1
|
Gbadamosi SO, Evans KA, Brady BL, Hoovler A. Noninvasive tests and diagnostic pathways to MASH diagnosis in the United States: a retrospective observational study. J Med Econ 2025; 28:314-322. [PMID: 39963742 DOI: 10.1080/13696998.2025.2468582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/03/2025]
Abstract
AIM Although liver biopsy is considered the most reliable diagnostic tool for metabolic dysfunction-associated steatohepatitis (MASH), it is invasive and can be costly. Clinicians are increasingly relying on routine biomarkers and other noninvasive tests (NITs) for diagnosis. We examined real-world diagnostic pathways for patients newly diagnosed with MASH with a primary focus on NITs. MATERIALS AND METHODS This retrospective, observational study analyzed healthcare claims data (Merative MarketScan Commercial and Medicare Databases) from patients in the United States newly diagnosed with MASH from October 1, 2016, to March 31, 2023. Patients ≥18 years old with ≥12 months of continuous enrollment with medical and pharmacy benefits prior to diagnosis were included. Diagnostic pathways leading up to MASH diagnosis, including NITs (blood-based and imaging-based tests) and liver biopsies were assessed. Prevalence of comorbid conditions, MASH-associated medication use, and the diagnosing physician specialty were also examined. RESULTS A total of 18,396 patients were included in the analysis. Routine laboratory tests (alanine aminotransferase [ALT], albumin, aspartate aminotransferase [AST], cholesterol, complete blood count, and hemoglobin A1c) were performed among ≥70% of patients prior to MASH diagnosis, including 89% of patients with a liver enzyme test (ALT and/or AST). More than 75% of patients had necessary laboratory tests to calculate AST to platelet ratio index (APRI) and fibrosis-4 index (FIB-4) scores. The most common imaging performed was ultrasound (62%); liver biopsy was only performed in 10% of patients. There was a high prevalence of cardio metabolic risk factors such as hyperlipidemia (66%), hypertension (62%), obesity (58%), type 2 diabetes (40%), and cardiovascular disease (21%). Nearly half of the patients (49%) were diagnosed by a primary care physician. LIMITATIONS AND CONCLUSIONS This study highlights real-world diagnostic pathways among patients newly diagnosed with MASH, supporting previous findings that liver biopsies are infrequently used in favor of noninvasive methods.
Collapse
|
2
|
Kalacun V, Ekart R, Bevc S, Skok P, Hojs R, Vodošek Hojs N. Oxidative stress and inflammation in hemodialysis: a comparison of patients with or without advanced nonalcoholic fatty liver disease (NAFLD). Ren Fail 2025; 47:2455523. [PMID: 39842820 PMCID: PMC11755732 DOI: 10.1080/0886022x.2025.2455523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and chronic kidney disease are global public health issues associated with high morbidity and mortality. Both diseases are also interlinked. Little is known about the meaning of NAFLD in hemodialysis (HD) patients. Therefore, the aim of our study was to investigate the difference in oxidative stress and inflammation in HD patients with or without advanced NAFLD. Seventy-seven HD patients were included (65.14 ± 12.34 years, 59.2% male) and divided according to abdominal ultrasound and two-dimensional shear wave elastography (2D-SWE) measurements into two groups: 1) no NAFLD or no advanced NAFLD (2D-SWE <9 kPa) and 2) advanced NAFLD (2D-SWE ≥9 kPa). Medical history data and blood results were collected. HD patients with advanced NAFLD had significantly higher levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG; p = 0.025), tumor necrosis factor-alpha (TNF-α; p = 0.023), and intercellular adhesion molecule 1 (ICAM-1; p = 0.015) in comparison to HD patients without advanced NAFLD. Interleukin 6 (IL-6) was higher in the advanced NAFLD group, but the difference was of borderline significance (p = 0.054). There was no significant difference in high-sensitivity C-reactive protein (hs-CRP), and vascular cell adhesion molecule 1 (VCAM-1) between groups. In binary logistic regression analysis, advanced NAFLD was significantly associated with 8-OHdG and ICAM-1. In conclusion, higher oxidative stress and inflammation levels are present in HD patients with advanced NAFLD.
Collapse
Affiliation(s)
- Vanja Kalacun
- Department of Gastroenterology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Sebastjan Bevc
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Pavel Skok
- Department of Gastroenterology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Radovan Hojs
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Nina Vodošek Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| |
Collapse
|
3
|
Wang Z, Tang Y, Zhang Y, Li Y, Chen C, Gao S, Qiao L. Nanomaterials as novel matrices to improve biomedical applications of MALDI-TOF/MS. Talanta 2025; 293:128092. [PMID: 40215718 DOI: 10.1016/j.talanta.2025.128092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025]
Abstract
With the rapid development of biomedical technology, there is an increasing demand for accurate analysis of biomolecules and their interactions. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) is a novel soft ionization biomass spectrometry technology that can accurately assess the molecular weight of samples quickly and sensitively, and it plays an important role in the analysis and detection of large molecule substances, drug research, and life sciences. In recent years, due to the outstanding performance of nanomaterials, they have been widely used as matrices in drug metabolism research and cancer detection. This paper aims to review the latest research progress of nanomaterials as new matrices for MALDI-TOF MS in enhancing biomedical analysis, discus its value and limitations, and look forward to its future research direction.
Collapse
Affiliation(s)
- Zhiyi Wang
- College of Phamaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Yuanting Tang
- College of Phamaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Ying Zhang
- College of Phamaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Yingjing Li
- College of Phamaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Cong Chen
- Academy of Chinese Medicine Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Shijie Gao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Li Qiao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| |
Collapse
|
4
|
Victor DW, Kodali S, Noureddin M, Brombosz EW, Lopez A, Basra T, Graviss EA, Nguyen DT, Saharia A, Connor AA, Abdelrahim M, Cheah YL, Simon CJ, Hobeika MJ, Mobley CM, Ghobrial RM. Disparities in liver transplantation for metabolic dysfunction-associated steatohepatitis-associated hepatocellular carcinoma. World J Transplant 2025; 15:101997. [DOI: 10.5500/wjt.v15.i3.101997] [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: 10/04/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is increasingly common, as is hepatocellular carcinoma (HCC) in the background of MASH. Liver transplantation (LT) provides superior long-term survival for patients with unresectable MASH-HCC, but not all patients have equal access to transplant. MASH-HCC disproportionately affects Hispanic patients, but minorities are less likely to undergo LT for HCC. Additionally, females also undergo LT at lower rates than males.
AIM To investigate whether race/ethnicity and sex affect LT waitlist outcomes.
METHODS Records of adults with MASH-HCC in the United States Organ Procurement and Transplantation Network database listed for LT between 1/2015 and 12/2021 were analyzed.
RESULTS Most of the 3810 patients waitlisted for LT for MASH-HCC were non-Hispanic (NH) white (71.2%) or Hispanic (23.4%), with only 49 (1.1%) NH Black candidates. Hispanics underwent LT at lower rates than NH whites (71.6% vs 78.4%, P < 0.001), but race/ethnicity did not affect waitlist mortality (P = 0.06). Patients with Hispanic [hazard ratio (HR) = 0.85, 95%CI: 0.77-0.95, P = 0.002] or Asian (HR = 0.79, 95%CI: 0.63-0.98, P = 0.04) race/ethnicity were less likely to undergo LT. Women were also less likely to receive LT (male: HR = 1.16, 95%CI: 1.04-1.29, P = 0.01). Patients in regions 1 and 9 were less likely to be transplanted as well (P = 0.07).
CONCLUSION Hispanic patients are less likely to undergo LT for MASH-HCC, concerning given their susceptibility to MASH and HCC. There were very few NH Black candidates. Disparities were also unequal across regions, which is particularly concerning in states where at-risk populations have rising cancer incidence. Additional research is needed to identify strategies for mitigating these differences in access to LT for MASH-HCC.
Collapse
Affiliation(s)
- David W Victor
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Sudha Kodali
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Mazen Noureddin
- Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
- Houston Research Institute, Houston, TX 77079, United States
| | - Elizabeth W Brombosz
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Analisa Lopez
- JC Walter Jr Transplant Center, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Tamneet Basra
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Duc T Nguyen
- Department of Pediatrics, Baylor College of Medicine, Houston, 77030, United States
| | - Ashish Saharia
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Ashton A Connor
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Maen Abdelrahim
- Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Yee Lee Cheah
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Caroline J Simon
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Mark J Hobeika
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - Constance M Mobley
- JC Walter Jr Transplant Center, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| | - R Mark Ghobrial
- J C Walter Jr Transplant Center, Sherrie and Alan Conover Center for Liver Disease and Transplantation, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, United States
| |
Collapse
|
5
|
Gao F, Ma Y, Yu C, Duan Q. miR-125b-5p regulates FFA-induced hepatic steatosis in L02 cells by targeting estrogen-related receptor alpha. Gene 2025; 959:149419. [PMID: 40113187 DOI: 10.1016/j.gene.2025.149419] [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: 12/10/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND & AIMS NAFLD is a global and complex liver disease caused by multiple factors. Intrahepatocellular steatosis is the primary prerequisite for the occurrence and development of NAFLD. It has been shown that miR-125b-5p is highly correlated with NAFLD, and ESRRA is a factor that regulates lipid metabolism. The purpose of our study is to investigate whether miR-125b-5p regulates FFA-induced steatosis in L02 cells by targeting ESRRA. APPROACHES AND RESULTS Estrogen-related receptor alpha (ESRRA) was identified as a direct target of miR-125b-5p through database prediction and a dual-luciferase reporter gene assay. L02 cells were induced with free fatty acids (OA:PA, 2:1) at concentrations of 0.3 mM, 0.6 mM, 0.9 mM, 1.2 mM and 1.5 mM for 24 h, 48 h and 72 h, respectively. The degree of hepatocyte steatosis and triglyceride content were separately manifested by oil red O staining and colorimetric method. Cell viability per group was detected by CCK-8 assay. Eventually, 0.9 mM and 24 h were screened out as the optimal concentration and time for establishing the in-vitro model of hepatic steatosis. Followingly, miR-125b-5p and ESRRA were knocked down by transient transfection. We monitored the expressions of lipid metabolism factors SREBP-1c, ACC1 and FAS and determine triglyceride content within the cells per group. The data showed that knockdown of ESRRA led to down-regulation of the expressions of SREBP-1, ACC1, FAS and triglyceride content. Meanwhile, knockdown of ESRRA and miR-125b-5p resulted that the expressions of ESRRA, SREBP-1, ACC1, FAS and triglyceride content rebounded. CONCLUSIONS MiR-125b-5p down-regulates the expressions of lipid metabolism-related factors by negatively regulating ESRRA, thereby improving hepatic steatosis.
Collapse
Affiliation(s)
- Fen Gao
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Yanhua Ma
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Chun Yu
- Gansu University of Chinese Medicine, Gansu 730000, China
| | | |
Collapse
|
6
|
Choi MA, Rose S, Langouët S. Per- and polyfluoroalkyl substances as potentiators of hepatotoxicity in an exposome framework: Current challenges of environmental toxicology. Toxicology 2025; 515:154167. [PMID: 40300710 DOI: 10.1016/j.tox.2025.154167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/17/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Chronic liver diseases, including metabolic dysfunction-associated steatosic liver disease (MASLD) and hepatocellular carcinoma (HCC), are on the rise, potentially due to daily exposure to complex mixtures of chemical compounds forming part of the exposome. Understanding the mechanisms involved in hepatotoxicity of these mixtures is essential to identify common molecular targets that may highlight potential interactions at the molecular level. We illustrated this issue with two families of environmental contaminants, per- and polyfluoroalkyl substances (PFAS) and heterocyclic aromatic amines (HAAs), both of which could be involved in the progression of chronic liver diseases, and whose toxicity may be potentiated by interactions at the level of xenobiotic metabolism. In the study of exposome effects on chronic liver disease, New Approach Methodologies (NAMs) including omics analyses and data from various in vitro, in vivo and in silico approaches, are crucial for improving predictivity of toxicological studies in humans while reducing animal experimentation. Additionally, the development of complex in vitro human liver cell models, such as organoids, is essential to avoid interspecies differences that minimize the risk for humans. All together, these approaches will contribute to construct Adverse Outcome Pathways (AOPs) and could be applied not only to PFAS mixtures but also to other chemical families, providing valuable insights into mixture hepatotoxicity prediction in the study of the exposome. A better understanding of toxicological mechanisms will clarify the role of environmental contaminant mixtures in the development of MASLD and HCC, supporting risk assessment for better treatment, monitoring and prevention strategies.
Collapse
Affiliation(s)
- Minna A Choi
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France.
| |
Collapse
|
7
|
van Kleef LA, Pustjens J, Janssen HLA, Brouwer WP. Diagnostic Accuracy of the LiverRisk Score to Detect Increased Liver Stiffness Among a United States General Population and Subgroups. J Clin Exp Hepatol 2025; 15:102512. [PMID: 40093506 PMCID: PMC11908561 DOI: 10.1016/j.jceh.2025.102512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 03/19/2025] Open
Abstract
Background The LiverRisk score (LRS) has recently been proposed to predict liver fibrosis and future development of liver-related outcomes in the general population. Here, we performed an external validation of this score. Methods We used data from National Health and Nutrition Examination Survey 2017-2020, a United States population-based cohort to assess the diagnostic accuracy of the LRS to detect a liver stiffness measurement (LSM) ≥8 and ≥12 kPa. Performance was tested among the entire general population and clinically relevant subgroups. Results The cohort comprised 7,025 participants (aged 49 [33-63], 49% male), and 9.7% had an LSM ≥8 and 3.2% had an LSM ≥12 kPa. The area under the receiver characteristic operator curve (AUC) in the overall population was 0.73 (95% confidence interval [CI] :0.71-0.75) and 0.78 (95% CI: 0.74-0.81) to detect an LSM ≥8 and ≥ 12 kPa, respectively, significantly outperforming the fibrosis 4 index (FIB-4) but not the nonalcoholic fatty liver disease fibrosis score, steatosis-associated fibrosis estimator (SAFE), or metabolic dysfunction-associated fibrosis 5 (MAF-5). Performance was consistent among most subgroups, but AUC levels to detect an LSM ≥8 kPa decreased to <0.70 among participants aged 18-40 or 60-80 years, blacks, and individuals with diabetes or liver steatosis. The LRS categorized 80.5% as very low risk, 17.7% as low risk, and 1.8% as at risk, prevalence of an LSM ≥8 in these groups was 6.3%, 20.8%, and 50.5%, respectively. The sensitivity to detect an LSM ≥8 kPa was 47.3% in the overall population (but dropped to 21.3% for individuals aged 18-40 years) despite applying the lowest cut-off, which should yield the highest sensitivity. Conclusion The LRS score is a promising new tool to predict liver fibrosis; however, its diagnostic accuracy attenuates especially among patients aged 18-40 or 60-80 years. The overall sensitivity was only 47.3% at the lowest LRS cut-off. Further studies assessing cost-benefit ratios according to the LRS compared to FIB-4 and other risk scores such as MAF-5 and SAFE are required to determine its usefulness in referral strategies.
Collapse
Affiliation(s)
- Laurens A van Kleef
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Jesse Pustjens
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Canada
| | - Willem P Brouwer
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Wu D, Liu J, Guo Z, Wang L, Yao Z, Wu Q, Lu Y, Lv W. Natural bioactive compounds reprogram bile acid metabolism in MAFLD: Multi-target mechanisms and therapeutic implications. Int Immunopharmacol 2025; 157:114708. [PMID: 40306110 DOI: 10.1016/j.intimp.2025.114708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/20/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
Metabolic-associated fatty liver disease (MAFLD) has become an increasingly prevalent liver disorder worldwide, being closely associated with obesity, metabolic syndrome, and insulin resistance. Bile acids (BAs), beyond their traditional role in lipid digestion, play a pivotal part in regulating lipid and glucose metabolism as well as inflammatory responses. Recent investigations have recognized BAs as key factors in the onset and progression of MAFLD, mainly via their interactions with nuclear receptors such as the farnesoid X receptor (FXR) and the G protein-coupled bile acid receptor (TGR5). Additionally, active compounds derived from traditional Chinese medicine (TCM) have shown promising potential in the treatment of MAFLD. This study systematically reviews and analyzes the molecular mechanisms and recent progress in the application of TCM active ingredients for MAFLD treatment, with a focus on their regulation of BAs. These active ingredients, including saponins, flavonoids, polysaccharides, and sterols, exert therapeutic effects through diverse mechanisms, such as modulating BA synthesis and mediating receptor-signaling pathways, and are expected to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dongjie Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Liu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ziwei Guo
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Liang Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Qingjuan Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yanping Lu
- Department of Hepatology, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Shenzhen 518100, China.
| | - Wenliang Lv
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
9
|
Wang J, Yang R, Miao Y, Zhang X, Paillard‐Borg S, Fang Z, Xu W. Metabolic Dysfunction-Associated Steatotic Liver Disease Is Associated With Accelerated Brain Ageing: A Population-Based Study. Liver Int 2025; 45:e70109. [PMID: 40296771 PMCID: PMC12038381 DOI: 10.1111/liv.70109] [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: 02/06/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is linked to cognitive decline and dementia risk. We aimed to investigate the association between MASLD and brain ageing and explore the role of low-grade inflammation. METHODS Within the UK Biobank, 30 386 chronic neurological disorders-free participants who underwent brain magnetic resonance imaging (MRI) scans were included. Individuals were categorised into no MASLD/related SLD and MASLD/related SLD (including subtypes of MASLD, MASLD with increased alcohol intake [MetALD] and MASLD with other combined aetiology). Brain age was estimated using machine learning by 1079 brain MRI phenotypes. Brain age gap (BAG) was calculated as the difference between brain age and chronological age. Low-grade inflammation (INFLA) was calculated based on white blood cell count, platelet, neutrophil granulocyte to lymphocyte ratio and C-reactive protein. Data were analysed using linear regression and structural equation models. RESULTS At baseline, 7360 (24.2%) participants had MASLD/related SLD. Compared to participants with no MASLD/related SLD, those with MASLD/related SLD had significantly larger BAG (β = 0.86, 95% CI = 0.70, 1.02), as well as those with MASLD (β = 0.59, 95% CI = 0.41, 0.77) or MetALD (β = 1.57, 95% CI = 1.31, 1.83). The association between MASLD/related SLD and larger BAG was significant across middle-aged (< 60) and older (≥ 60) adults, males and females, and APOE ɛ4 carriers and non-carriers. INFLA mediated 13.53% of the association between MASLD/related SLD and larger BAG (p < 0.001). CONCLUSION MASLD/related SLD, as well as MASLD and MetALD, is associated with accelerated brain ageing, even among middle-aged adults and APOE ɛ4 non-carriers. Low-grade systemic inflammation may partially mediate this association.
Collapse
Affiliation(s)
- Jiao Wang
- Center of Gerontology and GeriatricsNational Clinical Research Center for GeriatricsWest China Hospital, Sichuan UniversityChengduChina
- Aging Research Center, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Rongrong Yang
- Aging Research Center, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Public Health Science and Engineering CollegeTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yuyang Miao
- Tianjin Key Laboratory of Elderly Health, Department of Geriatrics, Tianjin Geriatrics InstituteTianjin Medical University General HospitalTianjinChina
| | - Xinjie Zhang
- Department of Pediatric Neurosurgery, West China Second University HospitalSichuan UniversityChengduChina
| | | | - Zhongze Fang
- Department of Toxicology and Health Inspection and Quarantine, School of Public HealthTianjin Medical UniversityTianjinChina
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Tianjin Key Laboratory of Elderly Health, Department of Geriatrics, Tianjin Geriatrics InstituteTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
10
|
Wang S, Du R, Liu J, Zhong W, Zhang C, Jiang X, Wang X, Wu Q, Tong G, Luo L. Multi-approach analysis reveals the mechanism by which Shugan Xiaozhi decoction protects against metabolic dysfunction-associated steatohepatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156712. [PMID: 40220418 DOI: 10.1016/j.phymed.2025.156712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is a human health-threatening hepatic disease with limited treatment strategies. As a clinical Traditional Chinese Medicine compound for MASH, Shugan Xiaozhi (SGXZ) decoction has a definite effect, but its mechanism in treating MASH is still not very clear. PURPOSE Exploring the potential mechanism of SGXZ decoction in treating MASH through multiomics and animal experimental validation. METHODS UPLC-ESI-MS method was used to identify the main components of SGXZ decoction. Periodic acid-schiff (PAS), picrosirius red (PSR), and oil red o staining were used to assess the effect of SGXZ decoction on MCD-induced MASH mouse model. The mechanism of SGXZ decoction on MASH was analyzed using multiomics techniques. TUNEL staining, western blot (WB), immunohistochemistry (IHC), kits, transmission electron microscopy (TEM), and immunofluorescence (IF) were used to validate the mechanism of SGXZ decoction on MASH. Finally, molecular docking and molecular dynamics simulation were used to verify the targeting between key components of SGXZ decoction and important targets for intervention. RESULTS Through UPLC-ESI-MS analysis, 30 main active ingredients were obtained from SGXZ decoction. SGXZ decoction improved MASH, as evidenced by the improvement in histopathology, hepatic function indexes, lipid and fibrosis indicators. Both proteomic and transcriptomic results suggested an important role for ferroptosis in SGXZ decoction intervention in MASH, ferroptosis-related pathways were the main significant pathways obtained from these analyses. In addition, SGXZ decoction treatment reduced cell death, inflammation, and oxidative stress levels and restored impaired mitochondrial morphology in MCD-induced MASH mice. Furthermore, Mechanism experiments proved that SGXZ decoction treatment improved iron metabolism and lipid peroxidation imbalance and activated the Xc- system in MASH mice. CONCLUSION SGXZ decoction does have a therapeutic effect on MASH, and its mechanism may be related to its regulation of p53/ SLC7A11/GPX4 pathway to reduce ferroptosis.
Collapse
Affiliation(s)
- Shuai Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Ruili Du
- The First Clinical Medical College of Henan University of Chinese Medicine, No. 19, Renmin Road, Jinshui District, Henan, 450003, PR China
| | - Jiahui Liu
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong 518033, PR China
| | - Weichao Zhong
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, PR China
| | - Xia Jiang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, PR China
| | - Xiaohui Wang
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao ln-Depth Cooperation Zone in Hengqin, 519000, PR China.
| | - Guangdong Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| | - Lidan Luo
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| |
Collapse
|
11
|
Hu Y, Li N, Zhang R, Wang J, Fang D, Zhou Q, Zhang H, Cai H, Lu Y. Linghe granules reduces hepatic lipid accumulation in Non-alcoholic fatty liver disease through regulating lipid metabolism and redox balance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156654. [PMID: 40220422 DOI: 10.1016/j.phymed.2025.156654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/15/2024] [Accepted: 03/15/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a prevalent liver disorder with no approved pharmacological therapies. Linghe granules, a hospital-based formulation derived from a classic prescription, have demonstrated potential in reducing hepatic fat accumulation and improving metabolic health. This study provides a novel, comprehensive assessment of Linghe granules, integrating clinical, preclinical, and molecular analyses for NAFLD management. PURPOSE This study aims to evaluate the therapeutic efficacy of Linghe granules in alleviating NAFLD through an integrated approach. METHODS A clinical trial involving 40 patients with NAFLD was conducted, with participants divided into a control group (lifestyle interventions) and a treatment group (lifestyle interventions plus oral Linghe granules). Various metabolic and liver function indicators were assessed before and after treatment. Additionally, a high-fat diet (HFD) was used to induce a NAFLD model in rat, followed by treatment with different doses of Linghe granules. In vitro studies on HepG2 and L02 cells were performed to the effects of the granules on lipid metabolism. Transcriptomic profiling, Weighted Gene Co-expression Network Analysis (WGCNA), Dynamic Network Biomarkers (DNB) analysis, and molecular docking were employed to explore the underlying mechanisms. RESULTS Linghe granules led to significant reductions in BMI, liver enzymes (AST, ALT), triglycerides, LDL-C, and GGT in patients with NAFLD, accompanied by a notable decrease in hepatic fat accumulation. In the rat model, treatment improved liver weight, liver function, and lipid metabolism. In vitro, Linghe granules decreased lipid accumulation and regulated key lipid metabolism markers, including sterol regulatory element-binding protein 1 (SREBP-1), stearoyl-CoA desaturase 1 (SCD1), and fatty acid-binding protein 5 (FABP5). Mechanistic analyses revealed that Linghe granules modulated oxidative stress-related pathways and genes involved in lipid metabolism. CONCLUSION This study represents the first integrated evaluation of Linghe granules' efficacy and mechanisms in treating NAFLD, demonstrating their potential to improve liver function, reduce lipid accumulation, and modulate key metabolic markers. These results suggest that Linghe granules may serve as an effective adjunctive treatment for NAFLD.
Collapse
Affiliation(s)
- Yuting Hu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ni'ao Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rumian Zhang
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361015, China
| | - Jia Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dongdong Fang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhang
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong Cai
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361015, China.
| | - Yiyu Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Kim JH, Lee Y, Nam CM, Kwon YJ, Lee JW. Impact of cardiometabolic risk factors for metabolic dysfunction-associated steatotic liver disease on mortality. Nutr Metab Cardiovasc Dis 2025; 35:103965. [PMID: 40187915 DOI: 10.1016/j.numecd.2025.103965] [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: 11/16/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a potential independent risk factor for cardiovascular disease (CVD)-associated and all-cause mortalities as they share common risk factors. We investigated the association between cardiometabolic risk factors for MASLD and CVD-associated and all-cause mortality risks in middle-aged and older Korean adults. METHODS AND RESULTS We used data from the Korean Genome and Epidemiology Study, a population-based prospective cohort study. Five cardiometabolic risk factors were assessed. MASLD was defined as liver steatosis with a fatty liver index (FLI) ≥60 and at least one cardiometabolic risk factor. The non-MASLD group included individuals with a FLI <60 or FLI ≥60 without cardiometabolic risk factors. The primary outcomes were CVD-associated and all-cause mortalities. Cox proportional hazard models were used to evaluate the association between cardiometabolic risk factors for MASLD and mortalities, adjusting for covariates. Multivariable Cox regression analysis revealed that the MASLD group had increased CVD-associated and all-cause mortality risks compared to the non-MASLD group. The presence of three or more and one or more cardiometabolic risk factors significantly increased the CVD-associated and all-cause mortality rate, respectively. The combination of hypertriglyceridemia, low high-density lipoprotein cholesterol (HDL-C), and high glucose concentrations significantly increased both CVD-associated (hazard ratio [HR] 3.64; 95 % confidence interval [CI] 1.44-9.22; p = 0.006) and all-cause (HR 4.57; 95 % CI: 1.74-12.05; p = 0.002) mortality risks. CONCLUSION Cardiometabolic risk factors for MASLD are strongly associated with higher CVD-associated and all-cause mortality risks, highlighting the need to manage hypertriglyceridemia, low HDL-C, and high glucose concentrations.
Collapse
Affiliation(s)
- Jung-Hwan Kim
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yaeji Lee
- Department of Biostatistics and Computing, Yonsei University, Seoul, 03722, Republic of Korea
| | - Chung-Mo Nam
- Department of Health Informatics and Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, 16995, Republic of Korea.
| | - Ji-Won Lee
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
13
|
He C, Zhou L, Gao T, Cao R, Cai C, Jiang G. Sex differences in the mediation of the MASLD - Depression association by fat distribution in U.S. adults. Acta Psychol (Amst) 2025; 256:105041. [PMID: 40300432 DOI: 10.1016/j.actpsy.2025.105041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
PURPOSE This study investigates the intricate relationship between metabolic dysfunction-associated steatotic liver disease (MASLD) and depression, emphasizing the mediating role of body fat distribution, particularly in female. METHODS We analyzed the pairwise relationship among body fat distribution (android fat and gynoid fat), MASLD, and depression within a robust, ethnically diverse sample (n = 3332) drawn from the National Health and Nutrition Examination Survey (NHANES). RESULTS Studies have indicated that individuals with depression or MASLD exhibit significantly higher levels of android fat compared to those without these health issues. Even after controlling for confounding factors, MASLD maintained significant correlations with both depression and body fat distribution (android fat and gynoid fat). Notably, sex moderated the relationship between MASLD, depression, and body fat distribution. Among the potential mediators of the effect of MASLD on depression, android fat emerged as a significant mediator, accounting for 16.6 % of the variance and yielding statistically significant results (p < 0.05). This mediating effect was particularly pronounced in female subjects, with a mediating proportion of 17.83 %, which was also statistically significant (p < 0.05). However, it was not observed in males. CONCLUSIONS This study reveals an important association between MASLD and depression, and android fat distribution is a potential mediator in this relationship, with a particularly pronounced effect on the female population.
Collapse
Affiliation(s)
- Changhuai He
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Liuxin Zhou
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Tianming Gao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Runmin Cao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Guoqing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu 225001, China.
| |
Collapse
|
14
|
Pitkänen S, Niskanen J, Mysore R, Niskanen EA, Palvimo JJ, Pijnenburg D, van Beuningen R, Rashidian A, Kronenberger T, Poso A, Levonen AL, Küblbeck J, Honkakoski P. Activation of steroid hormone receptors by metabolism-disrupting chemicals. Toxicol Appl Pharmacol 2025; 499:117335. [PMID: 40216312 DOI: 10.1016/j.taap.2025.117335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
Exposure to metabolism-disrupting chemicals (MDCs), compounds largely belonging to the group of endocrine-disrupting chemicals (EDCs), is associated with metabolic dysfunctions such as dyslipidemia, insulin resistance and hepatic steatosis. Steroid hormone receptors (SHRs) are known targets for MDCs but their regulatory environment in the presence of environmental chemicals remains elusive. Here, we studied the activation and molecular interactions of SHRs exposed to 17 suspected MDCs including pesticides, plasticizers, pharmaceuticals, flame retardants, industrial chemicals and their metabolites by combining in vitro and in silico approaches. We first established and pre-validated reporter gene assays in HepG2 hepatoma cells to assess the activation of estrogen (ER), androgen (AR), glucocorticoid (GR) and progesterone (PR) receptors. Next, using RNA-seq and publicly available protein interaction data, we identified relevant SHR-interacting coregulators expressed in hepatic cells and measured their MDC-dependent interactions with SHRs using the Microarray Assay for Real-time Coregulator-Nuclear receptor Interaction (MARCoNI) technology. Finally, we examined MDC binding to ER and GR using molecular dynamics simulations. These combined approaches lead to identification of MDCs capable of SHR activation at picomolar-to-low micromolar concentrations and paralleled with their ability to induce recruitment of multiple coregulators. MDCs induced distinct SHR-coregulator binding patterns involving multiple coactivators, corepressors and other modulatory proteins. Our results have broadened the test battery to detect MDCs and indicate that the activation of SHRs by MDCs is driven by diverse molecular interactions.
Collapse
Affiliation(s)
- Sini Pitkänen
- A. I. Virtanen -Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Jonna Niskanen
- A. I. Virtanen -Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Raghavendra Mysore
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Einari A Niskanen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Dirk Pijnenburg
- PamGene International B.V., Wolvenhoek 10, NL-5211 HH 's-Hertogenbosch, The Netherlands
| | - Rinie van Beuningen
- PamGene International B.V., Wolvenhoek 10, NL-5211 HH 's-Hertogenbosch, The Netherlands
| | - Azam Rashidian
- Partner-site Tübingen, German Center for Infection Research (DZIF), University Hospital of Tübingen, Tübingen, Germany
| | - Thales Kronenberger
- Partner-site Tübingen, German Center for Infection Research (DZIF), University Hospital of Tübingen, Tübingen, Germany; School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3, 70211 Kuopio, Finland
| | - Antti Poso
- Partner-site Tübingen, German Center for Infection Research (DZIF), University Hospital of Tübingen, Tübingen, Germany; School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3, 70211 Kuopio, Finland
| | - Anna-Liisa Levonen
- A. I. Virtanen -Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Jenni Küblbeck
- A. I. Virtanen -Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3, 70211 Kuopio, Finland.
| |
Collapse
|
15
|
Ni D, Qi Z, Ma S, Wang Y, Liang D, Zhang X, Man Y, Chen J, Dou K, Li G. Membrane-associated ring-CH-type finger 2 protects against metabolic dysfunction-associated fatty liver disease by targeting fatty acid synthase. Mol Metab 2025; 96:102137. [PMID: 40189099 DOI: 10.1016/j.molmet.2025.102137] [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: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as an important public health concern that poses a significant threat to human health and imposes a substantial economic burden. Research has demonstrated that ubiquitin ligase-mediated substrate protein ubiquitination is a pivotal factor influencing liver lipid homeostasis and metabolic abnormalities in MAFLD. Nevertheless, the specific enzyme molecules implicated in this regulatory process remain to be elucidated. We have published a transcriptome-overexpressing ubiquitin ligase, membrane-associated ring-CH-type finger 2 (MARCH2), in HepG2 cells, and subsequent reanalysis of these transcriptome data revealed a close association between MARCH2 and lipid metabolism. METHODS By employing a range of methodologies, including recombinant adeno-associated virus (rAAV) transduction, lentiviral transduction, immunoblotting, quantitative PCR, tissue section staining, ubiquitination assays, serum biochemical analysis, immunoprecipitation, and mass spectrometry, this study investigated the functions and mechanisms of MARCH2 in the progression of MAFLD at the molecular, cellular, and organismal levels. RESULTS Overexpression of MARCH2, but not its catalytically inactive ligase variant, inhibited lipid accumulation in HepG2 cells. Additionally, MARCH2 undergoes K48-linked self-polyubiquitination and subsequent proteasomal degradation in response to oleic acid/palmitic acid stimulation. Furthermore, knockout of MARCH2 exacerbates the progression of MAFLD-related phenotypes, including increased body weight, impaired glucose tolerance, reduced insulin sensitivity, hypercholesterolemia, hepatic lipid accumulation, and steatosis, in high-fat diet-fed mice, irrespective of sex. Mechanistically, MARCH2 facilitates the polyubiquitination and degradation of fatty acid synthase (FASN) in the de novo lipogenesis pathway. And liver-specific overexpression of MARCH2 by rAAV effectively reduces FASN levels and further ameliorates MAFLD in ob/ob mice. CONCLUSIONS MARCH2 undergoes self-ubiquitination and plays an important role in maintaining the liver lipid homeostasis of MAFLD, and drug intervention in the MARCH2-FASN axis is a promising approach for treating systemic metabolic abnormalities in MAFLD.
Collapse
Affiliation(s)
- Dongsheng Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Zhaolai Qi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Shuang Ma
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Yuefeng Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Dehuan Liang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Fifth School of Clinical Medicine (Beijing Hospital), Peking University, Beijing, 100730, PR China
| | - Xiyue Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Yong Man
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, PR China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, PR China.
| | - Kefei Dou
- Cardiometabolic Medicine Center, National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, PR China.
| | - Guoping Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, PR China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
16
|
Sun X, Yang Z, Li M, Gong S, Miao X, Wang B, Kong X, Zhu Q. Interferon regulatory factor 1 contributes to metabolic dysfunction associated steatotic liver disease. Life Sci 2025; 370:123575. [PMID: 40132726 DOI: 10.1016/j.lfs.2025.123575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
AIMS Non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction associated steatotic liver disease (MASLD), has reached epidemic levels in multiple regions worldwide and contributes to cirrhosis and hepatocellular carcinoma. We have previously reported that the CC motif chemokine ligand 11 (CCL11) is a key regulator of MASLD. Expression of interferon regulatory factor 1 (IRF1) can be up-regulated by CCL11 treatment in hepatocytes, the relevance of which is not clear. In the present study we investigated the role of IRF1 in NAFLD pathogenesis. METHODS AND MATERIALS MASLD was investigated in mice fed a high-fat high carbohydrate (HFHC) diet or in the genetically predisposed obese mice (db/db). KEY FINDINGS Hepatocytes from CCL11 knockout mice displayed a less severe MASLD phenotype, when treated with palmitic acid (PA), compared to wild type hepatocytes, which could be normalized by IRF1 over-expression. On the contrary, IRF1 knockdown in hepatocytes significantly down-regulated expression of pro-inflammatory mediators and dampened lipid accumulation induced by PA treatment. More importantly, IRF1 knockdown in hepatocytes led to amelioration of MASLD in mice. RNA-seq and CUT&Tag-seq identified pro-MASLD genes, including Osbpl3, Ddit4, and Ccl2, as potential targets for IRF1 in hepatocytes. SIGNIFICANCE Our data reveal a novel regulatory role of IRF1 in MASLD pathogenesis. Targeting IRF1 can be considered as a reasonable approach for MASLD intervention.
Collapse
Affiliation(s)
- Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Zhen Yang
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Min Li
- Center for Experimental Medicine, Department of Pathophysiology, Jiangsu Health Vocational College, China
| | - Shanwen Gong
- Institute of Biomedical Research, College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Xiulian Miao
- Institute of Biomedical Research, College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Bo Wang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University, the Wujin Clinical College of Xuzhou Medical University, Changzhou, China.
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qiang Zhu
- Department of Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
17
|
Zhuang C, Cui F, Chen J, He D, Sun T, Wang P. Rbm39 ameliorates metabolic dysfunction-associated steatotic liver disease by regulating Apob and Fabp4. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167815. [PMID: 40147697 DOI: 10.1016/j.bbadis.2025.167815] [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: 11/29/2024] [Revised: 03/12/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Excessive hepatic lipid accumulation is the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD), yet its underlying mechanisms still not fully understood. In this study, we identified RNA binding motif protein 39 (Rbm39) as a key modulator of hepatic lipid homeostasis during MASLD progression. To establish in vivo MASLD model, mice were fed either a high-fat diet (HFD) or a Gubra-Amylin NASH (GAN) diet. We employed adeno-associated virus to manipulate Rbm39 expression levels to assess its role in MASLD. Transcriptome analysis was conducted to pinpoint the genes targeted by Rbm39. Western blot, RT-PCR, dual-luciferase reporter gene assays, and alternative splicing analysis were utilized to delve into the molecular mechanisms. Our results showed that Rbm39 expression was notably decreased in the livers of MASLD mice. Knockdown of hepatic Rbm39 aggravated HFD-induced hepatic steatosis and GAN diet-induced MASH, along with a notable decrease in serum lipid levels. Conversely, overexpression of Rbm39 attenuated MASLD development and progression. RNA sequencing data analysis indicated that Rbm39 regulated the expression of apolipoprotein B (Apob) and fatty acid-binding protein 4 (Fabp4), both of which are crucial for lipid transport. Mechanistically, Rbm39 enhanced the transcription of Apob by upregulating hepatocyte nuclear factor 4α (Hnf4α), while it suppressed Fabp4 transcription by regulating alternative splicing of hypoxia inducible factor-1α (Hif-1α). These findings highlight the pivotal role of Rbm39 in maintaining hepatic lipid homeostasis and suggest its potential as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Chunbo Zhuang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Fangfang Cui
- Department of Gastroenterology, Kaifeng People's Hospital, Kaifeng, Henan 475000, PR China
| | - Jin Chen
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Dezhi He
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Ting Sun
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Pei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
18
|
Fan W, Yang S, Wei Y, Tian M, Liu Q, Li X, Ding J, Li X, Mao M, Han X, Du Y, Qiu C, Dong Y, Wang Y. Characterization of brain morphology associated with metabolic dysfunction-associated steatotic liver disease in the UK Biobank. Diabetes Obes Metab 2025; 27:3419-3430. [PMID: 40171859 DOI: 10.1111/dom.16362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Emerging evidence has linked metabolic dysfunction-associated steatotic liver disease (MASLD) with accelerated cognitive decline and dementia. We aimed to investigate the associations of MASLD with volumes of total brain tissue and subcortical grey matter, and white matter microstructures in the UK Biobank. METHODS This cross-sectional study included 29,195 individuals (aged 45-82 years) from the UK Biobank who undertook a magnetic resonance imaging (MRI) sub-study between 2014 and 2022. The brain MRI covers three modalities (T1, T2 FLAIR, and diffusion). Volumes of grey matter, subcortical grey matter structures, and regional cortex were derived from T1-weighted images. Fractional anisotropy (FA) and mean diffusivity (MD) were derived from diffusion tensor imaging (DTI) to assess global and tract-specific microstructure. MASLD was defined as the MRI-derived proton density fat fraction (MRI-PDFF) ≥5% and the presence of at least one cardiometabolic criterion. Data were analysed using multiple linear regression models. RESULTS MASLD was significantly associated with smaller volumes of total grey matter and subcortical grey matter (p < 0.05) and reduced Alzheimer's disease (AD)-signature cortical thickness (multivariable-adjusted β = -0.04; 95% confidence interval [CI]: -0.07, -0.01). Having MASLD was associated with higher total white matter hyperintensity (WMH) volume (multivariable-adjusted β = 0.12; 95% CI: 0.10, 0.15). For white matter microstructure, MASLD was associated with increased global FA (multivariable-adjusted β = 0.05; 95% CI: 0.03, 0.08) and reduced global MD (multivariable-adjusted β = -0.04; 95% CI: -0.07, -0.01). CONCLUSIONS Brain morphology associated with MASLD is characterized by smaller subcortical grey matter volume and higher coherence but lower magnitudes of white matter microstructure.
Collapse
Affiliation(s)
- Wenxiao Fan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Shuping Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Yiran Wei
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Minle Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Qianying Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaomeng Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Jiahao Ding
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xuewei Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Ming Mao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaolei Han
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Chengxuan Qiu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Yi Dong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yongxiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| |
Collapse
|
19
|
Zhao M, Liang C, Huang J, Dai L, Wang H, Zhang X, Zhao S, Guo C, Liu Z, Zhang T. Association between triglyceride-glucose related indices and severe infection requiring hospital admission or resulting in mortality among individuals with metabolic dysfunction-associated steatotic liver disease. Clin Nutr 2025; 49:1-10. [PMID: 40222088 DOI: 10.1016/j.clnu.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/28/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND AND AIMS Despite increasing evidence linking triglyceride-glucose (TyG) related indices with metabolic and cardiovascular outcomes, the associations with severe infection in individuals with metabolic dysfunction-associated steatotic liver disease (MASLD) remain unclear. METHODS We analyzed data from a large population-based cohort, which included participants who were diagnosed with MASLD and enrolled in 2016 and 2017, with follow-up until December 2022. Severe infections were defined as those requiring hospital admission or resulting in mortality. Multivariate Cox and restricted cubic spline (RCS) regression models were used to evaluate the associations between TyG-related indices and severe infection among participants with MASLD. Additionally, we examined these associations within subgroups defined by age, sex, hypertension, diabetes, obesity, and hyperlipidemia. RESULTS Among the 11,782 eligible participants with MASLD (mean age 57.02 years; 34.36 % male), a total of 898 (7.62 %) severe infections occurred during the median follow-up of 5.71 years. The multivariate Cox regression analyses revealed that high levels of TyG (HR = 1.175, 95%CI = 1.049-1.315), TyG-BMI (HR = 1.004, 95%CI = 1.001-1.006), and TyG-WC (HR = 1.002, 95%CI = 1.001-1.003) indices were significantly associated with the severe infection in patients with MASLD. The RCS curves showed positive linear correlations between three TyG-related indices with severe infection in MASLD. Subgroup analyses showed these associations were more pronounced among MASLD patients without obesity. Additionally, incorporating TyG-related indices into the basic model considerably improved the predictive ability for severe infection. CONCLUSIONS Our study indicated that a high TyG-related indices are associated with an increased risk of severe infection in MASLD patients. TyG-related indices would be the surrogate biomarkers for the follow-up of the MASLD population.
Collapse
Affiliation(s)
- Ming Zhao
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Chenyu Liang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Jiayi Huang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Luojia Dai
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Chengnan Guo
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China; Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China; Yiwu Research Institute, Fudan University, Yiwu, China.
| |
Collapse
|
20
|
Zhang X, Nguyen MH. Metabolic dysfunction-associated steatotic liver disease: A sexually dimorphic disease and breast and gynecological cancer. Metabolism 2025; 167:156190. [PMID: 40081614 DOI: 10.1016/j.metabol.2025.156190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become a global public health and economic burden worldwide in the past few decades. Epidemiological studies have shown that MASLD is a multisystem disease that is associated not only with liver-related complications but also with an increased risk of developing extrahepatic cancers. MASLD is a sexually dimorphic disease with sex hormones playing an important role in the development and progression of MASLD, especially by the levels and ratios of circulating estrogens and androgens. MASLD is associated with hormone-sensitive cancers including breast and gynecological cancer. The risk of breast and gynecological cancer is elevated in individuals with MASLD driven by shared metabolic risk factors including obesity and insulin resistance. Multiple potential mechanisms underline these associations including metabolic dysfunction, gut dysbiosis, chronic inflammation and dysregulated release of hepatokines. However, the effect of hormone therapy including hormone replacement therapy and anti-estrogen treatment on MASLD and female-specific cancers remains debatable at this time. This synopsis will review the associations between MASLD and breast and gynecological cancer, their underlying mechanisms, implications of hormonal therapies, and their future directions.
Collapse
Affiliation(s)
- Xinrong Zhang
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States; Department of Epidemiology and Population Health, Stanford University Medical Center, Palo Alto, CA, United States; Stanford Cancer Institute, Stanford University Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
21
|
Garcia-Morena D, Fernandez-Cantos MV, Escalera SL, Lok J, Iannone V, Cancellieri P, Maathuis W, Panagiotou G, Aranzamendi C, Aidy SE, Kolehmainen M, El-Nezami H, Wellejus A, Kuipers OP. In Vitro Influence of Specific Bacteroidales Strains on Gut and Liver Health Related to Metabolic Dysfunction-Associated Fatty Liver Disease. Probiotics Antimicrob Proteins 2025; 17:1498-1512. [PMID: 38319537 PMCID: PMC12055940 DOI: 10.1007/s12602-024-10219-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has become a major health risk and a serious worldwide issue. MAFLD typically arises from aberrant lipid metabolism, insulin resistance, oxidative stress, and inflammation. However, subjacent causes are multifactorial. The gut has been proposed as a major factor in health and disease, and over the last decade, bacterial strains with potentially beneficial effects on the host have been identified. In vitro cell models have been commonly used as an early step before in vivo drug assessment and can confer complementary advantages in gut and liver health research. In this study, several selected strains of the order Bacteroidales were used in a three-cell line in vitro analysis (HT-29, Caco-2, and HepG2 cell lines) to investigate their potential as new-generation probiotics and microbiota therapeutics. Antimicrobial activity, a potentially useful trait, was studied, and the results showed that Bacteroidales can be a source of either wide- or narrow-spectrum antimicrobials targeting other closely related strains. Moreover, Bacteroides sp. 4_1_36 induced a significant decrease in gut permeability, as evidenced by the high TEER values in the Caco-2 monolayer assay, as well as a reduction in free fatty acid accumulation and improved fatty acid clearance in a steatosis HepG2 model. These results suggest that Bacteroidales may spearhead the next generation of probiotics to prevent or diminish MAFLD.
Collapse
Affiliation(s)
- Diego Garcia-Morena
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Maria Victoria Fernandez-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Silvia Lopez Escalera
- Chr. Hansen A/S, Bøge Allé 10-12, 2970, Hørsholm, Denmark
- Friedrich-Schiller Universität Jena, Fakultät für Biowissenschaften, 18K, 07743, Bachstraβe, Germany
| | - Johnson Lok
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Valeria Iannone
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Pierluca Cancellieri
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Willem Maathuis
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745, Jena, Germany
- Department of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, 07745, Jena, Germany
| | - Carmen Aranzamendi
- Groningen Biomolecular Sciences and Biotechnology Institute, Host-Microbe Metabolic Interactions, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Sahar El Aidy
- Groningen Biomolecular Sciences and Biotechnology Institute, Host-Microbe Metabolic Interactions, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Marjukka Kolehmainen
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Hani El-Nezami
- Molecular and Cell Biology Division, School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Hong Kong SAR
| | - Anja Wellejus
- Chr. Hansen A/S, Bøge Allé 10-12, 2970, Hørsholm, Denmark
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
22
|
Si G, Lv X, Ge Y, Zhang R, Hao D, Chen X, Wang C, Li Y, Li X, Yuan X. Age at menarche and risk of non-alcoholic fatty liver disease: Insights from the NHANES 2017-2020 and Mendelian randomization analyses. Exp Gerontol 2025; 204:112748. [PMID: 40194671 DOI: 10.1016/j.exger.2025.112748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND This study aimed to explore the causal relationship between age at menarche (AAM) and non-alcoholic fatty liver disease (NAFLD), leveraging data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES) alongside Mendelian randomization (MR) analyses. Notably, this research represents the first attempt to link AAM to NAFLD using genetic methodologies, thereby providing novel insights into the interplay between these two conditions. METHODS Cross-sectional data from 2730 participants were analyzed using logistic regression to evaluate the association between AAM and NAFLD risk. A two-sample MR study was performed to investigate causal relationships, utilizing genetic data from large-scale genome-wide association studies (GWASs). The inverse variance weighted (IVW) method served as the primary MR analysis approach. RESULTS A significant negative association between AAM and NAFLD was found in Model 3 (OR = 0.85, 95 % CI: 0.74-0.97). Participants in the highest AAM quintile exhibited a 68 % reduction of NAFLD prevalence compared to those in the lowest AAM quintile (OR = 0.32, 95 % CI: 0.11-0.97). MR analysis confirmed a potential negative causal association (discovery: OR = 0.81, 95 % CI: 0.73-0.90; validation: OR = 0.80, 95 % CI: 0.66-0.96). CONCLUSIONS Our findings indicate a potential causal association between AAM and NAFLD, suggesting that early AAM may serve as a potential risk marker for NAFLD. This highlights the importance of incorporating AAM into clinical risk assessment tools and developing targeted prevention strategies for at-risk populations. Further research is needed to clarify the underlying mechanisms and to explore the potential benefits of early intervention.
Collapse
Affiliation(s)
- Guifei Si
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xiaopan Lv
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Yuxin Ge
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Rui Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Dongxiao Hao
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xuemei Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Changchun Wang
- Department of Internal Medicine, Feicheng Street Health Centre, Linyi 276000, Shandong, China
| | - Yuquan Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261000, Shandong, China; Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xiuping Li
- Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China
| | - Xuemin Yuan
- Department of Gastroenterology, Linyi People's Hospital Affiliated to Shandong Second Medical University, Linyi 276000, Shandong, China.
| |
Collapse
|
23
|
Kim HR, Tabiatnejad P, Arestakesyan H, Young CN. Modulation of liver lipid metabolic pathways by central nervous system ER stress. Am J Physiol Endocrinol Metab 2025; 328:E833-E844. [PMID: 40261717 DOI: 10.1152/ajpendo.00392.2024] [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: 10/08/2024] [Revised: 11/11/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), considered as the hepatic manifestation of metabolic syndrome, can increase the risk for cardiometabolic diseases. Accumulating reports have implicated the central nervous system in MASLD pathogenesis, specifically endoplasmic reticulum (ER) stress in subfornical organ (SFO) to hypothalamic paraventricular nucleus (PVN) projecting neurons (SFO→PVN). Here, we investigated how ER stress in this neural circuit influences hepatic lipid regulatory pathways that may contribute to MASLD development during obesity. Hepatic steatosis was elicited by feeding C57BL/6J male mice a high-fat diet for 11 wk. Intersectional viral targeting was used to inhibit ER stress in SFO→PVN neurons to examine the contribution of ER stress in this circuit to hepatic lipid acquisition and disposal genes during obesity. Inhibition of ER stress in SFO→PVN neurons of obese mice resulted in a reduction in hepatic triglycerides and lipid acquisition genes that was paralleled by a reduction in liver tyrosine hydroxylase, the rate-limiting enzyme in catecholamine synthesis. Moreover, hepatic tyrosine hydroxylase expression was positively correlated with lipid acquisition but not disposal pathways. These results indicate that ER stress in SFO→PVN neurons may contribute to MASLD through sympathetic nervous system influences, primarily on hepatic lipid acquisition.NEW & NOTEWORTHY Endoplasmic reticulum stress in SFO→PVN neurons modulates hepatic lipid acquisition and disposal pathways during obesity-induced hepatic steatosis. Hepatic tyrosine hydroxylase levels are positively correlated with liver triglyceride levels and lipid acquisition pathway-related genes in diet-induced obese animals.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Parisa Tabiatnejad
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Hovhannes Arestakesyan
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Colin N Young
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
24
|
Cao T, Ni X, Halengbieke A, Tang J, Han Y, Sun F, Gao B, Zheng D, Yan Y, Yang X. Effects of the triglyceride-glucose index on non-alcoholic fatty liver disease: Causal evidence from longitudinal cohort studies. Arch Gerontol Geriatr 2025; 133:105813. [PMID: 40073798 DOI: 10.1016/j.archger.2025.105813] [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: 12/16/2024] [Revised: 02/08/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Insulin resistance (IR) is strongly related to non-alcoholic fatty liver disease (NAFLD). Triglyceride-glucose (TyG) index serves as a novel substitute indicator for IR. However, research on the effect of TyG index on NAFLD remains sparse. This study aims to investigate the causal association between TyG index and incident NAFLD. METHODS The primary cohort consisted of 27,052 participants from the Beijing Health Management Cohort, while the external validation cohort included 75,023 participants from the Taiwan MJ Cohort. Entropy balancing for continuous treatments (EBCT) combined with logistic regression and targeted maximum likelihood estimation (TMLE) were used to evaluate the causal association between the TyG index and incident NAFLD. RESULTS During a median follow-up of 2.49 years in the primary cohort, 6,168 participants (median age: 36.0 years) developed incident NAFLD. EBCT combined with logistic regression revealed the odds ratio (95 % CI) of NAFLD risk was 1.742 (1.478-2.054) for each 1-unit increase in the baseline TyG index. In the TMLE model, the risk ratio (95 % CI) for NAFLD was 1.540 (1.406-1.687) in the Q4 (quartile 4) group compared with the Q1 group. These findings were consistent with those from the external validation cohort, reinforcing the robustness of the causal relationship between the TyG index and NAFLD incidence. CONCLUSIONS The advanced double-robust estimation method suggests that a higher baseline TyG index may be causally associated with an increased NAFLD risk, providing more reliable evidence for its role as a simple biomarker and demonstrating the utility of double-robust estimation causal inference models in epidemiology.
Collapse
Affiliation(s)
- Tengrui Cao
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Xuetong Ni
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Aheyeerke Halengbieke
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Jianmin Tang
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Yumei Han
- Science and Education Section, Beijing Physical Examination Center, No. 59, Beiwei Road, Xicheng District, Beijing 100050, China.
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China.
| | - Bo Gao
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Deqiang Zheng
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Yuxiang Yan
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Xinghua Yang
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| |
Collapse
|
25
|
Miryan M, Azizi A, Pasdar Y, Moradi M. Adherence to plant based diets reduce the risk of hepatic fibrosis in nonalcoholic fatty liver disease. Sci Rep 2025; 15:17403. [PMID: 40389596 DOI: 10.1038/s41598-025-02613-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 05/14/2025] [Indexed: 05/21/2025] Open
Abstract
Adherence to plant-based diets has significantly increased in popularity recently, with claims that they reduce the risk of non-communicable diseases. This study investigated whether high adherence to plant-based diets can reduce the risk of hepatic steatosis and fibrosis. In this study, 8516 participants from the Ravansar Noncommunicable Disease cohort completed a validated food frequency questionnaire (FFQ) to assess their plant-based diet scores. The study used the fatty liver index and fibrosis-4 index to predict hepatic steatosis and fibrosis. The plant-based diet index (PDI) was used to measure the overall quality of diets from healthy and unhealthy plant-derived foods and animal-derived foods. Associations were determined using binary logistic regression, considering potential confounders. Participants in the highest tertiles of plant-based diet scores had higher energy-adjusted intakes of fructose than those in the lowest tertiles (16.09 ± 12.11 vs. 26.65 ± 12; P-value < 0.001). In multivariable-adjusted models, participants in the highest tertile of PDI had lower odds of hepatic fibrosis than those in the lowest tertile (OR: 0.59; 95%CI: 0.43-0.81). There was no significant association between adherence to PDI and hepatic steatosis after adjustment for potential confounders (OR: 0.989; 95%CI 0.78 - 1.25). The odds of hepatic fibrosis decreased by 6% for each unit increase in healthy plant-based foods (OR: 0.94; 95%CI: 0.91-0.97). The odds of hepatic steatosis increased by 14% for each 1 SD increase in fructose intake (OR: 1.14; 95% CI: 1.02-1.28). This study highlights the potential benefits of high adherence to plant-based diets in reducing the risk of hepatic fibrosis, but high fructose content in some plant-based foods may have an unfavorable role in hepatic steatosis. These findings highlight the importance of selecting whole, fiber-rich plant foods and minimizing intake of fructose-dense products in plant-based diets to promote liver health. Therefore, selecting low-fructose food items in plant-based diets is recommended, though further research is needed to confirm these findings.
Collapse
Affiliation(s)
- Mahsa Miryan
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Azizi
- Social Development and Health Promotion Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Community and Family Medicine, School of Medicine, Kermanshah University of Medical Sciences, P.O.BOX: 1568, Kermanshah, Iran.
| | - Yahya Pasdar
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojgan Moradi
- Internal Medicine Department, School of Medicine, Imam Khomeini Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
26
|
Marengo M, Briet C, Munier M, Boursier J, Rodien P, Suteau V. Fatty Liver Disease Along Cushing Syndrome Evolution. J Clin Endocrinol Metab 2025; 110:e2037-e2044. [PMID: 39193719 DOI: 10.1210/clinem/dgae568] [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: 02/20/2024] [Revised: 06/09/2024] [Accepted: 08/27/2024] [Indexed: 08/29/2024]
Abstract
CONTEXT The clinical manifestations of Cushing syndrome are variable, but an important number of patients present a metabolic syndrome, strongly associated with hepatic steatosis. OBJECTIVE The aim of this study was to determine the prevalence of metabolic dysfunction associated steatotic liver disease (MASLD) at the diagnosis of Cushing syndrome. METHODS We conducted a single-center retrospective study at Angers Hospital (France) between 2010 and 2020. Forty-nine patients followed for Cushing syndrome with available abdominal imaging at diagnosis were included. A mean liver/spleen density ratio < 1 on computed tomography was diagnostic of hepatic steatosis. Simple clinico-biological scores predictive of hepatic fibrosis (FIB-4, NAFLD Fibrosis Score, and eLIFT) were calculated for patients with hepatic steatosis. RESULTS Of the 49 patients, 13 (26.5%) had hepatic steatosis at diagnosis of Cushing syndrome. All 13 had MASLD. These patients had a higher prevalence of type 2 diabetes and higher triglyceride levels in multivariate analysis. There was no difference according to the intensity or duration of Cushing syndrome. Among the 13 patients with MASLD, 2 (15.4%) had a significant fibrosis predictive score. Of the 4 patients with follow-up imaging after remission of Cushing syndrome, 3 had remission of steatosis between 1 and 5 years after remission of Cushing syndrome. No patient without MASLD at diagnosis had a worsening liver/spleen ratio after remission. CONCLUSION We estimated the prevalence of hepatic steatosis at the diagnosis of Cushing syndrome at 26.5%. The presence of metabolic factors was associated with the occurrence of hepatic steatosis.
Collapse
Affiliation(s)
- Maria Marengo
- Department of Endocrinology, Diabetology and Nutrition, Angers University Hospital, 49100 Angers, France
| | - Claire Briet
- Department of Endocrinology, Diabetology and Nutrition, Angers University Hospital, 49100 Angers, France
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, 49100 Angers, France
| | - Mathilde Munier
- Department of Endocrinology, Diabetology and Nutrition, Angers University Hospital, 49100 Angers, France
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, 49100 Angers, France
- Centre de Référence des Maladies Rares de la Thyroïde et des Récepteurs Hormonaux, University Hospital Angers, 49100 Angers, France
| | - Jérôme Boursier
- Department of Hepato-Gastroenterology, Angers University Hospital, 49100 Angers, France
- HIFIH Laboratory, Angers University, UPRES EA3859, SFR 4208, 49100 Angers, France
| | - Patrice Rodien
- Department of Endocrinology, Diabetology and Nutrition, Angers University Hospital, 49100 Angers, France
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, 49100 Angers, France
- Centre de Référence des Maladies Rares de la Thyroïde et des Récepteurs Hormonaux, University Hospital Angers, 49100 Angers, France
| | - Valentine Suteau
- Department of Endocrinology, Diabetology and Nutrition, Angers University Hospital, 49100 Angers, France
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, 49100 Angers, France
| |
Collapse
|
27
|
Koliaki C, Dalamaga M, Kakounis K, Liatis S. Metabolically Healthy Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): Navigating the Controversies in Disease Development and Progression. Curr Obes Rep 2025; 14:46. [PMID: 40387999 DOI: 10.1007/s13679-025-00637-9] [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: 05/05/2025] [Indexed: 05/20/2025]
Abstract
PURPOSE OF REVIEW The natural course of metabolic dysfunction-associated steatotic liver disease (MASLD) in the population with metabolically healthy obesity (MHO) has not been adequately explored. In the present narrative review, we summarize the evidence regarding the association between MHO and MASLD prevalence, incidence and progression. RECENT FINDINGS Cross-sectional, population-based, cohort studies have shown an increased prevalence of hepatic steatosis and fibrosis in subjects with MHO compared with metabolically healthy non-obese individuals (MHNO). In large-scale longitudinal cohort studies among metabolically healthy subjects, increasing body mass index (BMI) has been found to be independently associated with an increased incidence of MASLD and progressive hepatic fibrosis over a mean follow-up period of 2.2-7.7 years. With regard to advanced MASLD, the prevalence of steatohepatitis and clinically significant liver fibrosis is lower in MHO compared with subjects with metabolically unhealthy obesity (MUO). The presence of MASLD has been proposed as a strong risk factor for metabolic health deterioration in MHO. Furthermore, subjects with MHO and MASLD display an elevated 10-year cardiovascular risk and a three-fold increased risk of incident diabetes compared with MHO without MASLD. MASLD may also predict the failure to convert from MUO to MHO after a weight loss intervention.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Propaedeutic Department of Internal Medicine and Diabetes Center, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, Athens, 11527, Greece.
| | - Maria Dalamaga
- Department of Biologic Chemistry, Medical School, National Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Kakounis
- Department of Gastroenterology, Hippokration General Hospital of Athens, Athens, Greece
| | - Stavros Liatis
- First Propaedeutic Department of Internal Medicine and Diabetes Center, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, Athens, 11527, Greece
| |
Collapse
|
28
|
Mohan V, Joshi S, Kant S, Shaikh A, Sreenivasa Murthy L, Saboo B, Singh P, Sosale AR, Sanyal D, Shanmugasundar G, Singh SK, Pancholia AK, Mondal S, George R, Jaiswal A, Jhaveri K. Prevalence of Metabolic Dysfunction-Associated Steatotic Liver Disease: Mapping Across Different Indian Populations (MAP Study). Diabetes Ther 2025:10.1007/s13300-025-01748-1. [PMID: 40381173 DOI: 10.1007/s13300-025-01748-1] [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: 03/13/2025] [Accepted: 04/23/2025] [Indexed: 05/19/2025] Open
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global health concern. MASLD is strongly linked to obesity, sedentary lifestyles, and metabolic syndrome. In India, the prevalence of MASLD exhibits regional variations because of genetic, dietary, and socioeconomic factors. Liver stiffness measurement (LSM) and controlled attenuation parameter (CAP) are standardized non-invasive tools for assessing fibrosis and steatosis in MASLD. This study aimed to determine the prevalence and severity of MASLD across different Indian regions and examine regional disparities in MASLD burden. METHODS This retrospective, multicenter, cross-sectional study analyzed the data of 13,750 adults from 105 diabetes and endocrine clinics across six geographic zones in India between May 2023 and February 2024. Participants underwent LSM and CAP assessment using FibroScan™. Based on LSM, liver fibrosis was categorized as F0-F1 (2-7 kPa), F2 (7-10 kPa), F3 (10-14 kPa), and F4 (≥ 14 kPa). Based on CAP, MASLD was classified as mild (238-260 dB/m), moderate (261-292 dB/m), and severe (≥ 293 dB/m). RESULTS The prevalence of MASLD was 68.2% (CAP ≥ 238 dB/m), and the prevalence of fibrosis was 33.7% (LSM ≥ 7 kPa). The highest MASLD burden was observed in North India (73.3%), particularly in Uttarakhand (80.0%). Severe fibrosis (F4) was highly prevalent in Kerala (20.0%), whereas severe steatosis (S3) was highly prevalent in Jammu and Kashmir (50.3%). The prevalence of MASLD was significantly associated with regional variations (P < 0.001) but not with age. CONCLUSION This study highlights significant regional disparities in MASLD burden. The high burden in North India calls for region-specific public health interventions, standardized screening, and preventive strategies.
Collapse
Affiliation(s)
- Viswanathan Mohan
- Madras Diabetes Research Foundation (ICMR-Collaborating Centre of Excellence) & Dr. Mohan's Diabetes Specialities Centre (IDF Centre of Excellence in Diabetes Care), No 4, Conran Smith Road, Gopalapuram, Chennai, Tamil Nadu, 600086, India.
| | - Shashank Joshi
- Department of Diabetology and Endocrinology, Lilavati Hospital and Research Centre, Mumbai, 400050, India
| | - Saket Kant
- Department of Endocrinology, Ananda Medicare, Sector-9, Rohini, New Delhi, 110085, India
| | - Altamash Shaikh
- Department of Endocrinology, AMS Neolife Superspeciality Clinic, Mumbai, 400008, Maharashtra, India
| | - L Sreenivasa Murthy
- Department of Diabetology, Lifecare Hospital and Research Centre, Bangalore, Karnataka, 560092, India
| | - Banshi Saboo
- Department of Diabetology, Dia Care Hormone Clinic, Ahmedabad, Gujarat, 380015, India
| | - Parminder Singh
- Department of Endocrinology, Dr. Parminder's Diabetes & Endocrine Centre, Ludhiana, Punjab, 141001, India
| | - Aravind R Sosale
- Department of Diabetology, Diacon Hospital, Bangalore, Karnataka, 560010, India
| | - Debmalya Sanyal
- Endocrinology, KPC Medical College and NH-RTIICS, Kolkata, West Bengal, 700032, India
| | - G Shanmugasundar
- Department of Endocrinology, Magna Centres for Obesity & Endocrinology, Chennai, Tamil Nadu, 600083, India
| | - Santosh Kumar Singh
- Consultant Endocrinologist, Department of Endocrinology, Udayman Appartment, Patna, Bihar, 800001, India
| | - A K Pancholia
- Department of Medicine and Preventive Cardiology, Arihant Hospital and Research Centre, Indore, India
| | - Sunetra Mondal
- Department of Endocrinology, "Morning Glory" Polyclinic, Burdwan, West Bengal, 713104, India
| | - Rishi George
- Department of Diabetology, Welmont Hospital, Ernakulum, Kerala, 682017, India
| | - Ashok Jaiswal
- Department of Medical Affairs, Zydus Healthcare Limited, Mumbai, Maharashtra, 400063, India
| | - Kunal Jhaveri
- Department of Medical Affairs, Zydus Healthcare Limited, Mumbai, Maharashtra, 400063, India
| |
Collapse
|
29
|
Zhang J, Wang S, Zhang T, Zi M, Wang S, Zhang Q. Green tea epigallocatechin gallate attenuate metabolic dysfunction-associated steatotic liver disease by regulation of pyroptosis. Lipids Health Dis 2025; 24:180. [PMID: 40382644 DOI: 10.1186/s12944-025-02596-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the most common chronic liver disease, with other fat-liver diseases potentially progressing to cirrhosis and hepatocellular carcinoma. Our study aimed to alleviate MASLD by EGCG inhibiting oxidative stress-mediated pyroptosis in zebrafish. METHODS The one month old wild-type zebrafh larval (50 per group) and three months old adult male zebrafish (15 per group) were treated with high-fat diet (HFD) feeding (powdered egg yolk) following by treatment with 25 μM EGCG for 15 days. Indicators related to liver damage, oxidative stress, inflammation, pyroptosis and aging were assessed using Oil Red O staining, H&E staining, commercial assay kits, enzyme-linked immunosorbent assay (ELISA) kits, and Western blot analysis. RESULTS The results suggest that EGCG significantly reduced fatness, severe lipid deposition, triglyceride (TG), total cholesterol (TC) and free fatty acid (FFA) levels (p < 0.05). EGCG markedly reduced serum ALT, AST and ameliorated liver injury in zebrafish (p < 0.05). EGCG also showed an antioxidant effect by reducing reactive oxygen species (ROS) production malondialdehyde (MDA) and increasing superoxide dismutase (SOD) levels (p < 0.05). Moreover, EGCG obviously down-regulated the pro-inflammatory factors like tumor necrosis factor-a (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-18 (IL-18) levels (p < 0.05). EGCG indicated a significant upregulation involved in pyroptosis pathway, such as nuclear factor erythroid 2-related factor 2 (NRF2) and downregulated the expressions of nuclear factor-κB (NF-κB), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), cysteinyl aspartate specific proteinase-1 (caspase-1), as well as gasdermin D (GSDMD) (p < 0.01). Moreover, EGCG significantly improved aging-related markers induced by a HFD, including the level of senescence-associated β-Galactosidase (SA β-Gal) and expression of p53, p16, and p21 (p < 0.05), while ameliorating liver function in zebrafish. CONCLUSIONS These results suggest that EGCG may attenuate MASLD in larval and adult zebrafish induced by 15 consecutive days HFD, which is potentially mediated by modulating the Nrf2/NF-κB/NLRP3 signaling pathway which relieve pyroptosis.
Collapse
Affiliation(s)
- Jie Zhang
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shuangshuang Wang
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Ting Zhang
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Minghui Zi
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shuxiang Wang
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Qiao Zhang
- School of Public Health, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
30
|
Zhu L, Liu S, Wang Z, Yang Y, Han P, Tong W, Zhao T, Wang L, Cui T, Yang L, Zhang Y. Modeling hepatic steatosis with human adult stem cell-derived liver organoids. iScience 2025; 28:112344. [PMID: 40276762 PMCID: PMC12019286 DOI: 10.1016/j.isci.2025.112344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 11/05/2024] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) remains the most common chronic liver disease worldwide, and appropriate in vitro models are of great significance for investigating pathogenesis and drug screening of MASLD. In this study, human expandable cholangiocyte organoids were derived from adult stem cells of normal liver tissue. After differentiation, liver organoids (LOs) exhibited the functional characteristics and genomic features of mature hepatocytes. To induce steatosis, LOs were incubated with a gradient concentration oleic acid, and it was found that the model could recapitulate the development of lipid accumulation and inflammation. In addition, the drug sensitivity of the hepatic steatosis model was further verified through anti-steatosis drug testing. In summary, LOs have great potential for disease modeling, and the results indicate that the hepatic steatosis model may serve as a useful tool for exploring the molecular mechanisms and drug screening of MASLD.
Collapse
Affiliation(s)
- Liuyang Zhu
- First Central Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Sen Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ze Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Yueyue Yang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Pinsheng Han
- Nankai University of Medicine College, Tianjin 300071, China
| | - Wen Tong
- First Central Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Tianyu Zhao
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Libo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Tao Cui
- Tianhui Biotechnology Co., Ltd., Hefei 230000, China
| | - Long Yang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| |
Collapse
|
31
|
Zerehpooshnesfchi S, Safri F, Pan Z, Nguyen R, Yuen L, Lam V, Nahm C, Pang T, Ahlenstiel G, George J, Eslam M, Qiao L. Characterisation of non-cirrhotic MAFLD-related hepatocellular carcinoma: a retrospective cohort study. Ther Adv Chronic Dis 2025; 16:20406223251339402. [PMID: 40385594 PMCID: PMC12084690 DOI: 10.1177/20406223251339402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/16/2025] [Indexed: 05/20/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major global health issue, in which the underlying liver disease aetiology has shifted towards non-viral causes, particularly metabolic (dysfunction)-associated fatty liver disease (MAFLD). While traditionally associated with cirrhosis, a subset of HCC cases arises in patients with MAFLD but without cirrhosis, whose characteristics remain poorly understood. Objectives The study aims to explore the clinical, tumour and genetic characteristics of non-cirrhotic MAFLD-related HCC when compared to those that develop in the context of cirrhosis. Design A multi-centre, retrospective study of 89 MAFLD-related HCC patients enrolled between 2009 and 2023 was performed. Methods We conducted a study of well-defined MAFLD-related HCC patients to explore their MAFLD-related clinical and genetic associations. Statistical analysis was undertaken to compare the underlying cirrhosis and non-cirrhosis groups for HCC features, adjusting for relevant confounders. Results Patients with HCC arising in cases of MAFLD without cirrhosis exhibited a lower body mass index, higher triglyceride levels and increased smoking prevalence compared to their counterparts with cirrhosis. Despite arising in the absence of cirrhosis, these patients had more aggressive tumour features, including larger tumour size, multifocality and portal vein thrombosis. Logistic regression confirmed non-cirrhosis status to be an independent predictor of larger tumour size and increased lesion number. Conclusion Non-cirrhotic MAFLD-related HCC presents with distinct clinical and tumour characteristics, suggesting the existence of unique disease drivers that are yet to be discovered.
Collapse
Affiliation(s)
| | - Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Ziyan Pan
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Lawrence Yuen
- Department of Upper GI and HPB Surgery, Faculty of Medicine and Health Sciences, The University of Sydney, Westmead Hospital, Westmead, NSW, Australia
| | - Vincent Lam
- Department of Upper GI and HPB Surgery, Faculty of Medicine and Health Sciences, The University of Sydney, Westmead Hospital, Westmead, NSW, Australia
| | - Christopher Nahm
- Department of Upper GI and HPB Surgery, Faculty of Medicine and Health Sciences, The University of Sydney, Westmead Hospital, Westmead, NSW, Australia
| | - Tony Pang
- Department of Upper GI and HPB Surgery, Faculty of Medicine and Health Sciences, The University of Sydney, Westmead Hospital, Westmead, NSW, Australia
| | - Golo Ahlenstiel
- Department of Gastroenterology and Hepatology, Blacktown Hospital, Blacktown, NSW, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health Sciences, The University of Sydney, Westmead, NSW, Australia
| | - Mohammed Eslam
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
| |
Collapse
|
32
|
Munk Lauridsen M, Ravnskjaer K, Gluud LL, Sanyal AJ. Disease classification, diagnostic challenges, and evolving clinical trial design in MASLD. J Clin Invest 2025; 135:e189953. [PMID: 40371650 PMCID: PMC12077896 DOI: 10.1172/jci189953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) diagnosis and management have evolved rapidly alongside the increasing prevalence of obesity and related complications. Hepatology has expanded its focus beyond late-stage cirrhosis and portal hypertension to earlier, complex MASLD cases in younger patients, necessitating closer collaboration with endocrinology. The renaming of nonalcoholic fatty liver disease (NAFLD) to MASLD reflects its pathophysiology, reduces stigma, and has prompted new research directions. Noninvasive tests such as liver stiffness measurement now play a crucial role in diagnosis, reducing reliance on invasive liver biopsies. However, advanced omics technologies, despite their potential to enhance diagnostic precision and patient stratification, remain underutilized in routine clinical practice. Behavioral factors, including posttraumatic stress disorder (PTSD) and lifestyle choices, influence disease outcomes and must be integrated into patient management strategies. Primary care settings are critical for early screening to prevent progression to advanced disease, yet sizable challenges remain in implementing effective screening protocols. This Review explores these evolving aspects of MASLD diagnosis and management, emphasizing the need for improved diagnostic tools, multidisciplinary collaboration, and holistic care approaches to address existing gaps and ensure comprehensive patient care across all healthcare levels.
Collapse
Affiliation(s)
- Mette Munk Lauridsen
- Stravitz-Sanyal Liver Institute, Department of Gastroenterology & Hepatology, Virginia Commonwealth University Medical Clinic, Richmond, Virginia, USA
- University Hospital of Southern Denmark, Liver Research Group, Department of Gastroenterology and Hepatology, Esbjerg, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital, Hvidovre, Denmark, and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arun J. Sanyal
- Stravitz-Sanyal Liver Institute, Department of Gastroenterology & Hepatology, Virginia Commonwealth University Medical Clinic, Richmond, Virginia, USA
| |
Collapse
|
33
|
Messer EK, Petroff D, Schattenberg J, Zeuzem S, von der Ohe M, Ludwig L, Demir M, Buggisch P, Stein K, Serfert Y, Wedemeyer H, Berg T, Hofmann WP, Geier A, Wiegand J. Evaluating Resmetirom Eligibility Among Patients with MASH: Insights from the German Steatotic Liver Disease-Registry. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025. [PMID: 40373810 DOI: 10.1055/a-2592-6109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
The THR-β agonist resmetirom is the first treatment approved for metabolic dysfunction-associated steatohepatitis (MASH) in the US so far. It can be prescribed given MASH and F2/F3-fibrosis ("at-risk MASH").We analyzed how many patients qualify for resmetirom in a recently recruited Steatotic Liver Disease-cohort involving both tertiary and secondary care centers, the German SLD-Registry.Indication for resmetirom was assessed by three different approaches: (i) biopsy-proven MASH with F2/3 fibrosis and NAS-score ≥ 4; (ii) FibroScan-AST (FAST) score ≥ 0.67 and vibration controlled transient elastography (VCTE) < 15 kPa; (iii) US expert recommendations with VCTE 10-15 kPa and platelets ≥ 140×109/L or VCTE 8-15 kPa.1113 patients were recruited across 8 tertiary and 12 secondary care centers. NAS grading and staging were available for 180 cases (16%) with 179/180 conducted at tertiary care level. Of these, 61 (34%) qualified for resmetirom. FAST score without histologic assessment was available for 638 cases (57.3%), of which 612 (87%) were from tertiary and 26 (11%) from secondary care centers. Based on approach (ii), 41 (6%) of these individuals qualified for resmetirom compared to 117 (18.3%) using approach (iii). Combining approach (iii) with FAST ≥ 0.67 leads to 191 (30.0%) eligible patients. Using VCTE 8-15 kPa results in 182 (28.5%) eligible patients.Eligibility for resmetirom treatment depends on the available method used to identify "at-risk MASH". Availability of VCTE was highest among different levels of care.
Collapse
Affiliation(s)
- Eva Katharina Messer
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - David Petroff
- Clinical Trial Centre Leipzig, Leipzig University, Leipzig, Germany, Leipzig, Germany
| | - Jörn Schattenberg
- Department of Internal Medicine II, Saarland University Medical Center, Homburg, Germany
| | - Stefan Zeuzem
- Medical Clinic I, Johann Wolfgang Goethe University Hospital, Frankfurt am Main, Germany
| | | | | | - Münevver Demir
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Buggisch
- ifi-Institute for Interdisciplinary Medicine, Hamburg, Germany
| | | | | | - Heiner Wedemeyer
- Leberstiftungs-GmbH Deutschland, Hannover, Germany
- Dept. of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Wolf P Hofmann
- Leberstiftungs-GmbH Deutschland, Hannover, Germany
- Gastroenterologie am Bayerischen Platz, Berlin, Germany
| | - Andreas Geier
- Division of Infectious Diseases, Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Johannes Wiegand
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| |
Collapse
|
34
|
Lv C, Shi K, Guo Y, Guo Z, Luo P, Wang L, Wu Z, Yu P. Emerging Roles of Periodontal Pathogen-Derived Outer Membrane Vesicles in NAFLD. Int Dent J 2025; 75:100825. [PMID: 40378508 DOI: 10.1016/j.identj.2025.03.029] [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: 02/28/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 05/19/2025] Open
Abstract
The rising incidence of nonalcoholic fatty liver disease (NAFLD) poses a great socioeconomic burden worldwide. Also, periodontitis is the most common chronic inflammatory disease caused by a group of oral pathogens, affecting both oral health and systemic conditions, especially liver disease. Although accumulating evidence has elucidated an association between periodontal pathogens and NAFLD, the role of periodontal pathogen-derived outer membrane vesicles (OMVs) has not yet been clarified. In this comprehensive review, we aim to address this gap by summarising the progression and pathogenesis of NAFLD and revealing the relationship between periodontal disease and NAFLD multidimensionally. Additionally, this review sheds light on the multifunctional roles of periodontal pathogens OMVs and emphasises that periodontal pathogen-derived OMVs promote the development of NAFLD by stimulating Kupffer cells to produce inflammatory factors and inducing the activation of Hepatic stellate cells. However, it is still controversial whether periodontal pathogen-derived OMVs can be transferred to the liver through the bloodstream route or the oral-gut-liver axis. This highlights the pressing need for continued research efforts to develop new and optimised research schemes to observe the formation of the systemic distribution pathway of periodontal pathogen-derived OMVs. Finally, it is notable that there are currently no relevant clinical treatment guidelines to make specific provisions on controlling the level of periodontal pathogen-derived OMVs in patients with NAFLD. Guidelines developed based on our findings may contribute to the standardisation of practices. It can also provide effective strategies and potential therapeutic targets for NAFLD patients with periodontitis to alleviate the development of NAFLD diseases by inhibiting periodontal pathogens OMVs.
Collapse
Affiliation(s)
- Congcong Lv
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Kaikai Shi
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yadong Guo
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Zixin Guo
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Pingchan Luo
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Lijing Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Zhe Wu
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China.
| | - Pei Yu
- Department of Prosthodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction and Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
35
|
Zhang R, Zhang Q, Cui Z, Huang B, Wang Y, Ma H. Dimethyl fumarate improves non-alcoholic fatty liver disease by regulating SIRT1 signal to inhibit MAMs over-enrichment. Eur J Pharmacol 2025; 1000:177693. [PMID: 40340013 DOI: 10.1016/j.ejphar.2025.177693] [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: 01/02/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex metabolic disorder of the liver with an increasing global prevalence. Despite significant advancements in understanding NAFLD, effective therapeutic strategies remain limited. Mitochondria-associated membranes (MAMs) are specialized domains of the endoplasmic reticulum (ER) that closely interact with mitochondria and play a crucial role in regulating Ca2+ homeostasis. Our previous research demonstrated that dimethyl fumarate (DMF) alleviates NAFLD by modulating hepatic Ca2+ homeostasis. However, the precise mechanisms remain unclear. In this study, we found that DMF significantly alleviated lipid accumulation in NAFLD mice by inhibiting excessive MAMs enrichment. Mechanistically, DMF improved mitochondrial function by reducing mitochondrial Ca2+ overload and oxidative stress caused by MAMs over-enrichment. Furthermore, our results demonstrated that protective effects of DMF against NAFLD are dependent on Sirtuin-1 (SIRT1) regulation. Inhibition of SIRT1 markedly reversed the ability of DMF to suppress MAMs over-enrichment in both in vitro and in vivo models. Moreover, the beneficial effects of DMF on oxidative stress, mitochondrial dysfunction, and hepatic steatosis were abrogated by co-administration of EX527, a selective SIRT1 inhibitor. In summary, our findings demonstrate that DMF alleviates mitochondrial Ca2+ dysregulation caused by aberrant MAMs enrichment, thereby reducing oxidative stress and mitochondrial dysfunction, ultimately inhibiting lipid accumulation in hepatocytes. These results provide new insights into the therapeutic potential of DMF for NAFLD treatment.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Quanwei Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - ZiYi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - BenZeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yulei Wang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
36
|
Tian C, Deng S, Zhang Z, Zheng K, Wei L. Bifidobacterium bifidum 1007478 derived indole-3-lactic acid alleviates NASH via an aromatic hydrocarbon receptor-dependent pathway in zebrafish. Life Sci 2025; 369:123557. [PMID: 40074143 DOI: 10.1016/j.lfs.2025.123557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
AIMS This study investigates the potential of Bifidobacterium bifidum 1007478 (BB478) and its metabolite indole-3-lactic acid (ILA) in alleviating non-alcoholic steatohepatitis (NASH) induced by a high-fat diet (HFD) and fructose exposure. MATERIALS AND METHODS A zebrafish model of NASH was established by exposure to HFD and fructose. BB478 was administered, and the effects on liver lipid accumulation, oxidative stress, and inflammation were assessed. ILA production by BB478 was confirmed, and its impact on hepatic lipogenesis and inflammatory pathways was evaluated. The involvement of the aromatic hydrocarbon receptor (AhR) was also examined using an AhR inhibitor. KEY FINDINGS BB478 supplementation inhibited lipid accumulation in the liver, reduced triglycerides (TG) and total cholesterol (TC), and mitigated oxidative stress, as evidenced by lower levels of reactive oxygen species (ROS) and malondialdehyde (MDA). ILA, produced by BB478, could alleviate the hepatic damage and fat deposition in liver. Mechanistically, it suppressed hepatic lipogenesis by downregulating lipogenesis-related genes, including sterol response element binding protein 1 (SREBP1) and fatty acid synthase (FASN). ILA also inhibited the expression of pro-inflammatory cytokines to suppress inflammation. The therapeutic effects of ILA were reversed by the AhR inhibitor, indicating that ILA's actions are AhR-dependent. SIGNIFICANCE These findings reveal the potential of ILA, produced by Bifidobacterium bifidum, as a therapeutic agent for NASH. The mechanistic insights into AhR-mediated effects provide a foundation for further exploration of ILA as a novel approach for managing liver diseases.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China
| | - Zhao Zhang
- Research and Development Centre, GuangDong Longseek Testing Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Kangdi Zheng
- Research and Development Centre, GuangDong Longseek Testing Co., Ltd., Guangzhou, Guangdong 510700, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
37
|
Gogola T, Pitkänen S, Huovinen M, Laitinen H, Küblbeck J. Association between phthalate exposure and metabolic dysfunction-associated steatotic liver disease (MASLD) - Systematic literature review. ENVIRONMENTAL RESEARCH 2025; 273:121186. [PMID: 39986424 DOI: 10.1016/j.envres.2025.121186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rising globally. Recent studies have suggested connections between exposure to endocrine disrupting chemicals (EDCs) and the development of MASLD. Phthalates, which are commonly utilized as plasticizers, in building materials and consumer items, exhibit endocrine disrupting effects and have been shown to interfere with lipid metabolism in mechanistic studies. The objective of this systematic review was to examine the association between MASLD and exposure to phthalates in the adult human populations. We searched PubMed, Scopus and Web of Science for studies published from the inception of each database until December 12, 2024. The literature search yielded 10 cross-sectional studies, which were analyzed in detail. The key findings of this study indicate a potential correlation between the prevalence of MASLD and exposure to certain phthalates. Among the phthalates examined, the metabolites of bis(2-ethylhexyl) phthalate (DEHP) - namely MECPP, MEHHP, and MEOHP, demonstrated the strongest and most frequent associations with MASLD. All the current studies followed cross-sectional study designs, which limits the possibility to establish a causal relationship between MASLD and phthalate exposure. Therefore, longitudinal studies are needed to corroborate these findings and shed light on the involvement of phthalate exposure in MASLD.
Collapse
Affiliation(s)
- Tomasz Gogola
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Sini Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland.
| | - Marjo Huovinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | | | - Jenni Küblbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| |
Collapse
|
38
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [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: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
39
|
Lv J, Yao J, Zheng L, Shao J, Jin J, Zheng Q, Teng J, Zhou J, Cai F, Huang P, Jiang X. Association between atherogenic index of plasma and all-cause mortality and cardiovascular disease among individuals with non-alcoholic fatty liver disease or metabolic dysfunction-associated steatotic liver disease. Diabetol Metab Syndr 2025; 17:153. [PMID: 40369673 PMCID: PMC12076888 DOI: 10.1186/s13098-025-01715-7] [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: 01/08/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) and metabolic dysfunction-associated steatotic liver disease (MASLD) are common chronic liver diseases worldwide, both of which are closely associated with an increased risk of cardiovascular disease (CVD). Atherogenic index of plasma (AIP), as a biomarker of dyslipidemia, may predict CVD risk and mortality in these patients, but its specific role in patients with NAFLD and MASLD has not been studied in detail. This study adopted a cohort design, using data from the National Health and Nutrition Examination Survey (NHANES, 1988-1994) database, which was conducted by the Centers for Disease Control and Prevention. A total of 12,929 adult participants were analyzed. After participants were grouped according to AIP quartiles, the relationship between AIP levels and CVD risk was explored using multivariate logistic regression models and restricted cubic splines. The relationship between AIP levels and all-cause and CVD mortality was analyzed using multivariate Cox regression models. RESULTS Participants with the higher AIP quartiles showed high rates of CVD among participants with NAFLD (Quartile 1: 7.57%; Quartile 2: 10.00%; Quartile 3: 11.63%; Quartile 4: 15.08%). Participants with the higher AIP quartiles showed high rates of CVD among participants with MASLD (Quartile 1: 9.71%; Quartile 2: 11.30%; Quartile 3: 11.14%; Quartile 4: 15.00%). The findings suggested a linear association between the AIP index and the risk of CVD in participants with NAFLD or MASLD. AIP was significantly associated with CVD in the highest quartile of NAFLD or MASLD patients, and the adjusted hazard ratio indicated that high AIP levels were associated with high risk of CVD among participants with NAFLD (HR: 1.77, 95% CI: 1.24, 2.52) and MASLD (HR 1.76, 95% CI: 1.04, 2.98). In addition, higher AIP levels were also associated with increased all-cause mortality and CVD mortality among participants with NAFLD or MASLD. CONCLUSION This study showed that AIP is an effective tool for predicting CVD risk and mortality in patients with NAFLD and MASLD. Regular monitoring of AIP levels can help identify high-risk patients early and provide clinical risk assessment before intervention, thereby improving patient management and prognosis. Future studies need to further explore the role of AIP in different ethnic and economic conditions to optimize cardiovascular disease prevention and treatment strategies.
Collapse
Affiliation(s)
- Jialu Lv
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Jiangnan Yao
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Leyi Zheng
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Jiaxin Shao
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Jijie Jin
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Qianrong Zheng
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Jiayi Teng
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China
| | - Jianghua Zhou
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Fuman Cai
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China.
| | - Pan Huang
- College of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang province, 325000, China.
| | - Xuepei Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China.
| |
Collapse
|
40
|
Chi KY, Kim G, Son JS, Han J, Kim JH. Recent Advances in Three-Dimensional In Vitro Models for Studies of Liver Fibrosis. Tissue Eng Regen Med 2025:10.1007/s13770-025-00719-8. [PMID: 40358834 DOI: 10.1007/s13770-025-00719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Liver fibrosis is a reversible but complex pathological condition associated with chronic liver diseases, affecting over 1.5 billion people worldwide. It is characterized by excessive extracellular matrix deposition resulting from sustained liver injury, often advancing to cirrhosis and cancer. As its progression involves various cell types and pathogenic factors, understanding the intricate mechanisms is essential for the development of effective therapies. In this context, extensive efforts have been made to establish three-dimensional (3D) in vitro platforms that mimic the progression of liver fibrosis. METHODS This review outlines the pathophysiology of liver fibrosis and highlights recent advancements in 3D in vitro liver models, including spheroids, organoids, assembloids, bioprinted constructs, and microfluidic systems. It further assesses their biological relevance, with particular focus on their capacity to reproduce fibrosis-related characteristics. RESULTS 3D in vitro liver models offer significant advantages over conventional two-dimensional cultures. Although each model exhibits unique strengths, they collectively recapitulate key fibrotic features, such as extracellular matrix remodeling, hepatic stellate cell activation, and collagen deposition, in a physiologically relevant 3D setting. In particular, multilineage liver organoids and assembloids integrate architectural complexity with scalability, enabling deeper mechanistic insights and supporting therapeutic evaluation with improved translational relevance. CONCLUSION 3D in vitro liver models represent a promising strategy to bridge the gap between in vitro studies and in vivo realities by faithfully replicating liver-specific architecture and microenvironments. With enhanced reproducibility through standardized protocols, these models hold great potential for advancing drug discovery and facilitating the development of personalized therapies for liver fibrosis.
Collapse
Affiliation(s)
- Kyun Yoo Chi
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyeongmin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jeong Sang Son
- User Convenience Technology R&D Department, Korea Institute of Industrial Technology (KITECH), Ansan, 15588, South Korea
| | - Jiyou Han
- Department of Biomedical and Chemical Sciences, Hyupsung University, Hwasung-Si, 18330, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
41
|
Fukuda T, Thompson BR, Brouwers B, Qian HR, Wang W, Morse BL, LaBell ES, Durham TB, Konig M, Haupt A, Benson CT, MacKrell J. LY3522348, A New Ketohexokinase Inhibitor: A First-in-Human Study in Healthy Adults. Diabetes Ther 2025:10.1007/s13300-025-01752-5. [PMID: 40358849 DOI: 10.1007/s13300-025-01752-5] [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: 03/05/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION This study aimed to assess the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple doses of the ketohexokinase inhibitor LY3522348 in healthy participants. METHODS This first-in-human phase 1 study evaluated LY3522348, a highly selective, oral dual inhibitor of human ketohexokinase (KHK) isoforms C and A. The study was conducted in two parts: a single-ascending dose (SAD) study and a multiple-ascending dose (MAD) study, including a drug-drug interaction analysis with midazolam. Participants in the SAD study received single oral doses of LY3522348 ranging from 5 to 380 mg, while participants in the MAD study received once-daily doses of 50 mg, 120 mg, and 290 mg for 14 days. RESULTS A total of 65 healthy participants were included; of these 40 were in the SAD study (placebo = 10; LY3522348: 5 mg = 6; 15 mg = 6; 50 mg = 6; 150 mg = 6; 380 mg = 6) and 25 in the MAD study (placebo = 6; LY3522348: 50 mg = 6; 120 mg = 6; 290 mg = 7). LY3522348 was well tolerated, with the majority of the reported adverse events being mild. PK analysis showed an approximately dose-proportional increase in LY3522348 exposure, and the half-life ranged from 23.7 to 33.8 h. PD analysis indicated a dose-dependent increase in plasma fructose concentrations following the administration of a fructose beverage, supporting the inhibition of fructose metabolism by LY3522348. CONCLUSIONS LY3522348 demonstrated a favorable safety profile and well-behaved pharmacokinetics following once-daily oral dosing, and effective inhibition of fructose metabolism. The study was registered on ClinicalTrials.gov (NCT04559568).
Collapse
Affiliation(s)
- Tsuyoshi Fukuda
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Brian R Thompson
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Bram Brouwers
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Hui-Rong Qian
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Wei Wang
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Bridget L Morse
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | - Timothy B Durham
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Manige Konig
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Axel Haupt
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Charles T Benson
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - James MacKrell
- Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, 46285, USA.
| |
Collapse
|
42
|
Xu M, Gong R, Xie J, Xu S, Wang S. Clinical characteristics of lean and non-lean non-alcoholic fatty liver disease: a cross-sectional study. Nutr Metab (Lond) 2025; 22:40. [PMID: 40355898 PMCID: PMC12070601 DOI: 10.1186/s12986-025-00927-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) affects more than a quarter of the global population and has become the world's number one chronic liver disease, seriously jeopardizing public life and health. Despite the new terminology of metabolic dysfunction-associated steatotic liver disease (MASLD) has been proposed, the mechanisms underlying the heterogeneity across BMI stratification in non-alcoholic fatty liver disease (NAFLD) remain unclear. The aim of this study was to reveal the differences in metabolic and fibrotic characteristics between lean (BMI < 23 kg/m2) and non-lean NAFLD in an Asian population. METHODS The current study collected NAFLD patients from the physical examination population. Patients were divided into two groups by BMI to compare their clinical parameters, including lean (BMI < 23 kg/m2) and non-lean (BMI ≥ 23 kg/m2) and fibrosis subgroups (with a threshold of LSM = 8 kPa) and analyzed for risk factors by logistic regression models. RESULTS Of the 11,577 NAFLD patients who participated in the study, there were 916 lean and 10,661 non-lean. The non-lean group was younger than the lean group (median age 50 vs. 52 years, P < 0.001) and had a significantly higher prevalence of hypertension (28.0% vs. 18.3%), diabetes mellitus (10.1% vs. 6.1%), and liver fibrosis (9.1% vs. 5.1%) (all P < 0.001). Analysis of metabolic indexes showed that TyG, TyG-BMI, TG/HDL-C and APRI were higher in the non-lean group (all P < 0.001). Gender stratification revealed that ALT was significantly higher in the male non-lean group, while HDL-C was lower in the female non-lean group (1.35 vs. 1.47 mmol/L). Multiple regression suggested that the risk of fibrosis was independently associated with CAP values and fasting glucose, BMI, direct bilirubin, globulin, and age in the non-lean group, whereas the risk was mainly driven by GGT and ALP in the lean group. CONCLUSIONS Non-lean NAFLD patients showed more significant metabolic disturbances and risk of liver fibrosis. Although metabolic indicators (TyG, FIB-4) have limited predictive value for liver fibrosis, they are strongly associated with metabolic risk in MASLD.
Collapse
Affiliation(s)
- Mengyan Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Gong
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sanping Xu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Shi Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
43
|
Yang X, Guo C, Yang Y, Huang L, Luo L, Zhou Y, Xiao Y, Deng L, Li S. Targeting neutrophil extracellular traps: SERPINE1 and THBS1 as non-invasive biomarkers for early detection of liver fibrosis in metabolic dysfunction-associated Steatotic liver disease. Int Immunopharmacol 2025; 158:114828. [PMID: 40349409 DOI: 10.1016/j.intimp.2025.114828] [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: 02/17/2025] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Metabolic dysfunction - Associated Steatotic Liver Disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), poses a significant clinical burden due to its high prevalence and potential progression to fibrosis. While neutrophil extracellular traps (NETs) have been implicated in MASLD progression, their specific role in fibrosis remains unclear. This study integrates transcriptomic and single-cell data using weighted gene co-expression network analysis (WGCNA), causal WGCNA (CWGCNA), single-sample gene set enrichment analysis (ssGSEA), gene set variation analysis (GSVA), and linear models for microarray data (Limma) to identify key genes driving steatosis-to-fibrosis transition. Validation in human serum, mouse liver tissue, and mouse serum confirmed that SERPINE1 and THBS1 as robust non-invasive biomarkers with strong diagnostic performance. When combined with clinical features, these markers improved fibrosis prediction accuracy in MASLD patients. Additionally, SERPINE1 appears to mediate interactions between hepatic stellate cells and neutrophils, highlighting a novel therapeutic target. Overall, our findings reveal that NETs-related genes, particularly SERPINE1 and THBS1, hold strong diagnostic value for early-stage fibrosis in MASLD. Targeting SERPINE1 in hepatic stellate cells offers a promising strategy for therapeutic intervention. This study provides a novel framework for non-invasive MASLD fibrosis prediction and lays the foundation for targeted interventions to mitigate disease progression.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunhong Guo
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yichun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Huang
- Department of Pediatric Research Institute, Chongqing, China; National Clinical Research Center for Child Health and Disorders, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Luo
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, China
| | - Youping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuliang Xiao
- Department of Gastroenterology, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Liang Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Shan Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
44
|
Kalyesubula M, Von Bank H, Davidson JW, Burhans MS, Becker MM, Aljohani A, Simcox J, Ntambi JM. Stearoyl-CoA Desaturase 1 deficiency drives saturated lipid accumulation and increases liver and plasma acylcarnitines. J Lipid Res 2025:100824. [PMID: 40350036 DOI: 10.1016/j.jlr.2025.100824] [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: 06/07/2024] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025] Open
Abstract
Stearoyl-CoA desaturase-1 (SCD1) is a critical regulator of lipogenesis that catalyzes the synthesis of monounsaturated fatty acids (MUFA), mainly oleate (18:1n-9) and palmitoleate (16:1n-7) from saturated fatty acids (SFA), stearoyl-CoA (18:0) and palmitoyl-CoA (16:0), respectively. Elevated SCD1 expression and its products are associated with obesity, metabolic dysfunction-associated steatotic liver disease, insulin resistance, and cancer. Conversely, Scd1 deficiency diminishes de novo lipogenesis and protects mice against adiposity, hepatic steatosis, and hyperglycemia. Yet, the comprehensive impact of Scd1 deficiency on hepatic and circulating lipids remains incompletely understood. To further delineate the effects of SCD1 on lipid metabolism, we employed lipidomics on the liver from mice under a lipogenic high carbohydrate, very low-fat diet. We found that Scd1 deficiency leads to an accumulation of saturated lipids and an increase in hepatic and plasma acylcarnitines. Remarkably, transgenic replenishment of de novo oleate synthesis by human SCD5 in the liver of Scd1-deficient mice not only restored hepatic lipid desaturation levels but also attenuated acylcarnitine accumulation, highlighting the distinct role of SCD1 and oleate in regulating intracellular lipid homeostasis.
Collapse
Affiliation(s)
- Mugagga Kalyesubula
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Helaina Von Bank
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Maggie S Burhans
- Department of Nutritional Sciences, University of Wisconsin-Madison, 1415 Linden Drive, Madison, WI 53706, USA
| | - Madelaine M Becker
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Ahmed Aljohani
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11564, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh 11564, Saudi Arabia
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; HHMI, Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA.
| | - James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, 1415 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
45
|
Hao Y, Li X, Zhang Y, Zheng J, Miao Y, Tan J, Zhang Q. Combined effect of fasting blood glucose and serum uric acid on nonalcoholic fatty liver disease. Lipids Health Dis 2025; 24:168. [PMID: 40340743 PMCID: PMC12060386 DOI: 10.1186/s12944-025-02538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/17/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND This study sought to investigate the independent and synergistic impacts of fasting blood glucose (FBG) and serum uric acid (SUA) levels on non-alcoholic fatty liver disease (NAFLD) in participants with and without type 2 diabetes mellitus (T2DM). METHOD A total of 12,430 participants (mean age: 54.34 ± 15.23, 34.34% female) were enrolled through the Health Screening Center of Tianjin Medical University General Hospital. FBG was classified as < 6 mmol/L, 6-7 mmol/L, and ≥ 7 mmol/L. SUA was classified into two categories: normal SUA and hyperuricemia (SUA level ≥ 420 µmol/L for men, ≥ 360 µmol/L for women). T2DM was ascertained through self-reported data. The diagnosis of NAFLD is established via abdominal ultrasound imaging. Logistic regression models and interaction effect models are used for data analysis. RESULT Of the 12,430 participants, 4846 (38.99%) were diagnosed with NAFLD. In comparison to individuals with FBG < 6 mmol/L and no self-reported T2DM, those with FBG ≥ 7 mmol/L and no self-reported T2DM exhibited the highest prevalence of NAFLD (odds ratio [OR] 2.91, 95% CI 2.16-3.93) following multi-adjusted analysis. In the joint effect analysis of FBG and SUA, FBG ≥ 7 mmol/L and hyperuricemia were linked to a greater prevalence of NAFLD compared to FBG < 6 mmol/L and normal SUA, both in individuals with self-reported T2DM (OR 2.92, 95% CI 1.68-5.05) and those without self-reported T2DM (OR 7.87, 95% CI 3.57-17.34). An additive interaction existed between FBG and SUA regarding NAFLD in individuals without self-reported T2DM (AP 0.488, 95% CI: 0.068-0.909, P = 0.02). CONCLUSION Elevated FBG levels are associated with NAFLD irrespective of self-reported T2DM status. The concomitant elevation of FBG and SUA levels exhibits a significant correlation with NAFLD, particularly in individuals lacking self-reported T2DM.
Collapse
Affiliation(s)
- Yifan Hao
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Xuerui Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Yiting Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Jun Zheng
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in the Central Nervous System, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Elderly Health, Ministry of Education, Tianjin Geriatrics Institute, Tianjin, China.
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Key Laboratory of Elderly Health, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052, China.
| |
Collapse
|
46
|
Ye J, Zhu W, Cui Y, Zhang Q, Xiong Y, Jin L, Wang A, Lin M, Dong H, Liang G, Hu X, Luo W. Compound J27 alleviates high-fat diet-induced metabolic dysfunction-associated steatotic liver disease by targeting JNK. Int Immunopharmacol 2025; 154:114570. [PMID: 40188525 DOI: 10.1016/j.intimp.2025.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/12/2025] [Accepted: 03/26/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most characteristic form of liver diseases. As the member of MAPK family, the cJun-N-terminal-kinase (JNK) plays a crucial role in the pathogenesis of MASLD. A small molecule compound, J27, has demonstrated strong anti-inflammatory effects by inhibiting JNK phosphorylation, but its therapeutic potential in MASLD remains unclear. METHODS To evaluate the effect of J27, we used a high-fat diet (HFD)-induced MASLD mouse model with or without J27 treatment. Pathological changes were assessed through tissue staining, biochemical analysis, and other assays. In vitro, J27's effects were tested on macrophages, hepatocytes, and co-culture systems under palmitic acid stimulation. RESULTS J27 significantly reduced HFD-induced hepatic steatosis, liver injury, insulin resistance, and inflammatory responses by targeting JNK both in vivo and in vitro. On one hand, J27 blocked JNK activation, thereby improving insulin signaling and alleviating metabolic dysfunction in hepatocytes. On the other hand, J27 inhibited the inflammatory response in macrophages by disrupting the JNK/NF-κB axis, which, through cell-cell communication, further reduced hepatocyte injury. CONCLUSIONS J27, as a potent JNK inhibitor, markedly reduced HFD-induced MASLD, suggesting it as a promising therapeutic candidate for this disease.
Collapse
Affiliation(s)
- Jiaxi Ye
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Zhu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China
| | - Yaqian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Qianhui Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yongqiang Xiong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Leiming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Ao Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Mengsha Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Hui Dong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Guang Liang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China.
| | - Xiang Hu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| | - Wu Luo
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
47
|
Zhu X, Yin H, Han J, Zhang X, Han Q, Sun W, Liu Y, Tao W, Liu X, Wang G, Li L. Association Between Uric Acid to HDL-C Ratio and Metabolic Dysfunction-Associated Steatotic Liver Disease in Type 2 Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2025; 18:1459-1466. [PMID: 40356713 PMCID: PMC12067459 DOI: 10.2147/dmso.s520688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Background Patients with type 2 diabetes mellitus (T2DM) exhibit an elevated risk of developing metabolic dysfunction-associated steatotic liver disease (MASLD). The uric acid to high-density lipoprotein cholesterol ratio (UHR) has emerged as a novel metabolic biomarker implicated in MASLD pathogenesis. This study aimed to evaluate the association between UHR and MASLD in a T2DM population. Methods In this cross-sectional study, we analyzed clinical data from 1081 T2DM patients (464 without MASLD, 617 with MASLD). Physiological and biochemical parameters were collected and analyzed. UHR was calculated as [uric acid (mg/dL)/HDL-C (mg/dL)] × 100%. Univariate and multivariate logistic regression analyses were performed to examine the association between UHR and MASLD. Results T2DM patients with MASLD had significantly higher UHR levels than those without MASLD (12.12[9.06-16.83] vs 10.36[7.65-14.08], p<0.001). UHR showed a strong positive correlation with TG/HDL (r =0.673, p < 0.001), moderate correlations with TG (r = 0.516, p < 0.001) and TC/HDL (r =0.548, p < 0.001), weak but significant associations with BMI (r = 0.330), WHR (r = 0.289), HOMA-IR (r = 0.121), ALT (r = 0.123), and GGT (r = 0.267) (all p < 0.05). Multivariate logistic regression showed that elevated UHR levels were significantly associated with increased MASLD risk (adjusted OR = 1.057, 95% CI: 1.016-1.100, p = 0.006), after adjusting for age, diabetes duration, BMI, blood pressure, and biochemical confounders. Conclusion Elevated UHR is independently associated with MASLD in T2DM patients, suggesting its clinical relevance in MASLD screening among this high-risk population.
Collapse
Affiliation(s)
- Xiangyun Zhu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Han Yin
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Jing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xiaoyan Zhang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Qing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Yijun Liu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Wenxuan Tao
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Xinliang Liu
- Department of Endocrinology, Lianyungang Affiliated Hospital of Nanjing Medical University, Lianyungang, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Xuyi People’s Hospital of Clinical College of Yangzhou University, Huai’an, Jiangsu, 211700, People’s Republic of China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
- Pancreatic Research Institute, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| |
Collapse
|
48
|
Balakrishnan S, Ranganathan P, Prabhudesai KS, Vijay R, Gawali VP, Kareenhalli V. MASLD Risk Score (MRS) and Random Forest model (RF Model): Novel tools for screening and severity assessment of MASLD. Comput Biol Med 2025; 192:110314. [PMID: 40328026 DOI: 10.1016/j.compbiomed.2025.110314] [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: 12/06/2024] [Revised: 04/21/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND AND AIMS Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) spans from simple steatosis to progressive forms like non-alcoholic steatohepatitis (NASH), fibrosis, and cirrhosis, making early diagnosis and grading crucial. This study aimed to develop and validate predictive models for diagnosing and assessing MASLD severity using routinely available biomarkers from both public and clinical datasets. APPROACH & RESULTS We developed a novel MASLD Risk Score (MRS) using data from the CDC NHANES (2000-2020) and validated it in a clinically profiled Indian cohort. Unlike existing indices, the predictors were derived through data-driven feature selection from large dataset, ensuring statistical robustness. It integrates novel (Uric Acid, HOMA-IR) and established (liver enzymes, triglycerides, waist circumference, BMI) biomarkers to improve metabolic profiling and predictive accuracy. The MRS also uniquely enables grading of MASLD severity, addressing a key limitation of previous models. The MRS achieved AUROCs of 0.91 (public) and 0.85 (clinical) with accuracies of 94 % and 82 %, respectively. A Random Forest (RF) model built on the same features provided AUROCs of 0.87 (public) and 0.94 (clinical), with accuracies of 83 % and 82 %. MRS parameters were optimized using a diverse population, improving generalizability across demographics. Both models showed strong correlation with ultrasonography results and outperformed existing indices. CONCLUSIONS The MRS offers a novel, interpretable, and cost-effective solution for MASLD screening. Its development from a large, demographically diverse population and incorporation of varied biomarkers supports generalizability. While results are promising, external validation in multi-center clinical settings is needed to confirm broad utility.
Collapse
Affiliation(s)
| | | | | | - Ria Vijay
- Bhaktivedanta Hospital and Research Institute, India
| | | | - Venkatesh Kareenhalli
- MetFlux Research Private Limited, India; Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India.
| |
Collapse
|
49
|
Allen AM, Hoovler AR, Articolo A, Fisher T, Noureddin M, Dieterich D. Gaps in MASLD/MASH Education: A Quantitative and Qualitative Survey with Leaders of US Graduate Medical Education Programs. ADVANCES IN MEDICAL EDUCATION AND PRACTICE 2025; 16:729-748. [PMID: 40351776 PMCID: PMC12063625 DOI: 10.2147/amep.s491271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Purpose Metabolic dysfunction-associated steatotic liver disease (MASLD) and its inflammatory subtype, metabolic dysfunction-associated steatohepatitis (MASH), are associated with cardiometabolic risk factors, including obesity and type 2 diabetes. The prevalence of both conditions is rising rapidly and is underdiagnosed (<5%). We aimed to gather qualitative and quantitative insights from program leaders in US medical education training on their experience with MASH-related training and education. Participants and methods A cross-sectional study consisting of a quantitative survey and qualitative discussions with individuals in primary care (internal medicine and family medicine) and specialty programs (hepatology, gastroenterology, and endocrinology) were held from February 21 to August 28, 2023. Descriptive statistics were used for data analysis. Results A total of 190 leaders participated in the online survey and 11 leaders joined the focus groups. Almost all respondents reported that MASLD (96%) and MASH (92%) were included in their program's curricula. However, many believed that little time was devoted to discussing MASH in their program. Most respondents agreed that MASH is extremely underdiagnosed. Program leaders agreed that the interconnectedness of MASH with other cardiometabolic conditions necessitates instruction time on MASH beyond that of its dedicated curriculum time. All participants believed that emergence of regulatory-approved drugs for MASH will drive a decision to increase the time allotted for MASH in the curriculum. Conclusion Although program leaders agreed that MASH has an important place in medical education curricula, the relative paucity of treatment options reduces its coverage in training, thereby limiting healthcare practitioners' understanding of MASH.
Collapse
Affiliation(s)
- Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Amy Articolo
- Global Medical Affairs, Novo Nordisk Inc., Plainsboro, NJ, USA
| | - Travis Fisher
- US Medical Affairs, Novo Nordisk Inc., Plainsboro, NJ, USA
| | - Mazen Noureddin
- Department of Medicine, Houston Methodist Hospital, and Houston Research Institute, Houston, TX, USA
| | - Douglas Dieterich
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Elangovan H, George J. Guidelines for the evolving landscape of liver disease: From viral hepatitis to MAFLD. Chin Med J (Engl) 2025; 138:1013-1015. [PMID: 40134300 PMCID: PMC12068753 DOI: 10.1097/cm9.0000000000003616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Indexed: 03/27/2025] Open
Affiliation(s)
- Harendran Elangovan
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, NSW, Australia
| |
Collapse
|