1
|
Saha T, Mehrotra S, Gupta P, Kumar A. Exosomal miRNA combined with anti-inflammatory hyaluronic acid-based 3D bioprinted hepatic patch promotes metabolic reprogramming in NAFLD-mediated fibrosis. Biomaterials 2025; 318:123140. [PMID: 39892017 DOI: 10.1016/j.biomaterials.2025.123140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex metabolic disorder, where the underlying molecular mechanisms are mostly not well-understood and therefore, warrants the need for therapeutic interventions targeting several metabolic pathways as a unified response. Of late, promising outcomes have been observed with mesenchymal stem cell-derived exosomes. However, reduced bioavailability due to systemic delivery and the need for repeated fresh isolation hinders their feasibility for clinical applications. In this regard, an 'off-the-shelf' 3D bioprinted hyaluronic acid-based hepatic patch to deliver encapsulated exosomes alone/or with hepatocytes (as dual-therapy) is developed as a holistic approach for ameliorating the disease condition and promoting tissue regeneration. The bioprinted hepatic patch demonstrated sustained and localized release of exosomes (∼82 % in 21 days), and healthy liver tissue-like mechanical properties while being biocompatible and biodegradable. Assessment in NAFLD rat models displayed alleviation of the altered biochemical parameters such as fat deposition, deranged liver functions, disrupted lipid, glucose, and insulin metabolism along with a reduction in localized inflammation, and associated liver fibrosis. The study suggests that a synergistic effect between the miRNA population of released exosomes, cell therapy, and the bioprinted matrix materials is crucial in targeting multiple complex metabolic pathways associated with the severity of the disease.
Collapse
Affiliation(s)
- Triya Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| | - Purva Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India; Centre of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India.
| |
Collapse
|
2
|
Gao S, Wang X, Xu Q, Li R, Yao L, Zhang A, Zhou Q, Xiao Z, Li S, Meng X, Wu J, Qin L. Total Sanghuangporus vaninii extract inhibits hepatocyte ferroptosis and intestinal microbiota disturbance to attenuate liver fibrosis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119571. [PMID: 40023344 DOI: 10.1016/j.jep.2025.119571] [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: 11/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanghuangporus, the dried fruiting body of Sanghuangporus vaninii (Ljub) L.W.Zhou et Y.C.Dai. As the main species of Sanghuang, it has been well-known and used commonly as a traditional medicinal and edible macrofungi for thousands of years in many countries, including China, Korea and Japan. Although it has good hepatoprotective activity, its potential efficacy and mechanism on liver fibrosis remain elusive. AIM OF THE STUDY Total Sanghuangporus vaninii extract (TSH) was prepared by ethanol extraction to investigate its chemical components and to conduct an initial assessment of its efficacy and underlying mechanism in a murine model of liver fibrosis. MATERIALS AND METHODS The chemical components of TSH were initially analyzed by UHPLC-Q-Orbitrap HRMS. To elucidate the effects of TSH, an in vivo model of fibrosis was established in mice using carbon tetrachloride (CCl4), followed by assessments of serum liver function and histopathological analysis. Besides, indicators related to liver fibrosis, hepatic stellate cells (HSCs) activation, inflammation response and ferroptosis related indicators were detected by western blotting, immunohistochemistry and real-time quantitative PCR (RT-qPCR) analysis. Additionally, the 16S rDNA sequencing and untargeted metabolomics analysis of intestinal microbiota were employed to investigating the role of TSH in gut microbiome. In vitro, the human hepatocyte line L02 was stimulated with erastin and treated with or without TSH to elucidate its underlying mechanism. RESULTS The administration of TSH significantly improved serum indicators of liver injury in CCl4-induced fibrosis mice, reduced HSCs activation and collagen deposition, while inhibiting the expressions of transforming growth factor-β1(TGF-β1)/Smad signaling pathway. Notably, TSH treatment attenuated hepatocyte ferroptosis and lipid peroxidation both in vivo and in vitro, as evidenced by a marked decrease in liver iron and malondialdehyde (MDA) contents. In particular, TSH was demonstrated to activate the nuclear factor erythroid 2-related factor 2 (Nrf2)-glutathione peroxidase 4 (GPX4) signaling pathway, thereby protecting hepatocytes from ferroptosis with a particular enhancement of Nrf2 nuclear transcription. Furthermore, TSH influenced gut microbiota composition and ameliorated intestinal metabolic disorders. The increased abundance of Parasutterella and Olsenellas due to TSH treatment was significantly positively correlated with elevated phosphatidylcholines involved in linoleic acid metabolism, and negatively correlated with the reduction of fatty acyls. And the enrichment of intestinal linoleic acid metabolism presented a negative correlation in liver fibrosis biomarkers. CONCLUSIONS Our findings indicate that the TSH treatment exerts a significantly protective effect on CCl4-induced mice by ameliorating hepatic injury and ferroptosis damage, inhibiting HSCs activation and collagen deposition, and remodeling gut microbiota homeostasis and metabolic imbalance. Notably, TSH attenuated hepatocyte ferroptosis in liver fibrosis and exhibited upregulation of the Nrf2-GPX4 signaling pathway. Furthermore, TSH could enrich the abundance of Parasutterella and Olsenellas, which may contribute to intestinal linoleic acid metabolism, thereby contributing to the reduction of liver fibrosis damage. Our study provides more effective and unreported evidence of TSH in anti-fibrosis activity.
Collapse
Affiliation(s)
- Siqi Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xingxing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiuying Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Rongsheng Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lumeng Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Anna Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qun Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Zhun Xiao
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Shengsheng Li
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Jianjun Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
3
|
Guan Q, Dong R, Zhang M, Chang D, Zhang R, Wang Y, Zhang W, Wang J. Factors Influencing Knowledge-Action Gap in Patients With Metabolic Dysfunction-Associated Fatty Liver Disease: A Qualitative Study. JOURNAL OF NUTRITION EDUCATION AND BEHAVIOR 2025; 57:274-284. [PMID: 39864004 DOI: 10.1016/j.jneb.2024.12.010] [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: 02/14/2024] [Revised: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
OBJECTIVE To explore the knowledge-action gap regarding health behaviors and their influencing factors among patients with metabolic dysfunction-associated fatty liver disease (MAFLD), using the Health Belief Model as a theoretical framework. DESIGN A qualitative approach was adopted, involving semistructured interviews with individuals with MAFLD. SETTING Participants were recruited from a community hospital and a tertiary hospital in Nanjing, China, between July and October 2022. PARTICIPANTS A purposive sample of 21 adults with MAFLD, who were primarily overweight or obese (86%), males (52%), and aged ≥ 60 years (52%). PHENOMENON OF INTEREST This study focused on the knowledge-action gap in health behaviors among MAFLD patients. ANALYSIS Data were analyzed using content analysis, with the Health Belief Model guiding the identification of themes and categorization of specific domains. RESULTS This study found that perceptions of disease susceptibility and severity, perceived barriers to healthy lifestyles, and various modifying factors impeded the adoption of healthy behaviors. In contrast, perceived benefits, cues to action, and self-efficacy facilitated the implementation of these behaviors. CONCLUSIONS AND IMPLICATIONS This research highlights the factors contributing to the knowledge-action gap in health behaviors among MAFLD patients. The findings suggest potential targets for interventions aimed at enhancing the alignment between patients' knowledge and their actions, ultimately promoting better health outcomes.
Collapse
Affiliation(s)
- Qing Guan
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengting Zhang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongchun Chang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ru Zhang
- School of Nursing and Midwifery, Jiangsu College of Nursing, Huaian, Jiangsu, China
| | - Yunqi Wang
- Nanjing Foreign Language School, Nanjing, Jiangsu, China
| | - Wei Zhang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Wakabayashi S, Kimura T, Tamaki N, Iwadare T, Okumura T, Kobayashi H, Yamashita Y, Tanaka N, Kurosaki M, Umemura T. AI-Based Platelet-Independent Noninvasive Test for Liver Fibrosis in MASLD Patients. JGH Open 2025; 9:e70150. [PMID: 40191781 PMCID: PMC11969565 DOI: 10.1002/jgh3.70150] [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: 09/06/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Background and Aim Noninvasive tests (NITs), such as platelet-based indices and ultrasound/MRI elastography, are widely used to assess liver fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD). However, platelet counts are not routinely included in Japanese health check-ups, limiting their utility in large-scale screenings. Additionally, elastography, while effective, is costly and less accessible in routine practice. Most existing AI-based models incorporate these markers, restricting their applicability. This study aimed to develop a simple yet accurate AI model for liver fibrosis staging using only routine demographic and biochemical markers. Methods This retrospective study analyzed biopsy-proven data from 463 Japanese MASLD patients. Patients were randomly assigned to training (N = 370, 80%) and test (N = 93, 20%) cohorts. The AI model incorporated age, sex, BMI, diabetes, hypertension, hyperlipidemia, and routine blood markers (AST, ALT, γ-GTP, HbA1c, glucose, triglycerides, cholesterol). Results The Support Vector Machine model demonstrated high diagnostic performance, with an area under the curve (AUC) of 0.886 for detecting significant fibrosis (≥ F2). The AUCs for advanced fibrosis (≥ F3) and cirrhosis (F4) were 0.882 and 0.916, respectively. Compared to FIB-4, APRI, and FAST score (0.80-0.96), SVM achieved comparable accuracy while eliminating the need for platelet count or elastography. Conclusion This AI model accurately assesses liver fibrosis in MASLD patients without requiring platelet count or elastography. Its simplicity, cost-effectiveness, and strong diagnostic performance make it well-suited for large-scale health screenings and routine clinical use.
Collapse
Affiliation(s)
- Shun‐ichi Wakabayashi
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
| | - Takefumi Kimura
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
- Consultation Center for Liver DiseasesShinshu University HospitalMatsumotoJapan
| | - Nobuharu Tamaki
- Department of Gastroenterology and HepatologyMusashino Red Cross HospitalTokyoJapan
| | - Takanobu Iwadare
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
| | - Taiki Okumura
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
| | - Hiroyuki Kobayashi
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
| | - Yuki Yamashita
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
| | - Naoki Tanaka
- Department of Global Medical Research PromotionShinshu University Graduate School of MedicineMatsumotoJapan
- International Relations OfficeShinshu University School of MedicineMatsumotoJapan
- Research Center for Social SystemsShinshu UniversityMatsumotoJapan
| | - Masayuki Kurosaki
- Department of Gastroenterology and HepatologyMusashino Red Cross HospitalTokyoJapan
| | - Takeji Umemura
- Department of Medicine, Division of GastroenterologyShinshu University School of MedicineMatsumotoJapan
- Consultation Center for Liver DiseasesShinshu University HospitalMatsumotoJapan
| |
Collapse
|
5
|
Tsitsou S, Bali T, Adamantou M, Saridaki A, Poulia K, Karagiannakis DS, Papakonstantinou E, Cholongitas E. Effects of a 12-Week Mediterranean-Type Time-Restricted Feeding Protocol in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomised Controlled Trial-The 'CHRONO-NAFLD Project'. Aliment Pharmacol Ther 2025; 61:1290-1309. [PMID: 40017349 PMCID: PMC11950810 DOI: 10.1111/apt.70044] [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: 12/17/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND The Mediterranean diet (MD) is considered the best dietary approach for patients with metabolic dysfunction-associated steatotic liver disease (MASLD). Recently, time-restricted feeding (TRF) has gained attention for its lifestyle compatibility and health benefits. AIMS This study aimed to compare the effects of a hypocaloric MD with a 10-h TRF protocol to an unrestricted MD in MASLD patients with overweight/obesity and evaluate differences between early and late TRF. METHODS This 12-week randomised controlled trial in MASLD patients with overweight/obesity consisted of three groups, all following a hypocaloric Mediterranean-type diet. The control group had no eating time restrictions. The early TRF (eTRF) and late TRF (lTRF) groups had a 10-h eating window, from 8 AM to 6 PM and from 12 PM to 10 PM, respectively. Various health parameters were measured. Compliance was tracked via food diaries, and an 8-week follow-up occurred post-intervention. RESULTS Fifty-nine MASLD individuals (27 males; 52.9 years; body mass index 32.1 kg/m2) completed the trial (control, n = 19; eTRF, n = 20; lTRF, n = 20). All groups showed significant 12-week reductions in body weight, anthropometry and blood pressure. Glycated haemoglobin A1c and insulin resistance, as measured by the Matsuda index, homeostatic model assessment for insulin resistance and fasting glucose-to-insulin ratio, improved in the eTRF group at 12 weeks. CONCLUSIONS This study corroborates the efficacy of MD in ameliorating cardiometabolic risk factors such as body weight and blood pressure in MASLD patients. The combination with an eTRF protocol may improve glycaemic control (NCT05866744). TRIAL REGISTRATION The study is registered at clinicaltrials.gov (NCT05866744).
Collapse
Affiliation(s)
- Sofia Tsitsou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human NutritionAgricultural University of AthensAthensGreece
| | - Triada Bali
- First Department of Internal Medicine, Laiko General Hospital of Athens, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Magdalini Adamantou
- First Department of Internal Medicine, Laiko General Hospital of Athens, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Aristi Saridaki
- First Department of Internal Medicine, Laiko General Hospital of Athens, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Kalliopi‐Anna Poulia
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human NutritionAgricultural University of AthensAthensGreece
| | - Dimitrios S. Karagiannakis
- Fourth Department of Internal MedicineAttiko University Hospital, Medical School, National and Kapodistrian University of AthensAthensGreece
| | - Emilia Papakonstantinou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human NutritionAgricultural University of AthensAthensGreece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital of Athens, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
6
|
Sun L, He Z, Li Y, Huo Z, Liu L. The relationship between nutrient intake, lifestyle, and non-alcoholic fatty liver disease in patients with type 2 diabetes mellitus: Results from the National Health and Nutrition Examination Surveys 2007-2018. Clin Nutr ESPEN 2025; 66:446-453. [PMID: 39993562 DOI: 10.1016/j.clnesp.2025.02.008] [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: 05/17/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE More than half of the individuals with type 2 diabetes mellitus (T2DM) are accompanied by Non-alcoholic fatty liver disease (NAFLD). This study aimed to explore the relationship between nutrient intake, lifestyle, and the risk NAFLD in patients with T2DM. METHODS This study comprised 2110 adult patients with diabetes from the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2018. We employed weighted logistic regression to assess the associations between nutrient intake, lifestyle and NAFLD, while exploring potential non-linear relationships using restrictive cubic spline analysis. Additionally, we validated our findings through subgroup analyses and sensitivity analyses to ensure the robustness and reliability of our results. RESULT Out of 2110 diabetes patients, 1743 were diagnosed with NAFLD, and 53.43 % of them were male. After adjusting for potential confounders, we found a negative correlation between Vitamin K intake and the occurrence of NAFLD in patients with T2DM (OR = 0.885 [0.829, 0.959]). This dose-response relationship was further validated through stratification analysis by tertiles of vitamin K intake. Lycopene intake is identified as a risk factor for NAFLD in patients with T2DM. Specifically, for every 100 μg of lycopene ingested, there was a 0.2 % higher likelihood of NAFLD (OR = 1.002 [1.0001, 1.005], p < 0.05). Conversely, a 0.5 % reduction in NAFLD risk was observed with the same amount of lutein intake (OR = 0.995 [0.992, 0.999], p < 0.05). Furthermore, we also found that a high-quality diet can reduce the risk of NAFLD in patients with T2DM, with an odds ratio of 0.208 (0.101, 0.430). CONCLUSION Increasing intake of Vitamin K and lutein, reducing intake of lycopene, and improving dietary quality may lower the risk of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Liangyuanhui Sun
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Department of Gastroenterology, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, China.
| | - Zhiqing He
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 519041, Guangdong, China.
| | - Yanbin Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Zijun Huo
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Liehua Liu
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
7
|
Kumar A, Arora A, Choudhury A, Arora V, Rela M, Jothimani DK, Mahtab MA, Devarbhavi H, Eapen CE, Goel A, Yaghi C, Ning Q, Chen T, Jia J, Zhongping D, Hamid SS, Butt AS, Jafri W, Shukla A, Tan SS, Kim DJ, Saraya A, Hu J, Sood A, Goyal O, Midha V, Pati GK, Singh A, Lee GH, Treeprasertsuk S, Thanapirom K, Mandot A, Maghade R, Lesmana RC, Ghazinyan H, Mohan Prasad VG, Dokmeci AK, Sollano JD, Abbas Z, Shrestha A, Lau GK, Payawal DA, Shiha GE, Duseja A, Taneja S, Verma N, Rao PN, Kulkarni AV, Karim F, Saraswat VA, Alam S, Chowdhury D, Kedarisetty CK, Saigal S, Sharma P, Yattoo GN, Koshy A, Patwa AK, Elbasiony M, Rathi PM, Maharshi S, Dayal VM, Jha AK, Kalista KF, Gani RA, Yuen MF, Singh V, Sargsyan VA, Huang CH, Mukewar SS, Xin S, Rajaram RB, Panackel C, Dadhich S, Sachdeva S, Kumar A, Behera S, Kamani L, Saithanyamurthi HV, Prasad B, Sarin SK. Impact of Diabetes, Drug-Induced Liver Injury, and Sepsis on Outcomes in Metabolic Dysfunction Associated Fatty Liver Disease-Related Acute-on-Chronic Liver Failure. Am J Gastroenterol 2025; 120:816-826. [PMID: 39016385 DOI: 10.14309/ajg.0000000000002951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/09/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION The prevalence of metabolic dysfunction-associated fatty liver disease (MAFLD) and its complication, MAFLD-related acute-on-chronic liver failure (MAFLD-ACLF), is rising. Yet, factors determining patient outcomes in MAFLD-ACLF remain understudied. METHODS Patients with MAFLD-ACLF were recruited from the Asian Pacific Association for the Study of the Liver-ACLF Research Consortium (AARC registry). The diagnosis of MAFLD-ACLF was made when the treating unit had identified the etiology of chronic liver disease as MAFLD (or previous nomenclature such as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, or non-alcoholic steatohepatitis-cirrhosis). Patients with coexisting other etiologies of chronic liver disease (such as alcohol, hepatitis B virus, hepatitis C virus, etc.) were excluded. Data were randomly split into derivation (n = 258) and validation (n = 111) cohorts at a 70:30 ratio. The primary outcome was 90-day mortality. Only the baseline clinical, laboratory features and severity scores were considered. RESULTS The derivation group had 258 patients; 60% were male, with a mean age of 53. Diabetes was noted in 27% and hypertension in 29%. The dominant precipitants included viral hepatitis (hepatitis A virus and hepatitis E virus, 32%), drug-induced injury (drug-induced liver injury, 29%), and sepsis (23%). Model for End-Stage Liver Disease-Sodium (MELD-Na) and AARC scores on admission averaged 32 ± 6 and 10.4 ± 1.9. At 90 days, 51% survived. Nonviral precipitant, diabetes, bilirubin, international normalized ratio, and encephalopathy were independent factors influencing mortality. Adding diabetes and precipitant to MELD-Na and AARC scores, the novel MAFLD-MELD-Na score (+12 for diabetes, +12 for nonviral precipitant), and MAFLD-AARC score (+5 for each) were formed. These outperformed the standard scores in both cohorts. DISCUSSION Almost half of patients with MAFLD-ACLF die within 90 days. Diabetes and nonviral precipitants such as drug-induced liver injury and sepsis lead to adverse outcomes. The new MAFLD-MELD-Na and MAFLD-AARC scores provide reliable 90-day mortality predictions for patients with MAFLD-ACLF.
Collapse
Affiliation(s)
- Ashish Kumar
- Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, India
| | - Anil Arora
- Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, India
| | | | - Vinod Arora
- Institute of Liver and Biliary Sciences, New Delhi, India
| | | | | | - Mamun A Mahtab
- Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | | | | | | | - Qin Ning
- Tongji Hospital, Wuhan/Capital Medical University, Beijing, China
| | | | - Jidong Jia
- Tongji Hospital, Wuhan/Capital Medical University, Beijing, China
| | - Duan Zhongping
- Youan Hospital Capital Medical University, Beijing, China
| | | | - Amna S Butt
- Aga Khan University Hospital, Karachi, Pakistan
| | - Wasim Jafri
- Aga Khan University Hospital, Karachi, Pakistan
| | - Akash Shukla
- Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Seok S Tan
- Selayang Hospital University of Malaysia, Malaya, Malaysia
| | - Dong J Kim
- Chuncheon Sacred Heart Hospital, Chuncheon, Korea
| | - Anoop Saraya
- Institute of liver and Biliary Sciences, New Delhi, India
| | - Jinhua Hu
- The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Ajit Sood
- Dayanand Medical College, Ludhiana, India
| | | | | | | | - Ayaskant Singh
- SUM Ultimate Medicare and SUM Hospital, SOA University, Bhubaneswar, Odisha, India
| | - Guan H Lee
- National University Hospital, Kent Ridge, Queenstown, Singapore
| | | | | | | | | | - Rinaldi C Lesmana
- Digestive Disease and Oncology Center, Medistra Hospital, Jakarta, Indonesia
| | | | | | - Abdul K Dokmeci
- Ankara University School of Medicine, Hacettepe, Ankara, Turkey
| | - Jose D Sollano
- Cardinal Santos Medical Center, Metro Manila, Philippines
| | - Zaigham Abbas
- Dr. Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | - George K Lau
- Humanity and Health Medical Centre, Hong Kong, SAR, China
| | | | - Gamal E Shiha
- Egyptian Liver Research Institute and Hospital (ELRIAH), Egypt
| | - Ajay Duseja
- Post Graduate Institute of Medical Education and Research Chandigarh, India
| | - Sunil Taneja
- Post Graduate Institute of Medical Education and Research Chandigarh, India
| | - Nipun Verma
- Post Graduate Institute of Medical Education and Research Chandigarh, India
| | - Padaki N Rao
- Asian Institute of Gastroentrology, Somajiguda, Hyderabad, India
| | - Anand V Kulkarni
- Asian Institute of Gastroentrology, Somajiguda, Hyderabad, India
| | - Fazal Karim
- Sir Salimullah Medical College, Mitford Hospital, Dhaka, Bangladesh
| | - Vivek A Saraswat
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Shahinul Alam
- Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | | | - Sanjiv Saigal
- Max Super Speciallity Hospital, Saket, New Delhi, India
| | - Praveen Sharma
- Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, India
| | - Ghulam N Yattoo
- Sher-e-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Abraham Koshy
- VPS Lakeshore Hospital and Research Center Ltd, Kochi, Kerala, India
| | | | | | - Pravin M Rathi
- Topi Wala National (TN) Medical College and BYL Nair Charitable Hospital, Mumbai, India
| | | | - Vishwa M Dayal
- Indira Gandhi Institute of Medical Sciences (IGIMS), Bailey Road, Patna, Bihar, India
| | - Ashish K Jha
- Indira Gandhi Institute of Medical Sciences (IGIMS), Bailey Road, Patna, Bihar, India
| | | | - Rino A Gani
- Cipto Mangunkusumo General Hospital, Jakarta , Indonesia
| | - Man F Yuen
- Department of Medicine , University of Hong Kong, Hong Kong, China
| | - Virendra Singh
- Punjab Institute of Liver and Biliary Sciences, Mohali, Punjab, India
| | | | - Chien H Huang
- Chang Gung Medical Foundation Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | | | | | | | | | - Sunil Dadhich
- Dr. Sampuranand Medical College (SNMC), Jodhpur, Rajasthan, India
| | | | - Ajay Kumar
- Govind Ballabh Pant Hospital, New Delhi, India
| | | | | | | | - Babita Prasad
- Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K Sarin
- Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
8
|
Wu X, Song Y, Yuan Z, Wu S. Preclinical insights into the potential of itaconate and its derivatives for liver disease therapy. Metabolism 2025; 165:156152. [PMID: 39909101 DOI: 10.1016/j.metabol.2025.156152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/12/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Annually, approximately 3.5 % of the world's population dies of cirrhosis or liver cancer, and the burden of liver disease is steadily expanding owing to multiple factors such as alcohol consumption, irrational diets, viral transmission, and exposure to drugs and toxins. However, the lack of effective therapies and the adverse effects of some medications remain a threat to the management of liver disease. Recently, immunometabolism, as an emerging discipline, appears to be the focus of unprecedented research. As a natural metabolite that regulates cellular functions, itaconate is a crucial bridge connecting metabolism and immune response. Remodeling immune function through metabolic modulation may be a promising alternative for disease intervention strategies. In this review, we first briefly describe the historical origin of itaconate and the development of its derivatives. This was followed by a review of the molecular mechanisms by which itaconate regulated immune-metabolic responses. Furthermore, we analyzed the effects of itaconate regulation on immune cells of the hepatic system. Finally, we summarized the experimental evidence for itaconate and its derivatives in the therapeutic application of liver diseases. Itaconate is potentially an invaluable component of emerging therapeutic strategies for liver disease.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Azizi N, Naghibi H, Shakiba M, Morsali M, Zarei D, Abbastabar H, Ghanaati H. Evaluation of MRI proton density fat fraction in hepatic steatosis: a systematic review and meta-analysis. Eur Radiol 2025; 35:1794-1807. [PMID: 39254718 DOI: 10.1007/s00330-024-11001-1] [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: 02/10/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Amidst the global rise of metabolic dysfunction-associated steatotic liver disease (MASLD), driven by increasing obesity rates, there is a pressing need for precise, non-invasive diagnostic tools. Our research aims to validate MRI Proton Density Fat Fraction (MRI-PDFF) utility, compared to liver biopsy, in grading hepatic steatosis in MASLD. METHODS A systematic search was conducted across Embase, PubMed/Medline, Scopus, and Web of Science until January 13, 2024, selecting studies that compare MRI-PDFF with liver biopsy for hepatic steatosis grading, defined as grades 0 (< 5% steatosis), 1 (5-33% steatosis), 2 (34-66% steatosis), and 3 (> 66% steatosis). RESULTS Twenty-two studies with 2844 patients were included. The analysis showed high accuracy of MRI-PDFF with AUCs of 0.97 (95% CI = 0.96-0.98) for grade 0 vs ≥ 1, 0.91 (95% CI = 0.88-0.93) for ≤ 1 vs ≥ 2, and 0.91 (95% CI = 0.88-0.93) for ≤ 2 vs 3, diagnostic odds ratio (DOR) from 98.74 (95% CI = 58.61-166.33) to 23.36 (95% CI = 13.76-39.68), sensitivity and specificity from 0.93 (95% CI = 0.88-0.96) to 0.76 (95% CI = 0.63-0.85) and 0.93 (95% CI = 0.88-0.96) to 0.89 (95% CI = 0.84-0.93), respectively. Likelihood ratio (LR) + ranged from 13.3 (95% CI = 7.4-24.0) to 7.2 (95% CI = 4.9-10.5), and LR - from 0.08 (95% CI = 0.05-0.13) to 0.27 (95% CI = 0.17-0.42). The proposed MRI-PDFF threshold of 5.7% for liver fat content emerges as a potential cut-off for the discrimination between grade 0 vs ≥ 1 (p = 0.075). CONCLUSION MRI-PDFF is a precise non-invasive technique for diagnosing and grading hepatic steatosis, warranting further studies to establish its diagnostic thresholds. CLINICAL RELEVANCE STATEMENT This study underscores the high diagnostic accuracy of MRI-PDFF for distinguishing between various grades of hepatic steatosis for early detection and management of MASLD, though further research is necessary for broader application. KEY POINTS MRI-PDFF offers precision in diagnosing and monitoring hepatic steatosis. The diagnostic accuracy of MRI-PDFF decreases as the grade of hepatic steatosis advances. A 5.7% MRI-PDFF threshold differentiates steatotic from non-steatotic livers.
Collapse
Affiliation(s)
- Narges Azizi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hamed Naghibi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Madjid Shakiba
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Mina Morsali
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Diana Zarei
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hedayat Abbastabar
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hossein Ghanaati
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
10
|
Xie J, He Q, Fisher D, Pronyuk K, Musabaev E, Zhao L. Association of platelet to albumin ratio with metabolic dysfunction-associated steatotic liver disease based on the National Health and Nutrition Survey 2017-2018. Sci Rep 2025; 15:10573. [PMID: 40148478 PMCID: PMC11950324 DOI: 10.1038/s41598-025-92837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
The prevalence and incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) are significantly increasing globally, but the index of non-invasive disease is limited. Platelet-albumin ratio(PAR) is a non-invasive biomarker of inflammation, the aim of this study was to evaluate the relationship between PAR and MASLD. This population-based cross-sectional retrospective study analyzed data extracted from the National Health and Nutrition Survey (NHANES) database from 2017 to 2018. Multivariate logistic regression analysis was used to evaluate the correlation between PAR and MASLD in different models. Model I was unadjusted, model II adjusted for race, sex and age, and model III was adjusted based on model II plus smoking status, hypertension, and diabetes. Further subgroup analysis was carried out according to sex, age, hypertension and diabetes status. The study involved 3287 participants, of whom 873 (26.5%) were diagnosed with MASLD. The PAR level in MASLD group was significantly higher than non-MASLD group (P < 0.05). Multivariate logistic regression revealed that high PAR level was an independent risk factor for MASLD (OR = 2.58, 95%CI: 1.26-5.27, P = 0.03), which adjusted for sex, age, race, smoking status, hypertension, and diabetes.The same results were observed in multiple subgroups of further subgroup analysis, and it can effectively predict the risk of MASLD (AUC = 0.842, 95% CI: 0.826-0.859). In conclusion, the new biomarker PAR shows a positive correlation with the risk of MASLD in the population, and can be used as a biomarker of MASLD to help clinicians identify people at high risk of MASLD.
Collapse
Affiliation(s)
- Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qingliu He
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | - Khrystyna Pronyuk
- Infectious Diseases Department, O.Bogomolets National Medical University, Kyiv, Ukraine
| | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, Tashkent, 100122, Uzbekistan
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
11
|
Liao X, Yu S, Wang L, Zhang R, Yu K. Elevated red blood cell folate levels are associated with metabolic dysfunction-associated steatotic liver disease: results from NHANES 2017-2020. Front Physiol 2025; 16:1494863. [PMID: 40182691 PMCID: PMC11965589 DOI: 10.3389/fphys.2025.1494863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. However, the role of folate in MASLD remains controversial. This study aimed to investigate the association between two folate indicators [serum folate and red blood cell (RBC) folate] and MASLD prevalence using data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES). Methods A total of 3,879 participants without liver disease or significant alcohol consumption were included in the final analysis. Hepatic steatosis was assessed via transient elastography, with MASLD defined as a controlled attenuation parameter (CAP) ≥285 dB/m and the presence of at least one cardiometabolic risk factor. Logistic regression and generalized additive models (GAMs) were used to evaluate associations between folate levels and MASLD, with subgroup analyses stratified by age, gender, and body mass index (BMI). Results After full adjustment for confounders, RBC folate exhibited a significant positive association with MASLD (OR = 1.111 and 95% CI: 1.015-1.216 per 1-unit increase). In contrast, serum folate showed a transient negative association in minimally adjusted models (OR = 0.869 and 95% CI: 0.802-0.941), which disappeared after further adjustments. Subgroup analyses confirmed that age, gender, and BMI did not modify the RBC folate-MASLD relationship. Discussion These findings suggest that elevated RBC folate levels are independently associated with MASLD prevalence, whereas serum folate may lack clinical relevance due to susceptibility to confounding factors. RBC folate, as a stable biomarker of long-term folate status, may serve as a superior indicator for investigating folate-MASLD associations.
Collapse
Affiliation(s)
- Xin Liao
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Song Yu
- Department of General Medicine, the Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People’s Hospital, Chengdu, Sichuan, China
| | - Lin Wang
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Ruyue Zhang
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Ke Yu
- Department of General Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Li J, Li P, Wu X, Li Z, Li Y, Liu C, Bian J, Han L, Wang M. Protocatechuic Acid Reduces Liver Fatty Acid Uptake in HFD-Fed Mice Associated With the Inhibition of DHHC5-Mediated CD36 Palmitoylation. Mol Nutr Food Res 2025:e70012. [PMID: 40109131 DOI: 10.1002/mnfr.70012] [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: 10/22/2024] [Revised: 01/08/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is highly prevalent and has emerged as a pressing issue for human health. A highly palmitoylated cluster of differentiation 36 (CD36) promotes free fatty acid (FFA) uptake, which contributes to the development of MASLD. Protocatechuic acid (PCA), the main metabolite of anthocyanins, was reported to inhibit MASLD by regulating the expression of CD36. However, the impact of PCA on CD36 palmitoylation has not been extensively studied. In the present study, we found that PCA could significantly reduce lipid uptake and accumulation in hepatocytes by decreasing CD36 palmitoylation. Inhibitors were used to prove that PCA suppressed CD36 palmitoylation by lowering zinc finger DHHC-type palmitoyltransferase 5 (DHHC5) palmitoylation, but not in an acyl protein thioesterase 1 (APT1)-dependent manner. Further experiments showed that PCA-mediated inhibition of DHHC5 palmitoylation and acyltransferase activity was closely related to the reduction of the CD36/Fyn/Lyn complex. PCA diminished the palmitoylation of CD36 and DHHC5 and ultimately lessened lipid uptake and accumulation in hepatocytes.
Collapse
Affiliation(s)
- Jia Li
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Peiran Li
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Xue Wu
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zibin Li
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Yunlong Li
- Institute of Functional Food Shanxi, Shanxi Agricultural University, Taiyuan, Shanxi, People's Republic of China
| | - Chao Liu
- Shandong Academy of Agricultural Sciences, Institute of Agro-Food Science and Technology, Jinan, Shandong, People's Republic of China
| | - Ji Bian
- University of Sydney, Royal North Shore Hospital, Kolling Institute, Sydney Medical School, New South Wales, Australia
| | - Lin Han
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Min Wang
- College of Food Science & Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| |
Collapse
|
13
|
Popa ML, Ichim C, Anderco P, Todor SB, Pop-Lodromanean D. MicroRNAs in the Diagnosis of Digestive Diseases: A Comprehensive Review. J Clin Med 2025; 14:2054. [PMID: 40142862 PMCID: PMC11943142 DOI: 10.3390/jcm14062054] [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: 02/11/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
MicroRNAs (miRNAs) have emerged as crucial regulators in digestive pathologies, including inflammatory bowel disease (miR-31, miR-155, and miR-21), colorectal cancer (miR-21, miR-598, and miR-494), and non-alcoholic fatty liver disease (miR-21, miR-192, and miR-122). Their capacity to modulate gene expression at the post-transcriptional level makes them highly promising candidates for biomarkers and therapeutic interventions. However, despite considerable progress, their clinical application remains challenging. Research has shown that miRNA expression is highly dynamic, varying across patients, disease stages, and different intestinal regions. Their dual function as both oncogenes and tumor suppressors further complicates their therapeutic use, as targeting miRNAs may yield unpredictable effects. Additionally, while miRNA-based therapies hold great potential, significant hurdles persist, including off-target effects, immune activation, and inefficiencies in delivery methods. The intricate interplay between miRNAs and gut microbiota adds another layer of complexity, influencing disease mechanisms and treatment responses. This review examined the role of miRNAs in digestive pathologies, emphasizing their diagnostic and therapeutic potential. While they offer new avenues for disease management, unresolved challenges underscore the need for further research to refine their clinical application.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (M.L.P.); (S.B.T.); (D.P.-L.)
| | - Paula Anderco
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (M.L.P.); (S.B.T.); (D.P.-L.)
| | | | | |
Collapse
|
14
|
Zhang J, Zhang Y, Qiu CX, Zeng W, Ruan Y, Gao Y, Ma W, Wu K, Zhang J, Cui J, Ye C, Liang J, Wang Z. Association of occupational noise exposure and shift work with non-alcoholic fatty liver disease: a cross-sectional study of male workers in the Chinese automobile manufacturing industry. BMJ Open 2025; 15:e085753. [PMID: 40074255 PMCID: PMC11904356 DOI: 10.1136/bmjopen-2024-085753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE This study aimed to determine the relationship between occupational noise, shift work and non-alcoholic fatty liver disease (NAFLD) in male workers in the automobile manufacturing industry. DESIGN Cross-sectional study. SETTING This study was carried out at the Guangzhou Twelfth People's Hospital using data from April to September 2022. PARTICIPANTS A total of 4672 eligible participants were included in the study. PRIMARY AND SECONDARY OUTCOME MEASURES Diagnosis of NAFLD was made using ultrasound. Noise was detected according to the Measurement of Physical Factors in the Workplace-Part 8: Noise. Environmental noise intensity was assessed using an EDGE personal noise dosimeter manufactured by CASELLA (UK). The working status of workers was investigated by questionnaire. RESULTS The OR of NAFLD was 1.39 (1.03, 1.88) in the cumulative noise exposure (CNE)≥95 group compared with CNE<85 group. Improved risk of NAFLD in workers with shift work compared with those without shift work (OR=1.35, 95% CI: 1.09, 1.68). As stratified analyses showed, the ORs of NAFLD prevalence related to occupational noise and shift work exposure appear to be increased in young workers. When both shift work and noise exposure work are present simultaneously, the synergy index between them was 0.47 (95% CI: 0.25, 0.89). Combined effects analysis revealed that the OR of NAFLD was 2.02 (95% CI: 1.34, 2.99) in CNE≥95 and cumulative length of night shifts work>2920 hours. CONCLUSION Occupational noise exposure may be an independent risk factor for NAFLD. It may synergistically affect disease when combined with night shift work, particularly among younger workers. These findings underscore the importance for companies to prioritise the management and training of younger workers, along with targeted occupational health education initiatives, as crucial measures for reducing the incidence of NAFLD.
Collapse
Affiliation(s)
- Jinwei Zhang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yuxia Zhang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Cong Xi Qiu
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Wenfeng Zeng
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yanmei Ruan
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yunxia Gao
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiyu Ma
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Kangyong Wu
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingwen Zhang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiaxin Cui
- School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Cuiping Ye
- Department of Preventive Health, Guangzhou Red Cross Hospital, Guangzhou, Guangdong, China
| | - Jiabin Liang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Zhi Wang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Jadhav PA, Thomas AB, Pathan MK, Chaudhari SY, Wavhale RD, Chitlange SS. Unlocking the therapeutic potential of unexplored phytocompounds as hepatoprotective agents through integration of network pharmacology and in-silico analysis. Sci Rep 2025; 15:8425. [PMID: 40069278 PMCID: PMC11897136 DOI: 10.1038/s41598-025-92868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Liver diseases account for over two million deaths annually, amounting to 4% of mortality worldwide, underscoring the need for development of novel preventive and therapeutic strategies. The growing interest in natural hepatoprotective agents highlights the potential of traditional medicine for modern drug discovery, though unlocking their molecular complexity requires advanced tools. This study integrates cutting-edge computational techniques with traditional herbal knowledge to identify potential hepatoprotective compounds. Protein targets implicated in liver disorders were identified through network pharmacology and by leveraging the rich molecular diversity inherent in herbal compounds, phytocompounds were selected. The Gene Ontology, Kyoto Encyclopedia of Genes and Genome data were compiled and enrichment analysis was performed using the DAVID database. Molecular docking of selected phytocompounds with top five protein targets helped identify 14 compounds which were employed for building the pharmacophore model. In virtual screening, among 1089 compounds screened, 10 compounds were identified as potential hits based on their predicted scores and alignment with pharmacophore features. The interactions of resulting hits were then analyzed through redocking studies and validated through molecular dynamics simulation and ADMET studies. Notably, (2S,5E)-2-(3,4-Dihydroxybenzyl)-6-(3,4-dihydroxyphenyl)-4-oxo-5-hexenoic acid and 5'-hydroxymorin emerged as lead compounds for further investigation. Both compounds exhibited significant binding affinities with specific amino acids in selected targets, suggesting their potential to modulate key pathways involved in hepatic disorders. Our findings demonstrate the utility of this integrated approach which transits beyond traditional trial-and-error methods. This approach will accelerate the discovery of novel hepatoprotective compounds, providing deeper insights into their mechanistic pathways and action.
Collapse
Affiliation(s)
- Pranali A Jadhav
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| | - Asha B Thomas
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India.
| | | | - Somdatta Y Chaudhari
- Department of Pharmaceutical Chemistry, PES's Modern College of Pharmacy, Nigdi, Pune, Maharashtra, India
| | - Ravindra D Wavhale
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| | - Sohan S Chitlange
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, 411 018, India
| |
Collapse
|
16
|
Yang Y, Luo Y, Shi J, Yin Y, Du X, Guo J, Zhuang H. The triglyceride glucose-waist circumference is the best indicator for screening non-alcoholic fatty liver disease in middle-aged and elderly people. NUTR HOSP 2025. [PMID: 40066565 DOI: 10.20960/nh.05367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND this investigation aimed to assess the correlation between the triglyceride glucose (TyG) index and its related indicators, as well as the ratio of triglyceride to high-density lipoprotein cholesterol (TG/HDL-c), with hepatic steatosis and liver fibrosis among middle-aged and elderly participants. METHODS based on data from the 2017-2020 National Health and Nutrition Examination Survey, the study included adults of ages 40 years and older in the United States. To explore the correlation between TyG and its related indicators, as well as TG/HDL-c with hepatic steatosis and liver fibrosis, multiple regression models were employed. In addition, the receiver operating characteristic curves were used to further explore the diagnostic efficacy of these indicators in non-alcoholic fatty liver disease (NAFLD) and liver fibrosis. RESULTS following the adjustment for various possible covariates, TyG, triglyceride glucose-body mass index (TyG-BMI), triglyceride glucose-waist circumference (TyG-WC), as well as TG/HDL-c were positively correlated with controlled attenuation parameter and NAFLD, with corresponding β coefficients of 17.90, 0.19, 0.20, and 1.57, alongside odds ratios of 2.10, 1.01, 1.01, and 1.15, respectively (all p < 0.05). The β coefficient for the association between TyG and liver stiffness measurement was -0.43 (p = 0.023). Notably, the area under the curve (AUC) of TyG-WC was the highest of all parameters, showing strong diagnostic potential for identifying NAFLD (AUC = 0.79) and liver fibrosis (AUC = 0.75). CONCLUSIONS this study reveals a significant positive correlation between TyG-WC and the prevalence of NAFLD in middle-aged and elderly people in the United States. These findings highlight that lowering TyG-WC levels may help reduce the incidence of NAFLD in middle-aged and older Americans.
Collapse
Affiliation(s)
- Yin Yang
- Department of Medical Ultrasound. West China Hospital. Sichuan University
| | - Yuan Luo
- Department of Medical Ultrasound. West China Tianfu Hospital. Sichuan University
| | - Jinchun Shi
- Affiliated Hospital of North Sichuan Medical College
| | - Yunyu Yin
- Affiliated Hospital of North Sichuan Medical College
| | - Xiangyu Du
- Department of Liver Surgery. West China Hospital. Sichuan University
| | - Jia Guo
- Department of Pancreatitis Center. West China Hospital. Sichuan University
| | - Hua Zhuang
- Department of Medical Ultrasound. West China Hospital. Sichuan University
| |
Collapse
|
17
|
Chen Y, Yang F, Wang Y, Shi Y, Liu L, Luo W, Zhou J, Yan Y. Mesenchymal stem cell-derived small extracellular vesicles reduced hepatic lipid accumulation in MASLD by suppressing mitochondrial fission. Stem Cell Res Ther 2025; 16:116. [PMID: 40045380 PMCID: PMC11884000 DOI: 10.1186/s13287-025-04228-2] [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: 11/22/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease characterized by lipid accumulation in liver cells. Human umbilical cord mesenchymal stem cell-derived small extracellular vesicles (MSC-sEV) have great potential in repairing and regenerating liver diseases. However, it is still unclear whether MSC-sEV can inhibit hepatocyte lipid accumulation by regulating mitochondrial fission. METHODS We investigated the effects of MSC-sEV on mitochondrial fission and its potential mechanism in lipotoxic hepatocytes and high-fat diet (HFD)-induced MASLD mice. RESULTS We found that MSC-sEV can effectively inhibit the expression of the Dynamin-related protein 1 (DRP1), thereby reducing mitochondrial fission, mitochondrial damage, and lipid deposition in lipotoxic hepatocytes and livers of HFD-induced MASLD in mice. Further mechanistic studies revealed that RING finger protein 31 (RNF31) played a crucial role in mediating the inhibitory effect of MSC-sEV on DRP1 and mitochondrial fission. RNF31 can suppress DRP1 expression and mitochondrial fission, thereby improving mitochondrial dysfunction and reducing hepatocyte lipid deposition. These findings suggest that MSC-sEV may downregulate hepatocyte DRP1-mediated mitochondrial fission by transporting RNF31, ultimately inhibiting hepatocyte lipid accumulation. CONCLUSIONS The insights from this study provide a new perspective on the mechanism of MSC-sEV in reducing lipid accumulation and offer a potential therapeutic target by targeting DRP1 to inhibit hepatocyte steatosis and the progression of MASLD.
Collapse
Affiliation(s)
- Yifei Chen
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Fuji Yang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yanjin Wang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yujie Shi
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Likang Liu
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Wei Luo
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China
- Changzhou Key Laboratory of Exosome Foundation and Transformation Application, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, China
| | - Jing Zhou
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China
- Changzhou Key Laboratory of Exosome Foundation and Transformation Application, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213017, China.
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou, 213017, China.
- Changzhou Key Laboratory of Exosome Foundation and Transformation Application, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University, Changzhou, 213017, China.
| |
Collapse
|
18
|
Wang Q, Tang X, Wang Y, Zhang D, Li X, Liu S. The role of extracellular vesicles in non-alcoholic steatohepatitis: Emerging mechanisms, potential therapeutics and biomarkers. J Adv Res 2025; 69:157-168. [PMID: 38494073 PMCID: PMC11954800 DOI: 10.1016/j.jare.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH), an emerging global healthcare problem, has become the leading cause of liver transplantation in recent decades. No effective therapies in the clinic have been proven due to the incomplete understanding of the pathogenesis of NASH, and further studies are expected to continue to delve into the mechanisms of NASH. Extracellular vesicles (EVs), which are small lipid membrane vesicles carrying proteins, microRNAs and other molecules, have been identified to play a vital role in cell-to-cell communication and are involved in the development and progression of various diseases. In recent years, there has been increasing interest in the role of EVs in NASH. Many studies have revealed that EVs mediate important pathological processes in NASH, and the role of EVs in NASH is distinct and variable depending on their origin cells and target cells. This review outlines the emerging mechanisms of EVs in the development of NASH and the preclinical evidence related to stem cell-derived EVs as a potential therapeutic strategy for NASH. Moreover, possible strategies involving EVs as clinical diagnostic, staging and prognostic biomarkers for NASH are summarized.
Collapse
Affiliation(s)
- Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiangning Tang
- Department of endocrinology, the Second Affiliated Hospital of University of South China, 421001 Hunan Province, China
| | - Yu Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Danyi Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
19
|
Jin M, Lu Q, Xia N, Fan X, Zhang Z, Huang X, Sun L, Zhang L, Jiang Z, Yu Q. LncRNA Gm35585 transcriptionally activates the peroxidase EHHADH against diet-induced fatty liver. Exp Mol Med 2025; 57:652-666. [PMID: 40082671 PMCID: PMC11958773 DOI: 10.1038/s12276-025-01420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/13/2024] [Accepted: 01/01/2025] [Indexed: 03/16/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease is one of the most common chronic liver diseases worldwide and has no approved treatment thus far. Here we report that the hepatic overexpression of Gm35585, a novel lncRNA downregulated in the livers of mice fed a high-fat diet, is functionally important in alleviating hepatic lipid accumulation pathologies. Gm35585 activates the peroxisome proliferator-activated receptor α (PPARα) signaling pathway and promotes the expression of downstream PPARα-target gene, enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH), which is one of the four enzymes of the peroxisomal β-oxidation pathway. Activation of EHHADH promotes the oxidation of long-chain fatty acids (LCFAs), and the increased levels of hepatic LCFAs contribute to metabolic-dysfunction-associated steatotic liver disease. Mechanistically, Gm35585 binds to retinoid X receptor α (RXRα) and then forms a PPARα/RXRα heterodimer with PPARα and guides the heterodimer to recognize the promoter of EHHADH, which is called peroxisome proliferator-activated receptor response element, causing transcriptional activation of EHHADH. Taken together, Gm35585 is a hepatic lipid metabolism regulator that activates EHHADH transcription, promoting peroxisomal β-oxidation of LCFAs and ultimately ameliorating diet-induced fatty liver.
Collapse
Affiliation(s)
- Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qian Lu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ninglin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xue Fan
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ziling Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaofei Huang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Li Sun
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China.
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
20
|
Chen J, Wang P, Li Z, Wu J, Tang F, Yang N, Cen B, Xie C, Yang Y, Yang Z, Zhang C, Yao X, Xu Z. An in vitro 3D spheroid model with liver steatosis and fibrosis on microwell arrays for drug efficacy evaluation. J Biotechnol 2025; 399:153-163. [PMID: 39889902 DOI: 10.1016/j.jbiotec.2025.01.019] [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: 12/04/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is now the most common chronic liver disease worldwide, affecting more than 30 percent of adults. The most severe form of MASLD, metabolic dysfunction-associated steatohepatitis (MASH), is characterized by necrotizing inflammation and rapid fibrosis progression, often leading to cirrhosis and hepatocellular carcinoma. Currently, only Resmetirom is approved for the treatment of MASH one of the main reasons is the absence of representative in vivo or in vitro models for MASH. To address this challenge, we developed a high-throughput 3D spheroid model consisting of human hepatocellular carcinoma cells (HepG2) and human hepatic stellate cells (LX-2) on microwell arrays. This model, induced with free fatty acids (FFA) to simulate steatosis and fibrosis, enables the assessment of efficacy and mechanisms for potential anti-MASH drugs. Our findings demonstrate that this in vitro spheroid model replicates key pathological features of human MASLD, including steatosis, oxidative stress, and fibrosis. Upon validation, we selected pirfenidone (PFD) and yinfenidone (AC-003), which are commonly used to treat idiopathic pulmonary fibrosis (IPF), to test their anti-MASH efficacy. Treatment with these drugs showed that they could regulate lipid synthesis and metabolism genes, reduce lipid accumulation, oxidative stress, and fibrosis levels. This 3D spheroid model represents a straightforward and efficient tool for screening anti-MASH drugs and investigating the molecular mechanisms of drug action.
Collapse
Affiliation(s)
- Jiamin Chen
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China; Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong Province 518000, China
| | - Ping Wang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Zhanpeng Li
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Jieyi Wu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Fang Tang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Niao Yang
- Department of pharmacy, The Second Naval Hospital of Southern Theater Command, Sanya, Hainan Province 572000, China
| | - Bohong Cen
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Cuiyin Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yufan Yang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Ziyan Yang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Chuwen Zhang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xiangcao Yao
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
21
|
Bayram S, Ülger Y. Association of PNPLA3 rs738409 C > G and rs2896019 T > G Polymorphisms with Nonalcoholic Fatty Liver Disease in a Turkish Population from Adıyaman Province. Genet Test Mol Biomarkers 2025; 29:63-73. [PMID: 40101239 DOI: 10.1089/gtmb.2024.0481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Objectives: The purpose of this study was to investigate the effect of patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 C > G and rs2896019 T > G polymorphisms on genetic susceptibility to nonalcoholic fatty liver disease (NAFLD) in a Turkish population from Adıyaman province, located in the Southeast Anatolia Region of Turkey. Materials and Methods: This hospital-based molecular epidemiological case-control study analyzed the PNPLA3 rs738409 C > G and rs2896019 T > G polymorphisms in 335 NAFLD cases and 410 healthy controls. Genotype frequencies were determined using real-time polymerase chain reaction with the TaqMan assay. The association with NAFLD susceptibility was evaluated by calculating odds ratios (ORs) and 95% confidence intervals (CIs) using an unconditional logistic regression model. Results: We found that the PNPLA3 rs738409 C > G (CC vs. GG: OR = 1.90, 95% CI = 1.05-3.44) and rs2896019 T > G (TT vs. GG: OR = 3.24, 95% CI = 1.44-7.27) polymorphisms were linked to an increased risk of NAFLD in almost all genetic models (p < 0.05). In addition, the PNPLA3 Grs738409/Grs2896019 haplotype was associated with NAFLD development (p < 0.05). Significant differences in alanine aminotransferase and aspartate aminotransferase enzyme levels were observed across the genotypes of these polymorphisms (p < 0.05). Conclusion: This is the first study on PNPLA3 single nucleotide polymorphisms (SNPs) and NAFLD in the Turkish population of Adıyaman Province, Southeast Anatolia. Our findings suggest that the PNPLA3 rs738409 C > G and rs2896019 T > G polymorphisms, along with their haplotypes, may influence NAFLD susceptibility. Further independent studies with larger sample sizes and diverse populations are needed to confirm these results.
Collapse
Affiliation(s)
- Süleyman Bayram
- Faculty of Health Sciences, Department of Public Health Nursing, Adıyaman University, Adıyaman, Turkey
| | - Yakup Ülger
- Faculty of Medicine, Department of Gastroenterology, Çukurova University, Adana, Turkey
| |
Collapse
|
22
|
Theel WB, de Jong VD, Castro Cabezas M, Grobbee DE, Jukema JW, Trompet S. Risk of cardiovascular disease in elderly subjects with obesity and liver fibrosis and the potential benefit of statin treatment. Eur J Clin Invest 2025; 55:e14368. [PMID: 39636216 PMCID: PMC11810556 DOI: 10.1111/eci.14368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Liver fibrosis progression is influenced by older age and cardiometabolic risk factors such as obesity and is associated with an increased risk of cardiovascular events. While statins may protect against cardiovascular complications, their effects in elderly individuals with obesity and liver fibrosis are unclear. METHOD The PROspective Study of Pravastatin in the Elderly at Risk (PROSPER) database was used to evaluate the effect of pravastatin on major adverse cardiovascular events in an elderly population (>70 years). Subjects were categorized by BMI: lean (<25 kg/m2), overweight (25-29.9 kg/m2) and obese (≥30 kg/m2). Liver fibrosis was assessed using the FIB-4 index: low risk (<2.0), intermediate risk (2.0-2.66) and high risk (≥2.67). Time-to-event data were analysed using the Cox proportional hazards model, adjusted for confounders and compared the placebo and pravastatin groups. RESULTS A total of 5.804 subjects were included. In the placebo group, the highest risk group (high FIB-4 and obesity) had a significantly higher hazard ratio for (non-)fatal stroke (HR 2.74; 95% CI 1.19-6.29) compared to the low FIB-4, lean BMI group. This risk disappeared in the same pravastatin group. Pravastatin did not affect other cardiovascular endpoints. All-cause mortality was significantly higher in subjects with lean weight and high FIB-4 on placebo (HR 1.88; 95% CI 1.14-3.11), but not on pravastatin (HR .58; 95% CI .28-1.20). CONCLUSION Elderly individuals with obesity and liver fibrosis are at higher risk for (non-)fatal stroke, which is reduced with pravastatin. Pravastatin also potentially lowers all-cause mortality in subjects with lean weight and liver fibrosis.
Collapse
Affiliation(s)
- Willy B. Theel
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Obesity Center CGGRotterdamThe Netherlands
| | - Vivian D. de Jong
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Manuel Castro Cabezas
- Department of Internal medicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Julius ClinicalZeistThe Netherlands
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Diederick E. Grobbee
- Julius Global Health, Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
- Julius ClinicalZeistThe Netherlands
| | - Johan W. Jukema
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Stella Trompet
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Internal Medicine, Section of Gerontology & GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
23
|
Cui J, Zhao G, Xie W, Yang Y, Fu X, Meng H, Liu H, Tan M, Chen D, Rong C, Wang Y, Wang Y, Zhang LW. Exacerbated hepatotoxicity in in vivo and in vitro non-alcoholic fatty liver models by biomineralized copper sulfide nanoparticles. BIOMATERIALS ADVANCES 2025; 168:214117. [PMID: 39580989 DOI: 10.1016/j.bioadv.2024.214117] [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: 06/28/2024] [Revised: 09/19/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024]
Abstract
Copper sulfide nanoparticles (NPs) synthesized through biomineralization have significant commercial potential as photothermal agents, while the safety evaluation of these NPs, especially for patients with non-alcoholic fatty liver (NAFL), remains insufficient. To explore the differential hepatotoxicity of copper sulfide NPs in NAFL conditions, we synthesized large-sized (LNPs, 15.1 nm) and small-sized (SNPs, 3.5 nm) BSA@Cu2-xS NPs. A NAFL rat model fed with high fat diet (HFD) was successfully established for a 14-day subacute toxicity study by daily repeated administration of BSA@Cu2-xS NPs. Our findings from serum biochemistry and histopathological examinations revealed that copper sulfide at both sizes NPs induced more pronounced liver damage in NAFL rats than rats fed with normal diet. Transcriptome sequencing analysis showed that LNPs activated inflammation and DNA damage repair pathways in the livers of NAFL rats, while SNPs displayed minimal inflammation. A three-dimensional spheroid model of NAFL developed in our in-house cell spheroid culture honeycomb chips demonstrated that LNPs, but not SNPs, triggered a distinct release of inflammatory factors and increased reactive oxygen species through Kupffer cells. These results highlight that NAFL condition exacerbated the hepatotoxicity of BSA@Cu2-xS NPs, with SNPs emerging as safer photothermal agents compared to LNPs, suggesting superior potential for clinical applications.
Collapse
Affiliation(s)
- Jinbin Cui
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Gang Zhao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Xie
- The College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, Zhejiang, China
| | - Yang Yang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xing Fu
- Suzhou Vivoid Biotechnology Co., Ltd, Suzhou 215124, China
| | - Hezhang Meng
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - He Liu
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mengfei Tan
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Dandan Chen
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Chao Rong
- Department of Pathology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Yangyun Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yong Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Leshuai W Zhang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
24
|
Nakatsuka T, Tateishi R, Sato M, Hashizume N, Kamada A, Nakano H, Kabeya Y, Yonezawa S, Irie R, Tsujikawa H, Sumida Y, Yoneda M, Akuta N, Kawaguchi T, Takahashi H, Eguchi Y, Seko Y, Itoh Y, Murakami E, Chayama K, Taniai M, Tokushige K, Okanoue T, Sakamoto M, Fujishiro M, Koike K. Deep learning and digital pathology powers prediction of HCC development in steatotic liver disease. Hepatology 2025; 81:976-989. [PMID: 38768142 PMCID: PMC11825480 DOI: 10.1097/hep.0000000000000904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/05/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND AND AIMS Identifying patients with steatotic liver disease who are at a high risk of developing HCC remains challenging. We present a deep learning (DL) model to predict HCC development using hematoxylin and eosin-stained whole-slide images of biopsy-proven steatotic liver disease. APPROACH AND RESULTS We included 639 patients who did not develop HCC for ≥7 years after biopsy (non-HCC class) and 46 patients who developed HCC <7 years after biopsy (HCC class). Paired cases of the HCC and non-HCC classes matched by biopsy date and institution were used for training, and the remaining nonpaired cases were used for validation. The DL model was trained using deep convolutional neural networks with 28,000 image tiles cropped from whole-slide images of the paired cases, with an accuracy of 81.0% and an AUC of 0.80 for predicting HCC development. Validation using the nonpaired cases also demonstrated a good accuracy of 82.3% and an AUC of 0.84. These results were comparable to the predictive ability of logistic regression model using fibrosis stage. Notably, the DL model also detected the cases of HCC development in patients with mild fibrosis. The saliency maps generated by the DL model highlighted various pathological features associated with HCC development, including nuclear atypia, hepatocytes with a high nuclear-cytoplasmic ratio, immune cell infiltration, fibrosis, and a lack of large fat droplets. CONCLUSIONS The ability of the DL model to capture subtle pathological features beyond fibrosis suggests its potential for identifying early signs of hepatocarcinogenesis in patients with steatotic liver disease.
Collapse
Affiliation(s)
- Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Natsuka Hashizume
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Ami Kamada
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Hiroki Nakano
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Yoshinori Kabeya
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Sho Yonezawa
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Rie Irie
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshio Sumida
- Department of Internal Medicine, Division of Hepatology and Pancreatology, Aichi Medical University, Aichi, Japan
| | - Masashi Yoneda
- Department of Internal Medicine, Division of Hepatology and Pancreatology, Aichi Medical University, Aichi, Japan
| | - Norio Akuta
- Department of Hepatology, Toranomon Hospital and Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Takumi Kawaguchi
- Department of Medicine, Division of Gastroenterology, Kurume University School of Medicine, Fukuoka, Japan
| | | | - Yuichiro Eguchi
- Liver Center, Saga University Hospital, Saga, Japan
- Loco Medical General Institute, Saga, Japan
| | - Yuya Seko
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Eisuke Murakami
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Collaborative Research Laboratory of Medical Innovation, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Hiroshima Institute of Life Sciences, Hiroshima, Japan
| | - Makiko Taniai
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsutoshi Tokushige
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology, Saiseikai Suita Hospital, Suita, Osaka, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Medicine, Kanto Central Hospital, Tokyo, Japan
| |
Collapse
|
25
|
Guo Y, Huang H, Yang L, Shen Q, Liu Z, Wang Q, Chen S, Pan J, Zhai H, Li Y, Xu L, Yu C, Xu C. Amyloid precursor protein promotes MASH progression by upregulating death receptor 6-mediated hepatocyte apoptosis. J Biol Chem 2025; 301:108285. [PMID: 39938799 PMCID: PMC11923821 DOI: 10.1016/j.jbc.2025.108285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a complicated process that contributes to end-stage liver disease and, eventually, hepatocellular carcinoma. Hepatocyte apoptosis, a well-defined form of cell death in MASH, is considered the primary cause of liver inflammation and fibrosis. However, the mechanisms underlying the regulation of hepatocyte apoptosis in MASH remain largely unclear. We explored the proapoptotic effect of hepatocyte amyloid precursor protein (APP) in MASH. C57BL/6J mice were fed a Western diet plus sugar water, a high-fat high-fructose diet, or a methionine and choline deficiency diet to induce MASH. APP expression was analyzed in murine MASH specimens. App-/- mice and mice with adeno-associated virus-mediated APP overexpression were established to study the role of APP in MASH. Palmitic acid was used to mimic lipotoxicity-induced MASH in AML12 cells. We identified a dramatic increase in APP expression in hepatocytes of patients with MASH and three different mouse models. Suppression of APP attenuated hepatic steatosis, inflammation, and fibrosis in MASH mice, whereas its restoration activated MASH pathogenesis. Furthermore, increased death receptor 6 (DR6) was observed in MASH mouse livers. Mechanistically, APP interacted with DR6, a tumor necrosis factor receptor, to facilitate DR6 expression and activation. Activated DR6 increased apoptosis in hepatocytes, which was associated with an increase in proapoptotic effectors (cleaved-caspase 3/7). Our results highlight the role of the APP-DR6 axis in hepatocyte apoptosis, inflammation activation, and fibrosis formation in murine MASH model, providing new insights into therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hangkai Huang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Yang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qien Shen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhening Liu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qinqiu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenghui Chen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Pan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoliang Zhai
- Department of Gastroenterology, Haining Branch, The First Affiliated Hospital, Zhejiang University School of Medicine, Haining, China
| | - Youming Li
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
26
|
Hong J, Xu Z, Xu F, Wu H, Liu J, Qu L. Immune-related diagnostic indicators and targeted therapies for COPD combined with NASH were identified and verified via WGCNA and LASSO. Front Immunol 2025; 16:1514422. [PMID: 40093012 PMCID: PMC11906333 DOI: 10.3389/fimmu.2025.1514422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The incidence of chronic obstructive pulmonary disease (COPD) and non-alcoholic fatty liver disease (NAFLD) has increased significantly in past decades, posing a significant public health burden. An increasing amount of research points to a connection between COPD and NAFLD. This study aimed to identify the key genes of these two diseases, construct a diagnostic model, and predict potential therapeutic agents based on critical genes. Methods NAFLD and COPD datasets were obtained from the GEO database, differential genes were identified by differential analysis and WGCNA, PPI networks were constructed and enriched for differential genes and COPD-associated secreted proteins, small molecule compounds were screened, and immune cell infiltration was assessed. Meanwhile, LASSO and RF further screened the essential genes, and finally, two key genes were obtained. Subsequently, the nomogram diagnostic model and lncRNA-miRNA-mRNA network were constructed based on these two core genes, subjected to drug prediction and GSEA enrichment analysis, and validated in an external cohort using qRT-PCR. Results KEGG enrichment analysis indicated that the NF-kappa B and TNF signaling pathways may be associated with COPD and NASH co-morbidities. Ten small-molecule drugs associated with COPD and NASH were identified through cMAP analysis, including ansoprazole and atovaquone. In addition, we further identified the hub genes S100A9 and MYH2 for NAFLD and COPD by machine learning methods. The immune infiltration indicated that these two core genes might be involved in the immunomodulatory process of NASH by regulating the function or recruitment of specific immune cell types. A nomogram diagnostic model was constructed based on these two core genes. The AUC value for S100A9 was 0.887, for MYH2 was 0.877, and for the nomogram was 0.889, demonstrating excellent diagnostic efficacy. Two hundred fifty-four potential drugs targeting S100A9 and 67 MYH2 were searched in the DGIdb database. Meanwhile, the lncRNA-miRNA-mRNA network was constructed by predicting target miRNAs of biomarkers and further predicting lncRNAs targeting miRNAs. qRT-PCR analysis revealed that S100A9 was upregulated in both COPD and NAFLD, consistent with bioinformatic predictions, while MYH2 showed increased expression in COPD but decreased expression in NAFLD, diverging from the predicted downregulation in both diseases. These findings suggest that S100A9 serves as a common inflammatory marker for both diseases, whereas MYH2 may be regulated by disease-specific mechanisms, highlighting its potential for distinguishing COPD from NAFLD. Conclusion The hub genes S100A9 and MYH2 in COPD and NASH were identified by various bioinformatics methods and a diagnostic model was constructed to improve the diagnostic efficiency. We also revealed some potential biological mechanisms of COPD and NASH and potential drugs for COPD-related NASH. Our findings provide potential new diagnostic and therapeutic options for COPD-associated NASH and may help reduce its prevalence.
Collapse
Affiliation(s)
- Jianwei Hong
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zikai Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Fangrui Xu
- Department of Medical Imaging, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haifeng Wu
- Department of Emergency Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China
| | - Jinxia Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Lishuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
27
|
Pădureanu V, Forțofoiu MC, Pîrșcoveanu M, Pădureanu R, Rădulescu D, Donoiu I, Pîrșcoveanu DFV. Cardiovascular Manifestations of Patients with Non-Alcoholic Fatty Liver Disease. Metabolites 2025; 15:149. [PMID: 40137114 PMCID: PMC11943630 DOI: 10.3390/metabo15030149] [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: 01/11/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD), more recently redefined as metabolic-associated fatty liver disease (MAFLD), is now recognized as the most prevalent cause of chronic liver disease. Its strong association with cardiovascular disease (CVD) underscores its emerging role in global morbidity and mortality. Objective: This review critically examines the pathophysiological mechanisms that link NAFLD/MAFLD with CVD. It focuses on shared metabolic disturbances, inflammatory pathways, and alterations in the gut microbiota that contribute to hepatic and cardiovascular pathology. Review and Gaps: Current evidence highlights insulin resistance, dyslipidemia, systemic inflammation, and gut dysbiosis as pivotal factors connecting NAFLD/MAFLD to CVD. Despite these insights, inconsistencies in diagnostic criteria and a lack of validated non-invasive biomarkers hinder a clear understanding of the causal relationship between liver and cardiovascular diseases. Conclusions: Addressing these knowledge gaps through standardized diagnostic protocols and large-scale longitudinal studies is essential. Improved biomarker validation and clearer delineation of the underlying mechanisms will improve cardiovascular risk stratification and enable more personalized therapeutic strategies for patients with NAFLD/MAFLD.
Collapse
Affiliation(s)
- Vlad Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Mircea Cătălin Forțofoiu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Mircea Pîrșcoveanu
- Department of Surgery, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Rodica Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania; (V.P.); (M.C.F.)
| | - Dumitru Rădulescu
- Department of Surgery, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Ionuț Donoiu
- Department of Cardiology, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | | |
Collapse
|
28
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025:S1550-4131(25)00019-1. [PMID: 40015280 DOI: 10.1016/j.cmet.2025.01.020] [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: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
29
|
Li J, Li C, Wu X, Dong Y, Li Y, Jiao X, Li J, Han L, Wang M. Protocatechuic Acid Suppresses Lipid Uptake and Synthesis through the PPARγ Pathway in High-Fat Diet-Induced NAFLD Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4012-4026. [PMID: 39907525 DOI: 10.1021/acs.jafc.4c01354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become one of the most concerning health problems in the world. Dietary intervention is an effective way to prevent and improve NAFLD. As one of the main metabolites of anthocyanins, protocatechuic acid (PCA) exhibited strong activity to improve NAFLD, but the specific mechanism remains unclear. Currently, proteomics has been used to identify that PCA treatment could significantly influence the expression of 224 proteins, including 89 downregulated proteins and 135 upregulated proteins. KEGG analysis showed that PCA obviously inhibited the peroxisome proliferator-activated receptor (PPAR) signaling pathway. Immunofluorescence and Western blot analyses further confirmed that PCA repressed the protein expression of PPARγ and subsequently inhibited the expression of free fatty acid (FFA) uptake proteins (CD36 and FABP2) and FFA synthesis proteins (ACC and FASN), respectively. These effects of PCA contributed to the inhibitory activity of excessive lipid accumulation in the liver. Our results highlighted that PCA could effectively alleviate high-fat diet-induced (HFD) NAFLD by inhibiting lipid absorption and synthesis through the PPARγ signaling pathway.
Collapse
Affiliation(s)
- Jia Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Chaoyue Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Xue Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Yonghui Dong
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Yunlong Li
- Institute of Functional Food of Shanxi, Shanxi Agricultural University, Taiyuan 030006, P. R. China
| | - Xiaowen Jiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Jiating Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Lin Han
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| | - Min Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| |
Collapse
|
30
|
Amer J, Abdoh Q, Salous Z, Alsoud EA, AbuBaker S, Salhab A, Badrasawi M. A cross-sectional study of risk factors associated with sarcopenia in patients with metabolic dysfunction-associated steatotic liver disease. Front Med (Lausanne) 2025; 12:1488068. [PMID: 40041458 PMCID: PMC11876153 DOI: 10.3389/fmed.2025.1488068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease linked to several adverse health consequences that include metabolic disturbances affecting skeletal muscle. Sarcopenia, characterized by skeletal muscle loss, is commonly observed in individuals with MASLD. Our study aimed to identify modifiable lifestyle factors associated with sarcopenia in patients with MASLD. Methods This study was a cross-sectional study that was conducted in three clinics in Nablus. A total of 162 adults diagnosed with MASLD were recruited for the study. The patients were interviewed and instructed to provide the necessary information, such as sociodemographic factors, medical and surgical history, lifestyle information, MASLD-related data, and nutritional and functional status. Sarcopenia was defined using the Foundation for the National Institutes of Health (FNIH) criteria, which includes the weight-adjusted skeletal muscle index (wSMI) with the cut-off scores (male subjects: 35.7% and female subjects: 30.7%). Statistical analysis was conducted using SPSS v.21. A chi-squared or independent samples t-test was utilized to identify the factors linked to sarcopenia in the study sample. Results Our data found that 44% of MASLD patients had sarcopenia. This condition was significantly associated with female gender (p < 0.0001), older age (p < 0.0001), presence of chronic diseases (p < 0.035), and medication use (p < 0.05). Regarding nutritional factors, sarcopenia had a significant association with obesity, a higher body fat percentage, a high waist-to-hip ratio, a low mid-upper arm circumference, and a reduced calf circumference (p < 0.001). Sarcopenic individuals often exhibit reduced handgrip strength. Lifestyle factors such as a history of smoking and the type of smoking were found to be positively associated with sarcopenia (p < 0.0001). Conclusion Sarcopenia was prevalent in the study population and was linked to modifiable risk factors that can be managed to reduce its progression. Future research using different study designs, such as longitudinal design, is recommended to identify the determinants of sarcopenia. Intervention studies are also required to improve the nutritional and functional status of MASLD patients.
Collapse
Affiliation(s)
- Johnny Amer
- Department of Allied Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Qusay Abdoh
- Department of Internal Medicine, GI and Endoscopy Unit, An-Najah National University Hospital, Nablus, Palestine
| | - Zaina Salous
- Department of Medical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Eithar Abu Alsoud
- Department of Medical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sama AbuBaker
- Department of Medical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Ahmad Salhab
- Department of Medical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Manal Badrasawi
- Nutrition and Food Technology Department, An-Najah National University, Nablus, Palestine
| |
Collapse
|
31
|
Shi D, Tan Q, Zhang Y, Qi X, Xu X, Xu G, Bai R, Deng J, Chen M, Jiang T, Mei Y. Serum uric acid trajectories and risk of metabolic dysfunction-associated steatotic liver disease in China: a 2019-2021 cohort health survey. BMC Public Health 2025; 25:653. [PMID: 39962427 PMCID: PMC11834244 DOI: 10.1186/s12889-024-21214-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/26/2024] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE To investigate the associations between serum uric acid (UA) trajectories and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in large cohort survey 2019-2021. METHODS This cohort health survey included 11,644 participants without MASLD before January 1, 2021. Among them, 5578 (47.90%) were men and 6066 (52.10%) were women. The group-based trajectory model method was applied to identify serum UA trajectories from January 1, 2019, to December 30, 2021. New-onset MASLD events in 2021 were treated as outcomes. A logistic regression model was used to assess associations between UA trajectories and incidence of MASLD. RESULTS Four distinct serum UA trajectories among both sexes were identified: "low-stable" trajectory 1 (n = 783 men; n = 1143 women), "moderate-moderate increasing" trajectory 2 (n = 2794 men; n = 3266 women), "moderate high-moderate increasing" trajectory 3 (n = 1660 men; n = 1464 women), and "high-increasing" trajectory 4 (n = 341 men; n = 193 women). During the 3-year follow-up period, 840 (15.06%) men and 408 (6.72%) women developed MASLD, respectively. Compared with the trajectory 1 group, the trajectory 4 group had the highest risk (odds ration [OR] 2.99 [95% confidence interval {CI} 1.70, 5.24] for men; OR 2.37 [95% CI 1.04, 5.33] for women), followed by the trajectory 3 (OR 2.23 [95% CI 1.52, 3.30] for men; OR 2.37 [95% CI 1.45, 3.92] for women) and trajectory 2 (OR 1.43 [95% CI 1.07, 1.94] for men; OR 1.37 [95% CI 0.93, 2.03] for women) groups. CONCLUSIONS High serum UA trajectories were independent risk factors for MASLD in both sexes, which is critical for informing prevention and treatment strategies in public health initiatives and clinical practice.
Collapse
Affiliation(s)
- Dan Shi
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Qilong Tan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
- School of Public Health, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yong Zhang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoya Qi
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoyang Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Guoqiong Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Ruixue Bai
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Jing Deng
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Mengxue Chen
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Tao Jiang
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| | - Ying Mei
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| |
Collapse
|
32
|
Li Z, Jin Y, Zhao H, Gu Y, Zhang Y, Cheng S, Zhang L, He P, Liu X, Jia Y. Aurantio-Obtusin Regulates Gut Microbiota and Serum Metabolism to Alleviate High-Fat Diet-Induced Obesity-Associated Non-Alcoholic Fatty Liver Disease in Mice. Phytother Res 2025. [PMID: 39953693 DOI: 10.1002/ptr.8459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive condition with limited effective treatments. This study investigated the therapeutic effects of Aurantio-obtusin (AO), a bioactive compound from Cassiae Semen, on obesity-associated NAFLD. An obesity-related NAFLD model was established in ApoE-/- mice fed a high-fat diet (HFD) for 24 weeks, with AO administered during the last 16 weeks. Mouse body weight, adipose tissue weights, liver weights, serum lipid levels, hepatic steatosis, inflammatory damage, and colonic tissue barrier integrity were evaluated. Gut microbial communities and serum metabolic profiles were analyzed using 16S rRNA sequencing and untargeted metabolomics. Hepatic lipid metabolism-related gene expression was assessed using molecular biology techniques. AO treatment significantly ameliorated HFD-induced adiposity, hyperlipidemia, and NAFLD symptoms. It preserved intestinal barrier integrity, modulated gut microbial composition by enriching beneficial taxa, and improved serum metabolic profiles. AO favorably adjusted hepatic lipid metabolism by upregulating PPARα and CPT1A while downregulating SREBP1, FASN, and SCD1. Correlation analysis revealed significant associations among gut microbial composition, serum metabolites, and disease indicators. AO's therapeutic benefits in NAFLD might be attributed to its ability to modulate gut microbial community composition and serum metabolic profile, enhance intestinal barrier function, and regulate hepatic lipid metabolism gene expression. AO presents a promising therapeutic agent for obesity-associated NAFLD, warranting further investigation into its potential clinical applications.
Collapse
Affiliation(s)
- Zhaoyong Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Jin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Huashan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyan Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yaxin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Saibo Cheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lifang Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Peikun He
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xiaoyu Liu
- Pingshan Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Pingshan District Peoples' Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Yuhua Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Rotaru A, Stafie R, Stratina E, Zenovia S, Nastasa R, Minea H, Huiban L, Cuciureanu T, Muzica C, Chiriac S, Girleanu I, Singeap AM, Sfarti C, Stanciu C, Trifan A. Lean MASLD and IBD: Exploring the Intersection of Metabolic Dysfunction and the Gut-Liver Axis. Life (Basel) 2025; 15:288. [PMID: 40003697 PMCID: PMC11856855 DOI: 10.3390/life15020288] [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: 01/17/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) challenges traditional paradigms by manifesting in lean individuals. The link between MASLD and inflammatory bowel disease (IBD) underscores the importance of the gut-liver axis in disease progression and chronic inflammation. This study evaluates MASLD prevalence, clinical characteristics, and diagnostic predictors in lean individuals with and without IBD. This prospective study included 387 lean patients. Hepatic steatosis and fibrosis were assessed using vibration-controlled transient elastography (VCTE). Anthropometric, clinical and biological data were compared. The subgroup analyses focused on MASLD patients with and without IBD. MASLD was present in 34.1% of lean individuals and 46.3% of those who were lean with IBD. MASLD patients had increased visceral adiposity (CUN-BAE: 31.21 ± 5.42 vs. 24.57 ± 6.49, p < 0.001) and metabolic dysfunction, including dyslipidemia and elevated fasting glucose. IBD-MASLD patients exhibited greater hepatic steatosis and systemic inflammation. CUN-BAE outperformed FLI and HSI in predicting liver steatosis, especially in IBD patients (AUC = 0.806). Lean MASLD, particularly in IBD patients, highlights the need for tailored diagnostic and management strategies. The gut-liver axis plays a key role in disease progression, and the CUN-BAE index demonstrates superior accuracy for identifying liver steatosis.
Collapse
Affiliation(s)
- Adrian Rotaru
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Remus Stafie
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Ermina Stratina
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Sebastian Zenovia
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Robert Nastasa
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Horia Minea
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Laura Huiban
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Tudor Cuciureanu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Cristina Muzica
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Stefan Chiriac
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Irina Girleanu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Ana-Maria Singeap
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Catalin Sfarti
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Carol Stanciu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| | - Anca Trifan
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.R.); (E.S.); (S.Z.); (R.N.); (H.M.); (L.H.); (T.C.); (C.M.); (I.G.); (A.-M.S.); (C.S.); (C.S.); (A.T.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iasi, Romania
| |
Collapse
|
34
|
Zhang N, Li J, Xie X, Hu Y, Chen H, Zhang Y, Liu Y, Zhu X, Xu H, Wang Z, Baima K, Zhang X, Qin Z, Yu Z, Xiao X, Zhao X. Changes in drinking levels and metabolic dysfunction-associated steatotic liver disease: a longitudinal study from the China multi-ethnic cohort study. BMC Public Health 2025; 25:556. [PMID: 39934719 DOI: 10.1186/s12889-025-21752-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Little is known about the associations of changes in drinking levels with the newly defined metabolic dysfunction-associated steatotic liver disease (MASLD). We therefore sought to estimate the associations between changes in drinking levels and MASLD in less developed regions of China. METHODS This longitudinal study included 8727 participants from the China Multi-Ethnic Cohort (CMEC) in less developed regions, all participating in baseline and a follow-up survey. MASLD was defined as hepatic steatosis, along with the presence of at least one of five cardiometabolic risks, in addition to limiting excessive alcohol consumption. We applied the parametric g-formula to evaluate the association between changes in drinking levels and MASLD. We further estimated the association between changes in drinking levels and fibrosis scores (AST-to-platelet ratio and fibrosis-4 index) in patients with MASLD. RESULTS Compared with sustained non-drinking, sustained modest drinking was associated with a higher risk of MASLD (Mean Ratio (MR): 1.127 [95% CI: 1.040-1.242]). Compared to sustained non-drinking, the MR for those transitioning from non-drinking to modest drinking was 1.065 [95% CI: 0.983-1.169], while the MR for those changing from modest drinking to non-drinking was 1.059 [95% CI: 0.965, 1.173]. Non-invasive fibrosis scores tended to increase with modest drinking compared to sustained non-drinking. CONCLUSION In the less developed regions of China, sustained moderate drinking was associated with the risk of MASLD compared with sustained non-drinking. Increased drinking showed a trend towards a higher risk of MASLD. This study can inform drinking policies related to MASLD and liver fibrosis in less developed regions.
Collapse
Affiliation(s)
- Ning Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jingzhong Li
- Tibet Center for Disease Control and Prevention, Lhasa, China
| | - Xiaofen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yifan Hu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hongxiang Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuan Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yujie Liu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xingren Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Sichuan University, Chengdu, China
| | - Zhenghong Wang
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
| | - Kangzhuo Baima
- High Altitude Health Science Research Center of Tibet University, Lhasa, Tibet, China
| | - Xuehui Zhang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Zixiu Qin
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Zhimiao Yu
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Xiong Xiao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Lei Y, Tao S, Yang Y, Xie F, Xie W. Association between prognostic nutritional index and all-cause mortality and cardiovascular disease mortality in American adults with non-alcoholic fatty liver disease. Front Nutr 2025; 12:1526801. [PMID: 39996009 PMCID: PMC11847695 DOI: 10.3389/fnut.2025.1526801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background The current research was to investigate the relationship between prognostic nutritional index (PNI) and mortality, with a focus on all-cause and cardiovascular disease (CVD) mortality, for those with non-alcoholic fatty liver disease (NAFLD). Methods Data from 20,142 patients who participated in the National Health and Nutrition Examination Survey (NHANES), which was carried out between 2005 and 2014, were included in this research. To examine the relationship between PNI and both all-cause and cardiovascular mortality, we employed weighted Cox regression models with multiple variables. Kaplan-Meier survival curves were utilized to visualize the survival distribution across different levels of PNI. The non-linear association between PNI and mortality was addressed through penalized spline smoothing. Subgroup analyses were conducted to examine the potential influence of relevant clinical variables on the relationship between PNI and mortality. The precision of PNI in forecasting the outcome of survival was assessed as well using time-dependent receiver operating characteristic curve (ROC) analysis. Results Kaplan-Meier analysis linked higher PNI to significantly reduced all-cause and CVD mortality. Multivariable Cox models demonstrated that increasing PNI consistently lowered mortality risks. With a threshold value of 50.5, the link between PNI and mortality showed a non-linear pattern after adjusting for confounding factors. Subgroup analyses confirmed robust associations, particularly in race, education, BMI, and fibrosis. Time-dependent ROC analysis highlighted the strong predictive performance of PNI across various time points. Conclusion PNI played a significant role as an effective predictor of prognosis in individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Yuqing Lei
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Shaohong Tao
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yubo Yang
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, Foshan, Guangdong, China
| | - Fang Xie
- Department of Liver Disease, Jinling Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu, China
| | - Weining Xie
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong, China
| |
Collapse
|
36
|
Li Y, Zhao J. Xiaohua Funing decoction ameliorates non-alcoholic fatty liver disease by modulating the gut microbiota and bile acids. Front Microbiol 2025; 16:1511885. [PMID: 40012777 PMCID: PMC11863611 DOI: 10.3389/fmicb.2025.1511885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/07/2025] [Indexed: 02/28/2025] Open
Abstract
Introduction The gut microbiota and bile acids (BAs) have emerged as factors involved in the development of non-alcoholic fatty liver disease (NAFLD). Xiaohua Funing decoction (XFD) is a traditional Chinese medicine formula used for the treatment of NAFLD. Previous studies have indicated that XFD protects liver function, but the underlying mechanism remains unclear. Methods In this study, a Wistar rat model of NAFLD (Mod) was established via a high-fat diet. The effects of obeticholic acid (OCA) and XFD on Mod rats were subsequently evaluated. Wistar rats in the control (Con) group were fed a standard diet. There were eight rats in each group, and the treatment lasted for 12 weeks. Furthermore, metagenomic sequencing and BA metabolomic analyses were performed. Results Compared to the Con group, the Mod group presented significant differences in body and liver weights; serum total cholesterol (TC) and triglyceride (TG) levels; and liver TG, TC, and bile salt hydrolase levels (p < 0.05 or p < 0.01). Importantly, OCA and XFD administration normalized these indicators (p < 0.05 or p < 0.01). Pathology of the liver and white fat steatosis was observed in the Mod group, but steatosis was significantly alleviated in the OCA and XFD groups (p < 0.05 or p < 0.01). The abundances of Bacteroidales_bacterium, Prevotella_sp., bacterium_0.1xD8-71, and unclassified_g_Turicibacter in the Mod group were significantly different from those in the Con group (p < 0.05 or p < 0.01), whereas the abundance of Bacteroidales_bacterium was greater in the XFD group. A total of 17, 24, and 24 differentially abundant BAs were detected in the feces, liver, and serum samples from the Mod and Con groups, respectively (p < 0.05 or p < 0.01). In the feces, liver, and serum, XFD normalized the levels of 16, 23, and 14 BAs, respectively, including glycochenodeoxycholic acid, deoxycholic acid, murideoxycholic acid, lithocholic acid, 23-nordeoxycholic acid, and 3β-ursodeoxycholic acid. In addition, glycochenodeoxycholic acid was identified as a potential biomarker of NAFLD. Discussion In summary, our experiments revealed that XFD regulates the gut microbiota and BAs, providing beneficial effects on liver lipid accumulation in NAFLD.
Collapse
Affiliation(s)
- Yan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jindong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
37
|
Hu W, Gong W, Yang F, Cheng R, Zhang G, Gan L, Zhu Y, Qin W, Gao Y, Li X, Liu J. Dual GIP and GLP-1 receptor agonist tirzepatide alleviates hepatic steatosis and modulates gut microbiota and bile acid metabolism in diabetic mice. Int Immunopharmacol 2025; 147:113937. [PMID: 39752752 DOI: 10.1016/j.intimp.2024.113937] [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: 10/29/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/29/2025]
Abstract
Tirzepatide is a dual agonist of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptors and is a promising therapeutic option for type 2 diabetes mellitus (T2DM). Nevertheless, its effect and underlying mechanism on hepatic steatosis remain ambiguous. Herein, we explored the impact of tirzepatide on improving hepatic steatosis in diabetic mice, with a particular focus on the gut microbiota and bile acids (BAs) using animal models. The tirzepatide effectively reduced body weight, improved insulin resistance, decreased serum and hepatic lipid levels, and mitigated liver injury. Compared to semaglutide, tirzepatide exhibited superior efficacy in reducing hepatic lipid accumulation. 16S rRNA gene sequencing and targeted metabolomics of BAs revealed that tirzepatide ameliorated gut microbiota dysbiosis and BAs metabolism in diabetic mice. Notably, tirzepatide observably increased the abundance of beneficial genera such as Akkermansia, elevated the ratio of farnesoid X receptor (FXR) antagonists (glycoursodeoxycholic acid: GUDCA, β-muricholic acid: β-MCA, hyodeoxycholic acid: HDCA, ursodeoxycholic acid: UDCA) to natural agonists (cholic acid: CA, lithocholic acid: LCA, chenodeoxycholic acid: CDCA, glycocholic acid: GCA, taurodeoxycholic acid: TDCA), and reduced FXR expression in intestinal tissues. In conclusion, tirzepatide attenuated hepatic steatosis in diabetic mice and regulated the gut microbiota and BAs metabolism, which may help to provide a novel therapeutic approach and therapeutic target for metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Weiting Hu
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China; Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Wenyu Gong
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Fan Yang
- The First Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Rui Cheng
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Gerong Zhang
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Lu Gan
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yikun Zhu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Weiwei Qin
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Ying Gao
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Xing Li
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China.
| | - Jing Liu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China.
| |
Collapse
|
38
|
Xu J, Cheng X, Wang Q, Zhang F, Ren X, Huang K, Hu Y, Gao R, Yang K, Yin J, Yang B, He X, Li Y. Artemether Ameliorates Non-Alcoholic Steatohepatitis by Restraining Cross-Talk Between Lipotoxicity-Induced Hepatic Hepatocytes and Macrophages. Phytother Res 2025; 39:604-618. [PMID: 39609107 DOI: 10.1002/ptr.8393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/08/2024] [Accepted: 11/09/2024] [Indexed: 11/30/2024]
Abstract
Non-alcoholic steatohepatitis (NASH) has no effective treatment drug. Our previous study initially found that artemether (Art) treatment significantly attenuates NSAH by regulating liver lipid metabolism. This study further elucidates new mechanisms of Art in improving liver inflammation and provides evidence for drug repurposing. Herein, we utilized HFHF diet-induced animal model and macrophage models to detect the mechanisms of Art in NASH. We confirmed that Art significantly reduced hepatic steatosis, injury, and fibrosis in a high-fat high-fructose (HFHF) diet-induced animal model. Art significantly suppressed the activation of inflammatory macrophages and secretion of pro-inflammatory cytokine (IL-1β) by reducing serum double-stranded DNA (dsDNA) levels and triggering the AIM2/Caspase-1/GSDMD signaling in vivo. dsDNA-induced Caspase-1 and PI-positive cells pyroptosis, AIM2 inflammasome activation, IL-1β, and IL-18 secretion increase were inhibited by Art in vitro. Furthermore, we found Art effectively suppressed mitochondrial DNA (mtDNA), a typical form of dsDNA, released from free fatty acid (FFA)-stressed hepatocytes, which further inhibited AIM2 inflammasome mediated-pyroptosis through decreasing the cleavage of Caspase-1/GSDMD/IL-1β. Moreover, inhibition of the AIM2 gene partly reversed the inhibitory effect of Art on macrophage pyroptosis. Impaired mitochondrial structure and function were confirmed in FFA-stressed hepatocytes and the HFHF-diet-induced NASH mouse model, which was reversed by Art treatment. The present study provides evidence for Art as a potential anti-pyroptosis therapeutic agent for NASH treatment.
Collapse
Affiliation(s)
- Jia Xu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoyan Cheng
- Institute of Analysis and Testing, Beijing Academy of Science and Technology, Beijing, China
| | - Qi Wang
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Feng Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xinxin Ren
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanzhou Hu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ruxin Gao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kun Yang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jingya Yin
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Bingqing Yang
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yue Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Rayapati D, McGlynn KA, Groopman JD, Kim AK. Environmental exposures and the risk of hepatocellular carcinoma. Hepatol Commun 2025; 9:e0627. [PMID: 39813595 PMCID: PMC11737496 DOI: 10.1097/hc9.0000000000000627] [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: 04/29/2024] [Accepted: 11/25/2024] [Indexed: 01/18/2025] Open
Abstract
The global epidemiology of HCC is shifting due to changes in both established and emerging risk factors. This transformation is marked by an emerging prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) and type 2 diabetes, alongside traditional risks such as viral hepatitis (HBV and HCV), and exposure to chemical agents like aflatoxin, alcohol, tobacco, and air pollution. This review examines how environmental exposures and evolving liver pathology, exacerbated by lifestyle and metabolic conditions, are contributing to the rising worldwide incidence of HCC. Effective prevention strategies must not only address traditional risk factors through vaccination and therapeutic measures but also confront metabolic and socioeconomic disparities through comprehensive public health efforts. As the burden of liver cancer continues to grow, particularly in resource-limited settings, an expansive and inclusive approach is vital for mitigating its impact across diverse populations.
Collapse
Affiliation(s)
- Divya Rayapati
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Katherine A. McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - John D. Groopman
- Department of Environmental Health and Engineering, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Amy K. Kim
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Zeng Z, Zhao Z, Yuan Q, Yang S, Wang Z, Wang Z, Zeng S, Li A, Chen Q, Zhu G, Xiao X, Luo G, Luo H, Li J, Zu X, Xie H, Liu J. Hepatic Steatosis Aggravates Vascular Calcification via Extracellular Vesicle-Mediated Osteochondrogenic Switch of Vascular Smooth Muscle Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408660. [PMID: 39680681 PMCID: PMC11791995 DOI: 10.1002/advs.202408660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/18/2024]
Abstract
The global incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) has risen sharply. This condition is strongly associated with the risk of cardiovascular disease (CVD), but how MAFLD affects the development and progression of CVD, particularly concerning vascular calcification, remains unclear. Herein, extracellular vesicles (EVs) are identified from steatotic hepatocytes as a trigger that accelerated the progression of both vascular intimal and medial calcification. Steatotic hepatocytes are found to release more EVs, which are able to reach the vascular tissue, be taken up by vascular smooth muscle cells (VSMCs), and promote their osteogenic differentiation. Within these toxic vesicles, a protein cargo is identified called lectin galactoside-binding soluble 3 binding protein (Lgals3bp) that acted as a potent inducer of osteochondrogenic transformation in VSMCs. Both the inhibition of EV release and the liver-specific knockdown of Lgals3bp profoundly attenuated vascular calcification. This work partially explains the reason for the high incidence of vascular calcification in MAFLD and unveils a novel mechanism that may be used to prevent or treat cardiovascular complications in patients with MAFLD.
Collapse
Affiliation(s)
- Zhao‐Lin Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhi‐Bo Zhao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qing Yuan
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Qi Yang
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhen‐Xing Wang
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zuo Wang
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Yu Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - An‐Qi Li
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qian Chen
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guo‐Qiang Zhu
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xin‐Hua Xiao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guang‐Hua Luo
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hai‐Yan Luo
- Department of GastroenterologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jiao‐Yang Li
- Department of Occupational and Environmental HealthSchool of Public HealthWuhan UniversityWuhan430071P. R. China
| | - Xu‐Yu Zu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hui Xie
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiang‐Hua Liu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| |
Collapse
|
41
|
Li B, Sun J, Zheng Y, Fan X. Letter: Increased Prevalence of Visceral Obesity in Nonresponder Patients With Primary Biliary Cholangitis Based on Computed Tomography. Aliment Pharmacol Ther 2025; 61:589-590. [PMID: 39696728 DOI: 10.1111/apt.18440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Affiliation(s)
- Bo Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Sun
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
- Medical College, Tibet University, Lhasa, China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Alfadda AA, Alqutub AN, Sherbeeni SM, Aldosary AS, Alqahtani SA, Isnani A, Gul R, Khaleel MS, Alqasim SM, Almaghamsi AM. Predictors of liver fibrosis progression in cohort of type 2 diabetes mellitus patients with MASLD. J Diabetes Complications 2025; 39:108910. [PMID: 39675110 DOI: 10.1016/j.jdiacomp.2024.108910] [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: 02/27/2024] [Revised: 09/10/2024] [Accepted: 11/16/2024] [Indexed: 12/17/2024]
Abstract
AIM To investigate predictors of liver fibrosis progression in patients with type 2 diabetes mellitus (T2DM) over a minimum follow-up duration of three years. METHODS Two hundred and thirty-three patients completed the follow-up period and their clinical, laboratory and liver FibroScan data are reported. Patients were categorized into progressors 42 (18.0 %) and non-progressors 191 (82.0 %) based on liver fibrosis progression. Factors influencing fibrosis progression were identified by comparing these groups. RESULTS Progressors showed significantly increased aspartate aminotransferase (AST) (p = 0.010), increased alkaline phosphatase (ALP) (p = 0.001) and decreased platelet count (p = 0.002). Non-progressors exhibited significant decreases in diastolic blood pressure (DBP) (p = 0.050), body mass index (BMI) (p < 0.001), waist circumference (p < 0.001), gamma-glutamyl transferase (GGT) (p < 0.001), albumin (p < 0.001), alanine aminotransferase (ALT) (p = 0.022), glycosylated haemoglobin (HbA1c) (p = 0.002) and fasting blood sugar (FBS) (p = 0.030) with increase in HDL-cholesterol (p < 0.001), creatinine (p < 0.001), bilirubin (p < 0.001), and ALP (p = 0.007). Baseline parameters predictive of liver fibrosis progression included elevated AST and reduced platelet count. Delta changes from baseline to follow-up revealed that increases in ALP, BMI, waist circumference, and reduction in platelet count were correlated with fibrosis progression. Use of GLP-1 receptor agonist was associated with reduced progression. CONCLUSION In conclusion, increase in ALP and waist circumference and reduction in platelet count are predictive of liver fibrosis progression in patients with T2DM. GLP-1 receptor agonists use seems to have a promising protective effect against liver fibrosis progression. CLINICALTRIALS govID:NCT05697991.
Collapse
Affiliation(s)
- Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Obesity, Endocrine and Metabolism Center, King Fahad Medical City, Riyadh, Saudi Arabia.
| | - Adel N Alqutub
- Department of Gastroenterology and Hepatology, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Abdullah S Aldosary
- Department of Medical Imaging, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Saleh A Alqahtani
- Liver Transplant Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Arthur Isnani
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rukhsana Gul
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad S Khaleel
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sara M Alqasim
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
43
|
Xu J, Li QQ, Yang S, Teng HD, Lu ZY, Gu YZ, Xi JH, Mei ZN, Chen Y, Yang GZ. Garcibracgluinols A-C, structurally intriguing polycyclic polyprenylated acylphloroglucinols from Garcinia bracteata alleviate hepatocyte lipid accumulation and insulin resistance. J Mol Struct 2025; 1323:140755. [DOI: 10.1016/j.molstruc.2024.140755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
44
|
Castera L, Alazawi W, Bugianesi E, Caussy C, Federici M, Romero‐Gómez M, Schattenberg JM, Basuroy R, Prasad P, Estulin D, Lazarus JV. A European Survey to Identify Challenges in the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int 2025; 45:e16224. [PMID: 39752213 PMCID: PMC11698224 DOI: 10.1111/liv.16224] [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: 10/03/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) and its more severe subtype, metabolic dysfunction-associated steatohepatitis (MASH), are highly prevalent and strongly associated with obesity and type 2 diabetes (T2D). This study sought to identify challenges to the diagnosis, treatment and management of people living with MASLD and MASH and understand the key barriers to adopting relevant clinical guidelines. METHODS A real-world, cross-sectional study (BARRIERS-MASLD) consisting of a quantitative survey and qualitative interviews of physicians in France, Germany, Italy, Spain and the United Kingdom was conducted from March to September 2023. Descriptive statistics were used for data analysis. RESULTS A total of 626 physicians completed the survey; n = 10 from each country participated in the qualitative interviews. Physicians considered the presence of MASH to be highly impactful on how they treated people living with obesity (66%) and T2D (69%). Over one-third (35%) of the respondents could not identify any MASH-specific clinical guidelines issued by medical societies or associations top-of-mind, but overall awareness rose when prompted about country-specific guidelines. Physicians said they would need evidence of success (48%) and clinical guidelines that address common MASLD comorbidities (38%) to increase their adoption. CONCLUSIONS This study found that lack of awareness around MASLD and MASH clinical guidelines and clearly established care pathways, particularly for addressing common comorbidities, was a key factor preventing physicians from optimising care for people living with MASH in Europe. This research highlights opportunities to improve education and training about clinical guidelines and care coordination.
Collapse
Affiliation(s)
- Laurent Castera
- Department of HepatologyBeaujon Hospital, AP‐HP, Université Paris‐CitéClichyFrance
| | | | | | | | - Massimo Federici
- Department of Systems MedicineUniversity of Rome tor VergataRomeItaly
| | - Manuel Romero‐Gómez
- UCM Digestive Diseases and Ciberehd, Virgen Del Rocío University Hospital, Institute of Biomedicine of Seville (CSIC/HUVR/US)University of SevilleSevilleSpain
| | - Jörn M. Schattenberg
- Department of Internal Medicine IISaarland University Medical CenterHomburgGermany
| | | | | | | | - Jeffrey V. Lazarus
- City University of New York Graduate School for Public Health and Health Policy (CUNY SPH)New YorkNew YorkUSA
- Barcelona Institute for Global Health (ISGlobal), Hospital ClinicUniversity of BarcelonaBarcelonaSpain
- Faculty of Medicine and Health SciencesUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
45
|
Özdede M, Güven AT, Eroğlu BÇ. In-depth assessment of steatotic liver disease awareness in high-risk groups. J Eval Clin Pract 2025; 31:e14091. [PMID: 38993006 DOI: 10.1111/jep.14091] [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: 01/30/2024] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVES This study aims to determine the awareness levels and factors affecting it, along with prevalent misconceptions about Steatotic Liver Disease (SLD) among participants with high-risk indicators. METHODS A questionnaire with open-ended questions was utilized. Participants were recruited from two general internal medicine outpatient clinics, focusing on those with high-risk indicators for SLD. Data collection involved a questionnaire covering demographic information, self-reported clinical conditions, and open-ended questions about SLD awareness. Key focus areas included misconceptions, thematic awareness, and the relationship between awareness and educational attainment. RESULTS The study involved 228 participants, predominantly female (70.4%), with an average age of 53.8 years. Only 33.7% showed a comprehensive understanding of all aspects of SLD. However, 90.4% provided some accurate information, though often limited or incomplete. Higher education and awareness of SLD risks were key predictors of better understanding. The logistic regression model, with an accuracy of 0.76 and recall of 0.84, found higher education inversely related to low awareness. Common misconceptions highlighted included the belief that polypharmacy or certain medications cause SLD, fatigue as an effect, and increased water intake as a treatment. Notably, seven patients mentioned artichoke consumption as a potential treatment. CONCLUSION The findings highlight the gap between comprehensive and partial awareness of SLD among high-risk individuals. Educational level and informed understanding of SLD risks are crucial for improving awareness, emphasizing the need for specialized educational efforts and risk communication to high-risk patients.
Collapse
Affiliation(s)
- Murat Özdede
- Department of Internal Medicine, Division of General Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Türkiye
- Center for Genomics and Rare Diseases, Hacettepe University, Ankara, Türkiye
| | - Alper Tuna Güven
- Department of Internal Medicine, Division of General Internal Medicine, Başkent University Faculty of Medicine, Ankara, Türkiye
| | - Burcu Çelik Eroğlu
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
46
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
47
|
Zhao K, Zhang H, Ding W, Yu X, Hou Y, Liu X, Li X, Wang X. Adipokines regulate the development and progression of MASLD through organellar oxidative stress. Hepatol Commun 2025; 9:e0639. [PMID: 39878681 PMCID: PMC11781772 DOI: 10.1097/hc9.0000000000000639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), which is increasingly being recognized as a leading cause of chronic liver pathology globally, is increasing. The pathophysiological underpinnings of its progression, which is currently under active investigation, involve oxidative stress. Human adipose tissue, an integral endocrine organ, secretes an array of adipokines that are modulated by dietary patterns and lifestyle choices. These adipokines intricately orchestrate regulatory pathways that impact glucose and lipid metabolism, oxidative stress, and mitochondrial function, thereby influencing the evolution of hepatic steatosis and progression to metabolic dysfunction-associated steatohepatitis (MASH). This review examines recent data, underscoring the critical interplay of oxidative stress, reactive oxygen species, and redox signaling in adipokine-mediated mechanisms. The role of various adipokines in regulating the onset and progression of MASLD/MASH through mitochondrial dysfunction and endoplasmic reticulum stress and the underlying mechanisms are discussed. Due to the emerging correlation between adipokines and the development of MASLD positions, these adipokines are potential targets for the development of innovative therapeutic interventions for MASLD management. A comprehensive understanding of the pathogenesis of MASLD/MASH is instrumental for identifying therapies for MASH.
Collapse
Affiliation(s)
- Ke Zhao
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Heng Zhang
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- Central laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenyu Ding
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xiaoshuai Yu
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- Central laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanli Hou
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xihong Liu
- Department of Pathology, The Fourth People’s Hospital of Jinan, Jinan, Shandong, China
| | - Xinhua Li
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xiaolei Wang
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- First school of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
48
|
Jokisch F, Geyer LJM, Janssen KP. Liver regeneration in fatty liver disease: can metabolomics shed light on the contribution of the gut microbiome? EBioMedicine 2025; 112:105552. [PMID: 39798398 PMCID: PMC11774801 DOI: 10.1016/j.ebiom.2024.105552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025] Open
Affiliation(s)
- Florian Jokisch
- Department of Surgery, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | | | - Klaus-Peter Janssen
- Department of Surgery, Technical University of Munich, School of Medicine and Health, Munich, Germany.
| |
Collapse
|
49
|
Tong X, Sun Y, Wang Q, Zhao X, Chen W, Zhang M, Ren Y, Zhao X, Wu X, Zhao J, Sun C, Zheng M, Ren H, Yang Z, Ou X, Jia J, You H. Delicate and thin fibrous septa indicate a regression tendency in metabolic dysfunction-associated steatohepatitis patients with advanced fibrosis. Hepatol Int 2025; 19:166-180. [PMID: 39152361 DOI: 10.1007/s12072-024-10719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatohepatitis (MASH)-related fibrosis is reversible. However, the dynamic morphology change in fibrosis regression remains unclear. We aim to explore the morphological characteristics of fibrosis regression in advanced MASH patients. METHODS Clinical and histological data of 79 biopsy-proved MASH patients with advanced fibrosis (F3-F4) were reviewed. The second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) image technology was used to quantitatively identify the R (regressive) septa from P (progressive) septa and PS (perisinusoidal) fibrosis. Non-invasive tests were used to compare the fibrosis level with and without R septa groups. Transcriptomics was used to explore hub genes and the underlying mechanism of the formation of R septa. RESULTS The R septa were different from the P septa and PS fibrosis in detail collagen quantitation identified by SHG/TPEF technology. The R septa were found in MASH fibrosis-regressed patients, which met the definition of the "Beijing classification". Therefore, patients were divided into two groups according to septa morphology: with R septa (n = 10, 12.7%), and without R septa (n = 69, 87.3%). Patients with R septa had lower values in most non-invasive tests, especially for liver stiffness assessed by TE (12.3 vs. 19.4 kPa, p = 0.010) and FAST (FibroScan®-AST) score (0.43 vs. 0.70, p = 0.003). Transcriptomics analysis showed that the expressions of five hub fibrogenic genes, including Col3A1, BGN, Col4A1, THBS2, and Col4A2 in the R septa group, were significantly lower. CONCLUSIONS The R septa can be differentiated from the P septa and PS fibrosis by quantitative assessment of SHG/TPEF, and it represents a tendency of fibrosis regression in MASH patients. TRIAL REGISTRATION NCT03386890, 29/12/2017.
Collapse
Affiliation(s)
- Xiaofei Tong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Qianyi Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Wei Chen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Mengyang Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Yayun Ren
- HistoIndex Pte Ltd, Singapore, Singapore
| | - Xinyu Zhao
- Clinical Epidemiology and EBM Unit, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Jingjie Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Chenglin Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Minghua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Hao Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China.
| |
Collapse
|
50
|
Tiwari K, Kumar B, Tiwari A, Dhamija P, Vardhan G, Dehade A, Kumar V. In Silico Analysis of Saroglitazar and Ferulic Acid Binding to Human Ketohexokinase: Implications for Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Cureus 2025; 17:e79437. [PMID: 40130107 PMCID: PMC11931454 DOI: 10.7759/cureus.79437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease renamed as metabolic dysfunction-associated steatotic liver disease (MASLD) and a global health issue that causes excessive liver fat deposition without alcohol usage. Basic fatty liver to non-alcoholic steatohepatitis can lead to liver fibrosis, cirrhosis, and hepatocellular carcinoma. Role of research is vital due to the multifaceted, complex pathophysiology and the increasing incidence of a sedentary lifestyle. Computational network pharmacology, docking and dynamics studies of saroglitazar and ferulic acid with human ketohexokinase (KHK) were conducted to propose potential MASLD management. METHOD Utilized computational methodologies were utilized to examine binding interactions of saroglitazar (compound identifier (CID): 60151560) and ferulic acid (CID: 445858) with human ketohexokinase (KHK: P50053, Protein Data Bank (PDB) ID: 6W0W). Active site analysis was done by using the Conserved Domain Database (CDD) server (Collaborative Drug Discovery, Burlingame, California) and BIOVIA Discovery Studio 2019 (Dassault Systèmes, Vélizy-Villacoublay, France). The best PDB complex was used for molecular dynamics simulation and trajectory analysis on 100 ns, and functional associations were checked based on network analysis using the Search Tool for Interactions of Chemicals (STITCH) server (STITCH Consortium (EMBL), Heidelberg, Germany). RESULTS Human ketohexokinase (KHK) protein (UniProt ID: P50053) was obtained. Additional KHK PDB Structure (6W0W) was retrieved for docking calculation. PubChem Database 2 Structure-Data File (SDF) files (National Center for Biotechnology Information (NCBI), U.S. National Library of Medicine, Bethesda, Maryland), ferulic acid (CID: 445858) and saroglitazar (CID: 60151560) were used as ligands. Active site residues were identified using the CDD server and BIOVIA Discovery Studio 2019. Further, identified active site residues, i.e., Arg108, Trp225, Glu227, Gly229, Ala230, Pro246, Pro247, Val250, Thr253, Gly257, Cys282, Gly286, and Cys289 were used as potential active site for docking. D. E. Shaw Research Molecular Dynamics (DESMOND, Schrödinger, Inc., New York) was used for molecular dynamics simulation and trajectory analysis equilibrated after 40 ns in best-docked complex (saroglitazar (CID: 60151560) and KHK; binding energy: -21 kcal/mol). CONCLUSION The study shows that saroglitazar and ferulic acid are potent KHK inhibitors for metabolic diseases, including MASLD, suggesting multi-target treatments.
Collapse
Affiliation(s)
- Kalpana Tiwari
- Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Brijesh Kumar
- Pharmacology and Therapeutics, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Anurag Tiwari
- Gastroenterology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Puneet Dhamija
- Clinical Pharmacology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Gyan Vardhan
- Pharmacology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Amol Dehade
- Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| | - Vinay Kumar
- School of Biotechnology, Center for Bioinformatics, Institute of Medical Science, Banaras Hindu University, Varanasi, IND
| |
Collapse
|