levels. Among the common pathophysiological mechanisms involved in the development and progression of both diseases, oxidative stress and inflammation, insulin resistance, lipid metabolism deterioration, hepatokines, and gut dysbiosis along with genetic factors have been recognized to play a pivotal role. Pharmacologic interventions with drugs targeting common modifiable cardiometabolic risk factors, such as T2DM, dyslipidemia, and hypertension, are a reasonable strategy to prevent CVD development and progression of MASLD. Recently, a novel drug for metabolic dysfunction-associated steatohepatitis (MASH), resmetirom, has shown positive effects regarding CVD risk, opening new opportunities for the therapeutic approach of MASLD and CVD. This review provides current knowledge on the epidemiologic association of MASLD to CVD morbidity and mortality and enlightens the possible underlying pathophysiologic mechanisms linking MASLD with CVD. The role of cardiometabolic drugs such as anti-hypertensive drugs, hypolipidemic agents, glucose-lowering medications, acetylsalicylic acid, and the thyroid hormone receptor-beta agonist in the progression of MASLD is also discussed. Metformin failed to prove beneficial effects in MASLD progression. Studies on the administration of thiazolinediones in MASLD suggest effectiveness in improving steatosis, steatohepatitis, and fibrosis, while newer categories of glucose-lowering agents such as GLP-1Ra and SGLT-2i are currently being tested for their efficacy across the whole spectrum of MASLD. Statins alone or in combination with ezetimibe have yielded promising results. The conduction of long-duration, large, high-quality, randomized-controlled trials aiming to assess by biopsy the efficacy of cardiometabolic drugs to reverse MASLD progression is of great importance.
Collapse
Affiliation(s)
- Marios Zisis
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece; (M.Z.); (I.R.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodoros Androutsakos
- Department of Pathophysiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Ilias Rantos
- Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Greece; (M.Z.); (I.R.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, University of Athens Medical School, 11527 Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
42
|
Wang Y, Chen Z, Wei B, Zhang R, Zhang X, Xu W. Metabolic dysfunction associated steatotic liver disease is associated with atrial fibrillation recurrence following cryoballoon ablation. Sci Rep 2025; 15:6287. [PMID: 39984596 PMCID: PMC11845672 DOI: 10.1038/s41598-025-90667-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: 08/17/2024] [Accepted: 02/14/2025] [Indexed: 02/23/2025] Open
Abstract
Atrial fibrillation (AF) is a common arrhythmia often treated with cryoballoon ablation. The impact of Metabolic-associated fatty liver disease (MASLD), a condition newly defined by a fatty liver index ≥ 60, on AF recurrence post-ablation is unclear. We analyzed 303 patients undergoing cryoballoon ablation for AF. Cox proportional hazards models were used to assess the relationship between MASLD and AF recurrence. Paroxysmal atrial fibrillation was present in 61.1% of patients and 63% were male. Among the patients, 23.4% had MASLD. These patients exhibited larger left atrial diameter and left ventricular end-diastolic dimension. During a median follow-up of 14 months, AF recurrence was more frequent in MASLD patients (45.1% vs. 20.7%). MASLD independently predicted AF recurrence (HR, 2.24 [95% CI 1.35-3.74], P = 0.002), alongside persistent AF, longer AF duration, and larger left atrial diameter. MASLD consistently demonstrated a significant association with an increased risk of AF recurrence in both paroxysmal (HR, 2.38 [95% CI, 1.08-5.23], P = 0.031) and persistent AF (HR, 2.55 [95% CI, 1.23-5.26], P = 0.011). MASLD significantly increases the risk of AF recurrence after cryoballoon ablation, highlighting the importance of supporting targeted interventions of MASLD in the periprocedural management of AF.
Collapse
Affiliation(s)
- Yu Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China
| | - Zheng Chen
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Bingqian Wei
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - RuiXin Zhang
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinlin Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China.
| | - Wei Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China.
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
43
|
Jiang W, Zeng Q, Liu CH, Wang Y, Wang S, Chen E, Wang M, Zhou T, Bai L, Wu D, Tang H. Huc-MSCs-derived exosomes alleviate non-alcoholic steatohepatitis by regulating macrophages polarization through miR-24-3p/STING axis. Stem Cell Res Ther 2025; 16:74. [PMID: 39984996 PMCID: PMC11846240 DOI: 10.1186/s13287-025-04197-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND There's a scarcity of drugs effective against nonalcoholic steatohepatitis (NASH). Exosomes from Human umbilical cord mesenchymal stem cells (huc-MSCs) show potential in managing glycolipid metabolism and the immune response. Therefore, further investigations are required to explore their application in NASH and the underlying mechanisms. METHODS C57BL/6J mice were fed with a western diet for 12 weeks to induce NASH, and huc-MSCs exosomes (MSCs-exo) were administered during the feeding period. The effect of MSCs-exo was evaluated by monitoring changes in body weight, fat distribution, blood glucose, and insulin levels, and analyzing pathological alterations in liver tissue. Mechanism investigations were carried out using flow cytometry, immunofluorescence staining, and other experimental techniques. RESULTS MSCs-exo could reduce liver fat, inflammation, fibrosis, and improved metabolism to alleviate the progression of NASH. Besides, MSCs-exo could decrease macrophage accumulation in the liver, encouraging M2 over M1 macrophage polarization. Furthermore, our study found that MSCs-exo had a high expression of miR-24-3p, which may regulate macrophage polarization by targeting the interferon-stimulated genes (STING) gene in macrophages, with its overexpression amplifying MSCs-exo's NASH benefits. CONCLUSIONS These findings suggest that the therapeutic effect of MSCs-exo on NASH may be attributed to the regulation of macrophage M2 polarization through miR-24-3p targeting STING. This provides a scientific basis for future clinical application.
Collapse
Affiliation(s)
- Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Shisheng Wang
- Liver Surgery and Liver Transplant Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Taoyou Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
44
|
Dong R, Tian T, Ming C, Zhang R, Xue H, Luo Z, Shen C, Ni Y, Shao J, Wang J. Multifaceted environmental factors linked to metabolic dysfunction-associated fatty liver disease: an environment-wide association study. BMC Public Health 2025; 25:709. [PMID: 39979906 PMCID: PMC11843789 DOI: 10.1186/s12889-025-21930-1] [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: 06/23/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Environmental factors, or exposome, are non-negligible contributors to the occurrence and progression of metabolic dysfunction-associated fatty liver disease (MAFLD). Therefore, this environment-wide association study (EWAS) aimed to investigate the associations between multifarious environmental factors and MAFLD among the general adult population in the United States. METHODS Eligible participants were obtained from the National Health and Nutrition Examination Survey 2005-2020 cycles. Survey-weighted multivariate logistic regression models were constructed to identify and tentatively validate MAFLD-associated environmental factors. The least absolute shrinkage and selection operator (LASSO) regression was conducted to identify tentatively validated environmental factors with stronger associations with MAFLD. Moreover, the importance, discrimination power, correlation patterns, subgroup-specific differences, and survey cycle heterogeneity of the identified factors were further examined by multiple statistical strategies. RESULTS A total of 14,416 participants were included in this EWAS. Among 511 candidate environmental factors, 167 were identified and tentatively validated, and 45 were preserved after the LASSO selection and correlation evaluation. In this study, most previously known factors were replicated with reduced bias, and several poorly studied environmental factors were discovered, for example, upper leg length, access to care, mid-upper arm circumference, and total trabecular bone score. Their importance, discrimination ability, pairwise correlations, subgroup variations, and heterogeneity across survey cycles were further systematically and rigorously evaluated. CONCLUSIONS This EWAS comprehensively explored the associations between environmental factors and MAFLD in the general adult population from a panoramic perspective. The findings may provide clues for further understanding this disease and promote early prevention and risk prediction strategies in the future.
Collapse
Affiliation(s)
- Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Jiangsu, 211166, China
| | - Ting Tian
- Institute of Nutrition and Food Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Chen Ming
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Ru Zhang
- School of Nursing and Midwifery, Jiangsu College of Nursing, Huaian, China
| | - Hong Xue
- Department of Liver Disease, Third Affiliated Hospital of Nantong University, Nantong, China
| | - Zhenghan Luo
- East China Institute of Biomedical Technology, Nanjing, China
| | - Chao Shen
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Yunlong Ni
- Institute of Nutrition and Food Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jianguo Shao
- Department of Gastroenterology, Third Affiliated Hospital of Nantong University, 60 Qingnian Middle Avenue, Chongchuan District, Jiangsu, 226001, China.
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Jiangsu, 211166, China.
| |
Collapse
|
45
|
Li Q, Xiang J. METTL3 promotes the progression of non-alcoholic fatty liver disease by mediating m6A methylation of FAS. Sci Rep 2025; 15:6162. [PMID: 39979577 PMCID: PMC11842791 DOI: 10.1038/s41598-025-90419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N6-methyladenosine (m6A) is involved in the development of non-alcoholic fatty liver disease (NAFLD). Here, we aimed to investigate the effect of m6A methyltransferase METTL3 on liver damage in high-fat diet (HFD)-induced mouse model and hepatocyte damage treated with free fatty acid (FFA). Plasma lipid, lipogenesis, viability, and apoptosis were measured to assess injury. m6A methylation was evaluated using m6A dot blot, methylated RNA immunoprecipitation, dual-luciferase reporter assay, and RNA decay assay. The results indicated that METTL3 was highly expressed in the liver of HFD mice, which knockdown improved plasma lipid and reduced liver lipids. Additionally, silencing of METTL3 promoted cell viability, inhibited apoptosis, reduced lipid concentrations, and downregulated lipogenesis-related marker levels. Moreover, METTL3 promoted the m6A methylation of FAS and enhanced its stability. In conclusion, silencing of METTL3 attenuates the progression of NAFLD by FAS m6A methylation, suggesting that METTL3 may be a promising target for treating NAFLD.
Collapse
Affiliation(s)
- Qunhua Li
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China
| | - Junying Xiang
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China.
| |
Collapse
|
46
|
Mi K, Ye T, Zhu L, Pan CQ. Risk-stratified hepatocellular carcinoma surveillance in non-cirrhotic patients with MASLD. Gastroenterol Rep (Oxf) 2025; 13:goaf018. [PMID: 39980834 PMCID: PMC11842057 DOI: 10.1093/gastro/goaf018] [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: 05/12/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 02/22/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly emerging as the leading global liver disorder and is poised to become the primary cause of hepatocellular carcinoma (HCC). Research indicates that nearly 50% of HCC cases in MASLD patients occur without cirrhosis, often presenting with more advanced and larger tumors. Despite this, current guidelines primarily focus on HCC screening in cirrhotic patients, with limited guidance for non-cirrhotic MASLD individuals. This narrative review seeks to identify key risk factors for HCC development, consolidate available screening methods, and propose a practical, risk-stratified algorithm for HCC surveillance in non-cirrhotic MASLD patients. We conducted a comprehensive review of studies published between 2017 and 2023 using PubMed, Embase, and CNKI, focusing on HCC risk factors and emerging screening strategies for non-cirrhotic MASLD cohorts. Key risk factors for HCC development in these patients include male sex, age over 65, hypertension, diabetes, mild alcohol consumption, smoking, dyslipidemia, elevated alanine aminotransferase levels, and a platelet count ≤ 150 × 109/L. Among the screening methods evaluated, circulating free DNA, alpha-fetoprotein (AFP) combined with protein induced by vitamin K absence or antagonist-II (PIVKA-II), and the GALAD score (incorporating Glypican-3, AFP, alpha-1-Antitrypsin, and des-gamma-carboxy prothrombin) demonstrated the highest performance. Based on these findings, we proposed a risk-stratified HCC surveillance algorithm that integrates GALAD and PIVKA-II into the existing sonography and AFP screening protocols. This review aims to provide clinicians with actionable recommendations for HCC screening in non-cirrhotic MASLD patients.
Collapse
Affiliation(s)
- Ke Mi
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Tingdan Ye
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Lin Zhu
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Calvin Q Pan
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
- Division of Gastroenterology and Hepatology, Department of Medicine, NYU Langone Health, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
47
|
Ko CJ, Lin HY, Hsieh PM, Wang WL, Chen SY, Chou LW, Chen YS, Huang YW, Ho WC, Lin CW. Association of concomitant MASLD and hepatitis B virus with clinical prognosis in hepatocellular carcinoma after curative resection. Am J Cancer Res 2025; 15:737-748. [PMID: 40084366 PMCID: PMC11897618 DOI: 10.62347/ksln5850] [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: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
The term "metabolic dysfunction-associated steatotic liver disease" (MASLD) was introduced to replace the term "nonalcoholic fatty liver disease". The prevalence of MASLD is increasing worldwide. The prevalence of concomitant MASLD and hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) is also increasing. This study explored the effect of the coexistence of MASLD and HBV on clinicopathological features and long-term clinical prognoses in patients with MASLD-related and/or HBV-related HCC after curative hepatectomy. The study retrospectively collected the data of 653 patients with HCC who had undergone curative hepatectomy between 2011 and 2022. We assessed the association of histologically confirmed MASLD with HCC recurrence and mortality. Of 653 patients, 320 (49.0%), 103 (15.8%), and 230 (35.2%) had concomitant MASLD and HBV, MASLD only, and HBV only, respectively. The median follow-up period was 5.1 years. Patients with concomitant MASLD and HBV were at a significantly increased risk of HCC recurrence (P = 0.013 and P = 0.041) and mortality (P = 0.044 and P = 0.026) than those with MASLD or HBV alone. In multivariable analyses, concomitant MASLD and HBV, male sex, body mass index < 23, absence of antiviral therapy, and tumor size ≥ 5 cm were significantly associated with increased HCC recurrence. Concomitant MASLD and HBV, male sex, type 2 diabetes mellitus, serum aspartate aminotransferase ≥ 40 U/L, tumor size ≥ 5 cm, tumor cell differentiation II-III, microvascular invasion, lymph node invasion, and tumor recurrence were significantly associated with increased mortality. In conclusion, patients with concomitant MASLD and HBV are at a significantly greater risk of HCC recurrence and mortality after curative hepatectomy than those with MASLD or HBV alone.
Collapse
Affiliation(s)
- Chih-Jan Ko
- Department of Public Health, China Medical UniversityTaichung, Taiwan
- Department of General Surgery, China Medical University Hsinchu HospitalHsinchu, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, and Research Center for Traditional Chinese Medicine, China Medical UniversityTaichung, Taiwan
| | - Hung-Yu Lin
- Department of Surgery, E-Da Cancer Hospital, I-Shou UniversityKaohsiung, Taiwan
- Department of Surgery, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Pei-Min Hsieh
- Department of Surgery, E-Da Dachang Hospital, I-Shou UniversityKaohsiung, Taiwan
- Division of Occupational Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Wen-Lung Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
| | - Szu-Ying Chen
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- Division of Surgical Intensive Care, Department of Critical Care Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- Department of Nursing, Fooyin UniversityKaohsiung, Taiwan
| | - Li-Wei Chou
- Department of Physical Medicine and Rehabilitation, China Medical University HospitalTaichung, Taiwan
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical UniversityTaichung, Taiwan
- Department of Physical Medicine and Rehabilitation, Asia University Hospital, Asia UniversityTaichung, Taiwan
| | - Yaw-Sen Chen
- Department of Surgery, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Yu-Wei Huang
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- Department of Nursing, Fooyin UniversityKaohsiung, Taiwan
- Department of Anesthesiology, Emergency and Critical Care Center, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Wen-Chao Ho
- Department of Public Health, China Medical UniversityTaichung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, and Research Center for Traditional Chinese Medicine, China Medical UniversityTaichung, Taiwan
| |
Collapse
|
48
|
Mei J, Xie Y, Huang P, Jin Y, Wang X, Chen Y. The effects of HAPA theory-based case management in patients with metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025; 30:101790. [PMID: 39954736 DOI: 10.1016/j.aohep.2025.101790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 02/17/2025]
Abstract
INTRODUCTION AND OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common liver disease affecting people's health, with its incidence increasing year by year. This study aims to determine the effects of Health Action Process Approach (HAPA)-based health management on patients diagnosed with MASLD. PATIENTS AND METHODS 204 MASLD patients were selected from the hospital's physical examination center from January 1, 2023, through April 1, 2023. Patients are randomly assigned to two groups (n = 102 each) using an envelope-based. Individuals in the experimental group received case management based on the HAPA theory, while standard health management was employed for control group patients. All subjects were monitored over a 6-month period, comparing body mass index (BMI), waist-to-hip ratio (WHR), levels of liver function (AST, ALT), blood lipid levels (total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and health behaviours (as assessed with the HPLP-II health promotion lifestyle scale) before and after the intervention. RESULTS Post-intervention, the experimental group showed significant improvement in fatty liver (P < 0.01) and reductions in biochemical indices, BMI, and WHR levels (P < 0.05), and a corresponding improvement in HDL-C levels (P < 0.001), compared to the control group. Additionally, patients in the experimental group exhibited significantly better health behaviour scores related to stress management, exercise, diet, and health responsibility compared to controls (P < 0.01). CONCLUSIONS HAPA theory-based case management can improve blood lipid profiles, liver function, and health-related behaviours in MASLD patients, highlighting its potential as an effective management strategy for this condition.
Collapse
Affiliation(s)
- Jia Mei
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yuncai Xie
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Pingping Huang
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yudi Jin
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xia Wang
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Ying Chen
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
49
|
Wu Y, Dong P, Wu Q, Zhang Y, Xu G, Pan C, Tong H. Insights into Clinical Trials for Drugs Targeting MASLD: Progress, Challenges, and Future Directions. Clin Pharmacol Ther 2025. [PMID: 39953659 DOI: 10.1002/cpt.3606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
The transition in terminology from fatty liver disease to metabolic dysfunction-associated steatotic liver disease (MASLD) marks a considerable evolution in diagnostic standards. This new definition focuses on liver fat accumulation in the context of overweight/obesity, type 2 diabetes, or metabolic dysfunction, without requiring the exclusion of other concurrent liver diseases. The new definition also provides clear guidelines for defining alcohol consumption in relation to the disease. MASLD is currently acknowledged as the most widespread liver disorder globally, affecting ~25% of the population. Despite the extensive array of clinical trials conducted in recent years, the number of approved treatments for metabolic dysfunction-associated fatty liver disease is very limited. In the review critically evaluates the results of clinical trials of related drugs and assesses the future directions for drug development trials. The renaming of MASLD presents new challenges and opportunities for the design of clinical trials and the selection of target populations for drug development.
Collapse
Affiliation(s)
- Yu Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Pu Dong
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qifang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Ya Zhang
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gang Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenwei Pan
- Department of Infectious Diseases, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou, China
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| |
Collapse
|
50
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [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: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|